1
|
Borim PA, Gatto M, Mota GAF, Meirelles ALB, dos Santos ACC, Pagan LU, Ojopi EPB, Rodrigues EA, Souza LM, Damatto FC, Oliveira LRDS, Zornoff LAM, Okoshi K, Okoshi MP. Nlrc4 Inflammasome Expression After Acute Myocardial Infarction in Rats. Int J Mol Sci 2025; 26:3697. [PMID: 40332346 PMCID: PMC12028149 DOI: 10.3390/ijms26083697] [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: 01/22/2025] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025] Open
Abstract
Acute myocardial necrosis activates the immune response and inflammatory processes. Although the initial response is helpful in restoring tissue injury, dysregulated and exacerbated inflammation contributes to the progression of cardiac remodeling. Inflammasomes play important roles in post-infarction inflammation. NALP1/NLRP1, NLRP 3, and NLRC4 are the best-known inflammasomes. NLRP3, which has received the most study in cardiovascular disease, has been linked to increased IL-1β (IL1B) production and caspase-1 activity, as well as impaired cardiac function. The role of NLRP1 and NLRC4 inflammasomes after acute myocardial infarction (MI) is poorly understood. We evaluated the expression of myocardial inflammasomes and inflammatory markers 72 h after MI in rats. Male Wistar rats were divided into Sham (n = 15) and MI (n = 16) groups. MI was induced by ligating the left anterior descending coronary artery. Infarct size was assessed by histology. Myocardial protein and gene expression was analyzed by Western blot and RT-qPCR, respectively. IL-1β (Il1b) concentrations in serum and heart macerate supernatant were evaluated by ELISA. Statistical analysis was performed using Student's t test. Rats with an MI size less than 30% of the total left ventricle (LV) area were excluded; infarct size was 46 ± 11% of the total LV area in MI. The interstitial collagen fraction was higher in MI. Nlrc4, caspase-1 (Casp1), and IL-1β (Il1b) protein expressions were higher in MI. Nlrp3, Nlrp1, ASC (Pycard), pro-caspase-1, and pro-IL-1β (Il1b) expressions did not differ between groups. Expression of the Nlrp3 and ASC (Pycard) genes, as well as myocardial and serum IL-1β (Il1b) concentrations, was higher in MI. Acute post-myocardial infarction inflammation is characterized by increased protein expression of Nlrc4, caspase-1, and interleukin-1β; increased gene expression of Nlrp3 and ASC (Pycard); and elevated serum and myocardial concentrations of interleukin-1β in combination with an increased myocardial collagen interstitial fraction.
Collapse
Affiliation(s)
- Patricia Aparecida Borim
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Mariana Gatto
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Gustavo Augusto Ferreira Mota
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Ana Luiza Barioni Meirelles
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Anna Clara Consorti dos Santos
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Luana Urbano Pagan
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Elida Paula Benquique Ojopi
- Clinic Hospital, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil;
| | - Eder Anderson Rodrigues
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Lidiane Moreira Souza
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Felipe Cesar Damatto
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Leiliane Rodrigues dos Santos Oliveira
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Leonardo Antonio Mamede Zornoff
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Katashi Okoshi
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| | - Marina Politi Okoshi
- Internal Medicine Department, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (P.A.B.); (M.G.); (G.A.F.M.); (A.L.B.M.); (A.C.C.d.S.); (L.U.P.); (E.A.R.); (L.M.S.); (F.C.D.); (L.R.d.S.O.); (L.A.M.Z.); (K.O.)
| |
Collapse
|
2
|
Issa F, Abdulla M, Retnowati FD, Al-Khawaga H, Alhiraky H, Al-Harbi KM, Al-Haidose A, Maayah ZH, Abdallah AM. Cardio-Rheumatic Diseases: Inflammasomes Behaving Badly. Int J Mol Sci 2025; 26:3520. [PMID: 40331999 PMCID: PMC12026794 DOI: 10.3390/ijms26083520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/28/2025] [Accepted: 03/28/2025] [Indexed: 05/08/2025] Open
Abstract
Cardio-rheumatology is an evolving and interdisciplinary field lying at the intersection of rheumatology and cardiovascular medicine that recognizes that individuals with autoimmune and inflammatory rheumatic complications have a much higher likelihood of developing cardiovascular diseases (CVDs). Inflammasomes are multiprotein complexes stimulated by the immune system after the detection of pathogens or cellular injury. Inflammasomes undergo a two-stage activation process initiated by nuclear factor (NF)-κB, subsequently playing a crucial role in innate immunity through activation of caspase 1 and the consequent release of proinflammatory cytokines such as IL-18 and IL-1β. However, a loss of control of inflammasome activation can cause inflammatory diseases in humans. Recent studies have focused on the role of inflammasomes in inflammatory cascades implicated in the pathogenesis of several diseases. Here, we review inflammasome activation, its mechanism of action, and its role in CVD. In particular, we describe the role of inflammasomes in rheumatic heart disease, Kawasaki disease, familial Mediterranean fever, ankylosing spondylitis, and rheumatoid arthritis as exemplars to illustrate pathobiological mechanisms and the potential for targeting inflammasomes for therapeutic benefit.
Collapse
Affiliation(s)
- Farah Issa
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Marah Abdulla
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Faizah D. Retnowati
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Huda Al-Khawaga
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Hanin Alhiraky
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Khalid M. Al-Harbi
- Department of Pediatric, College of Medicine, Taibah University, Madinah 41477, Saudi Arabia;
| | - Amal Al-Haidose
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| | - Zaid H. Maayah
- Department of Pharmaceutical Sciences, College of Pharmacy, QU Health Sector, Qatar University, Doha 2713, Qatar;
| | - Atiyeh M. Abdallah
- Department of Biomedical Sciences, College of Health Sciences, QU Health Sector, Qatar University, Doha 2713, Qatar; (F.I.); (M.A.); (F.D.R.); (H.A.-K.); (H.A.); (A.A.-H.)
| |
Collapse
|
3
|
Zhang Y, Guo Z, Lai R, Zou X, Ma L, Cai T, Huang J, Huang W, Zou B, Zhou J, Li J. Comprehensive Analysis Based on Genes Associated With Cuproptosis, Ferroptosis, and Pyroptosis for the Prediction of Diagnosis and Therapies in Coronary Artery Disease. Cardiovasc Ther 2025; 2025:9106621. [PMID: 40124544 PMCID: PMC11929595 DOI: 10.1155/cdr/9106621] [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] [Received: 02/20/2024] [Revised: 11/06/2024] [Accepted: 02/18/2025] [Indexed: 03/25/2025] Open
Abstract
Coronary artery disease (CAD) is a complex condition influenced by genetic factors, lifestyle, and other risk factors that contribute to increased mortality. This study is aimed at evaluating the diagnostic potential of genes associated with cuproptosis, ferroptosis, and pyroptosis (CFP) using network modularization and machine learning methods. CAD-related datasets GSE42148, GSE20680, and GSE20681 were sourced from the GEO database, and genes related to CFP genes were gathered from MsigDB and FerrDb datasets and literature. To identify diagnostic genes linked to these pathways, weighted gene coexpression network analysis (WGCNA) was used to isolate CAD-related modules. The diagnostic accuracy of key genes in these modules was then assessed using LASSO, SVM, and random forest models. Immunity and drug sensitivity correlation analyses were subsequently performed to investigate possible underlying mechanisms. The function of a potential gene, STK17B, was analyzed through western blot and transwell assays. Two CAD-related modules with strong correlations were identified and validated. The SVM model outperformed LASSO and random forest models, demonstrating superior discriminative power (AUC = 0.997 in the blue module and AUC = 1.000 in the turquoise module), with nine key genes identified: CTDSP2, DHRS7, NLRP1, MARCKS, PELI1, RILPL2, JUNB, STK17B, and SLC40A1. Knockdown of STK17B inhibited cell migration and invasion in human umbilical vein endothelial cells. In summary, our findings suggest that CFP genes hold potential as diagnostic biomarkers and therapeutic targets, with STK17B playing a role in CAD progression.
