1
|
Lai Y, Zhuang L, Zhu J, Wang S, Guo C, Chen B, Li J, Shi J, Li M, Yang N, Zhou M. Novel approach to alleviate lupus nephritis: targeting the NLRP3 inflammasome in CD8 +CD69 +CD103 + T RM cells. J Transl Med 2024; 22:1139. [PMID: 39716284 DOI: 10.1186/s12967-024-05951-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/06/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Renal CD8+ tissue-resident memory T (TRM) cells display prolonged survival and activity in lupus nephritis (LN), exacerbating renal pathology. NLRP3 regulates the T cell response. This study explored the impact of NLRP3 inflammasome activity on the regulatory functions of TRM cells in LN. METHODS NLRP3 inflammasome activity in renal CD8+ TRM cells from lupus-prone MRL/lpr mice and in vitro induced human CD8+CD103+ T cells was assessed by quantifying NLRP3, caspase-1, gasdermin D (GSDMD), and IL-1β levels using flow cytometry, ELISA, and western blotting analysis. The specific NLRP3 inhibitor MCC950, caspase-1 inhibitor Ac-YVAD-cmk, and NF-κB inhibitor JSH23 were utilized to delineate the role of NLRP3 in modulating the pathogenicity of CD8+ TRM cells in LN. RESULTS Activation of the NLRP3 inflammasome was confirmed in renal CD8+CD69+CD103+ TRM cells derived from mice with LN and in vitro-induced human CD8+CD103+ TRM-like cells. MCC950 curtailed the infiltration and activity of CD8+CD69+CD103+ TRM cells and enhanced renal outcomes. MCC950 also suppressed the maturation and functional capabilities of CD8+CD103+ T cells in a manner reliant on inflammasome activity in vitro. IL-1β promoted the expression of TGF-βRII in CD8+ T cells via the NF-κB pathway. CONCLUSIONS NLRP3 inflammasome activity in renal CD8+CD69+CD103+ TRM cells contributes to LN pathogenesis by regulating cell differentiation and effector functions. Therapeutically targeting the NLRP3 inflammasome could significantly mitigate CD8+CD69+CD103+ TRM cell-mediated renal damage in LN.
Collapse
Affiliation(s)
- Yimei Lai
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China
| | - Lili Zhuang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China
| | - Jieying Zhu
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China
| | - Shuang Wang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China
| | - Chaohuan Guo
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China
| | - Binfeng Chen
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China
| | - Jin Li
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China
| | - Jia Shi
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China
| | - Mengyuan Li
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China
| | - Niansheng Yang
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China.
| | - Mianjing Zhou
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan Second Road, Guangzhou, 510080, P. R. China.
| |
Collapse
|
2
|
Chen J, Xie S, Qiu D, Xie M, Wu M, Li X, Zhang X, Wu Q, Xiong Y, Wu C, Ren J, Peng Y. The NLRP3 molecule influences the therapeutic effects of mesenchymal stem cells through Glut1-mediated energy metabolic reprogramming. J Adv Res 2024; 65:125-136. [PMID: 38070595 PMCID: PMC11519012 DOI: 10.1016/j.jare.2023.12.006] [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: 03/26/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 02/12/2024] Open
Abstract
INTRODUCTION Numerous studies demonstrated that NLRP3 has been implicated in the pathogenesis of inflammatory bowel disease (IBD). Mesenchymal stem cells (MSCs) regulated the NLRP3 inflammasome, which has emerged as a novel therapeutic approach for treating IBD. OBJECTIVES The exact role of NLRP3 in regulating MSCs' function is unclear. Our study aimed to explore how NLRP3 affects the therapeutic effects of MSCs in colitis. METHODS We extracted MSCs from the bone marrow of C57BL/6 mice and Nlrp3 KO mice, and identified them using differentiation assays and flow cytometry. In vitro, Both WT MSCs and Nlrp3 KO MSCs were stimulated with inflammatory factor Lipopolysaccharide (LPS), and only WT MSCs were stimulated with varying concentrations of the NLRP3 inhibitor MCC950, then, quantified IL-10 levels in the supernatant. RNA-seq was performed to examine gene expression patterns and Seahorse was used to assess oxidative phosphorylation (OXPHOS) and glycolysis levels. Western blot was used to evaluate protein expression. In vivo, we treated DSS-induced colitis with either WT or Nlrp3 KO MSCs, monitoring weight, measuring colon length, and further evaluation. We also treated DSS-induced colitis with pretreated MSCs (BAY876, oe-Glut1, or oe-NLRP3), following the same experimental procedures as described above. RESULTS Our results demonstrate that Nlrp3 deletion did not affect MSC phenotypes, but rather promoted osteogenic differentiation. However, the absence of Nlrp3 reduced IL-10 production in MSCs in the presence of LPS, leading to impaired protection on DSS-induced colitis. Conversely, overexpression of NLRP3 promotes the production of IL-10, enhancing therapeutic effects. Further investigation revealed that Nlrp3 deficiency downregulated Glut1 expression and glycolysis activation in MSCs, resulting in decreased IL-10 production. Notably, overexpressing Glut1 in Nlrp3 KO MSCs restored their therapeutic effect that was previously dampened due to Nlrp3 deletion. CONCLUSION Our findings demonstrate that NLRP3 heightens the therapeutic effects of MSC treatment on DSS-induced colitis.
Collapse
Affiliation(s)
- Jingrou Chen
- The Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Shujuan Xie
- The Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Dongbo Qiu
- The Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Maosheng Xie
- Department of Rheumatology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Mengye Wu
- The Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xiaoping Li
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-sen University, Guangzhou 510630, Guangdong, China
| | - Xiaoran Zhang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qili Wu
- Medical Research Center, Guangdong Provincial Hospital, Guangzhou 510080, China
| | - Yi Xiong
- The Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Changyou Wu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jie Ren
- Department of Medical Ultrasonic, The Third Affiliated Hospital, Sun Yat-sen University, No. 600 Tianhe Road, Guangzhou 510630, China.
| | - Yanwen Peng
- The Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
3
|
Kattelus R, Starskaia I, Lindén M, Batkulwar K, Pietilä S, Moulder R, Marson A, Rasool O, Suomi T, Elo LL, Lahesmaa R, Buchacher T. Phenotypic profiling of human induced regulatory T cells at early differentiation: insights into distinct immunosuppressive potential. Cell Mol Life Sci 2024; 81:399. [PMID: 39264416 PMCID: PMC11393232 DOI: 10.1007/s00018-024-05429-3] [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: 06/26/2024] [Revised: 08/20/2024] [Accepted: 08/27/2024] [Indexed: 09/13/2024]
Abstract
Regulatory T cells (Tregs) play a key role in suppressing systemic effector immune responses, thereby preventing autoimmune diseases but also potentially contributing to tumor progression. Thus, there is great interest in clinically manipulating Tregs, but the precise mechanisms governing in vitro-induced Treg (iTreg) differentiation are not yet fully understood. Here, we used multiparametric mass cytometry to phenotypically profile human iTregs during the early stages of in vitro differentiation at single-cell level. A panel of 25 metal-conjugated antibodies specific to markers associated with human Tregs was used to characterize these immunomodulatory cells. We found that iTregs highly express the transcription factor FOXP3, as well as characteristic Treg-associated surface markers (e.g. CD25, PD1, CD137, CCR4, CCR7, CXCR3, and CD103). Expression of co-inhibitory factors (e.g. TIM3, LAG3, and TIGIT) increased slightly at late stages of iTreg differentiation. Further, CD103 was upregulated on a subpopulation of iTregs with greater suppressive capacity than their CD103- counterparts. Using mass-spectrometry-based proteomics, we showed that sorted CD103+ iTregs express factors associated with immunosuppression. Overall, our study highlights that during early stages of differentiation, iTregs resemble memory-like Treg features with immunosuppressive activity, and provides opportunities for further investigation into the molecular mechanisms underlying Treg function.
Collapse
Affiliation(s)
- Roosa Kattelus
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - Inna Starskaia
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Turku Doctoral Programme of Molecular Medicine, University of Turku, Turku, Finland
| | - Markus Lindén
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Kedar Batkulwar
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Sami Pietilä
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Robert Moulder
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Alexander Marson
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, 94158, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Omid Rasool
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Riitta Lahesmaa
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Institute of Biomedicine, University of Turku, Turku, Finland.
| | - Tanja Buchacher
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
| |
Collapse
|
4
|
Atalay Ekiner S, Gęgotek A, Skrzydlewska E. Inflammasome activity regulation by PUFA metabolites. Front Immunol 2024; 15:1452749. [PMID: 39290706 PMCID: PMC11405227 DOI: 10.3389/fimmu.2024.1452749] [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: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Oxidative stress and the accompanying chronic inflammation constitute an important metabolic problem that may lead to pathology, especially when the body is exposed to physicochemical and biological factors, including UV radiation, pathogens, drugs, as well as endogenous metabolic disorders. The cellular response is associated, among others, with changes in lipid metabolism, mainly due to the oxidation and the action of lipolytic enzymes. Products of oxidative fragmentation/cyclization of polyunsaturated fatty acids (PUFAs) [4-HNE, MDA, 8-isoprostanes, neuroprostanes] and eicosanoids generated as a result of the enzymatic metabolism of PUFAs significantly modify cellular metabolism, including inflammation and the functioning of the immune system by interfering with intracellular molecular signaling. The key regulators of inflammation, the effectiveness of which can be regulated by interacting with the products of lipid metabolism under oxidative stress, are inflammasome complexes. An example is both negative or positive regulation of NLRP3 inflammasome activity by 4-HNE depending on the severity of oxidative stress. 4-HNE modifies NLRP3 activity by both direct interaction with NLRP3 and alteration of NF-κB signaling. Furthermore, prostaglandin E2 is known to be positively correlated with both NLRP3 and NLRC4 activity, while its potential interference with AIM2 or NLRP1 activity is unproven. Therefore, the influence of PUFA metabolites on the activity of well-characterized inflammasome complexes is reviewed.
Collapse
Affiliation(s)
| | - Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
5
|
Li Y, Qiang R, Cao Z, Wu Q, Wang J, Lyu W. NLRP3 Inflammasomes: Dual Function in Infectious Diseases. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:407-417. [PMID: 39102612 PMCID: PMC11299487 DOI: 10.4049/jimmunol.2300745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 06/11/2024] [Indexed: 08/07/2024]
Abstract
The Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome has been the most distinctive polymer protein complex. After recognizing the endogenous and exogenous danger signals, NLRP3 can cause inflammation by pyroptosis and secretion of mature, bioactive forms of IL-1β and IL-18. The NLRP3 inflammasome is essential in the genesis and progression of infectious illnesses. Herein, we provide a comprehensive review of the NLRP3 inflammasome in infectious diseases, focusing on its two-sided effects. As an essential part of host defense with a protective impact, abnormal NLRP3 inflammasome activation, however, result in a systemic high inflammatory response, leading to subsequent damage. In addition, scientific evidence of small molecules, biologics, and phytochemicals acting on the NLRP3 inflammasome has been reviewed. We believe that the NLRP3 inflammasome helps us understand the pathological mechanism of different stages of infectious diseases and that inhibitors targeting the NLRP3 inflammasome will become a new and valuable research direction for the treatment of infectious diseases.
Collapse
Affiliation(s)
- Yanbo Li
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing
| | - Rui Qiang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine Shunyi Hospital, Beijing, China
| | - Zhengmin Cao
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing
| | - Qingjuan Wu
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing
| | - Jiuchong Wang
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing
| | - Wenliang Lyu
- Department of Infectious Diseases, Guang’anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing
| |
Collapse
|
6
|
Wang K, Liu J, Yue J, Zhou L, Mao H, Li J, Sun Z, Chen Z, Zhang L. Nlrp3 inflammasome drives regulatory T cell depletion to accelerate periapical bone erosion. Int Endod J 2024; 57:1110-1123. [PMID: 38441141 DOI: 10.1111/iej.14062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/24/2024] [Accepted: 02/26/2024] [Indexed: 07/03/2024]
Abstract
AIM Apical periodontitis is an inflammatory disorder triggered by an immune response to bacterial infection, leading to the periapical tissue damage and alveolar resorption. However, the underlying mechanisms driving this process remain elusive, due to the complex and interconnected immune microenvironment within the local lesion site. In this study, the influence of Nlrp3 inflammasome-mediated immune response on the apical periodontitis was investigated. METHODOLOGY RNA sequencing, immunohistochemistry and ELISA assay were performed to investigate the activation of Nlrp3 inflammasome signalling pathways in the human periapical tissues, including radicular cysts, periapical granulomas and healthy oral mucosa. A mouse model of apical periodontitis was established to study the role of Nlrp3 knockout in periapical bone resorption and Treg cell stability, and the underlying mechanism was explored through in vitro experiments. In vivo Treg cell adoptive transfer was performed to investigate the effects of Treg cells on the progression of apical periodontitis. RESULTS Our findings find that the hyperactivated Nlrp3 inflammasome is present in human periapical lesions and plays a vital role in the immune-related periapical bone loss. Using a mouse model of apical periodontitis, we observe that Nlrp3 deficiency is resistant to bone resorption. This protection was accompanied by elevated generation and infiltration of local Treg cells that displayed a notable ability to suppress RANKL-dependent osteoclast differentiation. In terms of the mechanism of action, Nlrp3 deficiency directly inhibits the osteoclast differentiation and bone loss through JNK/MAPK and NF-κB pathways. In addition, Nlrp3 induces pyroptosis in the stem cells from apical papilla (SCAPs), and the subsequent release of cytokines affects the stability of Treg cell in periapical lesions, leading indirectly to enhanced bone resorption. In turn, adoptive transfer of both Nlrp3-deficient and wild-type Treg cells effectively prevent the bone erosion during apical periodontitis. CONCLUSIONS Together, our data identify that the Nlrp3 inflammasome modulates the Treg cell stability and osteoclastogenesis in the periapical inflammatory microenvironment, thus determining the progression of bone erosion.
