1
|
Brahmbhatt DH, Scolari FL, Fung NL, Otsuki M, Lawler PR, Ross HJ, Kuzmanov U, Gramolini AO, Luk AC, Billia F. Heart failure decompensation with cardiogenic shock exhibits distinct sequential inflammatory profiles. ESC Heart Fail 2025; 12:2077-2086. [PMID: 39925014 PMCID: PMC12055340 DOI: 10.1002/ehf2.15217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 11/26/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
AIMS The inflammatory profile of cardiogenic shock (CS) after myocardial infarction affects outcomes; however, little is known about the impact of inflammatory changes in CS caused by acute decompensated heart failure (ADHF-CS). We measured levels of inflammatory cytokines in patients with ADHF-CS admitted to a cardiac intensive care unit (CICU). METHODS We identified patients admitted to our CICU with ADHF-CS who had consented to having biospecimens stored. We identified two comparator groups of patients with HF seen as outpatients with stored biospecimens: firstly, those who had no history of decompensation and did not develop CS during follow-up after sample acquisition (stable HF), and secondly, a group of patients who developed CS during follow-up (pre-CS). All samples underwent 48-plex cytokine and white blood cell differential testing with the differences between groups analysed by comparing means. RESULTS Eighty-four ADHF-CS patients were identified who had samples obtained at a median of 2 [inter-quartile range (IQR) 0-7] days after CICU admission. Thirty-six pre-CS outpatients had samples taken 137 (IQR 41-258) days before admission with CS, and 338 stable HF control patients were included. Cytokine profiles differed between ADHF-CS and stable HF. Patients with CS had higher pro-inflammatory cytokine levels [including interleukin-1 (IL-1), interleukin-6 (IL-6) and interleukin-8 (IL-8)] and total white cell counts than stable HF patients. Analysis of the pre-CS outpatient group suggested an intermediate stage in subacute transition to CS. CONCLUSIONS ADHF-CS is characterized by high levels of pro-inflammatory cytokines and total white count, compared with ambulatory HF. Decompensation from HF has two distinct inflammatory phases that may help identify outpatients at risk of CS.
Collapse
Affiliation(s)
- Darshan H. Brahmbhatt
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac CentreUniversity Health NetworkTorontoOntarioCanada
- Division of Cardiology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteTorontoOntarioCanada
- Division of Cardiology, Mount Sinai HospitalSinai Health SystemTorontoOntarioCanada
| | - Fernando Luis Scolari
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac CentreUniversity Health NetworkTorontoOntarioCanada
- Division of Cardiology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteTorontoOntarioCanada
- Heart Transplant Program, Division of CardiologyHospital de Clinicas de Porto AlegrePorto AlegreBrazil
| | - Nicole L. Fung
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac CentreUniversity Health NetworkTorontoOntarioCanada
| | - Madison Otsuki
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac CentreUniversity Health NetworkTorontoOntarioCanada
| | - Patrick R. Lawler
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac CentreUniversity Health NetworkTorontoOntarioCanada
- Division of Cardiology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
- McGill University Health CentreMontrealQuebecCanada
| | - Heather J. Ross
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac CentreUniversity Health NetworkTorontoOntarioCanada
- Division of Cardiology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteTorontoOntarioCanada
| | - Uros Kuzmanov
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
- Translational Biology and Engineering ProgramTed Rogers Centre for Heart ResearchTorontoOntarioCanada
| | - Anthony O. Gramolini
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
- Translational Biology and Engineering ProgramTed Rogers Centre for Heart ResearchTorontoOntarioCanada
| | - Adriana C. Luk
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac CentreUniversity Health NetworkTorontoOntarioCanada
- Division of Cardiology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
| | - Filio Billia
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac CentreUniversity Health NetworkTorontoOntarioCanada
- Division of Cardiology, Department of MedicineUniversity of TorontoTorontoOntarioCanada
- Toronto General Hospital Research InstituteTorontoOntarioCanada
- Department of PhysiologyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
2
|
Blagov AV, Churov AV, Starodubtseva IA, Kovyanova TI, Pecherina TB, Sukhorukov VN. Cytokines are the Basis of the Development and Suppression of Inflammation in Atherosclerosis. Rev Cardiovasc Med 2025; 26:26421. [PMID: 40160593 PMCID: PMC11951496 DOI: 10.31083/rcm26421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 04/02/2025] Open
Abstract
Cardiovascular diseases continue to be the primary cause of mortality in industrialised countries, and atherosclerosis plays a role in their development. A persistent inflammatory condition affecting big and medium-sized arteries is known as atherosclerosis. It is brought on by dyslipidemia and is facilitated by the immune system's innate and adaptive components. At every stage of the progression of atherosclerosis, inflammation plays a crucial role. It has been demonstrated that soluble factors, or cytokines, activate cells involved in the pathophysiology of atherosclerosis and have a significant impact on disease progression. Anti-inflammatory cytokines (such as interleukin (IL)-5 and IL-13) mitigate atherosclerosis, whereas pro-inflammatory cytokines (such as IL-1, IL-6) quicken the disease's course. Of interest is the fact that a number of cytokines can exhibit both atherogenic and atheroprotective properties, which is the topic of study and discussion in this review. This review provides a comparative analysis of the functions of the main cytokines involved in the pathogenesis of atherosclerosis. Their functional relationships with each other are also shown. In addition, potential therapeutic strategies targeting these cytokines for the treatment of atherosclerosis are proposed, with an emphasis on recent clinical research in this area.
Collapse
Affiliation(s)
| | - Alexey V. Churov
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Institute on Aging Research, Russian Gerontology Clinical Research Centre, Pirogov Russian National Research Medical University, 129226 Moscow, Russia
| | - Irina A. Starodubtseva
- Department of Polyclinic Therapy, NN Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | - Tatiana I. Kovyanova
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Institute for Atherosclerosis Research, 121609 Moscow, Russia
| | - Tamara B. Pecherina
- Laboratory for Cardiac Fibrosis, Research Institute for Complex Issues of Cardiovascular Diseases, 650002 Kemerovo, Russia
| | - Vasily N. Sukhorukov
- Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
- Institute of Experimental Cardiology, Russian Medical Research Center of Cardiology, 121552 Moscow, Russia
| |
Collapse
|
3
|
Lunde IG, Rypdal KB, Van Linthout S, Diez J, González A. Myocardial fibrosis from the perspective of the extracellular matrix: Mechanisms to clinical impact. Matrix Biol 2024; 134:1-22. [PMID: 39214156 DOI: 10.1016/j.matbio.2024.08.008] [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: 05/07/2024] [Revised: 08/08/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Fibrosis is defined by the excessive accumulation of extracellular matrix (ECM) and constitutes a central pathophysiological process that underlies tissue dysfunction, across organs, in multiple chronic diseases and during aging. Myocardial fibrosis is a key contributor to dysfunction and failure in numerous diseases of the heart and is a strong predictor of poor clinical outcome and mortality. The excess structural and matricellular ECM proteins deposited by cardiac fibroblasts, is found between cardiomyocytes (interstitial fibrosis), in focal areas where cardiomyocytes have died (replacement fibrosis), and around vessels (perivascular fibrosis). Although myocardial fibrosis has important clinical prognostic value, access to cardiac tissue biopsies for histological evaluation is limited. Despite challenges with sensitivity and specificity, cardiac magnetic resonance imaging (CMR) is the most applicable diagnostic tool in the clinic, and the scientific community is currently actively searching for blood biomarkers reflecting myocardial fibrosis, to complement the imaging techniques. The lack of mechanistic insights into specific pro- and anti-fibrotic molecular pathways has hampered the development of effective treatments to prevent or reverse myocardial fibrosis. Development and implementation of anti-fibrotic therapies is expected to improve patient outcomes and is an urgent medical need. Here, we discuss the importance of the ECM in the heart, the central role of fibrosis in heart disease, and mechanistic pathways likely to impact clinical practice with regards to diagnostics of myocardial fibrosis, risk stratification of patients, and anti-fibrotic therapy.
Collapse
Affiliation(s)
- Ida G Lunde
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway.
| | - Karoline B Rypdal
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Javier Diez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| |
Collapse
|
4
|
Salah A, Bouzid F, Dhouib W, Benmarzoug R, Triki N, Rebai A, Kharrat N. Integrative Bioinformatics Approaches to Uncover Hub Genes and Pathways Involved in Cardiovascular Diseases. Cell Biochem Biophys 2024; 82:2107-2127. [PMID: 38809349 DOI: 10.1007/s12013-024-01319-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
Cardiovascular diseases (CVD) represent a significant global health challenge resulting from a complex interplay of genetic, environmental, and lifestyle factors. However, the molecular pathways and genetic factors involved in the onset and progression of CVDs remain incompletely understood. Here, we performed an integrative bioinformatic analysis to highlight specific genes and signaling pathways implicated in the pathogenesis of 80 CVDs. Differentially expressed genes (DEGs) were identified through the integrated analysis of microarray and GWAS datasets. Then, hub genes were identified after gene ontology functional annotation analysis and protein-protein internet (PPI) analysis. In addition, pathways were identified through KEGG and gene ontology enrichment analyses. A total of 821 hub genes related to 80 CVDs were identified, including 135 common and frequent CVD-associated genes. TNF, IL6, VEGFA, and TGFB.1 genes were the central core genes expressed in 50% or more of CVDs, confirming that the inflammation is a key pathological feature of CVDs. Analysis of hub genes by KEGG enrichment revealed predominant enrichment in 201 KEGG pathways, of which the AGE-RAGE signaling pathway in diabetic complications was identified as the common key KEGG implicated in 62 CVDs. In addition, the outcomes showed an overrepresentation in pathways categorized under human diseases, particularly in the subcategories of infectious diseases and cancers, which may be common risk factors for CVDs. In conclusion, this powerful approach for in silico fine-mapping of genes and pathways allowed the identification of determinant hubs genes and pathways implicated in the pathogenesis of CVDs which could be employed in developing more targeted and effective interventions for preventing, diagnosing, and treating CVDs. The function of these hub genes in CVDs needs further exploration to elucidate their biological characteristics.
Collapse
Affiliation(s)
- Awatef Salah
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia.
| | - Fériel Bouzid
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Wala Dhouib
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Riadh Benmarzoug
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Nesrine Triki
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Ahmed Rebai
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Najla Kharrat
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
5
|
Alkhatib B, Ciarelli J, Ghnenis A, Pallas B, Olivier N, Padmanabhan V, Vyas AK. Early- to mid-gestational testosterone excess leads to adverse cardiac outcomes in postpartum sheep. Am J Physiol Heart Circ Physiol 2024; 327:H315-H330. [PMID: 38819385 PMCID: PMC11687963 DOI: 10.1152/ajpheart.00763.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 05/13/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
Cardiovascular dysfunctions complicate 10-20% of pregnancies, increasing the risk for postpartum mortality. Various gestational insults, including preeclampsia are reported to be associated with adverse maternal cardiovascular outcomes. One such insult, gestational hyperandrogenism increases the risk for preeclampsia and other gestational morbidities but its impact on postpartum maternal health is not well known. We hypothesize that gestational hyperandrogenism such as testosterone (T) excess will adversely impact the maternal heart in the postpartum period. Pregnant ewes were injected with T propionate from day 30 to day 90 of gestation (term 147 days). Three months postpartum, echocardiograms, plasma cytokine profiles, cardiac morphometric, and molecular analysis were conducted [control (C) n = 6, T-treated (T) n = 7 number of animals]. Data were analyzed by two-tailed Student's t test and Cohen's effect size (d) analysis. There was a nonsignificant large magnitude decrease in cardiac output (7.64 ± 1.27 L/min vs. 10.19 ± 1.40, P = 0.22, d = 0.81) and fractional shortening in the T ewes compared with C (35.83 ± 2.33% vs. 41.50 ± 2.84, P = 0.15, d = 0.89). T treatment significantly increased 1) left ventricle (LV) weight-to-body weight ratio (2.82 ± 0.14 g/kg vs. 2.46 ± 0.08) and LV thickness (14.56 ± 0.52 mm vs. 12.50 ± 0.75), 2) proinflammatory marker [tumor necrosis factor-alpha (TNF-α)] in LV (1.66 ± 0.35 vs. 1.06 ± 0.18), 3) LV collagen (Masson's Trichrome stain: 3.38 ± 0.35 vs. 1.49 ± 0.15 and Picrosirius red stain: 5.50 ± 0.32 vs. 3.01 ± 0.23), 4) markers of LV apoptosis, including TUNEL (8.3 ± 1.1 vs. 0.9 ± 0.18), bcl-2-associated X protein (Bax)+-to-b-cell lymphoma 2 (Bcl2)+ ratio (0.68 ± 0.30 vs. 0.13 ± 0.02), and cleaved caspase 3 (15.4 ± 1.7 vs. 4.4 ± 0.38). These findings suggest that gestational testosterone excess adversely programs the maternal LV, leading to adverse structural and functional consequences in the postpartum period.NEW & NOTEWORTHY Using a sheep model of human translational relevance, this study provides evidence that excess gestational testosterone exposure such as that seen in hyperandrogenic disorders adversely impacts postpartum maternal hearts.
Collapse
Affiliation(s)
- Bashar Alkhatib
- Department of Pediatrics, Washington University, St Louis, Missouri, United States
| | - Joseph Ciarelli
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Adel Ghnenis
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Brooke Pallas
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Nicholas Olivier
- Department of Veterinary Medicine, Michigan State University, Lansing, Michigan, United States
| | - Vasantha Padmanabhan
- Department of Pediatrics, University of Michigan, Ann Arbor, Michigan, United States
| | - Arpita Kalla Vyas
- Department of Pediatrics, Washington University, St Louis, Missouri, United States
| |
Collapse
|
6
|
Efentakis P, Varela A, Lamprou S, Papanagnou ED, Chatzistefanou M, Christodoulou A, Davos CH, Gavriatopoulou M, Trougakos I, Dimopoulos MA, Terpos E, Andreadou I. Implications and hidden toxicity of cardiometabolic syndrome and early-stage heart failure in carfilzomib-induced cardiotoxicity. Br J Pharmacol 2024; 181:2964-2990. [PMID: 38679957 DOI: 10.1111/bph.16391] [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: 03/16/2023] [Revised: 07/25/2023] [Accepted: 09/12/2023] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND AND PURPOSE Cancer therapy-related cardiovascular adverse events (CAEs) in presence of comorbidities, are in the spotlight of the cardio-oncology guidelines. Carfilzomib (Cfz), indicated for relapsed/refractory multiple myeloma (MM), presents with serious CAEs. MM is often accompanied with co-existing comorbidities. However, Cfz use in MM patients with cardiometabolic syndrome (CMS) or in heart failure with reduced ejection fraction (HFrEF), is questionable. EXPERIMENTAL APPROACH ApoE-/- and C57BL6/J male mice received 14 weeks Western Diet (WD) (CMS models). C57BL6/J male mice underwent permanent LAD ligation for 14 days (early-stage HFrEF model). CMS- and HFrEF-burdened mice received Cfz for two consecutive or six alternate days. Daily metformin and atorvastatin administrations were performed additionally to Cfz, as prophylactic interventions. Mice underwent echocardiography, while proteasome activity, biochemical and molecular analyses were conducted. KEY RESULTS CMS did not exacerbate Cfz left ventricular (LV) dysfunction, whereas Cfz led to metabolic complications in both CMS models. Cfz induced autophagy and Ca2+ homeostasis dysregulation, whereas metformin and atorvastatin prevented Cfz-mediated LV dysfunction and molecular deficits in the CMS-burdened myocardium. Early-stage HFrEF led to depressed LV function and increased protein phosphatase 2A (PP2A) activity. Cfz further increased myocardial PP2A activity, inflammation and Ca2+-cycling dysregulation. Metformin co-administration exerted an anti-inflammatory potential on the myocardium without improving LV function. CONCLUSION AND IMPLICATIONS CMS and HFrEF seem to exacerbate Cfz-induced CAEs, by presenting metabolism-related hidden toxicity and PP2A-related cardiac inflammation, respectively. Metformin retains its prophylactic potential in the presence of CMS, while mitigating inflammation and Ca2+ signalling dysregulation in the HFrEF myocardium.
