1
|
Mitsis A, Myrianthefs M, Sokratous S, Karmioti G, Kyriakou M, Drakomathioulakis M, Tzikas S, Kadoglou NPE, Karagiannidis E, Nasoufidou A, Fragakis N, Ziakas A, Kassimis G. Emerging Therapeutic Targets for Acute Coronary Syndromes: Novel Advancements and Future Directions. Biomedicines 2024; 12:1670. [PMID: 39200135 PMCID: PMC11351818 DOI: 10.3390/biomedicines12081670] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/21/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
Acute coronary syndrome (ACS) remains a major cause of morbidity and mortality worldwide, requiring ongoing efforts to identify novel therapeutic targets to improve patient outcomes. This manuscript reviews promising therapeutic targets for ACS identified through preclinical research, including novel antiplatelet agents, anti-inflammatory drugs, and agents targeting plaque stabilization. Preclinical studies have expounded these agents' efficacy and safety profiles in mitigating key pathophysiological processes underlying ACS, such as platelet activation, inflammation, and plaque instability. Furthermore, ongoing clinical trials are evaluating the efficacy and safety of these agents in ACS patients, with potential implications for optimizing ACS management. Challenges associated with translating preclinical findings into clinical practice, including patient heterogeneity and trial design considerations, are also discussed. Overall, the exploration of emerging therapeutic targets offers promising avenues for advancing ACS treatment strategies and improving patient outcomes.
Collapse
Affiliation(s)
- Andreas Mitsis
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (M.M.); (S.S.); (G.K.); (M.K.); (M.D.)
| | - Michael Myrianthefs
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (M.M.); (S.S.); (G.K.); (M.K.); (M.D.)
| | - Stefanos Sokratous
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (M.M.); (S.S.); (G.K.); (M.K.); (M.D.)
| | - Georgia Karmioti
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (M.M.); (S.S.); (G.K.); (M.K.); (M.D.)
| | - Michaela Kyriakou
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (M.M.); (S.S.); (G.K.); (M.K.); (M.D.)
| | - Michail Drakomathioulakis
- Cardiology Department, Nicosia General Hospital, State Health Services Organization, Nicosia 2029, Cyprus; (M.M.); (S.S.); (G.K.); (M.K.); (M.D.)
| | - Stergios Tzikas
- Third Department of Cardiology, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | | | - Efstratios Karagiannidis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.K.); (A.N.); (N.F.); (G.K.)
| | - Athina Nasoufidou
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.K.); (A.N.); (N.F.); (G.K.)
| | - Nikolaos Fragakis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.K.); (A.N.); (N.F.); (G.K.)
| | - Antonios Ziakas
- First Department of Cardiology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
| | - George Kassimis
- Second Department of Cardiology, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece; (E.K.); (A.N.); (N.F.); (G.K.)
| |
Collapse
|
2
|
Frąk W, Dąbek B, Balcerczyk-Lis M, Motor J, Radzioch E, Młynarska E, Rysz J, Franczyk B. Role of Uremic Toxins, Oxidative Stress, and Renal Fibrosis in Chronic Kidney Disease. Antioxidants (Basel) 2024; 13:687. [PMID: 38929126 PMCID: PMC11200916 DOI: 10.3390/antiox13060687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 05/20/2024] [Accepted: 05/25/2024] [Indexed: 06/28/2024] Open
Abstract
Affecting millions of people worldwide, chronic kidney disease is a serious medical problem. It results in a decrease in glomerular filtration rate below 60 mL/min/1.73 m, albuminuria, abnormalities in urine sediment and pathologies detected by imaging studies lasting a minimum of 3 months. Patients with CKD develop uremia, and as a result of the accumulation of uremic toxins in the body, patients can be expected to suffer from a number of medical consequences such as progression of CKD with renal fibrosis, development of atherosclerosis or increased incidence of cardiovascular events. Another key element in the pathogenesis of CKD is oxidative stress, resulting from an imbalance between the production of antioxidants and the production of reactive oxygen species. Oxidative stress contributes to damage to cellular proteins, lipids and DNA and increases inflammation, perpetuating kidney dysfunction. Additionally, renal fibrogenesis involving the accumulation of fibrous tissue in the kidneys occurs. In our review, we also included examples of forms of therapy for CKD. To improve the condition of CKD patients, pharmacotherapy can be used, as described in our review. Among the drugs that improve the prognosis of patients with CKD, we can include: GLP-1 analogues, SGLT2 inhibitors, Finerenone monoclonal antibody-Canakinumab and Sacubitril/Valsartan.
Collapse
Affiliation(s)
- Weronika Frąk
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Bartłomiej Dąbek
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Marta Balcerczyk-Lis
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jakub Motor
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewa Radzioch
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Ewelina Młynarska
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrocardiology, Medical Univeristy of Lodz, ul. Zeromskiego 113, 90-549 Lodz, Poland
| |
Collapse
|
3
|
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
|
4
|
Giret C, Dos Santos Y, Blasco H, Paget C, Gonzalez L, Tressel N, Dieu M, Bigot A, Gissot V, Audemard‐Verger A, Maillot F. No evidence for systemic low-grade inflammation in adult patients with early-treated phenylketonuria: The INGRAPH study. JIMD Rep 2023; 64:446-452. [PMID: 37927482 PMCID: PMC10623104 DOI: 10.1002/jmd2.12366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/16/2023] [Accepted: 04/26/2023] [Indexed: 11/07/2023] Open
Abstract
The question of an increased cardiovascular risk has been recently raised in adults with phenylketonuria (PKU). As low-grade systemic inflammation increases cardiovascular risk, the INGRAPH study aimed to evaluate low-grade inflammation in adult PKU patients compared to healthy controls and to determine the potential influence of Phe-controlled diet on inflammation. Twenty early-treated adult PKU patients, including a subgroup of 15 classical PKU patients, and 20 healthy volunteers were included. PKU patients and healthy subjects were matched on age, sex and body mass index class. Plasma concentrations of CRP, IFNg, IL1a, IL1b, IL2, IL6, IL10, and TNFα were measured in PKU patients and compared to controls. Plasma CRP was not different in the PKU group as compared to controls. No significant differences were observed between the two groups concerning plasma cytokines concentrations. Plasma CRP and cytokine profile were not different between "on diet" and "off diet" PKU patients. All these results were similar considering only the classical PKU subgroup. No differences were shown in plasma CRP and pro-inflammatory cytokines between adult PKU patients and healthy controls. Further studies are needed, including more patients and extensive characterization of systemic low-grade inflammation, as cardiovascular risk appears to be a new concern in adult PKU population.
