1
|
Ali A, Azmat U, Ji Z, Khatoon A, Murtaza B, Akbar K, Irshad U, Raza R, Su Z. Beyond Genes: Epiregulomes as Molecular Commanders in Innate Immunity. Int Immunopharmacol 2024; 142:113149. [PMID: 39278059 DOI: 10.1016/j.intimp.2024.113149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/09/2024] [Accepted: 09/08/2024] [Indexed: 09/17/2024]
Abstract
The natural fastest way to deal with pathogens or danger signals is the innate immune system. This system prevents too much inflammation and tissue damage and efficiently eliminates pathogens. The epiregulome is the chromatin structure influenced by epigenetic factors and linked to cis-regulatory elements (CREs). The epiregulome helps to end the inflammatory response and also assists innate immune cells to show specific action by making cell-specific gene expression patterns. This inspection unfolds two concepts: (1) how epiregulomes are shaped by switching the expression levels of genes, manoeuvre enzyme activity and earmark of chromatin modifiers on specific genes; during and after the infection, and (2) how the expression of specific genes (aids in prompt management of innate cell growth, or the reaction to aggravation and illness) command by epiregulomes that formed during the above process. In this review, the consequences of intrinsic immuno-metabolic remodelling on epiregulomes and potential difficulties in identifying the master epiregulome that regulates innate immunity and inflammation have been discussed.
Collapse
Affiliation(s)
- Ashiq Ali
- Department of Histology and Embryology, Shantou University Medical College, China.
| | - Urooj Azmat
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Ziyi Ji
- Department of Histology and Embryology, Shantou University Medical College, China
| | - Aisha Khatoon
- Department of Pathology, University of Agriculture Faisalabad, Pakistan
| | - Bilal Murtaza
- School of Bioengineering, Dalian University of Science and Technology, Dalian, China
| | - Kaynaat Akbar
- Department of Zoology, Wildlife and Fisheries, University of Agriculture, Faisalabad, Pakistan
| | - Urooj Irshad
- Department Biological Sciences, Faculty of Sciences, Superior University Lahore, Punjab, Pakistan
| | - Rameen Raza
- Department of Pathology, University of Agriculture Faisalabad, Pakistan
| | - Zhongjing Su
- Department of Histology and Embryology, Shantou University Medical College, China.
| |
Collapse
|
2
|
Zuriaga MA, Yu Z, Matesanz N, Truong B, Ramos-Neble BL, Asensio-López MC, Uddin MM, Nakao T, Niroula A, Zorita V, Amorós-Pérez M, Moro R, Ebert BL, Honigberg MC, Pascual-Figal D, Natarajan P, Fuster JJ. Colchicine prevents accelerated atherosclerosis in TET2-mutant clonal haematopoiesis. Eur Heart J 2024; 45:4601-4615. [PMID: 39212933 PMCID: PMC11560281 DOI: 10.1093/eurheartj/ehae546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/14/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND AND AIMS Somatic mutations in the TET2 gene that lead to clonal haematopoiesis (CH) are associated with accelerated atherosclerosis development in mice and a higher risk of atherosclerotic disease in humans. Mechanistically, these observations have been linked to exacerbated vascular inflammation. This study aimed to evaluate whether colchicine, a widely available and inexpensive anti-inflammatory drug, prevents the accelerated atherosclerosis associated with TET2-mutant CH. METHODS In mice, TET2-mutant CH was modelled using bone marrow transplantations in atherosclerosis-prone Ldlr-/- mice. Haematopoietic chimeras carrying initially 10% Tet2-/- haematopoietic cells were fed a high-cholesterol diet and treated with colchicine or placebo. In humans, whole-exome sequencing data and clinical data from 37 181 participants in the Mass General Brigham Biobank and 437 236 participants in the UK Biobank were analysed to examine the potential modifying effect of colchicine prescription on the relationship between CH and myocardial infarction. RESULTS Colchicine prevented accelerated atherosclerosis development in the mouse model of TET2-mutant CH, in parallel with suppression of interleukin-1β overproduction in conditions of TET2 loss of function. In humans, patients who were prescribed colchicine had attenuated associations between TET2 mutations and myocardial infarction. This interaction was not observed for other mutated genes. CONCLUSIONS These results highlight the potential value of colchicine to mitigate the higher cardiovascular risk of carriers of somatic TET2 mutations in blood cells. These observations set the basis for the development of clinical trials that evaluate the efficacy of precision medicine approaches tailored to the effects of specific mutations linked to CH.
Collapse
Affiliation(s)
- María A Zuriaga
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Zhi Yu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
| | - Nuria Matesanz
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Buu Truong
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
| | - Beatriz L Ramos-Neble
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Mari C Asensio-López
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Cardiology Department, Hospital Virgen de la Arrixaca, IMIB-Arrixaca and University of Murcia, Murcia, Spain
| | - Md Mesbah Uddin
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
| | - Tetsushi Nakao
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Abhishek Niroula
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- SciLifeLab, University of Gothenburg, Gothenburg, Sweden
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Virginia Zorita
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Marta Amorós-Pérez
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Rosa Moro
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael C Honigberg
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Domingo Pascual-Figal
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- Cardiology Department, Hospital Virgen de la Arrixaca, IMIB-Arrixaca and University of Murcia, Murcia, Spain
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| | - Pradeep Natarajan
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, 75 Ames St., Cambridge, MA 02142, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, 185 Cambridge Street, CPZN 3.184, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, 25 Shattuck St., Boston, MA 02115, USA
| | - José J Fuster
- Program on Novel Mechanisms of Atherosclerosis, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Madrid, Spain
| |
Collapse
|
3
|
Kumar V, Stewart Iv JH. Platelet's plea to Immunologists: Please do not forget me. Int Immunopharmacol 2024; 143:113599. [PMID: 39547015 DOI: 10.1016/j.intimp.2024.113599] [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: 07/09/2024] [Revised: 10/07/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
Platelets are non-nucleated mammalian cells originating from the cytoplasmic expulsion of the megakaryocytes. Megakaryocytes develop during hematopoiesis through megakaryopoiesis, whereas platelets develop from megakaryocytes through thrombopoiesis. Since their first discovery, platelets have been studied as critical cells controlling hemostasis or blood coagulation. However, coagulation and innate immune response are evolutionarily linked processes. Therefore, it has become critical to investigate the immunological functions of platelets to maintain immune homeostasis. Advances in immunology and platelet biology research have explored different critical roles of platelets, including phagocytosis, release of different immune mediators, and controlling functions of different immune cells by direct interaction and immune mediators. The current article discusses platelet's development and their critical role as innate immune cells, which express different pattern recognition receptors (PRRs), recognizing different pathogen or microbe-associated molecular patterns (PAMPs or MAMPs) and death/damage-associated molecular patterns (DAMPs) and their direct interactions with innate and adaptive immune cells to maintain immune homeostasis.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Medical Education Building-C, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310 USA.
| | - John H Stewart Iv
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Medical Education Building-C, Morehouse School of Medicine, 720 Westview Drive, Atlanta, GA 30310 USA
| |
Collapse
|
4
|
Wen S, Arakawa H, Yokoyama S, Shirasaka Y, Higashida H, Tamai I. Functional identification of soluble uric acid as an endogenous inhibitor of CD38. eLife 2024; 13:RP96962. [PMID: 39527634 PMCID: PMC11554305 DOI: 10.7554/elife.96962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Excessive elevation or reduction of soluble uric acid (sUA) levels has been linked to some of pathological states, raising another subject that sUA at physiological levels may be essential for the maintenance of health. Yet, the fundamental physiological functions and molecular targets of sUA remain largely unknown. Using enzyme assays and in vitro and in vivo metabolic assays, we demonstrate that sUA directly inhibits the hydrolase and cyclase activities of CD38 via a reversible non-competitive mechanism, thereby limiting nicotinamide adenine dinucleotide (NAD+) degradation. CD38 inhibition is restricted to sUA in purine metabolism, and a structural comparison using methyl analogs of sUA such as caffeine metabolites shows that 1,3-dihydroimidazol-2-one is the main functional group. Moreover, sUA at physiological levels prevents crude lipopolysaccharide (cLPS)-induced systemic inflammation and monosodium urate (MSU) crystal-induced peritonitis in mice by interacting with CD38. Together, this study unveils an unexpected physiological role for sUA in controlling NAD+ availability and innate immunity through CD38 inhibition, providing a new perspective on sUA homeostasis and purine metabolism.
Collapse
Affiliation(s)
- Shijie Wen
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa UniversityKanazawaJapan
| | - Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa UniversityKanazawaJapan
| | - Shigeru Yokoyama
- Research Center for Child Mental Development, Kanazawa UniversityKanazawaJapan
- Division of Socio-Cognitive-Neuroscience, United Graduate School of Child Development, Osaka University, Kanazawa University, Hamamatsu University School of Medicine, Chiba University and University of FukuiKanazawaJapan
| | - Yoshiyuki Shirasaka
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa UniversityKanazawaJapan
| | - Haruhiro Higashida
- Research Center for Child Mental Development, Kanazawa UniversityKanazawaJapan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa UniversityKanazawaJapan
| |
Collapse
|
5
|
Yan HW, Feng YD, Tang N, Cao FC, Lei YF, Cao W, Li XQ. Viral myocarditis: From molecular mechanisms to therapeutic prospects. Eur J Pharmacol 2024; 982:176935. [PMID: 39182550 DOI: 10.1016/j.ejphar.2024.176935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/10/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Myocarditis is characterized as local or diffuse inflammatory lesions in the myocardium, primarily caused by viruses and other infections. It is a common cause of sudden cardiac death and dilated cardiomyopathy. In recent years, the global prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the widespread vaccination have coincided with a notable increase in the number of reported cases of myocarditis. In light of the potential threat that myocarditis poses to global public health, numerous studies have sought to elucidate the pathogenesis of this condition. However, despite these efforts, effective treatment strategies remain elusive. To collate the current research advances in myocarditis, and thereby provide possible directions for further research, this review summarizes the mechanisms involved in viral invasion of the organism and primarily focuses on how viruses trigger excessive inflammatory responses and in result in different types of cell death. Furthermore, this article outlines existing therapeutic approaches and potential therapeutic targets for the acute phase of myocarditis. In particular, immunomodulatory treatments are emphasized and suggested as the most extensively studied and clinically promising therapeutic options.
Collapse
Affiliation(s)
- Han-Wei Yan
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| | - Ying-Da Feng
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| | - Na Tang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| | - Feng-Chuan Cao
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| | - Ying-Feng Lei
- Department of Microbiology, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| | - Wei Cao
- Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China; Department of Pharmacy, School of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xiao-Qiang Li
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China; Key Laboratory of Gastrointestinal Pharmacology of Chinese Materia Medica of the State Administration of Traditional Chinese Medicine, Department of Pharmacology, School of Pharmacy, Air Force Medical University, Xi'an, Shaanxi, 710032, China.
| |
Collapse
|
6
|
Tercan H, van Broekhoven A, Bahrar H, Opstal T, Cossins BC, Rother N, Rodwell L, Bekkering S, El Messaoudi S, Riksen NP, Cornel JH. The Effect of Low-Dose Colchicine on the Phenotype and Function of Neutrophils and Monocytes in Patients with Chronic Coronary Artery Disease: A Double-Blind Randomized Placebo-Controlled Cross-Over Study. Clin Pharmacol Ther 2024; 116:1325-1333. [PMID: 39115262 DOI: 10.1002/cpt.3394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/03/2024] [Indexed: 10/16/2024]
Abstract
Recent landmark trials showed that colchicine provides a substantial benefit in reducing major cardiovascular events in patients with coronary artery disease. Yet, its exact mechanism of action is still poorly understood. This study aimed to unravel the effect of colchicine on monocyte and neutrophil phenotype and function. A randomized double-blind placebo-controlled cross-over intervention study was executed in patients with a history of myocardial infarction. In neutrophils, colchicine treatment decreased CD62L expression and NGAL release upon ex vivo stimulation and increased PMA-induced ROS production. The effects of colchicine on monocytes were limited to a decrease in HLA-DR expression in the intermediate and nonclassical monocytes. Also, on the level of RNA expression, colchicine did not affect monocyte phenotype, while affecting various immunomodulating genes in neutrophils. Overall, our study suggests that treatment with colchicine affects neutrophil function, particularly by reducing neutrophil recruitment, lowering concentrations of NGAL, and changing the expression of various genes with immunomodulatory potential, whereas the effect on monocytes is limited.