Collapse
Affiliation(s)
- Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Aimilia Varela
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Sofia Lamprou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Michail Chatzistefanou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Andriana Christodoulou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, Plasma Cell Dyscrasias Unit, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Ioannis Trougakos
- Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios Athanasios Dimopoulos
- Department of Clinical Therapeutics, Plasma Cell Dyscrasias Unit, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Evangelos Terpos
- Department of Clinical Therapeutics, Plasma Cell Dyscrasias Unit, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
7
|
Aggarwal K, Singh S, Singla A, Kanagala SG, Anamika F, Singh B, Aggarwal P, Jain R. Unveiling the Silent Intruder: H. pylori's Hidden Link to Ischemic Heart Disease. Cardiol Rev 2024:00045415-990000000-00227. [PMID: 38445894 DOI: 10.1097/crd.0000000000000686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
Cardiovascular disease is the leading cause of death. In addition to the well-known risk factors associated with cardiovascular disease, such as age, diabetes mellitus, smoking, hypertension, and obesity, there has been a growing concern regarding cardiac complications stemming from the Gram-negative bacteria Helicobacter pylori. While H. pylori is most commonly associated with chronic gastritis, peptic ulcer disease, gastric adenocarcinoma, and gastric lymphoma, it has also been implicated in extra gastric manifestations, encompassing cardiac, neurologic, ocular, and dermatologic issues. Key virulent factors for coronary artery disease include the vacuolating cytotoxin gene A and the cytotoxin-associated gene A. The most likely pathogenic mechanism of the relationship between H. pylori and coronary artery disease is initiating a chronic inflammatory process associated with infection and the modifications of classic risk factors. These alterations lead to the creation of prothrombotic and procoagulant environments. Here, we review the cardiac manifestations of H. pylori and the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Kanishk Aggarwal
- From the Department of Internal Medicine, Dayanand Medical College and Hospital, Ludhiana, Punjab, IndiaDepartment of Internal Medicine
| | - Sandeep Singh
- Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Ankur Singla
- From the Department of Internal Medicine, Dayanand Medical College and Hospital, Ludhiana, Punjab, IndiaDepartment of Internal Medicine
| | | | - Fnu Anamika
- Department of Internal Medicine, University College of Medical Sciences, New Delhi, India
| | - Bhupinder Singh
- Department of Internal Medicine, Government Medical College, Amritsar, India
| | - Priyanka Aggarwal
- Department of Internal Medicine, Maharishi Markandeshwar Institute of Medical Science & Research, Mullana, Haryana, India
| | - Rohit Jain
- Department of Internal Medicine, Penn State Milton S. Hershey Medical Center, Hershey, PA
| |
Collapse
|
8
|
Sarfraz A, Sarfraz Z, Bano S, Sarfraz M, Jaan A, Minhas A, Razzack AA, Patel G, Manish KC, Makkar SS, Garimella R, Pandav K, Almonte J, Paul T, Almonte T, Jimenez L, Pantoga JC, El Mazboudi N, Yatzkan G, Michel G, Michel J. Global Perspective on COVID-19 Therapies, Cardiovascular Outcomes, and Implications for Long COVID: A State-of-the-Art Review. J Community Hosp Intern Med Perspect 2024; 14:58-66. [PMID: 38966504 PMCID: PMC11221457 DOI: 10.55729/2000-9666.1308] [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: 09/07/2023] [Revised: 12/03/2023] [Accepted: 01/02/2024] [Indexed: 07/06/2024] Open
Abstract
The COVID-19 pandemic has resulted in many therapies, of which many are repurposed and used for other diseases in the last decade such in Influenza and Ebola. We intend to provide a robust foundation for cardiovascular outcomes of the therapies to better understand the rationale for the clinical trials that were conducted during the COVID-19 pandemic, and to gain more clarity on the steps moving forward should the repurposing provide clinical benefit in pandemic situations. With this state-of-the-art review, we aim to improve the understanding of the cardiovascular involvement of the therapies prior to, during, and after the COVID-19 pandemic to provide meaningful findings to the cardiovascular specialists and clinical trials for therapies, moving on from the period of pandemic urgency.
Collapse
Affiliation(s)
| | | | - Shehar Bano
- Fatima Jinnah Medical University, Lahore,
Pakistan
| | | | - Ali Jaan
- Rochester General Hospital, Rochester, NY,
USA
| | - Amna Minhas
- Fatima Jinnah Medical University, Lahore,
Pakistan
| | | | | | - KC Manish
- Larkin Health System, South Miami, Florida,
USA
| | | | | | | | | | - Trissa Paul
- Larkin Health System, South Miami, Florida,
USA
| | | | | | | | | | | | | | - Jack Michel
- Larkin Health System, South Miami, Florida,
USA
| |
Collapse
|
9
|
Hegemann N, Barth L, Döring Y, Voigt N, Grune J. Implications for neutrophils in cardiac arrhythmias. Am J Physiol Heart Circ Physiol 2024; 326:H441-H458. [PMID: 38099844 PMCID: PMC11219058 DOI: 10.1152/ajpheart.00590.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 02/03/2024]
Abstract
Cardiac arrhythmias commonly occur as a result of aberrant electrical impulse formation or conduction in the myocardium. Frequently discussed triggers include underlying heart diseases such as myocardial ischemia, electrolyte imbalances, or genetic anomalies of ion channels involved in the tightly regulated cardiac action potential. Recently, the role of innate immune cells in the onset of arrhythmic events has been highlighted in numerous studies, correlating leukocyte expansion in the myocardium to increased arrhythmic burden. Here, we aim to call attention to the role of neutrophils in the pathogenesis of cardiac arrhythmias and their expansion during myocardial ischemia and infectious disease manifestation. In addition, we will elucidate molecular mechanisms associated with neutrophil activation and discuss their involvement as direct mediators of arrhythmogenicity.
Collapse
Affiliation(s)
- Niklas Hegemann
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Lukas Barth
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| | - Yannic Döring
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Georg August University Göttingen, Göttingen, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Jana Grune
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Berlin, Germany
| |
Collapse
|
10
|
Athari SS, Mehrabi Nasab E, Jing K, Wang J. Interaction between cardiac resynchronization therapy and cytokines in heart failure patients. Cytokine 2024; 175:156479. [PMID: 38199086 DOI: 10.1016/j.cyto.2023.156479] [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: 04/20/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024]
Abstract
Congestive heart failure (CHF) is a complex multistage syndrome that has a great financial burden on human societies. It was known that the damaged myocardium sends a signal to stimulate the immune system and proliferation of leukocytes. In continuous, cytokine storm can be initiated and causes the probability of CHF. Persistent inflammation by increasing the levels of pro-inflammatory cytokines, plays an important role in the pathogenesis of CHF and causes remodeling, which is a progressive processs. Although treatment by drugs can reduce mortality and partially control the symptoms of heart failure patients, but complications and mortality are still high. Therefore, other treatment options such as Cardiac Resynchronization Therapy (CRT) are necessary. Today, it is known that CRT can be an effective treatment for many patients with heart failure. CRT is novel, non-pharmacological, and device-based therapy that would be beneficial to know more about its performance in the management of heart failure. In this study, we have reviewed the immunological processes involved in heart failure and the effect of CRT in controlling of the cytokine storm.
Collapse
Affiliation(s)
- Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Entezar Mehrabi Nasab
- Department of Cardiology, School of Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Cardiology, School of Medicine, Valiasr Hospital, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Kai Jing
- Department of Proctology, The People's Hospital of Huaiyin Jinan, 250021 Shandong, China
| | - Jin Wang
- Department of Cardiology, The Fifth People's Hospital of Jinan, 250022 Shandong, China.
| |
Collapse
|
11
|
Markousis-Mavrogenis G, Baumhove L, Al-Mubarak AA, Aboumsallem JP, Bomer N, Voors AA, van der Meer P. Immunomodulation and immunopharmacology in heart failure. Nat Rev Cardiol 2024; 21:119-149. [PMID: 37709934 DOI: 10.1038/s41569-023-00919-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 09/16/2023]
Abstract
The immune system is intimately involved in the pathophysiology of heart failure. However, it is currently underused as a therapeutic target in the clinical setting. Moreover, the development of novel immunomodulatory therapies and their investigation for the treatment of patients with heart failure are hampered by the fact that currently used, evidence-based treatments for heart failure exert multiple immunomodulatory effects. In this Review, we discuss current knowledge on how evidence-based treatments for heart failure affect the immune system in addition to their primary mechanism of action, both to inform practising physicians about these pleiotropic actions and to create a framework for the development and application of future immunomodulatory therapies. We also delineate which subpopulations of patients with heart failure might benefit from immunomodulatory treatments. Furthermore, we summarize completed and ongoing clinical trials that assess immunomodulatory treatments in heart failure and present several therapeutic targets that could be investigated in the future. Lastly, we provide future directions to leverage the immunomodulatory potential of existing treatments and to foster the investigation of novel immunomodulatory therapeutics.
Collapse
Affiliation(s)
- George Markousis-Mavrogenis
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Lukas Baumhove
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ali A Al-Mubarak
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
- Department of Cardiology, Erasmus Medical Center, Rotterdam, Netherlands
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
12
|
Wang Q, Xiao F, Zeng Y, Zhu Q, Zhang H. PD-1/PD-L1 inhibitors-associated cardiac adverse events: a retrospective and real-world study based on the FDA Adverse Event Reporting System (FAERS). Expert Opin Drug Saf 2024; 23:257-267. [PMID: 37070426 DOI: 10.1080/14740338.2023.2203483] [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: 12/09/2022] [Accepted: 03/18/2023] [Indexed: 04/19/2023]
Abstract
BACKGROUND Programmed cell death protein-1 (PD-1) and programmed cell death ligand-1 (PD-L1) inhibitors have reformed the treatment landscape for various malignancies and improved prognosis of patients. However, they also lead to events that although rare may prove to be fatal. RESEARCH DESIGN AND METHODS Data from July 2014 to June 2022 based on FDA Adverse Event Reporting System (FAERS) were analyzed. The signal index reporting odds ratio (ROR) was used to evaluate the correlation between cardiac AEs and given medications. The indications and the median time to onset (TTO) of different PD-1/PD-L1 inhibitors were compared. RESULTS Cardiac AEs are rare but may be fatal with particular profiles in primary tumor, onset time, and especially gender. We identified 11,538 reports that were related to cardiotoxicity of PD-1/PD-L1 inhibitors, in which 178 different preferred terms (PTs) were distinguished, and nivolumab reported the most PTs with signal. All targeted medications showed signals in myocardial disorders and pericardial disorders, which tend to occur in the first 1-2 months. Non-small cell neoplasm was the top and common indication during anti-PD-1 or anti-PD-L1 therapy with cardiotoxicity. CONCLUSIONS This study could help early diagnosis and surveillance of ICIs-related cardiotoxicity.
Collapse
Affiliation(s)
- Qiaoyun Wang
- Department of Pharmacy, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Fengjiao Xiao
- Department of Pharmacy, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Yanbin Zeng
- Department of Pharmacy, China Pharmaceutical University Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Qiaoling Zhu
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, Jiangsu Province, China
| | - Haixia Zhang
- Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China
- Nanjing Medical Center for Clinical Pharmacy, Nanjing, Jiangsu Province, China
| |
Collapse
|
13
|
Quagliariello V, Passariello M, Bisceglia I, Paccone A, Inno A, Maurea C, Rapuano Lembo R, Manna L, Iovine M, Canale ML, Scherillo M, Ascierto PA, Gabrielli D, De Lorenzo C, Maurea N. Combinatorial immune checkpoint blockade increases myocardial expression of NLRP-3 and secretion of H-FABP, NT-Pro-BNP, interleukin-1β and interleukin-6: biochemical implications in cardio-immuno-oncology. Front Cardiovasc Med 2024; 11:1232269. [PMID: 38322766 PMCID: PMC10844473 DOI: 10.3389/fcvm.2024.1232269] [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: 05/31/2023] [Accepted: 01/10/2024] [Indexed: 02/08/2024] Open
Abstract
Background Immune checkpoint blockade in monotherapy or combinatorial regimens with chemotherapy or radiotherapy have become an integral part of oncology in recent years. Monoclonal antibodies against CTLA-4 or PD-1 or PDL-1 are the most studied ICIs in randomized clinical trials, however, more recently, an anti-LAG3 (Lymphocyte activation gene-3) antibody, Relatlimab, has been approved by FDA in combination with Nivolumab for metastatic melanoma therapy. Moreover, Atezolizumab is actually under study in association with Ipilimumab for therapy of metastatic lung cancer. Myocarditis, vasculitis and endothelitis are rarely observed in these patients on monotherapy, however new combination therapies could expose patients to more adverse cardiovascular events. Methods Human cardiomyocytes co-cultured with human peripheral blood lymphocytes (hPBMCs) were exposed to monotherapy and combinatorial ICIs (PD-L1 and CTLA-4 or PD-1 and LAG-3 blocking agents, at 100 nM) for 48 h. After treatments, cardiac cell lysis and secretion of biomarkers of cardiotoxicity (H-FABP, troponin-T, BNP, NT-Pro-BNP), NLRP3-inflammasome and Interleukin 1 and 6 were determined through colorimetric and enzymatic assays. Mitochondrial functions were studied in cardiomyocyte cell lysates through quantification of intracellular Ca++, ATP content and NADH:ubiquinone oxidoreductase core subunit S1 (Ndufs1) levels. Histone deacetylases type 4 (HDAC-4) protein levels were also determined in cardiomyocyte cell lysates to study potential epigenetic changes induced by immunotherapy regimens. Results Both combinations of immune checkpoint inhibitors exert more potent cardiotoxic side effects compared to monotherapies against human cardiac cells co-cultured with human lymphocytes. LDH release from cardiac cells was 43% higher in PD-L1/CTLA-4 blocking agents, and 35.7% higher in PD-1/LAG-3 blocking agents compared to monotherapies. HDAC4 and intracellular Ca++ levels were increased, instead ATP content and Ndufs1 were reduced in myocardial cell lysates (p < 0.001 vs. untreated cells). Troponin-T, BNP, NT-Pro-BNP and H-FABP, were also strongly increased in combination therapy compared to monotherapy regimen. NLRP3 expression, IL-6 and IL-1β levels were also increased by PDL-1/CTLA-4 and PD-1/LAG-3 combined blocking agents compared to untreated cells and monotherapies. Conclusions Data of the present study, although in vitro, indicate that combinatorial immune checkpoint blockade, induce a pro- inflammatory phenotype, thus indicating that these therapies should be closely monitored by the multidisciplinary team consisting of oncologists, cardiologists and immunologists.