Collapse
Affiliation(s)
- Yongyi Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Zhehan Guo
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Renkui Lai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Xu Zou
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Liuling Ma
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Tianjin Cai
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Jingyi Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Wenxiang Huang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China
| | - Bingcheng Zou
- Schoole of Life Science, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jinming Zhou
- Schoole of Life Science, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jinxin Li
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, China
| |
Collapse
|
4
|
Dharmakumar R, Kloner RA, Fishbein M, Heusch G, Vora KP, Gropler R, Henry T, Shing-Chan F, Singh D, Jambunathan N, Subramanian R, Kreutz RP, Reed GW, Kovacs RJ, Fry E, Kalra A, Kumar A, Raman SV. Reperfused Myocardial Infarction: The Road to CCS Classification of Acute MI and Beyond. JACC. ADVANCES 2025; 4:101528. [PMID: 40021272 PMCID: PMC11905164 DOI: 10.1016/j.jacadv.2024.101528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 11/25/2024] [Indexed: 03/03/2025]
Abstract
The Canadian Cardiovascular Society recently put forth a new classification of acute reperfused myocardial infarction (MI) based on stages of myocardial injury. Backed by more than 5 decades of intense investigation in the field, the key message of this new classification is that not all MIs are the same and that the type and extent of myocardial injury should be considered in diagnosing and treating MI. We review the literature with the goal of highlighting the progressive advances that enabled the synthesis of the Canadian Cardiovascular Society classification into 4 distinct stages of tissue injury. We emphasize the major breakthroughs from insights gained from experimental, translational, and clinical studies to date. We also identify current gaps in knowledge and critical research directions that need to be pursued to improve patient care and reduce post-MI complications such as chronic heart failure and malignant arrhythmias, whose risk is linked to stage and extent of myocardial injury.
Collapse
Affiliation(s)
- Rohan Dharmakumar
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| | - Robert A Kloner
- Huntington Medical Research Institute, Pasadena, California, USA; Department of Medicine, Keck School of Medicine at University of Southern California, Los Angeles, California, USA
| | - Michael Fishbein
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Gerd Heusch
- Department of Pathophysiology, University of Essen, Essen, Germany
| | - Keyur P Vora
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert Gropler
- Department of Radiological Sciences, Washington University School of Medicine at St. Louis, St. Louis, Missouri, USA
| | - Timothy Henry
- Carl and Edyth Lindner Center for Research, The Christ Hospital, Cincinnati, Ohio, USA
| | - Fai Shing-Chan
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Dhirendra Singh
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Nithya Jambunathan
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Ramesh Subramanian
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rolf P Kreutz
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Grant W Reed
- Division of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Richard J Kovacs
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Edward Fry
- Ascension St. Vincent Heart Center, Indianapolis, Indiana, USA
| | - Ankur Kalra
- Krannert Cardiovascular Research Center, Indiana University School of Medicine, Indianapolis, Indiana, USA; Department of Cardiology, Franciscan Health, Lafayette, Indiana, USA
| | - Andreas Kumar
- Clinical Sciences Division, Northern Ontario School of Medicine, University and Health Sciences, Sudbury, Ontario, Canada
| | - Subha V Raman
- Heart and Vascular Division, OhioHealth, Columbus, Ohio, USA
| |
Collapse
|
5
|
Wang W, Wang XM, Zhang HL, Zhao R, Wang Y, Zhang HL, Song ZJ. Molecular and metabolic landscape of adenosine triphosphate-induced cell death in cardiovascular disease. World J Cardiol 2024; 16:689-706. [PMID: 39734818 PMCID: PMC11669974 DOI: 10.4330/wjc.v16.i12.689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/04/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
The maintenance of intracellular and extracellular adenosine triphosphate (ATP) levels plays a pivotal role in cardiac function. In recent years, burgeoning attention has been directed towards ATP-induced cell death (AICD), revealing it as a distinct cellular demise pathway triggered by heightened extracellular ATP concentrations, distinguishing it from other forms of cell death such as apoptosis and necrosis. AICD is increasingly acknowledged as a critical mechanism mediating the pathogenesis and progression of various cardiovascular maladies, encompassing myocardial ischemia-reperfusion injury, sepsis-induced cardiomyopathy, hypertrophic cardiomyopathy, arrhythmia, and diabetic cardiomyopathy. Consequently, a comprehensive understanding of the molecular and metabolic underpinnings of AICD in cardiac tissue holds promise for the prevention and amelioration of cardiovascular diseases. This review first elucidates the vital physiological roles of ATP in the cardiovascular system, subsequently delving into the intricate molecular mechanisms and metabolic signatures governing AICD. Furthermore, it addresses the potential therapeutic targets implicated in mitigating AICD for treating cardiovascular diseases, while also delineating the current constraints and future avenues for these innovative therapeutic targets, thereby furnishing novel insights and strategies for the prevention and management of cardiovascular disorders.
Collapse
Affiliation(s)
- Wei Wang
- College of Acupuncture-Moxibustion and Tuina, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Xue-Mei Wang
- College of Pharmacy, Gansu University of Chinese Medicine, Lanzhou 73000, Gansu Province, China
| | - Hao-Long Zhang
- University Sains Malaysia, Advanced Medical and Dental Institute, Penang 13200, Malaysia
| | - Rui Zhao
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Yong Wang
- Department of Pathology Center, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Hao-Ling Zhang
- Department of Biomedical Science, Advanced Medical and Dental Institute, University Sains Malaysia, Penang 13200, Malaysia
| | - Zhi-Jing Song
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|
6
|
Li F, Zhang Y, Wang Y, Cai X, Fan X. Cytokine Gene Variants as Predisposing Factors for the Development and Progression of Coronary Artery Disease: A Systematic Review. Biomolecules 2024; 14:1631. [PMID: 39766338 PMCID: PMC11726869 DOI: 10.3390/biom14121631] [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: 11/15/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025] Open
Abstract
Coronary artery disease (CAD) is the most prevalent form of cardiovascular disease. A growing body of research shows that interleukins (ILs), such as IL-8, IL-18 and IL-16, elicit pro-inflammatory responses and may play critical roles in the pathologic process of CAD. Single nucleotide polymorphisms (SNPs), capable of generating functional modifications in IL genes, appear to be associated with CAD risk. This study aims to evaluate the associations of ten previously identified SNPs of the three cytokines with susceptibility to or protection of CAD. A systematic review and meta-analysis were conducted using Pubmed, EMBASE, WOS, CENTRAL, CNKI, CBM, Weipu, WANFANG Data and Google Scholar databases for relevant literature published up to September 2024. Odds ratios (ORs) with 95% confidence intervals (CIs) were calculated for the four genetic models of the investigated SNPs in overall and subgroups analyses. Thirty-eight articles from 16 countries involving 14574 cases and 13001 controls were included. The present meta-analysis revealed no significant association between CAD and IL-8-rs2227306 or five IL-16 SNPs (rs8034928, rs3848180, rs1131445, rs4778889 and rs11556218). However, IL-8-rs4073 was significantly associated with an increased risk of CAD across all genetic models. In contrast, three IL-18 (rs187238, rs1946518 and rs1946519) variants containing minor alleles were associated with decreased risks of CAD under all models. Subgroups analyses by ethnicity indicated that IL-8-rs4073 conferred a significantly higher risk of CAD among Asians, including East, South and West Asians (allelic OR = 1.46, homozygous OR = 1.96, heterozygous OR = 1.47, dominant OR = 1.65), while it showed an inversely significant association with CAD risk in Caucasians (homozygous OR = 0.82, dominant OR = 0.85). Additionally, IL-18-rs187238 and IL-18-rs1946518 were significantly associated with reduced CAD risks in East Asians (for rs187238: allelic OR = 0.72, homozygous OR = 0.33, heterozygous OR = 0.73, dominant OR = 0.71; for rs1946518: allelic OR = 0.62, homozygous OR = 0.38, heterozygous OR = 0.49, dominant OR = 0.45). IL-18-rs187238 also demonstrated protective effects in Middle Eastern populations (allelic OR = 0.76, homozygous OR = 0.63, heterozygous OR = 0.72, dominant OR = 0.71). No significant associations were observed in South Asians or Caucasians for these IL-18 SNPs. Consistent with the overall analysis results, subgroups analyses further highlighted a significant association between IL-8-rs4073 and increased risk of acute coronary syndrome (heterozygous OR = 0.72). IL-18-rs187238 was significantly associated with decreased risks of myocardial infarction (MI) (allelic OR = 0.81, homozygous OR = 0.55, dominant OR = 0.80) and multiple vessel stenosis (allelic OR = 0.54, heterozygous OR = 0.45, dominant OR = 0.45). Similarly, IL-18-rs1946518 was significantly associated with reduced MI risk (allelic OR = 0.75, heterozygous OR = 0.68). These findings support the role of cytokine gene IL-8 and IL-18 variants as predisposing factors for the development and progression of CAD.
