1
|
Ciambarella BT, Almeida GSD, Dias DF, Trentin PG, Oliveira Santos TPD, Teixeira Ferreira TP, Arantes ACSD, Azevedo RBD, Fernandes AJM, Martins MA, E Silva PMR. TNF-alpha mediates airway hyperreactivity in silicotic mice: Effect of thalidomide treatment. Eur J Pharmacol 2025; 990:177263. [PMID: 39793879 DOI: 10.1016/j.ejphar.2025.177263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 12/12/2024] [Accepted: 01/08/2025] [Indexed: 01/13/2025]
Abstract
Inhalation of crystalline silica particles causes silicosis, which is a severe inflammatory lung disease that is associated with granulomatous and fibrotic responses. We investigated whether silica-induced silicosis might promote airway hyperreactivity (AHR) and the role of TNF-α and thalidomide in this process. Mice received an intranasal instillation of silica particles (1.25, 5, and 10 mg/mouse) and given methacholine on days 2, 7, and 28 after provocation or 5-HT challenges on day 7 after provocation. AHR was assessed using invasive whole-body plethysmography. Lung-tissue samples were collected for TNF-α measurements and histological analyses. Thalidomide was given orally from days 21-27 after silica administration. We found that following aerosolised methacholine or 5-HT treatment, a state of AHR was induced with silica-particle amounts of 5 and 10 mg/mouse, but not 1.25 mg/mouse. The effect was apparent within 2 days and remained for at least 28 days. Silica-particle amounts of 5 and 10 mg/mouse also induced significant granuloma response correlating with the silica required to induce AHR. In addition, a parallel was also observed between the elevation of lung tissue levels of TNF-α and AHR. Notably, silica-induced granulomatous and AHR responses were abolished in TNFR1-/- mice compared to wild-type mice. Moreover, the blockade of ongoing TNF-α generation by thalidomide prevented both events. Our findings suggest that exposure of mice to silica particles leads to a granulomatous lung response marked by non-specific AHR induced by TNF-α. In addition, the results indicate that thalidomide can control silica-induced pathological features of the lungs by blocking TNF-α generation.
Collapse
|
2
|
Sun J, Zhang Y, Zhang Q, Hu L, Zhao L, Wang H, Yuan Y, Niu H, Wang D, Zhang H, Liu J, Feng X, Su X, Qiu J, Sun J, Xu H, Zhang C, Wang K, Bi Y, Engleman EG, Shen L. Metabolic regulator LKB1 controls adipose tissue ILC2 PD-1 expression and mitochondrial homeostasis to prevent insulin resistance. Immunity 2024; 57:1289-1305.e9. [PMID: 38772366 DOI: 10.1016/j.immuni.2024.04.024] [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: 08/18/2023] [Revised: 02/06/2024] [Accepted: 04/25/2024] [Indexed: 05/23/2024]
Abstract
Adipose tissue group 2 innate lymphoid cells (ILC2s) help maintain metabolic homeostasis by sustaining type 2 immunity and promoting adipose beiging. Although impairment of the ILC2 compartment contributes to obesity-associated insulin resistance, the underlying mechanisms have not been elucidated. Here, we found that ILC2s in obese mice and humans exhibited impaired liver kinase B1 (LKB1) activation. Genetic ablation of LKB1 disrupted ILC2 mitochondrial metabolism and suppressed ILC2 responses, resulting in exacerbated insulin resistance. Mechanistically, LKB1 deficiency induced aberrant PD-1 expression through activation of NFAT, which in turn enhanced mitophagy by suppressing Bcl-xL expression. Blockade of PD-1 restored the normal functions of ILC2s and reversed obesity-induced insulin resistance in mice. Collectively, these data present the LKB1-PD-1 axis as a promising therapeutic target for the treatment of metabolic disease.
Collapse
Affiliation(s)
- Jiping Sun
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Youqin Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qingbing Zhang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lin Hu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Linfeng Zhao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hongdong Wang
- Department of Endocrinology, Drum Tower Hospital affiliated with Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
| | - Yue Yuan
- Department of Endocrinology, Drum Tower Hospital affiliated with Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
| | - Hongshen Niu
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dongdi Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Huasheng Zhang
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jianyue Liu
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xujiao Feng
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaohui Su
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Sun
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Heping Xu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Catherine Zhang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Kathleen Wang
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Yan Bi
- Department of Endocrinology, Drum Tower Hospital affiliated with Nanjing University Medical School, Branch of National Clinical Research Centre for Metabolic Diseases, Nanjing 210008, China
| | - Edgar G Engleman
- Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Lei Shen
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
3
|
Wang H, Cui J, Hao X, Guo L, Zhao J, Wang R, Liu H. Silicon, an important exposure marker in vivo in silicosis research. Int Arch Occup Environ Health 2021; 94:1513-1522. [PMID: 34110461 DOI: 10.1007/s00420-021-01729-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 03/27/2021] [Indexed: 11/25/2022]
Abstract
PURPOSE The degree of silicosis exposure is closely related to the progress of silicosis. At present, we use animal and human studies to explore whether silicon can be an important exposure marker in the development of silicosis. METHODS Rats were randomly divided into 2 groups: (1) controls; and (2) silicosis. Rats in the silicosis group were killed at 4, 8, 12, 16, 24 h, 3, 7, 14, 21, and 28 days. Hematoxylin-eosin (HE) and immunohistochemistry (IHC) were performed to observe the histomorphology of lung tissue. The expression levels of CC16 and SP-D were detected using ELISA kits. In addition, we conducted a population study. Workers who have been selected to work in an iron mine for more than 1 year as research objects. The population was divided into four groups: silicosis exposure group (workers exposed to silica dust for more than 1 year in an iron mine were selected); patients group (silicosis patients); observation group (evidence of disease not meeting formal diagnostic criteria) and control group. Both the levels of trace silicon in the urine and blood of rats and human subjects were measured with ICP-MS. RESULTS Serum levels of silicon were immediately increased in rats exposed to silicon dust. Similarly, our population study revealed that the silicon level in the silica exposure group and the observing group (exposed but no obvious symptoms) were significantly increased over that of the control group (P < 0.05). In subjects with extended exposure to silica, the serum and urine silicon level in exposed workers appeared to rapidly increase, reaching its peak in 1-5 years, followed by a gradual decline thereafter. Workers exposed to dust for less than 10 years were divided into subgroups by 2-year limit. The levels of serum silicon, urine silicon, TGF-β1, and TNF-α were significantly higher than that of control group. CONCLUSION Changes of the serum levels of silicon occurred earlier than the expression of cytokines such as TNF-α, TGF-β1, CC16, and SP-D. The level of silicon in workers rapidly increased after exposure to silica, and the change occurred before the expression of TGF-β1 and TNF-α. As a whole, the findings suggest that determining the level of silicon in vivo might be an effective exposure marker in the diagnosis and pathogenesis of silicosis.
Collapse
Affiliation(s)
- Hongli Wang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, China
| | - Jie Cui
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
| | - Xiaohui Hao
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, China
| | - Lingli Guo
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, China
| | - Jinyuan Zhao
- The Occupational Medicine Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Ruimin Wang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, China
| | - Heliang Liu
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China.
- Hebei Key Laboratory of Organ Fibrosis, North China University of Science and Technology, Tangshan, 063210, China.
| |
Collapse
|
4
|
Oflaz FE, Koshenov Z, Hirtl M, Rost R, Bachkoenig OA, Gottschalk B, Madreiter-Sokolowski CT, Malli R, Graier WF. Near-UV Light Induced ROS Production Initiates Spatial Ca 2+ Spiking to Fire NFATc3 Translocation. Int J Mol Sci 2021; 22:8189. [PMID: 34360954 PMCID: PMC8346968 DOI: 10.3390/ijms22158189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 01/19/2023] Open
Abstract
Ca2+-dependent gene regulation controls several functions to determine the fate of the cells. Proteins of the nuclear factor of activated T-cells (NFAT) family are Ca2+ sensitive transcription factors that control the cell growth, proliferation and insulin secretion in β-cells. Translocation of NFAT proteins to the nucleus occurs in a sequence of events that starts with activating calmodulin-dependent phosphatase calcineurin in a Ca2+-dependent manner, which dephosphorylates the NFAT proteins and leads to their translocation to the nucleus. Here, we examined the role of IP3-generating agonists and near-UV light in the induction of NFATc3 migration to the nucleus in the pancreatic β-cell line INS-1. Our results show that IP3 generation yields cytosolic Ca2+ rise and NFATc3 translocation. Moreover, near-UV light exposure generates reactive oxygen species (ROS), resulting in cytosolic Ca2+ spiking via the L-type Ca2+ channel and triggers NFATc3 translocation to the nucleus. Using the mitochondria as a Ca2+ buffering tool, we showed that ROS-induced cytosolic Ca2+ spiking, not the ROS themselves, was the triggering mechanism of nuclear import of NFATc3. Collectively, this study reveals the mechanism of near-UV light induced NFATc3 migration.
Collapse
Affiliation(s)
- Furkan E. Oflaz
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Zhanat Koshenov
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Martin Hirtl
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Rene Rost
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Olaf A. Bachkoenig
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Benjamin Gottschalk
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Corina T. Madreiter-Sokolowski
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
| | - Roland Malli
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| | - Wolfgang F. Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Neue Stiftingtalstraße 6/6, 8010 Graz, Austria; (F.E.O.); (Z.K.); (M.H.); (R.R.); (O.A.B.); (B.G.); (C.T.M.-S.); (R.M.)