Collapse
Affiliation(s)
- V. Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - M. Passariello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - I. Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
| | - A. Paccone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - A. Inno
- Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Sacro Cuore Don Calabria, Negrar, Italy
| | - C. Maurea
- Medical Oncology, Ospedale del Mare, Naples, Italy
| | - R. Rapuano Lembo
- Department of Molecular Medicine, Ceinge-Biotecnologie Avanzate s.c.a.r.l., Naples, Italy
| | - L. Manna
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - M. Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - M. L. Canale
- U.O.C. Cardiologia, Ospedale Versilia, Lido di Camaiore (LU), Camaiore, Italy
| | - M. Scherillo
- Cardiologia Interventistica e UTIC, A.O. San Pio, Presidio Ospedaliero Gaetano Rummo, Benevento, Italy
| | - P. A. Ascierto
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Naples, Italy
| | - D. Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, Roma – Fondazione per il Tuo Cuore – Heart Care Foundation, Firenze, Italy
| | - C. De Lorenzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
- Department of Molecular Medicine, Ceinge-Biotecnologie Avanzate s.c.a.r.l., Naples, Italy
| | - N. Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
14
|
Zhang H, Dhalla NS. The Role of Pro-Inflammatory Cytokines in the Pathogenesis of Cardiovascular Disease. Int J Mol Sci 2024; 25:1082. [PMID: 38256155 PMCID: PMC10817020 DOI: 10.3390/ijms25021082] [Citation(s) in RCA: 98] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
With cardiovascular disease (CVD) being a primary source of global morbidity and mortality, it is crucial that we understand the molecular pathophysiological mechanisms at play. Recently, numerous pro-inflammatory cytokines have been linked to several different CVDs, which are now often considered an adversely pro-inflammatory state. These cytokines most notably include interleukin-6 (IL-6),tumor necrosis factor (TNF)α, and the interleukin-1 (IL-1) family, amongst others. Not only does inflammation have intricate and complex interactions with pathophysiological processes such as oxidative stress and calcium mishandling, but it also plays a role in the balance between tissue repair and destruction. In this regard, pre-clinical and clinical evidence has clearly demonstrated the involvement and dynamic nature of pro-inflammatory cytokines in many heart conditions; however, the clinical utility of the findings so far remains unclear. Whether these cytokines can serve as markers or risk predictors of disease states or act as potential therapeutic targets, further extensive research is needed to fully understand the complex network of interactions that these molecules encompass in the context of heart disease. This review will highlight the significant advances in our understanding of the contributions of pro-inflammatory cytokines in CVDs, including ischemic heart disease (atherosclerosis, thrombosis, acute myocardial infarction, and ischemia-reperfusion injury), cardiac remodeling (hypertension, cardiac hypertrophy, cardiac fibrosis, cardiac apoptosis, and heart failure), different cardiomyopathies as well as ventricular arrhythmias and atrial fibrillation. In addition, this article is focused on discussing the shortcomings in both pathological and therapeutic aspects of pro-inflammatory cytokines in CVD that still need to be addressed by future studies.
Collapse
Affiliation(s)
- Hannah Zhang
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB R2H 2A6, Canada
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
15
|
Abbasi A, Shafie D, Heidari Moghaddam R, Sadeghi M, Safavi SM. Investigation of adherence to DASH diet components and reduction of heart failure risk in adults: A case-control study. ARYA ATHEROSCLEROSIS 2024; 20:38-46. [PMID: 39717422 PMCID: PMC11663282 DOI: 10.48305/arya.2024.42670.2964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/15/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND Heart failure (HF), or congestive Heart failure (CHF), is a heart disorder with a number of symptoms caused by the heart's inability to pump blood. Our aim in conducting this study is to investigate the adherence to dietary approaches to stop hypertension (DASH) diet components and the risk of HF in Iranian adult women and men. METHODS In this hospital-based, case-control study, we included 340 participants (194 men and 146 women) aged 30-70 years who were recently (less than 6 months) diagnosed with HF. In this study, there were 169 participants in the control group and 171 participants in the case group. A semi-quantitative food frequency questionnaire (FFQ) with 148 items was used to assess food intake. Multiple logistic regression statistical tests were used to evaluate the relationship between DASH score and HF. RESULTS After adjusting for confounding variables, the data showed that adherence to the DASH diet was associated with a reduced risk of HF. Our data show that a significant relationship was found between the consumption of fruits (OR: 0.62; 95% CI: 0.53-0.68), vegetables (OR: 0.53; 95% CI: 0.28-0.81), legumes and nuts (OR: 0.75; 95% CI: 0.65-0.68), and heart failure, but no significant relationship was found with the other components of the DASH diet and heart failure. CONCLUSION Findings suggest that there is an inverse relationship between adherence to the DASH-style diet and the likelihood of HF, and adherence to some components of the DASH diet was also effective in reducing the risk of HF. To obtain more complete results, it is necessary to conduct cohort studies and randomized clinical trials.
Collapse
Affiliation(s)
- Abbas Abbasi
- Department of Clinical Nutrition, School of Nutrition and Food Science, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Davood Shafie
- Heart Failure Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Heidari Moghaddam
- Cardiovascular Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Masoumeh Sadeghi
- Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyyed Morteza Safavi
- Department of Clinical Nutrition, School of Nutrition and Food Science, Nutrition and Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
16
|
Hall S, Samani S, Churillo A, Freeburg L, Cohen O, Devarakonda K, Khan S, Barringhaus KG, Shah N, Spinale FG. Obstructive sleep apnea alters microRNA levels: Effects of continuous positive airway pressure. MEDICAL RESEARCH ARCHIVES 2024; 12:4975. [PMID: 38770116 PMCID: PMC11105662 DOI: 10.18103/mra.v12i1.4975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Background Obstructive sleep apnea (OSA) has been linked to cytokine-mediated chronic inflammatory states. Continuous positive airway pressure (CPAP) is an established therapy for OSA, but its effects on inflammation remain unclear. A recent study from our group identified soluble cytokine receptors altered in OSA patients and modified by CPAP adherence. However, the upstream regulatory pathways responsible for these shifts in proinflammatory cascades with OSA and CPAP therapy remained unknown. Accordingly, this study mapped OSA and CPAP-modulated soluble cytokine receptors to specific microRNAs and then tested the hypothesis that OSA and CPAP adherence shift cytokine-related microRNA expression profiles. Study Design Plasma samples were collected from patients with OSA (n=50) at baseline and approximately 90 days after CPAP initiation and compared to referent control subjects (n=10). Patients with OSA were further divided into cohorts defined by adherence vs nonadherence to CPAP therapy. The microRNAs that mapped to soluble cytokine receptors of interest were subjected to quantitative polymerase chain reaction. Results At baseline, increased hsa-miR-15a-5p, hsa-miR-15b-5p, hsa-miR-16-5p, hsa-miR-195-5p, hsa-miR-424-5p, hsa-miR-223-3p, and hsa-miR-223-5p were observed in patients with OSA compared to controls (p<0.05). In CPAP adherent patients (n=22), hsa-miR233-3p and hsa-miR233-5p decreased at follow-up (p<0.05) whereas there was no change in miR levels from baseline in non-adherent CPAP patients (n=28). The miRs hsa-miR233-3p and hsa-miR233-5p mapped to both proinflammatory and innate immunity activation; the inflammasome. Conclusion A specific set of microRNAs, including hsa-miR233-3p and hsa-miR233-5p, may serve as a marker of inflammatory responses in patients with OSA, and be used to assess attenuation of inflammasome activation by CPAP.
Collapse
Affiliation(s)
- SarahRose Hall
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC
| | - Stephanie Samani
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC
| | - Amelia Churillo
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC
| | - Lisa Freeburg
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC
| | - Oren Cohen
- Division of Pulmonary, Sleep, and Critical Care Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Kavya Devarakonda
- Division of Pulmonary, Sleep, and Critical Care Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Samira Khan
- Division of Pulmonary, Sleep, and Critical Care Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Neomi Shah
- Division of Pulmonary, Sleep, and Critical Care Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Francis G. Spinale
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC
- Columbia VA Health Care System, Columbia, SC
| |
Collapse
|
17
|
Agarwal V, Kaushik AS, Chaudhary R, Rehman M, Srivastava S, Mishra V. Transcutaneous vagus nerve stimulation ameliorates cardiac abnormalities in chronically stressed rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:281-303. [PMID: 37421431 DOI: 10.1007/s00210-023-02611-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 07/01/2023] [Indexed: 07/10/2023]
Abstract
Chronically stressed patients often have low vagal tone and increased levels of proinflammatory cytokines, which increase their risk for developing cardiac dysfunction. Transcutaneous vagus nerve stimulation (taVNS) is a way to activate the parasympathetic system, which has the ability to reduce inflammation and antagonize excessive sympathetic responses. However, the effectiveness of taVNS in treating cardiac dysfunction caused by chronic unpredictable stress (CUS) has not been studied. To investigate this, we first validated a rat model of CUS, in which the rats were exposed to random stressors daily for 8 weeks. Post CUS, the rats were treated with taVNS (1.0 ms, 6 V, 6 Hz, for 40 min × 2 weeks, alternatively) and their cardiac function and cholinergic flow were evaluated. Furthermore, serum cardiac troponin I (cTnI), cardiac caspase-3, inducible nitric oxide synthase (iNOS), and transforming growth factor (TGF)-β1 expression in rats were also assessed. The chronically stressed rats showed depressed behavior with increased levels of serum corticosterone and proinflammatory cytokines. Electrocardiogram (ECG) and heart rate variability (HRV) studies revealed elevated heart rate, diminished vagal tone, and altered sinus rhythm in CUS rats. Furthermore, the CUS rats demonstrated cardiac hypertrophy and fibrosis with increased caspase-3, iNOS, and TGF-β expression in their myocardium and increased levels of serum cTnI. Interestingly, alternate taVNS therapy for 2 weeks, post CUS, helped alleviate these cardiac abnormalities. These suggest that taVNS could be a useful adjunctive and non-pharmacological approach for managing CUS induced cardiac dysfunction.
Collapse
Affiliation(s)
- Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, U.P., 226025, India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, U.P., 226025, India
| | - Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, U.P., 226025, India
| | - Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, U.P., 226025, India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, U.P., 226025, India
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, (A Central University), Vidya Vihar, Raebareli Road, Lucknow, U.P., 226025, India.
| |
Collapse
|
18
|
Arvunescu AM, Ionescu RF, Cretoiu SM, Dumitrescu SI, Zaharia O, Nanea IT. Inflammation in Heart Failure-Future Perspectives. J Clin Med 2023; 12:7738. [PMID: 38137807 PMCID: PMC10743797 DOI: 10.3390/jcm12247738] [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: 11/05/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
Chronic heart failure is a terminal point of a vast majority of cardiac or extracardiac causes affecting around 1-2% of the global population and more than 10% of the people above the age of 65. Inflammation is persistently associated with chronic diseases, contributing in many cases to the progression of disease. Even in a low inflammatory state, past studies raised the question of whether inflammation is a constant condition, or if it is, rather, triggered in different amounts, according to the phenotype of heart failure. By evaluating the results of clinical studies which focused on proinflammatory cytokines, this review aims to identify the ones that are independent risk factors for heart failure decompensation or cardiovascular death. This review assessed the current evidence concerning the inflammatory activation cascade, but also future possible targets for inflammatory response modulation, which can further impact the course of heart failure.
Collapse
Affiliation(s)
- Alexandru Mircea Arvunescu
- Department of Internal Medicine and Cardiology, “Prof. Dr. Th. Burghele” Clinical Hospital, 061344 Bucharest, Romania; (O.Z.); (I.T.N.)
- Department of Cardio-Thoracic Pathology, Cardio-Thoracic Pathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050471 Bucharest, Romania
| | - Ruxandra Florentina Ionescu
- Department of Cardiology I, Central Military Emergency Hospital “Dr Carol Davila”, 030167 Bucharest, Romania (S.I.D.)
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Sanda Maria Cretoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Silviu Ionel Dumitrescu
- Department of Cardiology I, Central Military Emergency Hospital “Dr Carol Davila”, 030167 Bucharest, Romania (S.I.D.)
- Department of Cardiology, Faculty of Medicine, Titu Maiorescu University, 040441 Bucharest, Romania
| | - Ondin Zaharia
- Department of Internal Medicine and Cardiology, “Prof. Dr. Th. Burghele” Clinical Hospital, 061344 Bucharest, Romania; (O.Z.); (I.T.N.)
- Department of Cardio-Thoracic Pathology, Cardio-Thoracic Pathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050471 Bucharest, Romania
| | - Ioan Tiberiu Nanea
- Department of Internal Medicine and Cardiology, “Prof. Dr. Th. Burghele” Clinical Hospital, 061344 Bucharest, Romania; (O.Z.); (I.T.N.)
- Department of Cardio-Thoracic Pathology, Cardio-Thoracic Pathology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050471 Bucharest, Romania
| |
Collapse
|
19
|
Reis-Mendes A, Ferreira M, Duarte JA, Duarte-Araújo M, Remião F, Carvalho F, Sousa E, Bastos ML, Costa VM. The role of inflammation and antioxidant defenses in the cardiotoxicity of doxorubicin in elderly CD-1 male mice. Arch Toxicol 2023; 97:3163-3177. [PMID: 37676301 PMCID: PMC10567829 DOI: 10.1007/s00204-023-03586-1] [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: 06/26/2023] [Accepted: 08/10/2023] [Indexed: 09/08/2023]
Abstract
Doxorubicin (DOX) is a potent chemotherapeutic agent used against several cancer types. However, due to its cardiotoxic adverse effects, the use of this drug may be also life-threatening. Although most cancer patients are elderly, they are poorly represented and evaluated in pre-clinical and clinical studies. Considering this, the present work aims to evaluate inflammation and oxidative stress as the main mechanisms of DOX-induced cardiotoxicity, in an innovative approach using an experimental model constituted of elderly animals treated with a clinically relevant human cumulative dose of DOX. Elderly (18-20 months) CD-1 male mice received biweekly DOX administrations, for 3 weeks, to reach a cumulative dose of 9.0 mg/kg. One week (1W) or two months (2 M) after the last DOX administration, the heart was collected to determine both drug's short and longer cardiac adverse effects. The obtained results showed that DOX causes cardiac histological damage and fibrosis at both time points. In the 1W-DOX group, the number of nuclear factor kappa B (NF-κB) p65 immunopositive cells increased and a trend toward increased NF-κB p65 expression was seen. An increase of inducible nitric oxide synthase (iNOS) and interleukin (IL)-33 and a trend toward increased IL-6 and B-cell lymphoma-2-associated X (Bax) expression were seen after DOX. In the same group, a decrease in IL-1β, p62, and microtubule-associated protein 1A/1B-light chain 3 (LC3)-I, p38 mitogen-activated protein kinase (MAPK) expression was observed. Contrariwise, the animals sacrificed 2 M after DOX showed a significant increase in glutathione peroxidase 1 and Bax expression with persistent cardiac damage and fibrosis, while carbonylated proteins, erythroid-2-related factor 2 (Nrf2), NF-κB p65, myeloperoxidase, LC3-I, and LC3-II expression decreased. In conclusion, our study demonstrated that in an elderly mouse population, DOX induces cardiac inflammation, autophagy, and apoptosis in the heart in the short term. When kept for a longer period, oxidative-stress-linked pathways remained altered, as well as autophagy markers and tissue damage after DOX treatment, emphasizing the need for continuous post-treatment cardiac monitoring.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Mariana Ferreira
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - José Alberto Duarte
- Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, Research Center in Physical Activity, Health and Leisure (CIAFEL), University of Porto, 4200-450, Porto, Portugal
- 1H-TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116, Gandra, Portugal
| | - Margarida Duarte-Araújo
- LAQV/REQUIMTE, University of Porto, 4050-313, Porto, Portugal
- Department of Immuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313, Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Chemistry Department, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, 4450-208, Porto, Portugal
| | - Maria Lourdes Bastos
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
20
|
Ito S, Yamatani F, Arai Y, Manabe E, Tsujino T. Dimethyl Fumarate Ameliorated Cardiorenal Anemia Syndrome and Improved Overall Survival in Dahl/Salt-Sensitive Rats. J Pharmacol Exp Ther 2023; 387:299-305. [PMID: 37857438 DOI: 10.1124/jpet.123.001692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 09/01/2023] [Accepted: 09/11/2023] [Indexed: 10/21/2023] Open
Abstract
Cardiovascular disease, chronic kidney disease, and anemia are known to adversely affect each other. Inflammation is commonly involved in these diseases. Cardiorenal anemia syndrome (CRAS) is the name given to this mutually harmful condition. Dimethyl fumarate (DMF) is a Food and Drug Administration-approved antioxidant and anti-inflammatory agent. The purpose of this study was to investigate the effects of DMF on Dahl/salt-sensitive (DS) rats as a CRAS model. Six-week-old DS rats were divided into three groups: the control group, the high-salt (HS) group, and the HS+DMF group. The HS and HS+DMF groups were fed a high-salt diet (8% NaCl) from 6 weeks of age. In the HS+DMF group, DMF (90 mg/kg per day) was orally administered from 6 to 15 weeks of age. Systolic blood pressure was measured every 2 weeks. The heart and renal injuries were assessed with histopathological analysis. The heart and renal expression of mRNAs was assessed by reverse-transcription polymerase chain reaction. DMF significantly improved overall survival, which was shortened by HS in DS rats. Systolic blood pressure increased in the HS group compared with the control group, and DMF tended to suppress this change. DMF ameliorated the cardiac and renal abnormalities confirmed in the HS group by histopathological analysis. Furthermore, the changes in mRNA expressions associated with disease exacerbation in the HS group were suppressed by DMF. DMF also improved anemia. This study suggests that DMF improves overall survival in DS rats through organ-protective effects and is effective against cardiorenal anemia syndrome. SIGNIFICANCE STATEMENT: Dimethyl fumarate was found to improve overall survival in Dahl/salt-sensitive rats, associated with its ability to ameliorate anemia and induce cardioprotective and renoprotective effects through anti-inflammatory and antifibrotic effects.
