1
|
Sharma P, Sethi RS. In Vivo Exposure of Deltamethrin Dysregulates the NFAT Signalling Pathway and Induces Lung Damage. J Toxicol 2024; 2024:5261994. [PMID: 39239465 PMCID: PMC11377118 DOI: 10.1155/2024/5261994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 07/23/2024] [Accepted: 08/10/2024] [Indexed: 09/07/2024] Open
Abstract
Deltamethrin is an insecticide used to control harmful agricultural insects that otherwise damage crops and to control vector-borne diseases. Long-term exposure to deltamethrin results in the inflammation of the lungs. The present study elucidates the molecular mechanism underlying the deltamethrin-induced lung damage. The lung samples were extracted from the Swiss albino mice following the treatment of low (2.5 mg/kg) and high (5 mg/kg) doses of deltamethrin. The mRNA expression of TCR, IL-4, and IL-13 showed upregulation, while the expression of NFAT and FOS was downregulated following a low dose of deltamethrin. Moreover, the expression of TCR was downregulated with the exposure of a high dose of deltamethrin. Furthermore, the immunohistochemistry data confirmed the pattern of protein expression for TCR, FOS, IL-4, and IL-13 following a low dose of deltamethrin exposure. However, no change was seen in the TCR, NFAT, FOS, JUN, IL-4, and IL-13 immunopositive cells of the high-dose treatment group. Also, ELISA results showed increased expression of IL-13 in the BAL fluid of animals exposed to low doses of deltamethrin. Overall, the present study showed that deltamethrin exposure induces lung damage and immune dysregulation via dysregulating the NFAT signalling pathway.
Collapse
Affiliation(s)
- Prakriti Sharma
- Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | - R S Sethi
- Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| |
Collapse
|
2
|
Nolze A, Matern S, Grossmann C. Calcineurin Is a Universal Regulator of Vessel Function-Focus on Vascular Smooth Muscle Cells. Cells 2023; 12:2269. [PMID: 37759492 PMCID: PMC10528183 DOI: 10.3390/cells12182269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Calcineurin, a serine/threonine phosphatase regulating transcription factors like NFaT and CREB, is well known for its immune modulatory effects and role in cardiac hypertrophy. Results from experiments with calcineurin knockout animals and calcineurin inhibitors indicate that calcineurin also plays a crucial role in vascular function, especially in vascular smooth muscle cells (VSMCs). In the aorta, calcineurin stimulates the proliferation and migration of VSMCs in response to vascular injury or angiotensin II administration, leading to pathological vessel wall thickening. In the heart, calcineurin mediates coronary artery formation and VSMC differentiation, which are crucial for proper heart development. In pulmonary VSMCs, calcineurin/NFaT signaling regulates the release of Ca2+, resulting in increased vascular tone followed by pulmonary arterial hypertension. In renal VSMCs, calcineurin regulates extracellular matrix secretion promoting fibrosis development. In the mesenteric and cerebral arteries, calcineurin mediates a phenotypic switch of VSMCs leading to altered cell function. Gaining deeper insights into the underlying mechanisms of calcineurin signaling will help researchers to understand developmental and pathogenetical aspects of the vasculature. In this review, we provide an overview of the physiological function and pathophysiology of calcineurin in the vascular system with a focus on vascular smooth muscle cells in different organs. Overall, there are indications that under certain pathological settings reduced calcineurin activity seems to be beneficial for cardiovascular health.
Collapse
Affiliation(s)
| | | | - Claudia Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| |
Collapse
|
3
|
Hou X, Hong X, Ou M, Meng S, Wang T, Liao S, He J, Yu H, Liu L, Yin L, Liu D, Tang D, Dai Y. Analysis of Gene Expression and TCR/B Cell Receptor Profiling of Immune Cells in Primary Sjögren's Syndrome by Single-Cell Sequencing. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:238-249. [PMID: 35705251 DOI: 10.4049/jimmunol.2100803] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 04/18/2022] [Indexed: 01/07/2023]
Abstract
Primary Sjögren's syndrome (pSS) is a chronic autoimmune disease that is estimated to affect 35 million people worldwide and is characterized by lymphocytic infiltration, elevated circulating autoantibodies, and proinflammatory cytokines. The key immune cell subset changes and the TCR/BCR repertoire alterations in pSS patients remain unclear. In this study, we sought to comprehensively characterize the transcriptional changes in PBMCs of pSS patients by single-cell RNA sequencing and single-cell V(D)J sequencing. Naive CD8+ T cells and mucosal-associated invariant T cells were markedly decreased but regulatory T cells were increased in pSS patients. There were a large number of differentially expressed genes shared by multiple subpopulations of T cells and B cells. Abnormal signaling pathways, including Ag processing and presentation, the BCR signaling pathway, the TCR signaling pathway, and Epstein-Barr virus infection, were highly enriched in pSS patients. Moreover, there were obvious differences in the CD30, FLT3, IFN-II, IL-1, IL-2, IL-6, IL-10, RESISTIN, TGF-β, TNF, and VEGF signaling networks between pSS patients and healthy controls. Single-cell TCR and BCR repertoire analysis showed that there was a lower diversity of T cells in pSS patients than in healthy controls; however, there was no significant difference in the degree of clonal expansion, CDR3 length distribution, or degree of sequence sharing. Notably, our results further emphasize the functional importance of αβ pairing in determining Ag specificity. In conclusion, our analysis provides a comprehensive single-cell map of gene expression and TCR/BCR profiles in pSS patients for a better understanding of the pathogenesis, diagnosis, and treatment of pSS.
