1
|
Verlinden SF. The genetic advantage of healthy centenarians: unraveling the central role of NLRP3 in exceptional healthspan. FRONTIERS IN AGING 2024; 5:1452453. [PMID: 39301197 PMCID: PMC11410711 DOI: 10.3389/fragi.2024.1452453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
Despite extensive research into extending human healthspan (HS) and compressing morbidity, the mechanisms underlying aging remain elusive. However, a better understanding of the genetic advantages responsible for the exceptional HS of healthy centenarians (HC), who live in good physical and mental health for one hundred or more years, could lead to innovative health-extending strategies. This review explores the role of NLRP3, a critical component of innate immunity that significantly impacts aging. It is activated by pathogen-associated signals and self-derived signals that increase with age, leading to low-grade inflammation implicated in age-related diseases. Furthermore, NLRP3 functions upstream in several molecular aging pathways, regulates cellular senescence, and may underlie the robust health observed in HC. By targeting NLRP3, mice exhibit a phenotype akin to that of HC, the HS of monkeys is extended, and aging symptoms are reversed in humans. Thus, targeting NLRP3 could offer a promising approach to extend HS. Additionally, a paradigm shift is proposed. Given that the HS of the broader population is 30 years shorter than that of HC, it is postulated that they suffer from a form of accelerated aging. The term 'auto-aging' is suggested to describe accelerated aging driven by NLRP3.
Collapse
|
2
|
Fan SY, Zhao ZC, Liu XL, Peng YG, Zhu HM, Yan SF, Liu YJ, Xie Q, Jiang Y, Zeng SZ. Metformin Mitigates Sepsis-Induced Acute Lung Injury and Inflammation in Young Mice by Suppressing the S100A8/A9-NLRP3-IL-1β Signaling Pathway. J Inflamm Res 2024; 17:3785-3799. [PMID: 38895139 PMCID: PMC11182881 DOI: 10.2147/jir.s460413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Background Globally, the subsequent complications that accompany sepsis result in remarkable morbidity and mortality rates. The lung is among the vulnerable organs that incur the sepsis-linked inflammatory storm and frequently culminates into ARDS/ALI. The metformin-prescribed anti-diabetic drug has been revealed with anti-inflammatory effects in sepsis, but the underlying mechanisms remain unclear. This study aimed to ascertain metformin's effects and functions in a young mouse model of sepsis-induced ALI. Methods Mice were randomly divided into 4 groups: sham, sham+ Met, CLP, and CLP+ Met. CLP was established as the sepsis-induced ALI model accompanied by intraperitoneal metformin treatment. At day 7, the survival state of mice was noted, including survival rate, weight, and M-CASS. Lung histological pathology and injury scores were determined by hematoxylin-eosin staining. The pulmonary coefficient was used to evaluate pulmonary edema. Furthermore, IL-1β, CCL3, CXCL11, S100A8, S100A9 and NLRP3 expression in tissues collected from lungs were determined by qPCR, IL-1β, IL-18, TNF-α by ELISA, caspase-1, ASC, NLRP3, P65, p-P65, GSDMD-F, GSDMD-N, IL-1β and S100A8/A9 by Western blot. Results The data affirmed that metformin enhanced the survival rate, lessened lung tissue injury, and diminished the expression of inflammatory factors in young mice with sepsis induced by CLP. In contrast to sham mice, the CLP mice were affirmed to manifest ALI-linked pathologies following CLP-induced sepsis. The expressions of pro-inflammatory factors, for instance, IL-1β, IL-18, TNF-α, CXCL11, S100A8, and S100A9 are markedly enhanced by CLP, while metformin abolished this adverse effect. Western blot analyses indicated that metformin inhibited the sepsis-induced activation of GSDMD and the upregulation of S100A8/A9, NLRP3, and ASC. Conclusion Metformin could improve the survival rate, lessen lung tissue injury, and minimize the expression of inflammatory factors in young mice with sepsis induced by CLP. Metformin reduced sepsis-induced ALI via inhibiting the NF-κB signaling pathway and inhibiting pyroptosis by the S100A8/A9-NLRP3-IL-1β pathway.
Collapse
Affiliation(s)
- Shi-Yuan Fan
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, 410005, People’s Republic of China
| | - Zi-Chi Zhao
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, 410005, People’s Republic of China
| | - Xing-Lv Liu
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, 410005, People’s Republic of China
| | - Ying-Gang Peng
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, 410005, People’s Republic of China
| | - Hui-Min Zhu
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, 410005, People’s Republic of China
| | - Shi-Fan Yan
- Department of Emergency, Institute of Emergency Medicine, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, 410005, People’s Republic of China
| | - Yan-Juan Liu
- Department of Emergency, Institute of Emergency Medicine, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, 410005, People’s Republic of China
| | - Qin Xie
- Department of Emergency, Institute of Emergency Medicine, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, 410005, People’s Republic of China
| | - Yu Jiang
- Department of Emergency, Institute of Emergency Medicine, Hunan Provincial People’s Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, 410005, People’s Republic of China
| | - Sai-Zhen Zeng
- The First Affiliated Hospital of Hunan Normal University (Hunan Provincial People’s Hospital), Changsha, Hunan, 410005, People’s Republic of China
| |
Collapse
|
3
|
Teuwen JTJ, van der Vorst EPC, Maas SL. Navigating the Maze of Kinases: CaMK-like Family Protein Kinases and Their Role in Atherosclerosis. Int J Mol Sci 2024; 25:6213. [PMID: 38892400 PMCID: PMC11172518 DOI: 10.3390/ijms25116213] [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: 04/13/2024] [Revised: 05/30/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
Circulating low-density lipoprotein (LDL) levels are a major risk factor for cardiovascular diseases (CVD), and even though current treatment strategies focusing on lowering lipid levels are effective, CVD remains the primary cause of death worldwide. Atherosclerosis is the major cause of CVD and is a chronic inflammatory condition in which various cell types and protein kinases play a crucial role. However, the underlying mechanisms of atherosclerosis are not entirely understood yet. Notably, protein kinases are highly druggable targets and represent, therefore, a novel way to target atherosclerosis. In this review, the potential role of the calcium/calmodulin-dependent protein kinase-like (CaMKL) family and its role in atherosclerosis will be discussed. This family consists of 12 subfamilies, among which are the well-described and conserved liver kinase B1 (LKB1) and 5' adenosine monophosphate-activated protein kinase (AMPK) subfamilies. Interestingly, LKB1 plays a key role and is considered a master kinase within the CaMKL family. It has been shown that LKB1 signaling leads to atheroprotective effects, while, for example, members of the microtubule affinity-regulating kinase (MARK) subfamily have been described to aggravate atherosclerosis development. These observations highlight the importance of studying kinases and their signaling pathways in atherosclerosis, bringing us a step closer to unraveling the underlying mechanisms of atherosclerosis.