Collapse
Affiliation(s)
- Chloé Giret
- Service de médecine interneCHRU de ToursToursFrance
- Université de ToursToursFrance
| | - Yann Dos Santos
- Université de ToursToursFrance
- UMR INSERM, “iBrain” 1253ToursFrance
| | - Hélène Blasco
- Université de ToursToursFrance
- UMR INSERM, “iBrain” 1253ToursFrance
- Laboratoire de biochimie, CHRU de ToursToursFrance
- Centre de référence des maladies héréditaires du métabolismeToursFrance
| | - Christophe Paget
- Université de ToursToursFrance
- Centre d'étude des Pathologies Respiratoires (CEPR)UMR INSERM 1100ToursFrance
| | - Loïc Gonzalez
- Université de ToursToursFrance
- Centre d'étude des Pathologies Respiratoires (CEPR)UMR INSERM 1100ToursFrance
| | - Nathalie Tressel
- Service de médecine interneCHRU de ToursToursFrance
- Centre de référence des maladies héréditaires du métabolismeToursFrance
| | - Maeva Dieu
- Service de médecine interneCHRU de ToursToursFrance
| | - Adrien Bigot
- Service de médecine interneCHRU de ToursToursFrance
- Centre de référence des maladies héréditaires du métabolismeToursFrance
| | - Valérie Gissot
- Centre d'investigation clinique, CHRU de ToursToursFrance
| | - Alexandra Audemard‐Verger
- Service de médecine interneCHRU de ToursToursFrance
- Université de ToursToursFrance
- Centre d'étude des Pathologies Respiratoires (CEPR)UMR INSERM 1100ToursFrance
| | - François Maillot
- Service de médecine interneCHRU de ToursToursFrance
- Université de ToursToursFrance
- UMR INSERM, “iBrain” 1253ToursFrance
- Centre de référence des maladies héréditaires du métabolismeToursFrance
| |
Collapse
|
5
|
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
|
6
|
Al-Hawary SIS, Jasim SA, Romero-Parra RM, Bustani GS, Hjazi A, Alghamdi MI, Kareem AK, Alwaily ER, Zabibah RS, Gupta J, Mahmoudi R, Hosseini-Fard S. NLRP3 inflammasome pathway in atherosclerosis: Focusing on the therapeutic potential of non-coding RNAs. Pathol Res Pract 2023; 246:154490. [PMID: 37141699 DOI: 10.1016/j.prp.2023.154490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 04/19/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023]
Abstract
NLRP3 (NOD-, LRR-, and pyrin domain-containing protein 3) inflammasome pathway has a critical role in the pathogenesis of atherosclerosis. Activation of this pathway is implicated in the subendothelial inflammation and atherosclerosis progression. The NLRP3 inflammasome are cytoplasmic sensors with the distinct capacity to identify a wide range of inflammation-related signals, which enhance NLRP3 inflammasome assembly and allow it to trigger inflammation. This pathway is triggered by a variety of intrinsic signals which exist in atherosclerotic plaques, like cholesterol crystals and oxidized LDL. Further pharmacological findings indicated that NLRP3 inflammasome enhanced caspase-1-mediated secretion of pro-inflammatory mediators like interleukin (IL)- 1β/18. Newly published cutting-edge studies suggested that non-coding RNAs (ncRNAs) including microRNAs (miRNAs, miRs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs) are major modulators of NLRP3 inflammasome in atherosclerosis. Therefore, in this review, we aimed to discuss the NLRP3 inflammasome pathway, biogenesis of ncRNAs as well as the modulatory role of ncRNAs in regulating the various mediators of NLRP3 inflammasome pathway including TLR4, NF-kB, NLRP3, and caspase 1. We also discussed the importance of NLRP3 inflammasome pathway-related ncRNAs as a diagnostic biomarker in atherosclerosis and current therapeutics in the modulation of NLRP3 inflammasome in atherosclerosis. Finally, we speak about the limitations and future prospects of ncRNAs in regulating inflammatory atherosclerosis via the NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Al-anbar-Ramadi, Iraq
| | | | | | - Ahmed Hjazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Kingdom of Saudi Arabia
| | | | - Ali Kamil Kareem
- Biomedical Engineering Department, Al-Mustaqbal University College, Hillah 51001, Iraq
| | - Enas R Alwaily
- Microbiology Research Group, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, UP, India
| | - Reza Mahmoudi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Seyedreza Hosseini-Fard
- Department of Clinical Biochemistry, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
Hagerty T, Kluge MA, LeWinter MM. Recurrent Pericarditis: a Stubborn Opponent Meets New Treatments in 2022. Curr Cardiol Rep 2022; 24:915-923. [PMID: 35612721 PMCID: PMC9130990 DOI: 10.1007/s11886-022-01719-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/11/2022] [Indexed: 11/29/2022]
Abstract
Purpose of Review Our goal in writing this review was to provide a comprehensive appraisal of current therapies for idiopathic recurrent pericarditis with a particular focus on the newest therapeutic agents. We sought to understand the role of the inflammasome in the pathophysiology of pericarditis and how it informs the use of interleukin-1 (IL-1)-directed therapies. Recent Findings The latest research on this topic has focused on the critical role of the NLRP3 (NACHT, leucine-rich repeat, and pyrin domain-containing protein) inflammasome. Very recently, components of the NLRP3 inflammasome were detected by immune staining in pericardial tissue from patients with recurrent idiopathic pericarditis. In a mouse model of pericarditis, anti-IL-1 agents anakinra and rilonacept reduced NLRP3 immunostaining. Subsequent study of these drugs in human subjects with idiopathic recurrent pericarditis demonstrated their efficacy. Summary Recurrent idiopathic pericarditis, while relatively rare, poses a continued treatment challenge and contributes to a diminished quality of life for those patients who are afflicted. Recent developments, including an animal model of the disease and the use of IL-1-directed therapies, represent an exciting leap forward in our understanding of treatment targets. These advances offer not only new tools in our fight against this disease, but also the promise of earlier intervention and attenuation of disease morbidity. As our experience with these new agents expands, we can address questions about the ideal timing of introduction of anti-IL-1 therapy and duration of therapy and better understand the potential side effect profile.