Collapse
Affiliation(s)
- Xianliang Hou
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China.,The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
| | - Xiaoping Hong
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Minglin Ou
- Central Laboratory, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, China; and
| | - Shuhui Meng
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Tingting Wang
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Shengyou Liao
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Jingquan He
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Haiyan Yu
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Lixiong Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Lianghong Yin
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Dongzhou Liu
- Department of Rheumatology and Immunology, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China;
| | - Donge Tang
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China;
| | - Yong Dai
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China;
| |
Collapse
|
4
|
Ramírez-Bello J, Jiménez-Morales S, Barbosa-Cobos RE, Sánchez-Zauco N, Hernández-Molina G, Luria-Pérez R, Fragoso JM, Cabello-Gutiérrez C, Montúfar-Robles I. TNFSF4 is a risk factor for rheumatoid arthritis but not for primary Sjögren's syndrome in the Mexican population. Immunobiology 2022; 227:152244. [PMID: 35835012 DOI: 10.1016/j.imbio.2022.152244] [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: 03/02/2022] [Revised: 06/04/2022] [Accepted: 07/06/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE Rheumatoid arthritis (RA) and primary Sjögren's syndrome (pSS) are autoimmune diseases (ADs) characterized by joint damage and involvement of the salivary glands, respectively. ADs share some susceptibility loci, such as TNFSF4, which is a classical susceptibility gene associated with systemic lupus erythematosus, but its role in RA and pSS is not yet clear. Thus, the aim of this study was to determine whether three TNFSFS4 polymorphisms are associated with RA and pSS. METHODS Our case-control study included 500 controls, 459 patients with RA, and 210 patients with pSS from Mexico. TNFSF4 single nucleotide polymorphisms (SNPs) rs1234315C/T, rs2205960G/T, and rs704840T/G were genotyped using TaqMan probes and discrimination allelic assay. RESULTS The three TNFSF4 SNPs were associated with susceptibility to RA (rs1234315C/T: odds ratio [OR] 1.4, p = 0.01; rs2205960G/T: OR 1.23, p = 0.03; rs704840T/G: OR 1.24, p = 0.02). An association between TNFSF4 rs1234315C/T and pSS was also observed (OR 1.28, p = 0.04), however, after Bonferroni correction, this association was lost. CONCLUSION Our data suggest that TNFSF4 could be a risk factor in RA but not pSS in a Mexican population.
Collapse
Affiliation(s)
- Julian Ramírez-Bello
- Departamento de Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, 14080 Mexico City, Mexico.
| | - Silvia Jiménez-Morales
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica, 14610 Mexico City, Mexico.
| | | | - Norma Sánchez-Zauco
- División de Auxiliares de Diagnóstico y Tratamiento, UMAE Hospital de Especialidades, Centro Médico Nacional Siglo XXI, 06720 IMSS, Mexico.
| | - Gabriela Hernández-Molina
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, 14080 Mexico City, Mexico.
| | - Rosendo Luria-Pérez
- Unidad de Investigación en Enfermedades Hemato-Oncológicas, Hospital Infantil de México Federico Gómez, 06720, Mexico.
| | - José M Fragoso
- Departamento de Biología Molecular, Instituto Nacional de Cardiología Ignacio Chávez, 14080 Mexico City, Mexico.
| | - Carlos Cabello-Gutiérrez
- Departamento de Investigación en Virología y Micología, Instituto Nacional de Enfermedades Respiratorias, 14080 Mexico City, Mexico.