Collapse
Affiliation(s)
- Jules T. J. Teuwen
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P. C. van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, 52074 Aachen, Germany
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität München, 80336 München, Germany
| | - Sanne L. Maas
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany;
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
4
|
Vizuete AFK, Fróes F, Seady M, Hansen F, Ligabue-Braun R, Gonçalves CA, Souza DO. A Mechanism of Action of Metformin in the Brain: Prevention of Methylglyoxal-Induced Glutamatergic Impairment in Acute Hippocampal Slices. Mol Neurobiol 2024; 61:3223-3239. [PMID: 37980327 DOI: 10.1007/s12035-023-03774-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 11/05/2023] [Indexed: 11/20/2023]
Abstract
Metformin, a biguanide compound (N-1,1-dimethylbiguanide), is widely prescribed for diabetes mellitus type 2 (T2D) treatment. It also presents a plethora of properties, such as anti-oxidant, anti-inflammatory, anti-apoptosis, anti-tumorigenic, and anti-AGE formation activity. However, the precise mechanism of action of metformin in the central nervous system (CNS) needs to be clarified. Herein, we investigated the neuroprotective role of metformin in acute hippocampal slices exposed to methylglyoxal (MG), a highly reactive dicarbonyl compound and a key molecule in T2D developmental pathophysiology. Metformin protected acute hippocampal slices from MG-induced glutamatergic neurotoxicity and neuroinflammation by reducing IL-1β synthesis and secretion and RAGE protein expression. The drug also improved astrocyte function, particularly with regard to the glutamatergic system, increasing glutamate uptake. Moreover, we observed a direct effect of metformin on glutamate transporters, where the compound prevented glycation, by facilitating enzymatic phosphorylation close to Lys residues, suggesting a new neuroprotective role of metformin via PKC ζ in preventing dysfunction in glutamatergic system induced by MG.
Collapse
Affiliation(s)
- Adriana Fernanda K Vizuete
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil.
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil.
- Department of Biochemistry, Institute of Basic Health Sciences, UFRGS, Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil.
| | - Fernanda Fróes
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
| | - Marina Seady
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
| | - Fernanda Hansen
- Department of Nutrition, Health Sciences Center, Federal University of Santa Catarina, University Campus, Trindade, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Rodrigo Ligabue-Braun
- Department of Pharmacosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Avenida Sarmento Leite 245, Porto Alegre, 90050-130, Brazil
| | - Carlos-Alberto Gonçalves
- Laboratory of Calcium-Binding Proteins in the CNS, Department of Biochemistry, Institute of Basic Health Sciences, Universidade Federal Do Rio Grande Do Sul (UFRGS), Porto Alegre, RS, Brazil
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
- Department of Biochemistry, Institute of Basic Health Sciences, UFRGS, Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil
| | - Diogo O Souza
- Post Graduate Program in Biochemistry, Institute of Basic Health Sciences, UFRGS, Porto Alegre, RS, Brazil
- Department of Biochemistry, Institute of Basic Health Sciences, UFRGS, Ramiro Barcelos, 2600-Anexo, Porto Alegre, RS, 90035-003, Brazil
| |
Collapse
|
5
|
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
|
6
|
Yang X, Ma Y, Chen X, Zhu J, Xue W, Ning K. Mechanisms of neutrophil extracellular trap in chronic inflammation of endothelium in atherosclerosis. Life Sci 2023:121867. [PMID: 37348812 DOI: 10.1016/j.lfs.2023.121867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/24/2023]
Abstract
Cardiovascular diseases are a primary cause of morbidity and mortality around the world. In addition, atherosclerosis (AS)-caused cardiovascular disease is the primary cause of death in human diseases, and almost two billion people suffer from carotid AS worldwide. AS is caused by chronic inflammation of the arterial vessel and is initiated by dysfunction of vascular endothelial cells. Neutrophils protect against pathogen invasion because they function as a component of the innate immune system. However, the contribution of neutrophils to cardiovascular disease has not yet been clarified. Neutrophil extracellular traps (NETs) represent an immune defense mechanism that is different from direct pathogen phagocytosis. NETs are extracellular web-like structures activated by neutrophils, and they play important roles in promoting endothelial inflammation via direct or indirect pathways. NETs consist of DNA, histones, myeloperoxidase, matrix metalloproteinases, proteinase 3, etc. Most of the components of NETs have no direct toxic effect on endothelial cells, such as DNA, but they can damage endothelial cells indirectly. In addition, NETs play a critical role in the process of AS; therefore, it is important to clarify the mechanisms of NETs in AS because NETs are a new potential therapeutic target AS. This review summarizes the possible mechanisms of NETs in AS.
Collapse
Affiliation(s)
- Xiaofan Yang
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Yupeng Ma
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Xin Chen
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Jingjing Zhu
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China
| | - Wenlong Xue
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shanghai Key Laboratory of Bioactive Small Molecules, Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai 200032, China.
| | - Ke Ning
- School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China; School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, No. 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
7
|
Zhu C, Chen W, Cui H, Huang Z, Ding R, Li N, Wang Q, Wu F, Zhao Y, Cong X. TRIM64 promotes ox-LDL-induced foam cell formation, pyroptosis, and inflammation in THP-1-derived macrophages by activating a feedback loop with NF-κB via IκBα ubiquitination. Cell Biol Toxicol 2023; 39:607-620. [PMID: 36229750 PMCID: PMC10406714 DOI: 10.1007/s10565-022-09768-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 08/26/2022] [Indexed: 11/30/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease and the main pathology behind most cardiovascular diseases and the overactivation of macrophages initiates the development of atherosclerosis. However, the specific functions of oxidized low-density lipoprotein (ox-LDL) in macrophages remain elusive. Macrophages derived from monocyte (THP-1) were treated with ox-LDL and were used to generate atherosclerosis in an in vitro model. NLRP3 inflammasome markers were examined using quantitative RT-PCR and Western blotting. Cytokines were measured using ELISA. Chromatin immunoprecipitation (ChIP) was utilized to detect nuclear factor kappa B (NF-κB) and TRIM64 interactions. A fat-rich diet was applied to ApoE-/- mice for in vivo studies. ox-LDL promoted TRIM64 expression in a time-dependent manner. According to loss- and gain-of-function analyses, TRIM64 enhanced the activation of NLRP3 inflammasomes and the expression of downstream molecules. TRIM64 directly interacted with IκBα and promoted IκBα ubiquitination at K67 to activate NF-κB signaling. We detected direct binding between NF-κB and the TRIM64 promoter, as well as enhanced TRIM64 expression. Our study revealed an interaction between TRIM64 and NF-κB in the development of atherosclerosis. TRIM64 and NF-κB formed a positive feedback to activate NF-κB pathway. ox-LDL induces foam cell formation and TRIM64 expression TRIM64 regulates ox-LDL-induced foam cell formation, pyroptosis and inflammation via the NF-κB signaling TRIM64 activates NF-κB signaling by ubiquitination of IκBα NF-κB inhibition attenuates atherosclerosis in HFD-induced ApoE (-/-) mice.
