1
|
Li K, Wang S, Li J, Wang L, Zhang Q, Hou L, Yu X, Liu Z, Lv T, Shang L. Low shear stress induces vascular endothelial cells apoptosis via miR-330 /SOD2 /HSP70 signaling pathway. Exp Cell Res 2025; 445:114410. [PMID: 39788367 DOI: 10.1016/j.yexcr.2025.114410] [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: 07/25/2024] [Revised: 11/30/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Atherosclerosis (AS) is a chronic disease initiated by vascular endothelial dysfunction, with low shear stress (SS) being a critical inducing factor in this dysfunction. Apoptosis, a form of programmed cell death, is closely associated with AS progression. However, the impact of low SS on endothelial apoptosis and its specific molecular mechanisms remains unclear. Our study revealed that low SS induces apoptosis in endothelial cells and contributes to endothelial dysfunction. Under low SS conditions, miR-330 expression was markedly upregulated, which subsequently targeted and inhibited SOD2 expression, leading to ROS accumulation and oxidative stress. Overexpression of SOD2 under low SS conditions markedly elevated HSP70 expression, contributing to endothelial homeostasis. However, when HSP70 expression was inhibited in the context of SOD2 overexpression, there was a significant increase in pro-apoptotic proteins (BAX and cleaved-caspase-3) and total apoptosis rate, along with a significant reduction in endothelial function markers such as nitric oxide and endothelial nitric oxide synthase. Notably, our experiments indicated no direct interaction between SOD2 and HSP70. Furthermore, inhibiting ROS production significantly raised HSP70 expression, suggesting that SOD2 regulates HSP70 in an indirect process involving ROS. In summary, our findings elucidate that low SS induces endothelial apoptosis and dysfunction through the miR-330/SOD2/HSP70 signaling pathway, providing valuable insights into AS intervention and prevention.
Collapse
Affiliation(s)
- Ke Li
- Department of Gastroenterology, 215 Hospital of Shaanxi Province, Xianyang, 712000, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Shaohu Wang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China; Department of Immunization and Planning, Heping District Center for Disease Control and Prevention, Tianjin, 300041, China
| | - Jiana Li
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China; Department of Gastroenterology, No.983rd Hospital of the Chinese People's Liberation Army Joint Logistics and Security Forces, Tianjin, 300143, China
| | - Lingling Wang
- Department of Gastroenterology, 215 Hospital of Shaanxi Province, Xianyang, 712000, China
| | - Qin Zhang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Liming Hou
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Xinyi Yu
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Zhendong Liu
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Ting Lv
- Department of Gastroenterology, 215 Hospital of Shaanxi Province, Xianyang, 712000, China.
| | - Luxiang Shang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China; Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, 250014, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
2
|
Kobzeva K, Ivenkov M, Gromov R, Bushueva O. HSP90 Family Members, Their Regulators and Ischemic Stroke Risk: A Comprehensive Molecular-Genetics and Bioinformatics Analysis. Front Biosci (Schol Ed) 2024; 16:19. [PMID: 39736019 DOI: 10.31083/j.fbs1604019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/15/2024] [Accepted: 11/21/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Disruptions in proteostasis are recognized as key drivers in cerebro- and cardiovascular disease progression. Heat shock proteins (HSPs), essential for maintaining protein stability and cellular homeostasis, are pivotal in neuroperotection. Consequently, deepening the understanding the role of HSPs in ischemic stroke (IS) risk is crucial for identifying novel therapeutic targets and advancing neuroprotective strategies. AIM Our objective was to examine the potential correlation between single nucleotide polymorphisms (SNPs) in genes that encode members of the Heat shock protein 90 (HSP90), small heat shock proteins (HSPB), and heat shock factors (HSF) families, and the risk and clinical characteristics of IS. METHODS 953 IS patients and 1265 controls from Central Russia were genotyped for nine SNPs in genes encoding HSP90AA1, HSFs, and HSPBs using the MassArray-4 system and probe-based polymerase chain reaction (PCR). RESULTS In smokers, SNP rs1133026 HSPB8 increased the risk of IS (risk allele A, odds ratio (OR) = 1.43, 95% Confidence Interval (CI) 1.02-2.02, p = 0.035), and rs556439 HSF2 increased the brain infarct size (risk allele A, p = 0.02). In non-smokers, SNPs rs4279640 HSF1 (protective allele T, OR = 0.58, 95% CI 0.37-0.92, p = 0.02) and rs4264324 HSP90AA1 (protective allele C, OR = 0.11, 95% CI 0.01-0.78, p = 0.001) lowered the risk of recurrent stroke; SNP rs7303637 HSPB8 increased the age of onset of IS (protective allele T, p = 0.04). In patients with body mass index (BMI) ≥25, SNPs rs556439 HSF2 (risk allele A, OR = 1.33, 95% CI 1.04-1.69, p = 0.02) and rs549302 HSF2 (risk allele G, OR = 1.34, 95% CI 1.02-1.75, p = 0.03) were linked to a higher risk of IS. CONCLUSIONS The primary molecular mechanisms through which the studied SNPs contribute to IS pathogenesis were found to be the regulation of cell death, inflammatory and oxidative stress responses.
Collapse
Affiliation(s)
- Ksenia Kobzeva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Maxim Ivenkov
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Rostislav Gromov
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Olga Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 305041 Kursk, Russia
| |
Collapse
|
3
|
Zelko IN, Hussain A, Malovichko MV, Wickramasinghe N, Srivastava S. Benzene metabolites increase vascular permeability by activating heat shock proteins and Rho GTPases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626801. [PMID: 39677674 PMCID: PMC11643022 DOI: 10.1101/2024.12.04.626801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Benzene is a ubiquitous environmental and occupational pollutant abundant in household products, petrochemicals, and cigarette smoke. It is also a well-known carcinogen and hematopoietic toxin. Population-based studies indicate an increased risk of heart failure in subjects exposed to inhaled benzene, which coincides with the infiltration of immune cells into the myocardium. However, the mechanisms of benzene-induced cardiovascular disease remain unknown. Our data suggests that benzene metabolites trans,trans-muconaldehyde (MA), and hydroquinone (HQ) propagate endothelial activation and apoptosis analyzed by endothelial-specific microparticles in C57BL/6J mice plasma. Subcutaneous injections of MA and HQ increased vascular permeability by 1.54 fold and 1.27 fold correspondingly. In addition, the exposure of primary cardiac microvascular endothelial cells to MA increased vascular permeability detected by transendothelial monolayer resistance and by fluorescently labeled dextrans diffusion. The bulk RNA sequencing of endothelial cells exposed to MA for 2, 6, and 24 hours showed MA-dependent upregulation of heat shock-related pathways at 2 and 6 hours, dysregulation of GTPases at 6 hours, and altered cytoskeleton organization at 24 hours of exposure. We found that the HSP70 protein induced by MA in endothelial cells is colocalized with F-actin foci. HSP70 inhibitor 17AAG and HSP90 inhibitor JG98 attenuated MA-induced endothelial permeability, while HSP activator TRC enhanced endothelial leakage. Moreover, MA induced Rac1 GTPase activity, while Rho GTPase inhibitor Y-27632 attenuated MA-induced endothelial permeability. We showed that benzene metabolites compromised the endothelial barrier by altering HSP- and GTPase-related signaling pathways.
Collapse
Affiliation(s)
- Igor N. Zelko
- University of Louisville Superfund Research Center, University of Louisville, Louisville, KY 40202
- Envirome Institute, University of Louisville, Louisville, KY 40202
- Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Ahtesham Hussain
- University of Louisville Superfund Research Center, University of Louisville, Louisville, KY 40202
- Envirome Institute, University of Louisville, Louisville, KY 40202
- Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Marina V. Malovichko
- University of Louisville Superfund Research Center, University of Louisville, Louisville, KY 40202
- Envirome Institute, University of Louisville, Louisville, KY 40202
- Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Nalinie Wickramasinghe
- University of Louisville Superfund Research Center, University of Louisville, Louisville, KY 40202
- Envirome Institute, University of Louisville, Louisville, KY 40202
- Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| | - Sanjay Srivastava
- University of Louisville Superfund Research Center, University of Louisville, Louisville, KY 40202
- Envirome Institute, University of Louisville, Louisville, KY 40202
- Department of Medicine, Division of Environmental Medicine, University of Louisville, Louisville, KY 40202
| |
Collapse
|
4
|
Wang S, Chen Y, Zhou D, Zhang J, Guo G, Chen Y. Pathogenic Autoimmunity in Atherosclerosis Evolves from HSP60-Reactive CD4 + T Cells. J Cardiovasc Transl Res 2024; 17:1172-1180. [PMID: 38767798 DOI: 10.1007/s12265-024-10516-8] [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: 02/24/2024] [Accepted: 04/15/2024] [Indexed: 05/22/2024]
Abstract
Clinical evidence suggests anti-Hsp60 antibodies could contribute to atherosclerosis (AS) development, with unclear mechanisms. This study aims to explore the role of anti-HSP60-mediated autoimmunity in AS progression. HSP60-MHC tetramers were used to characterize HSP60-specific CD4 + T cells and assess TCR responses in mice. These cells were transplanted into AS mice to examine immune cell differentiation and infiltration in plaques and blood. Mice were injected with recombinant HSP60 or anti-HSP60 sera to evaluate effects on plaque progression and macrophage activity. Experiments with muMT-/-Apoe-/- mice examined humoral immunity's role in this autoimmunity. HSP60-reactive CD4 + T cells in AS mice differentiated into follicular helper cells, not Th1/Th17. Anti-HSP60 treatments increased macrophage infiltration and M1 polarization, indicating an anti-HSP60-driven inflammatory progression, dependent on humoral immunity. Anti-HSP60 influences macrophage infiltration, polarization, and plaque formation via humoral immunity, shedding light on its potential role in AS progression.
Collapse
MESH Headings
- Animals
- Chaperonin 60/immunology
- Chaperonin 60/genetics
- Autoimmunity
- Atherosclerosis/immunology
- Atherosclerosis/genetics
- Atherosclerosis/pathology
- Atherosclerosis/blood
- Disease Models, Animal
- Macrophages/immunology
- Macrophages/metabolism
- Plaque, Atherosclerotic
- Mice, Knockout, ApoE
- Immunity, Humoral
- Mice, Inbred C57BL
- Cell Differentiation
- Phenotype
- T Follicular Helper Cells/immunology
- Adoptive Transfer
- Mitochondrial Proteins/genetics
- Mitochondrial Proteins/immunology
- Mitochondrial Proteins/metabolism
- Autoantibodies/blood
- Aortic Diseases/immunology
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Cells, Cultured
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Disease Progression
- Macrophage Activation
- Male
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Shixiang Wang
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, No. 63, Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Yongquan Chen
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, No. 63, Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Danyan Zhou
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, No. 63, Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Jiawei Zhang
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, No. 63, Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Guofeng Guo
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, No. 63, Duobao Road, Liwan District, Guangzhou, 510150, China
| | - Youquan Chen
- Department of Cardiology, The Third Affiliated Hospital of Guangzhou Medical University, No. 63, Duobao Road, Liwan District, Guangzhou, 510150, China.
| |
Collapse
|
5
|
Sircana MC, Erre GL, Castagna F, Manetti R. Crosstalk between Inflammation and Atherosclerosis in Rheumatoid Arthritis and Systemic Lupus Erythematosus: Is There a Common Basis? Life (Basel) 2024; 14:716. [PMID: 38929699 PMCID: PMC11204900 DOI: 10.3390/life14060716] [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: 05/01/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in patients with rheumatoid arthritis and systemic lupus erythematosus. Traditional cardiovascular risk factors, although present in lupus and rheumatoid arthritis, do not explain such a high burden of early cardiovascular disease in the context of these systemic connective tissue diseases. Over the past few years, our understanding of the pathophysiology of atherosclerosis has changed from it being a lipid-centric to an inflammation-centric process. In this review, we examine the pathogenesis of atherosclerosis in systemic lupus erythematosus and rheumatoid arthritis, the two most common systemic connective tissue diseases, and consider them as emblematic models of the effect of chronic inflammation on the human body. We explore the roles of the inflammasome, cells of the innate and acquired immune system, neutrophils, macrophages, lymphocytes, chemokines and soluble pro-inflammatory cytokines in rheumatoid arthritis and systemic lupus erythematosus, and the roles of certain autoantigens and autoantibodies, such as oxidized low-density lipoprotein and beta2-glycoprotein, which may play a pathogenetic role in atherosclerosis progression.