| | | |
Collapse
|
5
|
Cai Y, Yao H, Sun Z, Wang Y, Zhao Y, Wang Z, Li L. Role of NFAT in the Progression of Diabetic Atherosclerosis. Front Cardiovasc Med 2021; 8:635172. [PMID: 33791348 PMCID: PMC8006278 DOI: 10.3389/fcvm.2021.635172] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/15/2021] [Indexed: 12/15/2022] Open
Abstract
Nuclear factor of activated T cells (NFAT) is a transcription factor with a multidirectional regulatory function, that is widely expressed in immune cells, including cells in the cardiovascular system, and non-immune cells. A large number of studies have confirmed that calcineurin/NFAT signal transduction is very important in the development of vascular system and cardiovascular system during embryonic development, and plays some role in the occurrence of vascular diseases such as atherosclerosis, vascular calcification, and hypertension. Recent in vitro and in vivo studies have shown that NFAT proteins and their activation in the nucleus and binding to DNA-related sites can easily ɨnduce the expression of downstream target genes that participate in the proliferation, migration, angiogenesis, and vascular inflammation of vascular wall related cells in various pathophysiological states. NFAT expression is regulated by various signaling pathways, including CD137-CD137L, and OX40-OX40L pathways. As a functionally diverse transcription factor, NFAT interacts with a large number of signaling molecules to modulate intracellular and extracellular signaling pathways. These NFAT-centered signaling pathways play important regulatory roles in the progression of atherosclerosis, such as in vascular smooth muscle cell phenotypic transition and migration, endothelial cell injury, macrophage-derived foam cell formation, and plaque calcification. NFAT and related signaling pathways provide new therapeutic targets for vascular diseases such as atherosclerosis. Hence, further studies of the mechanism of NFAT in the occurrence and evolution of atherosclerosis remain crucial.
Collapse
Affiliation(s)
- Yaoyao Cai
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Haipeng Yao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ying Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yunyun Zhao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
6
|
CD137 Signaling Promotes Endothelial Apoptosis by Inhibiting Nrf2 Pathway, and Upregulating NF- κB Pathway. Mediators Inflamm 2020; 2020:4321912. [PMID: 32587470 PMCID: PMC7294359 DOI: 10.1155/2020/4321912] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/02/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
Background Endothelial dysfunction and apoptosis resulting from oxidative stress can lead to the development of atherosclerosis. Our group has previously showed that CD137 signaling contributes to the progression of atherosclerosis and the vulnerability of plaques. The aim of this study is to investigate the effects of CD137 signaling in atherosclerosis on endothelial cells (ECs) apoptosis and to explore the underlying mechanisms. Methods Serum samples were collected from 11 patients with acute myocardial infarction and 4 controls. Peritoneal injection of agonist-CD137 recombinant protein in ApoE−/− mice was used to determine whether CD137 signaling can promote apoptosis in vivo, and human umbilical vein endothelial cells treated with agonist-CD137 recombinant protein, M5580 (a Nrf2 pathway agonist) and CAPE (a NF-κB pathway inhibitor) were used to explore the effect of Nrf2 and NF-κB pathway in CD137 signaling-induced ECs apoptosis in vitro. Results ELISA showed that Bcl-2 in the serum of AMI patients was lower than that of the control group, while TNF-α and sCD137 were higher than that of the control group. Confocal microscopy and Western blot analysis showed that the nuclear translocation of Nrf2 in the agonist-CD137 group was significantly inhibited, and the expression of its downstream antioxidant enzymes was also decreased when compared with control. Immunofluorescence and Western blot results showed that the nuclear translocation of NF-κB in the agonist-CD137 group was enhanced, and ELISA results showed that the secretion of proinflammatory cytokines in the agonist-CD137 group was increased. Immunofluorescence results revealed that ROS production in the agonist-CD137 group was higher than that in control, M5580 (a Nrf2 pathway agonist) and CAPE (a NF-κB pathway inhibitor) groups. In vitro studies using HUVECs and in vivo studies using high-fat-fed ApoE−/− mice showed that the number of apoptotic endothelial cells was the highest in the agonist-CD137 group. By contrast, both M5580 and CAPE treatments were able to reduce CD137 induced ECs apoptosis. Conclusions Our results showed that CD137 signaling promotes ECs apoptosis through prooxidative and proinflammatory mechanisms, mediated by Nrf2 and NF-κB pathways, respectively.