Collapse
Affiliation(s)
- Satoyasu Ito
- The Second Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy (S.I., F.Y., Y.A., E.M., T.T.) and Department of Cardiovascular and Renal Medicine, School of Medicine (E.M., T.T.), Hyogo Medical University, Hyogo, Japan
| | - Fuyuka Yamatani
- The Second Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy (S.I., F.Y., Y.A., E.M., T.T.) and Department of Cardiovascular and Renal Medicine, School of Medicine (E.M., T.T.), Hyogo Medical University, Hyogo, Japan
| | - Yuri Arai
- The Second Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy (S.I., F.Y., Y.A., E.M., T.T.) and Department of Cardiovascular and Renal Medicine, School of Medicine (E.M., T.T.), Hyogo Medical University, Hyogo, Japan
| | - Eri Manabe
- The Second Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy (S.I., F.Y., Y.A., E.M., T.T.) and Department of Cardiovascular and Renal Medicine, School of Medicine (E.M., T.T.), Hyogo Medical University, Hyogo, Japan
| | - Takeshi Tsujino
- The Second Division of Pharmacotherapy, Department of Pharmacy, School of Pharmacy (S.I., F.Y., Y.A., E.M., T.T.) and Department of Cardiovascular and Renal Medicine, School of Medicine (E.M., T.T.), Hyogo Medical University, Hyogo, Japan
| |
Collapse
|
21
|
Madè A, Bibi A, Garcia-Manteiga JM, Tascini AS, Piella SN, Tikhomirov R, Voellenkle C, Gaetano C, Leszek P, Castelvecchio S, Menicanti L, Martelli F, Greco S. circRNA-miRNA-mRNA Deregulated Network in Ischemic Heart Failure Patients. Cells 2023; 12:2578. [PMID: 37947656 PMCID: PMC10648415 DOI: 10.3390/cells12212578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
Noncoding RNAs (ncRNAs), which include circular RNAs (circRNAs) and microRNAs (miRNAs), regulate the development of cardiovascular diseases (CVD). Notably, circRNAs can interact with miRNAs, influencing their specific mRNA targets' levels and shaping a competing endogenous RNAs (ceRNA) network. However, these interactions and their respective functions remain largely unexplored in ischemic heart failure (IHF). This study is aimed at identifying circRNA-centered ceRNA networks in non-end-stage IHF. Approximately 662 circRNA-miRNA-mRNA interactions were identified in the heart by combining state-of-the-art bioinformatics tools with experimental data. Importantly, KEGG terms of the enriched mRNA indicated CVD-related signaling pathways. A specific network centered on circBPTF was validated experimentally. The levels of let-7a-5p, miR-18a-3p, miR-146b-5p, and miR-196b-5p were enriched in circBPTF pull-down experiments, and circBPTF silencing inhibited the expression of HDAC9 and LRRC17, which are targets of miR-196b-5p. Furthermore, as suggested by the enriched pathway terms of the circBPTF ceRNA network, circBPTF inhibition elicited endothelial cell cycle arrest. circBPTF expression increased in endothelial cells exposed to hypoxia, and its upregulation was confirmed in cardiac samples of 36 end-stage IHF patients compared to healthy controls. In conclusion, circRNAs act as miRNA sponges, regulating the functions of multiple mRNA targets, thus providing a novel vision of HF pathogenesis and laying the theoretical foundation for further experimental studies.
Collapse
Affiliation(s)
- Alisia Madè
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Alessia Bibi
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
- Department of Biosciences, University of Milan, 20122 Milan, Italy
| | - Jose Manuel Garcia-Manteiga
- Center for Omics Sciences COSR, BioInformatics Laboratory, San Raffaele Scientific Institute, 20132 Milan, Italy; (J.M.G.-M.); (A.S.T.)
| | - Anna Sofia Tascini
- Center for Omics Sciences COSR, BioInformatics Laboratory, San Raffaele Scientific Institute, 20132 Milan, Italy; (J.M.G.-M.); (A.S.T.)
- Università Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Santiago Nicolas Piella
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Roman Tikhomirov
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Christine Voellenkle
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Przemyslaw Leszek
- Department of Heart Failure and Transplantology, National Institute of Cardiology, 04-628 Warsaw, Poland;
| | - Serenella Castelvecchio
- Department of Adult Cardiac Surgery, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (S.C.); (L.M.)
| | - Lorenzo Menicanti
- Department of Adult Cardiac Surgery, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (S.C.); (L.M.)
| | - Fabio Martelli
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| | - Simona Greco
- Molecular Cardiology Laboratory, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy; (A.M.); (A.B.); (S.N.P.); (R.T.); (C.V.); (S.G.)
| |
Collapse
|
22
|
Singh P, Shah DA, Jouni M, Cejas RB, Crossman DK, Magdy T, Qiu S, Wang X, Zhou L, Sharafeldin N, Hageman L, McKenna DE, Armenian SH, Balis FM, Hawkins DS, Keller FG, Hudson MM, Neglia JP, Ritchey AK, Ginsberg JP, Landier W, Bhatia R, Burridge PW, Bhatia S. Altered Peripheral Blood Gene Expression in Childhood Cancer Survivors With Anthracycline-Induced Cardiomyopathy - A COG-ALTE03N1 Report. J Am Heart Assoc 2023; 12:e029954. [PMID: 37750583 PMCID: PMC10727235 DOI: 10.1161/jaha.123.029954] [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: 02/22/2023] [Accepted: 08/08/2023] [Indexed: 09/27/2023]
Abstract
Background Anthracycline-induced cardiomyopathy is a leading cause of premature death in childhood cancer survivors, presenting a need to understand the underlying pathogenesis. We sought to examine differential blood-based mRNA expression profiles in anthracycline-exposed childhood cancer survivors with and without cardiomyopathy. Methods and Results We designed a matched case-control study (Children's Oncology Group-ALTE03N1) with mRNA sequencing on total RNA from peripheral blood in 40 anthracycline-exposed survivors with cardiomyopathy (cases) and 64 matched survivors without (controls). DESeq2 identified differentially expressed genes. Ingenuity Pathway Analyses (IPA) and Gene Set Enrichment Analyses determined the potential roles of altered genes in biological pathways. Functional validation was performed by gene knockout in human-induced pluripotent stem cell-derived cardiomyocytes using CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) technology. Median age at primary cancer diagnosis for cases and controls was 8.2 and 9.7 years, respectively. Thirty-six differentially expressed genes with fold change ≥±2 were identified; 35 were upregulated. IPA identified "hepatic fibrosis" and "iron homeostasis" pathways to be significantly modulated by differentially expressed genes, including toxicology functions of myocardial infarction, cardiac damage, and cardiac dilation. Leading edge analysis from Gene Set Enrichment Analyses identified lactate dehydrogenase A (LDHA) and cluster of differentiation 36 (CD36) genes to be significantly upregulated in cases. Interleukin 1 receptor type 1, 2 (IL1R1, IL1R2), and matrix metalloproteinase 8, 9 (MMP8, MMP9) appeared in multiple canonical pathways. LDHA-knockout human-induced pluripotent stem cell-derived cardiomyocytes showed increased sensitivity to doxorubicin. Conclusions We identified differential mRNA expression profiles in peripheral blood of anthracycline-exposed childhood cancer survivors with and without cardiomyopathy. Upregulation of LDHA and CD36 genes suggests metabolic perturbations in a failing heart. Dysregulation of proinflammatory cytokine receptors IL1R1 and IL1R2 and matrix metalloproteinases, MMP8 and MMP9 indicates structural remodeling that accompanies the clinical manifestation of symptomatic cardiotoxicity.
Collapse
Affiliation(s)
- Purnima Singh
- Institute for Cancer Outcomes and SurvivorshipUniversity of Alabama at BirminghamBirminghamAL
- Department of PediatricsUniversity of Alabama at BirminghamBirminghamAL
| | | | - Mariam Jouni
- Department of PharmacologyNorthwestern UniversityChicagoIL
| | | | - David K. Crossman
- Department of GeneticsUniversity of Alabama at BirminghamBirminghamAL
| | - Tarek Magdy
- Department of PharmacologyNorthwestern UniversityChicagoIL
- Louisiana State University Health ShreveportShreveportLA
| | - Shaowei Qiu
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinChina
- Division of Hematology and OncologyUniversity of Alabama at BirminghamBirminghamAL
| | - Xuexia Wang
- Department of BiostatisticsFlorida International UniversityMiamiFL
| | - Liting Zhou
- Institute for Cancer Outcomes and SurvivorshipUniversity of Alabama at BirminghamBirminghamAL
| | - Noha Sharafeldin
- Institute for Cancer Outcomes and SurvivorshipUniversity of Alabama at BirminghamBirminghamAL
| | - Lindsey Hageman
- Institute for Cancer Outcomes and SurvivorshipUniversity of Alabama at BirminghamBirminghamAL
| | | | | | - Frank M. Balis
- Department of PediatricsChildren’s Hospital of PhiladelphiaPhiladelphiaPA
| | | | - Frank G. Keller
- Department of Pediatrics, Children’s Healthcare of AtlantaEmory UniversityAtlantaGA
| | - Melissa M. Hudson
- Department of Epidemiology and Cancer ControlSt. Jude Children’s Research HospitalMemphisTN
| | | | - A Kim Ritchey
- Department of PediatricsUPMC Children’s Hospital of PittsburghPAPittsburgh
| | - Jill P. Ginsberg
- Department of PediatricsChildren’s Hospital of PhiladelphiaPhiladelphiaPA
| | - Wendy Landier
- Institute for Cancer Outcomes and SurvivorshipUniversity of Alabama at BirminghamBirminghamAL
- Department of PediatricsUniversity of Alabama at BirminghamBirminghamAL
| | - Ravi Bhatia
- Division of Hematology and OncologyUniversity of Alabama at BirminghamBirminghamAL
| | | | - Smita Bhatia
- Institute for Cancer Outcomes and SurvivorshipUniversity of Alabama at BirminghamBirminghamAL
- Department of PediatricsUniversity of Alabama at BirminghamBirminghamAL
| |
Collapse
|
23
|
Blackwell K, Blackwell M, Blackwell T. Testosterone Replacement Therapy and Cardiovascular Disease: Balancing Safety and Risks in Hypogonadal Men. Curr Cardiol Rep 2023; 25:1157-1163. [PMID: 37733143 DOI: 10.1007/s11886-023-01935-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/03/2023] [Indexed: 09/22/2023]
Abstract
PURPOSE OF REVIEW: The purpose of this review is to analyze the link between testosterone replacement therapy (TRT) and adverse cardiovascular (CV) events. RECENT FINDINGS: A few published studies suggest a link between TRT and CV events. These studies contained flaws, and many other studies reveal a reduction in CV events. Hypogonadism is associated with increased mortality in men with CVD. TRT in hypogonadal men can improve many CVD risk factors, reduce QT interval prolongation, lead to better outcomes in heart failure patients, and slow the progression of atherosclerosis. The use of TRT to achieve physiologic testosterone concentrations in men does not pose a threat to CV health and has demonstrated a cardioprotective effect.
Collapse
Affiliation(s)
- Kelli Blackwell
- UTMB 301 University Blvd, Rebecca Sealy Bldg 5.138, Galveston, TX, 77554, USA
| | - Michele Blackwell
- UTMB 301 University Blvd, Rebecca Sealy Bldg 5.138, Galveston, TX, 77554, USA
| | - Thomas Blackwell
- UTMB 301 University Blvd, Rebecca Sealy Bldg 5.138, Galveston, TX, 77554, USA.
| |
Collapse
|
24
|
Suvakov S, Kattah AG, Gojkovic T, Enninga EAL, Pruett J, Jayachandran M, Sousa C, Santos J, Abou Hassan C, Gonzales-Suarez M, Garovic VD. Impact of Aging and Cellular Senescence in the Pathophysiology of Preeclampsia. Compr Physiol 2023; 13:5077-5114. [PMID: 37770190 DOI: 10.1002/cphy.c230003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The incidence of hypertensive disorders of pregnancy is increasing, which may be due to several factors, including an increased age at pregnancy and more comorbid health conditions during reproductive years. Preeclampsia, the most severe hypertensive disorder of pregnancy, has been associated with an increased risk of future disease, including cardiovascular and kidney diseases. Cellular senescence, the process of cell cycle arrest in response to many physiologic and maladaptive stimuli, may play an important role in the pathogenesis of preeclampsia and provide a mechanistic link to future disease. In this article, we will discuss the pathophysiology of preeclampsia, the many mechanisms of cellular senescence, evidence for the involvement of senescence in the development of preeclampsia, as well as evidence that cellular senescence may link preeclampsia to the risk of future disease. Lastly, we will explore how a better understanding of the role of cellular senescence in preeclampsia may lead to therapeutic trials. © 2023 American Physiological Society. Compr Physiol 13:5077-5114, 2023.