- BioTechMed Graz, Mozartgasse 12/II, 8010 Graz, Austria
| |
Collapse
|
5
|
Zhao C, Yang S, Lu W, Liu J, Wei Y, Guo H, Zhang Y, Shi J. Increased NFATC4 Correlates With Poor Prognosis of AML Through Recruiting Regulatory T Cells. Front Genet 2020; 11:573124. [PMID: 33329712 PMCID: PMC7728998 DOI: 10.3389/fgene.2020.573124] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/27/2020] [Indexed: 12/13/2022] Open
Abstract
Despite that immune responses play important roles in acute myeloid leukemia (AML), immunotherapy is still not widely used in AML due to lack of an ideal target. Therefore, we identified key immune genes and cellular components in AML by an integrated bioinformatics analysis, trying to find potential targets for AML. Eighty-six differentially expressed immune genes (DEIGs) were identified from 751 differentially expressed genes (DEGs) between AML patients with fair prognosis and poor prognosis from the TCGA database. Among them, nine prognostic immune genes, including NCR2, NPDC1, KIR2DL4, KLC3, TWIST1, SNORD3B-1, NFATC4, XCR1, and LEFTY1, were identified by univariate Cox regression analysis. A multivariable prediction model was established based on prognostic immune genes. Kaplan–Meier survival curve analysis indicated that patients in the high-risk group had a shorter survival rate and higher mortality than those in the low-risk group (P < 0.001), indicating good effectiveness of the model. Furthermore, nuclear factors of activated T cells-4 (NFATC4) was recognized as the key immune gene identified by co-expression of differentially expressed transcription factors (DETFs) and prognostic immune genes. ATP-binding cassette transporters (ABC transporters) were the downstream KEGG pathway of NFATC4, identified by gene set variation analysis (GSVA) and gene set enrichment analysis (GSEA). To explore the immune responses NFATC4 was involved in, an immune gene set of T cell co-stimulation was identified by single-cell GSEA (ssGSEA) and Pearson correlation analysis, positively associated with NFATC4 in AML (R = 0.323, P < 0.001, positive). In order to find out the immune cell types affected by NFATC4, the CIBERSORT algorithm and Pearson correlation analysis were applied, and it was revealed that regulatory T cells (Tregs) have the highest correlation with NFATC4 (R = 0.526, P < 0.001, positive) in AML from 22 subsets of tumor-infiltrating immune cells. The results of this study were supported by multi-omics database validation. In all, our study indicated that NFATC4 was the key immune gene in AML poor prognosis through recruiting Tregs, suggesting that NFATC4 might serve as a new therapy target for AML.
Collapse
Affiliation(s)
- Chong Zhao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Shaoxin Yang
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Lu
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiali Liu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yanyu Wei
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hezhou Guo
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanjie Zhang
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Shi
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Yuan Y, Chen K, Chen X, Wang C, Qiu H, Cao Z, Song D, Sun Y, Guo J, Tickner J, Xu J, Zou J. Fumitremorgin C Attenuates Osteoclast Formation and Function via Suppressing RANKL-Induced Signaling Pathways. Front Pharmacol 2020; 11:238. [PMID: 32210820 PMCID: PMC7076231 DOI: 10.3389/fphar.2020.00238] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 02/21/2020] [Indexed: 12/26/2022] Open
Abstract
Excessive bone resorption conducted by osteoclasts is considered as the main cause of osteoclast-related bone diseases such as osteoporosis. Therefore, the suppression of excessive osteoclast formation and function is one of the strategies to treat osteoclast-related bone diseases. Fumitremorgin C (Fum) is a mycotoxin extracted from Aspergillus fumigatus. It has been shown to have extensive pharmacological properties, but its role in the treatment of osteoclast-related bone diseases remains unclear. In this study, we aim to find out whether Fum can inhibit the receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation and function. The results showed that Fum could significantly attenuate osteoclast formation and function at concentrations from 2.5 to 10 µM. The protein expression of bone resorption factors such as NFATc1, cathepsin K, V-ATPase-d2, and c-Fos was suppressed with the treatment of Fum at a concentration of 10 µM. In addition, Fum was also shown to suppress the activity of NF-κB, intracellular reactive oxygen species level, and MAPK pathway. Taken together, the present study showed that Fum could attenuate the formation and function of osteoclast via suppressing RANKL-induced signaling pathways, suggesting that Fum might be a potential novel drug in the treatment of osteoclast-related bone diseases.
Collapse
Affiliation(s)
- Yu Yuan
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- School of Physical Education and Sports Science, South China Normal University, Guangzhou, China
| | - Kai Chen
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xi Chen
- School of Sports Science, Wenzhou Medical University, Wenzhou, China
| | - Chao Wang
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Heng Qiu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Zhen Cao
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Dezhi Song
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Youqiang Sun
- Department of Orthopedics, First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianmin Guo
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jennifer Tickner
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Jun Zou
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
7
|
Chen K, Yuan Y, Wang Z, Song D, Zhao J, Cao Z, Chen J, Guo Q, Chen L, Tickner J, Xu J. Helvolic acid attenuates osteoclast formation and function via suppressing RANKL‐induced NFATc1 activation. J Cell Physiol 2018; 234:6477-6488. [DOI: 10.1002/jcp.27385] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 08/17/2018] [Indexed: 12/27/2022]
Affiliation(s)
- Kai Chen
- School of Biomedical Sciences, University of Western Australia Perth Western Australia Australia
| | - Yu Yuan
- School of Biomedical Sciences, University of Western Australia Perth Western Australia Australia
- School of Physical Education and Sports Science, South China Normal University Guangzhou Guangdong China
| | - Ziyi Wang
- School of Biomedical Sciences, University of Western Australia Perth Western Australia Australia
| | - Dezhi Song
- School of Biomedical Sciences, University of Western Australia Perth Western Australia Australia
- Research Centre for Regenerative Medicine, Guangxi Medical University Nanning Guangxi China
| | - Jinmin Zhao
- Research Centre for Regenerative Medicine, Guangxi Medical University Nanning Guangxi China
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University Guangxi China
- International Joint Laboratory on Regeneration of Bone and Soft Tissues, Guangxi Medical University Guangxi China
| | - Zhen Cao
- School of Biomedical Sciences, University of Western Australia Perth Western Australia Australia
- Department of Biomedical Materials Science Third Military Medical University Chongqing China
| | - Junhao Chen
- School of Biomedical Sciences, University of Western Australia Perth Western Australia Australia
| | - Qiang Guo
- School of Biomedical Sciences, University of Western Australia Perth Western Australia Australia
- Department of Spine Surgery Xiangya Hospital, Central South University Changsha China
| | - Li Chen
- School of Biomedical Sciences, University of Western Australia Perth Western Australia Australia
- Melbourne Medical School, University of Melbourne Parkville Victoria Australia
| | - Jennifer Tickner
- School of Biomedical Sciences, University of Western Australia Perth Western Australia Australia
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia Perth Western Australia Australia
| |
Collapse
|
8
|
Hwang E, Gao W, Xiao Y, Ngo HTT, Yi T. Helianthus annuus
L. flower prevents UVB‐induced photodamage in human dermal fibroblasts by regulating the MAPK/AP‐1, NFAT, and Nrf2 signaling pathways. J Cell Biochem 2018; 120:601-612. [DOI: 10.1002/jcb.27417] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 07/12/2018] [Indexed: 12/30/2022]
Affiliation(s)
- Eunson Hwang
- Graduate School of Biotechnology, Kyung Hee University Giheung‐gu Yongin‐si Gyeonggi‐do South Korea
| | - Wei Gao
- Graduate School of Biotechnology, Kyung Hee University Giheung‐gu Yongin‐si Gyeonggi‐do South Korea
| | - Yong‐kun Xiao
- Graduate School of Biotechnology, Kyung Hee University Giheung‐gu Yongin‐si Gyeonggi‐do South Korea
- Tianjin Ginkingsen Health Technology Co., Ltd Binhai Gaoxin Tianjin China
| | - Hien T. T. Ngo
- Graduate School of Biotechnology, Kyung Hee University Giheung‐gu Yongin‐si Gyeonggi‐do South Korea
| | - Tae‐Hoo Yi
- Graduate School of Biotechnology, Kyung Hee University Giheung‐gu Yongin‐si Gyeonggi‐do South Korea
| |
Collapse
|
9
|
Bonafini C, Marzotto M, Bellavite P. In vitro effects of Zinc in soluble and homeopathic formulations on macrophages and astrocytes. HOMEOPATHY 2017; 106:103-113. [DOI: 10.1016/j.homp.2017.03.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 02/27/2017] [Accepted: 03/08/2017] [Indexed: 12/13/2022]
Abstract
Zinc is an important metal in body homeostasis. Zinc in soluble form (Zn2+) and homeopathic Zincum metallicum were tested in macrophages and astrocytes in order to investigate its potential toxic or therapeutic effects. We evaluated cell viability (WST assay), cytokine production such as tumour necrosis factor alpha (TNF-α) and interleukin 10 (IL-10) by enzyme-linked immunosorbent assay (ELISA) and nitric oxide release by Griess reaction. The effect of zinc-depletion and high zinc pre-treatments on the cell adaptation capability was also investigated. In THP-1 macrophage cell line and in human primary macrophages, Zn2+ at sub-toxic doses (30 μM) caused stimulation of TNF-α and IL-10 with different dynamics reaching the maximum peak at the zinc concentration 100 μM, before the cell death. Highest doses (300 μM) impaired dramatically cell vitality. Similar effects on cell viability were obtained also in C6 astrocytes, where Zn2+ slightly increased the nitric oxide release only in cells activated by one of the pro-inflammatory stimuli used in our cellular model (interferon gamma plus TNF-α). Zinc depletion markedly reduced IL-10 production and cell viability. Zincum metallicum did not cause toxicity in any cell type and showed some small stimulation in WST assay that was statistically significant in a few experimental conditions.