Collapse
|
7
|
Gang W, Yu-Zhu W, Yang Y, Feng S, Xing-Li F, Heng Z. The critical role of calcineurin/NFAT (C/N) pathways and effective antitumor prospect for colorectal cancers. J Cell Biochem 2019; 120:19254-19273. [PMID: 31489709 DOI: 10.1002/jcb.29243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 06/10/2019] [Indexed: 12/11/2022]
Abstract
Transcription factors (TFs) like a nuclear factor of activated T-cells (NFAT) and its controller calcineurin are highly expressed in primary intestinal epithelial cells (IECs) due to delamination, damage by tumor-associated flora and selective activation in the intestinal tract tumor are crucial in the progression and growth of colorectal cancer (CRC). This study sought to summarize the current findings concerning the dysregulated calcineurin/NFAT (C/N) signaling involved in CRC initiation and progression. These signalings include proliferation, T-cell functions, and glycolysis with high lactate production that remodels the acidosis, which genes in tumor cells provide an evolutionary advantage, or even increased their attack phenotype. Moreover, the relationship between C/N and gut microbiome in CRC, especially role of NFAT and toll-like receptor signaling in regulating intestinal microbiota are also discussed. Furthermore, this review will discuss the proteins and genes relating to C/N induced acidosis in CRC, which includes ASIC2 regulated C/N1 and TFs associated with the glycolytic by-product that affect T-cell functions and CRC cell growth. It is revealed that calcineurin or NFAT targeting to antitumor, selective calcineurin inhibition or targets in NFAT signaling may be useful for clinical treatment of CRC. This can further aid in the identification of specific targets via cancer patient-personalized approach. Future studies should be focused on targeting to C/N or TLR signaling by the combination of therapeutic agents to regulate T-cell functions and gut microbiome for activating potent anticancer property with the prospect of potentiating the antitumor therapy for CRC.
Collapse
Affiliation(s)
- Wang Gang
- Department of Pharmaceutics, Shanghai Eight People's Hospital, Jiangsu University, Shanghai, China
| | - Wang Yu-Zhu
- Department of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yu Yang
- Department of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Shi Feng
- Department of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Fu Xing-Li
- Department of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zhang Heng
- Department of General Surgery, Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| |
Collapse
|
8
|
Lv YW, Chen Y, Lv HT, Li X, Tang YJ, Qian WG, Xu QQ, Sun L, Qian GH, Ding YY. Kawasaki disease OX40-OX40L axis acts as an upstream regulator of NFAT signaling pathway. Pediatr Res 2019; 85:835-840. [PMID: 30723312 DOI: 10.1038/s41390-019-0312-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 12/22/2018] [Accepted: 01/16/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND We investigated a costimulatory molecule OX40-OX40L acting as an upstream regulator to regulate the nuclear factor of activated T cell (NFAT) in the acute phase of Kawasaki disease (KD). METHODS One hundred and one samples were collected and divided into six groups: coronary artery lesion (KD-CAL) before intravenous immunoglobulin (IVIG), KD-CAL after IVIG, KD without CAL (KD-nCAL) before IVIG, KD-nCAL after IVIG, fever of unknown (Fou), and Healthy. In vitro OX40-stimulating and OX40L-inhibiting tests were conducted in Healthy and KD groups, respectively. Both the messenger RNA (mRNA) and protein expression levels of OX40, OX40L, NFAT1, and NFAT2 were investigated using quantitative reverse transcription PCR and immunoblotting assay, respectively. RESULTS The mRNA and protein expression levels of NFAT1, NFAT2, OX40, and OX40L were significantly increased in KD-CAL and KD-nCAL groups before IVIG compared with Fou and Healthy groups and decreased after IVIG. A positive correlation was found between them in KD. In vitro OX40-stimulating test demonstrated the significantly increased mRNA and protein expression levels of NFAT1 and NFAT2 in the peripheral blood mononuclear cells of the Healthy group. Meanwhile, OX40L-inhibiting test showed significantly decreased expression levels of NFAT1 and NFAT2 in the KD group. CONCLUSION OX40-OX40L acts as an upstream regulator in the NFAT signaling pathway involved in KD.
Collapse
Affiliation(s)
- Yu-Wen Lv
- Cardiology Department, Children's Hospital of Soochow University, 215025, Suzhou, Jiangsu Province, China
| | - Ye Chen
- Cardiology Department, Children's Hospital of Soochow University, 215025, Suzhou, Jiangsu Province, China
| | - Hai-Tao Lv
- Cardiology Department, Children's Hospital of Soochow University, 215025, Suzhou, Jiangsu Province, China
| | - Xuan Li
- Cardiology Department, Children's Hospital of Soochow University, 215025, Suzhou, Jiangsu Province, China
| | - Yun-Jia Tang
- Cardiology Department, Children's Hospital of Soochow University, 215025, Suzhou, Jiangsu Province, China
| | - Wei-Guo Qian
- Cardiology Department, Children's Hospital of Soochow University, 215025, Suzhou, Jiangsu Province, China
| | - Qiu-Qin Xu
- Cardiology Department, Children's Hospital of Soochow University, 215025, Suzhou, Jiangsu Province, China
| | - Ling Sun
- Cardiology Department, Children's Hospital of Soochow University, 215025, Suzhou, Jiangsu Province, China
| | - Guang-Hui Qian
- Institute of Pediatric Research, Children's Hospital of Soochow University, 215025, Suzhou, Jiangsu Province, China.