Collapse
Affiliation(s)
- Konghuai Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jiayi Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Junli Yue
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lu Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hanqing Mao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Jiaqi Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhijun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lu Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
7
|
Li D, Gao S. The interplay between T lymphocytes and macrophages in myocardial ischemia/reperfusion injury. Mol Cell Biochem 2024; 479:1925-1936. [PMID: 37540399 DOI: 10.1007/s11010-023-04822-z] [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: 06/07/2023] [Accepted: 07/24/2023] [Indexed: 08/05/2023]
Abstract
Acute myocardial infarction is one of the most important causes of death in the world, causing a huge health and economic burden to the world. It is still a ticklish problem how to effectively prevent reperfusion injury while recovering the blood flow of ischemic myocardium. During the process of myocardial ischemia/reperfusion injury (MI/RI), the modulation of immune cells plays an important role. Monocyte/macrophage, neutrophils and endothelial cells initiate the inflammatory response and induce the release of various inflammatory cytokines, resulting in increased vascular permeability, tissue edema and damage. Meanwhile, T cells were recruited to impaired myocardium and release pro-inflammatory and anti-inflammatory cytokines. T cells and macrophages play important roles in keeping cardiac homeostasis and orchestrate tissue repair. T cells differentiation and macrophages polarization precisely regulates the tissue microenvironment in MI/RI, and shows cross action, but the mechanism is unclear. To identify potential intervention targets and propose ideas for treatment and prevention of MI/RI, this review explores the crosstalk between T lymphocytes and macrophages in MI/RI.
Collapse
Affiliation(s)
- Dan Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, 314 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China
- Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Shan Gao
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, 314 An Shan Xi Road, Nan Kai District, Tianjin, 300193, China.
- Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
8
|
Hsu ML, Jhuang KF, Zouali M. Inflammasome functional activities in B lymphocytes. Immunol Res 2024; 72:828-840. [PMID: 38777958 DOI: 10.1007/s12026-024-09490-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024]
Abstract
Studies in animal models and human subjects have shown that, in addition to their implication in innate immunity, inflammasomes also can play a role in adaptive immunity. However, the contribution of the nucleotide-binding oligomerization domain-, leucine-rich repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasome pathway to adaptive immunity remains incompletely explored. Here, we show that NLRP3 plays an important role in different facets of B cell functions, including proliferation, antibody production, and secretion of inflammatory and anti-inflammatory cytokines. When exposed to B cell receptor engagement, Toll-like receptor activation, stimulation in conditions that mimic T cell-dependent responses, or NLRP3 activation, B cells manifest disparate responses and produce different cytokine patterns critical for modulating innate and adaptive immunity, indicating that the cytokines produced serve a critical link between the early innate immune response and the delayed adaptive immunity. Importantly, genetic ablation of nlrp3 reduced the inflammasome-mediated functions of B cells. We propose that, in the absence of other cell types, the potential of B lymphocytes to respond to NLRP3 engagement enables them to initiate inflammatory cascades through recruitment of other cell subsets, such as macrophages and neutrophils. Since NLRP3 activation of B cells is not followed by pyroptosis, even in the presence of a basal caspase-1 activity, this pathway acts as a bridge that optimizes interactions between the innate and adoptive branches of the immune response.
Collapse
Affiliation(s)
- Man Lun Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Xueshi Road, North District, 404, Taichung, Taiwan
| | - Kai Fu Jhuang
- Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Xueshi Road, North District, 404, Taichung, Taiwan
| | - Moncef Zouali
- Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Xueshi Road, North District, 404, Taichung, Taiwan.
| |
Collapse
|
9
|
Alipour S, Mardi A, Shajari N, Kazemi T, Sadeghi MR, Ahmadian Heris J, Masoumi J, Baradaran B. Unmasking the NLRP3 inflammasome in dendritic cells as a potential therapeutic target for autoimmunity, cancer, and infectious conditions. Life Sci 2024; 348:122686. [PMID: 38710282 DOI: 10.1016/j.lfs.2024.122686] [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: 01/17/2024] [Revised: 04/13/2024] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
Proper and functional immune response requires a complex interaction between innate and adaptive immune cells, which dendritic cells (DCs) are the primary actors in this coordination as professional antigen-presenting cells. DCs are armed with numerous pattern recognition receptors (PRRs) such as nucleotide-binding and oligomerization domain-like receptors (NLRs) like NLRP3, which influence the development of their activation state upon sensation of ligands. NLRP3 is a crucial component of the immune system for protection against tumors and infectious agents, because its activation leads to the assembly of inflammasomes that cause the formation of active caspase-1 and stimulate the maturation and release of proinflammatory cytokines. But, when NLRP3 becomes overactivated, it plays a pathogenic role in the progression of several autoimmune disorders. So, NLRP3 activation is strictly regulated by diverse signaling pathways that are mentioned in detail in this review. Furthermore, the role of NLRP3 in all of the diverse immune cells' subsets is briefly mentioned in this study because NLRP3 plays a pivotal role in modulating other immune cells which are accompanied by DCs' responses and subsequently influence differentiation of T cells to diverse T helper subsets and even impact on cytotoxic CD8+ T cells' responses. This review sheds light on the functional and therapeutic role of NLRP3 in DCs and its contribution to the occurrence and progression of autoimmune disorders, prevention of diverse tumors' development, and recognition and annihilation of various infectious agents. Furthermore, we highlight NLRP3 targeting potential for improving DC-based immunotherapeutic approaches, to be used for the benefit of patients suffering from these disorders.
Collapse
Affiliation(s)
- Shiva Alipour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirhossein Mardi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Shajari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Sadeghi
- Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Javad Masoumi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Zouali M. Swaying the advantage: multifaceted functions of inflammasomes in adaptive immunity. FEBS J 2024. [PMID: 38922787 DOI: 10.1111/febs.17204] [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: 09/18/2023] [Revised: 03/17/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Abstract
Eukaryotic cells are equipped with cytoplasmic sensors that recognize diverse pathogen- or danger-associated molecular patterns. In cells of the myeloid lineage, activation of these sensors leads to the assembly of a multimeric protein complex, called the inflammasome, that culminates in the production of inflammatory cytokines and pyroptosis. Recently, investigation of the inflammasomes in lymphocytes led to the discovery of functional pathways that were initially believed to be confined to the innate arm of the immune system. Thus, the adapter protein apoptosis-associated speck-like protein containing a CARD (ASC) was documented to play a critical role in antigen uptake by dendritic cells, and regulation of T- and B-cell motility at several stages, and absent in melanoma 2 (AIM2) was found to act as a modulator of regulatory T-cell differentiation. Remarkably, NLRP3 was demonstrated to act as a transcription factor that controls Th2 cell polarization, and as a negative regulator of regulatory T-cell differentiation by limiting Foxp3 expression. In B lymphocytes, NLRP3 plays a role in the transcriptional network that regulates B-cell development and homing, and its activation is essential for germinal center formation and maturation of high-affinity antibody responses. Such recently discovered inflammasome-mediated functions in T and B lymphocytes offer multiple cross-talk opportunities for the innate and adaptive arms of the immune system. A better understanding of the dialog between inflammasomes and intracellular components could be beneficial for therapeutic purposes in restoring immune homeostasis and mitigating inflammation in a wide range of disorders.
Collapse
Affiliation(s)
- Moncef Zouali
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
11
|
Paul O, Akolia IK, Qin Tao J, Jain N, Louneva N, Montone KT, Fisher AB, Rajapakse CS, Bermudez C, Chatterjee S. Reactive oxygen species in endothelial signaling in COVID-19: Protective role of the novel peptide PIP-2. PLoS One 2024; 19:e0289854. [PMID: 38771750 PMCID: PMC11108150 DOI: 10.1371/journal.pone.0289854] [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: 07/25/2023] [Accepted: 04/02/2024] [Indexed: 05/23/2024] Open
Abstract
INTRODUCTION Recent research suggests that endothelial activation plays a role in coronavirus disease 2019 (COVID-19) pathogenesis by promoting a pro-inflammatory state. However, the mechanism by which the endothelium is activated in COVID-19 remains unclear. OBJECTIVE To investigate the mechanism by which COVID-19 activates the pulmonary endothelium and drives pro-inflammatory phenotypes. HYPOTHESIS The "inflammatory load or burden" (cytokine storm) of the systemic circulation activates endothelial NADPH oxidase 2 (NOX2) which leads to the production of reactive oxygen species (ROS) by the pulmonary endothelium. Endothelial ROS subsequently activates pro-inflammatory pathways. METHODS The inflammatory burden of COVID-19 on the endothelial network, was recreated in vitro, by exposing human pulmonary microvascular endothelial cells (HPMVEC) to media supplemented with serum from COVID-19 affected individuals (sera were acquired from patients with COVID-19 infection that eventually died. Sera was isolated from blood collected at admission to the Intensive Care Unit of the Hospital of the University of Pennsylvania). Endothelial activation, inflammation and cell death were assessed in HPMVEC treated with serum either from patients with COVID-19 or from healthy individuals. Activation was monitored by measuring NOX2 activation (Rac1 translocation) and ROS production; inflammation (or appearance of a pro-inflammatory phenotype) was monitored by measuring the induction of moieties such as intercellular adhesion molecule (ICAM-1), P-selectin and the NLRP3 inflammasome; cell death was measured via SYTOX™ Green assays. RESULTS Endothelial activation (i.e., NOX2 activation and subsequent ROS production) and cell death were significantly higher in the COVID-19 model than in healthy samples. When HPMVEC were pre-treated with the novel peptide PIP-2, which blocks NOX2 activation (via inhibition of Ca2+-independent phospholipase A2, aiPLA2), significant abrogation of ROS was observed. Endothelial inflammation and cell death were also significantly blunted. CONCLUSIONS The endothelium is activated during COVID-19 via cytokine storm-driven NOX2-ROS activation, which causes a pro-inflammatory phenotype. The concept of endothelial NOX2-ROS production as a unifying pathophysiological axis in COVID-19 raises the possibility of using PIP-2 to maintain vascular health.
Collapse
Affiliation(s)
- Oindrila Paul
- Institute for Environmental Medicine and Department of Physiology, Philadelphia, Pennsylvania, United States of America
| | - Isha K. Akolia
- Institute for Environmental Medicine and Department of Physiology, Philadelphia, Pennsylvania, United States of America
| | - Jian Qin Tao
- Institute for Environmental Medicine and Department of Physiology, Philadelphia, Pennsylvania, United States of America
| | - Nikita Jain
- Institute for Environmental Medicine and Department of Physiology, Philadelphia, Pennsylvania, United States of America
| | - Natalia Louneva
- Peroxitech Inc., Philadelphia, Pennsylvania, United States of America
| | - Kathleen T. Montone
- Department of Pathology, Philadelphia, Pennsylvania, United States of America
| | - Aron B. Fisher
- Peroxitech Inc., Philadelphia, Pennsylvania, United States of America
| | - Chamith S. Rajapakse
- Department of Radiology, Philadelphia, Pennsylvania, United States of America
- Department of Orthopedic Surgery, Philadelphia, Pennsylvania, United States of America
| | - Christian Bermudez
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Shampa Chatterjee
- Institute for Environmental Medicine and Department of Physiology, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
12
|
Gao Q, Weng Z, Feng Y, Gong T, Zheng X, Zhang G, Gong L. KPNA2 suppresses porcine epidemic diarrhea virus replication by targeting and degrading virus envelope protein through selective autophagy. J Virol 2023; 97:e0011523. [PMID: 38038431 PMCID: PMC10734479 DOI: 10.1128/jvi.00115-23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/05/2023] [Indexed: 12/02/2023] Open
Abstract
IMPORTANCE Porcine epidemic diarrhea, characterized by vomiting, dehydration, and diarrhea, is an acute and highly contagious enteric disease caused by porcine epidemic diarrhea virus (PEDV) in neonatal piglets. This disease has caused large economic losses to the porcine industry worldwide. Thus, identifying the host factors involved in PEDV infection is important to develop novel strategies to control PEDV transmission. This study shows that PEDV infection upregulates karyopherin α 2 (KPNA2) expression in Vero and intestinal epithelial (IEC) cells. KPNA2 binds to and degrades the PEDV E protein via autophagy to suppress PEDV replication. These results suggest that KPNA2 plays an antiviral role against PEDV. Specifically, knockdown of endogenous KPNA2 enhances PEDV replication, whereas its overexpression inhibits PEDV replication. Our data provide novel KPNA2-mediated viral restriction mechanisms in which KPNA2 suppresses PEDV replication by targeting and degrading the viral E protein through autophagy. These mechanisms can be targeted in future studies to develop novel strategies to control PEDV infection.