Collapse
Affiliation(s)
- Sonja Suvakov
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Andrea G Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Tamara Gojkovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth A L Enninga
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Jacob Pruett
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Ciria Sousa
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Janelle Santos
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Coline Abou Hassan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Vesna D Garovic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
- Division of Research, Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| |
Collapse
|
25
|
Abubakar M, Rasool HF, Javed I, Raza S, Abang L, Hashim MMA, Saleem Z, Abdullah RM, Faraz MA, Hassan KM, Bhat RR. Comparative Roles of IL-1, IL-6, IL-10, IL-17, IL-18, 1L-22, IL-33, and IL-37 in Various Cardiovascular Diseases With Potential Insights for Targeted Immunotherapy. Cureus 2023; 15:e42494. [PMID: 37637634 PMCID: PMC10455045 DOI: 10.7759/cureus.42494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2023] [Indexed: 08/29/2023] Open
Abstract
In recent years, the study of interleukins (ILs), crucial cytokines involved in immune response and inflammation, has garnered significant attention within the sphere of cardiovascular diseases (CVDs). The research has provided insights into the involvement of ILs in diverse CVDs, including arrhythmias, myocardial infarction, atherosclerosis, and heart failure (HF). ILs have emerged as promising therapeutic targets for drug interventions through their involvement in disease development and progression. This comprehensive review provides a detailed overview of ILs, elucidating their functions within the immune system and offering insights into their specific contributions to various CVDs. Moreover, the article delves into the examination of current and potential drug therapies that selectively target ILs in the management of CVDs, presenting a comprehensive analysis of the advantages and disadvantages associated with these therapeutic approaches. A comprehensive literature review was conducted to investigate the involvement of ILs in CVDs. The relevant articles were searched on PubMed, PubMed Central, Medline, Cochrane, Google Scholar, and ScienceDirect databases. The search encompassed articles published from these databases' inception until July 12, 2023. We first examine generalized aspects of ILs, particularly CVDs. Then, we shift focus towards examining the direct impact of ILs on cardiac cells and tissue; on the immune system and inflammation; endothelial cells and vascular function; and finally, their interactions with other signaling pathways and molecules. Then, we discuss the molecular mechanisms of various ILs. Sequentially, we delve into a comprehensive analysis of the individualized role of each distinct IL in diverse CVDs, examining their specific contributions. Finally, we explore the potential for targeted drug therapy to modulate IL activity, aiming to enhance outcomes for patients burdened with CVD. The objective is the identification of gaps in current knowledge and highlight areas that require further investigation within the context of cardiovascular medicine. Through deepening our comprehension of the intricate involvement of ILs in CVDs and harnessing their potential for targeted drug therapy, novel treatment strategies can be devised, leading to improved patient outcomes in cardiovascular medicine.
Collapse
Affiliation(s)
- Muhammad Abubakar
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
- Department of Internal Medicine, Siddique Sadiq Memorial Trust Hospital, Gujranwala, PAK
| | - Hafiz Fahad Rasool
- Department of Public Health, Nanjing Medical University School of Public Health, Nanjing, CHN
| | - Izzah Javed
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
| | - Saud Raza
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
| | - Lucy Abang
- Department of Biochemistry, All Saints University School of Medicine, Roseau, DMA
| | | | - Zartasha Saleem
- Department of Emergency Medicine, The University of Lahore Teaching Hospital, Lahore, PAK
| | | | - Muhammad Ahmad Faraz
- Department of Forensic Medicine, Post Graduate Medical Institute, Lahore General Hospital, Lahore, PAK
| | - Khawaja Mushammar Hassan
- Department of Internal Medicine, Ameer-Ud-Din Medical College, Lahore General Hospital, Lahore, PAK
| | - Rakshita Ramesh Bhat
- Department of Medical Oncology, Mangalore Institute of Oncology, Mangalore, IND
- Department of Internal Medicine, Bangalore Medical College and Research Institute, Bangalore, IND
| |
Collapse
|
26
|
Shi YJ, Dong GJ, Guo M. Targeting epicardial adipose tissue: A potential therapeutic strategy for heart failure with preserved ejection fraction with type 2 diabetes mellitus. World J Diabetes 2023; 14:724-740. [PMID: 37383601 PMCID: PMC10294070 DOI: 10.4239/wjd.v14.i6.724] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/10/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a heterogeneous syndrome with various comorbidities, multiple cardiac and extracardiac pathophysiologic abnormalities, and diverse phenotypic presentations. Since HFpEF is a heterogeneous disease with different phenotypes, individualized treatment is required. HFpEF with type 2 diabetes mellitus (T2DM) represents a specific phenotype of HFpEF, with about 45%-50% of HFpEF patients suffering from T2DM. Systemic inflammation associated with dysregulated glucose metabolism is a critical pathological mechanism of HFpEF with T2DM, which is intimately related to the expansion and dysfunction (inflammation and hypermetabolic activity) of epicardial adipose tissue (EAT). EAT is well established as a very active endocrine organ that can regulate the pathophysiological processes of HFpEF with T2DM through the paracrine and endocrine mechanisms. Therefore, suppressing abnormal EAT expansion may be a promising therapeutic strategy for HFpEF with T2DM. Although there is no treatment specifically for EAT, lifestyle management, bariatric surgery, and some pharmaceutical interventions (anti-cytokine drugs, statins, proprotein convertase subtilisin/kexin type 9 inhibitors, metformin, glucagon-like peptide-1 receptor agonists, and especially sodium-glucose cotransporter-2 inhibitors) have been shown to attenuate the inflammatory response or expansion of EAT. Importantly, these treatments may be beneficial in improving the clinical symptoms or prognosis of patients with HFpEF. Accordingly, well-designed randomized controlled trials are needed to validate the efficacy of current therapies. In addition, more novel and effective therapies targeting EAT are needed in the future.
Collapse
Affiliation(s)
- Yu-Jiao Shi
- Department of Cardiovascular Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Guo-Ju Dong
- Department of Cardiovascular Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing 100091, China
| | - Ming Guo
- Department of Cardiovascular Medicine, Xiyuan Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing 100091, China
| |
Collapse
|
27
|
Matusovits D, Murlasits Z, Kupai K, Baráth Z, Kang HL, Osváth P, Szűcs M, Priksz D, Juhász B, Radák Z, Várkonyi T, Pavo I, Pósa A. Paclitaxel Protects against Isoproterenol-Induced Damage in Rat Myocardium: Its Heme-Oxygenase Mediated Role in Cardiovascular Research. Antioxidants (Basel) 2023; 12:antiox12051129. [PMID: 37237995 DOI: 10.3390/antiox12051129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/04/2023] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
(1) Background: In cardiovascular applications, paclitaxel inhibits smooth muscle cell proliferation and migration and significantly reduces the occurrence of restenosis and target lesion revascularization. However, the cellular effects of paclitaxel in the myocardium are not well understood; (2) Methods: Wistar rats were divided into four groups: control (CTRL), isoproterenol (ISO) treated (1 mg/kg) and two groups treated with paclitaxel (PAC), which was administrated (10 mg/kg/day) for 5 days by gavage/per os alone or in combination (ISO + PAC) 3 weeks after ISO treatment. Ventricular tissue was harvested 24 h later for measurements of heme oxygenase (HO-1), reduced glutathione (GSH), oxidized glutathione (GSSG), superoxide dismutase (SOD), NF-κB, TNF-α and myeloperoxidase (MPO); (3) Results: HO-1 protein concentration, HO-1 activity, SOD protein concentration and total glutathione significantly decreased in response to ISO treatment. When PAC was administered in conjunction with ISO, HO-1, SOD concentration and total glutathione were not different from control levels. MPO activity, NF-κB concentration and TNF-α protein concentration were significantly increased in the ISO-only group, while the levels of these molecules were restored when PAC was co-administered; (4) Conclusions: Oral administration of PAC can maintain the expression of important antioxidants, anti-inflammatory molecules, HO-1, SOD and GSH, and suppress the production of TNF-α, MPO and NF-κB, which are involved in myocardial damage. The principal component of this cellular defense seems to be the expression of HO-1.
Collapse
Affiliation(s)
- Danica Matusovits
- Department of Prosthodontics, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary
| | - Zsolt Murlasits
- Institute of Sport Science and Physical Education University of Pécs, 7601 Pécs, Hungary
| | - Krisztina Kupai
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6703 Szeged, Hungary
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary
| | - Zoltán Baráth
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary
| | - Hsu Lin Kang
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary
| | - Péter Osváth
- Department of Urology, University of Debrecen, 4006 Debrecen, Hungary
| | - Miklós Szűcs
- Department of Urology, University of Debrecen, 4006 Debrecen, Hungary
| | - Dániel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, 4006 Debrecen, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Debrecen, 4006 Debrecen, Hungary
| | - Zsolt Radák
- Institute for Sports and Health Sciences, Hungarian University of Sports Science, 1051 Budapest, Hungary
| | - Tamás Várkonyi
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6703 Szeged, Hungary
| | - Imre Pavo
- Department of Internal Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6703 Szeged, Hungary
| | - Anikó Pósa
- Department of Oral Biology and Experimental Dental Research, Faculty of Dentistry, University of Szeged, 6703 Szeged, Hungary
| |
Collapse
|
28
|
Agarwal M, Sharma A, Kagoo R A, Bhargava A. Interactions between genes altered during cardiotoxicity and neurotoxicity in zebrafish revealed using induced network modules analysis. Sci Rep 2023; 13:6257. [PMID: 37069190 PMCID: PMC10110561 DOI: 10.1038/s41598-023-33145-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 04/07/2023] [Indexed: 04/19/2023] Open
Abstract
As the manufacturing and development of new synthetic compounds increase to keep pace with the expanding global demand, adverse health effects due to these compounds are emerging as critical public health concerns. Zebrafish have become a prominent model organism to study toxicology due to their genomic similarity to humans, optical clarity, well-defined developmental stages, short generation time, and cost-effective maintenance. It also provides a shorter time frame for in vivo toxicology evaluation compared to the mammalian experimental systems. Here, we used meta-analysis to examine the alteration in genes during cardiotoxicity and neurotoxicity in zebrafish, caused by chemical exposure of any kind. First, we searched the literature comprehensively for genes that are altered during neurotoxicity and cardiotoxicity followed by meta-analysis using ConsensusPathDB. Since constant communication between the heart and the brain is an important physiological phenomenon, we also analyzed interactions among genes altered simultaneously during cardiotoxicity and neurotoxicity using induced network modules analysis in ConsensusPathDB. We observed inflammation and regeneration as the major pathways involved in cardiotoxicity and neurotoxicity. A large number of intermediate genes and input genes anchored in these pathways are molecular regulators of cell cycle progression and cell death and are implicated in tumor manifestation. We propose potential predictive biomarkers for neurotoxicity and cardiotoxicity and the major pathways potentially implicated in the manifestation of a particular toxicity phenotype.
Collapse
Affiliation(s)
- Manusmriti Agarwal
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Telangana, 502284, India
| | - Ankush Sharma
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Telangana, 502284, India
| | - Andrea Kagoo R
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Telangana, 502284, India
| | - Anamika Bhargava
- Department of Biotechnology, Indian Institute of Technology Hyderabad (IITH), Kandi, Telangana, 502284, India.
| |
Collapse
|
29
|
Abstract
With a global burden of 844 million, chronic kidney disease (CKD) is now considered a public health priority. Cardiovascular risk is pervasive in this population, and low-grade systemic inflammation is an established driver of adverse cardiovascular outcomes in these patients. Accelerated cellular senescence, gut microbiota-dependent immune activation, posttranslational lipoprotein modifications, neuroimmune interactions, osmotic and nonosmotic sodium accumulation, acute kidney injury, and precipitation of crystals in the kidney and the vascular system all concur in determining the unique severity of inflammation in CKD. Cohort studies documented a strong link between various biomarkers of inflammation and the risk of progression to kidney failure and cardiovascular events in patients with CKD. Interventions targeting diverse steps of the innate immune response may reduce the risk of cardiovascular and kidney disease. Among these, inhibition of IL-1β (interleukin-1 beta) signaling by canakinumab reduced the risk for cardiovascular events in patients with coronary heart disease, and this protection was equally strong in patients with and without CKD. Several old (colchicine) and new drugs targeting the innate immune system, like the IL-6 (interleukin 6) antagonist ziltivekimab, are being tested in large randomized clinical trials to thoroughly test the hypothesis that mitigating inflammation may translate into better cardiovascular and kidney outcomes in patients with CKD.
Collapse
Affiliation(s)
- Carmine Zoccali
- Renal Research Institute New York and Institute of Molecular Biology and genetics (BIOGEM), Ariano Irpino, Italy and Associazione Ipertensione, Nefrologia, Trapianto (IPNET), Reggio Calabria Italy (C.Z.)
| | - Francesca Mallamaci
- Division of Nephrology and Transplantation, Grande Ospedale Metropolitano, Reggio Calabria, Italy and National Research Council (CNR), Clinical Epidemiology of Hypertension and Renal Diseases Unit of the Institute of Clinical Physiology, Reggio Calabria, Italy (F.M.)
| |
Collapse
|
30
|
Wilson C, Zi M, Smith M, Hussain M, D’Souza A, Dobrzynski H, Boyett MR. Atrioventricular node dysfunction in pressure overload-induced heart failure—Involvement of the immune system and transcriptomic remodelling. Front Pharmacol 2023; 14:1083910. [PMID: 37081960 PMCID: PMC10110994 DOI: 10.3389/fphar.2023.1083910] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 03/13/2023] [Indexed: 04/07/2023] Open
Abstract
Heart failure is associated with atrioventricular (AV) node dysfunction, and AV node dysfunction in the setting of heart failure is associated with an increased risk of mortality and heart failure hospitalisation. This study aims to understand the causes of AV node dysfunction in heart failure by studying changes in the whole nodal transcriptome. The mouse transverse aortic constriction model of pressure overload-induced heart failure was studied; functional changes were assessed using electrocardiography and echocardiography and the transcriptome of the AV node was quantified using RNAseq. Heart failure was associated with a significant increase in the PR interval, indicating a slowing of AV node conduction and AV node dysfunction, and significant changes in 3,077 transcripts (5.6% of the transcriptome). Many systems were affected: transcripts supporting AV node conduction were downregulated and there were changes in transcripts identified by GWAS as determinants of the PR interval. In addition, there was evidence of remodelling of the sarcomere, a shift from fatty acid to glucose metabolism, remodelling of the extracellular matrix, and remodelling of the transcription and translation machinery. There was evidence of the causes of this widespread remodelling of the AV node: evidence of dysregulation of multiple intracellular signalling pathways, dysregulation of 109 protein kinases and 148 transcription factors, and an immune response with a proliferation of neutrophils, monocytes, macrophages and B lymphocytes and a dysregulation of 40 cytokines. In conclusion, inflammation and a widespread transcriptional remodelling of the AV node underlies AV node dysfunction in heart failure.