Collapse
Affiliation(s)
- Helin Tercan
- Department of Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Amber van Broekhoven
- Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Harsh Bahrar
- Department of Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Tjerk Opstal
- Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
| | - Benjamin C Cossins
- Department of Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Nils Rother
- Department of Nephrology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Laura Rodwell
- Section Biostatistics, IQ Health, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Niels P Riksen
- Department of Internal Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Jan H Cornel
- Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
- Department of Cardiology, Northwest Clinics, Alkmaar, The Netherlands
- Dutch Network for Cardiovascular Research (WCN), Utrecht, The Netherlands
| |
Collapse
|
7
|
Gupta S, Cassel SL, Sutterwala FS, Dagvadorj J. Regulation of the NLRP3 inflammasome by autophagy and mitophagy. Immunol Rev 2024. [PMID: 39417249 DOI: 10.1111/imr.13410] [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] [Indexed: 10/19/2024]
Abstract
The NLRP3 inflammasome is a multiprotein complex that upon activation by the innate immune system drives a broad inflammatory response. The primary initial mediators of this response are pro-IL-1β and pro-IL-18, both of which are in an inactive form. Formation and activation of the NLRP3 inflammasome activates caspase-1, which cleaves pro-IL-1β and pro-IL-18 and triggers the formation of gasdermin D pores. Gasdermin D pores allow for the secretion of active IL-1β and IL-18 initiating the organism-wide inflammatory response. The NLRP3 inflammasome response can be beneficial to the host; however, if the NLRP3 inflammasome is inappropriately activated it can lead to significant pathology. While the primary components of the NLRP3 inflammasome are known, the precise details of assembly and activation are less well defined and conflicting. Here, we discuss several of the proposed pathways of activation of the NLRP3 inflammasome. We examine the role of subcellular localization and the reciprocal regulation of the NLRP3 inflammasome by autophagy. We focus on the roles of mitochondria and mitophagy in activating and regulating the NLRP3 inflammasome. Finally, we detail the impact of pathologic NLRP3 responses in the development and manifestations of pulmonary disease.
Collapse
Affiliation(s)
- Suman Gupta
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Suzanne L Cassel
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Fayyaz S Sutterwala
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jargalsaikhan Dagvadorj
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
8
|
Zhang X, Shi S, Du Y, Chai R, Guo Z, Duan C, Wang H, Hu Y, Chang X, Du B. Shaping cardiac destiny: the role of post-translational modifications on endoplasmic reticulum - mitochondria crosstalk in cardiac remodeling. Front Pharmacol 2024; 15:1423356. [PMID: 39464632 PMCID: PMC11502351 DOI: 10.3389/fphar.2024.1423356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 09/23/2024] [Indexed: 10/29/2024] Open
Abstract
Cardiac remodeling is a shared pathological change in most cardiovascular diseases. Encompassing both adaptive physiological responses and decompensated pathological changes. Anatomically, atrial remodeling is primarily caused by atrial fibrillation, whereas ventricular remodeling is typically induced by myocardial infarction, hypertension, or cardiomyopathy. Mitochondria, the powerhouse of cardiomyocytes, collaborate with other organelles such as the endoplasmic reticulum to control a variety of pathophysiological processes such as calcium signaling, lipid transfer, mitochondrial dynamics, biogenesis, and mitophagy. This mechanism is proven to be essential for cardiac remodeling. Post-translational modifications can regulate intracellular signaling pathways, gene expression, and cellular stress responses in cardiac cells by modulating protein function, stability, and interactions, consequently shaping the myocardial response to injury and stress. These modifications, in particular phosphorylation, acetylation, and ubiquitination, are essential for the regulation of the complex molecular pathways that underlie cardiac remodeling. This review provides a comprehensive overview of the crosstalk between the endoplasmic reticulum and mitochondria during cardiac remodeling, focusing on the regulatory effects of various post-translational modifications on these interactions.
Collapse
Affiliation(s)
- Xiaohan Zhang
- Department of Cardiology, Guang’Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shuqing Shi
- Department of Internal Medicine, Guang’Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yihang Du
- Department of Cardiology, Guang’Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruoning Chai
- Department of Cardiology, Guang’Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zezhen Guo
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Chenglin Duan
- Department of Cardiology, Guang’Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Huan Wang
- Department of Cardiology, Guang’Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuanhui Hu
- Department of Cardiology, Guang’Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xing Chang
- Department of Cardiology, Guang’Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bai Du
- Department of Cardiology, Guang’Anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
9
|
Tuersuntuoheti M, Zhou L, Li J, Yang S, Zhou S, Gong H. Investigation of crucial genes and mitochondrial function impairment in diabetic cardiomyopathy. Gene 2024; 923:148563. [PMID: 38754569 DOI: 10.1016/j.gene.2024.148563] [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: 02/01/2024] [Revised: 04/10/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
BACKGROUND Diabetic cardiomyopathy (DCM) is a special type of cardiovascular disease, termed as a situation of abnormal myocardial structure and function that occurs in diabetic patients. However, the most fundamental mechanisms of DCM have not been fully explicated, and useful targets for the therapeutic strategies still need to be explored. METHODS In the present study, we combined bioinformatics analysis and in vitro experiments throughout the process of DCM. Differentially Expressed Genes (DEGs) analysis was performed and the weighted gene co-expression network analysis (WGCNA) was constructed to determine the crucial genes that were tightly connected to DCM. Additionally, Functional enrichment analysis was conducted to define biological pathways. To identify the specific molecular mechanism, the human cardiomyocyte cell line (AC16) was stimulated by high glucose (HG, 50 mM D-glucose) and used to imitate DCM condition. Then, we tentatively examined the effect of high glucose on cardiomyocytes, the expression levels of crucial genes were further validated by in vitro experiments. RESULTS Generally, NPPA, IGFBP5, SERPINE1, and C3 emerged as potential therapeutic targets. Functional enrichment analysis performed by bioinformatics indicated that the pathogenesis of DCM is mainly related to heart muscle contraction and calcium (Ca2+) release activation. In vitro, we discovered that high glucose treatment induced cardiomyocyte injury and exacerbated mitochondrial dysfunction remarkably. CONCLUSION Our research defined four crucial genes, as well as determined that mitochondrial function impairment compromises calcium homeostasis ultimately resulting in contractile dysfunction is a central contributor to DCM progression. Hopefully, this study will offer more effective biomarkers for DCM diagnosis and treatment.
Collapse
Affiliation(s)
- Maierhaba Tuersuntuoheti
- Department of Cardiology, Jinshan Hospital, Fudan University, Shanghai, China; Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Lei Zhou
- Department of Cardiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Juexing Li
- Department of Cardiology, Jinshan Hospital, Fudan University, Shanghai, China; Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shangneng Yang
- Department of Cardiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Suying Zhou
- Department of Cardiology, Jinshan Hospital, Fudan University, Shanghai, China; Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hui Gong
- Department of Cardiology, Jinshan Hospital, Fudan University, Shanghai, China; Department of Internal Medicine, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
10
|
Liao Y, Zhang W, Zhou M, Zhu C, Zou Z. Ubiquitination in pyroptosis pathway: A potential therapeutic target for sepsis. Cytokine Growth Factor Rev 2024:S1359-6101(24)00068-6. [PMID: 39294049 DOI: 10.1016/j.cytogfr.2024.09.001] [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: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Sepsis remains a significant clinical challenge, causing numerous deaths annually and representing a major global health burden. Pyroptosis, a unique form of programmed cell death characterized by cell lysis and the release of inflammatory mediators, is a crucial factor in the pathogenesis and progression of sepsis, septic shock, and organ dysfunction. Ubiquitination, a key post-translational modification influencing protein fate, has emerged as a promising target for managing various inflammatory conditions, including sepsis. This review integrates the current knowledge on sepsis, pyroptosis, and the ubiquitin system, focusing on the molecular mechanisms of ubiquitination within pyroptotic pathways activated during sepsis. By exploring how modulating ubiquitination can regulate pyroptosis and its associated inflammatory signaling pathways, this review provides insights into potential therapeutic strategies for sepsis, highlighting the need for further research into these complex molecular networks.
Collapse
Affiliation(s)
- Yan Liao
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Wangzheqi Zhang
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China
| | - Miao Zhou
- Department of Anesthesiology, the Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing Medical University, Nanjing, Jiangsu 210009, China
| | - Chenglong Zhu
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| | - Zui Zou
- School of Anesthesiology, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
11
|
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
|
12
|
Alfehaid LS, Farah S, Omer A, Weber BN, Alkhezi O, Tawfik YMK, Shah AM, Libby P, Buckley LF. Drug-Drug Interactions and the Clinical Tolerability of Colchicine Among Patients With COVID-19: A Secondary Analysis of the COLCORONA Randomized Clinical Trial. JAMA Netw Open 2024; 7:e2431309. [PMID: 39240567 PMCID: PMC11380098 DOI: 10.1001/jamanetworkopen.2024.31309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/01/2024] [Indexed: 09/07/2024] Open
Abstract
Importance Colchicine has many drug-drug interactions with commonly prescribed medications. Only pharmacokinetic studies have provided data on colchicine drug-drug interactions. Objective To evaluate the clinical tolerability of colchicine according to the presence or absence of a drug-drug interaction. Design, Setting, and Participants A secondary analysis of the COLCORONA trial was performed. The COLCORONA trial was a randomized, double-blind, placebo-controlled trial conducted in Brazil, Canada, Greece, South Africa, Spain, and the US between March 23, 2020, and January 20, 2021. The COLCORONA trial included ambulatory patients with COVID-19 with at least 1 high-risk characteristic and compared the effects of colchicine (0.5 mg twice daily for 3 days, then 0.5 mg daily thereafter) with placebo for 27 days. Data analysis was performed from February 24, 2023, to June 20, 2024. Exposure In this secondary analysis, baseline medications that had interactions with colchicine were identified using a previously published expert classification. Main Outcomes and Measures The primary outcome for this analysis was the composite of serious and nonserious treatment-related and treatment-unrelated gastrointestinal adverse events. The secondary outcomes were other adverse events and the composite of death or hospital admission due to COVID-19 infection. Logistic regression models adjusted for age, sex, estimated glomerular filtration rate, diabetes, heart failure, and myocardial infarction were assessed for effect modification of the association between the randomization arm and the outcomes of interest by drug-drug interaction status. Results The cohort included 2205 participants in the colchicine arm and 2227 in the placebo arm (median age, 54 [IQR, 47-61] years; 2389 [54%] women). The most common colchicine drug-drug interactions were rosuvastatin (12%) and atorvastatin (10%). In fully adjusted models, the odds of any gastrointestinal adverse event were 1.80 (95% CI, 1.51-2.15) times higher in the colchicine arm than the placebo arm among people without a drug-drug interaction and 1.68 (95% CI, 1.24-2.26) times higher in the colchicine arm than the placebo arm among people with a drug-drug interaction (P = .69 for interaction). Drug-drug interaction status did not significantly modify the effect of colchicine on the composite of COVID-19 hospitalization or death (odds ratio, 0.91; 95% CI, 0.59-1.40 for drug-drug interaction and 0.84; 95% CI, 0.60-1.19 for no drug-drug interaction; P = .80 for interaction). Conclusions and Relevance In this secondary analysis of the COLCORONA trial, operational classification of drug interactions system class 3 or 4 drug-drug interactions did not appear to significantly increase the risk of colchicine-related adverse effects. Trial Registration ClinicalTrials.gov Identifier: NCT04322682.
Collapse
Affiliation(s)
- Lama S. Alfehaid
- Department of Pharmacy, Brigham and Women’s Hospital, Boston, Massachusetts
- College of Pharmacy, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Subrina Farah
- Center for Clinical Investigation, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Azza Omer
- Department of Pharmacy, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Brittany N. Weber
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Omar Alkhezi
- Department of Pharmacy, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Pharmacy Practice, College of Pharmacy, Qassim University, Qassim, Saudi Arabia
| | - Yahya M. K. Tawfik
- Department of Pharmacy, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Amil M. Shah
- Division of Cardiology, The University of Texas Southwestern Medical Center, Dallas
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts
| | - Leo F. Buckley
- Department of Pharmacy, Brigham and Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
13
|
Hu D, Sheeja Prabhakaran H, Zhang YY, Luo G, He W, Liou YC. Mitochondrial dysfunction in sepsis: mechanisms and therapeutic perspectives. Crit Care 2024; 28:292. [PMID: 39227925 PMCID: PMC11373266 DOI: 10.1186/s13054-024-05069-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/17/2024] [Indexed: 09/05/2024] Open
Abstract
Sepsis is a severe medical condition characterized by a systemic inflammatory response, often culminating in multiple organ dysfunction and high mortality rates. In recent years, there has been a growing recognition of the pivotal role played by mitochondrial damage in driving the progression of sepsis. Various factors contribute to mitochondrial impairment during sepsis, encompassing mechanisms such as reactive nitrogen/oxygen species generation, mitophagy inhibition, mitochondrial dynamics change, and mitochondrial membrane permeabilization. Damaged mitochondria actively participate in shaping the inflammatory milieu by triggering key signaling pathways, including those mediated by Toll-like receptors, NOD-like receptors, and cyclic GMP-AMP synthase. Consequently, there has been a surge of interest in developing therapeutic strategies targeting mitochondria to mitigate septic pathogenesis. This review aims to delve into the intricate mechanisms underpinning mitochondrial dysfunction during sepsis and its significant impact on immune dysregulation. Moreover, we spotlight promising mitochondria-targeted interventions that have demonstrated therapeutic efficacy in preclinical sepsis models.
Collapse
Affiliation(s)
- Dongxue Hu
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Harshini Sheeja Prabhakaran
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Yuan-Yuan Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Key Laboratory for Disease Proteomics, Chongqing, 400038, China.
| | - Yih-Cherng Liou
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore.