Collapse
|
10
|
Jernigan NL, Resta TC, Gonzalez Bosc LV. Altered Redox Balance in the Development of Chronic Hypoxia-induced Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 967:83-103. [PMID: 29047083 DOI: 10.1007/978-3-319-63245-2_7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Normally, the pulmonary circulation is maintained in a low-pressure, low-resistance state with little resting tone. Pulmonary arteries are thin-walled and rely heavily on pulmonary arterial distension and recruitment for reducing pulmonary vascular resistance when cardiac output is elevated. Under pathophysiological conditions, however, active vasoconstriction and vascular remodeling lead to enhanced pulmonary vascular resistance and subsequent pulmonary hypertension (PH). Chronic hypoxia is a critical pathological factor associated with the development of PH resulting from airway obstruction (COPD, sleep apnea), diffusion impairment (interstitial lung disease), developmental lung abnormalities, or high altitude exposure (World Health Organization [WHO]; Group III). The rise in pulmonary vascular resistance increases right heart afterload causing right ventricular hypertrophy that can ultimately lead to right heart failure in patients with chronic lung disease. PH is typically characterized by diminished paracrine release of vasodilators, antimitogenic factors, and antithrombotic factors (e.g., nitric oxide and protacyclin) and enhanced production of vasoconstrictors and mitogenic factors (e.g., reactive oxygen species and endothelin-1) from the endothelium and lung parenchyma. In addition, phenotypic changes to pulmonary arterial smooth muscle cells (PASMC), including alterations in Ca2+ homeostasis, Ca2+ sensitivity, and activation of transcription factors are thought to play prominent roles in the development of both vasoconstrictor and arterial remodeling components of hypoxia-associated PH. These changes in PASMC function are briefly reviewed in Sect. 1 and the influence of altered reactive oxygen species homeostasis on PASMC function discussed in Sects. 2-4.
Collapse
Affiliation(s)
- Nikki L Jernigan
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Thomas C Resta
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA
| | - Laura V Gonzalez Bosc
- Department Cell Biology and Physiology, Vascular Physiology Group, University of New Mexico Health Sciences Center, University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
11
|
Two Distinct Isoforms of Matrix Metalloproteinase-2 Are Associated with Human Delayed Kidney Graft Function. PLoS One 2015; 10:e0136276. [PMID: 26379248 PMCID: PMC4574928 DOI: 10.1371/journal.pone.0136276] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 08/01/2015] [Indexed: 11/19/2022] Open
Abstract
Delayed graft function (DGF) is a frequent complication of renal transplantation, particularly in the setting of transplantation of kidneys derived from deceased donors and expanded-criteria donors. DGF results from tubular epithelial cell injury and has immediate and long term consequences. These include requirement for post-transplantation dialysis, increased incidence of acute rejection, and poorer long-term outcomes. DGF represents one of the clearest clinical examples of renal acute ischemia/reperfusion injury. Experimental studies have demonstrated that ischemia/reperfusion injury induces the synthesis of the full length secreted isoform of matrix metalloproteinase-2 (FL-MMP-2), as well as an intracellular N-terminal truncated MMP-2 isoform (NTT-MMP-2) that initiates an innate immune response. We hypothesized that the two MMP-2 isoforms mediate tubular epithelial cell injury in DGF. Archival renal biopsy sections from 10 protocol biopsy controls and 41 cases with a clinical diagnosis of DGF were analyzed for the extent of tubular injury, expression of the FL-MMP-2 and NTT-MMP-2 isoforms by immunohistochemistry (IHC), in situ hybridization, and qPCR to determine isoform abundance. Differences in transcript abundance were related to tubular injury score. Markers of MMP-2-mediated injury included TUNEL staining and assessment of peritubular capillary density. There was a clear relationship between tubular epithelial cell expression of both FL-MMP-2 and NTT-MMP-2 IHC with the extent of tubular injury. The MMP-2 isoforms were detected in the same tubular segments and were present at sites of tubular injury. qPCR demonstrated highly significant increases in both the FL-MMP-2 and NTT-MMP-2 transcripts. Statistical analysis revealed highly significant associations between FL-MMP-2 and NTT-MMP-2 transcript abundance and the extent of tubular injury, with NTT-MMP-2 having the strongest association. We conclude that two distinct MMP-2 isoforms are associated with tubular injury in DGF and offer novel therapeutic targets for the prevention of this disorder.
Collapse
|
12
|
Wang L, Hitron JA, Wise JTF, Son YO, Roy RV, Kim D, Dai J, Pratheeshkumar P, Zhang Z, Xu M, Luo J, Shi X. Ethanol enhances arsenic-induced cyclooxygenase-2 expression via both NFAT and NF-κB signalings in colorectal cancer cells. Toxicol Appl Pharmacol 2015. [PMID: 26220687 DOI: 10.1016/j.taap.2015.07.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Arsenic is a known carcinogen to humans, and chronic exposure to environmental arsenic is a worldwide health concern. As a dietary factor, ethanol carries a well-established risk for malignancies, but the effects of co-exposure to arsenic and ethanol on tumor development are not well understood. In the present study, we hypothesized that ethanol would enhance the function of an environmental carcinogen such as arsenic through increase in COX-2 expression. Our in vitro results show that ethanol enhanced arsenic-induced COX-2 expression. We also show that the increased COX-2 expression associates with intracellular ROS generation, up-regulated AKT signaling, with activation of both NFAT and NF-κB pathways. We demonstrate that antioxidant enzymes have an inhibitory effect on arsenic/ethanol-induced COX-2 expression, indicating that the responsive signaling pathways from co-exposure to arsenic and ethanol relate to ROS generation. In vivo results also show that co-exposure to arsenic and ethanol increased COX-2 expression in mice. We conclude that ethanol enhances arsenic-induced COX-2 expression in colorectal cancer cells via both the NFAT and NF-κB pathways. These results imply that, as a common dietary factor, ethanol ingestion may be a compounding risk factor for arsenic-induced carcinogenesis/cancer development.
Collapse
Affiliation(s)
- Lei Wang
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - John Andrew Hitron
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - James T F Wise
- Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Young-Ok Son
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Ram Vinod Roy
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Donghern Kim
- Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jin Dai
- Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Poyil Pratheeshkumar
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Zhuo Zhang
- Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Mei Xu
- Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Jia Luo
- Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Xianglin Shi
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
13
|
Liu YN, Zha WJ, Ma Y, Chen FF, Zhu W, Ge A, Zeng XN, Huang M. Galangin attenuates airway remodelling by inhibiting TGF-β1-mediated ROS generation and MAPK/Akt phosphorylation in asthma. Sci Rep 2015; 5:11758. [PMID: 26156213 PMCID: PMC4496730 DOI: 10.1038/srep11758] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 06/05/2015] [Indexed: 02/07/2023] Open
Abstract
Galangin, a natural flavonol, has attracted much attention for its potential anti-inflammatory properties. However, its role in the regulation of airway remodelling in asthma has not been explored. The present study aimed to elucidate the effects of galangin on chronic inflammation and airway remodelling and to investigate the underlying mechanisms both in vivo and in vitro. Ovalbumin (OVA)-sensitised mice were administered with galangin 30 min before challenge. Our results showed that severe inflammatory responses and airway remodelling occurred in OVA-induced mice. Treatment with galangin markedly attenuated the leakage of inflammatory cells into bronchoalveolar lavage fluid (BALF) and decreased the level of OVA-specific IgE in serum. Galangin significantly inhibited goblet cell hyperplasia, collagen deposition and α-SMA expression. Lowered level of TGF-β1 and suppressed expression of VEGF and MMP-9 were observed in BALF or lung tissue, implying that galangin has an optimal anti-remodelling effect in vivo. Consistently, the TGF-β1-induced proliferation of airway smooth muscle cells was reduced by galangin in vitro, which might be due to the alleviation of ROS levels and inhibition of MAPK pathway. Taken together, the present findings highlight a novel role for galangin as a promising anti-remodelling agent in asthma, which likely involves the TGF-β1-ROS-MAPK pathway.
Collapse
Affiliation(s)
- Ya-Nan Liu
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wang-Jian Zha
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan Ma
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fei-Fei Chen
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wen Zhu
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ai Ge
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiao-Ning Zeng
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Mao Huang
- Department of Respiratory Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Wei J, Du K, Cai Q, Ma L, Jiao Z, Tan J, Xu Z, Li J, Luo W, Chen J, Gao J, Zhang D, Huang C. Lead induces COX-2 expression in glial cells in a NFAT-dependent, AP-1/NFκB-independent manner. Toxicology 2014; 325:67-73. [PMID: 25193092 DOI: 10.1016/j.tox.2014.08.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 08/28/2014] [Accepted: 08/30/2014] [Indexed: 12/22/2022]
Abstract
Epidemiologic studies have provided solid evidence for the neurotoxic effect of lead for decades of years. In view of the fact that children are more vulnerable to the neurotoxicity of lead, lead exposure has been an urgent public health concern. The modes of action of lead neurotoxic effects include disturbance of neurotransmitter storage and release, damage of mitochondria, as well as induction of apoptosis in neurons, cerebrovascular endothelial cells, astroglia and oligodendroglia. Our studies here, from a novel point of view, demonstrates that lead specifically caused induction of COX-2, a well known inflammatory mediator in neurons and glia cells. Furthermore, we revealed that COX-2 was induced by lead in a transcription-dependent manner, which relayed on transcription factor NFAT, rather than AP-1 and NFκB, in glial cells. Considering the important functions of COX-2 in mediation of inflammation reaction and oxidative stress, our studies here provide a mechanistic insight into the understanding of lead-associated inflammatory neurotoxicity effect via activation of pro-inflammatory NFAT3/COX-2 axis.