Collapse
Affiliation(s)
- Claire Wilson
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Min Zi
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Matthew Smith
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Munir Hussain
- Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
| | - Alicia D’Souza
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
| | - Halina Dobrzynski
- Division of Cardiovascular Sciences, University of Manchester, Manchester, United Kingdom
- Department of Anatomy, Jagiellonian University Medical College, Kraków, Poland
- *Correspondence: Halina Dobrzynski, ; Mark R. Boyett,
| | - Mark R. Boyett
- Faculty of Life Sciences, University of Bradford, Bradford, United Kingdom
- *Correspondence: Halina Dobrzynski, ; Mark R. Boyett,
| |
Collapse
|
31
|
Massironi S, Mulinacci G, Gallo C, Viganò C, Fichera M, Villatore A, Peretto G, Danese S. The oft-overlooked cardiovascular complications of inflammatory bowel disease. Expert Rev Clin Immunol 2023; 19:375-391. [PMID: 36722283 DOI: 10.1080/1744666x.2023.2174971] [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: 12/05/2022] [Revised: 01/13/2023] [Accepted: 01/27/2023] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Inflammatory bowel disease (IBD) may be associated with several extraintestinal comorbidities, including cardiovascular disease (CVD). Chronic inflammation is recognized as an important factor in atherogenesis, thrombosis, and myocarditis. AREAS COVERED IBD patients may be at increased risk for developing early atherosclerosis, cardiovascular events, peripheral artery disease, venous thromboembolism, myocarditis, and arrhythmias. Anti-tumor necrosis factor agents and thiopurines have been shown to have a protective effect against acute arterial events, but more research is needed. However, an increased risk of venous thromboembolism and major cardiovascular events has been described with the use of Janus kinase inhibitors. EXPERT OPINION CVD risk is slightly increased in patients with IBD, especially during flares. Thromboprophylaxis is strongly recommended in hospitalized patients with active disease as the benefit of anticoagulation outweighs the risk of bleeding. The pathogenetic relationship between CVD and IBD and the impact of IBD drugs on CVD outcomes are not fully elucidated. CVD risk doesn't have the strength to drive a specific IBD treatment. However, proper CVD risk profiling should always be done and the best strategy to manage CVD risk in IBD patients is to combine appropriate thromboprophylaxis with early and durable remission of the underlying IBD.
Collapse
Affiliation(s)
- Sara Massironi
- Division of Gastroenterology, and Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, University of Milano-Bicocca School of Medicine, Monza, Italy
| | - Giacomo Mulinacci
- Division of Gastroenterology, and Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, University of Milano-Bicocca School of Medicine, Monza, Italy
| | - Camilla Gallo
- Division of Gastroenterology, and Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, University of Milano-Bicocca School of Medicine, Monza, Italy
| | - Chiara Viganò
- Division of Gastroenterology, and Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, University of Milano-Bicocca School of Medicine, Monza, Italy
| | - Maria Fichera
- Division of Gastroenterology, and Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Fondazione IRCCS San Gerardo dei Tintori, University of Milano-Bicocca School of Medicine, Monza, Italy
| | - Andrea Villatore
- Myocarditis Disease Unit, Department of Cardiac Electrophysiology and Arrhythmology, IRCCS Ospedale San Raffaele, Milan, Italy, and Vita-Salute San Raffaele University, Milan, Italy
| | - Giovanni Peretto
- Myocarditis Disease Unit, Department of Cardiac Electrophysiology and Arrhythmology, IRCCS Ospedale San Raffaele, Milan, Italy, and Vita-Salute San Raffaele University, Milan, Italy
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy, and Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
32
|
Kim IK, Song BW, Lim S, Kim SW, Lee S. The Role of Epicardial Adipose Tissue-Derived MicroRNAs in the Regulation of Cardiovascular Disease: A Narrative Review. BIOLOGY 2023; 12:498. [PMID: 37106699 PMCID: PMC10135702 DOI: 10.3390/biology12040498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 03/29/2023]
Abstract
Cardiovascular diseases have been leading cause of death worldwide for many decades, and obesity has been acknowledged as a risk factor for cardiovascular diseases. In the present review, human epicardial adipose tissue-derived miRNAs reported to be differentially expressed under pathologic conditions are discussed and summarized. The results of the literature review indicate that some of the epicardial adipose tissue-derived miRNAs are believed to be cardioprotective, while some others show quite the opposite effects depending on the underlying pathologic conditions. Furthermore, they suggest that that the epicardial adipose tissue-derived miRNAs have great potential as both a diagnostic and therapeutic modality. Nevertheless, mainly due to highly limited availability of human samples, it is very difficult to make any generalized claims on a given miRNA in terms of its overall impact on the cardiovascular system. Therefore, further functional investigation of a given miRNA including, but not limited to, the study of its dose effect, off-target effects, and potential toxicity is required. We hope that this review can provide novel insights to transform our current knowledge on epicardial adipose tissue-derived miRNAs into clinically viable therapeutic strategies for preventing and treating cardiovascular diseases.
Collapse
Affiliation(s)
- Il-Kwon Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
- International St. Mary’s Hospital, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Byeong-Wook Song
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
- International St. Mary’s Hospital, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Soyeon Lim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
- International St. Mary’s Hospital, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Sang-Woo Kim
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
- International St. Mary’s Hospital, Catholic Kwandong University, Incheon 22711, Republic of Korea
| | - Seahyoung Lee
- Institute for Bio-Medical Convergence, College of Medicine, Catholic Kwandong University, Gangneung-si 25601, Republic of Korea
- International St. Mary’s Hospital, Catholic Kwandong University, Incheon 22711, Republic of Korea
| |
Collapse
|
33
|
Gonzalez DE, Waldman HS, McAllister MJ. The Metabolic and Physiological Demands of a Simulated Fire Ground Test Versus a Live-Fire Training Evolution in Professional Firefighters. INTERNATIONAL JOURNAL OF EXERCISE SCIENCE 2023; 16:230-241. [PMID: 37113620 PMCID: PMC10124731 DOI: 10.70252/nwbq9464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Objective This study examined the similarities in metabolic and physiological demands of a fire ground test (FGT) and a live fire training evolution. Methods Twenty-seven firefighters completed either a FGT (n = 13) or a live fire training evolution (n = 14). Salivary samples were collected pre, immediately post, and 30-minutes post FGT and live fire training evolution, and analyzed for cortisol, uric acid, and interleukin-1β (IL-1β). Heart rate (HR) was measured pre- and post-task. Results Both tasks resulted in significant elevations in cortisol, IL-1β, and HR. Conclusions Both the FGT and live fire training evolution appear to result in similar metabolic and physiological demands. Further work may expand upon the additional elements (i.e., added heat) of the live fire training evolution. Fire departments may consider incorporating a variety of high intensity training to prepare personnel for these occupational demands.
Collapse
Affiliation(s)
- Drew E Gonzalez
- Department of Kinesiology & Sports Management, Texas A&M University, College Station, TX 77843
| | - Hunter S Waldman
- Human Performance Research Laboratory, Department of Kinesiology, University of North Alabama, Florence, AL 35630
| | - Matthew J McAllister
- Metabolic & Applied Physiology Laboratory, Department of Health & Human Performance, Texas State University, San Marcos, TX 78666
| |
Collapse
|
34
|
Wang Z, Xiao D, Ji Q, Li Y, Cai Z, Fang L, Huo H, Zhou G, Yan X, Shen L, He B. Jujuboside A attenuates sepsis-induced cardiomyopathy by inhibiting inflammation and regulating autophagy. Eur J Pharmacol 2022; 947:175451. [PMID: 36502962 DOI: 10.1016/j.ejphar.2022.175451] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Jujuboside A (JuA), as a main effective component of Jujubogenin, has long been known as a sedative-hypnotic drug. The aim of the current study was to investigate the potential effect of JuA on sepsis-induced cardiomyopathy (SIC) induced by lipopolysaccharide (LPS). METHOD Wide type C57BL/6 mice and neonatal rat cardiomyocytes (NRCMs) were exposed to LPS to establish myocardial toxicity models. Cardiac function of septic mice was detected by echocardiography. Moreover, the survival rate was calculated for 7 days. ELISA assays were used to analyze inflammatory factors in serum. Furthermore, western blotting, flow cytometry and TUNEL staining were performed to assess cell apoptosis and transmission electron microscopy detect the number of autophagosomes in myocardium. Finally, the expression of proteins related to pyroptosis, autophagy and oxidative stress was analyzed by western blotting and immunohistochemistry staining. RESULTS Results showed that JuA pretreatment significantly improved the survival rate and cardiac function, and suppressed systemic inflammatory response in septic mice. Further study revealed that JuA could decrease cell apoptosis and pyroptosis; instead, it strengthened autophagy in SIC. Moreover, JuA also significantly decreased oxidative stress and nitrodative stress, as evidenced by suppressing the superoxide production and downregulating iNOS and gp91 expression in vivo. In addition, the autophagy inhibitor 3-MA significantly abolished the effect of JuA on autophagic activity in SIC. CONCLUSION In conclusion, the findings indicated that JuA attenuates cardiac function via blocking inflammasome-mediated apoptosis and pyroptosis, at the same time by enhancing autophagy in SIC, heralding JuA as a potential therapy for sepsis.
Collapse
|
35
|
Combination of Spirulina platensis, Ganoderma lucidum and Moringa oleifera Improves Cardiac Functions and Reduces Pro-Inflammatory Biomarkers in Preclinical Models of Short-Term Doxorubicin-Mediated Cardiotoxicity: New Frontiers in Cardioncology? J Cardiovasc Dev Dis 2022; 9:jcdd9120423. [PMID: 36547420 PMCID: PMC9780956 DOI: 10.3390/jcdd9120423] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/02/2022] Open
Abstract
Anthracyclines are essential adjuvant therapies for a variety of cancers, particularly breast, gastric and esophageal cancers. Whilst prolonging cancer-related survival, these agents can induce drug-related cardiotoxicity. Spirulina, Reishi (Ganoderma lucidum) and Moringa are three nutraceuticals with anti-inflammatory effects that are currently used in cancer patients as complementary and alternative medicines to improve quality of life and fatigue. We hypothesize that the nutraceutical combination of Spirulina, Reishi and Moringa (Singo) could reduce inflammation and cardiotoxicity induced by anthracyclines. Female C57Bl/6 mice were untreated (Sham, n = 6) or treated for 7 days with short-term doxorubicin (DOXO, n = 6) or Singo (Singo, n = 6), or pre-treated with Singo for 3 days and associated with DOXO for remaining 7 days (DOXO−Singo, n = 6). The ejection fraction and radial and longitudinal strain were analyzed through transthoracic echocardiography (Vevo 2100, Fujifilm, Tokyo, Japan). The myocardial expressions of NLRP3, DAMPs (galectin-3 and calgranulin S100) and 13 cytokines were quantified through selective mouse ELISA methods. Myocardial fibrosis, necrosis and hypertrophy were analyzed through immunohistochemistry (IHC). Human cardiomyocytes were exposed to DOXO (200 nM) alone or in combination with Singo (at 10, 25 and 50 µg/mL) for 24 and 48 h. Cell viability and inflammation studies were also performed. In preclinical models, Singo significantly improved ejection fraction and fractional shortening. Reduced expressions of myocardial NLRP3 and NF-kB levels in cardiac tissues were seen in DOXO−Singo mice vs. DOXO (p < 0.05). The myocardial levels of calgranulin S100 and galectin-3 were strongly reduced in DOXO−Singo mice vs. DOXO (p < 0.05). Immunohistochemistry analysis indicates that Singo reduces fibrosis and hypertrophy in the myocardial tissues of mice during exposure to DOXO. In conclusion, in the preclinical model of DOXO-induced cardiotoxicity, Singo is able to improve cardiac function and reduce biomarkers involved in heart failure and fibrosis.
Collapse
|
36
|
MacDonnell S, Megna J, Ruan Q, Zhu O, Halasz G, Jasewicz D, Powers K, E H, del Pilar Molina-Portela M, Jin X, Zhang D, Torello J, Feric NT, Graziano MP, Shekhar A, Dunn ME, Glass D, Morton L. Activin A directly impairs human cardiomyocyte contractile function indicating a potential role in heart failure development. Front Cardiovasc Med 2022; 9:1038114. [PMID: 36440002 PMCID: PMC9685658 DOI: 10.3389/fcvm.2022.1038114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/10/2022] [Indexed: 09/27/2023] Open
Abstract
Activin A has been linked to cardiac dysfunction in aging and disease, with elevated circulating levels found in patients with hypertension, atherosclerosis, and heart failure. Here, we investigated whether Activin A directly impairs cardiomyocyte (CM) contractile function and kinetics utilizing cell, tissue, and animal models. Hydrodynamic gene delivery-mediated overexpression of Activin A in wild-type mice was sufficient to impair cardiac function, and resulted in increased cardiac stress markers (N-terminal pro-atrial natriuretic peptide) and cardiac atrophy. In human-induced pluripotent stem cell-derived (hiPSC) CMs, Activin A caused increased phosphorylation of SMAD2/3 and significantly upregulated SERPINE1 and FSTL3 (markers of SMAD2/3 activation and activin signaling, respectively). Activin A signaling in hiPSC-CMs resulted in impaired contractility, prolonged relaxation kinetics, and spontaneous beating in a dose-dependent manner. To identify the cardiac cellular source of Activin A, inflammatory cytokines were applied to human cardiac fibroblasts. Interleukin -1β induced a strong upregulation of Activin A. Mechanistically, we observed that Activin A-treated hiPSC-CMs exhibited impaired diastolic calcium handling with reduced expression of calcium regulatory genes (SERCA2, RYR2, CACNB2). Importantly, when Activin A was inhibited with an anti-Activin A antibody, maladaptive calcium handling and CM contractile dysfunction were abrogated. Therefore, inflammatory cytokines may play a key role by acting on cardiac fibroblasts, causing local upregulation of Activin A that directly acts on CMs to impair contractility. These findings demonstrate that Activin A acts directly on CMs, which may contribute to the cardiac dysfunction seen in aging populations and in patients with heart failure.
Collapse
Affiliation(s)
| | - Jake Megna
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Qin Ruan
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Olivia Zhu
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Gabor Halasz
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Dan Jasewicz
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Kristi Powers
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Hock E
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | | | - Ximei Jin
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Dongqin Zhang
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | | | - Nicole T. Feric
- TARA Biosystems Inc., Alexandria Center for Life Sciences, New York, NY, United States
| | - Michael P. Graziano
- TARA Biosystems Inc., Alexandria Center for Life Sciences, New York, NY, United States
| | | | | | - David Glass
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| | - Lori Morton
- Regeneron Pharmaceuticals, Tarrytown, NY, United States
| |
Collapse
|
37
|
Chaffey L, Roberti A, Greaves DR. Drug repurposing in cardiovascular inflammation: Successes, failures, and future opportunities. Front Pharmacol 2022; 13:1046406. [PMID: 36339576 PMCID: PMC9634418 DOI: 10.3389/fphar.2022.1046406] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/10/2022] [Indexed: 12/15/2022] Open
Abstract
Drug repurposing is an attractive, pragmatic approach to drug discovery that has yielded success across medical fields over the years. The use of existing medicines for novel indications enables dramatically reduced development costs and timescales compared with de novo drug discovery and is therefore a promising strategy in cardiovascular disease, where new drug approvals lag significantly behind that of other fields. Extensive evidence from pre-clinical and clinical studies show that chronic inflammation is a driver of pathology in cardiovascular disease, and many efforts have been made to target cardiovascular inflammation therapeutically. This approach has been met with significant challenges however, namely off-target effects associated with broad-spectrum immunosuppression, particularly in long-term conditions such as cardiovascular disease. Nevertheless, multiple anti-inflammatory medicines have been assessed for efficacy in cardiovascular clinical trials, with most of these being repurposed from their original indications in autoimmune conditions like rheumatoid arthritis. In this review, we discuss the mixed successes of clinical trials investigating anti-inflammatory drugs in cardiovascular disease, with examples such as anti-cytokine monoclonal antibodies, colchicine, and methotrexate. Looking to the future, we highlight potential new directions for drug repurposing in cardiovascular inflammation, including the emerging concepts of drug re-engineering and chrono-pharmacology.