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 119077, Singapore.
| |
Collapse
|
14
|
Yalcinkaya M, Tall AR. Genetic and epigenetic regulation of inflammasomes: Role in atherosclerosis. Atherosclerosis 2024; 396:118541. [PMID: 39111028 PMCID: PMC11374466 DOI: 10.1016/j.atherosclerosis.2024.118541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024]
Abstract
The cardiovascular complications of atherosclerosis are thought to arise from an inflammatory response to the accumulation of cholesterol-rich lipoproteins in the arterial wall. The positive outcome of CANTOS (Canakinumab Anti-inflammatory Thrombosis Outcome Study) provided key evidence to support this concept and suggested that inflammasomes and IL-1β are important inflammatory mediators in human atherosclerotic cardiovascular diseases (ACVD). In specific settings NLRP3 or AIM2 inflammasomes can induce inflammatory responses in the arterial wall and promote the formation of unstable atherosclerotic plaques. Clonal hematopoiesis (CH) has recently emerged as a major independent risk factor for ACVD. CH mutations arise during ageing and commonly involves variants in genes mediating epigenetic modifications (TET2, DNMT3A, ASXL1) or cytokine signaling (JAK2). Accumulating evidence points to the role of inflammasomes in the progression of CH-induced ACVD events and has shed light on the regulatory pathways and possible therapeutic approaches that specifically target inflammasomes in atherosclerosis. Epigenetic dynamics play a vital role in regulating the generation and activation of inflammasome components by causing changes in DNA methylation patterns and chromatin assembly. This review examines the genetic and epigenetic regulation of inflammasomes, the intersection of macrophage cholesterol accumulation with inflammasome activation and their roles in atherosclerosis. Understanding the involvement of inflammasomes in atherosclerosis pathogenesis may lead to customized treatments that reduce the burden of ACVD.
Collapse
Affiliation(s)
- Mustafa Yalcinkaya
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Alan R Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
15
|
Lee J, Sasaki F, Koike E, Cho M, Lee Y, Dho SH, Lee J, Lee E, Toyohara E, Sunakawa M, Ishibashi M, Hung HH, Nishioka S, Komine R, Okura C, Shimizu M, Ikawa M, Yoshimura A, Morita R, Kim LK. Gelsolin alleviates rheumatoid arthritis by negatively regulating NLRP3 inflammasome activation. Cell Death Differ 2024:10.1038/s41418-024-01367-6. [PMID: 39179640 DOI: 10.1038/s41418-024-01367-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 08/26/2024] Open
Abstract
Despite numerous biomarkers being proposed for rheumatoid arthritis (RA), a gap remains in our understanding of their mechanisms of action. In this study, we discovered a novel role for gelsolin (GSN), an actin-binding protein whose levels are notably reduced in the plasma of RA patients. We elucidated that GSN is a key regulator of NLRP3 inflammasome activation in macrophages, providing a plausible explanation for the decreased secretion of GSN in RA patients. We found that GSN interacts with NLRP3 in LPS-primed macrophages, hence modulating the formation of the NLRP3 inflammasome complex. Reducing GSN expression significantly enhanced NLRP3 inflammasome activation. GSN impeded NLRP3 translocation to the mitochondria; it contributed to the maintenance of intracellular calcium equilibrium and mitochondrial stability. This maintenance is crucial for controlling the inflammatory response associated with RA. Furthermore, the exacerbation of arthritic symptoms in GSN-deficient mice indicates the potential of GSN as both a diagnostic biomarker and a therapeutic target. Moreover, not limited to RA models, GSN has demonstrated a protective function in diverse disease models associated with the NLRP3 inflammasome. Myeloid cell-specific GSN-knockout mice exhibited aggravated inflammatory responses in models of MSU-induced peritonitis, folic acid-induced acute tubular necrosis, and LPS-induced sepsis. These findings suggest novel therapeutic approaches that modulate GSN activity, offering promise for more effective management of RA and a broader spectrum of inflammatory conditions.
Collapse
Affiliation(s)
- Jiyeon Lee
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Fumiyuki Sasaki
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Eri Koike
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Minjeong Cho
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yeongun Lee
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - So Hee Dho
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jina Lee
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eunji Lee
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eri Toyohara
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Mika Sunakawa
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Mariko Ishibashi
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Huynh Hiep Hung
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Saki Nishioka
- Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Ritsuko Komine
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Chiaki Okura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Masumi Shimizu
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan
| | - Masahito Ikawa
- Immunology Frontier Research Center, Osaka University, Suita, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Nippon Medical School, Tokyo, Japan.
| | - Lark Kyun Kim
- Department of Biomedical Sciences, Graduate School of Medical Science, Brain Korea 21 Project, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
16
|
Kim Y, Lee S, Park YH. NLRP3 Negative Regulation Mechanisms in the Resting State and Its Implications for Therapeutic Development. Int J Mol Sci 2024; 25:9018. [PMID: 39201704 PMCID: PMC11354250 DOI: 10.3390/ijms25169018] [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: 07/17/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 09/03/2024] Open
Abstract
The NACHT-, leucine-rich-repeat-, and pyrin domain-containing protein 3 (NLRP3) is a critical intracellular sensor of the innate immune system that detects various pathogen- and danger-associated molecular patterns, leading to the assembly of the NLRP3 inflammasome and release of interleukin (IL) 1β and IL-18. However, the abnormal activation of the NLRP3 inflammasome has been implicated in the pathogenesis of autoinflammatory diseases such as cryopyrin-associated autoinflammatory syndromes (CAPS) and common diseases such as Alzheimer's disease and asthma. Recent studies have revealed that pyrin functions as an indirect sensor, similar to the plant guard system, and is regulated by binding to inhibitory 14-3-3 proteins. Upon activation, pyrin transitions to its active form. NLRP3 is predicted to follow a similar regulatory mechanism and maintain its inactive form in the cage model, as it also acts as an indirect sensor. Additionally, newly developed NLRP3 inhibitors have been found to inhibit NLRP3 activity by stabilizing its inactive form. Most studies and reviews on NLRP3 have focused on the activation of the NLRP3 inflammasome. This review highlights the molecular mechanisms that regulate NLRP3 in its resting state, and discusses how targeting this inhibitory mechanism can lead to novel therapeutic strategies for NLRP3-related diseases.
Collapse
Affiliation(s)
- YeJi Kim
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.K.); (S.L.)
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Sumin Lee
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.K.); (S.L.)
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| | - Yong Hwan Park
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea; (Y.K.); (S.L.)
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon 16499, Republic of Korea
| |
Collapse
|
17
|
Zhou RN, Zhu ZW, Xu PY, Shen LX, Wang Z, Xue YY, Xiang YY, Cao Y, Yu XZ, Zhao J, Jin Y, Yan J, Yang Q, Fang PH, Shang WB. Rhein targets macrophage SIRT2 to promote adipose tissue thermogenesis in obesity in mice. Commun Biol 2024; 7:1003. [PMID: 39152196 PMCID: PMC11329635 DOI: 10.1038/s42003-024-06693-6] [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: 01/22/2024] [Accepted: 08/06/2024] [Indexed: 08/19/2024] Open
Abstract
Rhein, a component derived from rhubarb, has been proven to possess anti-inflammatory properties. Here, we show that rhein mitigates obesity by promoting adipose tissue thermogenesis in diet-induced obese mice. We construct a macrophage-adipocyte co-culture system and demonstrate that rhein promotes adipocyte thermogenesis through inhibiting NLRP3 inflammasome activation in macrophages. Moreover, clues from acetylome analysis identify SIRT2 as a potential drug target of rhein. We further verify that rhein directly interacts with SIRT2 and inhibits NLRP3 inflammasome activation in a SIRT2-dependent way. Myeloid knockdown of SIRT2 abrogates adipose tissue thermogenesis and metabolic benefits in obese mice induced by rhein. Together, our findings elucidate that rhein inhibits NLRP3 inflammasome activation in macrophages by regulating SIRT2, and thus promotes white adipose tissue thermogenesis during obesity. These findings uncover the molecular mechanism underlying the anti-inflammatory and anti-obesity effects of rhein, and suggest that rhein may become a potential drug for treating obesity.
Collapse
Affiliation(s)
- Ruo-Nan Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zi-Wei Zhu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ping-Yuan Xu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Li-Xuan Shen
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ziwei Wang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ying-Ying Xue
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ying-Ying Xiang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yue Cao
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xi-Zhong Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Juan Zhao
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yu Jin
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jing Yan
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qin Yang
- Department of Medicine, Physiology and Biophysics, University of California, Irvine, CA, 92697, USA
| | - Peng-Hua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Wen-Bin Shang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
- Key Laboratory for Metabolic Diseases in Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
18
|
Henedak NT, El-Abhar HS, Soubh AA, Abdallah DM. NLRP3 Inflammasome: A central player in renal pathologies and nephropathy. Life Sci 2024; 351:122813. [PMID: 38857655 DOI: 10.1016/j.lfs.2024.122813] [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: 02/12/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
The cytoplasmic oligomer NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome has been implicated in most inflammatory and autoimmune diseases. Here, we highlight the significance of NLRP3 in diverse renal disorders, demonstrating its activation in macrophages and non-immune tubular epithelial and mesangial cells in response to various stimuli. This activation leads to the release of pro-inflammatory cytokines, contributing to the development of acute kidney injury (AKI), chronic renal injury, or fibrosis. In AKI, NLRP3 inflammasome activation and pyroptotic renal tubular cell death is driven by contrast and chemotherapeutic agents, sepsis, and rhabdomyolysis. Nevertheless, inflammasome is provoked in disorders such as crystal and diabetic nephropathy, obesity-related renal fibrosis, lupus nephritis, and hypertension-induced renal damage that induce chronic kidney injury and/or fibrosis. The mechanisms by which the inflammatory NLRP3/ Apoptosis-associated Speck-like protein containing a Caspase recruitment domain (ASC)/caspase-1/interleukin (IL)-1β & IL-18 pathway can turn on renal fibrosis is also comprehended. This review further outlines the involvement of dopamine and its associated G protein-coupled receptors (GPCRs), including D1-like (D1, D5) and D2-like (D2-D4) subtypes, in regulating this inflammation-linked renal dysfunction pathway. Hence, we identify D-related receptors as promising targets for renal disease management by inhibiting the functionality of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Nada T Henedak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Giza, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo 11835, Egypt
| | - Ayman A Soubh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Giza, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| |
Collapse
|
19
|
Hao J, Guo X, Wang S, Guo X, Yuan K, Chen R, Hao L. LincRNA-p21/AIF-1/CMPK2/NLRP3 pathway promoted inflammation, autophagy and apoptosis of human tubular epithelial cell induced by urate via exosomes. Sci Rep 2024; 14:18146. [PMID: 39103417 PMCID: PMC11300820 DOI: 10.1038/s41598-024-69323-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024] Open
Abstract
Urate nephropathy, a common complication of hyperuricemia, has garnered increasing attention worldwide. However, the exact pathogenesis of this condition remains unclear. Currently, inflammation is widely accepted as the key factor in urate nephropathy. Therefore, the aim of this study was to elucidate the interaction of lincRNA-p21/AIF-1/CMPK2/NLRP3 via exosomes in urate nephropathy. This study evaluated the effect of lincRNA-p21/AIF-1/CMPK2/NLRP3 using clinical data collected from patients with urate nephropathy and human renal tubular epithelial cells (HK2) cultured with different concentrations of urate. In clinical research section, the level of lincRNA-p21/AIF-1 in exosomes of urine in patients with hyperuricemia or urate nephropathy was found to be increased, particularly in patients with urate nephropathy. In vitro study section, the level of exosomes, inflammation, autophagy, and apoptosis was increased in HK2 cells induced by urate. Additionally, the expression of lincRNA-p21, AIF-1, CMPK2, and NLRP3 was upregulated in exosomes and HK2 cells. Furthermore, manipulating the activity of lincRNA-p21, AIF-1, CMPK2, and NLRP3 through overexpression or interference vectors regulated the level of inflammation, autophagy, and apoptosis in HK2 cells. In conclusion, the pathway of lincRNA-p21/AIF-1/CMPK2/NLRP3 contributed to inflammation, autophagy, and apoptosis of human renal tubular epithelial cell induced by urate via exosomes. Additionally, the specific exosomes in urine might serve as novel biomarkers for urate nephropathy.