Collapse
Affiliation(s)
| | | | | | - Roberto Manetti
- Department of Medical, Surgical and Pharmacology, University of Sassari, 07100 Sassari, Italy; (G.L.E.); (F.C.)
| |
Collapse
|
6
|
Singh MK, Shin Y, Han S, Ha J, Tiwari PK, Kim SS, Kang I. Molecular Chaperonin HSP60: Current Understanding and Future Prospects. Int J Mol Sci 2024; 25:5483. [PMID: 38791521 PMCID: PMC11121636 DOI: 10.3390/ijms25105483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Molecular chaperones are highly conserved across evolution and play a crucial role in preserving protein homeostasis. The 60 kDa heat shock protein (HSP60), also referred to as chaperonin 60 (Cpn60), resides within mitochondria and is involved in maintaining the organelle's proteome integrity and homeostasis. The HSP60 family, encompassing Cpn60, plays diverse roles in cellular processes, including protein folding, cell signaling, and managing high-temperature stress. In prokaryotes, HSP60 is well understood as a GroEL/GroES complex, which forms a double-ring cavity and aids in protein folding. In eukaryotes, HSP60 is implicated in numerous biological functions, like facilitating the folding of native proteins and influencing disease and development processes. Notably, research highlights its critical involvement in sustaining oxidative stress and preserving mitochondrial integrity. HSP60 perturbation results in the loss of the mitochondria integrity and activates apoptosis. Currently, numerous clinical investigations are in progress to explore targeting HSP60 both in vivo and in vitro across various disease models. These studies aim to enhance our comprehension of disease mechanisms and potentially harness HSP60 as a therapeutic target for various conditions, including cancer, inflammatory disorders, and neurodegenerative diseases. This review delves into the diverse functions of HSP60 in regulating proteo-homeostasis, oxidative stress, ROS, apoptosis, and its implications in diseases like cancer and neurodegeneration.
Collapse
Affiliation(s)
- Manish Kumar Singh
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Centre for Genomics, SOS Zoology, Jiwaji University, Gwalior 474011, India;
| | - Yoonhwa Shin
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Sunhee Han
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Joohun Ha
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Pramod K. Tiwari
- Centre for Genomics, SOS Zoology, Jiwaji University, Gwalior 474011, India;
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Insug Kang
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.K.S.); (Y.S.); (S.H.); (J.H.)
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
7
|
Schroeder HT, De Lemos Muller CH, Heck TG, Krause M, Homem de Bittencourt PI. Heat shock response during the resolution of inflammation and its progressive suppression in chronic-degenerative inflammatory diseases. Cell Stress Chaperones 2024; 29:116-142. [PMID: 38244765 PMCID: PMC10939074 DOI: 10.1016/j.cstres.2024.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024] Open
Abstract
The heat shock response (HSR) is a crucial biochemical pathway that orchestrates the resolution of inflammation, primarily under proteotoxic stress conditions. This process hinges on the upregulation of heat shock proteins (HSPs) and other chaperones, notably the 70 kDa family of heat shock proteins, under the command of the heat shock transcription factor-1. However, in the context of chronic degenerative disorders characterized by persistent low-grade inflammation (such as insulin resistance, obesity, type 2 diabetes, nonalcoholic fatty liver disease, and cardiovascular diseases) a gradual suppression of the HSR does occur. This work delves into the mechanisms behind this phenomenon. It explores how the Western diet and sedentary lifestyle, culminating in the endoplasmic reticulum stress within adipose tissue cells, trigger a cascade of events. This cascade includes the unfolded protein response and activation of the NOD-like receptor pyrin domain-containing protein-3 inflammasome, leading to the emergence of the senescence-associated secretory phenotype and the propagation of inflammation throughout the body. Notably, the activation of the NOD-like receptor pyrin domain-containing protein-3 inflammasome not only fuels inflammation but also sabotages the HSR by degrading human antigen R, a crucial mRNA-binding protein responsible for maintaining heat shock transcription factor-1 mRNA expression and stability on heat shock gene promoters. This paper underscores the imperative need to comprehend how chronic inflammation stifles the HSR and the clinical significance of evaluating the HSR using cost-effective and accessible tools. Such understanding is pivotal in the development of innovative strategies aimed at the prevention and treatment of these chronic inflammatory ailments, which continue to take a heavy toll on global health and well-being.
Collapse
Affiliation(s)
- Helena Trevisan Schroeder
- Laboratory of Cellular Physiology (FisCel), Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Carlos Henrique De Lemos Muller
- Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX), Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Thiago Gomes Heck
- Post Graduate Program in Integral Health Care (PPGAIS-UNIJUÍ/UNICRUZ/URI), Regional University of Northwestern Rio Grande Do Sul State (UNIJUI) and Post Graduate Program in Mathematical and Computational Modeling (PPGMMC), UNIJUI, Ijuí, Rio Grande do Sul, Brazil
| | - Mauricio Krause
- Laboratory of Inflammation, Metabolism and Exercise Research (LAPIMEX), Department of Physiology, ICBS, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Paulo Ivo Homem de Bittencourt
- Laboratory of Cellular Physiology (FisCel), Department of Physiology, Institute of Basic Health Sciences (ICBS), Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
8
|
Tayal S, Bhatnagar S. Role of molecular mimicry in the SARS-CoV-2-human interactome for pathogenesis of cardiovascular diseases: An update to ImitateDB. Comput Biol Chem 2023; 106:107919. [PMID: 37463554 DOI: 10.1016/j.compbiolchem.2023.107919] [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: 01/15/2023] [Revised: 06/13/2023] [Accepted: 07/06/2023] [Indexed: 07/20/2023]
Abstract
Mimicry of host proteins is a strategy employed by pathogens to hijack host functions. Domain and motif mimicry was explored in the experimental and predicted SARS-CoV-2-human interactome. The host first interactor proteins were also added to capture the continuum of the interactions. The domains and motifs of the proteins were annotated using NCBI CD Search and ScanProsite, respectively. Host and pathogen proteins with a common host interactor and similar domain/motif constitute a mimicry pair indicating global structural similarity (domain mimicry pair; DMP) or local sequence similarity (motif mimicry pair; MMP). 593 DMPs and 7,02,472 MMPs were determined. AAA, DEXDc and Macro domains were frequent among DMPs whereas glycosylation, myristoylation and RGD motifs were abundant among MMP. The proteins involved in mimicry were visualised as a SARS-CoV-2 mimicry interaction network. The host proteins were enriched in multiple CVD pathways indicating the role of mimicry in COVID-19 associated CVDs. Bridging nodes were identified as potential drug targets. Approved antihypertensive and anti-inflammatory drugs are proposed for repurposing against COVID-19 associated CVDs. The SARS-CoV-2 mimicry data has been updated in ImitateDB (http://imitatedb.sblab-nsit.net/SARSCoV2Mimicry). Determination of key mechanisms, proteins, pathways, drug targets and repurposing candidates is critical for developing therapeutics for SARS CoV-2 associated CVDs.
Collapse
Affiliation(s)
- Sonali Tayal
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi 110078, India
| | - Sonika Bhatnagar
- Computational and Structural Biology Laboratory, Department of Biological Sciences and Engineering, Netaji Subhas University of Technology, Dwarka, New Delhi 110078, India.
| |
Collapse
|
9
|
Desai SR, Dhindsa DS, Ko YA, Sandesara PB, Mehta A, Liu C, Tahhan AS, Hayek SS, Ejaz K, Hooda A, Alkhoder A, Islam SJ, Rogers SC, Beshiri A, Murtagh G, Kim JH, Wilson P, Almuwaqqat Z, Sperling LS, Quyyumi AA. Aggregate Clinical and Biomarker-Based Model Predicts Adverse Outcomes in Patients With Coronary Artery Disease. Am J Cardiol 2023; 203:315-324. [PMID: 37517126 PMCID: PMC10900119 DOI: 10.1016/j.amjcard.2023.06.115] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/22/2023] [Accepted: 06/29/2023] [Indexed: 08/01/2023]
Abstract
Despite guideline-based therapy, patients with coronary artery disease (CAD) are at widely variable risk for cardiovascular events. This variability demands a more individualized risk assessment. Herein, we evaluate the prognostic value of 6 biomarkers: high-sensitivity C-reactive protein, heat shock protein-70, fibrin degradation products, soluble urokinase plasminogen activator receptor, high-sensitivity troponin I, and B-type natriuretic peptide. We then develop a multi-biomarker-based cardiovascular event prediction model for patients with stable CAD. In total, 3,115 subjects with stable CAD who underwent cardiac catheterization at Emory (mean age 62.8 years, 17% Black, 35% female, 57% obstructive CAD, 31% diabetes mellitus) were randomized into a training cohort to identify biomarker cutoff values and a validation cohort for prediction assessment. Main outcomes included (1) all-cause death and (2) a composite of cardiovascular death and nonfatal myocardial infarction (MI) within 5 years. Elevation of each biomarker level was associated with higher event rates in the training cohort. A biomarker risk score was created using optimal cutoffs, ranging from 0 to 6 for each biomarker exceeding its cutoff. In the validation cohort, each unit increase in the biomarker risk score was independently associated with all-cause death (hazard ratio 1.62, 95% confidence interval [CI] 1.45 to 1.80) and cardiovascular death/MI (hazard ratio 1.52, 95% CI 1.35 to 1.71). A biomarker risk prediction model for cardiovascular death/MI improved the c-statistic (∆ 6.4%, 95% CI 3.9 to 8.8) and net reclassification index by 31.1% (95% CI 24 to 37), compared with clinical risk factors alone. Integrating multiple biomarkers with clinical variables refines cardiovascular risk assessment in patients with CAD.
Collapse
Affiliation(s)
- Shivang R Desai
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Devinder S Dhindsa
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Yi-An Ko
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Pratik B Sandesara
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Anurag Mehta
- Department of Preventive Cardiology, VCU Health Pauley Heart Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Chang Liu
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia; Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Ayman S Tahhan
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Salim S Hayek
- Department of Medicine, Division of Cardiology, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Kiran Ejaz
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Ananya Hooda
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Ayman Alkhoder
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Shabatun J Islam
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Steven C Rogers
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Agim Beshiri
- Diagnostics Division, Abbott Laboratories, North Chicago, Illinois
| | - Gillian Murtagh
- Diagnostics Division, Abbott Laboratories, North Chicago, Illinois
| | - Jonathan H Kim
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Peter Wilson
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Zakaria Almuwaqqat
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Laurence S Sperling
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia
| | - Arshed A Quyyumi
- Division of Cardiology, Department of Medicine, Emory Clinical Cardiovascular Research Institute, Emory University School of Medicine, Atlanta, Georgia.
| |
Collapse
|
10
|
Keen A, Zhang F, Reader JS, Tzima E. Proteostasis and resilience in the mechanically-stressed vascular endothelium. CURRENT OPINION IN PHYSIOLOGY 2023; 34:None. [PMID: 40124949 PMCID: PMC11922819 DOI: 10.1016/j.cophys.2023.100673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Endothelial homeostasis is a central feature of vascular health. The vascular endothelium is under constant mechanical stress resulting from blood flow and, therefore, requires a high degree of resilience to adapt to stresses and resist development of disease. In this review, we discuss the molecular mechanisms by which the endothelium maintains proteostasis in response to haemodynamic forces by regulating three key areas: protein synthesis, recycling and degradation.
Collapse
Affiliation(s)
- Adam Keen
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - Feiran Zhang
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | - John S Reader
- Wellcome Centre for Human Genetics, Radcliffe Department of Medicine, University of Oxford, United Kingdom
| | | |
Collapse
|
11
|
Wong SHD, Yin B, Li Z, Yuan W, Zhang Q, Xie X, Tan Y, Wong N, Zhang K, Bian L. Mechanical manipulation of cancer cell tumorigenicity via heat shock protein signaling. SCIENCE ADVANCES 2023; 9:eadg9593. [PMID: 37418519 PMCID: PMC10328411 DOI: 10.1126/sciadv.adg9593] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 06/01/2023] [Indexed: 07/09/2023]
Abstract
Biophysical cues of rigid tumor matrix play a critical role in cancer cell malignancy. We report that stiffly confined cancer cells exhibit robust growth of spheroids in the stiff hydrogel that exerts substantial confining stress on the cells. The stressed condition activated Hsp (heat shock protein)-signal transducer and activator of transcription 3 signaling via the transient receptor potential vanilloid 4-phosphatidylinositol 3-kinase/Akt axis, thereby up-regulating the expression of the stemness-related markers in cancer cells, whereas these signaling activities were suppressed in cancer cells cultured in softer hydrogels or stiff hydrogels with stress relief or Hsp70 knockdown/inhibition. This mechanopriming based on three-dimensional culture enhanced cancer cell tumorigenicity and metastasis in animal models upon transplantation, and pharmaceutically inhibiting Hsp70 improved the anticancer efficacy of chemotherapy. Mechanistically, our study reveals the crucial role of Hsp70 in regulating cancer cell malignancy under mechanically stressed conditions and its impacts on cancer prognosis-related molecular pathways for cancer treatments.