Collapse
Affiliation(s)
- Fang Li
- Correspondence: (F.L.); (X.F.); Tel.: +86-731-88872780 (F.L. & X.F.)
| | | | | | | | - Xiongwei Fan
- The Laboratory of Heart Development Research, College of Life Sciences, Hunan Normal University, Changsha 410081, China; (Y.Z.); (Y.W.); (X.C.)
| |
Collapse
|
7
|
Abdelmonaem AA, Abdel-Aziz AM, Ibrahim YF, Abdelzaher WY, Amgad Mohammed N, Marey H, S Taghian A, Setouhi A, Radi A, Ahmed SM. Cardioprotective effect of tofisopam against isoprenaline-induced myocardial infarction in rats via modulation of NLRP3\IL-1β\caspase-1 pathway. Immunopharmacol Immunotoxicol 2024; 46:902-911. [PMID: 39448368 DOI: 10.1080/08923973.2024.2421528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/20/2024] [Indexed: 10/26/2024]
Abstract
PURPOSE Cardiovascular diseases (CVDs) are a leading cause of morbidity and mortality worldwide. Ischemic heart diseases, particularly acute myocardial infarction (MI), represent the most common cause of death. MI is influenced by multiple factors, including the release of inflammatory mediators. A significant percentage of individuals with CVD experience psychological effects, such as anxiety and depression, which are linked to an increased risk of coronary heart disease. Certain anti-anxiety medications have demonstrated immunomodulatory and anti-inflammatory effects. Tofisopam, a 2,3-benzodiazepine with anxiolytic properties, has been shown to exert in vitro anti-inflammatory and immunomodulatory effects. The present study investigates the potential of tofisopam as a protective adjuvant against isoprenaline-induced MI in rats and explores the possible underlying mechanisms. METHODS The study included four groups: a control group, a group pretreated with tofisopam, an isoprenaline toxic group, and an isoprenaline toxic group pretreated with tofisopam. RESULTS The findings demonstrated that isoprenaline significantly increased cardiac enzyme levels, as well as elevated oxidative and inflammatory stress parameters, along with evident apoptosis in cardiac cells. In contrast, the tofisopam-pretreated group showed a significant reversal of the cardiac damage induced by isoprenaline. CONCLUSIONS Tofisopam protects against isoprenaline-induced MI through its antioxidant, anti-inflammatory, and anti-apoptotic properties.
Collapse
Affiliation(s)
| | | | - Yasmine F Ibrahim
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| | | | - Nada Amgad Mohammed
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Heba Marey
- Department of Biochemistry, Faculty of Medicine, Minia University, Minia, Egypt
| | - Asmaa S Taghian
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Amr Setouhi
- Department of Cardiology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Ashraf Radi
- Department of Cardiology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Sara M Ahmed
- Department of Pharmacology, Faculty of Medicine, Minia University, Minia, Egypt
| |
Collapse
|
8
|
Anderson MJ, den Hartigh AB, Loomis WP, Fink SL. Broad-spectrum inflammasome inhibition by thiomuscimol. Cell Death Discov 2024; 10:470. [PMID: 39550359 PMCID: PMC11569204 DOI: 10.1038/s41420-024-02238-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
Inflammasome formation, arising from pathogen or internal activating signals, is a key step in canonical pyroptosis, a gasdermin-mediated inflammatory cell death. Inhibition of pyroptosis has great clinical relevance due to its involvement in many different disease states. Current inhibitors of pyroptosis either only inhibit the final lytic step, which still allows inflammatory signal release, or only inhibit a single inflammasome, which does not account for inherent redundancy in activation of other inflammatory pathways. Here, we show that thiomuscimol, a structural analog of the lysis inhibitor muscimol, exhibits unique inhibitory activity upstream of plasma membrane rupture. We find that thiomuscimol inhibits inflammasome formation, as well as downstream caspase-1 activation, initiated by multiple pyroptotic signals, regardless of whether NLR recruitment of caspase-1 to the inflammasome relies on the ASC adapter protein. The ability of thiomuscimol to block multiple different inflammasomes opens the door for development of therapeutics with increased applications to broadly inhibit pyroptosis in multiple pathological settings.
Collapse
Affiliation(s)
- Marisa J Anderson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Andreas B den Hartigh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Wendy P Loomis
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Susan L Fink
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
9
|
Cai K, Jiang H, Zou Y, Song C, Cao K, Chen S, Wu Y, Zhang Z, Geng D, Zhang N, Liu B, Sun G, Tang M, Li Z, Zhang Y, Sun Y, Zhang Y. Programmed death of cardiomyocytes in cardiovascular disease and new therapeutic approaches. Pharmacol Res 2024; 206:107281. [PMID: 38942341 DOI: 10.1016/j.phrs.2024.107281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024]
Abstract
Cardiovascular diseases (CVDs) have a complex pathogenesis and pose a major threat to human health. Cardiomyocytes have a low regenerative capacity, and their death is a key factor in the morbidity and mortality of many CVDs. Cardiomyocyte death can be regulated by specific signaling pathways known as programmed cell death (PCD), including apoptosis, necroptosis, autophagy, pyroptosis, and ferroptosis, etc. Abnormalities in PCD can lead to the development of a variety of cardiovascular diseases, and there are also molecular-level interconnections between different PCD pathways under the same cardiovascular disease model. Currently, the link between programmed cell death in cardiomyocytes and cardiovascular disease is not fully understood. This review describes the molecular mechanisms of programmed death and the impact of cardiomyocyte death on cardiovascular disease development. Emphasis is placed on a summary of drugs and potential therapeutic approaches that can be used to treat cardiovascular disease by targeting and blocking programmed cell death in cardiomyocytes.
Collapse
Affiliation(s)
- Kexin Cai
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Haoyue Jiang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yuanming Zou
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Chunyu Song
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Kexin Cao
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Shuxian Chen
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Yanjiao Wu
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Zhaobo Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Danxi Geng
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China
| | - Naijin Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Reproductive and Genetic Medicine (China Medical University), National Health Commission, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China
| | - Bo Liu
- The first hospital of China Medical University, Department of cardiac surgery, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Guozhe Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Man Tang
- Department of clinical pharmacology, College of Pharmacy, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Zhao Li
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Yixiao Zhang
- Department of Urology Surgery, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Heping District, Shenyang, Liaoning 110004, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China; Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| | - Ying Zhang
- Department of Cardiology, the First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, Liaoning 110001, People's Republic of China; Institute of health sciences, China medical university, 77 Puhe Road, Shenbei New District, Shenyang, Liaoning 110001, People's Republic of China.
| |
Collapse
|
10
|
Peng J, Zhou B, Xu T, Hu X, Zhu Y, Wang Y, Pan S, Li W, Qian W, Zong J, Li F. The Serum NLRP1 Level and Coronary Artery Calcification: From Association to Development of a Risk-Prediction Nomogram. Rev Cardiovasc Med 2024; 25:265. [PMID: 39139411 PMCID: PMC11317339 DOI: 10.31083/j.rcm2507265] [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: 10/11/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 08/15/2024] Open
Abstract
Background To investigate the correlation between inflammasomes and coronary artery calcification (CAC), and develop and validating a nomogram for predicting the risk of CAC in patients with coronary artery disease (CAD). Methods A total of 626 patients with CAD at the Affiliated Hospital of Xuzhou Medical University were enrolled in this study. The patients were divided into the calcification group and the non-calcification group based on the assessment of coronary calcification. We constructed a training set and a validation set through random assignment. The least absolute shrinkage and selection operator (LASSO) regression and multivariate analysis were performed to identify independent risk factors of CAC in patients with CAD. Based on these independent predictors, we developed a web-based dynamic nomogram prediction model. The area under the receiver operating characteristic curve (AUC-ROC), calibration curves, and decision curve analysis (DCA) were used to evaluate this nomogram. Results Age, smoking, diabetes mellitus (DM), hyperlipidemia, the serum level of nucleotide-binding oligomerization domain (NOD)-like receptor protein 1 (NLRP1), alkaline phosphatase (ALP) and triglycerides (TG) were identified as independent risk factors of CAC. The AUC-ROC of the nomogram is 0.881 (95% confidence interval (CI): 0.850-0.912) in the training set and 0.825 (95% CI: 0.760-0.876) in the validation set, implying high discriminative ability. Satisfactory performance of this model was confirmed using calibration curves and DCA. Conclusions The serum NLRP1 level is an independent predictor of CAC. We established a web-based dynamic nomogram, providing a more accurate estimation and comprehensive perspective for predicting the risk of CAC in patients with CAD.