Collapse
Affiliation(s)
- Jianbing Hao
- Department of Nephrology, Southern University of Science and Technology Hospital, Shenzhen, China.
| | - Xinyu Guo
- Department of Nephrology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Siyu Wang
- Department of Nephrology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Xiaojun Guo
- Department of Nephrology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Kun Yuan
- Department of Nephrology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Ruihong Chen
- Department of Nephrology, Southern University of Science and Technology Hospital, Shenzhen, China
| | - Lirong Hao
- Department of Nephrology, Southern University of Science and Technology Hospital, Shenzhen, China.
| |
Collapse
|
20
|
Li J, Yao Y, Lei X, Bao J, An S, Hu H, Sha T, Huang Q, Li T, Zeng Z, Wang X, Cai S. SIRTUIN 5 ALLEVIATES EXCESSIVE MITOCHONDRIAL FISSION VIA DESUCCINYLATION OF ATPASE INHIBITORY FACTOR 1 IN SEPSIS-INDUCED ACUTE KIDNEY INJURY. Shock 2024; 62:235-244. [PMID: 38754030 DOI: 10.1097/shk.0000000000002392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
ABSTRACT Sepsis-induced acute kidney injury (SAKI) poses a significant clinical challenge with high morbidity and mortality. Excessive mitochondrial fission has been identified as the central pathogenesis of sepsis-associated organ damage, which is also implicated in the early stages of SAKI. Sirtuin 5 (SIRT5) has emerged as a central regulator of cellular mitochondrial function; however, its role in the regulation of sepsis-induced excessive mitochondrial fission in kidney and the underlying mechanism remains unclear. In this study, SAKI was modeled in mice through cecal ligation and puncture, and in human renal tubular epithelial (HK-2) cells stimulated with lipopolysaccharide (LPS), to mimic the cell SAKI model. Our findings revealed that septic mice with a SIRT5 knockout exhibited shortened survival times and elevated levels of renal injury compared to wild-type mice, suggesting the significant involvement of SIRT5 in SAKI pathophysiology. Additionally, we observed that SIRT5 depletion led to increased renal mitochondrial fission, while the use of a mitochondrial fission inhibitor (Mdivi-1) reversed the detrimental effects caused by SIRT5 depletion, emphasizing the pivotal role of SIRT5 in preventing excessive mitochondrial fission. In vitro experiments demonstrated that the overexpression of SIRT5 effectively mitigated the adverse effects of LPS on HK-2 cells viability and mitochondrial fission. Conversely, downregulation of SIRT5 decreased HK-2 cells viability and exacerbated LPS-induced mitochondrial fission. Mechanistically, the protective function of SIRT5 may be in part, ascribed to its desuccinylating action on ATPase inhibitory factor 1. In conclusion, this study provides novel insights into the underlying mechanisms of SAKI, suggesting the possibility of identifying future drug targets in terms of improved mitochondrial dynamics by SIRT5.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qiaobing Huang
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Tao Li
- Department of Critical Care Medicine, The First People's Hospital of Chenzhou, Southern Medical University, Chenzhou, China
| | - Zhenhua Zeng
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiang Wang
- Department of Critical Care Medicine, The First People's Hospital of Chenzhou, Southern Medical University, Chenzhou, China
| | - Shumin Cai
- Department of Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
21
|
Kelly P, Lemmens R, Weimar C, Walsh C, Purroy F, Barber M, Collins R, Cronin S, Czlonkowska A, Desfontaines P, De Pauw A, Evans NR, Fischer U, Fonseca C, Forbes J, Hill MD, Jatuzis D, Kõrv J, Kraft P, Kruuse C, Lynch C, McCabe D, Mikulik R, Murphy S, Nederkoorn P, O'Donnell M, Sandercock P, Schroeder B, Shim G, Tobin K, Williams DJ, Price C. Long-term colchicine for the prevention of vascular recurrent events in non-cardioembolic stroke (CONVINCE): a randomised controlled trial. Lancet 2024; 404:125-133. [PMID: 38857611 DOI: 10.1016/s0140-6736(24)00968-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 04/30/2024] [Accepted: 05/07/2024] [Indexed: 06/12/2024]
Abstract
BACKGROUND Anti-inflammatory therapy with long-term colchicine prevented vascular recurrence in coronary disease. Unlike coronary disease, which is typically caused by atherosclerosis, ischaemic stroke is caused by diverse mechanisms including atherosclerosis and small vessel disease or is frequently due to an unknown cause. We aimed to investigate the hypothesis that long-term colchicine would reduce recurrent events after ischaemic stroke. METHODS We did a randomised, parallel-group, open-label, blinded endpoint assessed trial comparing long-term colchicine (0·5 mg orally per day) plus guideline-based usual care with usual care only. Hospital-based patients with non-severe, non-cardioembolic ischaemic stroke or high-risk transient ischaemic attack were eligible. The primary endpoint was a composite of first fatal or non-fatal recurrent ischaemic stroke, myocardial infarction, cardiac arrest, or hospitalisation (defined as an admission to an inpatient unit or a visit to an emergency department that resulted in at least a 24 h stay [or a change in calendar date if the hospital admission or discharge times were not available]) for unstable angina. The p value for significance was 0·048 to adjust for two prespecified interim analyses conducted by the data monitoring committee, for which the steering committee and trial investigators remained blinded. The trial was registered at ClinicalTrials.gov (NCT02898610) and is completed. FINDINGS 3154 patients were randomly assigned between Dec 19, 2016, and Nov 21, 2022, with the last follow-up on Jan 31, 2024. The trial finished before the anticipated number of outcomes was accrued (367 outcomes planned) due to budget constraints attributable to the COVID-19 pandemic. Ten patients withdrew consent for analysis of their data, leaving 3144 patients in the intention-to-treat analysis: 1569 (colchicine and usual care) and 1575 (usual care alone). A primary endpoint occurred in 338 patients, 153 (9·8%) of 1569 patients allocated to colchicine and usual care and 185 (11·7%) of 1575 patients allocated to usual care alone (incidence rates 3·32 vs 3·92 per 100 person-years, hazard ratio 0·84; 95% CI 0·68-1·05, p=0·12). Although no between-group difference in C-reactive protein (CRP) was observed at baseline, patients treated with colchicine had lower CRP at 28 days and at 1, 2, and 3 years (p<0·05 for all timepoints). The rates of serious adverse events were similar in both groups. INTERPRETATION Although no statistically significant benefit was observed on the primary intention-to-treat analysis, the findings provide new evidence supporting the rationale for anti-inflammatory therapy in further randomised trials. FUNDING Health Research Board Ireland, Deutsche Forschungsgemeinschaft (German Research Foundation), and Fonds Wetenschappelijk Onderzoek Vlaanderen (Research Foundation Flanders), Belgium.
Collapse
Affiliation(s)
- Peter Kelly
- Mater Misericordiae University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland; Health Research Board Stroke Clinical Trials Network Ireland, Dublin, Ireland.
| | - Robin Lemmens
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium; Department of Neurosciences, Department of Experimental Neurology, and Leuven Research Institute for Neuroscience and Disease (LIND), KU Leuven-University of Leuven, Leuven, Belgium
| | - Christian Weimar
- Institute for Medical Informatics, Biometry, and Epidemiology, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Cathal Walsh
- TCD Biostatistics Unit, Discipline of Public Health and Primary Care, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Francisco Purroy
- Stroke Unit, Department of Neurology, Hospitalt Universitari Arnau de Vilanova de Lleida, Lleida, Spain; Biomedical Research Institute of Lleida, Universitat de Lleida, Lleida, Spain
| | | | - Ronan Collins
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin, Ireland; Department of Neurology and Department of Geriatric and Stroke Medicine, Tallaght University Hospital-The Adelaide and Meath Hospital, Dublin, Ireland incorporating the National Children's Hospital and Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Simon Cronin
- Cork University Hospital, Cork, Ireland; School of Medicine, University College Cork, Cork, Ireland
| | | | | | | | | | - Urs Fischer
- Department of Neurology, University Hospital Bern and University of Bern, Bern, Switzerland
| | - Catarina Fonseca
- Department of Neurosciences and Mental Health (Neurology), Hospital Santa Maria-CHLN, Centro de Estudos Egas Moniz, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - John Forbes
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin, Ireland; School of Medicine, University of Limerick, Limerick, Ireland
| | - Michael D Hill
- Department of Clinical Neurosciences & Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary and Foothills Medical Centre, Calgary, AB, Canada
| | - Dalius Jatuzis
- Centre of Neurology, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Janika Kõrv
- Department of Neurology and Neurosurgery, University of Tartu, Tartu, Estonia
| | | | - Christina Kruuse
- Department of Neurology, Herlev and Gentofte Hospital, and Department of Brain and Spinal Cord Injury, Rigshospitalet Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Catherine Lynch
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin, Ireland
| | - Dominick McCabe
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin, Ireland; Department of Neurology and Department of Geriatric and Stroke Medicine, Tallaght University Hospital-The Adelaide and Meath Hospital, Dublin, Ireland incorporating the National Children's Hospital and Academic Unit of Neurology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Robert Mikulik
- International Clinical Research Center and Department of Neurology, St Anne's University Hospital and Masaryk University Brno, Brno, Czech Republic
| | - Sean Murphy
- Mater Misericordiae University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland; Health Research Board Stroke Clinical Trials Network Ireland, Dublin, Ireland; RCSI University of Medicine and Health Sciences and Beaumont Hospital, Dublin, Ireland
| | - Paul Nederkoorn
- Department of Neurology, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Martin O'Donnell
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin, Ireland; HRB Clinical Research Facility, University of Galway, Galway, Ireland
| | - Peter Sandercock
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | | | - Gek Shim
- University Hospital of North Durham, Durham, UK
| | - Katrina Tobin
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin, Ireland
| | - David J Williams
- Health Research Board Stroke Clinical Trials Network Ireland, Dublin, Ireland; RCSI University of Medicine and Health Sciences and Beaumont Hospital, Dublin, Ireland
| | - Christopher Price
- Population Health Sciences Institute, Newcastle University, Newcastle, UK
| |
Collapse
|
22
|
Kang H, Choi SW, Kim JY, Oh SJ, Kim SJ, Lee MS. ER-to-lysosome Ca 2+ refilling followed by K + efflux-coupled store-operated Ca 2+ entry in inflammasome activation and metabolic inflammation. eLife 2024; 12:RP87561. [PMID: 38953285 PMCID: PMC11219040 DOI: 10.7554/elife.87561] [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] [Indexed: 07/03/2024] Open
Abstract
We studied lysosomal Ca2+ in inflammasome. Lipopolysaccharide (LPS) + palmitic acid (PA) decreased lysosomal Ca2+ ([Ca2+]Lys) and increased [Ca2+]i through mitochondrial ROS, which was suppressed in Trpm2-KO macrophages. Inflammasome activation and metabolic inflammation in adipose tissue of high-fat diet (HFD)-fed mice were ameliorated by Trpm2 KO. ER→lysosome Ca2+ refilling occurred after lysosomal Ca2+ release whose blockade attenuated LPS + PA-induced inflammasome. Subsequently, store-operated Ca2+entry (SOCE) was activated whose inhibition suppressed inflammasome. SOCE was coupled with K+ efflux whose inhibition reduced ER Ca2+ content ([Ca2+]ER) and impaired [Ca2+]Lys recovery. LPS + PA activated KCa3.1 channel, a Ca2+-activated K+ channel. Inhibitors of KCa3.1 channel or Kcnn4 KO reduced [Ca2+]ER, attenuated increase of [Ca2+]i or inflammasome activation by LPS + PA, and ameliorated HFD-induced inflammasome or metabolic inflammation. Lysosomal Ca2+ release induced delayed JNK and ASC phosphorylation through CAMKII-ASK1. These results suggest a novel role of lysosomal Ca2+ release sustained by ER→lysosome Ca2+ refilling and K+ efflux through KCa3.1 channel in inflammasome activation and metabolic inflammation.
Collapse
Affiliation(s)
- Hyereen Kang
- Severance Biomedical Science Institute, Yonsei University College of MedicineSeoulRepublic of Korea
| | - Seong Woo Choi
- Department of Physiology and Ion Channel Disease Research Center, Dongguk University College of MedicineGyeongjuRepublic of Korea
| | - Joo Young Kim
- Department of Pharmacology and Brain Korea 21 Project for Medical Sciences, Yonsei University College of MedicineSeoulRepublic of Korea
| | - Soo-Jin Oh
- Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang University College of MedicineCheonanRepublic of Korea
| | - Sung Joon Kim
- Department of Physiology, Ischemic/Hypoxic Disease Institute, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Myung-Shik Lee
- Severance Biomedical Science Institute, Yonsei University College of MedicineSeoulRepublic of Korea
- Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang University College of MedicineCheonanRepublic of Korea
| |
Collapse
|
23
|
Fu J, Schroder K, Wu H. Mechanistic insights from inflammasome structures. Nat Rev Immunol 2024; 24:518-535. [PMID: 38374299 PMCID: PMC11216901 DOI: 10.1038/s41577-024-00995-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 02/21/2024]
Abstract
Inflammasomes are supramolecular complexes that form in the cytosol in response to pathogen-associated and damage-associated stimuli, as well as other danger signals that perturb cellular homoeostasis, resulting in host defence responses in the form of cytokine release and programmed cell death (pyroptosis). Inflammasome activity is closely associated with numerous human disorders, including rare genetic syndromes of autoinflammation, cardiovascular diseases, neurodegeneration and cancer. In recent years, a range of inflammasome components and their functions have been discovered, contributing to our knowledge of the overall machinery. Here, we review the latest advances in inflammasome biology from the perspective of structural and mechanistic studies. We focus on the most well-studied components of the canonical inflammasome - NAIP-NLRC4, NLRP3, NLRP1, CARD8 and caspase-1 - as well as caspase-4, caspase-5 and caspase-11 of the noncanonical inflammasome, and the inflammasome effectors GSDMD and NINJ1. These structural studies reveal important insights into how inflammasomes are assembled and regulated, and how they elicit the release of IL-1 family cytokines and induce membrane rupture in pyroptosis.
Collapse
Affiliation(s)
- Jianing Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kate Schroder
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
24
|
McManus RM, Latz E. NLRP3 inflammasome signalling in Alzheimer's disease. Neuropharmacology 2024; 252:109941. [PMID: 38565393 DOI: 10.1016/j.neuropharm.2024.109941] [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: 01/31/2024] [Revised: 03/20/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Every year, 10 million people develop dementia, the most common of which is Alzheimer's disease (AD). To date, there is no way to prevent cognitive decline and therapies are limited. This review provides a neuroimmunological perspective on the progression of AD, and discusses the immune-targeted therapies that are in preclinical and clinical trials that may impact the development of this disease. Specifically, we look to the role of the NLRP3 inflammasome, its triggers in the brain and how its activation can contribute to the progression of dementia. We summarise the range of inhibitors targeting the NLRP3 inflammasome and its downstream pathways that are under investigation, and discuss future therapeutic perspectives for this devastating condition.