| | - Yue-Yue Ding
- Cardiology Department, Children's Hospital of Soochow University, 215025, Suzhou, Jiangsu Province, China.
| |
Collapse
|
9
|
Xia L, Wu J, Pattaradilokrat S, Tumas K, He X, Peng YC, Huang R, Myers TG, Long CA, Wang R, Su XZ. Detection of host pathways universally inhibited after Plasmodium yoelii infection for immune intervention. Sci Rep 2018; 8:15280. [PMID: 30327482 PMCID: PMC6191451 DOI: 10.1038/s41598-018-33599-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/26/2018] [Indexed: 12/16/2022] Open
Abstract
Malaria is a disease with diverse symptoms depending on host immune status and pathogenicity of Plasmodium parasites. The continuous parasite growth within a host suggests mechanisms of immune evasion by the parasite and/or immune inhibition in response to infection. To identify pathways commonly inhibited after malaria infection, we infected C57BL/6 mice with four Plasmodium yoelii strains causing different disease phenotypes and 24 progeny of a genetic cross. mRNAs from mouse spleens day 1 and/or day 4 post infection (p.i.) were hybridized to a mouse microarray to identify activated or inhibited pathways, upstream regulators, and host genes playing an important role in malaria infection. Strong interferon responses were observed after infection with the N67 strain, whereas initial inhibition and later activation of hematopoietic pathways were found after infection with 17XNL parasite, showing unique responses to individual parasite strains. Inhibitions of pathways such as Th1 activation, dendritic cell (DC) maturation, and NFAT immune regulation were observed in mice infected with all the parasite strains day 4 p.i., suggesting universally inhibited immune pathways. As a proof of principle, treatment of N67-infected mice with antibodies against T cell receptors OX40 or CD28 to activate the inhibited pathways enhanced host survival. Controlled activation of these pathways may provide important strategies for better disease management and for developing an effective vaccine.
Collapse
Affiliation(s)
- Lu Xia
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA.,State Key Laboratory of Medical Genetics, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, The People's Republic of China
| | - Jian Wu
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Sittiporn Pattaradilokrat
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA.,Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Keyla Tumas
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Xiao He
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Yu-Chih Peng
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Timothy G Myers
- Genomic Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Carole A Long
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Rongfu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Xin-Zhuan Su
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA.
| |
Collapse
|
10
|
Dai J, Fang P, Saredy J, Xi H, Ramon C, Yang W, Choi ET, Ji Y, Mao W, Yang X, Wang H. Metabolism-associated danger signal-induced immune response and reverse immune checkpoint-activated CD40 + monocyte differentiation. J Hematol Oncol 2017; 10:141. [PMID: 28738836 PMCID: PMC5525309 DOI: 10.1186/s13045-017-0504-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/26/2017] [Indexed: 01/16/2023] Open
Abstract
Adaptive immunity is critical for disease progression and modulates T cell (TC) and antigen-presenting cell (APC) functions. Three signals were initially proposed for adaptive immune activation: signal 1 antigen recognition, signal 2 co-stimulation or co-inhibition, and signal 3 cytokine stimulation. In this article, we propose to term signal 2 as an immune checkpoint, which describes interactions of paired molecules leading to stimulation (stimulatory immune checkpoint) or inhibition (inhibitory immune checkpoint) of an immune response. We classify immune checkpoint into two categories: one-way immune checkpoint for forward signaling towards TC only, and two-way immune checkpoint for both forward and reverse signaling towards TC and APC, respectively. Recently, we and others provided evidence suggesting that metabolic risk factors (RF) activate innate and adaptive immunity, involving the induction of immune checkpoint molecules. We summarize these findings and suggest a novel theory, metabolism-associated danger signal (MADS) recognition, by which metabolic RF activate innate and adaptive immunity. We emphasize that MADS activates the reverse immune checkpoint which leads to APC inflammation in innate and adaptive immunity. Our recent evidence is shown that metabolic RF, such as uremic toxin or hyperhomocysteinemia, induced immune checkpoint molecule CD40 expression in monocytes (MC) and elevated serum soluble CD40 ligand (sCD40L) resulting in CD40+ MC differentiation. We propose that CD40+ MC is a novel pro-inflammatory MC subset and a reliable biomarker for chronic kidney disease severity. We summarize that CD40:CD40L immune checkpoint can induce TC and APC activation via forward stimulatory, reverse stimulatory, and TC contact-independent immune checkpoints. Finally, we modeled metabolic RF-induced two-way stimulatory immune checkpoint amplification and discussed potential signaling pathways including AP-1, NF-κB, NFAT, STAT, and DNA methylation and their contribution to systemic and tissue inflammation.