Collapse
Affiliation(s)
- Jingfeng Peng
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
| | - Bihan Zhou
- Department of Electrocardiography, The Affiliated Tumor Hospital of Nantong University, Nantong Tumor Hospital, 226000 Nantong, Jiangsu, China
| | - Tao Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
| | - Xiabing Hu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
| | - Yinghua Zhu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
| | - Yixiao Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
| | - Siyu Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
| | - Wenhua Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
| | - Wenhao Qian
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
| | - Jing Zong
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
| | - Fangfang Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 221000 Xuzhou, Jiangsu, China
| |
Collapse
|
11
|
Zheng Y, Chen Z, Yang J, Zheng J, Shui X, Yan Y, Huang S, Liang Z, Lei W, He Y. The Role of Hyperuricemia in Cardiac Diseases: Evidence, Controversies, and Therapeutic Strategies. Biomolecules 2024; 14:753. [PMID: 39062467 PMCID: PMC11274514 DOI: 10.3390/biom14070753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/10/2024] [Accepted: 06/11/2024] [Indexed: 07/28/2024] Open
Abstract
Hyperuricemia (HUA) may lead to myocardial cell damage, thereby promoting the occurrence and adverse outcomes of heart diseases. In this review, we discuss the latest clinical research progress, and explore the impact of HUA on myocardial damage-related diseases such as myocardial infarction, arrhythmias, and heart failure. We also combined recent findings from basic research to analyze potential mechanisms linking HUA with myocardial injury. In different pathological models (such as direct action of high uric acid on myocardial cells or combined with myocardial ischemia-reperfusion model), HUA may cause damage by activating the NOD-like receptor protein 3 inflammasome-induced inflammatory response, interfering with cardiac cell energy metabolism, affecting antioxidant defense systems, and stimulating reactive oxygen species production to enhance the oxidative stress response, ultimately resulting in decreased cardiac function. Additionally, we discuss the impact of lowering uric acid intervention therapy and potential safety issues that may arise. However, as the mechanism underlying HUA-induced myocardial injury is poorly defined, further research is warranted to aid in the development novel therapeutic strategies for HUA-related cardiovascular diseases.
Collapse
Affiliation(s)
- Yue Zheng
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zhirui Chen
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Jinya Yang
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Jing Zheng
- Department of Obstetrics and Gynecology, University of Wisconsin, Madison, WI 53715, USA;
| | - Xiaorong Shui
- Laboratory of Vascular Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China;
| | - Yiguang Yan
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (S.H.); (Z.L.)
| | - Shian Huang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (S.H.); (Z.L.)
| | - Zheng Liang
- Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (S.H.); (Z.L.)
| | - Wei Lei
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
- Precision Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Yuan He
- Guangdong Provincial Engineering Technology Research Center for Molecular Diagnosis and Innovative Drugs Translation of Cardiopulmonary Vascular Diseases, University Joint Laboratory of Guangdong Province and Macao Region on Molecular Targets and Intervention of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China; (Y.Z.); (Z.C.); (J.Y.); (Y.Y.)
- Laboratory of Cardiovascular Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| |
Collapse
|
12
|
Zheng H, Triplett KD, Prossnitz ER, Hall PR, Daly SM. G protein-coupled estrogen receptor agonist G-1 decreases ADAM10 levels and NLRP3-inflammasome component activation in response to Staphylococcus aureus alpha-hemolysin. Microbiologyopen 2024; 13:e23. [PMID: 38867416 PMCID: PMC11168966 DOI: 10.1002/mbo3.1423] [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: 01/08/2024] [Revised: 05/20/2024] [Accepted: 05/30/2024] [Indexed: 06/14/2024] Open
Abstract
The G protein-coupled estrogen receptor, also known as GPER1 or originally GPR30, is found in various tissues, indicating its diverse functions. It is typically present in immune cells, suggesting its role in regulating immune responses to infectious diseases. Our previous studies have shown that G-1, a selective GPER agonist, can limit the pathogenesis mediated by Staphylococcus aureus alpha-hemolysin (Hla). It aids in clearing bacteria in a mouse skin infection model and restricts the surface display of the Hla receptor, ADAM10 (a disintegrin and metalloprotease 10) in HaCaT keratinocytes. In this report, we delve into the modulation of GPER in human immune cells in relation to the NLRP3 inflammasome. We used macrophage-like differentiated THP-1 cells for our study. We found that treating these cells with G-1 reduces ATP release, decreases the activity of the caspase-1 enzyme, and lessens cell death following Hla intoxication. This is likely due to the reduced levels of ADAM10 and NLRP3 proteins, as well as the decreased display of the ADAM10 receptor in the G-1-treated THP-1 cells. Our studies, along with our previous work, suggest the potential therapeutic use of G-1 in reducing Hla susceptibility in humans. This highlights the importance of GPER in immune regulation and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Huayu Zheng
- Department of Pharmaceutical SciencesUniversity of New Mexico Health Sciences Center, College of PharmacyAlbuquerqueNew MexicoUSA
| | - Kathleen D. Triplett
- Department of Pharmaceutical SciencesUniversity of New Mexico Health Sciences Center, College of PharmacyAlbuquerqueNew MexicoUSA
| | - Eric R. Prossnitz
- Department of Internal Medicine, School of Medicine, Center of Biomedical Research Excellence in Autophagy, Inflammation and Metabolism and University of New Mexico Comprehensive Cancer CenterUniversity of New Mexico Health Sciences CenterAlbuquerqueNew MexicoUSA
| | - Pamela R. Hall
- Department of Pharmaceutical SciencesUniversity of New Mexico Health Sciences Center, College of PharmacyAlbuquerqueNew MexicoUSA
| | - Seth M. Daly
- Department of Pharmaceutical SciencesUniversity of New Mexico Health Sciences Center, College of PharmacyAlbuquerqueNew MexicoUSA
| |
Collapse
|
13
|
Vlachakis PK, Theofilis P, Kachrimanidis I, Giannakopoulos K, Drakopoulou M, Apostolos A, Kordalis A, Leontsinis I, Tsioufis K, Tousoulis D. The Role of Inflammasomes in Heart Failure. Int J Mol Sci 2024; 25:5372. [PMID: 38791409 PMCID: PMC11121241 DOI: 10.3390/ijms25105372] [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: 04/24/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024] Open
Abstract
Heart failure (HF) poses a significant world health challenge due to the increase in the aging population and advancements in cardiac care. In the pathophysiology of HF, the inflammasome has been correlated with the development, progression, and complications of HF disease. Discovering biomarkers linked to inflammasomes enhances understanding of HF diagnosis and prognosis. Directing inflammasome signaling emerges as an innovative therapeutic strategy for managing HF. The present review aims to delve into this inflammatory cascade, understanding its role in the development of HF, its potential role as biomarker, as well as the prospects of modulating inflammasomes as a therapeutic approach for HF.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Dimitris Tousoulis
- 1st Department of Cardiology, “Hippokration” General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.K.V.); (P.T.); (I.K.); (K.G.); (M.D.); (A.A.); (A.K.); (I.L.); (K.T.)
| |
Collapse
|
14
|
Hu S, Zhang X, Ding Y, Liu X, Xia R, Wang X. Inhibition of SPARC signal by aerobic exercise to ameliorate atherosclerosis. Int Immunopharmacol 2024; 132:111856. [PMID: 38537537 DOI: 10.1016/j.intimp.2024.111856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND AND AIMS Inflammation and atherosclerosis (AS) are closely associated to Secreted Protein Acidic and Rich in Cysteine (SPARC) and its related factors. This study attempted to define the role and the potential mechanism of SPARC and its related factors in ameliorating hyperlipidemia and AS by aerobic exercise intervention. METHODS The AS rat model was established with a high-fat diet plus vitamin D3 intraperitoneal injection. Treadmill exercises training (5 days/week at 14 m/min for 60 min/day) for 6 weeks was carried out for AS rat intervention method. Western blotting and qRT-PCR were used to analyze the mRNA and protein expression of SPARC and its related factors, respectively. H&E staining was applied to evaluate the morphological changes and inflammation damage. Von Kossa staining was used to measure the degree of vascular calcification. Fluorescence immunohistochemistry staining was used to detect the expression and distribution of SPARC signal molecules. RESULTS SPARC was highly expressed and co-localization with the smooth muscle marker α-SMC in the AS rat. And its downstream factors, NF-κB, Caspase-1, IL-1β and IL-18 were upregulated (P < 0.05 or P < 0.01), FNDC5 expression was downregulated in AS rat model. However, slight declined body weight, delayed AS progression, decreased hyperlipidemia and favorable morphology of skeletal muscle and blood vessels have been detected in AS rat with aerobic exercise intervention. Moreover, the expression of SPARC and its downstream factors were decreased (P < 0.05 or P < 0.01), while elevated the expression of FNDC5 (P < 0.01) was observed after aerobic exercise intervention. CONCLUSIONS This study suggested that aerobic exercise ameliorated hyperlipidemia and AS by effectively inhibiting SPARC signal, and vascular smooth muscle cells may contribute greatly to the protection of AS.