Collapse
Affiliation(s)
- Chao Zhu
- Department of Nephrology, Changhai Hospital, Shanghai, 200433, China
| | - Wei Chen
- Department of Cardiology, Shanghai Changzheng Hospital, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Haiming Cui
- Department of Cardiology, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, No. 110 Ganhe Road, Hongkou District, Shanghai, 200437, China
| | - Zhigang Huang
- Department of Cardiology, Shanghai Changzheng Hospital, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Ru Ding
- Department of Cardiology, Shanghai Changzheng Hospital, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Na Li
- Department of Cardiology, Shanghai Changzheng Hospital, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Qinqin Wang
- Department of Cardiology, Shanghai Changzheng Hospital, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Feng Wu
- Department of Cardiology, Yueyang Hospital, Shanghai University of Traditional Chinese Medicine, No. 110 Ganhe Road, Hongkou District, Shanghai, 200437, China.
| | - Yanmin Zhao
- Department of Cardiology, Shanghai Changzheng Hospital, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China.
| | - Xiaoliang Cong
- Department of Cardiology, Shanghai Changzheng Hospital, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China.
| |
Collapse
|
8
|
Tanase DM, Valasciuc E, Gosav EM, Ouatu A, Buliga-Finis ON, Floria M, Maranduca MA, Serban IL. Portrayal of NLRP3 Inflammasome in Atherosclerosis: Current Knowledge and Therapeutic Targets. Int J Mol Sci 2023; 24:ijms24098162. [PMID: 37175869 PMCID: PMC10179095 DOI: 10.3390/ijms24098162] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/26/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
We are witnessing the globalization of a specific type of arteriosclerosis with rising prevalence, incidence and an overall cardiovascular disease burden. Currently, atherosclerosis increasingly affects the younger generation as compared to previous decades. While early preventive medicine has seen improvements, research advances in laboratory and clinical investigation promise to provide us with novel diagnosis tools. Given the physio-pathological complexity and epigenetic patterns of atherosclerosis and the discovery of new molecules involved, the therapeutic field of atherosclerosis has room for substantial growth. Thus, the scientific community is currently investigating the role of nucleotide-binding and oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a crucial component of the innate immune system in different inflammatory disorders. NLRP3 is activated by distinct factors and numerous cellular and molecular events which trigger NLRP3 inflammasome assembly with subsequent cleavage of pro-interleukin (IL)-1β and pro-IL-18 pathways via caspase-1 activation, eliciting endothelial dysfunction, promotion of oxidative stress and the inflammation process of atherosclerosis. In this review, we introduce the basic cellular and molecular mechanisms of NLRP3 inflammasome activation and its role in atherosclerosis. We also emphasize its promising therapeutic pharmaceutical potential.
Collapse
Affiliation(s)
- Daniela Maria Tanase
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Emilia Valasciuc
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Evelina Maria Gosav
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Anca Ouatu
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Oana Nicoleta Buliga-Finis
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Mariana Floria
- Department of Internal Medicine, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
| | - Minela Aida Maranduca
- Internal Medicine Clinic, "St. Spiridon" County Clinical Emergency Hospital Iasi, 700111 Iasi, Romania
- Department of Morpho-Functional Sciences II, Discipline of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ionela Lacramioara Serban
- Department of Morpho-Functional Sciences II, Discipline of Physiology, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iasi, Romania
| |
Collapse
|
9
|
Cen Y, Xiong Y, Qin R, Tao H, Yang Q, Pan X. Anti-malarial artesunate ameliorates atherosclerosis by modulating arterial inflammatory responses via inhibiting the NF-κB-NLRP3 inflammasome pathway. Front Pharmacol 2023; 14:1123700. [PMID: 36817159 PMCID: PMC9931906 DOI: 10.3389/fphar.2023.1123700] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction: Chronic inflammation plays a critical role in the pathogenesis of atherosclerosis (AS), and involves a complex interplay between blood components, macrophages, and arterial wall. Therefore, it is valuable in the development of targeted therapies to treat AS. Methods: AS rat model was induced by atherogenic diet plus with lipopolysaccharide (LPS) and then treated by anti-malarial artesunate (Art), a succinate derivative of artemisinin. The arterial morphology was observed after Oil red O, hematoxylin-eosin, and Masson's staining. The arterial protein level was detected by immunohistochemistry or immunofluorescence. The expression level of mRNA was determined by PCR array or real-time PCR. Results: Herein, we showed that Art possessed a dose-dependently protective effect on AS rats. In detail, Art showed a comparable inhibitory effect on arterial plaque and serum lipids compared to those of rosuvastatin (RS), and further showed a better inhibition on arterial lipid deposition and arterial remodeling comprised of arterial wall thicken and vascular collagen deposition, than those of RS. The improvement of Art on AS rats was related to inhibit arterial macrophage recruitment, and inhibit nuclear factor κB (NF-κB)-related excessive arterial inflammatory responses. Critically, Art showed significant inhibition on the NLRP3 inflammasome activation in both arterial wall and arterial macrophages, by down-regulating the expression of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) and apoptosis associated speckle-like protein containing CARD (ASC), leading to less production of the NLRP3 inflammasome-derived caspase-1, interleukin-1β (IL-1β), IL-18, and subsequent transforming growth factor β1 (TGF-β1) in AS rats. Conclusion: We propose that Art is an anti-AS agent acts through modulating the arterial inflammatory responses via inhibiting the NF-κB - NLRP3 inflammasome pathway.
Collapse
|
10
|
Xu L, Wang W, Song W. A combination of metformin and insulin improve cardiovascular and cerebrovascular risk factors in individuals with type 1 diabetes mellitus. Diabetes Res Clin Pract 2022; 191:110073. [PMID: 36075464 DOI: 10.1016/j.diabres.2022.110073] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/14/2022] [Accepted: 08/31/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND This study aims to further clarify whether the addition of metformin to insulin treatment improve cardiovascular and cerebrovascular risk factors in individuals with T1DM. METHODS Electronic databases were searched for randomized controlled trials in which the efficacy and safety of metformin were compared with those of a placebo for risk factors of cardiovascular and cerebrovascular disease among individuals with T1DM, and a meta-analysis was conducted. RESULTS Thirteen cardiovascular studies were identified. In the metformin group, mean carotid intimal media thickness was significantly reduced by 0.03 mm, ascending aortic pulse wave velocity by 6.3 m/s, descending aortic wall shear stress by 1.77 dyn/cm2 (P = 0.02), insulin daily dose by 0.05 U/kg/d, body weight by 2.27 kg, fat-free mass by 1.32 kg, body mass index by 0.58 kg/m2, hip circumference by 0.29 m, and low-density lipoprotein by 0.16 mmol/L, all above are P < 0.05. In the metformin group, flow-mediated dilation was increased by 1.29 %, glucose infusion rate/insulin by 18.22 mg/(kg⋅min)/μIU/μL, and waist-to-hip ratio by 0.02, all above are P < 0.00001. The metformin group showed no differences in blood pressure, reactive hyperemia index, waist circumference, triglyceride, total cholesterol, high-density lipoprotein cholesterol, or body mass index Z score. For cerebrovascular studies were identified. But none of them had a risk factor assessment. CONCLUSIONS Metformin can ameliorate cardiovascular and cerebrovascular risk factors through non-hypoglycemic multiple pathways in individuals with T1DM.