Collapse
Affiliation(s)
- Siu Hong Dexter Wong
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, P. R. China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, P. R. China
| | - Bohan Yin
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, P. R. China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, P. R. China
| | - Zhuo Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong 999077, P. R. China
| | - Weihao Yuan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong 999077, P. R. China
| | - Qin Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, P. R. China
| | - Xian Xie
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Hong Kong 999077, P. R. China
| | - Youhua Tan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong 999077, P. R. China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518057, P. R. China
| | - Nathalie Wong
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong 999077, P. R. China
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P. R. China
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, P. R. China
| |
Collapse
|
12
|
Moreno-Gonzalez MA, Ortega-Rivera OA, Steinmetz NF. Two decades of vaccine development against atherosclerosis. NANO TODAY 2023; 50:101822. [PMID: 37860053 PMCID: PMC10586238 DOI: 10.1016/j.nantod.2023.101822] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Atherosclerosis is an immune-mediated chronic inflammatory disease that leads to the development of fatty plaques in the arterial walls, ultimately increasing the risk of thrombosis, stroke, and myocardial infarction. The immune response in this complex disease is both atheroprotective and pro-atherogenic, involving both innate and adaptive immunity. Current treatments include the adjustment of lifestyle factors, cholesterol-lowering drugs such as statins, and immunotherapy, whereas vaccine development has received comparatively little attention. In this review, we discuss the potential of antigen-specific vaccination as a preventative approach based on more than 20 years of research and innovation. Vaccination targets include proteins that are more abundant in atherosclerotic patients, such as oxidized low-density lipoprotein (LDL), apolipoprotein B-100, proprotein convertase subtilisin/kexin type-9 serine protease (PCSK9), cholesteryl ester transfer protein (CETP), and heat shock proteins HSP60 and HSP65. Immunization with such proteins or their peptide epitopes has been shown to induce T-cell activation, produce antigen-specific antibodies, reduce the size of atherosclerotic lesions, and/or reduce serum cholesterol levels. Vaccination against atherosclerosis therefore offers a new strategy to address the burden on healthcare systems caused by cardiovascular disease, the leading cause of death worldwide.
Collapse
Affiliation(s)
- Miguel A. Moreno-Gonzalez
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
| | - Oscar A. Ortega-Rivera
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92039, USA
- Institute for Materials Discovery and Design, University of California-San Diego, La Jolla, CA 92039, USA
- Department of Bioengineering, University of California-San Diego, La Jolla, CA 92039, USA
- Department of Radiology, University of California-San Diego, La Jolla, CA 92039, USA
- Moores Cancer Center, University of California-San Diego, La Jolla, CA 92039, USA
| |
Collapse
|
13
|
Patnaik S, Nathan S, Kar B, Gregoric ID, Li YP. The Role of Extracellular Heat Shock Proteins in Cardiovascular Diseases. Biomedicines 2023; 11:1557. [PMID: 37371652 DOI: 10.3390/biomedicines11061557] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/19/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
In the early 1960s, heat shock proteins (HSPs) were first identified as vital intracellular proteinaceous components that help in stress physiology and reprogram the cellular responses to enable the organism's survival. By the early 1990s, HSPs were detected in extracellular spaces and found to activate gamma-delta T-lymphocytes. Subsequent investigations identified their association with varied disease conditions, including autoimmune disorders, diabetes, cancer, hepatic, pancreatic, and renal disorders, and cachexia. In cardiology, extracellular HSPs play a definite, but still unclear, role in atherosclerosis, acute coronary syndromes, and heart failure. The possibility of HSP-targeted novel molecular therapeutics has generated much interest and hope in recent years. In this review, we discuss the role of Extracellular Heat Shock Proteins (Ec-HSPs) in various disease states, with a particular focus on cardiovascular diseases.
Collapse
Affiliation(s)
- Soumya Patnaik
- Division of Cardiology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Sriram Nathan
- Department of Advanced Cardiopulmonary Therapies and Transplantation, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Biswajit Kar
- Department of Advanced Cardiopulmonary Therapies and Transplantation, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Igor D Gregoric
- Department of Advanced Cardiopulmonary Therapies and Transplantation, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Yi-Ping Li
- Division of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
14
|
Alehossein P, Taheri M, Tayefeh Ghahremani P, Dakhlallah D, Brown CM, Ishrat T, Nasoohi S. Transplantation of Exercise-Induced Extracellular Vesicles as a Promising Therapeutic Approach in Ischemic Stroke. Transl Stroke Res 2023; 14:211-237. [PMID: 35596116 DOI: 10.1007/s12975-022-01025-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/06/2022] [Accepted: 04/15/2022] [Indexed: 11/24/2022]
Abstract
Clinical evidence affirms physical exercise is effective in preventive and rehabilitation approaches for ischemic stroke. This sustainable efficacy is independent of cardiovascular risk factors and associates substantial reprogramming in circulating extracellular vesicles (EVs). The intricate journey of pluripotent exercise-induced EVs from parental cells to the whole-body and infiltration to cerebrovascular entity offers several mechanisms to reduce stroke incidence and injury or accelerate the subsequent recovery. This review delineates the potential roles of EVs as prospective effectors of exercise. The candidate miRNA and peptide cargo of exercise-induced EVs with both atheroprotective and neuroprotective characteristics are discussed, along with their presumed targets and pathway interactions. The existing literature provides solid ground to hypothesize that the rich vesicles link exercise to stroke prevention and rehabilitation. However, there are several open questions about the exercise stressors which may optimally regulate EVs kinetic and boost brain mitochondrial adaptations. This review represents a novel perspective on achieving brain fitness against stroke through transplantation of multi-potential EVs generated by multi-parental cells, which is exceptionally reachable in an exercising body.
Collapse
Affiliation(s)
- Parsa Alehossein
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd., Chamran Hwy., PO: 19615-1178, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Taheri
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd., Chamran Hwy., PO: 19615-1178, Tehran, Iran
- Faculty of Sport Sciences and Health, Shahid Beheshti University, Tehran, Iran
| | - Pargol Tayefeh Ghahremani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd., Chamran Hwy., PO: 19615-1178, Tehran, Iran
| | - Duaa Dakhlallah
- Institute of Global Health and Human Ecology, School of Sciences & Engineering, The American University of Cairo, Cairo, Egypt
| | - Candice M Brown
- Department of Neuroscience, School of Medicine, and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Sanaz Nasoohi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Daneshjoo Blvd., Chamran Hwy., PO: 19615-1178, Tehran, Iran.
| |
Collapse
|
15
|
Chen YC, Chen KF, Andrew Lin KY, Su HP, Wu DN, Lin CH. Evaluation of toxicity of polystyrene microplastics under realistic exposure levels in human vascular endothelial EA.hy926 cells. CHEMOSPHERE 2023; 313:137582. [PMID: 36529175 DOI: 10.1016/j.chemosphere.2022.137582] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
Microplastics (MPs) have emerged as a global concern, with a recent study being the first to detect them in the bloodstream of healthy people. However, precise information regarding the toxic effects of MPs on the human vascular system is currently lacking. In this study, we used human vascular endothelial EA. hy926 cells to examine the toxic potential of polystyrene MPs (PSMPs) under realistic blood concentrations. Our findings indicated that PSMPs can cause oxidative stress by reducing the expression of antioxidants, thereby leading to apoptotic cytotoxicity in EA. hy926 cells. Furthermore, the protective potential of heat shock proteins can be reduced by PSMPs. PSMP-induced apoptosis might also lower the expression of rho-associated protein kinase-1 and nuclear factor-κB expression, thus dampening LRR- and pyrin domain-containing protein 3 in EA. hy926 cells. Moreover, we observed that PSMPs induce vascular barrier dysfunction via the depletion of zonula occludens-1 protein. However, although protein expression of the nuclear hormone receptor 77 was inhibited, no significant increase in ectin-like oxidized low-density lipoprotein receptor-1 was noted in PSMP-treated EA. hy926 cells. These results demonstrate that exposure to PSMPs may not sufficiently increase the risk of developing atherosclerosis. Overall, our research signifies that exposure to realistic blood concentrations of PSMPs is associated with low atherosclerotic cardiovascular risk in humans.
Collapse
Affiliation(s)
- Yi-Chun Chen
- Department of Biotechnology, National Formosa University, Yunlin, 63208, Taiwan; Department of Science and Environment Studies, The Education University of Hong Kong, New Territories, Hong Kong
| | - Ku-Fan Chen
- Department of Civil Engineering, National Chi Nan University, Nantou, 54561, Taiwan
| | - Kun-Yi Andrew Lin
- Department of Environmental Engineering, National Chung Hsing University, Taichung, 40227, Taiwan
| | - Han-Pang Su
- Third Research Division, Taiwan Research Institute, New Taipei City, 251030, Taiwan
| | - Dong-Ni Wu
- Department of Biotechnology, National Formosa University, Yunlin, 63208, Taiwan
| | - Chia-Hua Lin
- Department of Biotechnology, National Formosa University, Yunlin, 63208, Taiwan.
| |
Collapse
|
16
|
Niinuma SA, Lubbad L, Lubbad W, Moin ASM, Butler AE. The Role of Heat Shock Proteins in the Pathogenesis of Polycystic Ovarian Syndrome: A Review of the Literature. Int J Mol Sci 2023; 24:ijms24031838. [PMID: 36768170 PMCID: PMC9915177 DOI: 10.3390/ijms24031838] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/19/2023] Open
Abstract
Polycystic ovarian syndrome (PCOS) is the most common endocrine disorder in women of reproductive age and post-menopausal women. PCOS is a multifactorial heterogeneous disorder associated with a variety of etiologies, outcomes, and clinical manifestations. However, the pathophysiology of PCOS is still unclear. Heat shock proteins (HSPs) have recently been investigated for their role in the pathogenesis of PCOS. HSPs are a class of proteins that act as molecular chaperones and maintain cellular proteostasis. More recently, their actions beyond that of molecular chaperones have highlighted their pathogenic role in several diseases. In PCOS, different HSP family members show abnormal expression that affects the proliferation and apoptotic rates of ovarian cells as well as immunological processes. HSP dysregulation in the ovaries of PCOS subjects leads to a proliferation/apoptosis imbalance that mechanistically impacts follicle stage development, resulting in polycystic ovaries. Moreover, HSPs may play a role in the pathogenesis of PCOS-associated conditions. Recent studies on HSP activity during therapeutic interventions for PCOS suggest that modulating HSP activity may lead to novel treatment strategies. In this review, we summarize what is currently known regarding the role of HSPs in the pathogenesis of PCOS and their potential role in the treatment of PCOS, and we outline areas for future research.
Collapse
Affiliation(s)
- Sara Anjum Niinuma
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen 15503, Bahrain
| | - Laila Lubbad
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen 15503, Bahrain
| | - Walaa Lubbad
- School of Medicine, Royal College of Surgeons in Ireland Bahrain, Busaiteen 15503, Bahrain
| | - Abu Saleh Md Moin
- Research Department, Royal College of Surgeons in Ireland Bahrain, Busaiteen 15503, Bahrain
| | - Alexandra E. Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Busaiteen 15503, Bahrain
- Correspondence: or ; Tel.: +973-66760313
| |
Collapse
|
17
|
Saha A, Ahmed S. The Link Between Heat Shock Proteins, Renin-Angiotensin System, and the Coagulation Cascade in the Pathogenesis of the Coronavirus-19 Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1409:161-171. [PMID: 35882774 DOI: 10.1007/5584_2022_735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
INTRODUCTION Understanding the pathogenesis of COVID-19 is integral for its successful treatment. METHODS Available literature on the relationship between COVID-19, heat shock proteins (HSP), and the renin-angiotensin-aldosterone (RAAS) system were searched and used to hypothesize how HSP can be targeted in COVID-19. RESULTS During SARS-CoV-2 cellular entry, the ACE-2 receptor is downregulated. This leads to the augmentation of angiotensin-2/AT1 receptor axis along with attenuation of the ACE-2/angiotensin1-7/Mas axis. Heat shock proteins are key stabilizing molecules in various pathways.In the heart and vessels, HSP-90 and HSP-60 can facilitate angiotensin-2-mediated myocardial injury and endothelial cell activation. HSP-60-TLR4/CD14 complex formation stabilizes IκB-kinase (IKK) potentiating NF-κB activation. HSPs in lungs and kidneys have antioxidant, vasodilatory, and anti-inflammatory actions and may be protective against the effects of RAAS. Stress-induced HSP-70 has a role in complement-mediated microvascular injury such as has been demonstrated in COVID-19. SARS-CoV-2 can induce autophagy via Beclin-1 and ER (endoplasmic reticular) stress via BIP. These two can be potential targets in the HSP environment. CONCLUSION Various HSP molecules can modulate the effects of the renin-angiotensin-aldosterone (RAAS) system and thus may have a potential role in the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Aritra Saha
- Department of General Medicine, Assam Medical College and Hospital, Dibrugarh, India
| | - Sakir Ahmed
- Department of Clinical Immunology & Rheumatology, Kalinga Institute of Medical Sciences (KIMS), KIIT University, Bhubaneswar, India.
| |
Collapse
|
18
|
Suica VI, Uyy E, Ivan L, Boteanu RM, Cerveanu-Hogas A, Hansen R, Antohe F. Cardiac Alarmins as Residual Risk Markers of Atherosclerosis under Hypolipidemic Therapy. Int J Mol Sci 2022; 23:ijms231911174. [PMID: 36232476 PMCID: PMC9569654 DOI: 10.3390/ijms231911174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 12/01/2022] Open
Abstract
Increased levels of low-density lipoproteins are the main risk factor in the initiation and progression of atherosclerosis. Although statin treatment can effectively lower these levels, there is still a residual risk of cardiovascular events. We hypothesize that a specific panel of stress-sensing molecules (alarmins) could indicate the persistence of silent atherosclerosis residual risk. New Zealand White rabbits were divided into: control group (C), a group that received a high-fat diet for twelve weeks (Au), and a treated hyperlipidemic group with a lipid diet for eight weeks followed by a standard diet and hypolipidemic treatment (atorvastatin and PCSK9 siRNA-inhibitor) for four weeks (Asi). Mass spectrometry experiments of left ventricle lysates were complemented by immunologic and genomic studies to corroborate the data. The hyperlipidemic diet determined a general alarmin up-regulation tendency over the C group. A significant spectral abundance increase was measured for specific heat shock proteins, S100 family members, HMGB1, and Annexin A1. The hypolipidemic treatment demonstrated a reversed regulation trend with non-significant spectral alteration over the C group for some of the identified alarmins. Our study highlights the discriminating potential of alarmins in hyperlipidemia or following hypolipidemic treatment. Data are available via ProteomeXchange with identifier PXD035692.