Collapse
Affiliation(s)
- Jin Dai
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian road, Hangzhou, 310006, Zhejiang, China.,Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Pu Fang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Jason Saredy
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Hang Xi
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Cueto Ramon
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - William Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Eric T Choi
- Department of Surgery, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 210029, China
| | - Wei Mao
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian road, Hangzhou, 310006, Zhejiang, China.
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.,Department of Pharmacology, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA. .,Department of Pharmacology, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
11
|
Shen Y, Liu Y, Wang XQ, Ke X, Kang HY, Hong SL. Association between TNFSF4 and BLK gene polymorphisms and susceptibility to allergic rhinitis. Mol Med Rep 2017; 16:3224-3232. [PMID: 28713926 PMCID: PMC5547929 DOI: 10.3892/mmr.2017.6954] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 04/12/2017] [Indexed: 02/06/2023] Open
Abstract
Allergic rhinitis (AR) is a common inflammatory disease of the upper airway. Recent evidence suggests that gene‑gene interactions between tumor necrosis factor receptor superfamily 4 (TNFSF4) and B cell lymphocyte kinase (BLK) may have a synergistic effect on T and B cells in determining immunologic aberration, via the nuclear factor‑κB pathway. The present study was performed to evaluate the potential association between specific single nucleotide polymorphisms (SNPs) in the TNFSF4 and BKL genes with susceptibility to AR in Chinese subjects. A population‑based case‑control study was performed in 600 Chinese AR patients and 700 controls. Blood was drawn for DNA extraction, and 9 SNPs (6 in TNFSF4 and 3 in BKL genes) were selected and genotyped. The TNFSF4 SNPs rs1234314 and rs1234315, and the BLK SNPs rs13277113 and rs1600249 were observed to occur in different frequencies between the AR patients and the controls. The CC (rs1234314, rs1234315) and AA (rs1600249, rs13277113) genotypes provided protective effects against AR, whereas the AG (rs13277113) genotype presented a risk factor for AR. The haplotypes ACC in the rs1234313‑rs1234314‑rs1234315 block and GA in the rs2254546‑rs13277113 block significantly decreased the risk of AR, whereas the GGT and AG haplotypes served protective roles. SNP interaction analysis further indicated that there may be synergistic effects among the selected sets of polymorphisms. The present study suggests a novel association between specific TNFSF4 and BLK gene polymorphisms and AR risk, highlighting their potential utility as genetic biomarkers for AR susceptibility in a Chinese Han population.
Collapse
Affiliation(s)
- Yang Shen
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yun Liu
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xiao-Qiang Wang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xia Ke
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hou-Yong Kang
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Su-Ling Hong
- Department of Otorhinolaryngology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
12
|
Brand A, Singer K, Koehl GE, Kolitzus M, Schoenhammer G, Thiel A, Matos C, Bruss C, Klobuch S, Peter K, Kastenberger M, Bogdan C, Schleicher U, Mackensen A, Ullrich E, Fichtner-Feigl S, Kesselring R, Mack M, Ritter U, Schmid M, Blank C, Dettmer K, Oefner PJ, Hoffmann P, Walenta S, Geissler EK, Pouyssegur J, Villunger A, Steven A, Seliger B, Schreml S, Haferkamp S, Kohl E, Karrer S, Berneburg M, Herr W, Mueller-Klieser W, Renner K, Kreutz M. LDHA-Associated Lactic Acid Production Blunts Tumor Immunosurveillance by T and NK Cells. Cell Metab 2016; 24:657-671. [PMID: 27641098 DOI: 10.1016/j.cmet.2016.08.011] [Citation(s) in RCA: 1172] [Impact Index Per Article: 130.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 04/20/2016] [Accepted: 08/19/2016] [Indexed: 10/21/2022]
Abstract
Elevated lactate dehydrogenase A (LDHA) expression is associated with poor outcome in tumor patients. Here we show that LDHA-associated lactic acid accumulation in melanomas inhibits tumor surveillance by T and NK cells. In immunocompetent C57BL/6 mice, tumors with reduced lactic acid production (Ldhalow) developed significantly slower than control tumors and showed increased infiltration with IFN-γ-producing T and NK cells. However, in Rag2-/-γc-/- mice, lacking lymphocytes and NK cells, and in Ifng-/- mice, Ldhalow and control cells formed tumors at similar rates. Pathophysiological concentrations of lactic acid prevented upregulation of nuclear factor of activated T cells (NFAT) in T and NK cells, resulting in diminished IFN-γ production. Database analyses revealed negative correlations between LDHA expression and T cell activation markers in human melanoma patients. Our results demonstrate that lactic acid is a potent inhibitor of function and survival of T and NK cells leading to tumor immune escape.