Collapse
Affiliation(s)
- Linlin Xu
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Wang
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Wei Song
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| |
Collapse
|
11
|
Puerarin ameliorates acute lung injury by modulating NLRP3 inflammasome-induced pyroptosis. Cell Death Dis 2022; 8:368. [PMID: 35977927 PMCID: PMC9385627 DOI: 10.1038/s41420-022-01137-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022]
Abstract
We commenced to analyze putative anti-pyroptosis effects of puerarin (PU) as mediated by the PP2A-HDAC1-NLRP3 pathway in acute lung injury (ALI). ALI animal and cell models were constructed, followed by treatment of PU. Then, the effect of HDAC1, PP2A, and NLRP3 on cell inflammation and pyroptosis was explored. The interaction between HDAC1 and PP2A as well as between PP2A and NLRP3 was analyzed. Our findings suggested that PU downregulated HDAC1 expression to alleviate symptoms of ALI. HDAC1 overexpression promoted inflammation induced by LPS, which reversed the inhibitory effect of PU on ALI. HDAC1 overexpression also decreased PP2A expression, suggesting that PP2A was involved in the effects of HDAC1 on LPS-induced inflammation. PP2A exerted inhibitory effects on NLRP3. Meanwhile, PU hindered the progression of ALI by silencing HDAC1 or overexpressing PP2A both in vivo and in vitro. Taken together, PU restrained pyroptosis of cells induced by NLRP3 inflammasome to abate ALI.
Collapse
|
12
|
Ye J, Li L, Wang M, Ma Q, Tian Y, Zhang Q, Liu J, Li B, Zhang B, Liu H, Sun G. Diabetes Mellitus Promotes the Development of Atherosclerosis: The Role of NLRP3. Front Immunol 2022; 13:900254. [PMID: 35844498 PMCID: PMC9277049 DOI: 10.3389/fimmu.2022.900254] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 06/01/2022] [Indexed: 11/22/2022] Open
Abstract
Atherosclerosis is one of the main complications of diabetes mellitus, involving a variety of pathogenic factors. Endothelial dysfunction, inflammation, and oxidative stress are hallmarks of diabetes mellitus and atherosclerosis. Although the ability of diabetes to promote atherosclerosis has been demonstrated, a deeper understanding of the underlying biological mechanisms is critical to identifying new targets. NLRP3 plays an important role in both diabetes and atherosclerosis. While the diversity of its activation modes is one of the underlying causes of complex effects in the progression of diabetes and atherosclerosis, it also provides many new insights for targeted interventions in metabolic diseases.
Collapse
Affiliation(s)
- Jingxue Ye
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lanfang Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiuxiao Ma
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Tian
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiong Zhang
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jiushi Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bengang Zhang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haitao Liu
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Guibo Sun, ; Haitao Liu,
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Guibo Sun, ; Haitao Liu,
| |
Collapse
|
13
|
Abstract
Atherosclerosis is considered a chronic, inflammatory disease responsible for more than 15% of all global deaths, secondary to its complications of myocardial infarction, vascular disease, and stroke. Current treatment regimens consist of lipid-lowering pharmaceuticals, control of risk factors, and prevention of plaque rupture and thrombosis with antiplatelet agents. However, a significant burden on society remains due to the morbidity and mortality of coronary artery disease despite our best practices. In addition to dyslipidemia and hemostasis, inflammation has now moved to the proverbial forefront as the remaining obstacle to appropriate management of atherosclerosis. A complex dance of endothelial dysfunction, complement activation, and immune cell-mediated cytokine release underlie the pathogenesis of atherosclerotic plaque development, destabilization, and rupture. Cholesterol-induced sterile inflammation is thought to be central to this process via activation of a protein complex called the nucleotide-binding oligomerization domain-, leucine-rich repeat- and pyrin domain-containing 3 (NLRP3) inflammasome. The focus of this review article will be to examine the NLRP3 inflammasome, which directs the release of interleukin-1, leading to downstream pro-inflammatory effects, and its potential for therapeutic targeting using currently available and future tools in our pharmacologic arsenal. In particular, we focus on the results of several large, recently concluded clinical trials including the Canakinumab Antiinflammatory Thrombosis Outcome Study, Colchicine Cardiovascular Outcomes Trial, and the Low-Dose Colchicine Study, examining the efficacy of direct inhibition of interleukin-1 with canakinumab or a multimodal approach to inhibiting the NLRP3 inflammasome using colchicine, as well as an overview of novel small molecule inhibitors that are still in development.
Collapse
Affiliation(s)
- Gregory Hemenway
- From the Department of Medicine, Temple University Hospital, Philadelphia, PA
| | - William H Frishman
- Department of Medicine, New York Medical College/Westchester Medical Center, Valhalla, NY
| |
Collapse
|
14
|
Tosta E. The seven constitutive respiratory defense barriers against SARS-CoV-2 infection. Rev Soc Bras Med Trop 2021; 54:e04612021. [PMID: 34932765 PMCID: PMC8687496 DOI: 10.1590/0037-8682-0461-2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/17/2021] [Indexed: 12/18/2022] Open
Abstract
Before eliciting an adaptive immune response, SARS-CoV-2 must overcome seven constitutive respiratory defense barriers. The first is the mucus covering the respiratory tract's luminal surface, which entraps inhaled particles, including infectious agents, and eliminates them by mucociliary clearance. The second barrier comprises various components present in the airway lining fluid, the surfactants. Besides providing low surface tension that allows efficient gas exchange at the alveoli, surfactants inhibit the invasion of epithelial cells by respiratory viruses, enhance pathogen uptake by phagocytes, and regulate immune cells' functions. The respiratory tract microbiota constitutes the third defense barrier against SARS-CoV-2. It activates the innate and adaptive immune cells and elicits anti-infectious molecules such as secretory IgA antibodies, defensins, and interferons. The fourth defense barrier comprises the antimicrobial peptides defensins, and lactoferrin. They show direct antiviral activity, inhibit viral fusion, and modulate the innate and adaptive immune responses. Secretory IgA antibodies, the fifth defense barrier, besides protecting the local microbiota against noxious agents, also inhibit SARS-CoV-2 cell invasion. If the virus overcomes this barrier, it reaches its target, the respiratory epithelial cells. However, these cells also act as a defense barrier, the sixth one, since they hinder the virus' access to receptors and produce antiviral and immunomodulatory molecules such as interferons, lactoferrin, and defensins. Finally, the sensing of the virus by the cells of innate immunity, the last constitutive defense barrier, elicits a cascade of signals that activate adaptive immune cells and may inhibit the development of productive infection. The subject of the present essay is discussing these mechanisms.