Collapse
Affiliation(s)
- Jinlong Wei
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Kejun Du
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA; Department of Occupational and Environmental Health and Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Qinzhen Cai
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Lisha Ma
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhenzhen Jiao
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Jinrong Tan
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Zhou Xu
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Jingxia Li
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA
| | - Wenjin Luo
- Department of Occupational and Environmental Health and Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Jingyuan Chen
- Department of Occupational and Environmental Health and Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an 710032, China
| | - Jimin Gao
- Zhejiang Provincial Key Laboratory for Technology & Application of Model Organisms, School of Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Dongyun Zhang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA.
| | - Chuanshu Huang
- Nelson Institute of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987, USA.
| |
Collapse
|
15
|
Chung YH, Jang Y, Choi B, Song DH, Lee EJ, Kim SM, Song Y, Kang SW, Yoon SY, Chang EJ. Beclin-1 Is Required for RANKL-Induced Osteoclast Differentiation. J Cell Physiol 2014; 229:1963-71. [DOI: 10.1002/jcp.24646] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 04/11/2014] [Indexed: 12/31/2022]
Affiliation(s)
- Yeon-Ho Chung
- Department of Biomedical Sciences; University of Ulsan College of Medicine, Asan Medical Center; Seoul Korea
- Department of Anatomy and Cell Biology; Cell Dysfunction Research Center and BMIT, University of Ulsan College of Medicine; Seoul Korea
| | - Youngsaeng Jang
- Department of Biomedical Sciences; University of Ulsan College of Medicine, Asan Medical Center; Seoul Korea
- Department of Anatomy and Cell Biology; Cell Dysfunction Research Center and BMIT, University of Ulsan College of Medicine; Seoul Korea
| | - Bongkun Choi
- Department of Biomedical Sciences; University of Ulsan College of Medicine, Asan Medical Center; Seoul Korea
- Department of Anatomy and Cell Biology; Cell Dysfunction Research Center and BMIT, University of Ulsan College of Medicine; Seoul Korea
| | - Da-Hyun Song
- Department of Biomedical Sciences; University of Ulsan College of Medicine, Asan Medical Center; Seoul Korea
- Department of Anatomy and Cell Biology; Cell Dysfunction Research Center and BMIT, University of Ulsan College of Medicine; Seoul Korea
| | - Eun-Jin Lee
- Department of Biomedical Sciences; University of Ulsan College of Medicine, Asan Medical Center; Seoul Korea
- Department of Anatomy and Cell Biology; Cell Dysfunction Research Center and BMIT, University of Ulsan College of Medicine; Seoul Korea
| | - Sang-Min Kim
- Department of Biomedical Sciences; University of Ulsan College of Medicine, Asan Medical Center; Seoul Korea
- Department of Anatomy and Cell Biology; Cell Dysfunction Research Center and BMIT, University of Ulsan College of Medicine; Seoul Korea
| | - Youngsup Song
- Department of Biomedical Sciences; University of Ulsan College of Medicine, Asan Medical Center; Seoul Korea
- Department of Anatomy and Cell Biology; Cell Dysfunction Research Center and BMIT, University of Ulsan College of Medicine; Seoul Korea
| | - Sang-Wook Kang
- Department of Biomedical Sciences; University of Ulsan College of Medicine, Asan Medical Center; Seoul Korea
- Department of Anatomy and Cell Biology; Cell Dysfunction Research Center and BMIT, University of Ulsan College of Medicine; Seoul Korea
| | - Seung-Yong Yoon
- Department of Anatomy and Cell Biology; Cell Dysfunction Research Center and BMIT, University of Ulsan College of Medicine; Seoul Korea
| | - Eun-Ju Chang
- Department of Biomedical Sciences; University of Ulsan College of Medicine, Asan Medical Center; Seoul Korea
- Department of Anatomy and Cell Biology; Cell Dysfunction Research Center and BMIT, University of Ulsan College of Medicine; Seoul Korea
| |
Collapse
|
16
|
Ramiro-Diaz JM, Giermakowska W, Weaver JM, Jernigan NL, Gonzalez Bosc LV. Mechanisms of NFATc3 activation by increased superoxide and reduced hydrogen peroxide in pulmonary arterial smooth muscle. Am J Physiol Cell Physiol 2014; 307:C928-38. [PMID: 25163518 DOI: 10.1152/ajpcell.00244.2014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
We recently demonstrated increased superoxide (O2(·-)) and decreased H2O2 levels in pulmonary arteries of chronic hypoxia-exposed wild-type and normoxic superoxide dismutase 1 (SOD1) knockout mice. We also showed that this reciprocal change in O2(·-) and H2O2 is associated with elevated activity of nuclear factor of activated T cells isoform c3 (NFATc3) in pulmonary arterial smooth muscle cells (PASMC). This suggests that an imbalance in reactive oxygen species levels is required for NFATc3 activation. However, how such imbalance activates NFATc3 is unknown. This study evaluated the importance of O2(·-) and H2O2 in the regulation of NFATc3 activity. We tested the hypothesis that an increase in O2(·-) enhances actin cytoskeleton dynamics and a decrease in H2O2 enhances intracellular Ca(2+) concentration, contributing to NFATc3 nuclear import and activation in PASMC. We demonstrate that, in PASMC, endothelin-1 increases O2(·-) while decreasing H2O2 production through the decrease in SOD1 activity without affecting SOD protein levels. We further demonstrate that O2(·-) promotes, while H2O2 inhibits, NFATc3 activation in PASMC. Additionally, increased O2(·-)-to-H2O2 ratio activates NFATc3, even in the absence of a Gq protein-coupled receptor agonist. Furthermore, O2(·-)-dependent actin polymerization and low intracellular H2O2 concentration-dependent increases in intracellular Ca(2+) concentration contribute to NFATc3 activation. Together, these studies define important and novel regulatory mechanisms of NFATc3 activation in PASMC by reactive oxygen species.
Collapse
Affiliation(s)
- Juan Manuel Ramiro-Diaz
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Wieslawa Giermakowska
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - John M Weaver
- Center of Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico; and Department of Pharmaceutical Sciences, College of Pharmacy, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Nikki L Jernigan
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico
| | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, New Mexico;
| |
Collapse
|
17
|
Dash SK, Ghosh T, Roy S, Chattopadhyay S, Das D. Zinc sulfide nanoparticles selectively induce cytotoxic and genotoxic effects on leukemic cells: involvement of reactive oxygen species and tumor necrosis factor alpha. J Appl Toxicol 2014; 34:1130-44. [DOI: 10.1002/jat.2976] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2013] [Revised: 11/03/2013] [Accepted: 11/17/2013] [Indexed: 02/06/2023]
Affiliation(s)
- Sandeep Kumar Dash
- Immunology and Microbiology Laboratory, Department of Human Physiology with Community Health; Vidyasagar University; Midnapore-721 102 West Bengal India
| | - Totan Ghosh
- Department of Chemistry; University of Calcutta; 92, A. P. C. Road Kolkata 700 009 India
| | - Soumyabrata Roy
- Department of Chemistry; Indian Institute of Technology; Kharagpur Pin-721302 India
| | - Sourav Chattopadhyay
- Immunology and Microbiology Laboratory, Department of Human Physiology with Community Health; Vidyasagar University; Midnapore-721 102 West Bengal India
| | - Debasis Das
- Department of Chemistry; University of Calcutta; 92, A. P. C. Road Kolkata 700 009 India
| |
Collapse
|
18
|
Zhang L, He YL, Li QZ, Hao XH, Zhang ZF, Yuan JX, Bai YP, Jin YL, Liu N, Chen G, Yun X, Yao SQ. N-acetylcysteine alleviated silica-induced lung fibrosis in rats by down-regulation of ROS and mitochondrial apoptosis signaling. Toxicol Mech Methods 2014; 24:212-9. [PMID: 24392833 DOI: 10.3109/15376516.2013.879974] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Reactive oxygen species (ROS) is a normal metabolic product of cellular respiration, but too much ROS can induce cell apoptosis. Here, we used N-acetylcysteine (NAC) to inhibit ROS activity to explore the effects of NAC on silica-induced pulmonary fibrosis in rats and provide evidence for study on the mechanism of silicosis. 24 adult male Sprague-Dawley rats weighing 180-220 g were randomly divided into three groups with eight rats in each group. Silicosis model group and NAC group were adopted non-tracheal exposure method of disposable intrapulmonary injection of 50 g/L, silica suspension 1 mL to establish animal silicosis model, NAC group treated with 600 mg/kg NAC by gavage from the right day of modeling, all animals were sacrificed after 28 days. The level of ROS contents and mitochondrial transmembrane potential changes of AM, the mRNA expression level of type I and type III procollagen, cytochrome C, cysteinyl aspartate specific protease-9 and caspase-3 were detected. The severity of pathological changes and pulmonary fibrosis were observed by pathologic specimens. It was showed that ROS contents and MTP changes were lower in the NAC group compared with the silicosis model group, other indexes were lower in the NAC group than the model group, but higher than those of the control group, the degree of lung fibrotic lesions observed from the pathological slices showed the same trend. These data indicated that NAC can reduce ROS content of AM in silica exposure rats, the mitochondrial apoptosis pathway can also be inhibited, the severity of pulmonary fibrosis alleviated as a result.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Occupational and Environmental Health, School of Public Health, Hebei United University , Tangshan, Hebei , China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Wang X, Yun JW, Lei XG. Glutathione peroxidase mimic ebselen improves glucose-stimulated insulin secretion in murine islets. Antioxid Redox Signal 2014; 20:191-203. [PMID: 23795780 PMCID: PMC3887434 DOI: 10.1089/ars.2013.5361] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
AIMS Glutathione peroxidase (GPX) mimic ebselen and superoxide dismutase (SOD) mimic copper diisopropylsalicylate (CuDIPs) were used to rescue impaired glucose-stimulated insulin secretion (GSIS) in islets of GPX1 and(or) SOD1-knockout mice. RESULTS Ebselen improved GSIS in islets of all four tested genotypes. The rescue in the GPX1 knockout resulted from a coordinated transcriptional regulation of four key GSIS regulators and was mediated by the peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α)-mediated signaling pathways. In contrast, CuDIPs improved GSIS only in the SOD1 knockout and suppressed gene expression of the PGC-1α pathway. INNOVATION Islets from the GPX1 and(or) SOD1 knockout mice provided metabolically controlled intracellular hydrogen peroxide (H2O2) and superoxide conditions for the present study to avoid confounding effects. Bioinformatics analyses of gene promoters and expression profiles guided the search for upstream signaling pathways to link the ebselen-initiated H2O2 scavenging to downstream key events of GSIS. The RNA interference was applied to prove PGC-1α as the main mediator for that link. CONCLUSION Our study revealed a novel metabolic use and clinical potential of ebselen in rescuing GSIS in the GPX1-deficient islets and mice, along with distinct differences between the GPX and SOD mimics in this regard. These findings highlight the necessities and opportunities of discretional applications of various antioxidant enzyme mimics in treating insulin secretion disorders. REBOUND TRACK: This work was rejected during standard peer review and rescued by Rebound Peer Review (Antioxid Redox Signal 16: 293-296, 2012) with the following serving as open reviewers: Regina Brigelius-Flohe, Vadim Gladyshev, Dexing Hou, and Holger Steinbrenner.