Collapse
Affiliation(s)
- Viorel I. Suica
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
| | - Elena Uyy
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
| | - Luminita Ivan
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
| | - Raluca M. Boteanu
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
| | - Aurel Cerveanu-Hogas
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
| | - Rune Hansen
- SINTEF Digital, 7465 Trondheim, Norway
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Felicia Antohe
- Institute of Cellular Biology and Pathology “Nicolae Simionescu”, 050568 Bucharest, Romania
- Correspondence: ; Tel.: +40-213194518
| |
Collapse
|
19
|
Hu C, Yang J, Qi Z, Wu H, Wang B, Zou F, Mei H, Liu J, Wang W, Liu Q. Heat shock proteins: Biological functions, pathological roles, and therapeutic opportunities. MedComm (Beijing) 2022; 3:e161. [PMID: 35928554 PMCID: PMC9345296 DOI: 10.1002/mco2.161] [Citation(s) in RCA: 222] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 12/12/2022] Open
Abstract
The heat shock proteins (HSPs) are ubiquitous and conserved protein families in both prokaryotic and eukaryotic organisms, and they maintain cellular proteostasis and protect cells from stresses. HSP protein families are classified based on their molecular weights, mainly including large HSPs, HSP90, HSP70, HSP60, HSP40, and small HSPs. They function as molecular chaperons in cells and work as an integrated network, participating in the folding of newly synthesized polypeptides, refolding metastable proteins, protein complex assembly, dissociating protein aggregate dissociation, and the degradation of misfolded proteins. In addition to their chaperone functions, they also play important roles in cell signaling transduction, cell cycle, and apoptosis regulation. Therefore, malfunction of HSPs is related with many diseases, including cancers, neurodegeneration, and other diseases. In this review, we describe the current understandings about the molecular mechanisms of the major HSP families including HSP90/HSP70/HSP60/HSP110 and small HSPs, how the HSPs keep the protein proteostasis and response to stresses, and we also discuss their roles in diseases and the recent exploration of HSP related therapy and diagnosis to modulate diseases. These research advances offer new prospects of HSPs as potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Chen Hu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Jing Yang
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Ziping Qi
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Hong Wu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Beilei Wang
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Fengming Zou
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
| | - Husheng Mei
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Jing Liu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Wenchao Wang
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
| | - Qingsong Liu
- Anhui Province Key Laboratory of Medical Physics and TechnologyInstitute of Health and Medical TechnologyHefei Institutes of Physical ScienceChinese Academy of SciencesHefeiAnhuiP. R. China
- Hefei Cancer HospitalChinese Academy of SciencesHefeiAnhuiP. R. China
- University of Science and Technology of ChinaHefeiAnhuiP. R. China
- Precision Medicine Research Laboratory of Anhui ProvinceHefeiAnhuiP. R. China
| |
Collapse
|
20
|
Zieleniewska NA, Kazberuk M, Chlabicz M, Eljaszewicz A, Kamiński K. Trained Immunity as a Trigger for Atherosclerotic Cardiovascular Disease-A Literature Review. J Clin Med 2022; 11:jcm11123369. [PMID: 35743439 PMCID: PMC9224533 DOI: 10.3390/jcm11123369] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/05/2022] [Accepted: 06/09/2022] [Indexed: 11/17/2022] Open
Abstract
Atherosclerosis remains the leading cause of cardiovascular diseases and represents a primary public health challenge. This chronic state may lead to a number of life-threatening conditions, such as myocardial infarction and stroke. Lipid metabolism alterations and inflammation remain at the forefront of the pathogenesis of atherosclerotic cardiovascular disease, but the overall mechanism is not yet fully understood. Recently, significant effects of trained immunity on atherosclerotic plaque formation and development have been reported. An increased reaction to restimulation with the same stimulator is a hallmark of the trained innate immune response. The impact of trained immunity is a prominent factor in both acute and chronic coronary syndrome, which we outline in this review.
Collapse
Affiliation(s)
- Natalia Anna Zieleniewska
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, 15-259 Bialystok, Poland; (N.A.Z.); (M.C.)
- Department of Cardiology, Teaching University Hospital of Białystok, 15-259 Bialystok, Poland
| | - Małgorzata Kazberuk
- Scientific Group of Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, 15-259 Bialystok, Poland;
| | - Małgorzata Chlabicz
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, 15-259 Bialystok, Poland; (N.A.Z.); (M.C.)
- Department of Invasive Cardiology, Teaching University Hospital of Białystok, 15-259 Bialystok, Poland
| | - Andrzej Eljaszewicz
- Department of Regenerative Medicine and Immune Regulation, Medical University of Białystok, 15-259 Bialystok, Poland;
| | - Karol Kamiński
- Department of Population Medicine and Lifestyle Diseases Prevention, Medical University of Białystok, 15-259 Bialystok, Poland; (N.A.Z.); (M.C.)
- Department of Cardiology, Teaching University Hospital of Białystok, 15-259 Bialystok, Poland
- Correspondence:
| |
Collapse
|
21
|
Sun L, Zhang W, Zhao L, Zhao Y, Wang F, Lew AM, Xu Y. Self-Tolerance of Vascular Tissues Is Broken Down by Vascular Dendritic Cells in Response to Systemic Inflammation to Initiate Regional Autoinflammation. Front Immunol 2022; 13:823853. [PMID: 35154143 PMCID: PMC8825784 DOI: 10.3389/fimmu.2022.823853] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/03/2022] [Indexed: 12/19/2022] Open
Abstract
The correlation of infections with vascular autoinflammatory diseases such as vasculitis and atherosclerosis has been long recognized, and progressive inflammation with the formation of tertiary lymphoid organs in arterial adventitia intensively studied, the immunological basis of the nondiseased vasculatures that predispose to subsequent vascular autoimmunity during inflammation, however, is not well characterized. Here, we investigated the vascular immunity in situ of steady-state C57BL/6 mice and found that healthy vascular tissues contained a comprehensive set of immune cells with relatively higher proportion of innate components than lymphoid organs. Notably, a complete set of dendritic cell (DC) subsets was observed with monocyte-derived DCs (moDCs) constituting a major proportion; this is in contrast to moDCs being considered rare in the steady state. Interestingly, these vascular DCs constitutively expressed more suppressive factors with cDC1 for PD-L1 and moDCs for IL-10; this is concordant with the inhibitive phenotype of T cells in normal vascular tissues. The immunotolerant state of the vascular tissues, however, was readily eroded by systemic inflammation, demonstrated by the upregulation of proinflammatory cytokines and enhanced antigen presentation by vascular DCs to activate both cellular and humoral immunity in situ, which ultimately led to vascular destruction. Different vascular DC subsets elicited selective effects: moDCs were potent cytokine producers and B-cell activators, whereas cDCs, particularly, cDC1, were efficient at presenting antigens to stimulate T cells. Together, we unveil regional immunological features of vascular tissues to explain their dual facets under physiological versus pathological conditions for the better understanding and treatment of cardiovascular autoinflammation.
Collapse
Affiliation(s)
- Li Sun
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Wenjie Zhang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Lin Zhao
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Yanfang Zhao
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Fengge Wang
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| | - Andrew M Lew
- The Walter & Eliza Hall Institute of Medical Research and Dept of Microbiology & Immunology, University of Melbourne, Parkville, VIC, Australia
| | - Yuekang Xu
- Anhui Provincial Key Laboratory for Conservation and Exploitation of Biological Resources, College of Life Science, Anhui Normal University, Wuhu, China
| |
Collapse
|
22
|
Razeghian-Jahromi I, Karimi Akhormeh A, Razmkhah M, Zibaeenezhad MJ. Immune system and atherosclerosis: Hostile or friendly relationship. Int J Immunopathol Pharmacol 2022; 36:3946320221092188. [PMID: 35410514 PMCID: PMC9009140 DOI: 10.1177/03946320221092188] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/16/2022] [Indexed: 11/21/2022] Open
Abstract
Coronary artery disease has remained a major health challenge despite enormous progress in prevention, diagnosis, and treatment strategies. Formation of atherosclerotic plaque is a chronic process that is developmentally influenced by intrinsic and extrinsic determinants. Inflammation triggers atherosclerosis, and the fundamental element of inflammation is the immune system. The immune system involves in the atherosclerosis process by a variety of immune cells and a cocktail of mediators. It is believed that almost all main components of this system possess a profound contribution to the atherosclerosis. However, they play contradictory roles, either protective or progressive, in different stages of atherosclerosis progression. It is evident that monocytes are the first immune cells appeared in the atherosclerotic lesion. With the plaque growth, other types of the immune cells such as mast cells, and T lymphocytes are gradually involved. Each cell releases several cytokines which cause the recruitment of other immune cells to the lesion site. This is followed by affecting the expression of other cytokines as well as altering certain signaling pathways. All in all, a mix of intertwined interactions determine the final outcome in terms of mild or severe manifestations, either clinical or subclinical. Therefore, it is of utmost importance to precisely understand the kind and degree of contribution which is made by each immune component in order to stop the growing burden of cardiovascular morbidity and mortality. In this review, we present a comprehensive appraisal on the role of immune cells in the atherosclerosis initiation and development.
Collapse
Affiliation(s)
| | - Ali Karimi Akhormeh
- Cardiovascular Research Center, Shiraz University of Medical
Sciences, Shiraz, Iran
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer
Research, Shiraz University of Medical
Sciences, Shiraz, Iran
| | | |
Collapse
|
23
|
Hakhamaneshi MS, Abdolahi A, Vahabzadeh Z, Abdi M, Andalibi P. Toll-Like Receptor 4: A Macrophage Cell Surface Receptor Is Activated By Trimethylamine-N-Oxide. CELL JOURNAL 2021; 23:516-522. [PMID: 34837678 PMCID: PMC8588815 DOI: 10.22074/cellj.2021.7849] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 02/03/2021] [Indexed: 01/06/2023]
Abstract
OBJECTIVE Trimethylamine-N-Oxide (TMAO) is considered as a risk factor for atherosclerosis which further leads to inflammation during atherosclerosis. The exact mechanism(s) by which TMAO induces the inflammatory reactions remains to be determined. TMAO can cause the endoplasmic reticulum (ER) stress that triggers activation of Toll-Like Receptors (TLRs). In macrophages, this process stimulates the production of proinflammatory cytokines. This study designed to evaluate the expression level of TLR4 in TMAO-treated macrophages. MATERIALS AND METHODS In this experimental study, different concentrations of TMAO (37.5, 75, 150, and 300 μM) were exposed to murine macrophage (J774A.1 cell line) for 8, 18, 24, and 48 hours. The cells were also treated with 2.5 mM of 4-phenyl butyric acid as well as 2μg/ml of tunicamycin respectively as negative and positive controls for inducing ER-stress. We measured the viability of treated cells by the MTT test. Besides, the expression levels of TLR4 gene and protein were evaluated using western blotting and reverse transcription- quantitative polymerase chain reaction (RT-qPCR) analysis. One-Way ANOVA was used for statistical analysis. RESULTS No cell death was observed in treated cells. The cells treated with 150 and 300 μM doses of TMAO for 24 hours showed a significant elevation in the protein and/or mRNA levels of TLR4 when compared to normal control or tunicamycin-treated cells. CONCLUSION Our results may in part elucidate the mechanism by which TMAO induces the macrophage inflammatory reactions in response to the induction of ER stress, similar to what happens during atherosclerosis. It also provides documentation to support the direct contribution of TLR4 in TMAO-induced inflammation.