Supplementary Information The online version contains supplementary material available at 10.1007/s11886-022-01719-z.
Collapse
Affiliation(s)
- Tracy Hagerty
- Cardiology Unit, University of Vermont Medical Center, 111 Colchester Avenue, Burlington, VT 05401 USA
- Larner College of Medicine, University of Vermont, Burlington, VT USA
| | - Matthew A. Kluge
- Cardiology Unit, University of Vermont Medical Center, 111 Colchester Avenue, Burlington, VT 05401 USA
| | - Martin M. LeWinter
- Cardiology Unit, University of Vermont Medical Center, 111 Colchester Avenue, Burlington, VT 05401 USA
- Larner College of Medicine, University of Vermont, Burlington, VT USA
| |
Collapse
|
8
|
Walker M, Patel P, Kwon O, Koene RJ, Duprez DA, Kwon Y. Atrial Fibrillation and Hypertension: "Quo Vadis". Curr Hypertens Rev 2022; 18:39-53. [PMID: 35023459 DOI: 10.2174/1573402118666220112122403] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 10/29/2021] [Accepted: 12/24/2021] [Indexed: 11/22/2022]
Abstract
Hypertension is one of the most well-established risk factors for atrial fibrillation. Long-standing untreated hypertension leads to structural remodeling and electrophysiologic alterations causing an atrial myopathy that forms a vulnerable substrate for the development and maintenance of atrial fibrillation. Hypertension-induced hemodynamic, inflammatory, hormonal, and autonomic changes all appear to be important contributing factors. Furthermore, hypertension is also associated with several atrial fibrillation-related comorbidities. As such, hypertension may represent an important target for therapy in atrial fibrillation. Clinicians should be aware of pitfalls of the blood pressure measurement in atrial fibrillation. While the auscultatory method is preferred, the use of automated devices appears to be an acceptable method in the ambulatory setting. There are pathophysiologic bases and emerging clinical evidence suggesting the benefit of renin-angiotensin system inhibition in risk reduction of atrial fibrillation development particularly in patients with left ventricular hypertrophy or left ventricular dysfunction. A better understanding of hypertension's pathophysiologic link to atrial fibrillation may lead to the development of novel therapies for the primary prevention of atrial fibrillation. Finally, future studies are needed to address optimal blood pressure goal to minimize the risk of atrial fibrillation-related complications.
Collapse
Affiliation(s)
- McCall Walker
- Division of Cardiology, Department of Medicine, University of Texas Southwestern, Dallas, USA
| | - Paras Patel
- Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, USA
| | - Osung Kwon
- Division of Cardiology, Department of Internal Medicine, Asan Medical Center, University of Uslan College of Medicine, Seoul, Korea
| | - Ryan J Koene
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, Cleveland, USA
| | - Daniel A Duprez
- Department of Medicine, University of Minnesota, Minneapolis, USA
| | - Younghoon Kwon
- Division of Cardiology, Department of Medicine, University of Washington, Seattle, WA, USA
- Division of Cardiology, Department of Medicine, University of Virginia, Charlottesville, USA
- Department of Medicine, University of Minnesota, Minneapolis, USA
| |
Collapse
|
9
|
Vahdat-Lasemi F, Aghaee-Bakhtiari SH, Tasbandi A, Jaafari MR, Sahebkar A. Targeting interleukin-β by plant-derived natural products: Implications for the treatment of atherosclerotic cardiovascular disease. Phytother Res 2021; 35:5596-5622. [PMID: 34390063 DOI: 10.1002/ptr.7194] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 05/21/2021] [Accepted: 05/29/2021] [Indexed: 01/31/2023]
Abstract
Inflammation is the main contributing factor to atheroma formation in atherosclerosis. Interleukin-1 beta (IL-1β) is an inflammatory mediator found in endothelial cells and resident leukocytes. Canakinumab is a selective monoclonal antibody against IL-1β which attenuates inflammation and concurrently precipitates fatal infections and sepsis. Natural products derived from medicinal plants, herbal remedy and functional foods are widely used nowadays. Experimental and clinical trial evidence supports that some natural products such as curcumin, resveratrol, and quercetin have potential effects on IL-1β suppression. In this review, we tried to document findings that used medicinal plants and plant-based natural products for treating atherosclerosis and its related diseases through the suppression of IL-1β.