Collapse
Affiliation(s)
- Qi Gao
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Vaccine Development, Guangzhou, China
| | - Zhijun Weng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Vaccine Development, Guangzhou, China
| | - Yongzhi Feng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
- Ministry of Agriculture and Rural Affairs, Key Laboratory of Animal Vaccine Development, Guangzhou, China
| | - Ting Gong
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Xiaoyu Zheng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
| | - Guihong Zhang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Lang Gong
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, China
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
13
|
Zhang H, Gao J, Tang Y, Jin T, Tao J. Inflammasomes cross-talk with lymphocytes to connect the innate and adaptive immune response. J Adv Res 2023; 54:181-193. [PMID: 36681114 DOI: 10.1016/j.jare.2023.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 10/15/2022] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Innate and adaptive immunity are two different parts of the immune system that have different characteristics and work together to provide immune protection. Inflammasomes are a major part of the innate immune system that are expressed widely in myeloid cells and are responsible for inflammatory responses. Recent studies have shown that inflammasomes are also expressed and activated in lymphocytes, especially in T and B cells, to regulate the adaptive immune response. Activation of inflammasomes is also under the control of lymphocytes. Therefore, we propose that inflammasomes act as a bridge and they provide crosstalk between the innate and adaptive immune systems to obtain a fine balance in immune responses. AIM OF REVIEW This review systematially summarizes the interaction between inflammasomes and lymphocytes and describes the crosstalk between the innate and adaptive immune systems induced by inflammasomes, with the aim of providing new directions and important areas for further research. KEY SCIENTIFIC CONCEPTS OF REVIEW When considering the novel function of inflammasomes in various lymphocytes, attention should be given to the activity of specific inflammasomes in studies of lymphocyte function. Moreover, research on the function of various inflammasomes in lymphocytes will help advance knowledge on the mechanisms and treatment of various diseases, including autoimmune diseases and tumors. In addition, when studying inflammatory responses, inflammasomes in both lymphocytes and myeloid cells need to be considered.
Collapse
Affiliation(s)
- Hongliang Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; College of Medicine and Health, Lishui University, No. 1 Xueyuan Road, Liandu District, Lishui 323000, China
| | - Jie Gao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yujie Tang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tengchuan Jin
- Laboratory of Structural Immunology, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jinhui Tao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
14
|
Leite JA, Menezes L, Martins E, Rodrigues TS, Tavares L, Ebering A, Schelmbauer C, Martelossi Cebinelli GC, Zinina V, Golden A, Soshnikova N, Zamboni DS, Cunha FQ, Huber M, Silva JS, Waisman A, Carlos D, Saraiva Câmara NO. AIM2 promotes T H17 cells differentiation by regulating RORγt transcription activity. iScience 2023; 26:108134. [PMID: 37867943 PMCID: PMC10585393 DOI: 10.1016/j.isci.2023.108134] [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: 06/07/2023] [Revised: 09/15/2023] [Accepted: 10/02/2023] [Indexed: 10/24/2023] Open
Abstract
AIM2 is an interferon-inducible HIN-200 protein family member and is well-documented for its roles in innate immune responses as a DNA sensor. Recent studies have highlighted AIM2's function on regulatory T cells (Treg) and follicular T cells (Tfh). However, its involvement in Th17 cell differentiation remains unclear. This study reveals that AIM2 promotes Th17 cell differentiation. AIM2 deficiency decreases IL-17A production and downregulates key Th17 associated proteins (RORγt, IL-1R1, IL-23R). AIM2 is located in the nucleus of Th17 cells, where it interacts with RORγt, enhancing its binding to the Il17a promoter. The absence of AIM2 hinders naive CD4 T cells from differentiating into functional Th17 cells and from inducing colitis in Rag1-/- mice. This study uncovers AIM2's role as a regulator of Th17 cell transcriptional programming, highlighting its potential as a therapeutic target for Th17 cell-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Jefferson Antônio Leite
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Luísa Menezes
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Eloisa Martins
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
- Division of Nephrology, School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Tamara Silva Rodrigues
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Lucas Tavares
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Anna Ebering
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Carsten Schelmbauer
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Guilherme C. Martelossi Cebinelli
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Pharmacology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Valeriya Zinina
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Artemiy Golden
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Natalia Soshnikova
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Dario S. Zamboni
- Department of Cellular and Molecular Biology and Pathogenic Bioagents, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Fernando Q. Cunha
- Department of Pharmacology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Magdalena Huber
- Institute of Systems Immunology, Center for Tumor and Immunology, University of Marburg, Marburg, Germany
| | - João Santana Silva
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Fiocruz-Bi-Institutional Translational Medicine Project, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Daniela Carlos
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Department of Biochemistry and Immunology, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
- Division of Nephrology, School of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
15
|
Accogli T, Hibos C, Vegran F. Canonical and non-canonical functions of NLRP3. J Adv Res 2023; 53:137-151. [PMID: 36610670 PMCID: PMC10658328 DOI: 10.1016/j.jare.2023.01.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/22/2022] [Accepted: 01/02/2023] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Since its discovery, NLRP3 is almost never separated from its major role in the protein complex it forms with ASC, NEK7 and Caspase-1, the inflammasome. This key component of the innate immune response mediates the secretion of proinflammatory cytokines IL-1β and IL-18 involved in immune response to microbial infection and cellular damage. However, NLRP3 has also other functions that do not involve the inflammasome assembly nor the innate immune response. These non-canonical functions have been poorly studied. Nevertheless, NLRP3 is associated with different kind of diseases probably through its inflammasome dependent function as through its inflammasome independent functions. AIM OF THE REVIEW The study and understanding of the canonical and non-canonical functions of NLRP3 can help to better understand its involvement in various pathologies. In parallel, the description of the mechanisms of action and regulation of its various functions, can allow the identification of new therapeutic strategies. KEY SCIENTIFIC CONCEPTS OF THE REVIEW NLRP3 functions have mainly been studied in the context of the inflammasome, in myeloid cells and in totally deficient transgenic mice. However, for several year, the work of different teams has proven that NLRP3 is also expressed in other cell types where it has functions that are independent of the inflammasome. If these studies suggest that NLRP3 could play different roles in the cytoplasm or the nucleus of the cells, the mechanisms underlying NLRP3 non-canonical functions remain unclear. This is why we propose in this review an inventory of the canonical and non-canonical functions of NLRP3 and their impact in different pathologies.
Collapse
Affiliation(s)
- Théo Accogli
- Faculté des Sciences de Santé- University of Burgundy, Dijon 21000, FRANCE; CAdIR Team - Centre de Recherche INSERM - UMR 1231, Dijon 21000, FRANCE
| | - Christophe Hibos
- Faculté des Sciences de Santé- University of Burgundy, Dijon 21000, FRANCE; CAdIR Team - Centre de Recherche INSERM - UMR 1231, Dijon 21000, FRANCE; Université de Bourgogne Franche-Comté, Dijon 21000, FRANCE
| | - Frédérique Vegran
- Faculté des Sciences de Santé- University of Burgundy, Dijon 21000, FRANCE; CAdIR Team - Centre de Recherche INSERM - UMR 1231, Dijon 21000, FRANCE; Department of Biology and Pathology of Tumors - Centre anticancéreux GF Leclerc, Dijon 21000, FRANCE.
| |
Collapse
|
16
|
He X, You R, Shi Y, Zeng Z, Tang B, Yu J, Xiao Y, Xiao R. Pyroptosis: the potential eye of the storm in adult-onset Still's disease. Inflammopharmacology 2023; 31:2269-2282. [PMID: 37429997 DOI: 10.1007/s10787-023-01275-0] [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: 04/15/2023] [Accepted: 06/02/2023] [Indexed: 07/12/2023]
Abstract
Pyroptosis, a form of programmed cell death with a high pro-inflammatory effect, causes cell lysis and leads to the secretion of countless interleukin-1β (IL-1β) and IL-18 cytokines, resulting in a subsequent extreme inflammatory response through the caspase-1-dependent pathway or caspase-1-independent pathway. Adult-onset Still's disease (AOSD) is a systemic inflammatory disease with extensive disease manifestations and severe complications such as macrophage activation syndrome, which is characterized by high-grade inflammation and cytokine storms regulated by IL-1β and IL-18. To date, the pathogenesis of AOSD is unclear, and the available therapy is unsatisfactory. As such, AOSD is still a challenging disease. In addition, the high inflammatory states and the increased expression of multiple pyroptosis markers in AOSD indicate that pyroptosis plays an important role in the pathogenesis of AOSD. Accordingly, this review summarizes the molecular mechanisms of pyroptosis and describes the potential role of pyroptosis in AOSD, the therapeutic practicalities of pyroptosis target drugs in AOSD, and the therapeutic blueprint of other pyroptosis target drugs.
Collapse
Affiliation(s)
- Xinglan He
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ruixuan You
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yaqian Shi
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhuotong Zeng
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Bingsi Tang
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiangfan Yu
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yangfan Xiao
- Clinical Nursing Teaching and Research Section, The Second Xiangya Hospital, Central South University, Changsha, China.
- Department of Anesthesiology, The Second Xiangya Hospital, Central South University, Changsha, China.
| | - Rong Xiao
- Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Medical Epigenetics, Department of Dermatology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
17
|
Zhang M, Lan H, Peng S, Zhou W, Wang X, Jiang M, Hong J, Zhang Q. MiR-223-3p attenuates radiation-induced inflammatory response and inhibits the activation of NLRP3 inflammasome in macrophages. Int Immunopharmacol 2023; 122:110616. [PMID: 37459784 DOI: 10.1016/j.intimp.2023.110616] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 06/29/2023] [Accepted: 07/04/2023] [Indexed: 08/25/2023]
Abstract
Macrophage pyroptosis plays an important role in the development of radiation-induced cell and tissue damage, leading to acute lung injury. However, the underlying mechanisms of NOD-like receptor thermal protein domain-associated protein 3 (NLRP3)-mediated macrophage pyroptosis and the regulatory factors involved in radiation-induced pyroptosis are unclear. In this study, the expression of the NLRP3 inflammasome and pyroptosis-associated factors in murine macrophage cell lines was investigated after ionizing radiation. High-throughput RNA sequencing was performed to identify and characterize miRNAs and mRNA transcripts associated with NLRP3-mediated cell death. Our results demonstrated that cleaved-caspase-1 (p10) and N-terminal domain of gasdermin-D (GSDMD-N) were upregulated, and the number of NLRP3 inflammasomes and pyroptotic cells increased in murine macrophage cell lines after irradiation (8 Gy). Comparativeprofiling of 300miRNAs revealed that 41 miRNAsexhibited significantly different expression after 8 Gy of irradiation. Granulocyte-specific microRNA-223-3p (miR-223-3p) is a negative regulator of NLRP3. In vitro experiments revealed that the expression of miR-223-3p was significantly altered by irradiation. Moreover, miR-223-3p decreased the expression of NLRP3 and proinflammatory factors, resulting in reduced pyroptosis in irradiated murine macrophages. Subsequently, in vivo experiments revealed the efficacy of miR-223-3p supplementation in ameliorating alveolar macrophage (AM) pyroptosis, attenuating the infiltration of inflammatory monocytes, and significantly alleviating the severity of acute radiation-induced lung injury (ARILI). Our findings suggest that the miR-223-3p/NLRP3/caspase-1 axis is involved in radiation-induced AM pyroptosis and ARILI.