Collapse
Affiliation(s)
- Eduardo Tosta
- Professor Emeritus, Faculdade de Medicina, Universidade de Brasília, Brasília, DF , Brasil
| |
Collapse
|
15
|
Hu X, Luo H, Dou C, Chen X, Huang Y, Wang L, Xue S, Sun Z, Chen S, Xu Q, Geng T, Zhao X, Cui H. Metformin Triggers Apoptosis and Induction of the G0/G1 Switch 2 Gene in Macrophages. Genes (Basel) 2021; 12:genes12091437. [PMID: 34573418 PMCID: PMC8468785 DOI: 10.3390/genes12091437] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022] Open
Abstract
Metformin is a widely used antidiabetic drug for the treatment of type 2 diabetes and has been recently demonstrated to possess anti-inflammatory properties via AMPK-mediated modulation of M2 macrophage activation. However, the anti-inflammatory mechanisms of metformin on inflammatory macrophages are still not fully elucidated. In this study, we found that metformin induced apoptosis in macrophages. In particular, metformin induced apoptosis of M1 macrophages, based on M1 marker genes in apoptotic macrophages. Next, we comprehensively screened metformin-responsive genes in macrophages by RNA-seq and focused on the extrinsic apoptotic signaling pathway. The G0/G1 switch 2 gene (G0S2) was robustly up-regulated by metformin in macrophages. Overexpression of G0S2 significantly induced apoptosis of macrophages in a dose-dependent manner and blunted the function of the crucial anti-apoptotic gene Bcl-2, which was significantly reduced by metformin. These findings show that metformin promoted apoptosis of macrophages, especially M1 macrophages, via G0S2 induction and provides a novel anti-inflammatory mechanism of metformin through induction of macrophage apoptosis.
Collapse
Affiliation(s)
- Xuming Hu
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
- Department of Animal Science, McGill University, Montréal, QC H3A 0G4, Canada;
| | - Huan Luo
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Chunfeng Dou
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Xujing Chen
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Yi Huang
- Department of Pharmacy, Suzhou Vocational Health College, Suzhou 215009, China;
| | - Liping Wang
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Songlei Xue
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Zhen Sun
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Shihao Chen
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Qi Xu
- Department of Animal Science, McGill University, Montréal, QC H3A 0G4, Canada;
| | - Tuoyu Geng
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
| | - Xin Zhao
- Department of Animal Science, McGill University, Montréal, QC H3A 0G4, Canada;
- Correspondence: (X.Z.); (H.C.)
| | - Hengmi Cui
- Institute of Epigenetics and Epigenomics, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (X.H.); (H.L.); (C.D.); (X.C.); (L.W.); (S.X.); (Z.S.); (S.C.); (T.G.)
- Joint International Research Laboratory of Agricultural & Agri-Product Safety, Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
- Jiangsu Co-Innovation Center for Prevention & Control of Important Animal Infectious Diseases & Zoonoses, Yangzhou 225009, China
- Correspondence: (X.Z.); (H.C.)
| |
Collapse
|
16
|
Dang KR, Wu T, Hui YN, Du HJ. Newly-found functions of metformin for the prevention and treatment of age-related macular degeneration. Int J Ophthalmol 2021; 14:1274-1280. [PMID: 34414094 PMCID: PMC8342286 DOI: 10.18240/ijo.2021.08.20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
Metformin (MET), a first-line oral agent used to treat diabetes, exerts its function mainly by activating adenosine monophosphate-activated protein. The accumulation of oxidized phospholipids in the outer layer of the retina plays a key role in retinal pigment epithelium (RPE) cells death and the formation of choroidal neovascularization (CNV), which mean the development of age-related macular degeneration (AMD). Recent studies have shown that MET can regulate lipid metabolism, inhibit inflammation, and prohibit retinal cell death and CNV formation due to various pathological factors. Here, newly discovered functions of MET that may be used for the prevention and treatment of AMD were reviewed.
Collapse
Affiliation(s)
- Kuan-Rong Dang
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Tong Wu
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Yan-Nian Hui
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Hong-Jun Du
- Department of Ophthalmology, Xijing Hospital, Eye Institute of Chinese PLA, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
17
|
Das B, Sarkar C, Rawat VS, Kalita D, Deka S, Agnihotri A. Promise of the NLRP3 Inflammasome Inhibitors in In Vivo Disease Models. Molecules 2021; 26:4996. [PMID: 34443594 PMCID: PMC8399941 DOI: 10.3390/molecules26164996] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/28/2021] [Accepted: 08/10/2021] [Indexed: 12/26/2022] Open
Abstract
Nucleotide-binding oligomerization domain NOD-like receptors (NLRs) are conserved cytosolic pattern recognition receptors (PRRs) that track the intracellular milieu for the existence of infection, disease-causing microbes, as well as metabolic distresses. The NLRP3 inflammasome agglomerates are consequent to sensing a wide spectrum of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs). Certain members of the NLR family have been documented to lump into multimolecular conglomerates called inflammasomes, which are inherently linked to stimulation of the cysteine protease caspase-1. Following activation, caspase-1 severs the proinflammatory cytokines interleukin (IL)-1β and IL-18 to their biologically active forms, with consequent commencement of caspase-1-associated pyroptosis. This type of cell death by pyroptosis epitomizes a leading pathway of inflammation. Accumulating scientific documentation has recorded overstimulation of NLRP3 (NOD-like receptor protein 3) inflammasome involvement in a wide array of inflammatory conditions. IL-1β is an archetypic inflammatory cytokine implicated in multiple types of inflammatory maladies. Approaches to impede IL-1β's actions are possible, and their therapeutic effects have been clinically demonstrated; nevertheless, such strategies are associated with certain constraints. For instance, treatments that focus on systemically negating IL-1β (i.e., anakinra, rilonacept, and canakinumab) have been reported to result in an escalated peril of infections. Therefore, given the therapeutic promise of an NLRP3 inhibitor, the concerted escalated venture of the scientific sorority in the advancement of small molecules focusing on direct NLRP3 inflammasome inhibition is quite predictable.
Collapse
Affiliation(s)
- Biswadeep Das
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India;
| | - Chayna Sarkar
- Department of Clinical Pharmacology & Therapeutics, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences (NEIGRIHMS), Mawdiangdiang, Shillong 793018, Meghalaya, India;
| | - Vikram Singh Rawat
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India;
| | - Deepjyoti Kalita
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India; (D.K.); (S.D.)
| | - Sangeeta Deka
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India; (D.K.); (S.D.)
| | - Akash Agnihotri
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India;
| |
Collapse
|
18
|
Ahmadi A, Panahi Y, Johnston TP, Sahebkar A. Antidiabetic drugs and oxidized low-density lipoprotein: A review of anti-atherosclerotic mechanisms. Pharmacol Res 2021; 172:105819. [PMID: 34400317 DOI: 10.1016/j.phrs.2021.105819] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is one of the leading causes of mortality globally. Atherosclerosis is an important step towards different types of cardiovascular disease. The role of oxidized low-density lipoprotein (oxLDL) in the initiation and progression of atherosclerosis has been thoroughly investigated in recent years. Moreover, clinical trials have established that diabetic patients are at a greater risk of developing atherosclerotic plaques. Hence, we aimed to review the clinical and experimental impacts of various classes of antidiabetic drugs on the circulating levels of oxLDL. Metformin, pioglitazone, and dipeptidyl peptidase-4 inhibitors were clinically associated with a suppressive effect on oxLDL in patients with impaired glucose tolerance. However, there is an insufficient number of studies that have clinically evaluated the relationship between oxLDL and newer agents such as agonists of glucagon-like peptide 1 receptor or inhibitors of sodium-glucose transport protein 2. Next, we attempted to explore the multitude of mechanisms that antidiabetic agents exert to counter the undesirable effects of oxLDL in macrophages, endothelial cells, and vascular smooth muscle cells. In general, antidiabetic drugs decrease the uptake of oxLDL by vascular cells and reduce subsequent inflammatory signaling, which prevents macrophage adhesion and infiltration. Moreover, these agents suppress the oxLDL-induced transformation of macrophages into foam cells by either inhibiting oxLDL entrance, or by facilitating its efflux. Thus, the anti-inflammatory, anti-oxidant, and anti-apoptotic properties of antidiabetic agents abrogate changes induced by oxLDL, which can be extremely beneficial in controlling atherosclerosis in diabetic patients.