Collapse
Affiliation(s)
| | - Alina Abdolahi
- Department of Molecular Medicine and Genetics, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Zakaria Vahabzadeh
- Department of Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran. .,Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Abdi
- Cellular and Molecular Research Centre, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Pedram Andalibi
- Department of Biochemistry, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
24
|
Bartoli-Leonard F, Zimmer J, Aikawa E. Innate and adaptive immunity: the understudied driving force of heart valve disease. Cardiovasc Res 2021; 117:2506-2524. [PMID: 34432007 PMCID: PMC8783388 DOI: 10.1093/cvr/cvab273] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
Calcific aortic valve disease (CAVD), and its clinical manifestation that is calcific aortic valve stenosis, is the leading cause for valve disease within the developed world, with no current pharmacological treatment available to delay or halt its progression. Characterized by progressive fibrotic remodelling and subsequent pathogenic mineralization of the valve leaflets, valve disease affects 2.5% of the western population, thus highlighting the need for urgent intervention. Whilst the pathobiology of valve disease is complex, involving genetic factors, lipid infiltration, and oxidative damage, the immune system is now being accepted to play a crucial role in pathogenesis and disease continuation. No longer considered a passive degenerative disease, CAVD is understood to be an active inflammatory process, involving a multitude of pro-inflammatory mechanisms, with both the adaptive and the innate immune system underpinning these complex mechanisms. Within the valve, 15% of cells evolve from haemopoietic origin, and this number greatly expands following inflammation, as macrophages, T lymphocytes, B lymphocytes, and innate immune cells infiltrate the valve, promoting further inflammation. Whether chronic immune infiltration or pathogenic clonal expansion of immune cells within the valve or a combination of the two is responsible for disease progression, it is clear that greater understanding of the immune systems role in valve disease is required to inform future treatment strategies for control of CAVD development.
Collapse
Affiliation(s)
- Francesca Bartoli-Leonard
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jonas Zimmer
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
25
|
Sucato V, Corrado E, Manno G, Amata F, Testa G, Novo G, Galassi AR. Biomarkers of Coronary Microvascular Dysfunction in Patients With Microvascular Angina: A Narrative Review. Angiology 2021; 73:395-406. [PMID: 34338554 DOI: 10.1177/00033197211034267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The current gold standard for diagnosis of coronary microvascular dysfunction (CMD) in the absence of myocardial diseases, whose clinical manifestation is microvascular angina (MVA), is reactivity testing using adenosine or acetylcholine during coronary angiography. This invasive test can be difficult to perform, expensive, and harmful. The identification of easily obtainable blood biomarkers which reflect the pathophysiology of CMD, characterized by high reliability, precision, accuracy, and accessibility may reduce risks and costs related to invasive procedures and even facilitate the screening and diagnosis of CMD. In this review, we summarized the results of several studies that have investigated the possible relationships between blood biomarkers involved with CMD and MVA. More specifically, we have divided the analyzed biomarkers into 3 different groups, according to the main mechanisms underlying CMD: biomarkers of "endothelial dysfunction," "vascular inflammation," and "oxidative stress." Finally, in the last section of the review, we consider mixed mechanisms and biomarkers which are not included in the 3 major categories mentioned above, but could be involved in the pathogenesis of CMD.
Collapse
Affiliation(s)
- Vincenzo Sucato
- Unit of Cardiology, University Hospital Paolo Giaccone, University of Palermo, Palermo, Italy.,Department of Excellence of Sciences for Health Promotion and Maternal-Child Care, Internal Medicine and Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Egle Corrado
- Unit of Cardiology, University Hospital Paolo Giaccone, University of Palermo, Palermo, Italy.,Department of Excellence of Sciences for Health Promotion and Maternal-Child Care, Internal Medicine and Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Girolamo Manno
- Unit of Cardiology, University Hospital Paolo Giaccone, University of Palermo, Palermo, Italy.,Department of Excellence of Sciences for Health Promotion and Maternal-Child Care, Internal Medicine and Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Francesco Amata
- Department of Excellence of Sciences for Health Promotion and Maternal-Child Care, Internal Medicine and Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Gabriella Testa
- Unit of Cardiology, University Hospital Paolo Giaccone, University of Palermo, Palermo, Italy.,Department of Excellence of Sciences for Health Promotion and Maternal-Child Care, Internal Medicine and Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Giuseppina Novo
- Unit of Cardiology, University Hospital Paolo Giaccone, University of Palermo, Palermo, Italy.,Department of Excellence of Sciences for Health Promotion and Maternal-Child Care, Internal Medicine and Specialties (ProMISE), University of Palermo, Palermo, Italy
| | - Alfredo R Galassi
- Department of Excellence of Sciences for Health Promotion and Maternal-Child Care, Internal Medicine and Specialties (ProMISE), University of Palermo, Palermo, Italy
| |
Collapse
|
26
|
Patel N, Chin DD, Chung EJ. Exosomes in Atherosclerosis, a Double-Edged Sword: Their Role in Disease Pathogenesis and Their Potential as Novel Therapeutics. AAPS JOURNAL 2021; 23:95. [PMID: 34312734 DOI: 10.1208/s12248-021-00621-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/29/2021] [Indexed: 12/23/2022]
Abstract
Cardiovascular disease (CAD) due to atherosclerosis is a major cause of death worldwide. The development of atherosclerosis involves intercellular communication facilitated by exosomes secreted from vascular endothelial cells (VECs), vascular smooth muscle cells (VSMCs), immune cells, and platelets. In this review, we summarize the current understanding of exosome biogenesis and uptake, and discuss atherogenic and atheroprotective functions of exosomes secreted from these cell types. In addition, we examine the potential of enhancing the therapeutic and targeting ability of exosomes exhibiting atheroprotective function by drug loading and surface modification with targeting ligands. We conclude with current challenges associated with exosome engineering for therapeutic use.
Collapse
Affiliation(s)
- Neil Patel
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, DRB 140, California, Los Angeles, 90089, USA
| | - Deborah D Chin
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, DRB 140, California, Los Angeles, 90089, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, DRB 140, California, Los Angeles, 90089, USA. .,Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, University of Southern California, California, Los Angeles, 90033, USA. .,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, California, Los Angeles, 90089, USA. .,Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, California, Los Angeles, 90033, USA.
| |
Collapse
|
27
|
Role of Extracellular Vesicles in Compromising Cellular Resilience to Environmental Stressors. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9912281. [PMID: 34337063 PMCID: PMC8321721 DOI: 10.1155/2021/9912281] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/16/2021] [Accepted: 07/07/2021] [Indexed: 12/17/2022]
Abstract
Extracellular vesicles (EVs), like exosomes, are nanosized membrane-enveloped vesicles containing different bioactive cargo, such as proteins, lipids, mRNA, miRNA, and other small regulatory RNAs. Cell-derived EVs, including EVs originating from stem cells, may capture components from damaged cells or cells impacted by therapeutic treatments. Interestingly, EVs derived from stem cells can be preconditioned to produce and secrete EVs with different therapeutic properties, particularly with respect to heat-shock proteins and other molecular cargo contents. This behavior is consistent with stem cells that also respond differently to various microenvironments. Heat-shock proteins play roles in cellular protection and mediate cellular resistance to radiotherapy, chemotherapy, and heat shock. This review highlights the possible roles EVs play in mediating cellular plasticity and survival when exposed to different physical and chemical stressors, with a special focus on the respiratory distress due to the air pollution.
Collapse
|
28
|
Patients with Coronary Artery Disease Have Lower Levels of Antibody to Heat-Stressed Fibroblast Derived Proteins, versus Normal Subjects. Cardiovasc Ther 2021; 2021:5577218. [PMID: 34239605 PMCID: PMC8225444 DOI: 10.1155/2021/5577218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/03/2021] [Accepted: 06/05/2021] [Indexed: 01/06/2023] Open
Abstract
Cellular stress response plays an important role in the pathophysiology of coronary artery disease (CAD). Inhibition of cellular stress may provide a novel clinical approach regarding the diagnosis and treatment of CAD. Fibroblasts constitute 60-70% of cardiac cells and have a crucial role in cardiovascular function. Hence, the aim of this study was to show a potential therapeutic application of proteins derived from heat-stressed fibroblast in CAD patients. Fibroblasts were isolated from the foreskin and cultured under heat stress conditions. Surprisingly, 1.06% of the cells exhibited a necrotic death pattern. Furthermore, heat-stressed fibroblasts produced higher level of total proteins than control cells. In SDS-PAGE analysis, a 70 kDa protein band was observed in stressed cell culture supernatants which appeared as two acidic spots with close pI in the two-dimensional electrophoresis. To evaluate the immunogenic properties of fibroblast-derived heat shock proteins (HSPs), the serum immunoglobulin-G (IgG) was measured by ELISA in 50 CAD patients and 50 normal subjects who had been diagnosed through angiography. Interestingly, the level of anti-HSP antibody was significantly higher in non-CAD individuals in comparison with the patient's group (p < 0.05). The odds ratio for CAD was 5.06 (95%CI = 2.15‐11.91) in cut-off value of 30 AU/mL of anti-HSP antibody. Moreover, ROC analysis showed that anti-HSP antibodies had a specificity of 74% and a sensitivity of 64%, which is almost equal to 66% sensitivity of exercise stress test (EST) as a CAD diagnostic method. These data revealed that fibroblast-derived HSPs are suitable for the diagnosis and management of CAD through antibody production.
Collapse
|
29
|
Revisiting Ehrlichia ruminantium Replication Cycle Using Proteomics: The Host and the Bacterium Perspectives. Microorganisms 2021; 9:microorganisms9061144. [PMID: 34073568 PMCID: PMC8229282 DOI: 10.3390/microorganisms9061144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/28/2021] [Accepted: 05/07/2021] [Indexed: 12/16/2022] Open
Abstract
The Rickettsiales Ehrlichia ruminantium, the causal agent of the fatal tick-borne disease Heartwater, induces severe damage to the vascular endothelium in ruminants. Nevertheless, E. ruminantium-induced pathobiology remains largely unknown. Our work paves the way for understanding this phenomenon by using quantitative proteomic analyses (2D-DIGE-MS/MS, 1DE-nanoLC-MS/MS and biotin-nanoUPLC-MS/MS) of host bovine aorta endothelial cells (BAE) during the in vitro bacterium intracellular replication cycle. We detect 265 bacterial proteins (including virulence factors), at all time-points of the E. ruminantium replication cycle, highlighting a dynamic bacterium–host interaction. We show that E. ruminantium infection modulates the expression of 433 host proteins: 98 being over-expressed, 161 under-expressed, 140 detected only in infected BAE cells and 34 exclusively detected in non-infected cells. Cystoscape integrated data analysis shows that these proteins lead to major changes in host cell immune responses, host cell metabolism and vesicle trafficking, with a clear involvement of inflammation-related proteins in this process. Our findings led to the first model of E. ruminantium infection in host cells in vitro, and we highlight potential biomarkers of E. ruminantium infection in endothelial cells (such as ROCK1, TMEM16K, Albumin and PTPN1), which may be important to further combat Heartwater, namely by developing non-antibiotic-based strategies.
Collapse
|
30
|
Effects from the induction of heat shock proteins in a murine model due to progression of aortic atherosclerosis. Sci Rep 2021; 11:7025. [PMID: 33782520 PMCID: PMC8007726 DOI: 10.1038/s41598-021-86601-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 03/18/2021] [Indexed: 02/05/2023] Open
Abstract
Heat shock proteins (HSPs) are molecular chaperones that repair denatured proteins. The relationship between HSPs and various diseases has been extensively studied. However, the relationship between HSPs and atherosclerosis remains unclear. In this study, we induced the expression of HSPs and analyzed the effects on the development/progression of atherosclerosis in vivo. Remarkably, when HSPs were induced in apolipoprotein E deficient (ApoE-/-) mice prior to the formation of atheromas, the progression of atherosclerosis was inhibited; the short-term induction of HSPs significantly decreased the mRNA expression of intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) in the aorta. In contrast, the induction of HSPs after the formation of atheromas promoted the progression of atherosclerosis. In fact, the short-term induction of HSPs, after the formation of atheromas, significantly increased the mRNA expression of tumor necrosis factor-alpha, and interleukin 6 in the aorta. Of note, the induction of HSPs also promoted the formation of macrophage-derived foam cells. Overall, these results indicate that HSPs exerts different effects in the context of aortic atherosclerosis, depending on its degree of progression. Therefore, the induction and inhibition of HSPs should be considered for the prevention and treatment of atherosclerosis, respectively.