Collapse
Affiliation(s)
- Mingwei Zhang
- Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China; Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hailin Lan
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shaoli Peng
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Weitong Zhou
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xuezhen Wang
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Meina Jiang
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jinsheng Hong
- Department of Radiotherapy, Cancer Center, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Department of Radiotherapy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, China; Key Laboratory of Radiation Biology of Fujian Higher Education Institutions, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| | - Qiuyu Zhang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
18
|
Zhu J, Chen H, Cui J, Zhang X, Liu G. Oroxylin A inhibited autoimmune hepatitis-induced liver injury and shifted Treg/Th17 balance to Treg differentiation. Exp Anim 2023; 72:367-378. [PMID: 36927981 PMCID: PMC10435359 DOI: 10.1538/expanim.22-0171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
Autoimmune hepatitis (AIH) is a kind of autoimmune disease mediated by T cells, and its incidence is gradually increasing in the world. Oroxylin A (OA) is one of the major bioactive flavonoids that has been reported to inhibit inflammatory. Here, an AIH model of mouse was induced by Concanavalin A (Con A). It found that serum aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels were decreased in mice with the treatment of OA. Hematoxylin-eosin staining showed that the liver injury was attenuated by OA, and TUNEL staining indicated that the cells apoptosis of liver was weakened in mice with OA treatment. ELISA analysis of cytokines and chemokines suggested that OA reduced the expression of IL-6, IL-17A, chemokine ligand 2 (CCL2), C-X-C motif chemokine ligand 1 (CXCL1) and CXCL10, but promoted the expression of IL-10 and TGF-β in mice. The mRNA levels of Il-17a in liver and spleen tissues were also significantly decreased, on the contrary, the mRNA levels of Il-10 in liver and spleen tissues were increased. The proportion of Treg/Th17 detected by flow cytometry revealed that OA promoted the differentiation of Treg and inhibited the differentiation of Th17 both in the liver and spleen. The results of this study demonstrated the inhibitory effects of OA on AIH-induced liver injury and the inflammatory response of AIH, and revealed that OA affected the balance of Treg/Th17 and shifted the balance toward Treg differentiation. It provided new potential drugs for the prevention of AIH.
Collapse
Affiliation(s)
- Jinxia Zhu
- The First Clinical Medical College, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, Henan, 450046, P.R. China
| | - Hongxiu Chen
- The First Clinical Medical College, Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengzhou, Henan, 450046, P.R. China
| | - Jianjiao Cui
- Spleen, Stomach and Hepatobiliary Department, The First Affiliated Hospital of Henan University of Chinese Medicine, No. 19, Renmin Road, Zhengzhou, Henan, 450003, P.R. China
| | - Xiaorui Zhang
- Spleen, Stomach and Hepatobiliary Department, The First Affiliated Hospital of Henan University of Chinese Medicine, No. 19, Renmin Road, Zhengzhou, Henan, 450003, P.R. China
| | - Guangwei Liu
- Spleen, Stomach and Hepatobiliary Department, The First Affiliated Hospital of Henan University of Chinese Medicine, No. 19, Renmin Road, Zhengzhou, Henan, 450003, P.R. China
| |
Collapse
|
19
|
Niu J, Meng G. Roles and Mechanisms of NLRP3 in Influenza Viral Infection. Viruses 2023; 15:1339. [PMID: 37376638 DOI: 10.3390/v15061339] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/05/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Pathogenic viral infection represents a major challenge to human health. Due to the vast mucosal surface of respiratory tract exposed to the environment, host defense against influenza viruses has perpetually been a considerable challenge. Inflammasomes serve as vital components of the host innate immune system and play a crucial role in responding to viral infections. To cope with influenza viral infection, the host employs inflammasomes and symbiotic microbiota to confer effective protection at the mucosal surface in the lungs. This review article aims to summarize the current findings on the function of NACHT, LRR and PYD domains-containing protein 3 (NLRP3) in host response to influenza viral infection involving various mechanisms including the gut-lung crosstalk.
Collapse
Affiliation(s)
- Junling Niu
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, 320 Yueyang Road, Life Science Research Building B-205, Shanghai 200031, China
| | - Guangxun Meng
- The Center for Microbes, Development and Health, CAS Key Laboratory of Molecular Virology & Immunology, University of Chinese Academy of Sciences, 320 Yueyang Road, Life Science Research Building B-205, Shanghai 200031, China
| |
Collapse
|
20
|
Attia MS, Ewida HA, Abdel Hafez MA, El-Maraghy SA, El-Sawalhi MM. Altered Lnc-EGFR, SNHG1, and LincRNA-Cox2 Profiles in Patients with Relapsing-Remitting Multiple Sclerosis: Impact on Disease Activity and Progression. Diagnostics (Basel) 2023; 13:diagnostics13081448. [PMID: 37189549 DOI: 10.3390/diagnostics13081448] [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: 03/03/2023] [Revised: 04/12/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Relapsing-remitting multiple sclerosis (RRMS) is the most prevalent MS subtype. Ample evidence has indicated that long noncoding RNAs (lncRNAs) are crucial players in autoimmune and inflammatory disorders. This study investigated the expression of lnc-EGFR, SNHG1, and lincRNA-Cox2 in RRMS patients during active relapses and in remission. Additionally, the expression of FOXP3, a master transcription factor for regulatory T cells, and NLRP3-inflammasome-related genes were determined. Relationships between these parameters and MS activity and annualized relapse rate (ARR) were also evaluated. The study included 100 Egyptian participants: 70 RRMS patients (35 during relapse and 35 in remission) and 30 healthy controls. RRMS patients showed significant downregulation of lnc-EGFR and FOXP3 and dramatic upregulation of SNHG1, lincRNA-Cox2, NLRP3, ASC, and caspase-1 compared to controls. Lower serum TGF-β1 and elevated IL-1β levels were observed in RRMS patients. Notably, patients during relapses displayed more significant alterations than those in remission. Lnc-EGFR was positively correlated with FOXP3 and TGF-β1 and negatively correlated with ARR, SNHG1, lincRNA-Cox2, and NLRP3 inflammasome components. Meanwhile, SNHG1 and lincRNA-Cox2 were positively correlated with ARR, NLRP3, ASC, caspase-1, and IL-1β. Excellent diagnostic performance for lnc-EGFR, FOXP3, and TGF-β1 was demonstrated, while all biomarkers exhibited strong prognostic potential for predicting relapses. Finally, the differential expression of lnc-EGFR, SNHG1, and lincRNA-Cox2 in RRMS patients, especially during relapses, suggests their involvement in RRMS pathogenesis and activity. Correlation between their expression and ARR implies relationships to disease progression. Our findings also highlight their promising roles as biomarkers for RRMS.
Collapse
Affiliation(s)
- Mohamed S Attia
- Pharmacology, Toxicology and Biochemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), Cairo 11835, Egypt
| | - Heba A Ewida
- Pharmacology, Toxicology and Biochemistry Department, Faculty of Pharmacy, Future University in Egypt (FUE), Cairo 11835, Egypt
| | | | - Shohda A El-Maraghy
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Maha M El-Sawalhi
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
21
|
Leng S, Xu W, Wu L, Liu L, Du J, Yang F, Huang D, Zhang L. NLRP3 Disturbs Treg/Th17 Cell Balance to Aggravate Apical Periodontitis. J Dent Res 2023; 102:656-666. [PMID: 36883625 DOI: 10.1177/00220345231151692] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
Apical periodontitis is an inflammatory condition that is considered an immunological reaction of the periapical tissue to invading bacteria and their pathogenic components. Recent research has revealed that NLR family pyrin domain containing 3 (NLRP3) is crucial to the pathogenesis of apical periodontitis and serves as a link between innate and adaptive immunity. The balance between regulatory T-cell (Treg) and T helper cell 17 (Th17 cell) determines the direction of the inflammatory response. Therefore, this study aimed to investigate whether NLRP3 exacerbated periapical inflammation by disturbing Treg/Th17 balance and the underlying regulatory mechanisms. In the present study, NLRP3 was raised in apical periodontitis tissues as opposed to healthy pulp tissues. Low NLRP3 expression in dendritic cells (DCs) increased transforming growth factor β secretion while decreasing interleukin (IL)-1β and IL-6 production. The Treg ratio and IL-10 secretion rose when CD4+ T cells were cocultured with DCs primed with IL-1β neutralizing antibody (anti-IL-1β) and specific small interfering RNA (siRNA) targeting NLRP3 (siRNA NLRP3), but the proportion of Th17 cells and IL-17 release dropped. Furthermore, siRNA NLRP3-mediated suppression of NLRP3 expression aided Treg differentiation and elevated Foxp3 expression as well as IL-10 production in CD4+ T cells. Inhibition of NLRP3 activity by MCC950 boosted the percentage of Tregs while decreasing the ratio of Th17 cells, leading to reduced periapical inflammation and bone resorption. Nigericin administration, however, exacerbated periapical inflammation and bone destruction with an unbalanced Treg/Th17 response. These findings demonstrate that NLRP3 is a pivotal regulator by regulating the release of inflammatory cytokines from DCs or directly suppressing Foxp3 expression to disturb Treg/Th17 balance, thus exacerbating apical periodontitis.
Collapse
Affiliation(s)
- S Leng
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - W Xu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key laboratory of Oral Biomedical Research of Zhejiang Province Cancer Center of Zhejiang University, Hangzhou, China
| | - L Wu
- Department of Geriatric Stomatology, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Liu
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J Du
- Department of Health Care (Department of General Dentistry II), School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - F Yang
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - D Huang
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Zhang
- Department of Operative Dentistry and Endodontics, State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Disease, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Zhang S, Gan X, Gao J, Duan J, Gu A, Chen C. CoQ10 alleviates hepatic ischemia reperfusion injury via inhibiting NLRP3 activity and promoting Tregs infiltration. Mol Immunol 2023; 155:7-16. [PMID: 36640727 DOI: 10.1016/j.molimm.2023.01.005] [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: 10/12/2022] [Revised: 12/05/2022] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
Hepatic ischemia-reperfusion injury (IRI) has been concerned as a main complication of liver surgery and transplantation. Previous studies show that reactive oxygen species (ROS) associated inflammation response and contribute to the liver damage during IRI. Coenzyme Q10 (CoQ10) has shown many beneficial effects on abrogating ROS production and ameliorating liver injury. This study found lower CoQ10 level in the process of liver IRI in a mouse model of hepatic IRI. Meanwhile, our results showed that CoQ10 administration significantly attenuate hepatic IRI proved by HE staining, serum ALT/AST. The NOD-like receptor protein 3 (NLRP3) inflammasome is activated by ROS which triggers the activation of inflammatory caspases. In this study, NLRP3 was significantly suppressed by CoQ10 while Foxp3 exhibited increased expression in liver. Furthermore, Kupffer cells (KCs) pretreated with CoQ10 under the condition of hypoxia and reoxygenation contributed to improved CD4+CD25+Foxp3+ regulatory T cells (Tregs) ratio in co-culture system. Furthermore, NLRP3 inflammasome activator treatment in vivo resulted in higher expression of caspase-1 and NLRP3 and reduction of Tregs in liver, which reversed the protection of CoQ10 in the liver injury. Taken together, our study discovered that CoQ10 can suppress NLRP3 activity in KCs and improves Foxp3+ Tregs differentiation depending on M2 macrophage polarization of KCs to ameliorate hepatic IRI.
Collapse
Affiliation(s)
- Shaopeng Zhang
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing, Nanjing university of Chinese Medicine, Nanjing, China; Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaojie Gan
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Ji Gao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Duan
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing, Nanjing university of Chinese Medicine, Nanjing, China
| | - Aidong Gu
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing, Nanjing university of Chinese Medicine, Nanjing, China.
| | - Changhao Chen
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing, Nanjing university of Chinese Medicine, Nanjing, China.
| |
Collapse
|
23
|
Hsu ML, Zouali M. Inflammasome is a central player in B cell development and homing. Life Sci Alliance 2023; 6:6/2/e202201700. [PMID: 36450446 PMCID: PMC9713303 DOI: 10.26508/lsa.202201700] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022] Open
Abstract
Whereas the role of the nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing protein (NLRP) 3 pathway in innate immunity has been extensively studied, little attention has been paid to its contribution to adaptive immunity. Studies in animal models and human subjects have shown the contribution of NLRP3 to the T cell compartment, and its role in B lymphocyte functions has been proposed. Here, we report that ablation of nlrp3 in mice led to altered B cell development in the bone marrow, and distorted expression of B cell subsets that play innate-like functions, that is, marginal zone B cells in the spleen and B-1a cells in the peritoneal cavity. Mechanistically, in the absence of NLRP3 expression, the transcription factor IRF4, previously found to interact with NLRP3 in the nucleus of lymphocytes, was up-regulated. NLRP3 ablation reduced the expression of the chemokine receptors CXCR4 and CCR7 in an IRF4-dependent manner, indicating that the presence of NLRP3 is critical for optimal expression of chemokine receptors on B cells. We conclude that activation of the NLRP3 inflammasome plays a role in B cell development, homing, and retention in lymphoid organs.