Collapse
Affiliation(s)
- Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127, Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany.
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491, Trondheim, Norway; Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, 01605, USA; Deutsches Rheuma-Forschungszentrum (DRFZ), Charitéplatz 1, 10117, Berlin, Germany
| |
Collapse
|
25
|
Bulnes JF, González L, Velásquez L, Orellana MP, Venturelli PM, Martínez G. Role of inflammation and evidence for the use of colchicine in patients with acute coronary syndrome. Front Cardiovasc Med 2024; 11:1356023. [PMID: 38993522 PMCID: PMC11236697 DOI: 10.3389/fcvm.2024.1356023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/29/2024] [Indexed: 07/13/2024] Open
Abstract
Acute Coronary Syndrome (ACS) significantly contributes to cardiovascular death worldwide. ACS may arise from the disruption of an atherosclerotic plaque, ultimately leading to acute ischemia and myocardial infarction. In the pathogenesis of atherosclerosis, inflammation assumes a pivotal role, not solely in the initiation and complications of atherosclerotic plaque formation, but also in the myocardial response to ischemic insult. Acute inflammatory processes, coupled with time to reperfusion, orchestrate ischemic and reperfusion injuries, dictating infarct magnitude and acute left ventricular (LV) remodeling. Conversely, chronic inflammation, alongside neurohumoral activation, governs persistent LV remodeling. The interplay between chronic LV remodeling and recurrent ischemic episodes delineates the progression of the disease toward heart failure and cardiovascular death. Colchicine exerts anti-inflammatory properties affecting both the myocardium and atherosclerotic plaque by modulating the activity of monocyte/macrophages, neutrophils, and platelets. This modulation can potentially result in a more favorable LV remodeling and forestalls the recurrence of ACS. This narrative review aims to delineate the role of inflammation across the different phases of ACS pathophysiology and describe the mechanistic underpinnings of colchicine, exploring its purported role in modulating each of these stages.
Collapse
Affiliation(s)
- Juan Francisco Bulnes
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leticia González
- Centro de Imágenes Biomédicas, Departamento de Radiología, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leonardo Velásquez
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María Paz Orellana
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paula Muñoz Venturelli
- Centro de Estudios Clínicos, Instituto de Ciencias e Innovación en Medicina (ICIM), Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Gonzalo Martínez
- División de Enfermedades Cardiovasculares, Pontificia Universidad Católica de Chile, Santiago, Chile
- Heart Research Institute, Sydney, NSW, Australia
| |
Collapse
|
26
|
Guan J, Abudouaini H, Lin K, Yang K. Emerging insights into the role of IL-1 inhibitors and colchicine for inflammation control in type 2 diabetes. Diabetol Metab Syndr 2024; 16:140. [PMID: 38918878 PMCID: PMC11197348 DOI: 10.1186/s13098-024-01369-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM), a prevalent chronic metabolic disorder, is closely linked to persistent low-grade inflammation, significantly contributing to its development and progression. This review provides a comprehensive examination of the inflammatory mechanisms underlying T2DM, focusing on the role of the NLRP3 inflammasome and interleukin-1β (IL-1β) in mediating inflammatory responses. We discuss the therapeutic potential of IL-1 inhibitors and colchicine, highlighting their mechanisms in inhibiting the NLRP3 inflammasome and reducing IL-1β production. Recent studies indicate that these agents could effectively mitigate inflammation, offering promising avenues for the prevention and management of T2DM. By exploring the intricate connections between metabolic disturbances and chronic inflammation, this review underscores the need for novel anti-inflammatory strategies to address T2DM and its complications.
Collapse
Affiliation(s)
- Jianbin Guan
- Honghui-Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Haimiti Abudouaini
- Honghui-Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Kaiyuan Lin
- Honghui-Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| | - Kaitan Yang
- Honghui-Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
- Truma Rehabilitation Department, Honghui-Hospital,Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
27
|
Sugisawa R, Shanahan KA, Davis GM, Davey GP, Bowie AG. SARM1 regulates pro-inflammatory cytokine expression in human monocytes by NADase-dependent and -independent mechanisms. iScience 2024; 27:109940. [PMID: 38832024 PMCID: PMC11145347 DOI: 10.1016/j.isci.2024.109940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 03/14/2024] [Accepted: 05/06/2024] [Indexed: 06/05/2024] Open
Abstract
SARM1 is a Toll-IL-1 receptor (TIR) domain-containing protein with roles in innate immunity and neuronal death in diverse organisms. Unlike other innate immune TIR proteins that function as adaptors for Toll-like receptors (TLRs), SARM1 has NADase activity, and this activity regulates murine neuronal cell death. However, whether human SARM1, and its NADase activity, are involved in innate immune regulation remains unclear. Here, we show that human SARM1 regulates proinflammatory cytokine expression in both an NADase-dependent and -independent manner in monocytes. SARM1 negatively regulated TLR4-dependent TNF mRNA induction independently of its NADase activity. In contrast, SARM1 inhibited IL-1β secretion through both NADase-dependent inhibition of pro-IL-1β expression, and NADase-independent suppression of the NLRP3 inflammasome and hence processing of pro-IL-1β to mature IL-1β. Our study reveals multiple mechanisms whereby SARM1 regulates pro-inflammatory cytokines in human monocytes and shows, compared to other mammalian TIR proteins, a distinct NADase-dependent role for SARM1 in innate immunity.
Collapse
Affiliation(s)
- Ryoichi Sugisawa
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Department of Biochemistry, Kindai University Faculty of Medicine, Osaka, Japan
| | - Katharine A. Shanahan
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Gavin M. Davis
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Gavin P. Davey
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew G. Bowie
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
28
|
Valenti D, Atlante A. Sound Matrix Shaping of Living Matter: From Macrosystems to Cell Microenvironment, Where Mitochondria Act as Energy Portals in Detecting and Processing Sound Vibrations. Int J Mol Sci 2024; 25:6841. [PMID: 38999952 PMCID: PMC11241420 DOI: 10.3390/ijms25136841] [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: 05/06/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Vibration and sound are the shaping matrix of the entire universe. Everything in nature is shaped by energy vibrating and communicating through its own sound trail. Every cell within our body vibrates at defined frequencies, generating its peculiar "sound signature". Mitochondria are dynamic, energy-transforming, biosynthetic, and signaling organelles that actively transduce biological information. Novel research has shown that the mitochondrial function of mammalian cells can be modulated by various energetic stimuli, including sound vibrations. Regarding acoustic vibrations, definite types of music have been reported to produce beneficial impacts on human health. In very recent studies, the effects of different sound stimuli and musical styles on cellular function and mitochondrial activity were evaluated and compared in human cells cultured in vitro, investigating the underlying responsible molecular mechanisms. This narrative review will take a multilevel trip from macro to intracellular microenvironment, discussing the intimate vibrational sound activities shaping living matter, delving deeper into the molecular mechanisms underlying the sound modulation of biological systems, and mainly focusing our discussion on novel evidence showing the competence of mitochondria in acting as energy portals capable of sensing and transducing the subtle informational biofields of sound vibration.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via G. Amendola 122/O, 70126 Bari, Italy
| | - Anna Atlante
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via G. Amendola 122/O, 70126 Bari, Italy
| |
Collapse
|
29
|
Zhao X, Li Y, Yu J, Teng H, Wu S, Wang Y, Zhou H, Li F. Role of mitochondria in pathogenesis and therapy of renal fibrosis. Metabolism 2024; 155:155913. [PMID: 38609039 DOI: 10.1016/j.metabol.2024.155913] [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: 12/06/2023] [Revised: 03/18/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024]
Abstract
Renal fibrosis, specifically tubulointerstitial fibrosis, represents the predominant pathological consequence observed in the context of progressive chronic kidney conditions. The pathogenesis of renal fibrosis encompasses a multifaceted interplay of mechanisms, including but not limited to interstitial fibroblast proliferation, activation, augmented production of extracellular matrix (ECM) components, and impaired ECM degradation. Notably, mitochondria, the intracellular organelles responsible for orchestrating biological oxidation processes in mammalian cells, assume a pivotal role within this intricate milieu. Mitochondrial dysfunction, when manifest, can incite a cascade of events, including inflammatory responses, perturbed mitochondrial autophagy, and associated processes, ultimately culminating in the genesis of renal fibrosis. This comprehensive review endeavors to furnish an exegesis of mitochondrial pathophysiology and biogenesis, elucidating the precise mechanisms through which mitochondrial aberrations contribute to the onset and progression of renal fibrosis. We explored how mitochondrial dysfunction, mitochondrial cytopathy and mitochondrial autophagy mediate ECM deposition and renal fibrosis from a multicellular perspective of mesangial cells, endothelial cells, podocytes, macrophages and fibroblasts. Furthermore, it succinctly encapsulates the most recent advancements in the realm of mitochondrial-targeted therapeutic strategies aimed at mitigating renal fibrosis.
Collapse
Affiliation(s)
- Xiaodong Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yunkuo Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Jinyu Yu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Haolin Teng
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Shouwang Wu
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun 130021, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
30
|
Zeng ZJ, Lin X, Yang L, Li Y, Gao W. Activation of Inflammasomes and Relevant Modulators for the Treatment of Microglia-mediated Neuroinflammation in Ischemic Stroke. Mol Neurobiol 2024:10.1007/s12035-024-04225-1. [PMID: 38789893 DOI: 10.1007/s12035-024-04225-1] [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: 01/29/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024]
Abstract
As the brain's resident immune patrol, microglia mediate endogenous immune responses to central nervous system injury in ischemic stroke, thereby eliciting either neuroprotective or neurotoxic effects. The association of microglia-mediated neuroinflammation with the progression of ischemic stroke is evident through diverse signaling pathways, notably involving inflammasomes. Within microglia, inflammasomes play a pivotal role in promoting the maturation of interleukin-1β (IL-1β) and interleukin-18 (IL-18), facilitating pyroptosis, and triggering immune infiltration, ultimately leading to neuronal cell dysfunction. Addressing the persistent and widespread inflammation holds promise as a breakthrough in enhancing the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ze-Jie Zeng
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaobing Lin
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Liu Yang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yi Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| | - Wen Gao
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
31
|
Thirugnanam S, Rout N. A Perfect Storm: The Convergence of Aging, Human Immunodeficiency Virus Infection, and Inflammasome Dysregulation. Curr Issues Mol Biol 2024; 46:4768-4786. [PMID: 38785555 PMCID: PMC11119826 DOI: 10.3390/cimb46050287] [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: 04/23/2024] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
The emergence of combination antiretroviral therapy (cART) has greatly transformed the life expectancy of people living with HIV (PWH). Today, over 76% of the individuals with HIV have access to this life-saving therapy. However, this progress has come with a new challenge: an increase in age-related non-AIDS conditions among patients with HIV. These conditions manifest earlier in PWH than in uninfected individuals, accelerating the aging process. Like PWH, the uninfected aging population experiences immunosenescence marked by an increased proinflammatory environment. This phenomenon is linked to chronic inflammation, driven in part by cellular structures called inflammasomes. Inflammatory signaling pathways activated by HIV-1 infection play a key role in inflammasome formation, suggesting a crucial link between HIV and a chronic inflammatory state. This review outlines the inflammatory processes triggered by HIV-1 infection and aging, with a focus on the inflammasomes. This review also explores current research regarding inflammasomes and potential strategies for targeting inflammasomes to mitigate inflammation. Further research on inflammasome signaling presents a unique opportunity to develop targeted interventions and innovative therapeutic modalities for combating HIV and aging-associated inflammatory processes.
Collapse
Affiliation(s)
- Siva Thirugnanam
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Namita Rout
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA 70433, USA;
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Tulane Center for Aging, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
32
|
Yang S, Liu H, Fang XM, Yan F, Zhang Y. Signaling pathways in uric acid homeostasis and gout: From pathogenesis to therapeutic interventions. Int Immunopharmacol 2024; 132:111932. [PMID: 38560961 DOI: 10.1016/j.intimp.2024.111932] [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: 02/17/2024] [Revised: 03/20/2024] [Accepted: 03/24/2024] [Indexed: 04/04/2024]
Abstract
Uric acid is a product of purine degradation, and uric acid may have multiple physiologic roles, including the beneficial effects as an antioxidant and neuroprotector, maintenance of blood pressure during low salt ingestion, and modulation of immunity. However, overproduction of metabolic uric acid, and/or imbalance of renal uric acid secretion and reabsorption, and/or underexcretion of extrarenal uric acid, e.g. gut, will contribute to hyperuricemia, which is a common metabolic disease. Long-lasting hyperuricemia can induce the formation and deposition of monosodium urate (MSU) crystals within the joints and periarticular structures. MSU crystals further induce an acute, intensely painful, and sterile inflammation conditions named as gout by NLRP3 inflammasome-mediated cleavage of pro-IL-1β to bioactive IL-1β. Moreover, hyperuricemia and gout are associated with multiple cardiovascular and renal disorders, e.g., hypertension, myocardial infarction, stroke, obesity, hyperlipidemia, type 2 diabetes mellitus and chronic kidney disease. Although great efforts have been made by scientists of modern medicine, however, modern therapeutic strategies with a single target are difficult to exert long-term positive effects, and even some of these agents have severe adverse effects. The Chinese have used the ancient classic prescriptions of traditional Chinese medicine (TCM) to treat metabolic diseases, including gout, by multiple targets, for more than 2200 years. In this review, we discuss the current understanding of urate homeostasis, the pathogenesis of hyperuricemia and gout, and both modern medicine and TCM strategies for this commonly metabolic disorder. We hope these will provide the good references for treating hyperuricemia and gout.