Collapse
Affiliation(s)
- Shujuan Hu
- School of Education and Physical Education, Yangtze University, Jingzhou, Hubei, 434023, China; School of Physical Education and Science, Jishou University, Jishou, China
| | - Xiao Zhang
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China
| | - Yiting Ding
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China
| | - Xuan Liu
- School of Education and Physical Education, Yangtze University, Jingzhou, Hubei, 434023, China
| | - Ruohan Xia
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China.
| | - Xianwang Wang
- Health Science Center, Yangtze University, Jingzhou, Hubei, 434023, China; Shannan Maternal and Child Health Hospital, Shannan, Xizang, 856099, China.
| |
Collapse
|
15
|
Ding XY, Zhang H, Qiu YM, Xie MD, Wang H, Xiong ZY, Li TT, He CN, Dong W, Tang XL. Cardioprotective Potential of Cymbopogon citratus Essential Oil against Isoproterenol-induced Cardiomyocyte Hypertrophy: Possible Involvement of NLRP3 Inflammasome and Oxidative Phosphorylation Complex Subunits. Curr Med Sci 2024; 44:450-461. [PMID: 38639827 DOI: 10.1007/s11596-024-2851-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 02/25/2024] [Indexed: 04/20/2024]
Abstract
OBJECTIVE Cymbopogon citratus (DC.) Stapf is a medicinal and edible herb that is widely used for the treatment of gastric, nervous and hypertensive disorders. In this study, we investigated the cardioprotective effects and mechanisms of the essential oil, the main active ingredient of Cymbopogon citratus, on isoproterenol (ISO)-induced cardiomyocyte hypertrophy. METHODS The compositions of Cymbopogon citratus essential oil (CCEO) were determined by gas chromatography-mass spectrometry. Cardiomyocytes were pretreated with 16.9 µg/L CCEO for 1 h followed by 10 µmol/L ISO for 24 h. Cardiac hypertrophy-related indicators and NLRP3 inflammasome expression were evaluated. Subsequently, transcriptome sequencing (RNA-seq) and target verification were used to further explore the underlying mechanism. RESULTS Our results showed that the CCEO mainly included citronellal (45.66%), geraniol (23.32%), and citronellol (10.37%). CCEO inhibited ISO-induced increases in cell surface area and protein content, as well as the upregulation of fetal gene expression. Moreover, CCEO inhibited ISO-induced NLRP3 inflammasome expression, as evidenced by decreased lactate dehydrogenase content and downregulated mRNA levels of NLRP3, ASC, CASP1, GSDMD, and IL-1β, as well as reduced protein levels of NLRP3, ASC, pro-caspase-1, caspase-1 (p20), GSDMD-FL, GSDMD-N, and pro-IL-1β. The RNA-seq results showed that CCEO inhibited the increase in the mRNA levels of 26 oxidative phosphorylation complex subunits in ISO-treated cardiomyocytes. Our further experiments confirmed that CCEO suppressed ISO-induced upregulation of mt-Nd1, Sdhd, mt-Cytb, Uqcrq, and mt-Atp6 but had no obvious effects on mt-Col expression. CONCLUSION CCEO inhibits ISO-induced cardiomyocyte hypertrophy through the suppression of NLRP3 inflammasome expression and the regulation of several oxidative phosphorylation complex subunits.
Collapse
Affiliation(s)
- Xiao-Yun Ding
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Hao Zhang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Yu-Mei Qiu
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Meng-Die Xie
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Hu Wang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Zheng-Yu Xiong
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Ting-Ting Li
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Chun-Ni He
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China
| | - Wei Dong
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
| | - Xi-Lan Tang
- School of Pharmacy, Jiangxi Science & Technology Normal University, Nanchang, 330013, China.
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Nanchang, 330013, China.
| |
Collapse
|
16
|
Takejima AL, Machado-Júnior PAB, Blume GG, Simeoni RB, Francisco JC, Tonial MS, Marqueze LFB, Noronha L, Olandoski M, Abdelwahid E, Carvalho KATDE, Pinho RA, Guarita-Souza LC. Bone-marrow mononuclear cells and acellular human amniotic membrane improve global cardiac function without inhibition of the NLRP3 Inflammasome in a rat model of heart failure. AN ACAD BRAS CIENC 2024; 96:e20230053. [PMID: 38451595 DOI: 10.1590/0001-3765202420230053] [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: 01/18/2023] [Accepted: 04/19/2023] [Indexed: 03/08/2024] Open
Abstract
Recent studies have suggested that therapies with stem cells and amniotic membrane can modulate the inflammation following an ischemic injury in the heart. This study evaluated the effects of bone-marrow mononuclear cells (BMMC) and acellular human amniotic membrane (AHAM) on cardiac function and NLRP3 complex in a rat model of heart failure.On the 30th day,the echocardiographic showed improvements on ejection fraction and decreased pathological ventricular remodeling on BMMC and AHAM groups.Oxidative stress analysis was similar between the three groups,and the NLRP3 inflammasome activity were not decreased with the therapeutic use of both BMMC and AHAM,in comparison to the control group.
Collapse
Affiliation(s)
- Aline L Takejima
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Paulo André B Machado-Júnior
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Gustavo G Blume
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Rossana Baggio Simeoni
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Julio Cesar Francisco
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Murilo S Tonial
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Luis Felipe B Marqueze
- Pontifícia Universidade Católica do Paraná (PUCPR), Laboratory of Exercise Biochemistry in Health, School of Medicine, 1555 Imaculada Conceição Street, Prado Velho, 80215-901 Curitiba, PR, Brazil
| | - Lucia Noronha
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Marcia Olandoski
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| | - Eltyeb Abdelwahid
- Northwestern University, Feinberg School of Medicine, Feinberg Cardiovascular Research Institute, 303 E. Chicago Ave., Tarry 14-725, 60611 Chicago, IL, USA
| | - Katherine A T DE Carvalho
- The Pelé Pequeno Príncipe Institute, Cell Therapy and Biotechnology in Regenerative Medicine Department, Child and Adolescent Health Research & Pequeno Príncipe Faculties, 1632 Silva Jardim Ave., Água Verde, 80240-020 Curitiba, PR, Brazil
| | - Ricardo A Pinho
- Pontifícia Universidade Católica do Paraná (PUCPR), Laboratory of Exercise Biochemistry in Health, School of Medicine, 1555 Imaculada Conceição Street, Prado Velho, 80215-901 Curitiba, PR, Brazil
| | - Luiz César Guarita-Souza
- Pontifícia Universidade Católica do Paraná (PUCPR), Experimental Laboratory of Institute of Biological and Health Sciences, 1555 Imaculada Conceição Street, 80215-901 Curitiba, PR, Brazil
| |
Collapse
|
17
|
Gurubaran IS. Mitochondrial damage and clearance in retinal pigment epithelial cells. Acta Ophthalmol 2024; 102 Suppl 282:3-53. [PMID: 38467968 DOI: 10.1111/aos.16661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 03/13/2024]
Abstract
Age-related macular degeneration (AMD) is a devastating eye disease that causes permanent vision loss in the central part of the retina, known as the macula. Patients with such severe visual loss face a reduced quality of life and are at a 1.5 times greater risk of death compared to the general population. Currently, there is no cure for or effective treatment for dry AMD. There are several mechanisms thought to underlie the disease, for example, ageing-associated chronic oxidative stress, mitochondrial damage, harmful protein aggregation and inflammation. As a way of gaining a better understanding of the molecular mechanisms behind AMD and thus developing new therapies, we have created a peroxisome proliferator-activated receptor gamma coactivator 1-alpha and nuclear factor erythroid 2-related factor 2 (PGC1α/NFE2L2) double-knockout (dKO) mouse model that mimics many of the clinical features of dry AMD, including elevated levels of oxidative stress markers, damaged mitochondria, accumulating lysosomal lipofuscin and extracellular drusen-like structures in retinal pigment epithelial cells (RPE). In addition, a human RPE cell-based model was established to examine the impact of non-functional intracellular clearance systems on inflammasome activation. In this study, we found that there was a disturbance in the autolysosomal machinery responsible for clearing mitochondria in the RPE cells of one-year-old PGC1α/NFE2L2-deficient mice. The confocal immunohistochemical analysis revealed an increase in autophagosome marker microtubule-associated proteins 1A/1B light chain 3B (LC3B) as well as multiple mitophagy markers such as PTE-induced putative kinase 1 (PINK1) and E3 ubiquitin ligase (PARKIN), along with signs of damaged mitochondria. However, no increase in autolysosome formation was detected, nor was there a colocalization of the lysosomal marker LAMP2 or the mitochondrial marker, ATP synthase β. There was an upregulation of late autolysosomal fusion Ras-related protein (Rab7) in the perinuclear space of RPE cells, together with autofluorescent aggregates. Additionally, we observed an increase in the numbers of Toll-like receptors 3 and 9, while those of NOD-like receptor 3 were decreased in PGC1α/NFE2L2 dKO retinal specimens compared to wild-type animals. There was a trend towards increased complement component C5a and increased involvement of the serine protease enzyme, thrombin, in enhancing the terminal pathway producing C5a, independent of C3. The levels of primary acute phase C-reactive protein and receptor for advanced glycation end products were also increased in the PGC1α/NFE2L2 dKO retina. Furthermore, selective proteasome inhibition with epoxomicin promoted both nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondrial-mediated oxidative stress, leading to the release of mitochondrial DNA to the cytosol, resulting in potassium efflux-dependent activation of the absent in melanoma 2 (AIM2) inflammasome and the subsequent secretion of interleukin-1β in ARPE-19 cells. In conclusion, the data suggest that there is at least a relative decrease in mitophagy, increases in the amounts of C5 and thrombin and decreased C3 levels in this dry AMD-like model. Moreover, selective proteasome inhibition evoked mitochondrial damage and AIM2 inflammasome activation in ARPE-19 cells.