Collapse
Affiliation(s)
- Man Lun Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Moncef Zouali
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
24
|
Cui B, Chen XJ, Sun J, Li SP, Zhou GP, Sun LY, Wei L, Zhu ZJ. Dendritic cells originating exosomal miR-193b-3p induces regulatory T cells to alleviate liver transplant rejection. Int Immunopharmacol 2023; 114:109541. [PMID: 36700764 DOI: 10.1016/j.intimp.2022.109541] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Exosomes exert considerable influence in mediating regulatory T (Treg) cells differentiation, which attach great importance to attenuating acute cellular rejection after liver transplantation (LT). And, miRNAs are known to play essential roles in cell-cell communication delivered by exosomes. However, the function of exosomal miRNAs in regulating Treg cells after LT remains unknown. Here, we performed an expression profiling analysis of exosome-miRNAs from human plasma after LT and investigated their immunoregulatory effects on Treg cells. METHODS Fifty-eight LT patients and nine donors were included in this report. miRNA profiles in plasma exosomes were analyzed using next-generation sequencing. Flow cytometry, HE and multiplex immunofluorescent staining were used to identify Treg cells in the liver and peripheral blood. A lentiviral vector system was used to overexpress miR-193b-3p in dendritic cells (DCs), and exosomes isolated from these transfected cells were co-cultured with spleen lymphocytesin vitro. A quantitative Real-time PCR and enzyme-linked immunosorbent assay were used to detect the expression of cytokines. RESULTS Treg cell infiltration was increased in the liver along with Th17 and CD8+ T cell, and it was down-regulated in peripheral blood in the acute rejection group. High-throughput sequencing revealed that miR-193b-3p was markedly up-regulated in plasma exosomes of non-rejection LT patients. The NLRP3 inflammasome was screened as a target for miR-193b-3p based on target prediction and functional enrichment analyses. Exosomal miR-193b-3p derived from DCs increased Treg cells as demonstrated in vitro. miR-193b-3p overexpression down-regulated NLRP3 as well as the inflammatory cytokines IL-1β and IL-17A while increasing levels of the cytokines IL-10 and TGF-β. CONCLUSION DC derived exosomal miR-193b-3p promoted Treg cells by inhibiting NLRP3 expression. These findings not only provide a new perspective on the mechanisms, but also hold great promise for the treatment or prevention of liver allograft rejection.
Collapse
Affiliation(s)
- Bin Cui
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing 101100, China; Department of Neurosurgery, Aviation General Hospital, Beijing 100012, China
| | - Xiao-Jie Chen
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing 101100, China; Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing 101100, China; National Clinical Research Center for Digestive Diseases, Beijing 101100, China
| | - Jie Sun
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing 101100, China; Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing 101100, China; National Clinical Research Center for Digestive Diseases, Beijing 101100, China
| | - Shi-Peng Li
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing 101100, China; Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing 101100, China; National Clinical Research Center for Digestive Diseases, Beijing 101100, China
| | - Guang-Peng Zhou
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing 101100, China; Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing 101100, China; National Clinical Research Center for Digestive Diseases, Beijing 101100, China
| | - Li-Ying Sun
- Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing 101100, China; National Clinical Research Center for Digestive Diseases, Beijing 101100, China; Department of Critical Liver Diseases, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing 101100, China
| | - Lin Wei
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing 101100, China; Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing 101100, China; National Clinical Research Center for Digestive Diseases, Beijing 101100, China
| | - Zhi-Jun Zhu
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing 101100, China; Clinical Center for Pediatric Liver Transplantation, Capital Medical University, Beijing 101100, China; National Clinical Research Center for Digestive Diseases, Beijing 101100, China.
| |
Collapse
|
25
|
Lee JH, Kim HS, Jang SW, Lee GR. Histone deacetylase 6 plays an important role in TGF-β-induced murine Treg cell differentiation by regulating cell proliferation. Sci Rep 2022; 12:22550. [PMID: 36581745 PMCID: PMC9800578 DOI: 10.1038/s41598-022-27230-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/28/2022] [Indexed: 12/30/2022] Open
Abstract
Regulatory T (Treg) cells maintain immune homeostasis by preventing abnormal or excessive immune responses. Histone deacetylase 6 (HDAC6) regulates expression of Foxp3, and thus, Treg cell differentiation; however, its role in Treg cell differentiation is unclear and somewhat controversial. Here, we investigated the role of HDAC6 in TGF-β-induced murine Treg cells. HDAC6 expression was higher in Treg cells than in other T helper cell subsets. Pharmacological inhibitors of HDAC6 selectively inhibited Treg cell differentiation and suppressive function. A specific HDAC6 inhibitor induced changes in global gene expression by Treg cells. Of these changes, genes related to cell division were prominently affected. In summary, HDAC6 plays an important role in TGF-β-induced murine Treg cell differentiation by regulating cell proliferation.
Collapse
Affiliation(s)
- Ji Hyeon Lee
- grid.263736.50000 0001 0286 5954Department of Life Science, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul, 04107 Korea
| | - Hyeong Su Kim
- grid.263736.50000 0001 0286 5954Department of Life Science, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul, 04107 Korea
| | - Sung Woong Jang
- grid.263736.50000 0001 0286 5954Department of Life Science, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul, 04107 Korea
| | - Gap Ryol Lee
- grid.263736.50000 0001 0286 5954Department of Life Science, Sogang University, 35 Baekbeom-Ro, Mapo-Gu, Seoul, 04107 Korea
| |
Collapse
|
26
|
Liu SY, Huang CC, Yang YY, Huang SF, Lee TY, Li TH, Hou MC, Lin HC. Obeticholic acid treatment ameliorates the cardiac dysfunction in NASH mice. PLoS One 2022; 17:e0276717. [PMID: 36490253 PMCID: PMC9733885 DOI: 10.1371/journal.pone.0276717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/11/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Suppression of cardiac iinflammasome, which can be inhibited by Farnesoid X receptor (FXR) agonist, can ameliorate cardiac inflammation and fibrosis. Increased cardiac inflammasome decrease the abundance of regulatory T (Treg) cells and exacerbate cardiac dysfunction. Interaction between cardiomyocytes and Treg cells is involved in the development of nonalcoholic steatohepatitis (NASH)-related cardiac dysfunction. AIMS This study evaluates whether the FXR agonist obeticholic acid (OCA) treatment improves NASH-associated cardiac dysfunction. METHODS The in vivo and in vitro mechanisms and effects of two weeks of OCA treatment on inflammasome and Treg dysregulation-related cardiac dysfunction in NASH mice (NASH-OCA) at systemic, tissue and cellular levels were investigated. RESULTS The OCA treatment suppressed the serum and cardiac inflammasome levels, reduced the cardiac infiltrated CD3+ T cells, increased the cardiac Treg-represented anti-inflammatory cytokines (IL-10/IL-10R) and improved cardiac inflammation, fibrosis and function [decreased left ventricle (LV) mass and increased fractional shortening (FS)] in NASH-OCA mice. The percentages of OCA-decreased cardiac fibrosis and OCA-increased FS were positively correlated with the percentage of OCA-increased levels of cardiac FXR and IL-10/IL-10R. In the Treg cells from NASH-OCA mice spleen, in comparison with the Treg cells of the NASH group, higher intracellular FXR but lower inflammasome levels, and more proliferative/active and less apoptotic cells were observed. Incubation of H9c2 cardiomyoblasts with Treg-NASHcm [supernatant of Treg from NASH mice as condition medium (cm)], increased inflammasome levels, decreased the proliferative/active cells, suppressed the intracellular FXR, and downregulated differentiation/contraction marker. The Treg-NASHcm-induced hypocontractility of H9c2 can be attenuated by co-incubation with OCA, and the OCA-related effects were abolished by siIL-10R pretreatment. CONCLUSIONS Chronic FXR activation with OCA is a potential strategy for activating IL-10/IL-10R signalling, reversing cardiac regulatory T cell dysfunction, and improving inflammasome-mediated NASH-related cardiac dysfunction.
Collapse
Affiliation(s)
- Szu-Yu Liu
- Department of Medical Education, Clinical Innovation Center, Medical Innovation and Research Office, Taipei Veterans General Hospital, Taipei, Taiwan,Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Chia-Chang Huang
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan,Faculty of Medicine, Institute of Clinical Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Ying Yang
- Department of Medical Education, Clinical Innovation Center, Medical Innovation and Research Office, Taipei Veterans General Hospital, Taipei, Taiwan,Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan,* E-mail: (Y-YY); (H-CL)
| | - Shiang-Fen Huang
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan,Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzung-Yan Lee
- Graduate Institute of Traditional Chinese Medicine, Chang Guang Memorial Hospital, Linkou, Taiwan
| | - Tzu-Hao Li
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan,Faculty of Medicine, Institute of Clinical Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan,Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Shin Kong Wu Ho-Su Memorial Foundation, Taipei, Taiwan
| | - Ming-Chih Hou
- Faculty of Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan,Taipei Veterans General Hospital, Taipei, Taiwan
| | - Han-Chieh Lin
- Taipei Veterans General Hospital, Taipei, Taiwan,* E-mail: (Y-YY); (H-CL)
| |
Collapse
|
27
|
Franchon Marques Tejada N, Ziroldo Lopes JV, Duarte Gonçalves LE, Mamede Costa Andrade da Conceição I, Franco GR, Ghirotto B, Câmara NOS. AIM2 as a putative target in acute kidney graft rejection. Front Immunol 2022; 13:839359. [PMID: 36248890 PMCID: PMC9561248 DOI: 10.3389/fimmu.2022.839359] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Acute rejection (AR) is a process triggered via the recognition of grafted organ-derived antigens by the immune system, which could present as a life-threatening condition. In the context of a kidney transplant, despite improvement with immunosuppressive therapies, AR maintains a significant incidence of 10%, and currently available drugs generally act in similar and canonical pathways of lymphocyte activation. This prompted the research for different approaches to identify potential novel targets that could improve therapeutic interventions. Here, we conducted a transcriptome analysis comparing groups of acute rejection (including T cell-mediated rejection and antibody-mediated rejection) to stable grafts that included differentially expressed genes, transcription factor and kinase enrichment, and Gene Set Enrichment Analysis. These analyses revealed inflammasome enhancement in rejected grafts and AIM2 as a potential component linked to acute rejection, presenting a positive correlation to T-cell activation and a negative correlation to oxidative phosphorylation metabolism. Also, the AIM2 expression showed a global accuracy in discerning acute rejection grafts (area under the curve (AUC) = 0.755 and 0.894, p < 0.0001), and meta-analysis comprising different studies indicated a considerable enhancement of AIM2 in rejection (standardized mean difference (SMD) = 1.45, [CI 95%, 1.18 to 1.71]), especially for T cell-mediated rejection (TCMR) (SMD = 2.01, [CI 95%, 1.58 to 2.45]). These findings could guide future studies of AIM2 as either an adjuvant target for immunosuppression or a potential biomarker for acute rejection and graft survival.
Collapse
Affiliation(s)
- Nathália Franchon Marques Tejada
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - João Vitor Ziroldo Lopes
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Luis Eduardo Duarte Gonçalves
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Izabela Mamede Costa Andrade da Conceição
- Laboratory of Biochemical Genetics, Department of Biochemistry and Immunology, Institute of Biomedical Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Glória Regina Franco
- Laboratory of Biochemical Genetics, Department of Biochemistry and Immunology, Institute of Biomedical Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Bruno Ghirotto
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Câmara
- Laboratory of Transplantation Immunobiology, Institute of Biomedical Sciences, Department of Immunology, University of São Paulo, São Paulo, Brazil
- Laboratory of Biochemical Genetics, Department of Biochemistry and Immunology, Institute of Biomedical Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Niels Olsen Saraiva Câmara, ;
| |
Collapse
|
28
|
Rivas-Arancibia S, Hernández-Orozco E, Rodríguez-Martínez E, Valdés-Fuentes M, Cornejo-Trejo V, Pérez-Pacheco N, Dorado-Martínez C, Zequeida-Carmona D, Espinosa-Caleti I. Ozone Pollution, Oxidative Stress, Regulatory T Cells and Antioxidants. Antioxidants (Basel) 2022; 11:antiox11081553. [PMID: 36009272 PMCID: PMC9405302 DOI: 10.3390/antiox11081553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/31/2022] [Accepted: 08/05/2022] [Indexed: 12/06/2022] Open
Abstract
Ozone pollution, is a serious health problem worldwide. Repeated exposure to low ozone doses causes a loss of regulation of the oxidation–reduction systems, and also induces a chronic state of oxidative stress. This fact is of special importance for the regulation of different systems including the immune system and the inflammatory response. In addition, the oxidation–reduction balance modulates the homeostasis of these and other complex systems such as metabolism, survival capacity, cell renewal, and brain repair, etc. Likewise, it has been widely demonstrated that in chronic degenerative diseases, an alteration in the oxide-reduction balance is present, and this alteration causes a chronic loss in the regulation of the immune response and the inflammatory process. This is because reactive oxygen species disrupt different signaling pathways. Such pathways are related to the role of regulatory T cells (Treg) in inflammation. This causes an increase in chronic deterioration in the degenerative disease over time. The objective of this review was to study the relationship between environmental ozone pollution, the chronic state of oxidative stress and its effect on Treg cells, which causes the loss of regulation in the inflammatory response as well as the role played by antioxidant systems in various pathologies.