Collapse
Affiliation(s)
- Xinhui Wang
- 1 Department of Animal Science, Cornell University , Ithaca, New York
| | | | | |
Collapse
|
20
|
Fric J, Zelante T, Ricciardi-Castagnoli P. Phagocytosis of Particulate Antigens - All Roads Lead to Calcineurin/NFAT Signaling Pathway. Front Immunol 2014; 4:513. [PMID: 24409187 PMCID: PMC3885923 DOI: 10.3389/fimmu.2013.00513] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 12/27/2013] [Indexed: 11/25/2022] Open
Affiliation(s)
- Jan Fric
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR) , Biopolis , Singapore
| | - Teresa Zelante
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR) , Biopolis , Singapore
| | - Paola Ricciardi-Castagnoli
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (ASTAR) , Biopolis , Singapore
| |
Collapse
|
21
|
Aggarwal S, Gross CM, Sharma S, Fineman JR, Black SM. Reactive oxygen species in pulmonary vascular remodeling. Compr Physiol 2013; 3:1011-34. [PMID: 23897679 DOI: 10.1002/cphy.c120024] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The pathogenesis of pulmonary hypertension is a complex multifactorial process that involves the remodeling of pulmonary arteries. This remodeling process encompasses concentric medial thickening of small arterioles, neomuscularization of previously nonmuscular capillary-like vessels, and structural wall changes in larger pulmonary arteries. The pulmonary arterial muscularization is characterized by vascular smooth muscle cell hyperplasia and hypertrophy. In addition, in uncontrolled pulmonary hypertension, the clonal expansion of apoptosis-resistant endothelial cells leads to the formation of plexiform lesions. Based upon a large number of studies in animal models, the three major stimuli that drive the vascular remodeling process are inflammation, shear stress, and hypoxia. Although, the precise mechanisms by which these stimuli impair pulmonary vascular function and structure are unknown, reactive oxygen species (ROS)-mediated oxidative damage appears to play an important role. ROS are highly reactive due to their unpaired valence shell electron. Oxidative damage occurs when the production of ROS exceeds the quenching capacity of the antioxidant mechanisms of the cell. ROS can be produced from complexes in the cell membrane (nicotinamide adenine dinucleotide phosphate-oxidase), cellular organelles (peroxisomes and mitochondria), and in the cytoplasm (xanthine oxidase). Furthermore, low levels of tetrahydrobiopterin (BH4) and L-arginine the rate limiting cofactor and substrate for endothelial nitric oxide synthase (eNOS), can cause the uncoupling of eNOS, resulting in decreased NO production and increased ROS production. This review will focus on the ROS generation systems, scavenger antioxidants, and oxidative stress associated alterations in vascular remodeling in pulmonary hypertension.
Collapse
Affiliation(s)
- Saurabh Aggarwal
- Pulmonary Disease Program, Vascular Biology Center, Georgia Health Sciences University, Augusta, Georgia, USA
| | | | | | | | | |
Collapse
|
22
|
Friedman JK, Nitta CH, Henderson KM, Codianni SJ, Sanchez L, Ramiro-Diaz JM, Howard TA, Giermakowska W, Kanagy NL, Gonzalez Bosc LV. Intermittent hypoxia-induced increases in reactive oxygen species activate NFATc3 increasing endothelin-1 vasoconstrictor reactivity. Vascul Pharmacol 2013; 60:17-24. [PMID: 24239798 DOI: 10.1016/j.vph.2013.11.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 10/14/2013] [Accepted: 11/05/2013] [Indexed: 02/09/2023]
Abstract
Sleep apnea (SA), defined as intermittent respiratory arrest during sleep, is associated with increased incidence of hypertension, peripheral vascular disease, stroke, and sudden cardiac death. We have shown that intermittent hypoxia with CO2 supplementation (IH), a model for SA, increases blood pressure and circulating ET-1 levels, upregulates lung pre-pro ET-1 mRNA, increases vasoconstrictor reactivity to ET-1 in rat small mesenteric arteries (MA) and increases vascular reactive oxygen species (ROS). NFAT activity is increased in the aorta (AO) and MA of mice exposed to IH in an ET-1-dependent manner, and the genetic ablation of the isoform NFATc3 prevents IH-induced hypertension. We hypothesized that IH causes an increase in arterial ROS generation, which activates NFATc3 to increase vasoconstrictor reactivity to ET-1. In support of our hypothesis, we found that IH increases ROS in AO and MA. In vivo administration of the SOD mimetic tempol during IH exposure prevents IH-induced increases in NFAT activity in mouse MA and AO. We found that IH causes an NFATc3-dependent increase in vasoconstrictor reactivity to ET-1, accompanied by an increase in vessel wall [Ca²⁺]. Our results indicate that IH exposure causes an increase in arterial ROS to activate NFATc3, which then increases vasoconstrictor reactivity and Ca²⁺ response to ET-1. These studies highlight a novel regulatory pathway, and demonstrate the potential clinical relevance of NFAT inhibition to prevent hypertension in SA patients.
Collapse
Affiliation(s)
- J K Friedman
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - C H Nitta
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - K M Henderson
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - S J Codianni
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - L Sanchez
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - J M Ramiro-Diaz
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - T A Howard
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - W Giermakowska
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - N L Kanagy
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - L V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| |
Collapse
|
23
|
Ramiro-Diaz JM, Nitta CH, Maston LD, Codianni S, Giermakowska W, Resta TC, Gonzalez Bosc LV. NFAT is required for spontaneous pulmonary hypertension in superoxide dismutase 1 knockout mice. Am J Physiol Lung Cell Mol Physiol 2013; 304:L613-25. [PMID: 23475768 PMCID: PMC3652021 DOI: 10.1152/ajplung.00408.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 03/04/2013] [Indexed: 01/29/2023] Open
Abstract
Elevated reactive oxygen species are implicated in pulmonary hypertension (PH). Superoxide dismutase (SOD) limits superoxide bioavailability, and decreased SOD activity is associated with PH. A decrease in SOD activity is expected to increase superoxide and reduce hydrogen peroxide levels. Such an imbalance of superoxide/hydrogen peroxide has been implicated as a mediator of nuclear factor of activated T cells (NFAT) activation in epidermal cells. We have shown that NFATc3 is required for chronic hypoxia-induced PH. However, it is unknown whether NFATc3 is activated in the pulmonary circulation in a mouse model of decreased SOD1 activity and whether this leads to PH. Therefore, we hypothesized that an elevated pulmonary arterial superoxide/hydrogen peroxide ratio activates NFATc3, leading to PH. We found that SOD1 knockout (KO) mice have elevated pulmonary arterial wall superoxide and decreased hydrogen peroxide levels compared with wild-type (WT) littermates. Right ventricular systolic pressure (RVSP) was elevated in SOD1 KO and was associated with pulmonary arterial remodeling. Vasoreactivity to endothelin-1 was also greater in SOD1 KO vs. WT mice. NFAT activity and NFATc3 nuclear localization were increased in pulmonary arteries from SOD1 KO vs. WT mice. Administration of A-285222 (selective NFAT inhibitor) decreased RVSP, arterial wall thickness, vasoreactivity, and NFAT activity in SOD1 KO mice to WT levels. The SOD mimetic, tempol, also reduced NFAT activity, NFATc3 nuclear localization, and RVSP to WT levels. These findings suggest that an elevated superoxide/hydrogen peroxide ratio activates NFAT in pulmonary arteries, which induces vascular remodeling and increases vascular reactivity leading to PH.