Collapse
Affiliation(s)
- Iswariyaraja Sridevi Gurubaran
- Department of Medicine, Clinical Medicine Unit, University of Eastern Finland Institute of Clinical Medicine, Kuopio, Northern Savonia, Finland
| |
Collapse
|
18
|
Zhang T, Pang C, Xu M, Zhao Q, Hu Z, Jiang X, Guo M. The role of immune system in atherosclerosis: Molecular mechanisms, controversies, and future possibilities. Hum Immunol 2024; 85:110765. [PMID: 38369442 DOI: 10.1016/j.humimm.2024.110765] [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: 07/17/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/20/2024]
Abstract
Numerous cardiovascular disorders have atherosclerosis as their pathological underpinning. Numerous studies have demonstrated that, with the aid of pattern recognition receptors, cytokines, and immunoglobulins, innate immunity, represented by monocytes/macrophages, and adaptive immunity, primarily T/B cells, play a critical role in controlling inflammation and abnormal lipid metabolism in atherosclerosis. Additionally, the finding of numerous complement components in atherosclerotic plaques suggests yet again how heavily the immune system controls atherosclerosis. Therefore, it is essential to have a thorough grasp of how the immune system contributes to atherosclerosis. The specific molecular mechanisms involved in the activation of immune cells and immune molecules in atherosclerosis, the controversy surrounding some immune cells in atherosclerosis, and the limitations of extrapolating from relevant animal models to humans were all carefully reviewed in this review from the three perspectives of innate immunity, adaptive immunity, and complement system. This could provide fresh possibilities for atherosclerosis research and treatment in the future.
Collapse
Affiliation(s)
- Tianle Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Chenxu Pang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Mengxin Xu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Qianqian Zhao
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Zhijie Hu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| | - Maojuan Guo
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, PR China.
| |
Collapse
|
19
|
Yi YS. MicroRNA-mediated epigenetic regulation of inflammasomes in inflammatory responses and immunopathologies. Semin Cell Dev Biol 2024; 154:227-238. [PMID: 36437174 DOI: 10.1016/j.semcdb.2022.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/18/2022] [Accepted: 11/19/2022] [Indexed: 11/27/2022]
Abstract
Inflammation represents the first-line defense mechanism of the host against pathogens and cellular stress. One of the most critical inflammatory responses is characterized by the activation of inflammasomes, intracellular multiprotein complexes that induce inflammatory signaling pathways in response to various pathogen-associated molecular patterns or danger-associated molecular patterns under physiological and pathological conditions. Inflammasomes are tightly regulated in normal cells, and dysregulation of these complexes is observed in various pathological conditions, especially inflammatory diseases and cancers. Epigenetic regulation has been suggested as a key mechanism in modulating inflammasome activity, and microRNAs (miRNAs) have been implicated in the post-transcriptional regulation of inflammasomes. Therefore, miRNA-mediated epigenetic regulation of inflammasomes in pathological conditions has received considerable attention, and current strategies for targeting inflammasomes have been shown to be effective in the treatment of diseases associated with inflammasome activation. This review summarizes recent studies suggesting the roles of miRNAs in the epigenetic control of inflammasomes and highlights the potential of miRNAs as a therapeutic tool for treating human diseases.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, South Korea.
| |
Collapse
|
20
|
Rusetskaya NY, Loginova NY, Pokrovskaya EP, Chesovskikh YS, Titova LE. Redox regulation of the NLRP3-mediated inflammation and pyroptosis. BIOMEDITSINSKAIA KHIMIIA 2023; 69:333-352. [PMID: 38153050 DOI: 10.18097/pbmc20236906333] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
The review considers modern data on the mechanisms of activation and redox regulation of the NLRP3 inflammasome and gasdermins, as well as the role of selenium in these processes. Activation of the inflammasome and pyroptosis represent an evolutionarily conserved mechanism of the defense against pathogens, described for various types of cells and tissues (macrophages and monocytes, microglial cells and astrocytes, podocytes and parenchymal cells of the kidneys, periodontal tissues, osteoclasts and osteoblasts, as well as cells of the digestive and urogenital systems, etc.). Depending on the characteristics of redox regulation, the participants of NLRP3 inflammation and pyroptosis can be subdivided into 2 groups. Members of the first group block the mitochondrial electron transport chain, promote the formation of reactive oxygen species and the development of oxidative stress. This group includes granzymes, the mitochondrial antiviral signaling protein MAVS, and others. The second group includes thioredoxin interacting protein (TXNIP), erythroid-derived nuclear factor-2 (NRF2), Kelch-like ECH-associated protein 1 (Keap1), ninjurin (Ninj1), scramblase (TMEM16), inflammasome regulatory protein kinase NLRP3 (NEK7), caspase-1, gasdermins GSDM B, D and others. They have redox-sensitive domains and/or cysteine residues subjected to redox regulation, glutathionylation/deglutathionylation or other types of regulation. Suppression of oxidative stress and redox regulation of participants in NLRP3 inflammation and pyroptosis depends on the activity of the antioxidant enzymes glutathione peroxidase (GPX) and thioredoxin reductase (TRXR), containing a selenocysteine residue Sec in the active site. The expression of GPX and TRXR is regulated by NRF2 and depends on the concentration of selenium in the blood. Selenium deficiency causes ineffective translation of the Sec UGA codon, translation termination, and, consequently, synthesis of inactive selenoproteins, which can cause various types of programmed cell death: apoptosis of nerve cells and sperm, necroptosis of erythrocyte precursors, pyroptosis of infected myeloid cells, ferroptosis of T- and B-lymphocytes, kidney and pancreatic cells. In addition, suboptimal selenium concentrations in the blood (0.86 μM or 68 μg/l or less) have a significant impact on expression of more than two hundred and fifty genes as compared to the optimal selenium concentration (1.43 μM or 113 μg/l). Based on the above, we propose to consider blood selenium concentrations as an important parameter of redox homeostasis in the cell. Suboptimal blood selenium concentrations (or selenium deficiency states) should be used for assessment of the risk of developing inflammatory processes.
Collapse
Affiliation(s)
- N Yu Rusetskaya
- V.I. Razumovsky Saratov State Medical University, Saratov, Russia
| | - N Yu Loginova
- V.I. Razumovsky Saratov State Medical University, Saratov, Russia
| | - E P Pokrovskaya
- V.I. Razumovsky Saratov State Medical University, Saratov, Russia
| | - Yu S Chesovskikh
- V.I. Razumovsky Saratov State Medical University, Saratov, Russia
| | - L E Titova
- V.I. Razumovsky Saratov State Medical University, Saratov, Russia
| |
Collapse
|
21
|
Liudvytska O, Bandyszewska M, Skirecki T, Krzyżanowska-Kowalczyk J, Kowalczyk M, Kolodziejczyk-Czepas J. Anti-inflammatory and antioxidant actions of extracts from Rheum rhaponticum and Rheum rhabarbarum in human blood plasma and cells in vitro. Biomed Pharmacother 2023; 165:115111. [PMID: 37421780 DOI: 10.1016/j.biopha.2023.115111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/10/2023] Open
Abstract
Rheum rhaponticum L. (rhapontic rhubarb) and Rheum rhabarbarum L. (garden rhubarb) are edible and medicinal rhubarb species used for many centuries in traditional medicine. This work is focused on the biological activity of extracts from petioles and roots of R. rhaponticum and R. rhabarbarum as well as rhapontigenin and rhaponticin, typical stilbenes present in these rhubarbs, in a context of their effects on blood physiology and cardiovascular health. Anti-inflammatory properties of the examined substances were evaluated in human peripheral blood mononuclear cells (PBMCs) and THP1-ASC-GFP inflammasome reporter cells. Due to the coexistence of inflammation and oxidative stress in cardiovascular diseases, the study design included also antioxidant assays. This part of the work involved the assessment of the protective efficiency of the examined substances against the peroxynitrite-triggered damage to human blood plasma components, including fibrinogen, a protein of critical importance for blood clotting and maintaining the haemostatic balance. Pre-incubation of PBMCs with the examined substances (1-50 μg/mL) considerably decreased the synthesis of prostaglandin E2 as well as the release of pro-inflammatory cytokines (IL-2 and TNF-α) and metalloproteinase-9. A reduced level of secreted apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) specks in the THP-1-ASC-GFP cells was also observed. The examined substances significantly diminished the extent of ONOO‾induced oxidative modifications of blood plasma proteins and lipids and normalized, or even strengthened blood plasma antioxidant capacity. Furthermore, a reduction of oxidative damage to fibrinogen, including modifications of tyrosine and tryptophan residues along with the formation of protein aggregates was found.