Collapse
|
29
|
Palma G, Sorice GP, Genchi VA, Giordano F, Caccioppoli C, D’Oria R, Marrano N, Biondi G, Giorgino F, Perrini S. Adipose Tissue Inflammation and Pulmonary Dysfunction in Obesity. Int J Mol Sci 2022; 23:ijms23137349. [PMID: 35806353 PMCID: PMC9267094 DOI: 10.3390/ijms23137349] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 02/04/2023] Open
Abstract
Obesity is a chronic disease caused by an excess of adipose tissue that may impair health by altering the functionality of various organs, including the lungs. Excessive deposition of fat in the abdominal area can lead to abnormal positioning of the diaphragm and consequent reduction in lung volume, leading to a heightened demand for ventilation and increased exposure to respiratory diseases, such as chronic obstructive pulmonary disease, asthma, and obstructive sleep apnoea. In addition to mechanical ventilatory constraints, excess fat and ectopic deposition in visceral depots can lead to adipose tissue dysfunction, which promotes metabolic disorders. An altered adipokine-secretion profile from dysfunctional adipose tissue in morbid obesity fosters systemic, low-grade inflammation, impairing pulmonary immune response and promoting airway hyperresponsiveness. A potential target of these adipokines could be the NLRP3 inflammasome, a critical component of the innate immune system, the harmful pro-inflammatory effect of which affects both adipose and lung tissue in obesity. In this review, we will investigate the crosstalk between adipose tissue and the lung in obesity, highlighting the main inflammatory mediators and novel therapeutic targets in preventing pulmonary dysfunction.
Collapse
|
30
|
Liang X, Sun T, Cui Y, Zhou S, Liang X. Bone Marrow Mesenchymal Stem Cells (BMSCs)-Triggered Up-Regulation of miR-1297/NLR Family Pyrin Domain Containing 3 (NLRP3) Facilitates the Aggressive Proliferation of Lung Cancer Cells via Inducing Inflammatory Factor Release. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
miR-1297 derived from BMSC-originated exosomes participates in modulating multiple malignancies. Our study aims to clarify the effect of miR-1297 derived from BMSC-originated exosomes on the oxidative stress and inflammatory damage of lung cancer cells. miR-1297 and NLRP3 level was
measured in lung cancer tissues and para-cancerous tissues, as well as in lung cancer cell lines and pulmonary epithelial cells. After miR-1297-mimics transfection or BMSC co-cultivation, cell viability was assessed by MTT and cytokines were evaluated by ELISA along with analysis of SOD activity
and cell apoptosis. miR-1297 and NLRP3 were significantly elevated in lung cancer tissues and cell lines. Overexpression of miR-1297 enhanced oxidative stress and inflammatory response, along with increased cell viability and decreased apoptosis. Additionally, co-culture with BMSC protect
the viability of lung cancer cells by facilitating miR-1297/NLRP3. In conclusion, a significant elevation of miR-1297 is found in lung cancer tissues and cells. Its overexpression induced the release of inflammatory factors, thereby protecting the proliferating activity of lung cancer cells
and restraining apoptosis, indicating that miR-1297 may serve a promising target for early diagnosis of lung cancers.
Collapse
Affiliation(s)
- Xiujun Liang
- Department of Basic Medical School, Chengde Medical College, Chengde, Hebei, 067000, China
| | - Tongyou Sun
- Department of Chemoradiotherapy, Chengde Central Hospital, Chengde, Hebei, 067000, China
| | - Yujie Cui
- Department of Oncology Department, Hebei Provincial People’s Hospital, Shijiazhuang, Hebei, 050057, China
| | - Shuo Zhou
- Department of Graduate School, Chengde Medical College, Chengde, Hebei, 067000, China
| | - Xiujun Liang
- Department of Basic Medical School, Chengde Medical College, Chengde, Hebei, 067000, China
| |
Collapse
|
31
|
Challagundla N, Saha B, Agrawal-Rajput R. Insights into inflammasome regulation: cellular, molecular, and pathogenic control of inflammasome activation. Immunol Res 2022; 70:578-606. [PMID: 35610534 DOI: 10.1007/s12026-022-09286-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Abstract
Maintenance of immune homeostasis is an intricate process wherein inflammasomes play a pivotal role by contributing to innate and adaptive immune responses. Inflammasomes are ensembles of adaptor proteins that can trigger a signal following innate sensing of pathogens or non-pathogens eventuating in the inductions of IL-1β and IL-18. These inflammatory cytokines substantially influence the antigen-presenting cell's costimulatory functions and T helper cell differentiation, contributing to adaptive immunity. As acute and chronic disease conditions may accompany parallel tissue damage, we analyze the critical role of extracellular factors such as cytokines, amyloids, cholesterol crystals, etc., intracellular metabolites, and signaling molecules regulating inflammasome activation/inhibition. We develop an operative framework for inflammasome function and regulation by host cell factors and pathogens. While inflammasomes influence the innate and adaptive immune components' interplay modulating the anti-pathogen adaptive immune response, pathogens may target inflammasome inhibition as a survival strategy. As trapped between health and diseases, inflammasomes serve as promising therapeutic targets and their modus operandi serves as a scientific rationale for devising better therapeutic strategies.
Collapse
Affiliation(s)
- Naveen Challagundla
- Immunology lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382007, India
| | - Bhaskar Saha
- National Centre for Cell Science, Lab-5, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Reena Agrawal-Rajput
- Immunology lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382007, India.
| |
Collapse
|
32
|
Molnár K, Nógrádi B, Kristóf R, Mészáros Á, Pajer K, Siklós L, Nógrádi A, Wilhelm I, Krizbai IA. Motoneuronal inflammasome activation triggers excessive neuroinflammation and impedes regeneration after sciatic nerve injury. J Neuroinflammation 2022; 19:68. [PMID: 35305649 PMCID: PMC8934511 DOI: 10.1186/s12974-022-02427-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 03/01/2022] [Indexed: 12/12/2022] Open
Abstract
Background Peripheral nerve injuries are accompanied by inflammatory reactions, over-activation of which may hinder recovery. Among pro-inflammatory pathways, inflammasomes are one of the most potent, leading to release of active IL-1β. Our aim was to understand how inflammasomes participate in central inflammatory reactions accompanying peripheral nerve injury. Methods After axotomy of the sciatic nerve, priming and activation of the NLRP3 inflammasome was examined in cells of the spinal cord. Regeneration of the nerve was evaluated after coaptation using sciatic functional index measurements and retrograde tracing. Results In the first 3 days after the injury, elements of the NLRP3 inflammasome were markedly upregulated in the L4–L5 segments of the spinal cord, followed by assembly of the inflammasome and secretion of active IL-1β. Although glial cells are traditionally viewed as initiators of neuroinflammation, in this acute phase of inflammation, inflammasome activation was found exclusively in affected motoneurons of the ventral horn in our model. This process was significantly inhibited by 5-BDBD, a P2X4 receptor inhibitor and MCC950, a potent NLRP3 inhibitor. Although at later time points the NLRP3 protein was upregulated in microglia too, no signs of inflammasome activation were detected in these cells. Inhibition of inflammasome activation in motoneurons in the first days after nerve injury hindered development of microgliosis in the spinal cord. Moreover, P2X4 or inflammasome inhibition in the acute phase significantly enhanced nerve regeneration on both the morphological and the functional levels. Conclusions Our results indicate that the central reaction initiated by sciatic nerve injury starts with inflammasome activation in motoneurons of the ventral horn, which triggers a complex inflammatory reaction and activation of microglia. Inhibition of neuronal inflammasome activation not only leads to a significant reduction of microgliosis, but has a beneficial effect on the recovery as well. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02427-9.
Collapse
Affiliation(s)
- Kinga Molnár
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary
| | - Bernát Nógrádi
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary.,Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary.,Department of Neurology, University of Szeged, Szeged, Hungary
| | - Rebeka Kristóf
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary.,Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Ádám Mészáros
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary.,Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Krisztián Pajer
- Department of Anatomy, Histology and Embryology, University of Szeged, Szeged, Hungary
| | - László Siklós
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary
| | - Antal Nógrádi
- Department of Anatomy, Histology and Embryology, University of Szeged, Szeged, Hungary
| | - Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary. .,Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania.
| | - István A Krizbai
- Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network (ELKH), Temesvári krt. 62, 6726, Szeged, Hungary. .,Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania.
| |
Collapse
|
33
|
Zhang J, Zhang X, Wang L, Kang C, Li N, Xiao Z, Dai L. Multiomics-based analyses of KPNA2 highlight its multiple potentials in hepatocellular carcinoma. PeerJ 2021; 9:e12197. [PMID: 34616632 PMCID: PMC8462373 DOI: 10.7717/peerj.12197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/31/2021] [Indexed: 11/20/2022] Open
Abstract
Dysregulation and prognostic roles of Karyopherin α2 (KPNA2) were reported in many malignancies including hepatocellular carcinoma (HCC). A multi-omics analysis of KPNA2 is needed to gain a deeper understanding of its multilevel molecular characteristics and provide novel clues for HCC diagnosis, prognosis, and target therapy. Herein multi-omic alterations of KPNA2 were analyzed at genetic, epigenetic, transcript, and protein levels with evaluation of their relevance with clinicopathological features of HCC by integrative analyses. The significant correlations of KPNA2 expression with its gene copy number variation (CNV) and methylation status were shown through Spearman correlation analyses. With Cox regression, Kaplan-Meier survival, and receiver operating characteristic (ROC) analyses, based on the factors of KPNA2 CNV, methylation, expression, and tumor stage, risk models for HCC overall survival (OS) and disease-free survival (DFS) were constructed which could discriminate the 1-year, 3-year, and 5-year OS/DFS status effectively. With Microenvironment Cell Populations-counter (MCP-counter), the immune infiltrations of HCC samples were evaluated and their associations with KPNA2 were shown. KPNA2 expression in liver was found to be influenced by low fat diet and presented significant correlations with fatty acid metabolism and fatty acid synthase activity in HCC. KPNA2 was detected lowered in HCC patient's plasma by enzyme linked immunosorbent assay (ELISA), consistent with its translocation to nuclei of HCC cells. In conclusion, KPNA2 multilevel dysregulation in HCC and its correlations with immune infiltration and the fatty acid metabolism pathway indicated its multiple roles in HCC. The clinicopathological significance of KPNA2 was highlighted through the in-depth analyses at multilevels.
Collapse
Affiliation(s)
- Jinzhong Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, Henan Province, China
| | - Xiuzhi Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, Henan Province, China.,NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lingxiao Wang
- Department of Pathology, Henan Medical College, Zhengzhou, Henan Province, China
| | - Chunyan Kang
- Department of Pathology, Henan Medical College, Zhengzhou, Henan Province, China
| | - Ningning Li
- Department of Pathology, Henan Medical College, Zhengzhou, Henan Province, China
| | - Zhefeng Xiao
- NHC Key Laboratory of Cancer Proteomics, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan Province, China
| |
Collapse
|
34
|
Liu T, Wang L, Liang P, Wang X, Liu Y, Cai J, She Y, Wang D, Wang Z, Guo Z, Bates S, Xia X, Huang J, Cui J. USP19 suppresses inflammation and promotes M2-like macrophage polarization by manipulating NLRP3 function via autophagy. Cell Mol Immunol 2021; 18:2431-2442. [PMID: 33097834 PMCID: PMC8484569 DOI: 10.1038/s41423-020-00567-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023] Open
Abstract
Macrophage polarization to proinflammatory M1-like or anti-inflammatory M2-like cells is critical to mount a host defense or repair tissue. The exact molecular mechanisms controlling this process are still elusive. Here, we report that ubiquitin-specific protease 19 (USP19) acts as an anti-inflammatory switch that inhibits inflammatory responses and promotes M2-like macrophage polarization. USP19 inhibited NLRP3 inflammasome activation by increasing autophagy flux and decreasing the generation of mitochondrial reactive oxygen species. In addition, USP19 inhibited the proteasomal degradation of inflammasome-independent NLRP3 by cleaving its polyubiquitin chains. USP19-stabilized NLRP3 promoted M2-like macrophage polarization by direct association with interferon regulatory factor 4, thereby preventing its p62-mediated selective autophagic degradation. Consistent with these observations, compared to wild-type mice, Usp19-/- mice had decreased M2-like macrophage polarization and increased interleukin-1β secretion, in response to alum and chitin injections. Thus, we have uncovered an unexpected mechanism by which USP19 switches the proinflammatory function of NLRP3 into an anti-inflammatory function, and suggest that USP19 is a potential therapeutic target for inflammatory interventions.