Collapse
Affiliation(s)
- Fatemeh Vahdat-Lasemi
- Department of Medical Biotechnology and Nanotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Aida Tasbandi
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Medicine, The University of Western Australia, Perth, Australia
| |
Collapse
|
10
|
Morris G, Bortolasci CC, Puri BK, Marx W, O'Neil A, Athan E, Walder K, Berk M, Olive L, Carvalho AF, Maes M. The cytokine storms of COVID-19, H1N1 influenza, CRS and MAS compared. Can one sized treatment fit all? Cytokine 2021; 144:155593. [PMID: 34074585 PMCID: PMC8149193 DOI: 10.1016/j.cyto.2021.155593] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/03/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023]
Abstract
An analysis of published data appertaining to the cytokine storms of COVID-19, H1N1 influenza, cytokine release syndrome (CRS), and macrophage activation syndrome (MAS) reveals many common immunological and biochemical abnormalities. These include evidence of a hyperactive coagulation system with elevated D-dimer and ferritin levels, disseminated intravascular coagulopathy (DIC) and microthrombi coupled with an activated and highly permeable vascular endothelium. Common immune abnormalities include progressive hypercytokinemia with elevated levels of TNF-α, interleukin (IL)-6, and IL-1β, proinflammatory chemokines, activated macrophages and increased levels of nuclear factor kappa beta (NFκB). Inflammasome activation and release of damage associated molecular patterns (DAMPs) is common to COVID-19, H1N1, and MAS but does not appear to be a feature of CRS. Elevated levels of IL-18 are detected in patients with COVID-19 and MAS but have not been reported in patients with H1N1 influenza and CRS. Elevated interferon-γ is common to H1N1, MAS, and CRS but levels of this molecule appear to be depressed in patients with COVID-19. CD4+ T, CD8+ and NK lymphocytes are involved in the pathophysiology of CRS, MAS, and possibly H1N1 but are reduced in number and dysfunctional in COVID-19. Additional elements underpinning the pathophysiology of cytokine storms include Inflammasome activity and DAMPs. Treatment with anakinra may theoretically offer an avenue to positively manipulate the range of biochemical and immune abnormalities reported in COVID-19 and thought to underpin the pathophysiology of cytokine storms beyond those manipulated via the use of, canakinumab, Jak inhibitors or tocilizumab. Thus, despite the relative success of tocilizumab in reducing mortality in COVID-19 patients already on dexamethasone and promising results with Baricitinib, the combination of anakinra in combination with dexamethasone offers the theoretical prospect of further improvements in patient survival. However, there is currently an absence of trial of evidence in favour or contravening this proposition. Accordingly, a large well powered blinded prospective randomised controlled trial (RCT) to test this hypothesis is recommended.
Collapse
Affiliation(s)
- Gerwyn Morris
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Chiara C Bortolasci
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | | | - Wolfgang Marx
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Adrienne O'Neil
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Melbourne School of Population and Global Health, Melbourne, Australi
| | - Eugene Athan
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Barwon Health, Geelong, Australia
| | - Ken Walder
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, Centre for Molecular and Medical Research, School of Medicine, Geelong, Australia
| | - Michael Berk
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Florey Institute for Neuroscience and Mental Health and the Department of Psychiatry, The University of Melbourne, Melbourne, Australia
| | - Lisa Olive
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Deakin University, School of Psychology, Geelong, Australia
| | - Andre F Carvalho
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, University of Toronto, Toronto, Canada, Centre for Addiction and Mental Health (CAMH), Toronto, Canada
| | - Michael Maes
- Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Department of Psychiatry, King Chulalongkorn University Hospital, Bangkok, Thailand; Department of Psychiatry, Medical University of Plovdiv, Plovdiv, Bulgaria.
| |
Collapse
|
11
|
Opincariu D, Rodean I, Rat N, Hodas R, Benedek I, Benedek T. Systemic Vulnerability, as Expressed by I-CAM and MMP-9 at Presentation, Predicts One Year Outcomes in Patients with Acute Myocardial Infarction-Insights from the VIP Clinical Study. J Clin Med 2021; 10:jcm10153435. [PMID: 34362217 PMCID: PMC8347806 DOI: 10.3390/jcm10153435] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/18/2022] Open
Abstract
(1) Background: The prediction of recurrent events after acute myocardial infarction (AMI) does not sufficiently integrate systemic inflammation, coronary morphology or ventricular function in prediction algorithms. We aimed to evaluate the accuracy of inflammatory biomarkers, in association with angiographical and echocardiographic parameters, in predicting 1-year MACE after revascularized AMI. (2) Methods: This is an extension of a biomarker sub-study of the VIP trial (NCT03606330), in which 225 AMI patients underwent analysis of systemic vulnerability and were followed for 1 year. Hs-CRP, MMP-9, IL-6, I-CAM, V-CAM and E-selectin were determined at 1 h after revascularization. The primary end-point was the 1-year MACE rate. (3) Results: The MACE rate was 24.8% (n = 56). There were no significant differences between groups in regard to IL-6, V-CAM and E-selectin. The following inflammatory markers were significantly higher in MACE patients: hs-CRP (11.1 ± 13.8 vs. 5.1 ± 4.4 mg/L, p = 0.03), I-CAM (452 ± 283 vs. 220.5 ± 104.6, p = 0.0003) and MMP-9 (2255 ± 1226 vs. 1099 ± 706.1 ng/mL p = 0.0001). The most powerful predictor for MACE was MMP-9 of >1155 ng/mL (AUC-0.786, p < 0.001) even after adjustments for diabetes, LVEF, acute phase complications and other inflammatory biomarkers. For STEMI, the most powerful predictors for MACE included I-CAM > 239.7 ng/mL, V-CAM > 877.9 ng/mL and MMP-9 > 1393 ng/mL. (4) Conclusions: High levels of I-CAM and MMP-9 were the most powerful predictors for recurrent events after AMI for the overall study population. For STEMI subjects, the most important predictors included increased levels of I-CAM, V-CAM and MMP-9, while none of the analyzed parameters had proven to be predictive. Inflammatory biomarkers assayed during the acute phase of AMI presented a more powerful predictive capacity for MACE than the LVEF.