Collapse
|
31
|
Mitochondrial DAMPs and altered mitochondrial dynamics in OxLDL burden in atherosclerosis. Mol Cell Biochem 2021; 476:1915-1928. [PMID: 33492610 DOI: 10.1007/s11010-021-04061-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/11/2021] [Indexed: 10/22/2022]
Abstract
Atherosclerosis results in life-threatening cardiovascular pathologies, including ischemic heart disease, stroke, myocardial infarction, and peripheral arterial disease. The role of increased serum low-density lipoprotein (LDL) and resultant accumulation of oxidized-LDL (oxLDL) in atheroma formation is well established. Recent findings elucidate the significance of mitochondrial damage-associated molecular patterns (mtDAMPs) in triggering sterile inflammation in concert with oxLDL. The mtDAMPs including mitochondrial DNA (mtDNA), cytochrome C, cardiolipin, heat shock protein 60 (HSP60), mitochondrial transcription factor A (TFAM), and N-formyl peptides, are expected to possess proatherogenic roles. However, limited data are available in the literature. The mtDAMPs initiate sterile inflammation in atherosclerotic lesions via numerous signaling pathways, most of which converge to the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome. Priming the activation of the NLRP3 inflammasome, mtDAMPs promote secretion of proinflammatory cytokines, including interleukin-1β (IL-1β), implicated in atherosclerotic lesions through vascular smooth muscle and fibroblast proliferation, arterial wall thickening, and plaque formation. In this article we critically reviewed and discussed the central role of the NLRP3 inflammasome in mtDAMP-induced sterile inflammation in atherosclerosis with specific components including caspase-1, pregnane X receptor (PXR), adenosine monophosphate activated protein kinase (AMPK), protein phosphatase 2A (PP2A), thioredoxin-interacting protein (TXNIP), and downstream cytokines including IL-1β and IL-18 as potential mediators of atherosclerosis. Better understanding of the proinflammatory effects of mtDAMPs and its pathological association with oxLDL possess immense translational significance for novel therapeutic intervention.
Collapse
|
32
|
Salibe-Filho W, Araujo TLS, G. Melo E, B. C. T. Coimbra L, Lapa MS, Acencio MMP, Freitas-Filho O, Capelozzi VL, Teixeira LR, Fernandes CJCS, Jatene FB, Laurindo FRM, Terra-Filho M. Shear stress-exposed pulmonary artery endothelial cells fail to upregulate HSP70 in chronic thromboembolic pulmonary hypertension. PLoS One 2020; 15:e0242960. [PMID: 33270690 PMCID: PMC7714249 DOI: 10.1371/journal.pone.0242960] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/12/2020] [Indexed: 12/31/2022] Open
Abstract
The pathophysiological mechanisms underlying chronic thromboembolic pulmonary hypertension (CTEPH) are still unclear. Endothelial cell (EC) remodeling is believed to contribute to this pulmonary disease triggered by thrombus and hemodynamic forces disbalance. Recently, we showed that HSP70 levels decrease by proatherogenic shear stress. Molecular chaperones play a major role in proteostasis in neurological, cancer and inflammatory/ infectious diseases. To shed light on microvascular responses in CTEPH, we characterized the expression of molecular chaperones and annexin A2, a component of the fibrinolytic system. There is no animal model that reproduces microvascular changes in CTEPH, and this fact led us to isolated endothelial cells from patients with CTEPH undergoing pulmonary endarterectomy (PEA). We exposed CTEPH-EC and control human pulmonary endothelial cells (HPAEC) to high- (15 dynes/cm2) or low- (5 dynes/cm2) shear stress. After high-magnitude shear stress HPAEC upregulated heat shock protein 70kDa (HSP70) and the HSP ER paralogs 78 and 94kDa glucose-regulated protein (GRP78 and 94), whereas CTEPH-ECs failed to exhibit this response. At static conditions, both HSP70 and HSP90 families in CTEPH-EC are decreased. Importantly, immunohistochemistry analysis showed that HSP70 expression was downregulated in vivo, and annexin A2 was upregulated. Interestingly, wound healing and angiogenesis assays revealed that HSP70 inhibition with VER-155008 further impaired CTEPH-EC migratory responses. These results implicate HSP70 as a novel master regulator of endothelial dysfunction in type 4 PH. Overall, we first show that global failure of HSP upregulation is a hallmark of CTEPH pathogenesis and propose HSP70 as a potential biomarker of this condition.
Collapse
Affiliation(s)
- William Salibe-Filho
- Pulmonary Division, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo - São Paulo, Brazil
| | - Thaís L. S. Araujo
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Everton G. Melo
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Luiza B. C. T. Coimbra
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Monica S. Lapa
- Pulmonary Division, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo - São Paulo, Brazil
| | - Milena M. P. Acencio
- Pulmonary Division, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo - São Paulo, Brazil
| | - Orival Freitas-Filho
- Cardiovascular Surgery Division, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo - São Paulo, Brazil
| | - Vera Luiza Capelozzi
- Department of Pathology, Faculdade de Medicina da Universidade de São Paulo - São Paulo, Brazil
| | - Lisete Ribeiro Teixeira
- Pulmonary Division, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo - São Paulo, Brazil
| | - Caio J. C. S. Fernandes
- Pulmonary Division, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo - São Paulo, Brazil
| | - Fabio Biscegli Jatene
- Cardiovascular Surgery Division, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo - São Paulo, Brazil
| | - Francisco R. M. Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo - São Paulo, Brazil
| | - Mario Terra-Filho
- Pulmonary Division, Heart Institute (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo - São Paulo, Brazil
| |
Collapse
|
33
|
Amin MN, Siddiqui SA, Ibrahim M, Hakim ML, Ahammed MS, Kabir A, Sultana F. Inflammatory cytokines in the pathogenesis of cardiovascular disease and cancer. SAGE Open Med 2020; 8:2050312120965752. [PMID: 33194199 PMCID: PMC7594225 DOI: 10.1177/2050312120965752] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammatory cytokines are highly inducible small glycoproteins or regulatory proteins of low molecular weight secreted by different cell types. They regulate intercellular communication and mediate a number of physiological functions in the human immune system. Numerous prospective studies report that inflammatory cytokines strongly predict coronary artery disease, myocardial infarction, heart failure and other adverse cardiac events. Inflammatory cascade is believed to be a causative factor in the development of atherosclerotic process. Several aspects of atherogenesis are accelerated by cytokines. This article provides an overall overview of current understanding of cytokines in various cardiovascular events. Besides, inflammatory cytokines trigger cellular events that can induce malignancy and carcinogenesis. Elevated expression of several cytokines such as interleukin-1, interleukin-6, interleukin-10, tumor necrosis factor-α, macrophage migration inhibitory factor and transforming growth factor-β are involved in tumor initiation and progression. Thus, they exert a pivotal role in cancer pathogenesis. This review highlights the role of several cytokines in various events of tumorigenesis. Actually, this article summarizes the contributions of cytokines in the pathogenesis of cardiovascular disease and cancer.
Collapse
Affiliation(s)
- Mohammad Nurul Amin
- Department of Pharmacy, Atish Dipankar
University of Science and Technology, Dhaka, Bangladesh
- Pratyasha Health Biomedical Research
Center, Dhaka, Bangladesh
| | - Shafayet Ahmed Siddiqui
- Department of Pharmacy, Atish Dipankar
University of Science and Technology, Dhaka, Bangladesh
- Pratyasha Health Biomedical Research
Center, Dhaka, Bangladesh
| | - Md Ibrahim
- College of Medicine, University of South
Alabama, Mobile, AL, USA
| | - Md Lukman Hakim
- Department of Pharmaceutical Sciences,
North South University, Dhaka, Bangladesh
| | - Md. Salim Ahammed
- Department of Pharmacy, University of
Information Technology and Sciences, Dhaka, Bangladesh
| | - Asma Kabir
- Department of Pharmacy, Atish Dipankar
University of Science and Technology, Dhaka, Bangladesh
- Pratyasha Health Biomedical Research
Center, Dhaka, Bangladesh
| | - Farhana Sultana
- Department of Pharmacy, Atish Dipankar
University of Science and Technology, Dhaka, Bangladesh
- Pratyasha Health Biomedical Research
Center, Dhaka, Bangladesh
| |
Collapse
|
34
|
Sun L, Wang X, Saredy J, Yuan Z, Yang X, Wang H. Innate-adaptive immunity interplay and redox regulation in immune response. Redox Biol 2020; 37:101759. [PMID: 33086106 PMCID: PMC7575795 DOI: 10.1016/j.redox.2020.101759] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/01/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Innate and adaptive immune cell activation and infiltration is the key characteristic of tissue inflammation. The innate immune system is the front line of host defense in which innate immune cells are activated by danger signals, including pathogen- and danger-associated molecular pattern, and metabolite-associated danger signal. Innate immunity activation can directly contribute to tissue inflammation or immune resolution by phagocytosis and secretion of biologically active molecules, or indirectly via antigen-presenting cell (APC) activation-mediated adaptive immune responses. This review article describes the cellular and molecular interplay of innate-adaptive immune systems. Three major mechanisms are emphasized in this article for their role in facilitating innate-adaptive immunity interplay. 1) APC can be formed from classical and conditional innate immune cells to bridge innate-adaptive immune response. 2) Immune checkpoint molecular pairs connect innate and adaptive immune cells to direct one-way and two-way immune checkpoint reactions. 3) Metabolic reprogramming during immune responses leads to excessive cytosolic and mitochondrial reactive oxygen species (ROS) production. Increased NADPH oxidase-derived extracellular and intracellular ROS are mostly responsible for oxidative stress, which contributes to functional changes in immune cells. Further understanding of innate-adaptive immunity interplay and its underlying molecular basis would lead to the identification of therapeutic targets for immunological and inflammatory disease.
Collapse
Affiliation(s)
- Lizhe Sun
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China; Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Xianwei Wang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jason Saredy
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA; Department of Microbiology and Immunology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA.
| |
Collapse
|
35
|
Nzvere FP, Tariq E, Nishanth K, Arshid A, Cancarevic I. Long-Term Cardiovascular Diseases of Heatstroke: A Delayed Pathophysiology Outcome. Cureus 2020; 12:e9595. [PMID: 32789098 PMCID: PMC7416985 DOI: 10.7759/cureus.9595] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Heatstroke, defined as an elevated core body temperature above 40°C accompanied by altered mental status (e.g., confusion, disorientation, seizure and coma), is the most severe and life-threatening condition in the spectrum of heat-related illnesses. Heatstroke patients may present with multi-organ dysfunction, but with rapid cooling and organ failure management, a full recovery often occurs within weeks. Long-term impairment is rare, with neurological impairment occurring most frequently. Despite an abundance of research on the persistent neurological and hepatic impairments, our knowledge of the long-term cardiovascular events in patients with heatstroke history is poor. We wondered whether heatstroke leads to cardiovascular diseases long after full recovery. Using Pubmed, Web of Science and Scopus, we gathered cohort studies looking at cardiovascular disease incidence or mortality as an outcome, including heatstroke animal studies. Based on the available literature, we found that a history of heatstroke is associated with an increased risk of cardiovascular diseases, including ischemic heart disease, heart failure and atrial fibrillation. Delayed metabolic disturbances occurring in exertional heatstroke mice are linked to the formation of atherosclerosis and the development of heart failure. These processes provide potential pathophysiological pathways leading to ischemic heart disease and heart failure in heatstroke patients. Our findings may massively impact our understanding of heatstroke recovery and the follow up of heatstroke patients. Therefore larger, more adequately powered cohort studies with cardiovascular disease as an outcome, in tandem with animal studies examining the underlying pathophysiology, are required to confirm or reject these findings and answer the proposed questions.
Collapse
Affiliation(s)
- Farirai P Nzvere
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ezza Tariq
- Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA.,Medicine, Nishtar Medical College, Multan, PAK
| | - Katukuri Nishanth
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Assam Arshid
- Surgery, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ivan Cancarevic
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
36
|
Engineering the gut microbiota to treat chronic diseases. Appl Microbiol Biotechnol 2020; 104:7657-7671. [PMID: 32696297 PMCID: PMC7484268 DOI: 10.1007/s00253-020-10771-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/18/2020] [Accepted: 07/02/2020] [Indexed: 12/21/2022]
Abstract
Gut microbes play vital roles in host health and disease. A number of commensal bacteria have been used as vectors for genetic engineering to create living therapeutics. This review highlights recent advances in engineering gut bacteria for the treatment of chronic diseases such as metabolic diseases, cancer, inflammatory bowel diseases, and autoimmune disorders. KEY POINTS: • Bacterial homing to tumors has been exploited to deliver therapeutics in mice models. • Engineered bacteria show promise in mouse models of metabolic diseases. • Few engineered bacterial treatments have advanced to clinical studies.