Collapse
Affiliation(s)
- Tao Liu
- grid.12981.330000 0001 2360 039XMOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, Guangdong People’s Republic of China
| | - Liqiu Wang
- grid.12981.330000 0001 2360 039XMOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, Guangdong People’s Republic of China
| | - Puping Liang
- grid.12981.330000 0001 2360 039XMOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, Guangdong People’s Republic of China
| | - Xiaojuan Wang
- grid.12981.330000 0001 2360 039XDepartment of Experimental Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University, 510275 Guangzhou, People’s Republic of China
| | - Yukun Liu
- grid.12981.330000 0001 2360 039XMOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, Guangdong People’s Republic of China
| | - Jing Cai
- grid.12981.330000 0001 2360 039XMOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, Guangdong People’s Republic of China
| | - Yuanchu She
- grid.12981.330000 0001 2360 039XMOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, Guangdong People’s Republic of China
| | - Dan Wang
- grid.12981.330000 0001 2360 039XDepartment of Experimental Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University, 510275 Guangzhou, People’s Republic of China
| | - Zhi Wang
- grid.12981.330000 0001 2360 039XGuanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, 510060 Guangzhou, Guangdong People’s Republic of China
| | - Zhiyong Guo
- grid.12981.330000 0001 2360 039XOrgan Transplant Center, The First Affiliated Hospital, Sun Yat-Sen University, 510080 Guangzhou, Guangdong People’s Republic of China
| | - Samuel Bates
- grid.38142.3c000000041936754XChanning Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115 USA
| | - Xiaojun Xia
- grid.12981.330000 0001 2360 039XDepartment of Experimental Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University, 510275 Guangzhou, People’s Republic of China
| | - Junjiu Huang
- grid.12981.330000 0001 2360 039XMOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, Guangdong People’s Republic of China
| | - Jun Cui
- grid.12981.330000 0001 2360 039XMOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, 510275 Guangzhou, Guangdong People’s Republic of China ,grid.12981.330000 0001 2360 039XDepartment of Experimental Medicine, State Key Laboratory of Oncology in South China, Sun Yat-sen University, 510275 Guangzhou, People’s Republic of China
| |
Collapse
|
35
|
Inflammaging, an Imbalanced Immune Response That Needs to Be Restored for Cancer Prevention and Treatment in the Elderly. Cells 2021; 10:cells10102562. [PMID: 34685542 PMCID: PMC8533838 DOI: 10.3390/cells10102562] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/03/2021] [Accepted: 09/24/2021] [Indexed: 12/21/2022] Open
Abstract
Nowadays, new advances in society and health have brought an increased life expectancy. However, at the same time, aging comes with complications that impact the development of autoimmunity, neurodegenerative diseases and cancer. These complications affect the quality of life and impact the public health system. Specifically, with aging, a low-grade chronic sterile systemic inflammation with self-reactivity in the absence of acute infection occurs termed inflammaging. Inflammaging is related to an imbalanced immune response that can be either naturally acquired with aging or accelerated due to external triggers. Different molecules, metabolites and inflammatory forms of cell death are highly involved in these processes. Importantly, adoptive cellular immunotherapy is a modality of treatment for cancer patients that administers ex vivo expanded immune cells in the patient. The manipulation of these cells confers them enhanced proinflammatory properties. A general consequence of proinflammatory events is the development of autoimmune diseases and cancer. Herein, we review subsets of immune cells with a pertinent role in inflammaging, relevant proteins involved in these inflammatory events and external triggers that enhance and accelerate these processes. Moreover, we mention relevant preclinical studies that demonstrate associations of chronic inflammation with cancer development.
Collapse
|
36
|
Mendonça LO, Terreri MT, Osaku FM, Barros SF, Köhler KF, Prado AI, Barros MT, Kalil J, Castro FFM. Immunological repertoire linked to PSTPIP1-associated myeloid-related inflammatory (PAMI) syndrome. Pediatr Rheumatol Online J 2021; 19:126. [PMID: 34399798 PMCID: PMC8365952 DOI: 10.1186/s12969-021-00617-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 06/16/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mutations along PSTPIP1 gene are associated to two specific conditions, PAPA syndrome and PAMI syndrome, both autoinflammatory disorders associated to disturbances in cytoskeleton formation. Immunological aspects of PAMI syndrome has not yet been reported neither the clinical impact on therapeutical decisions. METHODS Clinical data of patients records were retrospectively accessed. Genomic DNA were extracted and sequenced following standard procedures. Peripheral lymphocytes were quantified in T, B e FOXP3 phenotypes. RESULTS We describe two related patients with PAMI syndrome harboring the usual E250K mutation. Anti-IL1 therapy could partially control the disease in the index patient. A broad spectrum of immunological effects as well as an aberrant expression of FOXP3 could be observed. CONCLUSIONS Here we report two related brazilian patients with PAMI syndromes harboring the E250K mutation in PSTPIP1, their immunological aspects and the therapeutical response to canakinumab.
Collapse
Affiliation(s)
- Leonardo Oliveira Mendonça
- Discipline of Clinical Immunology and Allergy, Universidade de São Paulo, School of Medicine, Rua Doutor Eneas de Carvalho Aguiar, 255, 8 andar, São Paulo, São Paulo, 05403-000, Brazil. .,Laboratory for Immunological Investigation (LIM-19), Heart Institute, University of São Paulo, São Paulo, Brazil. .,Center for Rare and Immunological Disorders, Hospital 9 de Julho, São Paulo, Brazil.
| | - Maria Teresa Terreri
- grid.411249.b0000 0001 0514 7202Pediatric Rheumatology Department, Universidade Federal de São Paulo, School of Medicine, São Paulo, São Paulo Brazil
| | - Fabiane Mitie Osaku
- grid.414705.3Pediatric Rheumatology, Hospital Infantil Joana de Gusmão, Florianópolis, Santa Catarina Brazil
| | - Samar Freschi Barros
- grid.11899.380000 0004 1937 0722Laboratory for Immunological Investigation (LIM-19), Heart Institute, University of São Paulo, São Paulo, Brazil
| | - Karen Francine Köhler
- grid.11899.380000 0004 1937 0722Laboratory for Immunological Investigation (LIM-19), Heart Institute, University of São Paulo, São Paulo, Brazil
| | - Alex Isidoro Prado
- grid.11899.380000 0004 1937 0722Discipline of Clinical Immunology and Allergy, Universidade de São Paulo, School of Medicine, Rua Doutor Eneas de Carvalho Aguiar, 255, 8 andar, São Paulo, São Paulo 05403-000 Brazil ,Center for Rare and Immunological Disorders, Hospital 9 de Julho, São Paulo, Brazil
| | - Myrthes Toledo Barros
- grid.11899.380000 0004 1937 0722Discipline of Clinical Immunology and Allergy, Universidade de São Paulo, School of Medicine, Rua Doutor Eneas de Carvalho Aguiar, 255, 8 andar, São Paulo, São Paulo 05403-000 Brazil
| | - Jorge Kalil
- grid.11899.380000 0004 1937 0722Discipline of Clinical Immunology and Allergy, Universidade de São Paulo, School of Medicine, Rua Doutor Eneas de Carvalho Aguiar, 255, 8 andar, São Paulo, São Paulo 05403-000 Brazil
| | - Fabio Fernandes Morato Castro
- grid.11899.380000 0004 1937 0722Discipline of Clinical Immunology and Allergy, Universidade de São Paulo, School of Medicine, Rua Doutor Eneas de Carvalho Aguiar, 255, 8 andar, São Paulo, São Paulo 05403-000 Brazil
| |
Collapse
|
37
|
Ma B, Chen D, Liu Y, Zhao Z, Wang J, Zhou G, Xu K, Zhu T, Wang Q, Ma C. Yanghe Decoction Suppresses the Experimental Autoimmune Thyroiditis in Rats by Improving NLRP3 Inflammasome and Immune Dysregulation. Front Pharmacol 2021; 12:645354. [PMID: 34234669 PMCID: PMC8255388 DOI: 10.3389/fphar.2021.645354] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 06/07/2021] [Indexed: 12/16/2022] Open
Abstract
Inflammation is an important contributor to autoimmune thyroiditis. Yanghe decoction (YH) is a traditional Chinese herbal formulation which has various anti-inflammatory effects. It has been used for the treatment of autoimmune diseases such as ankylosing spondylitis In this study we aimed to investigate the effects of YH on autoimmune thyroiditis in a rat model and elucidate the underlying mechanisms. The experimental autoimmune thyroiditis (EAT) model was established by thyroglobulin (pTG) injections and excessive iodine intake. Thyroid lesions were observed using hematoxylin and eosin (H and E) staining and serum TgAb, TPOAb, TSH, T3, and T4 levels were measured by enzyme-linked immunosorbent assay IL-35 levels were evaluated using real-time polymerase chain reaction (RT-PCR) and Th17/Treg balance in peripheral blood mononuclear cells (PBMCs) was determined by flow cytometry and RT-PCR. Changes in Wnt/β-catenin signaling were evaluated using Western blot. Immunofluorescence staining and western blot were employed to examine NLRP3 inflammasome activation in the thyroid. YH minimized thyroid follicle injury and decreased concentrations of serum TgAb, TPOAb, TSH, T3, and T4 in EAT model. The mRNA of IL-35 was increased after YH treatment. YH also increased the percentage of Treg cells, and decreased Th17 proportion as well as Th17/Treg ratio in PBMCs. Meanwhile, the mRNA levels of Th17 related cytokines (RORγt, IL-17A, IL-21, and IL-22) were suppressed and Treg related cytokines (FoxP3, TGF-β, and IL-10) were promoted in PBMCs. Additionally, the protein expressions of Wnt-1 and β-catenin were unregulated after YH treatment. NLRP3 immunostaining signal and protein levels of IL-17, p-NF-κB, NLRP3, ASC, cleaved-Caspase-1, cleaved-IL-1β, and IL-18 were downregulated in the thyroid after YH intervention. Overall, the present study demonstrated that YH alleviated autoimmune thyroiditis in rats by improving NLRP3 inflammasome and immune dysregulation.
Collapse
Affiliation(s)
- Bing'e Ma
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China.,Department of Thyroid and Breast Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine for Nanjing University of Chinese Medicine, Jiangsu, China
| | - Dexuan Chen
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Yangjing Liu
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Zhengping Zhao
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine for Nanjing University of Chinese Medicine, Jiangsu, China
| | - Jianhua Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine for Nanjing University of Chinese Medicine, Jiangsu, China
| | - Guowei Zhou
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Kun Xu
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Taiyang Zhu
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Qiong Wang
- Department of Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Chaoqun Ma
- Department of General Surgery, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| |
Collapse
|
38
|
Ni D, Tang T, Lu Y, Xu K, Shao Y, Saaoud F, Saredy J, Liu L, Drummer C, Sun Y, Hu W, Lopez-Pastrana J, Luo JJ, Jiang X, Choi ET, Wang H, Yang X. Canonical Secretomes, Innate Immune Caspase-1-, 4/11-Gasdermin D Non-Canonical Secretomes and Exosomes May Contribute to Maintain Treg-Ness for Treg Immunosuppression, Tissue Repair and Modulate Anti-Tumor Immunity via ROS Pathways. Front Immunol 2021; 12:678201. [PMID: 34084175 PMCID: PMC8168470 DOI: 10.3389/fimmu.2021.678201] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 04/01/2021] [Indexed: 12/11/2022] Open
Abstract
We performed a transcriptomic analyses using the strategies we pioneered and made the following findings: 1) Normal lymphoid Tregs, diseased kidney Tregs, splenic Tregs from mice with injured muscle have 3, 17 and 3 specific (S-) pathways, respectively; 2) Tumor splenic Tregs share 12 pathways with tumor Tregs; tumor splenic Tregs and tumor Tregs have 11 and 8 S-pathways, respectively; 3) Normal and non-tumor disease Tregs upregulate some of novel 2641 canonical secretomic genes (SGs) with 24 pathways, and tumor Tregs upregulate canonical secretomes with 17 pathways; 4) Normal and non-tumor disease tissue Tregs upregulate some of novel 6560 exosome SGs with 56 exosome SG pathways (ESP), tumor Treg ESP are more focused than other Tregs; 5) Normal, non-tumor diseased Treg and tumor Tregs upregulate some of novel 961 innate immune caspase-1 SGs and 1223 innate immune caspase-4 SGs to fulfill their tissue/SG-specific and shared functions; 6) Most tissue Treg transcriptomes are controlled by Foxp3; and Tumor Tregs had increased Foxp3 non-collaboration genes with ROS and 17 other pathways; 7) Immune checkpoint receptor PD-1 does, but CTLA-4 does not, play significant roles in promoting Treg upregulated genes in normal and non-tumor disease tissue Tregs; and tumor splenic and tumor Tregs have certain CTLA-4-, and PD-1-, non-collaboration transcriptomic changes with innate immune dominant pathways; 8) Tumor Tregs downregulate more immunometabolic and innate immune memory (trained immunity) genes than Tregs from other groups; and 11) ROS significantly regulate Treg transcriptomes; and ROS-suppressed genes are downregulated more in tumor Tregs than Tregs from other groups. Our results have provided novel insights on the roles of Tregs in normal, injuries, regeneration, tumor conditions and some of canonical and innate immune non-canonical secretomes via ROS-regulatory mechanisms and new therapeutic targets for immunosuppression, tissue repair, cardiovascular diseases, chronic kidney disease, autoimmune diseases, transplantation, and cancers.