Collapse
Affiliation(s)
- Diana Opincariu
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
- Correspondence: or (D.O.); or (R.H.); Tel.: +40-756-787-587 (D.O.); +40-742-385-600 (R.H.)
| | - Ioana Rodean
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
| | - Nora Rat
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
| | - Roxana Hodas
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Correspondence: or (D.O.); or (R.H.); Tel.: +40-756-787-587 (D.O.); +40-742-385-600 (R.H.)
| | - Imre Benedek
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
| | - Theodora Benedek
- Department of Cardiology, University of Medicine, Pharmacy, Sciences and Technology “George Emil Palade”, 540142 Târgu Mureș, Romania; (I.R.); (N.R.); (I.B.); (T.B.)
- Cardiomed Medical Center, 22 December 1989 Street, No. 76, 540124 Târgu Mureș, Romania
| |
Collapse
|
12
|
Abstract
Billions of cells undergo apoptosis daily and are swiftly removed by macrophages through an evolutionarily conserved program termed "efferocytosis". Consequently, macromolecules within an apoptotic cell significantly burden a phagocyte with nutrients, such as lipids, oligonucleotides, and amino acids. In response to this nutrient overload, metabolic reprogramming must occur for the process of efferocytosis to remain non-phlogistic and to execute successive rounds of efferocytosis. The inability to undergo metabolic reprogramming after efferocytosis drives inflammation and impairs its resolution, often promoting many chronic inflammatory diseases. This is particularly evident for atherosclerosis, as metabolic reprogramming alters macrophage function in every stage of atherosclerosis, from the early formation of benign lesions to the progression of clinically relevant atheromas and during atherosclerosis regression upon aggressive lipid-lowering. This Review focuses on the metabolic pathways utilized upon apoptotic cell ingestion, the consequences of these metabolic pathways in macrophage function thereafter, and the role of metabolic reprogramming during atherosclerosis. Due to the growing interest in this new field, I introduce a new term, "efferotabolism", as a means to define the process by which macrophages break down, metabolize, and respond to AC-derived macromolecules. Understanding these aspects of efferotabolism will shed light on novel strategies to combat atherosclerosis and compromised inflammation resolution.
Collapse
|
13
|
Cherney DZ, Kanbay M, Lovshin JA. Renal physiology of glucose handling and therapeutic implications. Nephrol Dial Transplant 2020; 35:i3-i12. [PMID: 32003835 PMCID: PMC6993194 DOI: 10.1093/ndt/gfz230] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Indexed: 02/06/2023] Open
Abstract
The rationale for using sodium-glucose cotransporter 2 (SGLT2) inhibitors in patients with type 2 diabetes (T2D) has evolved over the last decade. Due to the effects on glucosuria and body weight loss, SGLT2 inhibitors were originally approved for glycemic control in T2D. Since glucosuria is attenuated in chronic kidney disease (CKD) Stages 3–5, initial regulatory approval for SGLT2 inhibitor use was limited to patients with T2D and preserved estimated glomerular filtration rate. Over time, however, it has become increasingly apparent that these therapies have a variety of important pharmacodynamic and clinical effects beyond glycemic lowering, including antihypertensive and antialbuminuric properties, and the ability to reduce glomerular hypertension. Importantly, these sodium-related effects are preserved across CKD stages, despite attenuated glycemic effects, which are lost at CKD Stage 4. With the completion of cardiovascular (CV) outcome safety trials—EMPA-REG OUTCOME, CANVAS Program and DECLARE TIMI-58—in addition to reductions in CV events, SGLT2 inhibition consistently reduces hard renal endpoints. Importantly, these CV and renal effects are independent of glycemic control. Subsequent data from the recent CREDENCE trial—the first dedicated renal protection trial with SGLT-2 inhibition—demonstrated renal and CV benefits in albuminuric T2D patients, pivotal results that have expanded the clinical importance of these therapies. Ongoing trials will ultimately determine whether SGLT2 inhibition will have a role in renal protection in other clinical settings, including nondiabetic CKD and type 1 diabetes.
Collapse
Affiliation(s)
- David Z Cherney
- Toronto General Hospital Research Institute, UHN, Toronto, ON, Canada.,Department of Physiology and Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada.,Department of Medicine, Division of Nephrology, University of Toronto, Toronto, ON, Canada.,Banting and Best Diabetes Centre, Toronto, ON, Canada
| | - Mehmet Kanbay
- Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey
| | - Julie A Lovshin
- Toronto General Hospital Research Institute, UHN, Toronto, ON, Canada.,Banting and Best Diabetes Centre, Toronto, ON, Canada.,Department of Medicine, Division of Endocrinology, Sunnybrook Health Sciences Centre, University of Toronto, Toronot, ON, Canada.,Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| |
Collapse
|
14
|
Yuan S, Carter P, Bruzelius M, Vithayathil M, Kar S, Mason AM, Lin A, Burgess S, Larsson SC. Effects of tumour necrosis factor on cardiovascular disease and cancer: A two-sample Mendelian randomization study. EBioMedicine 2020; 59:102956. [PMID: 32805626 PMCID: PMC7452586 DOI: 10.1016/j.ebiom.2020.102956] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 07/26/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Tumour necrosis factor (TNF) inhibitors are used in the treatment of certain autoimmune diseases but given the role of TNF in tumour biology and atherosclerosis, such therapies may influence the risk of cancer and cardiovascular disease. We conducted a Mendelian randomization study to explore whether TNF levels are causally related to cardiovascular disease and cancer. METHODS Single-nucleotide polymorphisms associated with TNF levels at genome-wide significance were identified from a genome-wide association study of 30 912 European-ancestry individuals. Three TNF-associated single-nucleotide polymorphisms associated with higher risk of autoimmune diseases were used as instrumental variables. Summary-level data for 14 cardiovascular diseases, overall cancer and 14 site-specific cancers were obtained from UK Biobank and consortia. FINDINGS Genetically-predicted TNF levels were positively associated with coronary artery disease (odds ratio (OR) 2.25; 95% confidence interval (CI) 1.50, 3.37) and ischaemic stroke (OR 2.27; 95% CI 1.50, 3.43), and inversely associated with overall cancer (OR 0.54; 95% CI 0.42, 0.69), breast cancer (OR 0.51; 95% CI 0.39, 0.67), and colorectal cancer (OR 0.20; 95% CI 0.09, 0.45). There were suggestive associations of TNF with venous thromboembolism (OR 2.18; 95% CI 1.32, 3.59), endometrial cancer (OR 0.25; 95% CI 0.07, 0.94), and lung cancer (OR 0.45; 95% CI 0.21, 0.94). INTERPRETATION This study found evidence of causal associations of increased TNF levels with higher risk of common cardiovascular diseases and lower risk of overall and certain cancers.