Collapse
Affiliation(s)
- Shuangling Yang
- School of Health Sciences, Guangzhou Xinhua University, Guangzhou, Guangdong 510520, China
| | - Haimei Liu
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China
| | - Xian-Ming Fang
- Department of Cardiology, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi 530011, China.
| | - Fuman Yan
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China.
| | - Yaxing Zhang
- Department of Physiology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Research Centre of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, China; Issue 12(th) of Guangxi Apprenticeship Education of Traditional Chinese Medicine (Shi‑Cheng Class of Guangxi University of Chinese Medicine), College of Continuing Education, Guangxi University of Chinese Medicine, Nanning, Guangxi 530001, China.
| |
Collapse
|
33
|
Vecchié A, Bonaventura A, Golino M, Thomas G, Abbate A. Novel Therapeutic Insights Into the Treatment of Pericarditis: Targeting the Innate Immune System. J Cardiovasc Pharmacol 2024; 83:377-383. [PMID: 38422218 DOI: 10.1097/fjc.0000000000001553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 02/12/2024] [Indexed: 03/02/2024]
Abstract
ABSTRACT Acute pericarditis is characterized by pericardial inflammation that can be treated with anti-inflammatory drugs. A considerable percentage of patients develops recurrent pericarditis with several relapses. In developed countries, the idiopathic form is the most frequent and has a high risk of recurrences. Two pathophysiological mechanisms have been described for idiopathic recurrent pericarditis: autoimmune and autoinflammatory. The autoimmune mechanism is more frequently encountered in patients with rheumatologic disorders, especially systemic lupus erythematosus. The innate immune system plays a central role in the pathophysiology of pericarditis, especially in the autoinflammatory phenotype. Current evidence highlights the central role played by interleukin 1 and NLRP3 (NACHT, leucine-rich repeat, and pyrin domain-containing protein 3) in idiopathic recurrent pericarditis. Accordingly, interleukin 1 blockers have been approved for the treatment of this condition. Neutrophils are likely to be important in such setting; however, their role has only been partially investigated. In the present review, we have collected the current knowledge on the role of innate immune system in pericarditis pathophysiology and how this can be used to provide targeted treatments for patients with recurrent pericarditis.
Collapse
Affiliation(s)
| | - Aldo Bonaventura
- Department of Internal Medicine, ASST Sette Laghi, Varese, Italy
| | - Michele Golino
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
- Department of Medicine and Surgery, University of Insubria, Varese, Italy; and
| | - Georgia Thomas
- VCU Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, Richmond, VA
| | - Antonio Abbate
- Robert M. Berne Cardiovascular Research Center and Division of Cardiology, Department of Internal Medicine, Heart and Vascular Center, University of Virginia, Charlottesville, VA
| |
Collapse
|
34
|
Buckley LF, Libby P. Colchicine's Role in Cardiovascular Disease Management. Arterioscler Thromb Vasc Biol 2024; 44:1031-1041. [PMID: 38511324 PMCID: PMC11047118 DOI: 10.1161/atvbaha.124.319851] [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] [Indexed: 03/22/2024]
Abstract
Colchicine-an anti-inflammatory alkaloid-has assumed an important role in the management of cardiovascular inflammation ≈3500 years after its first medicinal use in ancient Egypt. Primarily used in high doses for the treatment of acute gout flares during the 20th century, research in the early 21st century demonstrated that low-dose colchicine effectively treats acute gout attacks, lowers the risk of recurrent pericarditis, and can add to secondary prevention of major adverse cardiovascular events. As the first Food and Drug Administration-approved targeted anti-inflammatory cardiovascular therapy, colchicine currently has a unique role in the management of atherosclerotic cardiovascular disease. The safe use of colchicine requires careful monitoring for drug-drug interactions, changes in kidney and liver function, and counseling regarding gastrointestinal upset. Future research should elucidate the mechanisms of anti-inflammatory effects of colchicine relevant to atherosclerosis, the potential role of colchicine in primary prevention, in other cardiometabolic conditions, colchicine's safety in cardiovascular patients, and opportunities for individualizing colchicine therapy using clinical and molecular diagnostics.
Collapse
Affiliation(s)
- Leo F. Buckley
- Department of Pharmacy, Brigham and Women’s Hospital, Boston MA
| | - Peter Libby
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston MA
| |
Collapse
|
35
|
Alabarse PG, Oliveira P, Qin H, Yan T, Migaud M, Terkeltaub R, Liu-Bryan R. The NADase CD38 is a central regulator in gouty inflammation and a novel druggable therapeutic target. Inflamm Res 2024; 73:739-751. [PMID: 38493256 PMCID: PMC11058052 DOI: 10.1007/s00011-024-01863-y] [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: 10/17/2023] [Revised: 01/23/2024] [Accepted: 02/13/2024] [Indexed: 03/18/2024] Open
Abstract
OBJECTIVES Cellular NAD+ declines in inflammatory states associated with increased activity of the leukocyte-expressed NADase CD38. In this study, we tested the potential role of therapeutically targeting CD38 and NAD+ in gout. METHODS We studied cultured mouse wild type and CD38 knockout (KO) murine bone marrow derived macrophages (BMDMs) stimulated by monosodium urate (MSU) crystals and used the air pouch gouty inflammation model. RESULTS MSU crystals induced CD38 in BMDMs in vitro, associated with NAD+ depletion, and IL-1β and CXCL1 release, effects reversed by pharmacologic CD38 inhibitors (apigenin, 78c). Mouse air pouch inflammatory responses to MSU crystals were blunted by CD38 KO and apigenin. Pharmacologic CD38 inhibition suppressed MSU crystal-induced NLRP3 inflammasome activation and increased anti-inflammatory SIRT3-SOD2 activity in macrophages. BMDM RNA-seq analysis of differentially expressed genes (DEGs) revealed CD38 to control multiple MSU crystal-modulated inflammation pathways. Top DEGs included the circadian rhythm modulator GRP176, and the metalloreductase STEAP4 that mediates iron homeostasis, and promotes oxidative stress and NF-κB activation when it is overexpressed. CONCLUSIONS CD38 and NAD+ depletion are druggable targets controlling the MSU crystal- induced inflammation program. Targeting CD38 and NAD+ are potentially novel selective molecular approaches to limit gouty arthritis.
Collapse
Affiliation(s)
- Paulo Gil Alabarse
- VA San Diego Healthcare System, 111K, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA
| | - Patricia Oliveira
- University of California San Diego, La Jolla, San Diego, CA, USA
- The Janssen Pharmaceutical Companies of Johnson & Johnson, La Jolla, San Diego, CA, USA
| | - Huaping Qin
- University of California San Diego, La Jolla, San Diego, CA, USA
| | - Tiffany Yan
- University of California San Diego, La Jolla, San Diego, CA, USA
- Gritstone Bio, Emeryville, CA, USA
| | - Marie Migaud
- Department of Pharmacology, F. Whiddon College of Medicine, Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Robert Terkeltaub
- VA San Diego Healthcare System, 111K, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA
- University of California San Diego, La Jolla, San Diego, CA, USA
| | - Ru Liu-Bryan
- VA San Diego Healthcare System, 111K, 3350 La Jolla Village Drive, San Diego, CA, 92161, USA.
- University of California San Diego, La Jolla, San Diego, CA, USA.
| |
Collapse
|
36
|
Dong H, Wang S, Hu C, Wang M, Zhou T, Zhou Y. Neuroprotective Effects of Intermittent Fasting in the Aging Brain. ANNALS OF NUTRITION & METABOLISM 2024; 80:175-185. [PMID: 38631305 DOI: 10.1159/000538782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 04/06/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND A major risk factor for neurodegenerative disorders is old age. Nutritional interventions that delay aging, such as calorie restriction (CR) and intermittent fasting (IF), as well as pharmaceuticals that affect the pathways linking nutrition and aging processes, have been developed in recent decades and have been shown to alleviate the effects of aging on the brain. SUMMARY CR is accomplished by alternating periods of ad libitum feeding and fasting. In animal models, IF has been shown to increase lifespan and slow the progression and severity of age-related pathologies such as cardiovascular and neurodegenerative diseases and cancer. According to recent research, dietary changes can help older people with dementia retain brain function. However, the mechanisms underlying the neuroprotective effect of IF on the aging brain and related questions in this area of study (i.e., the potential of IF to treat neurodegenerative disorders) remain to be examined. KEY MESSAGES This review addresses the hypothesis that IF may have translational potential in protecting the aged brain while summarizing the research supporting the putative neuroprotective mechanisms of IF in animal models. Additionally, given the emerging understanding of the connection between aging and dementia, our investigations may offer a fresh perspective on the use of dietary interventions for enhancing brain function and preventing dementia in elderly individuals. Finally, the absence of guidelines regarding the application of IF in patients hampers its broad utilization in clinical practice, and further studies are needed to improve our knowledge of the long-term effects of IF on dementia before it can be widely prescribed. In conclusion, IF may be an ancillary intervention for preserving memory and cognition in elderly individuals.
Collapse
Affiliation(s)
- Hao Dong
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Shiyan Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Chenji Hu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Mao Wang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhou
- Department of Pharmaceutical and Medical Equipment, Ba Yi Orthopedic Hospital, Chengdu, China
| | - Yue Zhou
- Department of Pharmacy, Xindu District People's Hospital of Chengdu, Chengdu, China
| |
Collapse
|
37
|
Sundaram B, Tweedell RE, Prasanth Kumar S, Kanneganti TD. The NLR family of innate immune and cell death sensors. Immunity 2024; 57:674-699. [PMID: 38599165 PMCID: PMC11112261 DOI: 10.1016/j.immuni.2024.03.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 04/12/2024]
Abstract
Nucleotide-binding oligomerization domain (NOD)-like receptors, also known as nucleotide-binding leucine-rich repeat receptors (NLRs), are a family of cytosolic pattern recognition receptors that detect a wide variety of pathogenic and sterile triggers. Activation of specific NLRs initiates pro- or anti-inflammatory signaling cascades and the formation of inflammasomes-multi-protein complexes that induce caspase-1 activation to drive inflammatory cytokine maturation and lytic cell death, pyroptosis. Certain NLRs and inflammasomes act as integral components of larger cell death complexes-PANoptosomes-driving another form of lytic cell death, PANoptosis. Here, we review the current understanding of the evolution, structure, and function of NLRs in health and disease. We discuss the concept of NLR networks and their roles in driving cell death and immunity. An improved mechanistic understanding of NLRs may provide therapeutic strategies applicable across infectious and inflammatory diseases and in cancer.
Collapse
Affiliation(s)
- Balamurugan Sundaram
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Rebecca E Tweedell
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | |
Collapse
|
38
|
Zhu C, Xu S, Jiang R, Yu Y, Bian J, Zou Z. The gasdermin family: emerging therapeutic targets in diseases. Signal Transduct Target Ther 2024; 9:87. [PMID: 38584157 PMCID: PMC10999458 DOI: 10.1038/s41392-024-01801-8] [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: 01/15/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024] Open
Abstract
The gasdermin (GSDM) family has garnered significant attention for its pivotal role in immunity and disease as a key player in pyroptosis. This recently characterized class of pore-forming effector proteins is pivotal in orchestrating processes such as membrane permeabilization, pyroptosis, and the follow-up inflammatory response, which are crucial self-defense mechanisms against irritants and infections. GSDMs have been implicated in a range of diseases including, but not limited to, sepsis, viral infections, and cancer, either through involvement in pyroptosis or independently of this process. The regulation of GSDM-mediated pyroptosis is gaining recognition as a promising therapeutic strategy for the treatment of various diseases. Current strategies for inhibiting GSDMD primarily involve binding to GSDMD, blocking GSDMD cleavage or inhibiting GSDMD-N-terminal (NT) oligomerization, albeit with some off-target effects. In this review, we delve into the cutting-edge understanding of the interplay between GSDMs and pyroptosis, elucidate the activation mechanisms of GSDMs, explore their associations with a range of diseases, and discuss recent advancements and potential strategies for developing GSDMD inhibitors.