Collapse
Affiliation(s)
- Juan Manuel Ramiro-Diaz
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Tmem64 modulates calcium signaling during RANKL-mediated osteoclast differentiation. Cell Metab 2013; 17:249-60. [PMID: 23395171 PMCID: PMC3569742 DOI: 10.1016/j.cmet.2013.01.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 12/03/2012] [Accepted: 01/08/2013] [Indexed: 12/14/2022]
Abstract
Osteoclast maturation and function primarily depend on receptor activator of NF-κB ligand (RANKL)-mediated induction of nuclear factor of activated T cells c1 (NFATc1), which is further activated via increased intracellular calcium ([Ca(2+)](i)) oscillation. However, the coordination mechanism that mediates Ca(2+) oscillation during osteoclastogenesis remains ill defined. Here, we identified transmembrane protein 64 (Tmem64) as a regulator of Ca(2+) oscillation during osteoclastogenesis. We found that Tmem64-deficient mice exhibit increased bone mass due in part to impaired osteoclast formation. Using in vitro osteoclast culture systems, we show here that Tmem64 interacts with sarcoplasmic endoplasmic reticulum Ca(2+) ATPase 2 (SERCA2) and modulates its activity. Consequently, Tmem64 deficiency significantly diminishes RANKL-induced [Ca(2+)](i) oscillation, which results in reduced Ca(2+)/calmodulin-dependent protein kinases (CaMK) IV and mitochondrial ROS, both of which contribute to achieving the CREB activity necessary for osteoclast formation. These data demonstrate that Tmem64 is a positive modulator of osteoclast differentiation via SERCA2-dependent Ca(2+) signaling.
Collapse
|
25
|
Silicon, a Possible Link between Environmental Exposure and Autoimmune Diseases: The Case of Rheumatoid Arthritis. ARTHRITIS 2012; 2012:604187. [PMID: 23119159 PMCID: PMC3483651 DOI: 10.1155/2012/604187] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 09/13/2012] [Indexed: 12/28/2022]
Abstract
Silicon is one of the most common chemicals on earth. Several compounds such as silica, asbestos, silicone or, nanoparticles are built from tetrahedral units with silicon as the central atom. Despite these, structural similarities, they have rarely been analyzed as a group. These compounds generate significant biological alterations that include immune hyperactivation, production of the reactive species of oxygen and tissue injury. These pathological processes may trigger autoimmune responses and lead to the development of rheumatoid arthritis. Populations at risk include those that constantly work in industrial process, mining, and agriculture as well as those that undergo silicone implants. Herein a review on the main features of these compounds and how they may induce autoimmune responses is presented.
Collapse
|
26
|
Mukherjee SP, Byrne HJ. Polyamidoamine dendrimer nanoparticle cytotoxicity, oxidative stress, caspase activation and inflammatory response: experimental observation and numerical simulation. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 9:202-11. [PMID: 22633897 DOI: 10.1016/j.nano.2012.05.002] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Revised: 04/02/2012] [Accepted: 05/07/2012] [Indexed: 02/01/2023]
Abstract
UNLABELLED Mechanisms underlying the in vitro cytotoxicity of Polyamidoamine nano-dendrimers in human keratinocytes are explored. Previous studies demonstrated a systematic, dendrimer-generation-dependent cytotoxicity, oxidative stress, and genotoxicity. The emerging picture is of dendrimer endocytosis, endosomal rupture and subsequent mitochondrial attack and cell death. To understand the underlying mechanisms, the evolution of reactive oxygen species, intracellular glutathione, caspase activation, mitochondrial membrane potential decay, and inflammatory responses have been examined. Early-stage responses are associated with endosomal encapsulation, later-stage with mitochondrial attack. In all cases, the magnitude and evolution of responses depend on dendrimer generation and dose. The early-stage response is modelled using a rate equation approach, qualitatively reproducing the time, dose and generation dependences, using only two variable parameters. The dependence of the response on the nanoparticle physicochemical properties can thus be separated from internal cellular parameters, and responses can be quantified in terms of rate constants rather than commonly employed effective concentrations. FROM THE CLINICAL EDITOR This contribution reports on the intracellular mechanism of PAMAM dendrimer cytotoxicity in human keratinocytes. In all cases, the magnitude and evolution of responses depend on dendrimer generation and dose. Experimental data were supported by numerical simulation using only two variables. It is suggested that responses can be quantified in terms of rate constants rather than effective concentrations.
Collapse
Affiliation(s)
- Sourav Prasanna Mukherjee
- Centre for Radiation and Environmental Science (RESC), Focas Research Institute, Dublin Institute of Technology, Dublin, Ireland.
| | | |
Collapse
|
27
|
Han X, Xu B, Beevers CS, Odaka Y, Chen L, Liu L, Luo Y, Zhou H, Chen W, Shen T, Huang S. Curcumin inhibits protein phosphatases 2A and 5, leading to activation of mitogen-activated protein kinases and death in tumor cells. Carcinogenesis 2012; 33:868-75. [PMID: 22298641 PMCID: PMC3324444 DOI: 10.1093/carcin/bgs029] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 01/25/2012] [Accepted: 01/26/2012] [Indexed: 12/22/2022] Open
Abstract
Curcumin can induce p53-independent apoptosis. However, the underlying mechanism remains to be defined. Here, we show that curcumin-induced apoptosis in a panel of tumor cells with mutant p53. Curcumin rapidly induced activation of the mitogen-activated protein kinases (MAPKs) including extracellular signal-regulated kinase 1/2 (Erk1/2) and c-Jun N-terminal kinase (JNK). Inhibition of JNK (with SP600125) or Erk1/2 (with U0126) partially prevented curcumin-induced cell death in the cells. Similarly, expression of dominant negative c-Jun or downregulation of Erk1/2 in part attenuated curcumin-induced cell death. It appears that curcumin-induced activation of MAPKs and apoptosis was due to induction of reactive oxygen species (ROS), as pretreatment with N-acetyl-L-cysteine, a ROS scavenger, blocked these events. Furthermore, we found that curcumin-induced activation of MAPK pathways was related to inhibition of the serine/threonine protein phosphatases 2A (PP2A) and 5 (PP5). Overexpression of PP2A or PP5 partially prevented curcumin-induced activation of JNK and Erk1/2 phosphorylation as well as cell death. The results suggest that curcumin induction of ROS activates MAPKs, at least partially by inhibiting PP2A and PP5, thereby leading to p53-independent apoptosis in tumor cells.
Collapse
Affiliation(s)
- Xiuzhen Han
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan 250012, Shandong Province, People’s Republic of China
- Department of Biochemistry and Molecular Biology
| | - Baoshan Xu
- Department of Biochemistry and Molecular Biology
| | | | | | - Long Chen
- Department of Biochemistry and Molecular Biology
| | - Lei Liu
- Department of Biochemistry and Molecular Biology
| | - Yan Luo
- Department of Biochemistry and Molecular Biology
| | - Hongyu Zhou
- Department of Biochemistry and Molecular Biology
| | - Wenxing Chen
- Department of Biochemistry and Molecular Biology
| | - Tao Shen
- Department of Biochemistry and Molecular Biology
| | - Shile Huang
- Department of Biochemistry and Molecular Biology
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| |
Collapse
|
28
|
Morishige T, Yoshioka Y, Inakura H, Tanabe A, Narimatsu S, Yao X, Monobe Y, Imazawa T, Tsunoda SI, Tsutsumi Y, Mukai Y, Okada N, Nakagawa S. Suppression of nanosilica particle-induced inflammation by surface modification of the particles. Arch Toxicol 2012; 86:1297-307. [DOI: 10.1007/s00204-012-0823-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 02/27/2012] [Indexed: 12/22/2022]
|
29
|
Orhue V, Kanaji A, Caicedo MS, Virdi AS, Sumner DR, Hallab NJ, Jahr H, Sena K. Calcineurin/nuclear factor of activated T cells (NFAT) signaling in cobalt-chromium-molybdenum (CoCrMo) particles-induced tumor necrosis factor-α (TNFα) secretion in MLO-Y4 osteocytes. J Orthop Res 2011; 29:1867-73. [PMID: 21557302 DOI: 10.1002/jor.21458] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Accepted: 04/22/2011] [Indexed: 02/04/2023]
Abstract
Aseptic loosening is the devastating long term complication of total hip arthroplasty and orthopedic implant debris has been shown to trigger an intense inflammatory reaction leading to resorption of the bone matrix. Inflammatory cytokines, such as tumor necrosis factor-α (TNFα), have been implicated in this process and osteocytes may play a role in its production. We previously demonstrated that cobalt-chromium-molybdenum (CoCrMo) particles upregulate TNFα production by MLO-Y4 osteocytes in vitro, but the underlying mechanism has not been elucidated. Based on previous studies by others, we hypothesized that the calcineurin-nuclear factor of activated T cells (NFAT) pathway mediates CoCrMo particle-induced TNFα production in MLO-Y4 osteocytes. MLO-Y4 osteocytes exposed to CoCrMo particle treatment resulted in a rapid and significant increase in calcineurin activity. We also demonstrate that CoCrMo particle-induced upregulation of TNFα is reduced to control levels with calcineurin-NFAT inhibitors and this was also confirmed at mRNA level. Moreover, we demonstrate the localization of NFATs in MLO-Y4 osteocytes and that NFAT1 and 2 translocate to the nucleus upon CoCrMo particle treatment. Our results suggest that calcineurin-NFAT signaling is involved in TNFα production by MLO-Y4 osteocytes after CoCrMo particle treatment.