Collapse
Affiliation(s)
- Almut Brand
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Katrin Singer
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Gudrun E Koehl
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Marlene Kolitzus
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Gabriele Schoenhammer
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Annette Thiel
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Carina Matos
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christina Bruss
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sebastian Klobuch
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Katrin Peter
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Michael Kastenberger
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Ulrike Schleicher
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, 91054 Erlangen, Germany
| | - Evelyn Ullrich
- Department of Internal Medicine 5, Hematology/Oncology, University Hospital Erlangen, 91054 Erlangen, Germany; Cellular Immunology, Pediatric Stem Cell Transplantation and Immunology, Department for Children and Adolescents Medicine of the University Hospital Frankfurt, Goethe-University, 60590 Frankfurt, Germany
| | - Stefan Fichtner-Feigl
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany; Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Rebecca Kesselring
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Matthias Mack
- Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany; Department of Internal Medicine II - Nephrology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Uwe Ritter
- Institute of Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Maximilian Schmid
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Institute of Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Christian Blank
- Division of Immunology, the Netherlands Cancer Institute, Antoni van Leeuwenhoek Hospital, Amsterdam 1066CX, the Netherlands
| | - Katja Dettmer
- Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany
| | - Peter J Oefner
- Institute of Functional Genomics, University of Regensburg, 93053 Regensburg, Germany
| | - Petra Hoffmann
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Stefan Walenta
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Jacques Pouyssegur
- Institute of Research on Cancer and Aging, University of Nice-Sophia Antipolis, Centre A. Lacassagne, 06189 Nice, France; Centre Scientifique de Monaco (CSM), 98000 Monaco, Monaco
| | - Andreas Villunger
- Medical University Innsbruck, Biocenter, Division of Developmental Immunology, 6020 Innsbruck, Austria; Tyrolean Cancer Research Institute, 6020 Innsbruck, Austria
| | - André Steven
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology Halle/Saale, 06112 Halle, Germany
| | - Barbara Seliger
- Martin Luther University Halle-Wittenberg, Institute of Medical Immunology Halle/Saale, 06112 Halle, Germany
| | - Stephan Schreml
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Elisabeth Kohl
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Sigrid Karrer
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Mark Berneburg
- Department of Dermatology, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany
| | - Wolfgang Mueller-Klieser
- Institute of Pathophysiology, University Medical Center of the Johannes Gutenberg University Mainz, 55128 Mainz, Germany
| | - Kathrin Renner
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany
| | - Marina Kreutz
- Department of Internal Medicine III, University Hospital Regensburg, 93053 Regensburg, Germany; Regensburg Center for Interventional Immunology, University of Regensburg, 93053 Regensburg, Germany.
| |
Collapse
|
13
|
CD137 Regulates NFATc1 Expression in Mouse VSMCs through TRAF6/NF-κB p65 Signaling Pathway. Mediators Inflamm 2015; 2015:639780. [PMID: 26600673 PMCID: PMC4639649 DOI: 10.1155/2015/639780] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/06/2015] [Accepted: 10/08/2015] [Indexed: 12/29/2022] Open
Abstract
Our previous study proved that CD137-CD137L interaction can regulate the expression of NFATc1. Here, we investigated whether CD137 signaling regulates the expression of NFATc1 in mice VSMCs through TRAF6/NF-κB p65 pathway. Data shows that the CD137 expression can be stimulated by TNF-α in a time-dependent manner in mouse VSMCs. Knockdown of TRAF6 by siTRAF6 significantly attenuated agonist-CD137mAb induced increase of NF-κB p65 and NFATc1 in VSMCs. Pretreatment with a NF-κB inhibitor PDTC for 30 min inhibited the expression of p-p65 in both cytoplasm and nucleus in VSMCs. Thus, the protein level of NFATc1 can be suppressed through inhibition of p-p65. Finally, we also show that the levels of IL-2 and IL-6 can be increased by agonist-CD137 stimulation and decreased when NFATc1 was suppressed. Our data suggest that activated CD137 signaling regulates the expression of NFATc1 and its downstream factors through TRAF6/NF-κB p65 pathways in VSMCs. These findings provide a novel target for treatment of atherosclerosis.