Collapse
Affiliation(s)
- Ali Ahmadi
- Pharmacotherapy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Yunes Panahi
- Pharmacotherapy Department, Faculty of Pharmacy, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; School of Medicine, The University of Western Asutralia, Perth, Australia; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948567, Iran.
| |
Collapse
|
19
|
Liang JJ, Fraser IDC, Bryant CE. Lipid regulation of NLRP3 inflammasome activity through organelle stress. Trends Immunol 2021; 42:807-823. [PMID: 34334306 DOI: 10.1016/j.it.2021.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/10/2021] [Accepted: 07/11/2021] [Indexed: 12/14/2022]
Abstract
Inflammation driven by the NLRP3 inflammasome in macrophages is an important contributor to chronic metabolic diseases that affect growing numbers of individuals. Many of these diseases involve the pathologic accumulation of endogenous lipids or their oxidation products, which can activate NLRP3. Other endogenous lipids, however, can inhibit the activation of NLRP3. The intracellular mechanisms by which these lipids modulate NLRP3 activity are now being identified. This review discusses emerging evidence suggesting that organelle stress, particularly involving mitochondria, lysosomes, and the endoplasmic reticulum, may be key in lipid-induced modification of NLRP3 inflammasome activity.
Collapse
Affiliation(s)
- Jonathan J Liang
- Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Iain D C Fraser
- Signaling Systems Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| | - Clare E Bryant
- Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
20
|
Feng X, Chen W, Ni X, Little PJ, Xu S, Tang L, Weng J. Metformin, Macrophage Dysfunction and Atherosclerosis. Front Immunol 2021; 12:682853. [PMID: 34163481 PMCID: PMC8215340 DOI: 10.3389/fimmu.2021.682853] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Metformin is one of the most widely prescribed hypoglycemic drugs and has the potential to treat many diseases. More and more evidence shows that metformin can regulate the function of macrophages in atherosclerosis, including reducing the differentiation of monocytes and inhibiting the inflammation, oxidative stress, polarization, foam cell formation and apoptosis of macrophages. The mechanisms by which metformin regulates the function of macrophages include AMPK, AMPK independent targets, NF-κB, ABCG5/8, Sirt1, FOXO1/FABP4 and HMGB1. On the basis of summarizing these studies, we further discussed the future research directions of metformin: single-cell RNA sequencing, neutrophil extracellular traps (NETs), epigenetic modification, and metformin-based combination drugs. In short, macrophages play an important role in a variety of diseases, and improving macrophage dysfunction may be an important mechanism for metformin to expand its pleiotropic pharmacological profile. In addition, the combination of metformin with other drugs that improve the function of macrophages (such as SGLT2 inhibitors, statins and IL-1β inhibitors/monoclonal antibodies) may further enhance the pleiotropic therapeutic potential of metformin in conditions such as atherosclerosis, obesity, cancer, dementia and aging.
Collapse
Affiliation(s)
- Xiaojun Feng
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Wenxu Chen
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Xiayun Ni
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Peter J. Little
- Sunshine Coast Health Institute, University of the Sunshine Coast, Birtinya, QLD, Australia
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD, Australia
| | - Suowen Xu
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| | - Liqin Tang
- Department of Pharmacy, the First Affiliated Hospital of University of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China (USTC), Hefei, China
| | - Jianping Weng
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China( USTC), Hefei, China
| |
Collapse
|
21
|
Yue H, Yang Z, Ou Y, Liang S, Deng W, Chen H, Zhang C, Hua L, Hu W, Sun P. Tanshinones inhibit NLRP3 inflammasome activation by alleviating mitochondrial damage to protect against septic and gouty inflammation. Int Immunopharmacol 2021; 97:107819. [PMID: 34098486 DOI: 10.1016/j.intimp.2021.107819] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/27/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023]
Abstract
Tanshinones, the active ingredients derived from the roots of Salvia miltiorrhiza, have been widely used as traditional medicinal herbs for treating human diseases. Although tanshinones showed anti-inflammatory effects in many studies, large knowledge gaps remain regarding their underlying mechanisms. Here, we identified 15 tanshinones that suppressed the activation of NLRP3 inflammasome and studied their structure-activity relationships. Three tanshinones (tanshinone IIA, isocryptotanshinone, and dihydrotanshinone I) reduced mitochondrial reactive-oxygen species production in lipopolysaccharide (LPS)/nigericin-stimulated macrophages and correlated with altered mitochondrial membrane potentials, mitochondria complexes activities, and adenosine triphosphate and protonated-nicotinamide adenine dinucleotide production. The tanshinones may confer mitochondrial protection by promoting autophagy and the AMP-activated protein kinase pathway. Importantly, our findings demonstrate that dihydrotanshinone I improved the survival of mice with LPS shock and ameliorated inflammatory responses in septic and gouty animals. Our results suggest a potential pharmacological mechanism whereby tanshinones can effectively treat inflammatory diseases, such as septic and gouty inflammation.
Collapse
Affiliation(s)
- Hu Yue
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Zhongjin Yang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yitao Ou
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Shuli Liang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Wenmin Deng
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Hao Chen
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Cheng Zhang
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Lei Hua
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Wenhui Hu
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China.