Collapse
|
37
|
Tang J, Tang Y, Yi I, Chen DF. The role of commensal microflora-induced T cell responses in glaucoma neurodegeneration. PROGRESS IN BRAIN RESEARCH 2020; 256:79-97. [PMID: 32958216 DOI: 10.1016/bs.pbr.2020.06.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Over the last decade, new evidence has become increasingly more compelling that commensal microflora profoundly influences the maturation and function of resident immune cells in host physiology. The concept of gut-retina axis is actively being explored. Studies have revealed a critical role of commensal microbes linked with neuronal stress, immune responses, and neurodegeneration in the retina. Microbial dysbiosis changes the blood-retina barrier permeability and modulates T cell-mediated autoimmunity to contribute to the pathogenesis of retinal diseases, such as glaucoma. Heat shock proteins (HSPs), which are evolutionarily conserved, are thought to function both as neuroprotectant and pathogenic antigens of T cells contributing to cell protection and tissue damage, respectively. Activated microglia recruit and interact with T cells during this process. Glaucoma, characterized by the progressive loss of retinal ganglion cells, is the leading cause of irreversible blindness. With nearly 70 million people suffering glaucoma worldwide, which doubles the number of patients with Alzheimer's disease, it represents the most frequent neurodegenerative disease of the central nervous system (CNS). Thus, understanding the mechanism of neurodegeneration in glaucoma and its association with the function of commensal microflora may help unveil the secrets of many neurodegenerative disorders in the CNS and develop novel therapeutic interventions.
Collapse
Affiliation(s)
- Jing Tang
- Department of Ophthalmology, West China Hospital, Sichuan University, Sichuan, China; Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Yizhen Tang
- Department of Ophthalmology and Vision Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China; Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Irvin Yi
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Dong Feng Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
38
|
de Lima Filho JB, Freire L, Nahas EAP, Orsatti FL, Orsatti CL. Heat Shock Protein 60 Antibodies Are Associated With a Risk Factor for Cardiovascular Disease in Bedridden Elderly Patients. Front Mol Biosci 2020; 7:103. [PMID: 32613007 PMCID: PMC7307547 DOI: 10.3389/fmolb.2020.00103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/05/2020] [Indexed: 12/26/2022] Open
Abstract
Frailty, in elderly people, represents multiple deficiencies in different organs and is characterized by decreased physiological reserves and greater vulnerability to stressors. Bedridden elderly, with cardiovascular disease (CVD), have a worse prognosis than non-bedridden patients. Heat-shock proteins (HSPs) are molecular chaperones that under physiological conditions facilitate the transport, folding and assembly of proteins. Serum HSP 60-kDa concentrations and their antibodies are increased, in response to non-physiological conditions, suggesting the involvement of HSPs and their antibodies in the development of CVD. The aim of this work was to evaluate heat shock protein 60 and anti-HSP60 antibody levels, associated with a risk factor for cardiovascular disease, in bedridden elderly patients. Clinical, analytical and cross-sectional analyses were performed with 57 elderly (>65 years). HSP60 and anti-HSP60 plasma levels were measured by ELISA. Bivariate analysis using a linear regression model adjusted for risk factors used Framingham Score. Among the 57 elderly, with an average age of 69.89 years, 39% are bedridden; 26% with pre-existing cardiovascular disease and 44% are dyslipidemic. The relationship of risk factors in the Framingham Score was positive for the anti-HSP60 antibody (p = 0.042) measurement. Our data show a positive correlation among the elevation of the Framingham score and the profile of anti-HSP60 antibodies. These results suggest a greater immune activation that is associated with cardiovascular risk and bedridden fragility.
Collapse
Affiliation(s)
| | - Letícia Freire
- Department of Health Science, Oeste Paulista University - UNOESTE, Jau, Brazil
| | - Eliana Aguiar Petri Nahas
- Department of Gynecology and Obstetrics, Botucatu Medical School, Paulista State University (UNESP), Botucatu, Brazil
| | - Fábio Lera Orsatti
- Department of Sport Sciences, Health Science Institute, Federal University of Triangulo Mineiro (UFTM), Uberaba, Brazil
| | | |
Collapse
|
39
|
The value of heat shock protein (HSP) 60 on in-hospital and short-term prognosis in patients with acute ST segment elevation myocardial infarction. JOURNAL OF SURGERY AND MEDICINE 2020. [DOI: 10.28982/josam.601334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
40
|
Vince RV, Kirk RJ, Aye MM, Atkin SL, Madden LA. Impaired heat shock protein 72 expression in women with polycystic ovary syndrome following a supervised exercise programme. Cell Stress Chaperones 2020; 25:73-80. [PMID: 31734892 PMCID: PMC6985053 DOI: 10.1007/s12192-019-01048-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/09/2019] [Accepted: 10/25/2019] [Indexed: 11/29/2022] Open
Abstract
Induction of heat shock protein expression and the heat shock (stress) response are seen in exercise. This exercise-induced response is thought protective against cellular stress through the expression of heat shock proteins. The highly inducible heat shock protein 72 (HSP72) has been shown to be expressed in a number of stress-related conditions, but not investigated in women with polycystic ovary syndrome (PCOS). Twenty-one women (10 controls, 11 with PCOS) concluded an 8-week supervised, moderate-intensity exercise programme. Monocytes and lymphocytes were analysed by flow cytometry for HSP72 expression from blood samples prior to, mid-way and at the completion of the programme. The monocyte HSP72 expression showed an increase from baseline values through mid-way (p = 0.025), and at the completion of the programme (p = 0.011) only in the control group, the PCOS group showed no significant change. This pattern was similar for lymphocyte HSP72 expression where a significant increase was found at the completion of the programme (p = 0.01) only in the control group. The magnitude of increased HSP72 expression following completion of the programme was linked to baseline values only in the control group. In conclusion, increased HSP72 expression to exercise over an 8-week period was seen in control but not in PCOS women, suggesting that there is an impairment of HSP72 expression in response to exercise in these women.
Collapse
Affiliation(s)
- Rebecca V Vince
- Sport, Health and Exercise Science, Faculty of Health Sciences, University of Hull, Hull, HU6 7RX, UK.
| | - Richard J Kirk
- Sport, Health and Exercise Science, Faculty of Health Sciences, University of Hull, Hull, HU6 7RX, UK
| | - Myint M Aye
- Hull York Medical School, Faculty of Health Sciences, University of Hull, Hull, UK
| | | | - Leigh A Madden
- Biomedical Science, Faculty of Health Sciences, University of Hull, Hull, UK
| |
Collapse
|
41
|
Taheri-Bonakdar M, Khazaee M, Saberi-Karimian M, Tayefi M, Ghazizadeh H, Moohebati M, Ebrahimi M, Safarian M, Nematy M, Sahebkar A, Ferns GA, Ghayour-Mobarhan M, Reza Parizadeh SM. The association between serum anti-HSP27 levels with hypertriglyceridemia. TRANSLATIONAL METABOLIC SYNDROME RESEARCH 2020. [DOI: 10.1016/j.tmsr.2020.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
42
|
Spiering R, Jansen MAA, Wood MJ, Fath AA, Eltherington O, Anderson AE, Pratt AG, van Eden W, Isaacs JD, Broere F, Hilkens CMU. Targeting of tolerogenic dendritic cells to heat-shock proteins in inflammatory arthritis. J Transl Med 2019; 17:375. [PMID: 31727095 PMCID: PMC6857208 DOI: 10.1186/s12967-019-2128-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Background Autologous tolerogenic dendritic cells (tolDC) are a promising therapeutic strategy for inflammatory arthritis (IA) as they can regulate autoantigen-specific T cell responses. Here, we investigated two outstanding priorities for clinical development: (i) the suitability of using heat-shock proteins (HSP), abundant in inflamed synovia, as surrogate autoantigens to be presented by tolDC and (ii) identification of functional biomarkers that confirm tolDC regulatory activity. Methods Cell proliferation dye-labelled human peripheral blood mononuclear cells of IA (rheumatoid arthritis (RA) and psoriatic arthritis (PsA)) patients or healthy donors were cultured with HSP40-, HSP60- and HSP70-derived peptides or recall antigens (e.g. tuberculin purified protein derivative (PPD)) in the presence or absence of tolDC or control DC for 9 days. Functional characteristics of proliferated antigen-specific T-cells were measured using flow cytometry, gene expression profiling and cytokine secretion immunoassays. Repeated measures analysis of variance (ANOVA) with Bonferroni correction for comparisons between multiple groups and paired Student t test for comparisons between two groups were used to determine significance. Results All groups showed robust CD4+ T-cell responses towards one or more HSP-derived peptide(s) as assessed by a stimulation index > 2 (healthy donors: 78%, RA: 73%, PsA: 90%) and production of the cytokines IFNγ, IL-17A and GM-CSF. Addition of tolDC but not control DC induced a type 1 regulatory (Tr1) phenotype in the antigen-specific CD4+ T-cell population, as identified by high expression of LAG3, CD49b and secretion of IL-10. Furthermore, tolDC inhibited bystander natural killer (NK) cell activation in a TGFβ dependent manner. Conclusions HSP-specific CD4+ T-cells are detectable in the majority of RA and PsA patients and can be converted into Tr1 cells by tolDC. HSP-loaded tolDC may therefore be suitable for directing T regulatory responses to antigens in inflamed synovia of IA patients. Tr1 markers LAG3, CD49b and IL-10 are suitable biomarkers for future tolDC clinical trials.
Collapse
Affiliation(s)
- Rachel Spiering
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Manon A A Jansen
- Division of Immunology, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Matthew J Wood
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Anshorulloh A Fath
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Oliver Eltherington
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Amy E Anderson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Arthur G Pratt
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Willem van Eden
- Division of Immunology, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - John D Isaacs
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Femke Broere
- Division of Immunology, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands.,Department of Clinical Sciences of Companion Animals, Faculty Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Catharien M U Hilkens
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK. .,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK. .,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
43
|
Konstantinova EV, Chipigina NS, Shurdumova MH, Kovalenko E, Sapozhnikov AM. Heat Shock Protein 70 kDa as a Target for Diagnostics and Therapy of Cardiovascular and Cerebrovascular Diseases. Curr Pharm Des 2019; 25:710-714. [DOI: 10.2174/1381612825666190329123924] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/25/2019] [Indexed: 01/12/2023]
Abstract
Acute focal ischemia is a main factor of pathogenesis of a number of widespread cardiovascular and
cerebrovascular diseases, in particular, myocardial infarction and ischemic stroke. It is known that under the
conditions of ischemia expression of intracellular heat shock proteins (HSPs), especially HSP70, grows greatly
irrespective of the cell type. This stress-induced cell response is connected with cytoprotective properties of
HSP70. The protective functions of HSP70 contribute to the cell survival under adverse conditions and inhibit
development of programmed cell death. It was shown, that the level of HSP70 increases in cardiomyocytes and
brain cells in response to ischemia, that was connected with cardioprotective and neuroprotective effects. Besides,
in recent years, clinical studies of HSP70 have demonstrated elevated level of HSP70 in peripheral blood lymphocytes
in groups of patients with ischemic stroke and myocardial infarction. This review indicates that HSP70
can serve as a target for developing new approaches to diagnostics and therapy of cardiovascular and cerebrovascular
diseases.
Collapse
Affiliation(s)
- Ekaterina V. Konstantinova
- Pirogov Russian National Research Medical University, Ulitsa Ostrovityanova, 1, Moskva, Russian Federation
| | - Natalia S. Chipigina
- Pirogov Russian National Research Medical University, Ulitsa Ostrovityanova, 1, Moskva, Russian Federation
| | - Marina H. Shurdumova
- Pirogov Russian National Research Medical University, Ulitsa Ostrovityanova, 1, Moskva, Russian Federation
| | - E.I. Kovalenko
- M.M. Shemyakin-Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Federation
| | | |
Collapse
|
44
|
Krüger K, Reichel T, Zeilinger C. Role of heat shock proteins 70/90 in exercise physiology and exercise immunology and their diagnostic potential in sports. J Appl Physiol (1985) 2019; 126:916-927. [DOI: 10.1152/japplphysiol.01052.2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Heat shock proteins (HSPs) are molecular chaperones facilitating the unfolding or folding of secondary structures of proteins, their client proteins, in cellular stress situations. Various internal and external physiological and mechanical stress factors induce a homeostatic imbalance, followed by an increased expression of HSP70 and HSP90. Exercise is a stress factor, too, and its cumulative physiological perturbation manifests at a cellular level by threatening the protein homeostasis of various cell types. Consequently, an increase of HSP70/90 was described in plasma and mononuclear cells and various organs and tissues, such as muscle, liver, cardiac tissue, and brain, after an acute bout of exercise. The specific response of HSP70/90 seems to be strongly related to the modality of exercise, with several dependent factors such as duration, intensity, exercise type, subjects’ training status, and environmental factors, e.g., temperature. It is suggested that HSP70/90 play a major role in immune regulation and cell protection during exercise and in the efficiency of regeneration and reparation processes. During long-term training, HSP70/90 are involved in preconditioning and adaptation processes that might also be important for disease prevention and therapy. With regard to their highly sensitive and individual response to specific exercise and training modalities, this review discusses whether and how HSP70 and HSP90 can be applied as biomarkers for monitoring exercise and training.