Collapse
|
14
|
Ait-Oufella H, Sage AP, Mallat Z, Tedgui A. Adaptive (T and B cells) immunity and control by dendritic cells in atherosclerosis. Circ Res 2014; 114:1640-60. [PMID: 24812352 DOI: 10.1161/circresaha.114.302761] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Chronic inflammation in response to lipoprotein accumulation in the arterial wall is central in the development of atherosclerosis. Both innate and adaptive immunity are involved in this process. Adaptive immune responses develop against an array of potential antigens presented to effector T lymphocytes by antigen-presenting cells, especially dendritic cells. Functional analysis of the role of different T-cell subsets identified the Th1 responses as proatherogenic, whereas regulatory T-cell responses exert antiatherogenic activities. The effect of Th2 and Th17 responses is still debated. Atherosclerosis is also associated with B-cell activation. Recent evidence established that conventional B-2 cells promote atherosclerosis. In contrast, innate B-1 B cells offer protection through secretion of natural IgM antibodies. This review discusses the recent development in our understanding of the role of T- and B-cell subsets in atherosclerosis and addresses the role of dendritic cell subpopulations in the control of adaptive immunity.
Collapse
Affiliation(s)
- Hafid Ait-Oufella
- From INSERM UMR-S 970, Paris Cardiovascular Research Center (PARCC), Université Paris Descartes, Sorbonne Paris Cité, Paris, France (H.A.-O., Z.M., A.T.); Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint-Antoine, Paris, France (H.A.-O.); and Department of Medicine, University of Cambridge, Cambridge, United Kingdom (A.P.S., Z.M.)
| | | | | | | |
Collapse
|
15
|
Li Y, Yan J, Wu C, Wang Z, Yuan W, Wang D. CD137-CD137L interaction regulates atherosclerosis via cyclophilin A in apolipoprotein E-deficient mice. PLoS One 2014; 9:e88563. [PMID: 24520398 PMCID: PMC3919780 DOI: 10.1371/journal.pone.0088563] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 01/10/2014] [Indexed: 12/18/2022] Open
Abstract
Background Our previous studies showed that increased levels of cyclophilin A (CyPA) may be a valuable marker for predicting the severity of acute coronary syndromes and that interruption of CD137-CD137L interactions diminished the formation and progression of atherosclerosis in apolipoprotein E-deficient (ApoE−/−) mice. Here, we sought to determine whether the proinflammatory factor CyPA is involved in atherosclerosis regulated by CD137-CD137L interactions. Methods and Results A constrictive collar was placed around the right carotid arteries of ApoE−/− mice that were fed a high-fat diet to induce atherosclerotic plaque formation. After that, the mice were intraperitoneally injected with anti-CD137 or anti-CD137L in the presence or absence of the recombinant lentiviral vectors LVTHM-CyPA or pGC-FU-CyPA, respectively. Interestingly, activation of CD137-CD137L was negatively correlated with CyPA expression in vivo and in vitro. Stimulating CD137-CD137L interaction significantly increased CyPA, which was concurrent with the upregulation of proinflammatory cytokines, chemokines and matrix metalloproteinases and resulted in the promotion of atherosclerosis in ApoE-/- mice. Silencing CyPA could eliminate these effects, and restoration of CyPA effectively and consistently attenuated the atherosclerotic suppression phenotypes elicited by the blockade of CD137-CD137L. Conclusion These observations suggest that CD137-CD137L interactions mediated via regulation of CyPA contribute to the progression of atherosclerosis.
Collapse
Affiliation(s)
- Yuefeng Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jinchuan Yan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- * E-mail:
| | - Chao Wu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Dongqing Wang
- Department of Radiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
16
|
Smeets E, Meiler S, Lutgens E. Lymphocytic tumor necrosis factor receptor superfamily co-stimulatory molecules in the pathogenesis of atherosclerosis. Curr Opin Lipidol 2013; 24:518-24. [PMID: 24184937 DOI: 10.1097/mol.0000000000000025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW The role of lymphocytes in the chronic inflammatory disease atherosclerosis has emerged over the past decade. Co-stimulatory molecules of the heterogeneous tumor necrosis factor receptor superfamily play a pivotal role in lymphocyte activation, proliferation and differentiation. Here we describe the immune modulatory properties and mechanisms of four tumor necrosis factor receptor superfamily members in atherosclerosis. RECENT FINDINGS CD40/CD40L, OX40L/OX40, CD70/CD27 and CD137/CD137L are present in human atherosclerotic plaques and have shown strong immune modulatory functions in atherosclerosis, resulting in either atherogenic or atheroprotective effects in mouse models of atherosclerosis. SUMMARY Insight into the immune modulatory mechanisms of co-stimulatory interactions in atherosclerosis can contribute to clinical exploitation of these interactions in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Esther Smeets
- aDepartment of Medical Biochemistry, Academic Medical Center, Meibergdreef, Amsterdam, The Netherlands bInstitute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians University, Munich, ,Germany
| | | | | |
Collapse
|