Collapse
Affiliation(s)
- Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm 17177, Sweden; Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Paul Carter
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Maria Bruzelius
- Coagulation Unit, Department of Hematology, Karolinska University Hospital, Stockholm, Sweden; Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | | | - Siddhartha Kar
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Amy M Mason
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom; National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge and Cambridge University Hospitals, Cambridge, United Kingdom
| | - Ang Lin
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stephen Burgess
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom; MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm 17177, Sweden; Department of Surgical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
15
|
Liwinski T, Zheng D, Elinav E. The microbiome and cytosolic innate immune receptors. Immunol Rev 2020; 297:207-224. [PMID: 32658330 DOI: 10.1111/imr.12901] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/09/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023]
Abstract
The discovery of innate immune sensors (pattern recognition receptors, PRRs) has profoundly transformed the notion of innate immunity, in providing a mechanistic basis for host immune interactions with a wealth of environmental signals, leading to a variety of immune-mediated outcomes including instruction and activation of the adaptive immune arm. As part of this growing understanding of host-environmental cross talk, an intimate connection has been unveiled between innate immune sensors and signals perceived from the commensal microbiota, which may be regarded as a hub integrating a variety of environmental cues. Among cytosolic PRRs impacting on host homeostasis by interacting with the commensal microbiota are nucleotide-binding domain, leucine-rich repeat-containing protein receptors (NLRs), together with a number of cytosolic DNA sensors and the family of absent in melanoma (AIM)-like receptors (ALRs). NLR sensors have been a particular focus of research, and some NLRs have emerged as key orchestrators of inflammatory responses and host homeostasis. Some NLRs achieve this through the formation of cytoplasmic multiprotein complexes termed inflammasomes. More recently discovered PRRs include retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), cyclic GMP-AMP synthase (cGAS), and STING. In the present review, they summarize recent advancements in knowledge on structure and function of cytosolic PRRs and their roles in host-microbiota cross talk and immune surveillance. In addition, we discuss their relevance for human health and disease and future therapeutic applications involving modulation of their activation and signaling.
Collapse
Affiliation(s)
- Timur Liwinski
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel.,1st Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Danping Zheng
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel.,Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, Israel.,Cancer-Microbiome Division Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| |
Collapse
|
16
|
Huang S, Che J, Chu Q, Zhang P. The Role of NLRP3 Inflammasome in Radiation-Induced Cardiovascular Injury. Front Cell Dev Biol 2020; 8:140. [PMID: 32226786 PMCID: PMC7080656 DOI: 10.3389/fcell.2020.00140] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/19/2020] [Indexed: 12/24/2022] Open
Abstract
The increasing risk of long-term adverse effects from radiotherapy on the cardiovascular structure is receiving increasing attention. However, the mechanisms underlying this increased risk remain poorly understood. Recently, the nucleotide-binding domain and leucine-rich-repeat-containing family pyrin 3 (NLRP3) inflammasome was suggested to play a critical role in radiation-induced cardiovascular injury. However, the relationship between ionizing radiation and the NLRP3 inflammasome in acute and chronic inflammation is complex. We reviewed literature detailing pathological changes and molecular mechanisms associated with radiation-induced damage to the cardiovascular structure, with a specific focus on NLRP3 inflammasome-related cardiovascular diseases. We also summarized possible therapeutic strategies for the prevention of radiation-induced heart disease (RIHD).
Collapse
Affiliation(s)
- Shanshan Huang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Che
- College of Life Sciences, Wuhan University, Wuhan, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
17
|
Circular RNA circ-RELL1 regulates inflammatory response by miR-6873-3p/MyD88/NF-κB axis in endothelial cells. Biochem Biophys Res Commun 2020; 525:512-519. [PMID: 32113679 DOI: 10.1016/j.bbrc.2020.02.109] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/16/2020] [Indexed: 11/23/2022]
Abstract
Endothelial inflammation is an important contributor to the pathology of atherosclerotic cardiovascular disease (ASCVD). Circular RNAs (circRNAs) function and role in endothelium inflammation still unknown. In our present study, we firstly identified that circ-RELL1 plays a proinflammatory role in ox-LDL-induced HUVECs through high-throughput circRNA microarray assays. Knockdown circ-RELL1 can reduce the expression of ICAM1 and VCAM1 in ox-LDL induced endothelium inflammation. Mechanistically, circ-RELL1 directly bound to miR-6873-3p in cytoplasm. Subsequently miR-6873-3p reduced MyD88 (myeloid differentiation primary response 88) protein expression and alleviated MyD88 medicated NF-κB activation. Furthermore, circ-RELL1 can abolish the inhibition of inflammation response by miR-6873-3p. Our findings illustrate a novel regulatory pathway that circ-RELL1 modulate inflammatory response by miR-6873-3p/MyD88/NF-κB axis in ox-LDL induced endothelial cells, which provides a potential therapeutic candidate for endothelium inflammation in atherosclerotic cardiovascular disease.