Collapse
Affiliation(s)
- Oleksandra Liudvytska
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| | - Magdalena Bandyszewska
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland.
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland.
| | - Justyna Krzyżanowska-Kowalczyk
- Department of Biochemistry and Crop Quality, Institute of Soil Science and Plant Cultivation, State Research Institute, Czartoryskich 8, 24-100 Puławy, Poland.
| | - Mariusz Kowalczyk
- Department of Biochemistry and Crop Quality, Institute of Soil Science and Plant Cultivation, State Research Institute, Czartoryskich 8, 24-100 Puławy, Poland.
| | - Joanna Kolodziejczyk-Czepas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| |
Collapse
|
22
|
Ya'ar Bar S, Pintel N, Abd Alghne H, Khattib H, Avni D. The therapeutic potential of sphingolipids for cardiovascular diseases. Front Cardiovasc Med 2023; 10:1224743. [PMID: 37608809 PMCID: PMC10440740 DOI: 10.3389/fcvm.2023.1224743] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/17/2023] [Indexed: 08/24/2023] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide and Inflammation plays a critical role in the development of CVD. Despite considerable progress in understanding the underlying mechanisms and various treatment options available, significant gaps in therapy necessitate the identification of novel therapeutic targets. Sphingolipids are a family of lipids that have gained attention in recent years as important players in CVDs and the inflammatory processes that underlie their development. As preclinical studies have shown that targeting sphingolipids can modulate inflammation and ameliorate CVDs, targeting sphingolipids has emerged as a promising therapeutic strategy. This review discusses the current understanding of sphingolipids' involvement in inflammation and cardiovascular diseases, the existing therapeutic approaches and gaps in therapy, and explores the potential of sphingolipids-based drugs as a future avenue for CVD treatment.
Collapse
Affiliation(s)
- Sapir Ya'ar Bar
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Noam Pintel
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
| | - Hesen Abd Alghne
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| | - Hamdan Khattib
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Department of Gastroenterology and Hepatology, Tel Aviv University Sackler Faculty of Medicine, Tel Aviv, Israel
| | - Dorit Avni
- Department of Natural Compound, Nutrition, and Health, MIGAL, Kiryat Shmona, Israel
- Tel-Hai College Department of Biotechnology, Kiryat Shmona, Israel
| |
Collapse
|
23
|
Chen L, Wang M, Zhang W, Zhang X, Jiao C, Yu Y, Guan B, Zhong Z. The value of inflammatory biomarkers in the occurrence and prognosis of sudden sensorineural hearing loss: a meta-analysis. Eur Arch Otorhinolaryngol 2023; 280:3119-3129. [PMID: 36574064 DOI: 10.1007/s00405-022-07806-9] [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: 08/30/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022]
Abstract
OBJECTIVES Sudden sensorineural hearing loss (SSNHL) is one of the common emergencies in otorhinolaryngology. Several studies have shown that chronic inflammation is associated with its onset and prognosis. However, the association between some inflammatory biomarkers and SSNHL is still unclear. Therefore, we conducted this meta-analysis to explore the value of inflammatory biomarkers in the occurrence and prognosis of SSNHL. METHODS Pubmed, Embase, Cochrane and Web of Science databases were searched comprehensively, the eligible literatures were screened out by formulating the inclusion criteria and exclusion criteria. After extracting sample size, mean and standard deviation, we performed meta-analysis with standardized mean deviation (SMD) and 95% confidence interval (CI) as effect sizes. RESULTS A total of 17 articles were included in this meta-analysis, including 2852 subjects, 1423 patients and 1429 healthy controls. The results of meta-analysis showed that the neutrophil-to-lymphocyte ratio (NLR) of the experimental group was significantly higher than the control group (SMD = 1.05, 95% CI 0.87-1.24, P < 0.001), the NLR of the recovery group was significantly lower than the unrecovered group (SMD = 0.68, 95% CI 0.27-1.08, P < 0.05); The platelet-to-lymphocyte ratio (PLR) of the experimental group was significantly higher than the control group (SMD = 0.55, 95% CI 0.34-0.76, P < 0.05), the PLR of the recovery group was significantly lower than the unrecovered group (SMD = 0.44, 95% CI 0.05-0.82, P < 0.05); The C-reactive protein-to-serum albumin ratio (CRP/Alb) of the experimental group was significantly higher than the control group (SMD = 0.39, 95% CI 0.04-0.74, P < 0.05). CONCLUSIONS The results showed that high NLR, PLR, and CRP/Alb indicated the occurrence of SSNHL, NLR and PLR could predict prognosis of SSNHL.
Collapse
Affiliation(s)
- Li Chen
- Department of Otolaryngology, Head and Neck Surgery, Dalian Medical University, Dalian, 116000, China
| | - Maohua Wang
- Department of OtolaryngologyHead and Neck Surgery, Hearing and Balance Medical Engineering Technology Center of Guangdong, The First People's Hospital of Foshan, Foshan, 528000, China
| | - Wentao Zhang
- Department of Otolaryngology, Head and Neck Surgery, Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Xu Zhang
- Department of Otolaryngology, Head and Neck Surgery, Dalian Medical University, Dalian, 116000, China
| | - Cheng Jiao
- Department of Otolaryngology, Head and Neck Surgery, Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Youjun Yu
- Department of OtolaryngologyHead and Neck Surgery, Hearing and Balance Medical Engineering Technology Center of Guangdong, The First People's Hospital of Foshan, Foshan, 528000, China
| | - Bing Guan
- Department of Otolaryngology, Head and Neck Surgery, Clinical Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Zhenhua Zhong
- Department of Otolaryngology, Head and Neck Surgery, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China.
| |
Collapse
|
24
|
Xu J, Zhou Z, Zheng Y, Yang S, Huang K, Li H. Roles of inflammasomes in viral myocarditis. Front Cell Infect Microbiol 2023; 13:1149911. [PMID: 37256114 PMCID: PMC10225676 DOI: 10.3389/fcimb.2023.1149911] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/28/2023] [Indexed: 06/01/2023] Open
Abstract
Viral myocarditis (VMC), characterized by viral infection-induced inflammation, is a life-threatening disease associated with dilated cardiomyopathy or heart failure. Innate immunity plays a crucial role in the progression of inflammation, in which inflammasomes provide a platform for the secretion of cytokines and mediate pyroptosis. Inflammasomes are rising stars gaining increasing attention. The nucleotide oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome, the caspase recruitment domain-containing protein 8 (CARD8) inflammasome, and the caspase-11 inflammasome are three inflammasomes that were reported to affect the process and prognosis of VMC. These inflammasomes can be activated by a wide range of cellular events. Accumulating evidence has suggested that inflammasomes are involved in different stages of VMC, including the trigger and progression of myocardial injury and remodeling after infection. In this review, we summarized the pathways involving inflammasomes in VMC and discussed the potential therapies targeting inflammasomes and related pathways.
Collapse
Affiliation(s)
- Jingyu Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihao Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yidan Zheng
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sai Yang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kun Huang
- Institution of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huili Li
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Zong J, Wang Y, Pan S, Yang Y, Peng J, Li F, Xu L, Li S, Qian W. The Relationship between the Serum NLRP1 Level and Coronary Lesions in Patients with Coronary Artery Disease. Int J Clin Pract 2023; 2023:2250055. [PMID: 37214347 PMCID: PMC10195180 DOI: 10.1155/2023/2250055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/22/2023] [Accepted: 05/02/2023] [Indexed: 05/24/2023] Open
Abstract
Background The pathogenesis of coronary artery disease is complex, and inflammation is one of the regulatory factors. The nucleotide-binding oligomerization domain (NOD)-like receptor protein 1 (NLRP1) plays an important role in the cellular inflammatory response, cell apoptosis, cell death, and autoimmune diseases. Whether the level of NLRP1 is related to the severity of coronary artery stenosis in patients with coronary artery disease (CAD) has not been reported. Objective To test the serum level of NLRP1 in unstable angina (UA) patients and investigate the effect of NLRP1 on coronary stenosis severity of the coronary artery disease (CAD). Methods 307 patients hospitalized in the Department of Cardiology of the Affiliated Hospital of Xuzhou Medical University for coronary angiography from January 1, 2021, to December 31, 2022 were included. We detect the level of NLRP1 in the serum of the included patients. Patients were divided into UA group and control group according to coronary angiography results and other clinical data. We use logistic regression to screen the influencing factors of UA. Then, subgroups were divided according to the Gensini score and the number of coronary artery lesions, and the difference of serum NLRP1 level between the groups was compared. Spearman correlation analysis was used to explore the correlation between the serum NLRP1 level and Gensini score. We analyze the diagnostic value of NLRP1 for UA by drawing ROC curve. Results The median level of serum NLRP1 in patients with UA (n = 257) was 49.71 pg/ml, IQR 30.15, 80.21, and that in patients without UA (n = 50) was 24.75 pg/ml, IQR 13.49, 41.95. Serum NLRP1 levels were significantly different among different subgroups. The patient's Gensini score was correlated with the patient's serum NLRP1 level. Conclusion The serum NLRP1 level is increased in patients with UA, which is increased with the increasing severity of coronary lesions.