Collapse
Affiliation(s)
- Karsten Krüger
- Department of Exercise and Health, Institute of Sports Science, Leibniz University Hannover, Hannover, Germany
| | - Thomas Reichel
- Department of Exercise and Health, Institute of Sports Science, Leibniz University Hannover, Hannover, Germany
| | - Carsten Zeilinger
- Center of Biomolecular Drug Research, Leibniz University Hannover, Hannover, Germany
| |
Collapse
|
45
|
Wang JC, Chien WC, Chu P, Chung CH, Lin CY, Tsai SH. The association between heat stroke and subsequent cardiovascular diseases. PLoS One 2019; 14:e0211386. [PMID: 30759128 PMCID: PMC6373898 DOI: 10.1371/journal.pone.0211386] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 12/24/2018] [Indexed: 12/13/2022] Open
Abstract
Background Recent studies have indicated that several critical illnesses are associated with an increased risk of cardiovascular diseases (CVDs). Nonetheless, studies of the association between heat-related illnesses (HRIs) and subsequent CVDs are still limited. We sought to evaluate whether heat stroke (HS) was associated with an increased CVD incidence. Methods The data from the nationwide, population-based, retrospective, cohort study described herein were obtained from the National Health Insurance Research Database in Taiwan. The outcome evaluated in this study was the cumulative incidence of CVDs, which was compared between patients with HS, patients with other HRIs and a control group during a 14-year follow-up period. Results Our analyses included 150 HS cases, 150 patients with other HRIs and 150 patients without HRIs. The HS patients had a significantly higher incidence of developing CVDs than the other HRI and control patients (32.67% vs. 23.33% vs. 16.67%, p = 0.005). Patients with HS had an increased incidence of acute myocardial infarction (AMI) compared with that of the controls (6% vs. 2.67%, p = 0.042) and an increased incidence of acute ischemic stroke (AIS) compared with those of the other HRI and control patients (12% vs. 6% vs. 4.67%, p = 0.038). An increased risk of chronic kidney disease (CKD) was also found in the patients with HS and other HRIs compared to that in the controls (17.33% vs. 14.67% vs. 6.67%, p = 0.016). Conclusion Prior HS was associated with an increased incidence of CVDs, particularly AMI and AIS, and an increased incidence of CKD.
Collapse
Affiliation(s)
- Jen-Chun Wang
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Wu-Chien Chien
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
- Taiwanese Injury Prevention and Safety Promotion Association, Taipei, Taiwan
- * E-mail: (SHT); (W-CC)
| | - Pauling Chu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chi-Hsiang Chung
- Department of Medical Research, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- School of Public Health, National Defense Medical Center, Taipei, Taiwan
- Taiwanese Injury Prevention and Safety Promotion Association, Taipei, Taiwan
| | - Chih-Yuan Lin
- Department of Surgery, Division of Cardiovascular Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hung Tsai
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
- * E-mail: (SHT); (W-CC)
| |
Collapse
|
46
|
Araujo TLS, Venturini G, Moretti AIS, Tanaka LY, Pereira AC, Laurindo FRM. Cell-surface HSP70 associates with thrombomodulin in endothelial cells. Cell Stress Chaperones 2019; 24:273-282. [PMID: 30645756 PMCID: PMC6363626 DOI: 10.1007/s12192-018-00964-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 04/26/2018] [Accepted: 12/14/2018] [Indexed: 10/27/2022] Open
Abstract
Heat shock protein-70 (HSP70) is crucial for proteostasis and displays cell-protective effects. Meanwhile, enhanced levels of cell-surface (cs) and secreted HSP70 paradoxically associate with pathologic cardiovascular conditions. However, mechanisms regulating csHSP70 pool are unknown. We hypothesized that total and csHSP70 expressions are modulated by hemodynamic forces, major contributors to endothelial pathophysiology. We also investigated whether thrombomodulin, a crucial thromboresistance cell-surface protein, is a csHSP70 target. We used proteomic/western analysis, confocal microscopy, and cs-biotinylation to analyze the pattern and specific characteristics of intracellular and csHSP70. HSP70 interaction with thrombomodulin was investigated by confocal colocalization, en face immunofluorescence, proximity assay, and immunoprecipitation. Thrombomodulin activity was assessed by measured protein C activation two-step assay. Our results show that csHSP70 pool in endothelial cells (EC) exhibits a peculiar cluster-like pattern and undergoes enhanced expression by physiological arterial-level laminar shear stress. Conversely, total and csHSP70 expressions were diminished under low shear stress, a known proatherogenic hemodynamic pattern. Furthermore, total HSP70 levels were decreased in aortic arch (associated with proatherogenic turbulent flow) compared with thoracic aorta (associated with atheroprotective laminar flow). Importantly, csHSP70 co-localized with thrombomodulin in cultured EC and aorta endothelium; proximity ligation assays and immunoprecipitation confirmed their physical interaction in EC. Remarkably, immunoneutralization of csHSP70 enhanced thrombomodulin activity in EC and aorta ex vivo. Overall, proatherogenic hemodynamic forces promote reduced total HSP70 expression, which might implicate in disturbed proteostasis; meanwhile, the associated decrease in cs-HSP70 pool associates with thromboresistance signaling. Cell-surface HSP70 (csHSP70) expression regulation and csHSP70 targets in vascular cells are unknown. We showed that HSP70 levels are shear stress-modulated and decreased under proatherogenic conditions. Remarkably, csHSP70 binds thrombomodulin and inhibits its activity in endothelial cells. This mechanism can potentially explain some deleterious effects previously associated with high extracellular HSP70 levels, as csHSP70 potentially could restrict thromboresistance and support thrombosis/inflammation in stress situations.
Collapse
Affiliation(s)
- Thaís L S Araujo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, Av. Enéas Carvalho Aguiar, 44, Annex II, 9th Floor, São Paulo, Brazil.
| | - Gabriela Venturini
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Ana I S Moretti
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, Av. Enéas Carvalho Aguiar, 44, Annex II, 9th Floor, São Paulo, Brazil
| | - Leonardo Y Tanaka
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, Av. Enéas Carvalho Aguiar, 44, Annex II, 9th Floor, São Paulo, Brazil
| | - Alexandre Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, Heart Institute (InCor), University of São Paulo School of Medicine, Av. Enéas Carvalho Aguiar, 44, Annex II, 9th Floor, São Paulo, Brazil
| |
Collapse
|
47
|
Bahrami LS, Sezavar Seyedi Jandaghi SH, Janani L, Pahlavan M, Arabi SM, Sadeghi H, Vafa M. Vitamin D supplementation and serum heat shock protein 60 levels in patients with coronary heart disease: a randomized clinical trial. Nutr Metab (Lond) 2018; 15:56. [PMID: 30093913 PMCID: PMC6080550 DOI: 10.1186/s12986-018-0292-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The aim in this study was to investigate the effect of vitamin D (25(OH)D3) supplementation on heat shock protein 60 (HSP 60) and other inflammatory markers (IL-17, TNF-α, PAB) in patients with coronary heart disease (CHD). METHODS In this double-blind, randomized clinical trial, we recruited 80 male and female patients aged 30-60 with CHD and 25(OH)D3 serum levels < 30 ng/ml from Rasool-e-Akram Hospital in Tehran, Iran. Serum levels of HSP 60 as primary outcome, and 25(OH)D3, IL-17, TNF-α, PAB, lipid profiles and parathyroid hormone (PTH) as secondary outcomes were measured at baseline and post-intervention. We randomly assigned eligible participants to a placebo group (N = 40) or an intervention group (N = 40) (50,000 IU/wk. vitamin D supplement) for eight weeks. RESULTS The results demonstrated that vitamin D supplementation resulted in a significant increase in 25(OH) D3 serum levels in the intervention group compared to the placebo group (46.86 vs. 7.28 ng/ml). PTH levels decreased in the intervention group compared to the placebo group (- 19.81 vs. 2.92 pg/ml) after eight weeks of supplementation. Furthermore, we observed a significant change in waist circumference (- 0.97 vs. -0.26 cm), fat percentage (-.13 vs. 0.1%), systolic blood pressure (- 3.85 vs. -2.11 mmHg) and diastolic blood presure (- 4 vs. -1.86 mmHg) in the vitamin D group compared to the placebo group (all P values < 0.05). Other variables did not significantly change after the intervention. CONCLUSION Based on our findings, weekly vitamin D supplementation of 50,000 IU for eight weeks in patients with CHD resulted in decreased systolic and diastolic blood pressure, waist circumference and fat percentage. No significant effect on HSP 60, inflammatory markers or lipid profiles was observed. TRIAL REGISTRATION IRCT, IRCT201612122365N14. Registered 12 December 2016.
Collapse
Affiliation(s)
- Leila Sadat Bahrami
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | | | - Leila Janani
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Mina Pahlavan
- Department of Medical Surgical Nursing, Nursing and Midwifery School, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Mostafa Arabi
- Department of Nutrition, Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Homa Sadeghi
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Mohammadreza Vafa
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
48
|
Barna J, Csermely P, Vellai T. Roles of heat shock factor 1 beyond the heat shock response. Cell Mol Life Sci 2018; 75:2897-2916. [PMID: 29774376 PMCID: PMC11105406 DOI: 10.1007/s00018-018-2836-6] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/07/2018] [Indexed: 01/09/2023]
Abstract
Various stress factors leading to protein damage induce the activation of an evolutionarily conserved cell protective mechanism, the heat shock response (HSR), to maintain protein homeostasis in virtually all eukaryotic cells. Heat shock factor 1 (HSF1) plays a central role in the HSR. HSF1 was initially known as a transcription factor that upregulates genes encoding heat shock proteins (HSPs), also called molecular chaperones, which assist in refolding or degrading injured intracellular proteins. However, recent accumulating evidence indicates multiple additional functions for HSF1 beyond the activation of HSPs. Here, we present a nearly comprehensive list of non-HSP-related target genes of HSF1 identified so far. Through controlling these targets, HSF1 acts in diverse stress-induced cellular processes and molecular mechanisms, including the endoplasmic reticulum unfolded protein response and ubiquitin-proteasome system, multidrug resistance, autophagy, apoptosis, immune response, cell growth arrest, differentiation underlying developmental diapause, chromatin remodelling, cancer development, and ageing. Hence, HSF1 emerges as a major orchestrator of cellular stress response pathways.
Collapse
Affiliation(s)
- János Barna
- Department of Genetics, Eötvös Loránd University, Pázmány Péter Stny. 1/C, Budapest, 1117, Hungary
- MTA-ELTE Genetics Research Group, Eötvös Loránd University, Budapest, Hungary
| | - Péter Csermely
- Department of Medical Chemistry, Semmelweis University, Budapest, Hungary
| | - Tibor Vellai
- Department of Genetics, Eötvös Loránd University, Pázmány Péter Stny. 1/C, Budapest, 1117, Hungary.
- MTA-ELTE Genetics Research Group, Eötvös Loránd University, Budapest, Hungary.
| |
Collapse
|
49
|
Forouzanfar F, Butler AE, Banach M, Barreto GE, Sahbekar A. Modulation of heat shock proteins by statins. Pharmacol Res 2018; 134:134-144. [DOI: 10.1016/j.phrs.2018.06.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 06/19/2018] [Indexed: 12/19/2022]
|
50
|
Vourc'h M, Roquilly A, Asehnoune K. Trauma-Induced Damage-Associated Molecular Patterns-Mediated Remote Organ Injury and Immunosuppression in the Acutely Ill Patient. Front Immunol 2018; 9:1330. [PMID: 29963048 PMCID: PMC6013556 DOI: 10.3389/fimmu.2018.01330] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/28/2018] [Indexed: 12/31/2022] Open
Abstract
Trauma is one of the leading causes of death and disability in the world. Multiple trauma or isolated traumatic brain injury are both indicative of human tissue damage. In the early phase after trauma, damage-associated molecular patterns (DAMPs) are released and give rise to sterile systemic inflammatory response syndrome (SIRS) and organ failure. Later, protracted inflammation following sepsis will favor hospital-acquired infection and will worsen patient’s outcome through immunosuppression. Throughout medical care or surgical procedures, severe trauma patients will be subjected to endogenous or exogenous DAMPs. In this review, we summarize the current knowledge regarding DAMP-mediated SIRS or immunosuppression and the clinical consequences in terms of organ failure and infections.
Collapse
Affiliation(s)
- Mickael Vourc'h
- Laboratoire UPRES EA3826 "Thérapeutiques cliniques et expérimentales des infections", IRS2 - Nantes Biotech, Université de Nantes, Nantes, France.,Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu, University Hospital of Nantes, Nantes, France
| | - Antoine Roquilly
- Laboratoire UPRES EA3826 "Thérapeutiques cliniques et expérimentales des infections", IRS2 - Nantes Biotech, Université de Nantes, Nantes, France.,Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu, University Hospital of Nantes, Nantes, France
| | - Karim Asehnoune
- Laboratoire UPRES EA3826 "Thérapeutiques cliniques et expérimentales des infections", IRS2 - Nantes Biotech, Université de Nantes, Nantes, France.,Intensive Care Unit, Anesthesia and Critical Care Department, Hôtel Dieu, University Hospital of Nantes, Nantes, France
| |
Collapse
|