Collapse
|
18
|
Flanagan TW, Sebastian MN, Battaglia DM, Foster TP, Maillet EL, Nichols CD. Activation of 5-HT 2 Receptors Reduces Inflammation in Vascular Tissue and Cholesterol Levels in High-Fat Diet-Fed Apolipoprotein E Knockout Mice. Sci Rep 2019; 9:13444. [PMID: 31530895 PMCID: PMC6748996 DOI: 10.1038/s41598-019-49987-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 08/25/2019] [Indexed: 01/14/2023] Open
Abstract
Coronary artery disease (CAD) is a progressive cardiovascular syndrome characterized by cholesterol-induced focal arterial lesions that impair oxygen delivery to the heart. As both innate and adaptive immune cells play critical roles in the formation and progression of arterial plaques and endothelial cell dysfunction, CAD is commonly viewed as a chronic inflammatory disorder. Our lab has previously discovered that 5-HT2A receptor activation with the 5-HT2 receptor selective agonist (R)-2,5-dimethoxy-4-iodoamphetamine [(R)-DOI] has potent anti-inflammatory activity in both cell culture and whole animal models. Here we have examined the putative therapeutic effects of (R)-DOI in the ApoE−/− high fat model of cardiovascular disease. Subcutaneously implanted osmotic minipumps were used to infuse sustained low rates (0.15 μg / hr) of (R)-DOI∙HCl to mice fed a high-fat “Western” diet. (R)-DOI treated mice had significant reductions in expression levels of mRNA for inflammatory markers like Il6 in vascular tissue, normalized glucose homeostasis, and reduced circulating cholesterol levels. As cardiovascular disease is a leading cause of death both globally and in the Western world, activation of 5-HT2A receptors at sub-behavioral levels may represent a new strategy to treat inflammation-based cardiovascular disease.
Collapse
Affiliation(s)
- Thomas W Flanagan
- Department of Pharmacology and Experimental Therapeutics Louisiana State University Health Sciences Center 1901 Perdido St, New Orleans, LA, 70112, USA
| | - Melaine N Sebastian
- Department of Pharmacology and Experimental Therapeutics Louisiana State University Health Sciences Center 1901 Perdido St, New Orleans, LA, 70112, USA
| | - Diana M Battaglia
- Department of Microbiology, Immunology, and Parasitology Louisiana State University Health Sciences Center 1901 Perdido St, New Orleans, LA, 70112, USA
| | - Timothy P Foster
- Department of Microbiology, Immunology, and Parasitology Louisiana State University Health Sciences Center 1901 Perdido St, New Orleans, LA, 70112, USA
| | - Emeline L Maillet
- Eleusis Benefit Corporation 11 East 44th St., Suite 104, New York, NY, 10017, USA
| | - Charles D Nichols
- Department of Pharmacology and Experimental Therapeutics Louisiana State University Health Sciences Center 1901 Perdido St, New Orleans, LA, 70112, USA.
| |
Collapse
|
19
|
Erhart P, Cakmak S, Grond-Ginsbach C, Hakimi M, Böckler D, Dihlmann S. Inflammasome activity in leucocytes decreases with abdominal aortic aneurysm progression. Int J Mol Med 2019; 44:1299-1308. [PMID: 31432101 PMCID: PMC6713432 DOI: 10.3892/ijmm.2019.4307] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/19/2019] [Indexed: 12/11/2022] Open
Abstract
Abdominal aortic aneurysms (AAAs) are characterized by chronic inflammatory cell infiltration. The present extended immunohistochemistry study aimed to characterize inflammation in AAA and aortic control samples. In specific, the composition of the infiltrating immune cells and the expression of five inflammasome components in these immune cells were evaluated, in order to characterize their role in AAA development. A total of 104 biopsies from 48 AAA patients and 40 healthy specimens from organ donors were evaluated for their grade of inflammation. Infiltrating leukocytes were characterized by specific markers (CD3, CD20 and CD68), intramural localization and inflammasome protein expression [NLR family pyrin domain containing 3 (NLRP3), absent in melanoma 2 (AIM2), apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), Caspase-1 and Caspase-5]. Macrophages, B and T lymphocytes were detected to a similar extent in grade 1, 2 and 3 AAA specimens, whereas in control samples, B and T lymphocytes were rarely observed in grade 1 lesions. Expression frequencies of NLRP3, AIM2 and Caspase-5 were significantly higher in grade 1 lesions of AAA samples compared with grade 1 lesions in control samples. Finally, AIM2, ASC, and Caspase-5 displayed significantly lower expression frequencies in grade 3 compared with grade 2 AAA specimens, and all inflammasome components were less frequently detected in grade 3 than in grade 1 lesions of AAA. This indicates that inflammasome activities decrease with AAA progression in infiltrating leukocytes. No statistically significant association was found for grade 2 and grade 3 lesions and total leukocyte count, C-reactive protein levels, maximal aortic diameter, plasma cholesterol level or biomechanical parameters (derived from finite element analysis) of the respective patients. Overall, the aortic wall of AAA contained lymphocytes and macrophages with different states of activity. The present data suggested that therapeutic inhibition of specific inflammasome components might counteract AAA development and progression.