| | - Ping Sun
- The Fifth Affiliated Hospital, Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
22
|
ZBTB20 Positively Regulates Oxidative Stress, Mitochondrial Fission, and Inflammatory Responses of ox-LDL-Induced Macrophages in Atherosclerosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5590855. [PMID: 33777314 PMCID: PMC7972849 DOI: 10.1155/2021/5590855] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/26/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023]
Abstract
Atherosclerosis (AS) is one of the most serious and common cardiovascular diseases affecting human health. AS is featured by the accumulation of plaques in vessel walls. The pathophysiology of AS is relevant in the low-density lipoprotein (LDL) uptake by macrophages, as well as the conversion of macrophages to foam cells. However, the mechanisms about how macrophages regulate AS have not been fully elucidated. In this study, we aimed to illuminate the roles of ZBTB20 and to excavate the underlying regulative mechanisms of ZBTB20 in AS. The microarray analysis revealed that ZBTB20 was a hub gene in the oxidative stress and inflammatory responses induced by oxidized LDL (ox-LDL) in AS. Correspondingly, our validation studies showed that ZBTB20 increased in either the human atherosclerotic lesion or the ox-LDL-stimulated macrophages. Moreover, the knockdown of ZBTB20 decreased M1 polarization, suppressed the proinflammatory factors, inhibited mitochondrial fission, and reduced the oxidative stress level of macrophages induced by ox-LDL. The mechanistic studies revealed that the ZBTB20 knockdown suppressed NF-κB/MAPK activation and attenuated the mitochondrial fission possibly via regulating the nucleus translocation of NRF2, a pivotal transcription factor on redox homeostasis. Our in vivo studies showed that the sh-ZBTB20 adenovirus injection could reduce the progression of AS in apolipoprotein E-deficient (ApoE−/−) mice. All in all, these results suggested that ZBTB20 positively regulated the oxidative stress level, mitochondrial fission, and inflammatory responses of macrophages induced by ox-LDL, and the knockdown of ZBTB20 could attenuate the development of AS in ApoE−/− mice.
Collapse
|
23
|
Non-alcoholic fatty liver disease: a metabolic burden promoting atherosclerosis. Clin Sci (Lond) 2021; 134:1775-1799. [PMID: 32677680 DOI: 10.1042/cs20200446] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/06/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the fastest growing chronic liver disease, with a prevalence of up to 25% worldwide. Individuals with NAFLD have a high risk of disease progression to cirrhosis, hepatocellular carcinoma (HCC), and liver failure. With the exception of intrahepatic burden, cardiovascular disease (CVD) and especially atherosclerosis (AS) are common complications of NAFLD. Furthermore, CVD is a major cause of death in NAFLD patients. Additionally, AS is a metabolic disorder highly associated with NAFLD, and individual NAFLD pathologies can greatly increase the risk of AS. It is increasingly clear that AS-associated endothelial cell damage, inflammatory cell activation, and smooth muscle cell proliferation are extensively impacted by NAFLD-induced systematic dyslipidemia, inflammation, oxidative stress, the production of hepatokines, and coagulations. In clinical trials, drug candidates for NAFLD management have displayed promising effects for the treatment of AS. In this review, we summarize the key molecular events and cellular factors contributing to the metabolic burden induced by NAFLD on AS, and discuss therapeutic strategies for the improvement of AS in individuals with NAFLD.
Collapse
|
24
|
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: 22] [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
|
25
|
Yan Z, Qi Z, Yang X, Ji N, Wang Y, Shi Q, Li M, Zhang J, Zhu Y. The NLRP3 inflammasome: Multiple activation pathways and its role in primary cells during ventricular remodeling. J Cell Physiol 2021; 236:5547-5563. [PMID: 33469931 DOI: 10.1002/jcp.30285] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/18/2022]
Abstract
Inflammasomes are a group of multiprotein signaling complexes located in the cytoplasm. Several inflammasomes have been identified, including NLRP1, NLRP2, NLRP3, AIM2, and NLRC4. Among them, NLRP3 was investigated in most detail, and it was reported that it can be activated by many different stimuli. Increased NLRP3 protein expression and inflammasome assembly lead to caspase-1 mediated maturation and release of IL-1β, which triggers inflammation and pyroptosis. The activation of the NLRP3 inflammasome has been widely reported in studies of tumors and neurological diseases, but relatively few studies on the cardiovascular system. Ventricular remodeling (VR) is an important factor contributing to heart failure (HF) after myocardial infarction (MI). Consequently, delaying VR is of great significance for improving heart function. Studies have shown that the NLRP3 inflammasome plays an essential role in the process of VR. Here, we reviewed the latest studies on the activation pathway of the NLRP3 inflammasome, focusing on the effects of the NLRP3 inflammasome in primary cells during VR, and finally discuss future research directions in this field.
Collapse
Affiliation(s)
- Zhipeng Yan
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhongwen Qi
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoya Yang
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Nan Ji
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yueyao Wang
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qi Shi
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Meng Li
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Junping Zhang
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yaping Zhu
- Department of Cardiology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
26
|
Mestareehi A, Zhang X, Seyoum B, Msallaty Z, Mallisho A, Burghardt KJ, Kowluru A, Yi Z. Metformin Increases Protein Phosphatase 2A Activity in Primary Human Skeletal Muscle Cells Derived from Lean Healthy Participants. J Diabetes Res 2021; 2021:9979234. [PMID: 34368369 PMCID: PMC8342103 DOI: 10.1155/2021/9979234] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 07/16/2021] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE To investigate if PP2A plays a role in metformin-induced insulin sensitivity improvement in human skeletal muscle cells. Participants. Eight lean insulin-sensitive nondiabetic participants (4 females and 4 males; age: 21.0 ± 1.0 years; BMI: 22.0 ± 0.7 kg/m2; 2-hour OGTT: 97.0 ± 6.0 mg/dl; HbA1c: 5.3 ± 0.1%; fasting plasma glucose: 87.0 ± 2.0 mg/dl; M value; 11.0 ± 1.0 mg/kgBW/min). DESIGN A hyperinsulinemic-euglycemic clamp was performed to assess insulin sensitivity in human subjects, and skeletal muscle biopsy samples were obtained. Primary human skeletal muscle cells (shown to retain metabolic characteristics of donors) were cultured from these muscle biopsies that included 8 lean insulin-sensitive participants. Cultured cells were expanded, differentiated into myotubes, and treated with 50 μM metformin for 24 hours before harvesting. PP2Ac activity was measured by a phosphatase activity assay kit (Millipore) according to the manufacturer's protocol. RESULTS The results indicated that metformin significantly increased the activity of PP2A in the myotubes for all 8 lean insulin-sensitive nondiabetic participants, and the average fold increase is 1.54 ± 0.11 (P < 0.001). CONCLUSIONS These results provided the first evidence that metformin can activate PP2A in human skeletal muscle cells derived from lean healthy insulin-sensitive participants and may help to understand metformin's action in skeletal muscle in humans.