Collapse
Affiliation(s)
- Vbenosawemwinghaye Orhue
- Department of Anatomy and Cell Biology, Rush University Medical Center, 600 South Paulina Street AF507, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Cox LAT. An exposure-response threshold for lung diseases and lung cancer caused by crystalline silica. RISK ANALYSIS : AN OFFICIAL PUBLICATION OF THE SOCIETY FOR RISK ANALYSIS 2011; 31:1543-1560. [PMID: 21477084 DOI: 10.1111/j.1539-6924.2011.01610.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Whether crystalline silica (CS) exposure increases risk of lung cancer in humans without silicosis, and, if so, whether the exposure-response relation has a threshold, have been much debated. Epidemiological evidence is ambiguous and conflicting. Experimental data show that high levels of CS cause lung cancer in rats, although not in other species, including mice, guinea pigs, or hamsters; but the relevance of such animal data to humans has been uncertain. This article applies recent insights into the toxicology of lung diseases caused by poorly soluble particles (PSPs), and by CS in particular, to model the exposure-response relation between CS and risk of lung pathologies such as chronic inflammation, silicosis, fibrosis, and lung cancer. An inflammatory mode of action is described, having substantial empirical support, in which exposure increases alveolar macrophages and neutrophils in the alveolar epithelium, leading to increased reactive oxygen species (ROS) and nitrogen species (RNS), pro-inflammatory mediators such as TNF-alpha, and eventual damage to lung tissue and epithelial hyperplasia, resulting in fibrosis and increased lung cancer risk among silicotics. This mode of action involves several positive feedback loops. Exposures that increase the gain factors around such loops can create a disease state with elevated levels of ROS, TNF-alpha, TGF-beta, alveolar macrophages, and neutrophils. This mechanism implies a "tipping point" threshold for the exposure-response relation. Applying this new model to epidemiological data, we conclude that current permissible exposure levels, on the order of 0.1 mg/m³, are probably below the threshold for triggering lung diseases in humans.
Collapse
|
31
|
Cai T, Li X, Ding J, Luo W, Li J, Huang C. A cross-talk between NFAT and NF-κB pathways is crucial for nickel-induced COX-2 expression in Beas-2B cells. Curr Cancer Drug Targets 2011; 11:548-59. [PMID: 21486220 DOI: 10.2174/156800911795656001] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2010] [Accepted: 11/30/2010] [Indexed: 01/09/2023]
Abstract
Cyclooxygenase-2 (COX-2) is a critical enzyme implicated in chronic inflammation-associated cancer development. Our studies have shown that the exposure of Beas-2B cells, a human bronchial epithelial cell line, to lung carcinogenic nickel compounds results in increased COX-2 expression. However, the signaling pathways leading to nickel-induced COX-2 expression are not well understood. In the current study, we found that the exposure of Beas-2B cells to nickel compounds resulted in the activation of both nuclear factor of activated T cell (NFAT) and nuclear factor-κB (NF-κB). The expression of COX-2 induced upon nickel exposure was inhibited by either a NFAT pharmacological inhibitor or the knockdown of NFAT3 by specific siRNA. We further found that the activation of NFAT and NF-κB was dependent on each other. Since our previous studies have shown that NF-κB activation is critical for nickel-induced COX-2 expression in Beas-2B cells exposed to nickel compounds under same experimental condition, we anticipate that there might be a cross-talk between the activation of NFAT and NF-κB for the COX-2 induction due to nickel exposure in Beas-2B cells. Furthermore, we showed that the scavenging of reactive oxygen species (ROS) by introduction of mitochondrial catalase inhibited the activation of both NFAT and NF-κB, and the induction of COX-2 due to nickel exposure. Taken together, our results defining the evidence showing a key role of the cross-talk between NFAT and NF-κB pathways in regulating nickel-induced COX-2 expression, further provide insight into the understanding of the molecular mechanisms linking nickel exposure to its lung carcinogenic effects.
Collapse
Affiliation(s)
- Tongjian Cai
- Nelson Institute of Environmental Medicine, New York University School of Medicine, 57 Old Forge Road, Tuxedo, NY 10987, USA
| | | | | | | | | | | |
Collapse
|
32
|
de Frutos S, Diaz JMR, Nitta CH, Sherpa ML, Bosc LVG. Endothelin-1 contributes to increased NFATc3 activation by chronic hypoxia in pulmonary arteries. Am J Physiol Cell Physiol 2011; 301:C441-50. [PMID: 21525433 DOI: 10.1152/ajpcell.00029.2011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Chronic hypoxia (CH) activates the Ca(2+)-dependent transcription factor nuclear factor of activated T cells isoform c3 (NFATc3) in mouse pulmonary arteries. However, the mechanism of this response has not been explored. Since we have demonstrated that NFATc3 is required for CH-induced pulmonary arterial remodeling, establishing how CH activates NFATc3 is physiologically significant. The goal of this study was to test the hypothesis that endothelin-1 (ET-1) contributes to CH-induced NFATc3 activation. We propose that this mechanism requires increased pulmonary arterial smooth muscle cell (PASMC) intracellular Ca(2+) concentration ([Ca(2+)](i)) and stimulation of RhoA/Rho kinase (ROK), leading to calcineurin activation and actin cytoskeleton polymerization, respectively. We found that: 1) CH increases pulmonary arterial pre-pro-ET-1 mRNA expression and lung RhoA activity; 2) inhibition of ET receptors, calcineurin, L-type Ca(2+) channels, and ROK blunts CH-induced NFATc3 activation in isolated intrapulmonary arteries from NFAT-luciferase reporter mice; and 3) both ET-1-induced NFATc3 activation in isolated mouse pulmonary arteries ex vivo and ET-1-induced NFATc3-green fluorescence protein nuclear import in human PASMC depend on ROK and actin polymerization. This study suggests that CH increases ET-1 expression, thereby elevating PASMC [Ca(2+)](i) and RhoA/ROK activity. As previously demonstrated, elevated [Ca(2+)](i) is required to activate calcineurin, which dephosphorylates NFATc3, allowing its nuclear import. Here, we demonstrate that ROK increases actin polymerization, thus providing structural support for NFATc3 nuclear transport.
Collapse
Affiliation(s)
- Sergio de Frutos
- Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, NM, 87131, USA
| | | | | | | | | |
Collapse
|
33
|
Kim HB, Kumar A, Wang L, Liu GH, Keller SR, Lawrence JC, Finck BN, Harris TE. Lipin 1 represses NFATc4 transcriptional activity in adipocytes to inhibit secretion of inflammatory factors. Mol Cell Biol 2010; 30:3126-39. [PMID: 20385772 PMCID: PMC2876672 DOI: 10.1128/mcb.01671-09] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Revised: 01/25/2010] [Accepted: 03/31/2010] [Indexed: 11/20/2022] Open
Abstract
Lipin 1 is a bifunctional protein that regulates gene transcription and, as a Mg(2+)-dependent phosphatidic acid phosphatase (PAP), is a key enzyme in the biosynthesis of phospholipids and triacylglycerol. We describe here the functional interaction between lipin 1 and the nuclear factor of activated T cells c4 (NFATc4). Lipin 1 represses NFATc4 transcriptional activity through protein-protein interaction, and lipin 1 is present at the promoters of NFATc4 transcriptional targets in vivo. Catalytically active and inactive lipin 1 can suppress NFATc4 transcriptional activity, and this suppression may involve recruitment of histone deacetylases to target promoters. In fat pads from mice deficient for lipin 1 (fld mice) and in 3T3-L1 adipocytes depleted of lipin 1 there is increased expression of several NFAT target genes including tumor necrosis factor alpha, resistin, FABP4, and PPARgamma. Finally, both lipin 1 protein and total PAP activity are decreased with increasing adiposity in the visceral, but not subcutaneous, fat pads of ob/ob mice. These observations place lipin 1 as a potentially important link between triacylglycerol synthesis and adipose tissue inflammation.