Collapse
Affiliation(s)
| | | | - David R. Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
38
|
Jiao Y, Zhang Q, Zhang J, Zha Y, Wang J, Li Y, Zhang S. Platelet-rich plasma ameliorates lipopolysaccharide-induced cardiac injury by inflammation and ferroptosis regulation. Front Pharmacol 2022; 13:1026641. [PMID: 36330090 PMCID: PMC9623117 DOI: 10.3389/fphar.2022.1026641] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/04/2022] [Indexed: 11/25/2022] Open
Abstract
Sepsis-induced myocardial dysfunction (SIMD) is a fatal disease with no specific treatment worldwide to this day. As a biological product, platelet-rich plasma (PRP) has attracted much attention due to its diverse and potential biological effects. However, its role in lipopolysaccharide (LPS)-induced cardiac injury has not been fully investigated. This study aimed to explore the mechanism of PRP in SIMD. PRP (30 µL) was injected in situ into the heart, and LPS (10 mg/kg) was injected intraperitoneally into mice. Neonatal rat cardiomyocytes were treated with LPS (1 μg/ml) for 24 h. The results showed that, compared with the LPS group, PRP significantly decreased the levels of Lactate dehydrogenase (LDH) and Creatine Kinase MB (CK-MB), and improved cardiac function. In addition, PRP markedly decreased the Malonic dialdehyde (MDA) content, and increased the Superoxide dismutase (SOD) activity and Glutathione (GSH) level, demonstrating that PRP alleviated LPS-induced oxidative stress. The Western blot and qPCR results showed that LPS-induced ferroptosis and inflammation effects in vivo and in vitro were ameliorated after PRP treatment. Moreover, PRP can alleviate erastin-induced ferroptosis and improve cell viability. Mechanistically, p-AKT and p-mTOR expressions were down-regulated after treatment with LPS, while PRP pretreatment could reverse this effect. In summary, our study demonstrated that PRP could play a unique role in reducing LPS-induced cardiac injury through regulation of AKT/mTOR signaling pathways. These findings provide a new therapeutic direction for treating SIMD.
Collapse
Affiliation(s)
- Yuheng Jiao
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingyu Zhang
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Jiayan Zhang
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yafang Zha
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Wang
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanyan Li
- Department of Cardiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yanyan Li, ; Song Zhang,
| | - Song Zhang
- Department of Cardiology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yanyan Li, ; Song Zhang,
| |
Collapse
|
39
|
Mohebi R, McCarthy CP, Gaggin HK, van Kimmenade RRJ, Januzzi JL. Inflammatory biomarkers and risk of cardiovascular events in patients undergoing coronary angiography. Am Heart J 2022; 252:51-59. [PMID: 35753356 PMCID: PMC9336200 DOI: 10.1016/j.ahj.2022.06.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Inflammation, measured by traditional biomarkers such as C-reactive protein, has been linked to cardiovascular (CV) events. Recent technological advancement has allowed for measuring larger numbers of inflammatory biomarkers. A contemporary evaluation with established and novel biomarkers of inflammation is needed. METHODS 1,090 individuals who underwent coronary angiography were enrolled. Twenty-four inflammatory biomarkers were collected prior to angiography. Unsupervised machine learning cluster analyses determined unique patterns of inflammatory biomarkers. Cox proportional hazard regression assessed both association of inflammatory biomarker clusters and individual biomarker associations with major adverse cardiovascular events (MACE; non-fatal myocardial infarction or stroke, and CV death) during a median follow-up of 3.67 years. RESULTS Four distinct clusters were recognized. Incremental increases in inflammatory biomarkers were observed from cluster 1 to cluster 4. During follow-up, 263 MACE were ascertained. Considering cluster 1 as a reference, study participants with inflammatory cluster 2 (Hazard ratio [HR] 1.55, 95% confidence interval [CI]: 1.01-2.37), cluster 3 (HR 1.89, CI: 1.25-2.85), and cluster 4 (HR 2.93, CI: 1.95-4.42) were at increased risk of MACE. Interleukin (IL)-1α IL-6, IL-8, IL-10, IL-12, Adhesion molecule-1 high-sensitivity C-reactive protein, ferritin, myeloperoxidase, macrophage inflammatory protein (MIP)-1a, MIP 3, and macrophage colony-stimulating factor-1 were independently associated with MACE. CONCLUSIONS Among persons undergoing coronary angiography procedures, distinct clusters of inflammatory biomarker distributions with significant prognostic meaning may be identified. These results may identify unique targets for anti-inflammatory treatments aimed at CV disease.
Collapse
Affiliation(s)
- Reza Mohebi
- Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Cian P McCarthy
- Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | - Hanna K Gaggin
- Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA
| | | | - James L Januzzi
- Massachusetts General Hospital, Boston, MA; Harvard Medical School, Boston, MA; Baim Institute for Clinical Research, Boston, MA.
| |
Collapse
|
40
|
Daniels CC, Isaacs Z, Finelli R, Leisegang K. The efficacy of Zingiber officinale on dyslipidaemia, blood pressure, and inflammation as cardiovascular risk factors: A systematic review. Clin Nutr ESPEN 2022; 51:72-82. [PMID: 36184251 DOI: 10.1016/j.clnesp.2022.08.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/20/2022] [Accepted: 08/26/2022] [Indexed: 10/14/2022]
Abstract
BACKGROUND & AIMS Hypertension, dyslipidaemia, and chronic inflammation contribute to the development of cardiovascular disease (CVD). Zingiber officinale has been suggested to reduce these CVD risk factors; however, the clinical evidence remains unclear. This systematic review aims to analyse the effect of Z. officinale as a sole intervention on these risk factors. METHODS In this PRISMA-based systematic review, we included randomised clinical trials from PubMed, Scopus and Cochrane Database of Systematic Reviews (July 2020) analysing triglycerides, low- and high-density lipoprotein (LDL, HDL), total cholesterol, C-reactive protein (CRP), tumor necrosis factor-alpha (TNF-α), interleukin 1, 6, 10, systolic and/or diastolic blood pressure as outcomes. Quality of studies was evaluated by JADAD and the Cochrane risk-of-bias tools. RESULTS A total of 24 studies were included, mostly (79.2%) showing low risk of bias. These were based on obesity and cardio-metabolic derangements (33.3%), type 2 diabetes mellitus (37.5%), and miscellaneous conditions (29.2%). While total cholesterol and triglycerides levels mostly improved after Z. officinale, results were inconsistent for other blood lipids markers. Inflammatory markers (CRP, TNF-α) were more consistently reduced by Z. officinale, while only 3 studies reported a non-significant reduction of blood pressure. CONCLUSIONS Although there remains a paucity of studies, Z. officinale may be beneficial for improving dyslipidaemia and inflammation.
Collapse
Affiliation(s)
- Chelsea Courtney Daniels
- School of Natural Medicine, Faculty of Community and Health Sciences, University of the Western Cape, Bellville, South Africa
| | - Zaiyaan Isaacs
- School of Natural Medicine, Faculty of Community and Health Sciences, University of the Western Cape, Bellville, South Africa
| | | | - Kristian Leisegang
- School of Natural Medicine, Faculty of Community and Health Sciences, University of the Western Cape, Bellville, South Africa.
| |
Collapse
|
41
|
Cornuault L, Rouault P, Duplàa C, Couffinhal T, Renault MA. Endothelial Dysfunction in Heart Failure With Preserved Ejection Fraction: What are the Experimental Proofs? Front Physiol 2022; 13:906272. [PMID: 35874523 PMCID: PMC9304560 DOI: 10.3389/fphys.2022.906272] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) has been recognized as the greatest single unmet need in cardiovascular medicine. Indeed, the morbi-mortality of HFpEF is high and as the population ages and the comorbidities increase, so considerably does the prevalence of HFpEF. However, HFpEF pathophysiology is still poorly understood and therapeutic targets are missing. An unifying, but untested, theory of the pathophysiology of HFpEF, proposed in 2013, suggests that cardiovascular risk factors lead to a systemic inflammation, which triggers endothelial cells (EC) and coronary microvascular dysfunction. This cardiac small vessel disease is proposed to be responsible for cardiac wall stiffening and diastolic dysfunction. This paradigm is based on the fact that microvascular dysfunction is highly prevalent in HFpEF patients. More specifically, HFpEF patients have been shown to have decreased cardiac microvascular density, systemic endothelial dysfunction and a lower mean coronary flow reserve. Importantly, impaired coronary microvascular function has been associated with the severity of HF. This review discusses evidence supporting the causal role of endothelial dysfunction in the pathophysiology of HFpEF in human and experimental models.
Collapse
|
42
|
Haji Aghajani M, Toloui A, Ahmadzadeh K, Madani Neishaboori A, Yousefifard M. Premature Coronary Artery Disease and Plasma Levels of Interleukins; a Systematic Scoping Review and Meta-Analysis. ARCHIVES OF ACADEMIC EMERGENCY MEDICINE 2022; 10:e51. [PMID: 36033992 PMCID: PMC9397602 DOI: 10.22037/aaem.v10i1.1605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Introduction Interleukins (ILs) can act as a predictive indicator of Premature Coronary Artery Disease (pCAD) and may be useful in screening of high-risk patients. However, there is no consensus on the relationship of serum levels of ILs and pCAD, yet. As a result, this study has been conducted in order to review the literature on the relationship between serum levels of different ILs and pCAD. Methods Medline, Scopus, Embase, and Web of Science databases were searched until December 7th 2020. Two reviewers independently screened and summarized eligible articles. A meta-analysis was performed to assess the relationship of ILs and pCAD. Results 12 case-control articles were included. IL-6 plasma changes do happen in pCAD patients with a standardized mean difference (SMD) of 0.51 (95% CI: 0.12-0.90; p=0.010) compared with the control group. This difference was also observed when evaluating the plasma levels of IL-1 and IL-17, with an SMD of 1.42 (95% CI: 1.11-1.73; p<0.001) and 0.59 (95% CI: 0.14-1.04; p=0.011), respectively. Meanwhile, no significant difference existed in plasma levels of IL-10 (SMD=0.26; 95% CI: -0.17-0.70; p=0.236), and IL-18 (SMD=1.44; 95% CI: -0.19-3.07; p=0.083) between pCAD patients and those in the control group. Conclusion Low level of evidence showed that there may be a significant relationship between increased plasma levels of ILs and the occurrence of pCAD. As a result, prospective cohort studies with serial assessments of serum ILs during follow up period, focusing on controlling classical risk factors of pCAD and increase in level of ILs, should be conducted.
Collapse
Affiliation(s)
- Mohammad Haji Aghajani
- Prevention of Cardiovascular Disease Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Cardiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirmohammad Toloui
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Koohyar Ahmadzadeh
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Mahmoud Yousefifard
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Pediatric Chronic Kidney Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Ebadi N, Ghafouri-Fard S, Taheri M, Arsang-Jang S, Omrani MD. Expression analysis of inflammatory response-associated genes in coronary artery disease. Arch Physiol Biochem 2022; 128:601-607. [PMID: 31913058 DOI: 10.1080/13813455.2019.1708953] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Coronary artery disease (CAD) is among prominent causes of death throughout the world. Inflammatory processes participate in the pathogenesis of this disorder. METHODS In the current case-control study, we compared expression levels of three inflammation-associated genes namely Antisense noncoding RNA in the INK4 locus (ANRIL), NKILA and IL-1B between CAD patients and matched healthy subjects. RESULTS ANRIL, IL-1B and NKILA were significantly down-regulated in CAD patients compared with controls (p values of <.0001, .023 and <.0001, respectively). When evaluating study participants based on their gender, the differences in expression levels of ANRIL and NKILA were significant in both male and female patients compared with the matched controls. However, IL-1B was only down-regulated in female patients compared with female controls. CONCLUSION Taken together, our study revealed dysregulation of inflammation-associated genes in the peripheral blood of CAD patients and supported the previously suggested role of inflammation in the pathogenesis of CAD.
Collapse
Affiliation(s)
- Nader Ebadi
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Arsang-Jang
- Department of Epidemiology and Biostatistics, Cellular and Molecular Research Center, Faculty of Health, Qom University of Medical Sciences, Qom, Iran
| | - Mir Davood Omrani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Younes R, LeBlanc CA, Hiram R. Evidence of Failed Resolution Mechanisms in Arrhythmogenic Inflammation, Fibrosis and Right Heart Disease. Biomolecules 2022; 12:biom12050720. [PMID: 35625647 PMCID: PMC9138906 DOI: 10.3390/biom12050720] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a complex program of active processes characterized by the well-orchestrated succession of an initiation and a resolution phase aiming to promote homeostasis. When the resolution of inflammation fails, the tissue undergoes an unresolved inflammatory status which, if it remains uncontrolled, can lead to chronic inflammatory disorders due to aggravation of structural damages, development of a fibrous area, and loss of function. Various human conditions show a typical unresolved inflammatory profile. Inflammatory diseases include cancer, neurodegenerative disease, asthma, right heart disease, atherosclerosis, myocardial infarction, or atrial fibrillation. New evidence has started to emerge on the role, including pro-resolution involvement of chemical mediators in the acute phase of inflammation. Although flourishing knowledge is available about the role of specialized pro-resolving mediators in neurodegenerative diseases, atherosclerosis, obesity, or hepatic fibrosis, little is known about their efficacy to combat inflammation-associated arrhythmogenic cardiac disorders. It has been shown that resolvins, including RvD1, RvE1, or Mar1, are bioactive mediators of resolution. Resolvins can stop neutrophil activation and infiltration, stimulate monocytes polarization into anti-inflammatory-M2-macrophages, and activate macrophage phagocytosis of inflammation-debris and neutrophils to promote efferocytosis and clearance. This review aims to discuss the paradigm of failed-resolution mechanisms (FRM) potentially promoting arrhythmogenicity in right heart disease-induced inflammatory status.
Collapse
Affiliation(s)
- Rim Younes
- Montreal Heart Institute (MHI), Montreal, QC H1T 1C8, Canada; (R.Y.); (C.-A.L.)
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Charles-Alexandre LeBlanc
- Montreal Heart Institute (MHI), Montreal, QC H1T 1C8, Canada; (R.Y.); (C.-A.L.)
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Roddy Hiram
- Montreal Heart Institute (MHI), Montreal, QC H1T 1C8, Canada; (R.Y.); (C.-A.L.)
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Correspondence: ; Tel.: +1-514-376-3330 (ext. 5015)
| |
Collapse
|
45
|
Ku EJ, Kim BR, Lee JI, Lee YK, Oh TJ, Jang HC, Choi SH. The Anti-Atherosclerosis Effect of Anakinra, a Recombinant Human Interleukin-1 Receptor Antagonist, in Apolipoprotein E Knockout Mice. Int J Mol Sci 2022; 23:ijms23094906. [PMID: 35563294 PMCID: PMC9104865 DOI: 10.3390/ijms23094906] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 12/10/2022] Open
Abstract
Interleukin (IL)-1β plays an important role in atherosclerosis pathogenesis. We aimed to investigate the effect of anakinra, a recombinant human IL-1 receptor antagonist, on the progression of atherosclerosis in apolipoprotein E knockout (ApoE−/−) mice. ApoE−/− mice (8-week male) were treated with saline (control), anakinra 10, 25, and 50 mg/kg, respectively (n = 10 in each group). Mice were fed a standard chow (4 weeks) followed by an atherogenic diet (35kcal% fat, 1.25% cholesterol, 12 weeks). Atheromatous plaques in ApoE−/− mice and the expression of inflammatory genes and signaling pathways in human umbilical vein endothelial cells (HUVECs), rat aortic smooth muscle cells (RAOSMCs), and 3T3-L1 adipocytes were assessed. Anakinra reduced the plaque size of the aortic arch (30.6% and 25.2% at the 25 mg/kg and 50 mg/kg doses, both p < 0.05) and serum triglyceride in ApoE−/− mice and suppressed inflammatory genes (IL-1β and IL-6) expressions in HUVECs and RAOSMCs (all p < 0.05). In RAOSMCs, anakinra reduced metalloproteinase-9 expression in a dose-dependent manner and inhibited cell migration. Anakinra-treated mice exhibited trends of lower CD68+ macrophage infiltration in visceral fat and monocyte chemoattractant protein-1 expression was reduced in 3T3-L1 adipocytes. Anakinra could be a useful component for complementary treatment with a standard regimen to reduce the residual cardiovascular risk.