Collapse
Affiliation(s)
- Philipp Erhart
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Sinan Cakmak
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | | | - Maani Hakimi
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Dittmar Böckler
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Susanne Dihlmann
- Department of Vascular and Endovascular Surgery, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| |
Collapse
|
20
|
Bomfim GF, Cau SBA, Bruno AS, Fedoce AG, Carneiro FS. Hypertension: a new treatment for an old disease? Targeting the immune system. Br J Pharmacol 2019; 176:2028-2048. [PMID: 29969833 PMCID: PMC6534786 DOI: 10.1111/bph.14436] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 06/14/2018] [Accepted: 06/24/2018] [Indexed: 12/22/2022] Open
Abstract
Arterial hypertension represents a serious public health problem, being a major cause of morbidity and mortality worldwide. The availability of many antihypertensive therapeutic strategies still fails to adequately treat around 20% of hypertensive patients, who are considered resistant to conventional treatment. In the pathogenesis of hypertension, immune system mechanisms are activated and both the innate and adaptive immune responses play a crucial role. However, what, when and how the immune system is triggered during hypertension development is still largely undefined. In this context, this review highlights scientific advances in the manipulation of the immune system in order to attenuate hypertension and end-organ damage. Here, we discuss the potential use of immunosuppressants and immunomodulators as pharmacological tools to control the activation of the immune system, by non-specific and specific mechanisms, to treat hypertension and improve end-organ damage. Nevertheless, more clinical trials should be performed with these drugs to establish their therapeutic efficacy, safety and risk-benefit ratio in hypertensive conditions. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
| | - Stefany Bruno Assis Cau
- Department of Pharmacology, Institute of Biological ScienceFederal University of Minas GeraisBelo HorizonteMGBrazil
| | - Alexandre Santos Bruno
- Department of Pharmacology, Institute of Biological ScienceFederal University of Minas GeraisBelo HorizonteMGBrazil
| | - Aline Garcia Fedoce
- Department of Pharmacology, Ribeirão Preto Medical SchoolUniversity of São PauloSão PauloBrazil
| | - Fernando S Carneiro
- Department of Pharmacology, Ribeirão Preto Medical SchoolUniversity of São PauloSão PauloBrazil
| |
Collapse
|
21
|
Meeusen JW, Donato LJ, Bryant SC, Baudhuin LM, Berger PB, Jaffe AS. Plasma Ceramides. Arterioscler Thromb Vasc Biol 2018; 38:1933-1939. [DOI: 10.1161/atvbaha.118.311199] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Ceramides are sphingolipids involved with cellular signaling. Synthesis of ceramides occurs in all tissues. Ceramides accumulate within tissues and the blood plasma during metabolic dysfunction, dyslipidemia, and inflammation. Elevations of ceramides are predictive of cardiovascular mortality. We sought to verify the utility of plasma concentrations of 4 ceramides: N-palmitoyl-sphingosine [Cer(16:0)], N-stearoyl-sphingosine [Cer(18:0)], N-nervonoyl-sphingosine [Cer(24:1)], and N-lignoceroyl-sphingosine [Cer(24:0)] in predicting major adverse cardiovascular events in a diverse patient population referred for coronary angiography.
Approach and Results—
Plasma ceramides were measured in 495 participants before nonurgent coronary angiography. Coronary artery disease, defined as >50% stenosis in ≥1 coronary artery, was identified 265 (54%) cases. Ceramides were not significantly associated with coronary artery disease. Patients were followed for a combined primary end point of myocardial infarction, percutaneous intervention, coronary artery bypass, stroke, or death within 4 years. Ceramides were significantly predictive of outcomes after adjusting for age, sex, body mass index, hypertension, smoking, LDL (low-density lipoprotein) cholesterol, HDL (high-density lipoprotein) cholesterol, triglycerides, serum glucose, and family history of coronary artery disease. The fully adjusted per SD hazard ratios (95% confidence interval) were 1.50 (1.16–1.93) for Cer(16:0), 1.42 (1.11–1.83) for Cer(18:0), 1.43 (1.08–1.89) for Cer(24:1), and 1.58 (1.22–2.04) for the ceramide risk score.
Conclusions—
Elevated plasma concentrations of ceramides are independently associated with major adverse cardiovascular events in patients with and without coronary artery disease.
Collapse
Affiliation(s)
- Jeffrey W. Meeusen
- From the Department of Laboratory Medicine and Pathology (J.W.M., L.J.D., L.M.B., A.S.J.)
| | - Leslie J. Donato
- From the Department of Laboratory Medicine and Pathology (J.W.M., L.J.D., L.M.B., A.S.J.)
| | | | - Linnea M. Baudhuin
- From the Department of Laboratory Medicine and Pathology (J.W.M., L.J.D., L.M.B., A.S.J.)
| | - Peter B. Berger
- Department of Cardiology (P.B.B., A.S.J.), Mayo Clinic, Rochester, MN
| | - Allan S. Jaffe
- From the Department of Laboratory Medicine and Pathology (J.W.M., L.J.D., L.M.B., A.S.J.)
- Department of Cardiology (P.B.B., A.S.J.), Mayo Clinic, Rochester, MN
| |
Collapse
|
22
|
Fernandez DM, Clemente JC, Giannarelli C. Physical Activity, Immune System, and the Microbiome in Cardiovascular Disease. Front Physiol 2018; 9:763. [PMID: 30013482 PMCID: PMC6036301 DOI: 10.3389/fphys.2018.00763] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/30/2018] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular health is a primary research focus, as it is a leading contributor to mortality and morbidity worldwide, and is prohibitively costly for healthcare. Atherosclerosis, the main driver of cardiovascular disease, is now recognized as an inflammatory disorder. Physical activity (PA) may have a more important role in cardiovascular health than previously expected. This review overviews the contribution of PA to cardiovascular health, the inflammatory role of atherosclerosis, and the emerging evidence of the microbiome as a regulator of inflammation.
Collapse
Affiliation(s)
- Dawn M. Fernandez
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jose C. Clemente
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Chiara Giannarelli
- Department of Medicine, Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|