Collapse
Affiliation(s)
- Chenglong Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
| | - Sheng Xu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Ruoyu Jiang
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Naval Medical University, Shanghai, 200433, China
| | - Yizhi Yu
- National Key Laboratory of Immunity & Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Jinjun Bian
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Zui Zou
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- School of Anesthesiology, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
39
|
Wen S, Arakawa H, Tamai I. Uric acid in health and disease: From physiological functions to pathogenic mechanisms. Pharmacol Ther 2024; 256:108615. [PMID: 38382882 DOI: 10.1016/j.pharmthera.2024.108615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/02/2024] [Accepted: 02/17/2024] [Indexed: 02/23/2024]
Abstract
Owing to renal reabsorption and the loss of uricase activity, uric acid (UA) is strictly maintained at a higher physiological level in humans than in other mammals, which provides a survival advantage during evolution but increases susceptibility to certain diseases such as gout. Although monosodium urate (MSU) crystal precipitation has been detected in different tissues of patients as a trigger for disease, the pathological role of soluble UA remains controversial due to the lack of causality in the clinical setting. Abnormal elevation or reduction of UA levels has been linked to some of pathological status, also known as U-shaped association, implying that the physiological levels of UA regulated by multiple enzymes and transporters are crucial for the maintenance of health. In addition, the protective potential of UA has also been proposed in aging and some diseases. Therefore, the role of UA as a double-edged sword in humans is determined by its physiological or non-physiological levels. In this review, we summarize biosynthesis, membrane transport, and physiological functions of UA. Then, we discuss the pathological involvement of hyperuricemia and hypouricemia as well as the underlying mechanisms by which UA at abnormal levels regulates the onset and progression of diseases. Finally, pharmacological strategies for urate-lowering therapy (ULT) are introduced, and current challenges in UA study and future perspectives are also described.
Collapse
Affiliation(s)
- Shijie Wen
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Arakawa
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan
| | - Ikumi Tamai
- Faculty of Pharmaceutical Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
40
|
Putnam CD, Broderick L, Hoffman HM. The discovery of NLRP3 and its function in cryopyrin-associated periodic syndromes and innate immunity. Immunol Rev 2024; 322:259-282. [PMID: 38146057 PMCID: PMC10950545 DOI: 10.1111/imr.13292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 12/27/2023]
Abstract
From studies of individual families to global collaborative efforts, the NLRP3 inflammasome is now recognized to be a key regulator of innate immunity. Activated by a panoply of pathogen-associated and endogenous triggers, NLRP3 serves as an intracellular sensor that drives carefully coordinated assembly of the inflammasome, and downstream inflammation mediated by IL-1 and IL-18. Initially discovered as the cause of the autoinflammatory spectrum of cryopyrin-associated periodic syndrome (CAPS), NLRP3 is now also known to play a role in more common diseases including cardiovascular disease, gout, and liver disease. We have seen cohesion in results from clinical studies in CAPS patients, ex vivo studies of human cells and murine cells, and in vivo murine models leading to our understanding of the downstream pathways, cytokine secretion, and cell death pathways that has solidified the role of autoinflammation in the pathogenesis of human disease. Recent advances in our understanding of the structure of the inflammasome have provided ways for us to visualize normal and mutant protein function and pharmacologic inhibition. The subsequent development of targeted therapies successfully used in the treatment of patients with CAPS completes the bench to bedside translational loop which has defined the study of this unique protein.
Collapse
Affiliation(s)
- Christopher D. Putnam
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Lori Broderick
- Division of Allergy, Immunology & Rheumatology, Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Rady Children’s Hospital, San Diego, California, USA
| | - Hal M. Hoffman
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- Division of Allergy, Immunology & Rheumatology, Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
- Rady Children’s Hospital, San Diego, California, USA
| |
Collapse
|
41
|
Que X, Zheng S, Song Q, Pei H, Zhang P. Fantastic voyage: The journey of NLRP3 inflammasome activation. Genes Dis 2024; 11:819-829. [PMID: 37692521 PMCID: PMC10491867 DOI: 10.1016/j.gendis.2023.01.009] [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: 12/07/2022] [Accepted: 01/07/2023] [Indexed: 09/12/2023] Open
Abstract
NLRP3 inflammasome, an intracellular multiprotein complex, can be activated by a range of pathogenic microbes or endogenous hazardous chemicals. Its activation results in the release of cytokines such as IL-1β and IL-18, as well as Gasdermin D which eventually causes pyroptosis. The activation of NLRP3 inflammasome is under strict control and regulation by numerous pathways and mechanisms. Its excessive activation can lead to a persistent inflammatory response, which is linked to the onset and progression of severe illnesses. Recent studies have revealed that the subcellular localization of NLRP3 changes significantly during the activation process. In this review, we review the current understanding of the molecular mechanism of NLRP3 inflammasome activation, focusing on the subcellular localization of NLRP3 and the associated regulatory mechanisms. We aim to provide a comprehensive understanding of the dynamic transportation, activation, and degradation processes of NLRP3.
Collapse
Affiliation(s)
- Xiangyong Que
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Sihao Zheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Qibin Song
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Huadong Pei
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Pingfeng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
42
|
Lee KG, Hong BK, Lee S, Lee N, Kim SW, Kim D, Kim WU. Nuclear receptor coactivator 6 is a critical regulator of NLRP3 inflammasome activation and gouty arthritis. Cell Mol Immunol 2024; 21:227-244. [PMID: 38195836 PMCID: PMC10902316 DOI: 10.1038/s41423-023-01121-x] [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: 04/27/2023] [Accepted: 11/30/2023] [Indexed: 01/11/2024] Open
Abstract
Transcriptional coactivators regulate the rate of gene expression in the nucleus. Nuclear receptor coactivator 6 (NCOA6), a coactivator, has been implicated in embryonic development, metabolism, and cancer pathogenesis, but its role in innate immunity and inflammatory diseases remains unclear. Here, we demonstrated that NCOA6 was expressed in monocytes and macrophages and that its level was increased under proinflammatory conditions. Unexpectedly, nuclear NCOA6 was found to translocate to the cytoplasm in activated monocytes and then become incorporated into the inflammasome with NLRP3 and ASC, forming cytoplasmic specks. Mechanistically, NCOA6 associated with the ATP hydrolysis motifs in the NACHT domain of NLRP3, promoting the oligomerization of NLRP3 and ASC and thereby instigating the production of IL-1β and active caspase-1. Of note, Ncoa6 deficiency markedly inhibited NLRP3 hyperactivation caused by the Nlrp3R258W gain-of-function mutation in macrophages. Genetic ablation of Ncoa6 substantially attenuated the severity of two NLRP3-dependent diseases, folic-induced acute tubular necrosis and crystal-induced arthritis, in mice. Consistent with these findings, NCOA6 was highly expressed in macrophages derived from gout patients, and NCOA6-positive macrophages were significantly enriched in gout macrophages according to the transcriptome profiling results. Conclusively, NCOA6 is a critical regulator of NLRP3 inflammasome activation and is therefore a promising target for NLRP3-dependent diseases, including gout.
Collapse
Affiliation(s)
- Kang-Gu Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Bong-Ki Hong
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Saseong Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Naeun Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Seung-Whan Kim
- Department of Pharmacology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
- Bio-Medical Institute of Technology, University of Ulsan, Seoul, 05505, Republic of Korea
| | - Donghyun Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Department of Biomedicine & Health Sciences, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
43
|
Lucena-Agell D, Guillén MJ, Matesanz R, Álvarez-Bernad B, Hortigüela R, Avilés P, Martínez-Díez M, Santamaría-Núñez G, Contreras J, Plaza-Menacho I, Giménez-Abián JF, Oliva MA, Cuevas C, Díaz JF. PM534, an Optimized Target-Protein Interaction Strategy through the Colchicine Site of Tubulin. J Med Chem 2024; 67:2619-2630. [PMID: 38294341 PMCID: PMC10895673 DOI: 10.1021/acs.jmedchem.3c01775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/02/2024] [Accepted: 01/10/2024] [Indexed: 02/01/2024]
Abstract
Targeting microtubules is the most effective wide-spectrum pharmacological strategy in antitumoral chemotherapy, and current research focuses on reducing main drawbacks: neurotoxicity and resistance. PM534 is a novel synthetic compound derived from the Structure-Activity-Relationship study on the natural molecule PM742, isolated from the sponge of the order Lithistida, family Theonellidae, genus Discodermia (du Bocage 1869). PM534 targets the entire colchicine binding domain of tubulin, covering four of the five centers of the pharmacophore model. Its nanomolar affinity and high retention time modulate a strikingly high antitumor activity that efficiently overrides two resistance mechanisms in cells (detoxification pumps and tubulin βIII isotype overexpression). Furthermore, PM534 induces significant inhibition of tumor growth in mouse xenograft models of human non-small cell lung cancer. Our results present PM534, a highly effective new compound in the preclinical evaluation that is currently in its first human Phase I clinical trial.
Collapse
Affiliation(s)
- Daniel Lucena-Agell
- Unidad
BICS. Centro de Investigaciones Biológicas Margarita Salas,
Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - María José Guillén
- PharmaMar
S.A., Avda de los Reyes
1, Colmenar Viejo, 28770 Madrid, Spain
| | - Ruth Matesanz
- Unidad
BICS. Centro de Investigaciones Biológicas Margarita Salas,
Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Beatriz Álvarez-Bernad
- Unidad
BICS. Centro de Investigaciones Biológicas Margarita Salas,
Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Rafael Hortigüela
- Unidad
BICS. Centro de Investigaciones Biológicas Margarita Salas,
Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Pablo Avilés
- PharmaMar
S.A., Avda de los Reyes
1, Colmenar Viejo, 28770 Madrid, Spain
| | | | | | - Julia Contreras
- Centro
Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro
3, 28029 Madrid, Spain
| | - Iván Plaza-Menacho
- Centro
Nacional de Investigaciones Oncológicas (CNIO), Melchor Fernández Almagro
3, 28029 Madrid, Spain
| | - Juan F. Giménez-Abián
- Unidad
BICS. Centro de Investigaciones Biológicas Margarita Salas,
Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - María A. Oliva
- Unidad
BICS. Centro de Investigaciones Biológicas Margarita Salas,
Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Carmen Cuevas
- PharmaMar
S.A., Avda de los Reyes
1, Colmenar Viejo, 28770 Madrid, Spain
| | - J. Fernando Díaz
- Unidad
BICS. Centro de Investigaciones Biológicas Margarita Salas,
Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, 28040 Madrid, Spain
| |
Collapse
|
44
|
Park K, Shin I, Kim Y, Kang H, Oh SJ, Jang E, Sim T, Youn J, Lee MS. A novel NLRP3 inhibitor as a therapeutic agent against monosodium urate-induced gout. Front Immunol 2024; 14:1307739. [PMID: 38371945 PMCID: PMC10869544 DOI: 10.3389/fimmu.2023.1307739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/20/2023] [Indexed: 02/20/2024] Open
Abstract
Background Since NEK7 is critical for NLRP3 inflammasome activation, NEK7 inhibitors could be employed as therapeutic agents against gout, a representative disease caused by NLRP3 inflammasome. Methods We designed NEK7 inhibitors based on biochemical kinome profiling of 2,7-substituted thieno[3,2-d]pyrimidine derivatives (SLC3031~3035 and SLC3037). Inflammasome activation was assessed by ELISA of IL-1b and immunoblotting of IL-1b maturation after treatment of bone marrow-derived macrophages with LPS+monosodium urate (MSU). NLPR3 binding to NEK7 and oligomerization were examined using immunoprecipitation and Blue Native gel electrophoresis, respectively. In vivo effect was investigated by studying gross and histopathological changes of food pad tissue of MSU-injected mice, together with assays of maturation of IL-1b and ASC speck in the tissue. Results SLC3037 inhibited inflammasome by MSU and other inflammasome activators through blockade of NLRP3 binding to NEK7 or oligomerization, and subsequent ASC oligomerization/phosphorylation. SLC3037 significantly reduced foot pad thickness and inflammation by MSU, which was superior to the effects of colchicine. SLC3037 significantly reduced content or maturation of IL-1b and ASC speck in the food pad. The number and height of intestinal villi were decreased by colchicine but not by SLC3037. Conclusion SLC3037, a NLRP3 inhibitor blocking NEK7 binding to NLRP3, could be a novel agent against diseases associated with NLRP3 inflammasome activation such as gout, cardiovascular diseases, metabolic syndrome or neurodegenerative diseases.
Collapse
Affiliation(s)
- Kihyoun Park
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Injae Shin
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Yoonseon Kim
- Department of Biomedical Science, Hanyang University, Seoul, Republic of Korea
| | - Hyereen Kang
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soo-Jin Oh
- Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Republic of Korea
| | - Eunkyeong Jang
- Department of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Taebo Sim
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jeehee Youn
- Department of Biomedical Science, Hanyang University, Seoul, Republic of Korea
- Department of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Myung-Shik Lee
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Soonchunhyang Institute of Medi-bio Science and Division of Endocrinology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Cheonan, Republic of Korea
| |
Collapse
|
45
|
Lilov A, Palaveev K, Mitev V. High Doses of Colchicine Act As "Silver Bullets" Against Severe COVID-19. Cureus 2024; 16:e54441. [PMID: 38510906 PMCID: PMC10951753 DOI: 10.7759/cureus.54441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/17/2024] [Indexed: 03/22/2024] Open
Abstract
A 48-year-old patient with a weight of 120 kg with type 2 diabetes mellitus, hypertension, and gout was hospitalized on the third day of the COVID-19 diagnosis. His general condition is relatively good, oxygen saturation is 89%. Despite starting standard treatment, on the seventh day from the onset of symptoms, the patient deteriorated sharply (oxygen saturation dropped to 74%). The negative development of the disease is interrupted with a loading dose of colchicine of 6 mg. This is a typical case of the life-saving effect of high but safe doses of colchicine in high-risk COVID-19 patients.