Collapse
Affiliation(s)
- Hyun Bae Kim
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Division of Endocrinology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Anil Kumar
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Division of Endocrinology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Lifu Wang
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Division of Endocrinology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Guang-Hui Liu
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Division of Endocrinology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Susanna R. Keller
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Division of Endocrinology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, Salk Institute for Biological Studies, La Jolla, California 92037
| | - John C. Lawrence
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Division of Endocrinology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Brian N. Finck
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Division of Endocrinology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, Salk Institute for Biological Studies, La Jolla, California 92037
| | - Thurl E. Harris
- Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Division of Endocrinology, University of Virginia School of Medicine, Charlottesville, Virginia 22908, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, Salk Institute for Biological Studies, La Jolla, California 92037
| |
Collapse
|
34
|
Li X, Hu Y, Jin Z, Jiang H, Wen J. Silica-induced TNF-alpha and TGF-beta1 expression in RAW264.7 cells are dependent on Src-ERK/AP-1 pathways. Toxicol Mech Methods 2010; 19:51-8. [PMID: 19778233 DOI: 10.1080/15376510802354201] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The cytokines secreted by lung macrophages have been shown to play a critical role in the pathogenesis of silicosis, tumor necrosis factor-alpha (TNF-alpha), and transforming growth factor-beta1 (TGF-beta1) are prominent cytokines in silicosis, but the underlying mechanism remains to be determined. The aim of the present study was to investigate the roles of Src-mitogen-activated protein kinase (MAPKs)/activator protein-1 (AP-1) signaling pathways in silica-induced TNF-alpha and TGF-beta1 expression in macrophage cells (RAW264.7). It was found that silica activated Src, p38 kinase, and extracellular signal-regulated kinase (ERK) in RAW264.7 cells. The induction of TNF-alpha and TGF-beta1 by silica was suppressed by Src inhibitor (PP1), ERK inhibitor (PD98059), but not by p38 kinase inhibitor (SB203580). Dominant negative mutant c-Jun (TAM67) inhibited silica-induced AP-1 DNA binding activity and downregulated the TNF-alpha and TGF-beta1 expression. In addition, PD98059 but not SB203580 inhibited the AP-1 DNA binding activity induced by silica. Based on these findings, it was conclude that Src-ERK/AP-1 signaling pathways are involved in the TNF-alpha and TGF-beta1 expression induced by silica in macrophages.
Collapse
Affiliation(s)
- Xiang Li
- Department of Pathology, Xiangya Medical School, Central South University, Changsha 410013, PR China
| | | | | | | | | |
Collapse
|
35
|
Falvo JV, Tsytsykova AV, Goldfeld AE. Transcriptional control of the TNF gene. ACTA ACUST UNITED AC 2010; 11:27-60. [PMID: 20173386 DOI: 10.1159/000289196] [Citation(s) in RCA: 194] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The cytokine TNF is a critical mediator of immune and inflammatory responses. The TNF gene is an immediate early gene, rapidly transcribed in a variety of cell types following exposure to a broad range of pathogens and signals of inflammation and stress. Regulation of TNF gene expression at the transcriptional level is cell type- and stimulus-specific, involving the recruitment of distinct sets of transcription factors to a compact and modular promoter region. In this review, we describe our current understanding of the mechanisms through which TNF transcription is specifically activated by a variety of extracellular stimuli in multiple cell types, including T cells, B cells, macrophages, mast cells, dendritic cells, and fibroblasts. We discuss the role of nuclear factor of activated T cells and other transcription factors and coactivators in enhanceosome formation, as well as the contradictory evidence for a role for nuclear factor kappaB as a classical activator of the TNF gene. We describe the impact of evolutionarily conserved cis-regulatory DNA motifs in the TNF locus upon TNF gene transcription, in contrast to the neutral effect of single nucleotide polymorphisms. We also assess the regulatory role of chromatin organization, epigenetic modifications, and long-range chromosomal interactions at the TNF locus.
Collapse
Affiliation(s)
- James V Falvo
- Immune Disease Institute and Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| | | | | |
Collapse
|
36
|
Eurich K, Segawa M, Toei-Shimizu S, Mizoguchi E. Potential role of chitinase 3-like-1 in inflammation-associated carcinogenic changes of epithelial cells. World J Gastroenterol 2009; 15:5249-59. [PMID: 19908331 PMCID: PMC2776850 DOI: 10.3748/wjg.15.5249] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The family of mammalian chitinases includes members both with and without glycohydrolase enzymatic activity against chitin, a polymer of N-acetylglucosamine. Chitin is the structural component of fungi, crustaceans, insects and parasitic nematodes, but is completely absent in mammals. Exposure to antigens containing chitin- or chitin-like structures sometimes induces strong T helper type-I responses in mammals, which may be associated with the induction of mammalian chitinases. Chitinase 3-like-1 (CHI3L1), a member of the mammalian chitinase family, is induced specifically during the course of inflammation in such disorders as inflammatory bowel disease, hepatitis and asthma. In addition, CHI3L1 is expressed and secreted by several types of solid tumors including glioblastoma, colon cancer, breast cancer and malignant melanoma. Although the exact function of CHI3L1 in inflammation and cancer is still largely unknown, CHI3L1 plays a pivotal role in exacerbating the inflammatory processes and in promoting angiogenesis and remodeling of the extracellular matrix. CHI3L1 may be highly involved in the chronic engagement of inflammation which potentiates development of epithelial tumorigenesis presumably by activating the mitogen-activated protein kinase and the protein kinase B signaling pathways. Anti-CHI3L1 antibodies or pan-chitinase inhibitors may have the potential to suppress CHI3L1-mediated chronic inflammation and the subsequent carcinogenic change in epithelial cells.
Collapse
|
37
|
Abstract
Exposing rodents to brief episodes of hypoxia mimics the hypoxemia and the cardiovascular and metabolic effects observed in patients with obstructive sleep apnoea (OSA), a condition that affects between 5% and 20% of the population. Apart from daytime sleepiness, OSA is associated with a high incidence of systemic and pulmonary hypertension, peripheral vascular disease, stroke and sudden cardiac death. The development of animal models to study sleep apnoea has provided convincing evidence that recurrent exposure to intermittent hypoxia (IH) has significant vascular and haemodynamic impact that explain much of the cardiovascular morbidity and mortality observed in patients with sleep apnoea. However, the molecular and cellular mechanisms of how IH causes these changes is unclear and under investigation. This review focuses on the most recent findings addressing these mechanisms. It includes a discussion of the contribution of the nervous system, circulating and vascular factors, inflammatory mediators and transcription factors to IH-induced cardiovascular disease. It also highlights the importance of reactive oxygen species as a primary mediator of the systemic and pulmonary hypertension that develops in response to exposure to IH.
Collapse
Affiliation(s)
- Laura V González Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, NM, USA.
| | | | | | | |
Collapse
|
38
|
Ji WJ, Zhou X, Zeng S, Wang SX. ATTENUATION OF SILICA-INDUCED PULMONARY FIBROBLASTS PROLIFERATION BY TAURINE AND NIACIN IN VITRO. Exp Lung Res 2009; 35:198-209. [DOI: 10.1080/01902140802499401] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
39
|
|
40
|
Abstract
Pulmonary pathology includes a large spectrum of both neoplastic and non-neoplastic diseases that affect the lung. Many of these are a result of the unusual relationship of the lung with the outside world. Every breath that a human takes brings the outside world into the body in the form of infectious agents, organic and inorganic particles, and noxious agents of all types. Although the lung has many defense mechanisms to protect itself from these insults, these are not infallible; therefore, lung pathology arises. Damage to the lung is particularly important given the role of the lung in the survival of the organism. Any impairment of lung function has widespread effects throughout the body, since all organs depend on the lungs for the oxygen they need. Pulmonary pathology catalogs the changes in the lung tissues and the mechanisms through which these occur. This chapter presents a review of lung pathology and the current state of knowledge about the pathogenesis of each disease. It suggests that a clear understanding of both morphology and mechanism is required for the development of new therapies and preventive measures.
Collapse
|
41
|
Fakhrzadeh L, Laskin JD, Laskin DL. Regulation of caveolin-1 expression, nitric oxide production and tissue injury by tumor necrosis factor-alpha following ozone inhalation. Toxicol Appl Pharmacol 2008; 227:380-9. [PMID: 18207479 PMCID: PMC3507418 DOI: 10.1016/j.taap.2007.11.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Revised: 11/06/2007] [Accepted: 11/08/2007] [Indexed: 01/15/2023]
Abstract
Alveolar macrophages (AM) and inflammatory mediators including nitric oxide and peroxynitrite contribute to ozone-induced lung injury. The generation of these mediators is regulated, in part, by the transcription factor NF-kappaB. We previously demonstrated a critical role for NF-kappaB p50 in ozone-induced injury. In the present studies mechanisms regulating NF-kappaB activation in the lung after ozone inhalation were analyzed. Treatment of wild type (WT) mice with ozone (0.8 ppm, 3 h) resulted in a rapid increase in NF-kappaB binding activity in AM, which persisted for at least 12 h. This was not evident in mice lacking TNFalpha which are protected from ozone-induced injury; there was also no evidence of nitric oxide or peroxynitrite production in lungs from these animals. These data demonstrate that TNFalpha plays a role in NF-kappaB activation and toxicity. TNFalpha signaling involves PI-3-kinase (PI3K)/protein kinase B (PKB), and p44/42 MAP kinase (MAPK) which are important in NF-kappaB activation. Ozone Inhalation resulted in rapid and transient increases in p44/42 MAPK and PI3K/PKB in AM from WT mice, which was evident immediately after exposure. Caveolin-1, a transmembrane protein that negatively regulates PI3K/PKB and p44/42 MAPK signaling, was downregulated in AM from WT mice after ozone exposure. In contrast, ozone had no effect on caveolin-1, PI3K/PKB or p44/42 MAPK expression in AM from TNFalpha knockout mice. These data, together with our findings that TNFalpha suppressed caveolin-1 expression in cultured AM, suggest that TNFalpha and downstream signaling mediate activation of NF-kappaB and the regulation of inflammatory genes important in ozone toxicity, and that this process is linked to caveolin-1.
Collapse
Affiliation(s)
- Ladan Fakhrzadeh
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey
| | - Jeffrey D. Laskin
- Department of Environmental and Occupational Medicine, University of Medicine and Dentistry of New Jersey-Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Debra L. Laskin
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey
| |
Collapse
|