Collapse
Affiliation(s)
- Eu Jeong Ku
- Department of Internal Medicine, Chungbuk National University Hospital, Cheongju 28644, Korea;
- Department of Internal Medicine, Chungbuk National University College of Medicine, Cheongju 28644, Korea
| | - Bo-Rahm Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
| | - Jee-In Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
| | - Yun Kyung Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
| | - Tae Jung Oh
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hak C. Jang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sung Hee Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea; (B.-R.K.); (J.-I.L.); (Y.K.L.); (T.J.O.); (H.C.J.)
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
- Correspondence: ; Tel.: +82-31-787-7033
| |
Collapse
|
46
|
Awuah A, Moore JS, Nesbit MA, Ruddock MW, Brennan PF, Mailey JA, McNeil AJ, Jing M, Finlay DD, Trucco E, Kurth MJ, Watt J, Lamont JV, Fitzgerald P, Spence MS, McLaughlin JAD, Moore TCB. A novel algorithm for cardiovascular screening using conjunctival microcirculatory parameters and blood biomarkers. Sci Rep 2022; 12:6545. [PMID: 35449196 PMCID: PMC9023476 DOI: 10.1038/s41598-022-10491-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 03/15/2022] [Indexed: 11/30/2022] Open
Abstract
Microvascular haemodynamic alterations are associated with coronary artery disease (CAD). The conjunctival microcirculation can easily be assessed non-invasively. However, the microcirculation of the conjunctiva has not been previously explored in clinical algorithms aimed at identifying patients with CAD. This case–control study involved 66 patients with post-myocardial infarction and 66 gender-matched healthy controls. Haemodynamic properties of the conjunctival microcirculation were assessed with a validated iPhone and slit lamp-based imaging tool. Haemodynamic properties were extracted with semi-automated software and compared between groups. Biomarkers implicated in the development of CAD were assessed in combination with conjunctival microcirculatory parameters. The conjunctival blood vessel parameters and biomarkers were used to derive an algorithm to aid in the screening of patients for CAD. Conjunctival blood velocity measured in combination with the blood biomarkers (N-terminal pro-brain natriuretic peptide and adiponectin) had an area under receiver operator characteristic curve (AUROC) of 0.967, sensitivity 93.0%, specificity 91.5% for CAD. This study demonstrated that the novel algorithm which included a combination of conjunctival blood vessel haemodynamic properties, and blood-based biomarkers could be used as a potential screening tool for CAD and should be validated for potential utility in asymptomatic individuals.
Collapse
Affiliation(s)
- Agnes Awuah
- Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine, BT52 1SA, UK
| | - Julie S Moore
- Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine, BT52 1SA, UK
| | - M Andrew Nesbit
- Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine, BT52 1SA, UK
| | - Mark W Ruddock
- Clinical Studies Group, Randox Laboratories Ltd, 55 Diamond Road, Crumlin, BT29 4QY, UK
| | - Paul F Brennan
- Department of Cardiology, Royal Victoria Hospital, Belfast Health and Social Care Trust, 274 Grosvenor Road, Belfast, BT12 6BA, UK
| | - Jonathan A Mailey
- Department of Cardiology, Royal Victoria Hospital, Belfast Health and Social Care Trust, 274 Grosvenor Road, Belfast, BT12 6BA, UK
| | - Andrew J McNeil
- VAMPIRE Project, Computing (SSEN), University of Dundee, Dundee, DD1 4HN, UK
| | - Min Jing
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), Ulster University, Jordanstown, BT37 0QB, UK
| | - Dewar D Finlay
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), Ulster University, Jordanstown, BT37 0QB, UK
| | - Emanuele Trucco
- VAMPIRE Project, Computing (SSEN), University of Dundee, Dundee, DD1 4HN, UK
| | - Mary Jo Kurth
- Clinical Studies Group, Randox Laboratories Ltd, 55 Diamond Road, Crumlin, BT29 4QY, UK
| | - Joanne Watt
- Clinical Studies Group, Randox Laboratories Ltd, 55 Diamond Road, Crumlin, BT29 4QY, UK
| | - John V Lamont
- Clinical Studies Group, Randox Laboratories Ltd, 55 Diamond Road, Crumlin, BT29 4QY, UK
| | - Peter Fitzgerald
- Clinical Studies Group, Randox Laboratories Ltd, 55 Diamond Road, Crumlin, BT29 4QY, UK
| | - Mark S Spence
- Department of Cardiology, Royal Victoria Hospital, Belfast Health and Social Care Trust, 274 Grosvenor Road, Belfast, BT12 6BA, UK
| | - James A D McLaughlin
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), Ulster University, Jordanstown, BT37 0QB, UK
| | - Tara C B Moore
- Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine, BT52 1SA, UK.
| |
Collapse
|
47
|
Infante M, Padilla N, Alejandro R, Caprio M, Della-Morte D, Fabbri A, Ricordi C. Diabetes-Modifying Antirheumatic Drugs: The Roles of DMARDs as Glucose-Lowering Agents. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:571. [PMID: 35629988 PMCID: PMC9143119 DOI: 10.3390/medicina58050571] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/14/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023]
Abstract
Systemic inflammation represents a shared pathophysiological mechanism which underlies the frequent clinical associations among chronic inflammatory rheumatic diseases (CIRDs), insulin resistance, type 2 diabetes (T2D), and chronic diabetes complications, including cardiovascular disease. Therefore, targeted anti-inflammatory therapies are attractive and highly desirable interventions to concomitantly reduce rheumatic disease activity and to improve glucose control in patients with CIRDs and comorbid T2D. Therapeutic approaches targeting inflammation may also play a role in the prevention of prediabetes and diabetes in patients with CIRDs, particularly in those with traditional risk factors and/or on high-dose corticosteroid therapy. Recently, several studies have shown that different disease-modifying antirheumatic drugs (DMARDs) used for the treatment of CIRDs exert antihyperglycemic properties by virtue of their anti-inflammatory, insulin-sensitizing, and/or insulinotropic effects. In this view, DMARDs are promising drug candidates that may potentially reduce rheumatic disease activity, ameliorate glucose control, and at the same time, prevent the development of diabetes-associated cardiovascular complications and metabolic dysfunctions. In light of their substantial antidiabetic actions, some DMARDs (such as hydroxychloroquine and anakinra) could be alternatively termed "diabetes-modifying antirheumatic drugs", since they may be repurposed for co-treatment of rheumatic diseases and comorbid T2D. However, there is a need for future randomized controlled trials to confirm the beneficial metabolic and cardiovascular effects as well as the safety profile of distinct DMARDs in the long term. This narrative review aims to discuss the current knowledge about the mechanisms behind the antihyperglycemic properties exerted by a variety of DMARDs (including synthetic and biologic DMARDs) and the potential use of these agents as antidiabetic medications in clinical settings.
Collapse
Affiliation(s)
- Marco Infante
- Clinical Cell Transplant Program (CCTP), Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136, USA; (R.A.); (C.R.)
- Department of Systems Medicine, Diabetes Research Institute Federation (DRIF), University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
- Section of Endocrinology, UniCamillus, Saint Camillus International University of Health Sciences, Via di Sant’Alessandro 8, 00131 Rome, Italy
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Via Cola di Rienzo 28, 00192 Rome, Italy
| | - Nathalia Padilla
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Colonia Centroamérica L-823, Managua 14048, Nicaragua;
| | - Rodolfo Alejandro
- Clinical Cell Transplant Program (CCTP), Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136, USA; (R.A.); (C.R.)
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Roma, Via di Val Cannuta 247, 00166 Rome, Italy;
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy;
| | - David Della-Morte
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy;
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Department of Neurology, Evelyn F. McKnight Brain Institute, University of Miami Miller School of Medicine, 1120 NW 14th St., Miami, FL 33136, USA
| | - Andrea Fabbri
- Department of Systems Medicine, Diabetes Research Institute Federation (DRIF), University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| | - Camillo Ricordi
- Clinical Cell Transplant Program (CCTP), Diabetes Research Institute, University of Miami Miller School of Medicine, 1450 NW 10th Ave, Miami, FL 33136, USA; (R.A.); (C.R.)
| |
Collapse
|
48
|
Overview of the Composition of Whole Grains’ Phenolic Acids and Dietary Fibre and Their Effect on Chronic Non-Communicable Diseases. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19053042. [PMID: 35270737 PMCID: PMC8910396 DOI: 10.3390/ijerph19053042] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/22/2022] [Accepted: 02/26/2022] [Indexed: 02/01/2023]
Abstract
Chronic non-communicable diseases are the major cause of death globally. Whole grains are recommended in dietary guidelines worldwide due to increasing evidence that their consumption can improve health beyond just providing energy and nutrients. Epidemiological studies have suggested that the incorporation of whole grains, as part of a healthy diet, plays a key role in reducing one’s risk for cardiovascular diseases (CVDs), obesity, type 2 diabetes (T2D) and cancer. Phenolic acids and dietary fibre are important components found in whole grains that are largely responsible for these health advantages. Both phenolic acids and dietary fibre, which are predominantly present in the bran layer, are abundant in whole-grain cereals and pseudo-cereals. Several studies indicate that whole grain dietary fibre and phenolic acids are linked to health regulation. The main focus of this study is two-fold. First, we provide an overview of phenolic acids and dietary fibres found in whole grains (wheat, barley, oats, rice and buckwheat). Second, we review existing literature on the linkages between the consumption of whole grains and the development of the following chronic non-communicable diseases: CVDs, obesity, T2D and cancer. Altogether, scientific evidence that the intake of whole grains reduces the risk of certain chronic non-communicable disease is encouraging but not convincing. Based on previous studies, the current review encourages further research to cover the gap between the emerging science of whole grains and human health.
Collapse
|
49
|
The Selective NLRP3-inflammasome inhibitor MCC950 Mitigates Post-resuscitation Myocardial Dysfunction and Improves Survival in a Rat Model of Cardiac Arrest and Resuscitation. Cardiovasc Drugs Ther 2022; 37:423-433. [PMID: 34973094 PMCID: PMC10164003 DOI: 10.1007/s10557-021-07282-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/21/2021] [Indexed: 11/03/2022]
Abstract
PURPOSE To investigate the effects of the selective NLRP3 inflammasome inhibitor MCC950 on post-resuscitation myocardial function and survival in a rat model of cardiopulmonary resuscitation (CPR). METHODS Thirty-six Sprague Dawley rats were randomized into three groups: (1) MCC950, (2) control, and (3) sham. Each group consisted of a 6 h non-survival subgroup (n = 6) and a 48 h survival subgroup (n = 6). Ventricular fibrillation (VF) was induced and untreated for 6 min. CPR was initiated and continued for 8 min. Resuscitation was attempted with a 4 J defibrillation. MCC950 (10 mg/kg) or vehicle was administered via intraperitoneal injection immediately after the return of spontaneous circulation (ROSC). Myocardial function and sublingual microcirculation were measured after ROSC in the non-survival subgroups. Plasma levels of interleukin Iβ (IL-1β) and cardiac troponin I (cTnI) were measured at baseline and 6 h in the non-survival subgroups. Heart tissue was harvested to measure the NLRP3 inflammasome constituents, including NLRP3, apoptosis-associated speck-like protein (ASC), Caspase-1, and IL-1β. Survival duration and neurologic deficit score (NDS) were recorded and evaluated among survival groups. RESULTS Post-resuscitation myocardial function and sublingual microcirculation were improved in MCC950 compared with control (p < 0.05). IL-1β and cTnI were decreased in MCC950 compared to control (p < 0.01). The MCC950 treated groups showed significantly reduced ASC, caspase-1, and IL-1β compared with the control group (p < 0.05). Survival at 48 h after ROSC was greater in MCC950 (p < 0.05) with improved NDS (p < 0.05). CONCLUSION Administration of MCC950 following ROSC mitigates post-resuscitation myocardial dysfunction and improves survival.
Collapse
|
50
|
Oikonomou E, Tsaplaris P, Anastasiou A, Xenou M, Lampsas S, Siasos G, Pantelidis P, Theofilis P, Tsatsaragkou A, Katsarou O, Sagris M, Vavuranakis MA, Vavuranakis M, Tousoulis D. Interleukin-1 in Coronary Artery Disease. Curr Top Med Chem 2022; 22:2368-2389. [PMID: 36263481 DOI: 10.2174/1568026623666221017144734] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 08/22/2022] [Accepted: 09/15/2022] [Indexed: 11/22/2022]
Abstract
Cardiovascular disease is the leading cause of mortality worldwide. Inflammation has long been established as a key component in the pathophysiology of coronary artery disease. The interleukin-1 family consists of 11 members that regulate the inflammatory response through both pro- and anti-inflammatory properties with the Nod-like receptor (NLR) family pyrin domain containing 3 inflammasome having a pivotal role in the process of converting interleukin-1 beta and interleukin- 18, two key inflammatory mediators, into their mature forms. Interleukin-1 affects various cell types that participate in the pathogenesis of atherosclerosis as it enhances the expression of leukocyte adhesion molecules on the surface of endothelial cells and augments the permeability of the endothelial cell barrier, attracting monocytes and macrophages into the vessel wall and aids the migration of smooth muscle cells toward atheroma. It also enhances the aggregation of low-density lipoprotein particles in endothelium and smooth muscle cells and exhibits procoagulant activity by inducing synthesis, cell-surface expression and release of tissue factor in endothelial cells, promoting platelet adhesion. The value of interleukin-1 as a diagnostic biomarker is currently limited, but interleukin-1 beta, interleukin-18 and interleukin-37 have shown promising data regarding their prognostic value in coronary artery disease. Importantly, target anti-inflammatory treatments have shown promising results regarding atherosclerosis progression and cardiovascular events. In this review article, we focus on the immense role of interleukin-1 in atherosclerosis progression, inflammation cascade and in the clinical application of target anti-inflammatory treatments.
Collapse
Affiliation(s)
- Evangelos Oikonomou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Paraskevas Tsaplaris
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Artemis Anastasiou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Maria Xenou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Stamatios Lampsas
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Panteleimon Pantelidis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| | - Panagiotis Theofilis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| | - Aikaterini Tsatsaragkou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Ourania Katsarou
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Marios Sagris
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| | - Michael-Andrew Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Manolis Vavuranakis
- 3rd Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Sotiria Chest Disease Hospital, Athens, Greece
| | - Dimitris Tousoulis
- 1st Department of Cardiology, National and Kapodistrian University of Athens, Medical School, Hippokration General Hospital, Athens, Greece
| |
Collapse
|