Collapse
Affiliation(s)
- Jing Zong
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
- Institute of Cardiovascular Disease, Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
| | - Yixiao Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
- Institute of Cardiovascular Disease, Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
| | - Siyu Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
- Institute of Cardiovascular Disease, Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
| | - Yiming Yang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
- Institute of Cardiovascular Disease, Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
| | - Jingfeng Peng
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
- Institute of Cardiovascular Disease, Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
| | - Fangfang Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
- Institute of Cardiovascular Disease, Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
| | - Luhong Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
- Institute of Cardiovascular Disease, Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
| | - Shanshan Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
| | - Wenhao Qian
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
- Institute of Cardiovascular Disease, Xuzhou Medical University, Xuzhou 221000, Jiangsu, China
| |
Collapse
|
26
|
Activation and Pharmacological Regulation of Inflammasomes. Biomolecules 2022; 12:biom12071005. [PMID: 35883561 PMCID: PMC9313256 DOI: 10.3390/biom12071005] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 01/27/2023] Open
Abstract
Inflammasomes are intracellular signaling complexes of the innate immune system, which is part of the response to exogenous pathogens or physiological aberration. The multiprotein complexes mainly consist of sensor proteins, adaptors, and pro-caspase-1. The assembly of the inflammasome upon extracellular and intracellular cues drives the activation of caspase-1, which processes pro-inflammatory cytokines IL-1β and IL-18 to maturation and gasdermin-D for pore formation, leading to pyroptosis and cytokine release. Inflammasome signaling functions in numerous infectious or sterile inflammatory diseases, including inherited autoinflammatory diseases, metabolic disorders, cardiovascular diseases, cancers, neurodegenerative disorders, and COVID-19. In this review, we summarized current ideas on the organization and activation of inflammasomes, with details on the molecular mechanisms, regulations, and interventions. The recent developments of pharmacological strategies targeting inflammasomes as disease therapeutics were also covered.
Collapse
|
27
|
Sridevi Gurubaran I, Hytti M, Kaarniranta K, Kauppinen A. Epoxomicin, a Selective Proteasome Inhibitor, Activates AIM2 Inflammasome in Human Retinal Pigment Epithelium Cells. Antioxidants (Basel) 2022; 11:antiox11071288. [PMID: 35883779 PMCID: PMC9311580 DOI: 10.3390/antiox11071288] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/26/2022] [Accepted: 06/26/2022] [Indexed: 02/04/2023] Open
Abstract
Emerging evidence suggests that the intracellular clearance system plays a vital role in maintaining homeostasis and in regulating oxidative stress and inflammation in retinal pigment epithelium (RPE) cells. Dysfunctional proteasomes and autophagy in RPE cells have been associated with the pathogenesis of age-related macular degeneration. We have previously shown that the inhibition of proteasomes using MG-132 activates the NLR family pyrin domain containing 3 (NLRP3) inflammasome in human RPE cells. However, MG-132 is a non-selective proteasome inhibitor. In this study, we used the selective proteasome inhibitor epoxomicin to study the effect of non-functional intracellular clearance systems on inflammasome activation. Our data show that epoxomicin-induced proteasome inhibition promoted both nicotinamide adenine dinucleotide phosphate oxidase and mitochondria-mediated oxidative stress and release of mitochondrial DNA to the cytosol, which resulted in potassium efflux-dependent absence in melanoma 2 (AIM2) inflammasome activation and subsequent interleukin-1β secretion in ARPE-19 cells. The non-specific proteasome inhibitor MG-132 activated both NLRP3 and AIM2 inflammasomes and oxidative stress predominated as the activation mechanism, but modest potassium efflux was also detected. Collectively, our data suggest that a selective proteasome inhibitor is a potent inflammasome activator in human RPE cells and emphasize the role of the AIM2 inflammasome in addition to the more commonly known NLRP3 inflammasome.
Collapse
Affiliation(s)
- Iswariyaraja Sridevi Gurubaran
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (I.S.G.); (K.K.)
| | - Maria Hytti
- Immuno-Ophthalmology, School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (I.S.G.); (K.K.)
- Department of Ophthalmology, University of Eastern Finland, 70211 Kuopio, Finland
- Department of Ophthalmology, Kuopio University Hospital, 70029 Kuopio, Finland
| | - Anu Kauppinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, 70210 Kuopio, Finland; (I.S.G.); (K.K.)
- Immuno-Ophthalmology, School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, 70210 Kuopio, Finland;
- Correspondence:
| |
Collapse
|
28
|
Papadakos SP, Dedes N, Kouroumalis E, Theocharis S. The Role of the NLRP3 Inflammasome in HCC Carcinogenesis and Treatment: Harnessing Innate Immunity. Cancers (Basel) 2022; 14:3150. [PMID: 35804922 PMCID: PMC9264914 DOI: 10.3390/cancers14133150] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/13/2022] [Accepted: 06/20/2022] [Indexed: 01/27/2023] Open
Abstract
The HCC constitutes one of the most frequent cancers, with a non-decreasing trend in disease mortality despite advances in systemic therapy and surgery. This trend is fueled by the rise of an obesity wave which is prominent the Western populations and has reshaped the etiologic landscape of HCC. Interest in the nucleotide-binding domain leucine-rich repeat containing (NLR) family member NLRP3 has recently been revived since it would appear that, by generating inflammasomes, it participates in several physiologic processes and its dysfunction leads to disease. The NLRP3 inflammasome has been studied in depth, and its influence in HCC pathogenesis has been extensively documented during the past quinquennial. Since inflammation comprises a major regulator of carcinogenesis, it is of paramount importance an attempt to evaluate the contribution of the NLRP3 inflammasome to the generation and management of HCC. The aim of this review was to examine the literature in order to determine the impact of the NLRP3 inflammasome on, and present a hypothesis about its input in, HCC.
Collapse
Affiliation(s)
- Stavros P. Papadakos
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (N.D.)
| | - Nikolaos Dedes
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (N.D.)
| | - Elias Kouroumalis
- Department of Gastroenterology, Medical School, University of Crete, 71500 Heraklion, Greece;
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.P.P.); (N.D.)
| |
Collapse
|
29
|
Nappi F, Bellomo F, Avtaar Singh SS. Insights into the Role of Neutrophils and Neutrophil Extracellular Traps in Causing Cardiovascular Complications in Patients with COVID-19: A Systematic Review. Biomedicines 2022; 11:2460. [PMID: 35566589 PMCID: PMC9855935 DOI: 10.3390/biomedicines11010113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The coronavirus disease 2019 (COVID-19) pandemic caused by the SARS-CoV-2 virus has resulted in significant mortality and burdening of healthcare resources. While initially noted as a pulmonary pathology, subsequent studies later identified cardiovascular involvement with high mortalities reported in specific cohorts of patients. While cardiovascular comorbidities were identified early on, the exact manifestation and etiopathology of the infection remained elusive. This systematic review aims to investigate the role of inflammatory pathways, highlighting several culprits including neutrophil extracellular traps (NETs) which have since been extensively investigated. METHOD A search was conducted using three databases (MEDLINE; MEDLINE In-Process & Other Non-Indexed Citations and EMBASE). Data from randomized controlled trials (RCT), prospective series, meta-analyses, and unmatched observational studies were considered for the processing of the algorithm and treatment of inflammatory response during SARS-CoV-2 infection. Studies without the SARS-CoV-2 Infection period and case reports were excluded. RESULTS A total of 47 studies were included in this study. The role of the acute inflammatory response in the propagation of the systemic inflammatory sequelae of the disease plays a major part in determining outcomes. Some of the mechanisms of activation of these pathways have been highlighted in previous studies and are highlighted. CONCLUSION NETs play a pivotal role in the pathogenesis of the inflammatory response. Despite moving into the endemic phase of the disease in most countries, COVID-19 remains an entity that has not been fully understood with long-term effects remaining uncertain and requiring ongoing monitoring and research.
Collapse
Affiliation(s)
- Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord of Saint-Denis, 93200 Saint-Denis, France
| | - Francesca Bellomo
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | | |
Collapse
|