Collapse
Affiliation(s)
- Dong Ni
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - TingTing Tang
- Metabolic Disease Research & Thrombosis Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yifan Lu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Keman Xu
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Ying Shao
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Fatma Saaoud
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jason Saredy
- Metabolic Disease Research & Thrombosis Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Lu Liu
- Metabolic Disease Research & Thrombosis Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Charles Drummer
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Yu Sun
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Wenhui Hu
- Metabolic Disease Research & Thrombosis Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jahaira Lopez-Pastrana
- Department of Psychiatry, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Jin J Luo
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaohua Jiang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Metabolic Disease Research & Thrombosis Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Eric T Choi
- Division of Vascular and Endovascular Surgery, Department of Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Hong Wang
- Metabolic Disease Research & Thrombosis Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Centers for Cardiovascular Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Metabolic Disease Research & Thrombosis Research, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Inflammation, Translational & Clinical Lung Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
39
|
Jin S, Sun S, Ling H, Ma J, Zhang X, Xie Z, Zhan N, Zheng W, Li M, Qin Y, Zhao H, Chen Y, Yang X, Wang J. Protectin DX restores Treg/T h17 cell balance in rheumatoid arthritis by inhibiting NLRP3 inflammasome via miR-20a. Cell Death Dis 2021; 12:280. [PMID: 33723242 PMCID: PMC7961047 DOI: 10.1038/s41419-021-03562-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 01/31/2023]
Abstract
Regulatory T-cell (Treg)/T-helper 17 (Th17) cell balance plays an important role in the progression of rheumatoid arthritis (RA). Our study explored the protective effect of protectin DX (PDX), which restored Treg/Th17 cell balance in RA, and the role of the nucleotide-binding domain (NOD)-like receptor protein 3 (NLRP3) inflammasome pathway in this process. Using mass spectrometry, we discovered that level of PDX decreased in active-RA patients and increased in inactive-RA patients compared with HCs, and serum PDX was a potential biomarker in RA activity detection (area under the curve [AUC] = 0.86). In addition, a collagen-induced arthritis (CIA) mice model was constructed and PDX obviously delayed RA progression in the CIA model, upregulating Tregs and anti-inflammatory cytokines while downregulating Th17 cells and pro-inflammatory cytokines. Moreover, NLRP3 knockout and rescue experiments demonstrated that NLRP3 participated in PDX-mediated Treg/Th17 cell balance restoration, joint injury amelioration and inflammatory-response attenuation using Nlrp3-/- mice. Furthermore, microarray and verified experiments confirmed that PDX reduced NLRP3 expression via miRNA-20a (miR-20a). In summary, we confirmed for the first time that PDX could effectively ameliorate CIA progression by restoring Treg/Th17 cell balance, which was mediated by inhibition of the NLRP3 inflammasome pathway via miR-20a.
Collapse
MESH Headings
- Animals
- Antirheumatic Agents/pharmacology
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/metabolism
- Case-Control Studies
- Cytokines/metabolism
- Docosahexaenoic Acids/blood
- Docosahexaenoic Acids/pharmacology
- Humans
- Inflammasomes/antagonists & inhibitors
- Inflammasomes/genetics
- Inflammasomes/metabolism
- Inflammation Mediators/metabolism
- Mice, Inbred DBA
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Mice
Collapse
Affiliation(s)
- Shengwei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Siyuan Sun
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hanzhi Ling
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jinglan Ma
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xu Zhang
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhen Xie
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ning Zhan
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenjie Zheng
- Department of Pediatric Nephrology and Rheumatology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Man Li
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yang Qin
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Heping Zhao
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yan Chen
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinyu Yang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China.
| | - Jianguang Wang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Department of Biochemistry, School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
40
|
Xie K, Chen YQ, Chai YS, Lin SH, Wang CJ, Xu F. HMGB1 suppress the expression of IL-35 by regulating Naïve CD4+ T cell differentiation and aggravating Caspase-11-dependent pyroptosis in acute lung injury. Int Immunopharmacol 2021; 91:107295. [PMID: 33360086 DOI: 10.1016/j.intimp.2020.107295] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a severe form of inflammatory lung disease. Its development and progression are regulated by cytokines. The purpose of this study was to determine the effects of HMGB1 involved in the regulation of Treg cells and IL-35. METHODS A cecal ligation and puncture (CLP)-induced ALI model was used to investigate the changes in IL-35, Tregs, and the expression of RAGE and caspase-11 after HMGB1 inhibition (glycyrrhizin was used as an inhibitor of HMGB1). CD4+ naïve T cells sorted from C57BL/6 mice spleens were cultured to explore the role of HMGB1 in the differentiation from CD4+ naïve T cells to Tregs. RESULTS HMGB1 promoted lung injury and uncontrolled inflammation in the CLP mouse model. HMGB1, NF-κB p65, RAGE, and caspase-11 expression in the lungs of CLP mice decreased significantly after pretreatment with glycyrrhizin. We found that the Treg proportion and IL-35 expression were upregulated in the serum and lung of CLP mice after inhibiting HMGB1. In our in vitro experiments, we found that recombinant HMGB1 significantly suppressed the proportion of CD4+CD25+FOXP3+Tregs differentiated from CD4+ naïve T cells. CONCLUSIONS The inhibition of HMGB1 increased the proportion of Treg and expression of IL-35 and alleviated lung injury in the CLP-induced ALI model. Furthermore, inhibition of HMGB1 reduced caspase-11-dependent pyroptosis in the lungs of the CLP-induced ALI model.
Collapse
Affiliation(s)
- Ke Xie
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan-Qing Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yu-Sen Chai
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shi-Hui Lin
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chuan-Jiang Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Fang Xu
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
41
|
Nógrádi B, Nyúl-Tóth Á, Kozma M, Molnár K, Patai R, Siklós L, Wilhelm I, Krizbai IA. Upregulation of Nucleotide-Binding Oligomerization Domain-, LRR- and Pyrin Domain-Containing Protein 3 in Motoneurons Following Peripheral Nerve Injury in Mice. Front Pharmacol 2020; 11:584184. [PMID: 33328988 PMCID: PMC7732612 DOI: 10.3389/fphar.2020.584184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022] Open
Abstract
Neuronal injuries are accompanied by release and accumulation of damage-associated molecules, which in turn may contribute to activation of the immune system. Since a wide range of danger signals (including endogenous ones) are detected by the nucleotide-binding oligomerization domain-, LRR- and pyrin domain-containing protein 3 (NLRP3) pattern recognition receptor, we hypothesized that NLRP3 may become activated in response to motor neuron injury. Here we show that peripheral injury of the oculomotor and the hypoglossal nerves results in upregulation of NLRP3 in corresponding motor nuclei in the brainstem of mice. Although basal expression of NLRP3 was observed in microglia, astroglia and neurons as well, its upregulation and co-localization with apoptosis-associated speck-like protein containing a caspase activation and recruitment domain, suggesting inflammasome activation, was only detected in neurons. Consequently, increased production of active pro-inflammatory cytokines interleukin-1β and interleukin-18 were detected after hypoglossal nerve axotomy. Injury-sensitive hypoglossal neurons responded with a more pronounced NLRP3 upregulation than injury-resistant motor neurons of the oculomotor nucleus. We further demonstrated that the mitochondrial protector diazoxide was able to reduce NLRP3 upregulation in a post-operative treatment paradigm. Our results indicate that NLRP3 is activated in motoneurons following acute nerve injury. Blockade of NLRP3 activation might contribute to the previously observed anti-inflammatory and neuroprotective effects of diazoxide.
Collapse
Affiliation(s)
- Bernát Nógrádi
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Foundation for the Future of Biomedical Sciences in Szeged, Szeged Scientists Academy, Szeged, Hungary
| | - Ádám Nyúl-Tóth
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Department of Biochemistry and Molecular Biology, Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Mihály Kozma
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Kinga Molnár
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Theoretical Medicine Doctoral School, University of Szeged, Szeged, Hungary
| | - Roland Patai
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - László Siklós
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Imola Wilhelm
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania
| | - István A Krizbai
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary.,Institute of Life Sciences, Vasile Goldiş Western University of Arad, Arad, Romania
| |
Collapse
|
42
|
Zhang X, Zhang J, Gao F, Fan S, Dai L, Zhang J. KPNA2-Associated Immune Analyses Highlight the Dysregulation and Prognostic Effects of GRB2, NRAS, and Their RNA-Binding Proteins in Hepatocellular Carcinoma. Front Genet 2020; 11:593273. [PMID: 33193737 PMCID: PMC7649362 DOI: 10.3389/fgene.2020.593273] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 09/21/2020] [Indexed: 12/14/2022] Open
Abstract
Karyopherin α2 (KPNA2) was reported to be overexpressed and have unfavorable prognostic effects in many malignancies including hepatocellular carcinoma (HCC). Although its contributions to inflammatory response were reported in many studies, its specific associations with immune infiltrations and immune pathways during cancer progression were unclear. Here, we aimed to identify new markers for HCC diagnosis and prognosis through KPNA2-associated immune analyses. RNA-seq expression data of HCC datasets were downloaded from The Cancer Genome Atlas and International Cancer Genome Consortium. The gene expressions were counts per million normalized. The infiltrations of 24 kinds of immune cells in the samples were evaluated with ImmuCellAI (Immune Cell Abundance Identifier). The Spearman correlations of the immune infiltrations with KPNA2 expression were investigated, and the specific positive correlation of B-cell infiltration with KPNA2 expression in HCC tumors was identified. Fifteen genes in KEGG (Kyoto Encyclopedia of Genes and Genomes) B-cell receptor signaling pathway presented significant correlations with KPNA2 expression in HCC. Among them, GRB2 and NRAS were indicated to be independent unfavorable prognostic factors for HCC overall survival. Clinical Proteomic Tumor Analysis Consortium HCC dataset was investigated to validate the results at protein level. The upregulation and unfavorable prognostic effects of KPNA2 and GRB2 were confirmed, whereas, unlike its mRNA form, NRAS protein was presented to be downregulated and have favorable prognostic effects. Through receiver operating characteristic curve analysis, the diagnostic potential of the three proteins was shown. The RNA-binding proteins (RBPs) of KPNA2, NRAS, and GRB2, downloaded via The Encyclopedia of RNA Interactomes, were investigated for their clinical significance in HCC at protein level. An eight-RBP signature with independent prognostic value and dysregulations in HCC was identified. All the RBPs were significantly correlated with MKI67 expression and at least one of KPNA2, GRB2, and NRAS at protein level in HCC, indicating their roles in HCC progression and the regulation of the three proteins. We concluded that KPNA2, GRB2, NRAS, and their RBPs might have coordinating roles in HCC immunoregulation and progression. They might be new markers for HCC diagnosis and prognosis predication and new targets for HCC immunotherapy.
Collapse
Affiliation(s)
- Xiuzhi Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Jialing Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Fenglan Gao
- Department of Pathology, Henan Medical College, Zhengzhou, China
| | - Shasha Fan
- Oncology Department, The First Affiliated Hospital of Hunan Normal University, Hunan Provincial People's Hospital, Changsha, China.,Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Hunan Normal University, Changsha, China
| | - Liping Dai
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinzhong Zhang
- Department of Pathology, Henan Medical College, Zhengzhou, China
| |
Collapse
|
43
|
Yang SR, Hsu WH, Wu CY, Shang HS, Liu FC, Chen A, Hua KF, Ka SM. Accelerated, severe lupus nephritis benefits from treatment with honokiol by immunoregulation and differentially regulating NF-κB/NLRP3 inflammasome and sirtuin 1/autophagy axis. FASEB J 2020; 34:13284-13299. [PMID: 32813287 DOI: 10.1096/fj.202001326r] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 07/08/2020] [Accepted: 07/16/2020] [Indexed: 12/21/2022]
Abstract
Using honokiol (HNK), a major anti-inflammatory bioactive compound in Magnolia officinalis, we show a potent therapeutic outcome against an accelerated, severe form of lupus nephritis (ASLN). The latter may follow infectious insults that act as environmental triggers in the patients. In the current study, an ASLN model in NZB/W F1 mice was treated with HNK by daily gavage after onset of the disease. We show that HNK ameliorated the ASLN by improving renal function, albuminuria, and renal pathology, especially reducing cellular crescents, neutrophil influx, fibrinoid necrosis in glomeruli, and glomerulonephritis activity scores. Meanwhile, HNK differentially regulated T cell functions, reduced serum anti-dsDNA autoantibodies, and inhibited NLRP3 inflammasome activation in the mice. The latter involved: (a) suppressed production of reactive oxygen species and NF-κB activation-mediated priming signal of the inflammasome, (b) reduced mitochondrial damage, and (c) enhanced sirtuin 1 (SIRT1)/autophagy axis activation. In conclusion, HNK represents a new drug candidate for acute, severe episodes of LN capable of alleviating renal lesions in ASLN mice by negatively regulating T cell functions and by enhancing SIRT1/autophagy axis-lessened NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Shin-Ruen Yang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wan-Han Hsu
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chung-Yao Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hung-Sheng Shang
- Division of Clinical Pathology, Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Feng-Cheng Liu
- Division of Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ann Chen
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Shuk-Man Ka
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.,Graduate Institute of Aerospace and Undersea Medicine, Department of Medicine, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
44
|
Park SH, Jung HJ, Kim TS. IL-33 changes CD25hi Tregs to Th17 cells through a dendritic cell-mediated pathway. Immunol Lett 2020; 218:5-10. [DOI: 10.1016/j.imlet.2019.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 11/30/2019] [Accepted: 12/17/2019] [Indexed: 02/06/2023]
|