Collapse
Affiliation(s)
- Alexander Lilov
- Pulmonology, Specialized Hospital for Active Treatment Pneumo-Pneumonia-Phthisiatric Diseases (SHATPPD) "Sofia District" Hospital, Sofia, BGR
| | - Kiril Palaveev
- Pulmonology, Specialized Hospital for Active Treatment Pneumo-Pneumonia-Phthisiatric Diseases (SHATPPD) "Sofia District" Hospital, Sofia, BGR
| | - Vanyo Mitev
- Chemistry and Biochemistry, Medical University of Sofia, Sofia, BGR
| |
Collapse
|
46
|
Bausson J, Keller N, Von Hunolstein JJ, Sacrez M, Michel B, Gourieux B, Fourtage M, Felten R. Safety and efficacy of colchicine in crystal-induced arthritis flare in 54 patients with severe chronic kidney disease. RMD Open 2024; 10:e003872. [PMID: 38296804 PMCID: PMC10831468 DOI: 10.1136/rmdopen-2023-003872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/17/2024] [Indexed: 02/02/2024] Open
Abstract
INTRODUCTION Colchicine, commonly used in gout flare, is contraindicated in severe chronic kidney disease (CKD) (estimated glomerular filtration rate <30 mL/min). However, in this context, there are few alternatives, and colchicine use persists. We evaluated the tolerance of colchicine and its efficacy in patients with severe CKD. PATIENTS AND METHODS All prescriptions of colchicine for managing crystal-induced arthritis flare (gout or calcium pyrophosphate deposition (CPPD) disease) in a hospitalised patient with severe CKD were screened from September 2020 to September 2021. After patient consent and treatment information, clinical and biological safety and efficacy data were prospectively collected from day 1 (D1) to D11. RESULTS We included 54 patients (median age 75 years (IQR 67-83)) with 62 colchicine prescriptions (cases). Twelve (22%) patients were on dialysis. The main reason for hospitalisation was heart failure (31.5%), acute renal failure (22.2%), infection (18.5%) or an acute joint episode (9.3%). In total, 59.3% of patients had diabetes. The prescriptions concerned 58 cases of gout flares, 1 case of CPPD and 3 cases of both. Initial colchicine dosages were ≤0.5 mg/day in 47/62 (75.8%) cases; no dosage exceeded 1 mg/day (median duration of 6 days (IQR 3-11)). Colchicine was well tolerated in 47/61 (77%) cases. No serious adverse event was reported. Colchicine was considered completely effective by the medical team in 48/58 (83%) of cases. CONCLUSION The use of colchicine, at reduced doses, was mostly effective to treat crystal-induced arthritis flare in 54 patients with severe CKD and was well tolerated, without any serious adverse events.
Collapse
Affiliation(s)
- Johanna Bausson
- Service de Pharmacie-Stérilisation, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Nicolas Keller
- Service de Néphrologie, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | | | - Matthieu Sacrez
- Service de Pharmacie-Stérilisation, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Bruno Michel
- Service de Pharmacie-Stérilisation, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Bénédicte Gourieux
- Service de Pharmacie-Stérilisation, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Marion Fourtage
- Service de Pharmacie-Stérilisation, Hôpitaux universitaires de Strasbourg, Strasbourg, France
| | - Renaud Felten
- Service de Rhumatologie de Hautepierre, RESO, Centre de Référence des Maladies Autoimmunes Systémiques Rares Est Sud-Ouest, Hôpitaux universitaires de Strasbourg, Strasbourg, France
- Centre d'Investigation Clinique, Inserm 1434, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
- Département Universitaire de Pharmacologie, Addictologie, Toxicologie et Thérapeutique (DUPATT), Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
47
|
Ohashi Y, Ooyama H, Makinoshima H, Takada T, Matsuo H, Ichida K. Plasma and Urinary Metabolomic Analysis of Gout and Asymptomatic Hyperuricemia and Profiling of Potential Biomarkers: A Pilot Study. Biomedicines 2024; 12:300. [PMID: 38397902 PMCID: PMC10887286 DOI: 10.3390/biomedicines12020300] [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: 12/29/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Gout results from monosodium urate deposition caused by hyperuricemia, but most individuals with hyperuricemia remain asymptomatic. The pathogenesis of gout remains uncertain. To identify potential biomarkers distinguishing gout from asymptomatic hyperuricemia, we conducted a genetic analysis of urate transporters and metabolomic analysis as a proof-of-concept study, including 33 patients with gout and 9 individuals with asymptomatic hyperuricemia. The variant allele frequencies of rs72552713, rs2231142, and rs3733591, which are related to serum urate levels (SUA) and gout, did not differ between the gout and asymptomatic hyperuricemia groups. In metabolomic analysis, the levels of citrate cycle intermediates, especially 2-ketoglutarate, were higher in patients with gout than in those with asymptomatic hyperuricemia (fold difference = 1.415, p = 0.039). The impact on the TCA cycle was further emphasized in high-risk gout (SUA ≥ 9.0 mg/dL). Of note, urinary nicotinate was the most prominent biomarker differentiating high-risk gout from asymptomatic hyperuricemia (fold difference = 6.515, p = 0.020). Although urate transporters play critical roles in SUA elevation and promote hyperuricemia, this study suggests that the progression from asymptomatic hyperuricemia to gout might be closely related to other genetic and/or environmental factors affecting carbohydrate metabolism and urinary urate excretion.
Collapse
Affiliation(s)
- Yuki Ohashi
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan;
- Department of Pharmacy, International University of Health and Welfare, Tochigi 324-8501, Japan
| | | | - Hideki Makinoshima
- Tsuruoka Metabolomics Laboratory, National Cancer Center, Yamagata 997-0052, Japan;
| | - Tappei Takada
- Department of Pharmacy, University of Tokyo Hospital, Faculty of Medicine, University of Tokyo, Tokyo 113-8655, Japan;
| | - Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Saitama 359-8513, Japan;
| | - Kimiyoshi Ichida
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan;
- Division of Kidney and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo 105-8461, Japan
- Chiba Health Promotion Center, East Japan Railway Company, Chiba 260-0045, Japan
| |
Collapse
|
48
|
Alvarez S, Vanasco V, Adán Areán JS, Magnani N, Evelson P. Mitochondrial Mechanisms in Immunity and Inflammatory Conditions: Beyond Energy Management. Antioxid Redox Signal 2024. [PMID: 38062738 DOI: 10.1089/ars.2023.0367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Significance: The growing importance of mitochondria in the immune response and inflammation is multifaceted. Unraveling the different mechanisms by which mitochondria have a relevant role in the inflammatory response beyond the energy management of the process is necessary for improving our understanding of the host immune defense and the pathogenesis of various inflammatory diseases and syndromes. Critical Issues: Mitochondria are relevant in the immune response at different levels, including releasing activation molecules, changing its structure and function to accompany the immune response, and serving as a structural base for activating intermediates as NLRP3 inflammasome. In this scientific journey of dissecting mitochondrial mechanisms, new questions and interesting aspects arise, such as the involvement of mitochondrial-derived vesicles in the immune response with the putative role of preventing uncontrolled situations. Recent Advances: Researchers are continuously rethinking the role of mitochondria in acute and chronic inflammation and related disorders. As such, mitochondria have important roles as centrally positioned signaling hubs in regulating inflammatory and immune responses. In this review, we present the current understanding of mitochondrial mechanisms involved, beyond the largely known mitochondrial dysfunction, in the onset and development of inflammatory situations. Future Directions: Mitochondria emerge as an interesting and multifaceted platform for studying and developing pharmaceutical and therapeutic approaches. There are many ongoing studies aimed to describe the effects of specific mitochondrial targeted molecules and treatments to ameliorate the consequences of exacerbated inflammatory components of pathologies and syndromes, resulting in an open area of increasing research interest.
Collapse
Affiliation(s)
- Silvia Alvarez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Fisicoquímica, CABA, Argentina
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Química General e Inorgánica, CABA, Argentina
| | - Virginia Vanasco
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Fisicoquímica, CABA, Argentina
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Química General e Inorgánica, CABA, Argentina
| | - Juan Santiago Adán Areán
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Fisicoquímica, CABA, Argentina
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Química General e Inorgánica, CABA, Argentina
| | - Natalia Magnani
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Química General e Inorgánica, CABA, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, CABA, Argentina
| | - Pablo Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Química General e Inorgánica, CABA, Argentina
- CONICET-Universidad de Buenos Aires, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Buenos Aires, CABA, Argentina
| |
Collapse
|
49
|
Ye S, Lyu Y, Chen L, Wang Y, He Y, Li Q, Tian L, Liu F, Wang X, Ai F. Construction of a molecular inflammatory predictive model with histone modification-related genes and identification of CAMK2D as a potential response signature to infliximab in ulcerative colitis. Front Immunol 2024; 14:1282136. [PMID: 38274809 PMCID: PMC10808628 DOI: 10.3389/fimmu.2023.1282136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
Background Ulcerative colitis (UC) is a lifelong inflammatory disease affecting the rectum and colon with numerous treatment options that require an individualized treatment plan. Histone modifications regulate chromosome structure and gene expression, resulting in effects on inflammatory and immune responses. However, the relationship between histone modification-related genes and UC remains unclear. Methods Transcriptomic data from GSE59071 and GSE66407 were obtained from the Gene Expression Omnibus (GEO), encompassing colonic biopsy expression profiles of UC patients in inflamed and non-inflamed status. Differentially expressed gene (DEG) analyses, functional enrichment analyses, weighted gene co-expression network analysis (WGCNA), and random forest were performed to identify histone modification-related core genes associated with UC inflammation. Features were screened through the least absolute shrinkage and selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE), establishing a molecular inflammatory predictive model using logistic regression. The model was validated in the GSE107499 dataset, and the performance of the features was assessed using receiver operating characteristic (ROC) and calibration curves. Immunohistochemistry (IHC) staining of colonic biopsy tissues from UC patients treated with infliximab was used to further confirm the clinical application value. Univariate logistic regression on GSE14580 highlighted features linked to infliximab response. Results A total of 253 histone modification-related DEGs were identified between inflammatory and non-inflammatory patients with UC. Seven key genes (IL-1β, MSL3, HDAC7, IRF4, CAMK2D, AUTS2, and PADI2) were selected using WGCNA and random forest. Through univariate logistic regression, three core genes (CAMK2D, AUTS2, and IL-1β) were further incorporated to construct the molecular inflammatory predictive model. The area under the curve (AUC) of the model was 0.943 in the independent validation dataset. A significant association between CAMK2D protein expression and infliximab response was observed, which was validated in another independent verification set of GSE14580 from the GEO database. Conclusion The molecular inflammatory predictive model based on CAMK2D, AUTS2, and IL-1β could reliably distinguish the mucosal inflammatory status of UC patients. We further revealed that CAMK2D was a predictive marker of infliximab response. These findings are expected to provide a new evidence base for personalized treatment and management strategies for UC patients.
Collapse
Affiliation(s)
- Shuyu Ye
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yongqing Lyu
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Libin Chen
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yiwei Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yue He
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Quansi Li
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Li Tian
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Fen Liu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyan Wang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Feiyan Ai
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
50
|
La Bella S, Di Ludovico A, Di Donato G, Basaran O, Ozen S, Gattorno M, Chiarelli F, Breda L. The pyrin inflammasome, a leading actor in pediatric autoinflammatory diseases. Front Immunol 2024; 14:1341680. [PMID: 38250061 PMCID: PMC10796709 DOI: 10.3389/fimmu.2023.1341680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024] Open
Abstract
The activation of the pyrin inflammasome represents a highly intriguing mechanism employed by the innate immune system to effectively counteract pathogenic agents. Despite its key role in innate immunity, pyrin has also garnered significant attention due to its association with a range of autoinflammatory diseases (AIDs) including familial Mediterranean fever caused by disruption of the MEFV gene, or in other genes involved in its complex regulation mechanisms. Pyrin activation is strictly dependent on homeostasis-altering molecular processes, mostly consisting of the disruption of the small Ras Homolog Family Member A (RhoA) GTPases by pathogen toxins. The downstream pathways are regulated by the phosphorylation of specific pyrin residues by the kinases PKN1/2 and the binding of the chaperone 14-3-3. Furthermore, a key role in pyrin activation is played by the cytoskeleton and gasdermin D, which is responsible for membrane pores in the context of pyroptosis. In addition, recent evidence has highlighted the role of steroid hormone catabolites and alarmins S100A8/A9 and S100A12 in pyrin-dependent inflammation. The aim of this article is to offer a comprehensive overview of the most recent evidence on the pyrin inflammasome and its molecular pathways to better understand the pathogenesis behind the significant group of pyrin-related AIDs.
Collapse
Affiliation(s)
- Saverio La Bella
- Department of Pediatrics, "G. D'Annunzio" University of Chieti, Chieti, Italy
| | - Armando Di Ludovico
- Department of Pediatrics, "G. D'Annunzio" University of Chieti, Chieti, Italy
| | - Giulia Di Donato
- Department of Pediatrics, "G. D'Annunzio" University of Chieti, Chieti, Italy
| | - Ozge Basaran
- Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine, Ankara, Türkiye
| | - Seza Ozen
- Department of Pediatrics, Division of Rheumatology, Hacettepe University Faculty of Medicine, Ankara, Türkiye
| | - Marco Gattorno
- UOC Rheumatology and Autoinflammatory Diseases, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Francesco Chiarelli
- Department of Pediatrics, "G. D'Annunzio" University of Chieti, Chieti, Italy
| | - Luciana Breda
- Department of Pediatrics, "G. D'Annunzio" University of Chieti, Chieti, Italy
| |
Collapse
|