Collapse
Affiliation(s)
- Aktham Mestareehi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Xiangmin Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Berhane Seyoum
- Division of Endocrinology, Wayne State University School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Zaher Msallaty
- Division of Endocrinology, Wayne State University School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Abdullah Mallisho
- Division of Endocrinology, Wayne State University School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Kyle Jon Burghardt
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy/Health Sciences, Wayne State University, Detroit, MI, USA
| | - Anjaneyulu Kowluru
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Program for Translational Research in Diabetes, Biomedical Research Service, John D. Dingell VA Medical Center, Detroit, MI 48201, USA
| | - Zhengping Yi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
27
|
The effect of metformin on carotid intima-media thickness (CIMT): A systematic review and meta-analysis of randomized clinical trials. Eur J Pharmacol 2020; 886:173458. [PMID: 32763300 DOI: 10.1016/j.ejphar.2020.173458] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/24/2020] [Accepted: 07/31/2020] [Indexed: 12/17/2022]
Abstract
Metformin administration has been reported to influence the carotid intima-media thickness (CIMT) in humans. However, since previously conducted studies have yielded inconsistent results, the exact effect of metformin on CIMT remains unclear. Causes that could lead to inconsistency in reported research could be the duration and dose of the intervention, as well as the sample size. To address this inconsistency, we conducted a systematic review and meta-analysis to evaluate the influence of metformin on CIMT in human subjects. We identified eligible studies by searching several electronic databases (EMBASE, PubMed-MEDLINE, Web of Science and Google Scholar) up to December 12, 2019. Data were pooled using the random-effects model. Combining data from 1087 participants (9 studies), our meta-analysis revealed that the administration of metformin resulted in a significant reduction in CIMT (WMD = -0.049 mm; 95% CI: -0.095, -0.004). Stratified analyses showed that an intervention lasting ≥12 months (WMD: -0.084 mm, 95% CI: -0.145, -0.024) and an intake of metformin ≤1500 mg/day (WMD: -0.081 mm, 95% CI: -0.132, -0.029) resulted in a significantly greater reduction in CIMT. However, an intervention duration of less than 12 months and an intake of metformin ˃1500 mg/day yielded no significant effects on CIMT. The results of the current study confirm that metformin administration is associated with a significant reduction in CIMT. Taking into account that CIMT reflects the burden of atherosclerosis, the clinical utility of metformin might also be related to its anti-atherogenic effects.
Collapse
|
28
|
NLPR3 Inflammasomes and Their Significance for Atherosclerosis. Biomedicines 2020; 8:biomedicines8070205. [PMID: 32664349 PMCID: PMC7399980 DOI: 10.3390/biomedicines8070205] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/04/2020] [Accepted: 07/08/2020] [Indexed: 01/08/2023] Open
Abstract
Atherosclerosis is a serious disorder, with numerous potential complications such as cardiovascular disease, ischemic stroke, and myocardial infarction. The origin of atherosclerosis is related to chronic inflammation, lipid metabolism alterations, and oxidative stress. Inflammasomes are the cytoplasmic multiprotein complex triggering the activation of inflammatory response. NLRP3 inflammasomes have a specific activation pathway that involves numerous stimuli, including a wide range of PAMPs and DAMPs. Recent studies of atherosclerotic pathology are focused on the mitochondria that appear to be a promising target for therapeutic approach development. Mitochondria are the main source of reactive oxygen species (ROS) associated with oxidative stress. It was previously shown that NLRP3 inflammasome activation results in mitochondrial damage, but the exact mechanisms of this need to be specified. In this review, we focused on the features of NLRP3 inflammasomes and their significance for atherosclerosis, especially concerning mitochondria.
Collapse
|
29
|
MLKL Aggravates Ox-LDL-Induced Cell Pyroptosis via Activation of NLRP3 Inflammasome in Human Umbilical Vein Endothelial Cells. Inflammation 2020; 43:2222-2231. [DOI: 10.1007/s10753-020-01289-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
30
|
NLRP3 Inflammasome: A Potential Alternative Therapy Target for Atherosclerosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1561342. [PMID: 32328119 PMCID: PMC7150718 DOI: 10.1155/2020/1561342] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/21/2020] [Accepted: 03/04/2020] [Indexed: 12/12/2022]
Abstract
Atherosclerosis (AS) is a complex and chronic inflammatory disease that occurs in multiple systems of the human body. It is an important pathological basis for a variety of diseases and a serious threat to human health. So far, many theories have been formed to explain the pathogenesis of atherosclerosis, among which “inflammation theory” has gradually become a research focus. This theory presents that inflammatory response runs through the whole progress of AS, inflammatory cells play as the main executors of AS, and inflammatory mediators are the key molecules of AS. In the inflammatory process of atherosclerosis, the role of NLRP3 in the atherosclerosis has gradually got the attention of researchers. NLRP3 is a kind of signal-transductional pattern recognition receptors (PRRs). After recognizing and binding to the damage factors, NLRP3 inflammasome will be assembled to activate IL-1β and caspase-1 pathways, resulting in promoting the inflammation process of AS, reducing the stability of the plaques, and finally increasing the incidence of adverse cardiovascular events. Taken above, the article will review the potential benefits of drugs targeting the NLRP3 inflammasome in the therapy of AS.
Collapse
|
31
|
Clark AR, Ohlmeyer M. Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration. Pharmacol Ther 2019; 201:181-201. [PMID: 31158394 PMCID: PMC6700395 DOI: 10.1016/j.pharmthera.2019.05.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric enzyme that catalyzes the selective removal of phosphate groups from protein serine and threonine residues. Emerging evidence suggests that it functions as a tumor suppressor by constraining phosphorylation-dependent signalling pathways that regulate cellular transformation and metastasis. Therefore, PP2A-activating drugs (PADs) are being actively sought and investigated as potential novel anti-cancer treatments. Here we explore the concept that PP2A also constrains inflammatory responses through its inhibitory effects on various signalling pathways, suggesting that PADs may be effective in the treatment of inflammation-mediated pathologies.
Collapse
Affiliation(s)
- Andrew R Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
| | | |
Collapse
|
32
|
Uetrecht J. Mechanistic Studies of Idiosyncratic DILI: Clinical Implications. Front Pharmacol 2019; 10:837. [PMID: 31402866 PMCID: PMC6676790 DOI: 10.3389/fphar.2019.00837] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 07/01/2019] [Indexed: 12/18/2022] Open
Abstract
The idiosyncratic nature of idiosyncratic drug-induced liver injury (IDILI) makes mechanistic studies very difficult, and little is known with certainty. However, the fact that the IDILI caused by some drugs is associated with specific HLA genotypes provides strong evidence that it is mediated by the adaptive immune system. This is also consistent with the histology and the general characteristics of IDILI. However, there are other mechanistic hypotheses. Various in vitro and in vivo systems have been used to test hypotheses. Two other hypotheses are mitochondrial injury and inhibition of the bile salt export pump. It is possible that these mechanisms are responsible for some cases of IDILI or that these mechanisms are complementary and are involved in initiating an immune response. In general, it is believed that the initiation of an immune response requires activation of antigen-presenting cells by molecules such as danger-associated molecular pattern molecules (DAMPs). An attractive hypothesis for the mechanism by which DAMPs induce an immune response is through the activation of inflammasomes. The dominant immune response in the liver is immune tolerance, and it is only when immune tolerance fails that significant liver injury occurs. Consistent with this concept, an animal model was developed in which immune checkpoint inhibition unmasked the ability of drugs to cause liver injury. Although it appears that the liver damage is mediated by the adaptive immune system, an innate immune response is required for an adaptive immune response. The innate immune response is not dependent on specific HLA genes or T cell receptors and may occur in most patients and animals treated with a drug that can cause IDILI. Studies of the subclinical innate immune response to drugs may provide important mechanistic clues and provide a method to screen drugs for their potential to cause IDILI.
Collapse
Affiliation(s)
- Jack Uetrecht
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
| |
Collapse
|