1
|
Chen X, Fang M, Hong J, Guo Y. JNK Pathway-Associated Phosphatase Deficiency Facilitates Atherosclerotic Progression by Inducing T-Helper 1 and 17 Polarization and Inflammation in an ERK- and NF-κB Pathway-Dependent Manner. J Atheroscler Thromb 2024; 31:1460-1478. [PMID: 38797677 PMCID: PMC11456371 DOI: 10.5551/jat.64754] [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] [Indexed: 05/29/2024] Open
Abstract
AIM JNK pathway-associated phosphatase (JKAP) regulates T cell-mediated immunity and inflammation, which are involved in atherosclerosis pathogenesis. This study investigated the effects of JKAP on T-helper (Th) cell polarization, inflammation, and atherosclerotic progression. METHODS Serum JKAP levels were measured in 30 patients with coronary heart disease (CHD) and 30 controls. CHD blood naïve CD4+ T cells were acquired, followed by JKAP overexpression and knockdown with or without treatment with PD98059 (ERK inhibitor) or BAY-11-7082 (NF-κB inhibitor) in vitro. CD4+ T-cell conditional JKAP ablation mice were established in vivo, followed by the construction of an atherosclerosis model. RESULTS JKAP was reduced and negatively correlated with the Gensini score, CRP, Th1 cells, Th17 cells, and proinflammatory cytokines in patients with CHD. In vitro, JKAP overexpression suppressed Th1 and Th17 cell differentiation and proinflammatory cytokines, whereas JKAP knockdown exerted the opposite effect; however, JKAP modification did not affect Th2 cell differentiation. Interestingly, JKAP negatively regulated the ERK and NF-κB pathways; meanwhile, the PD98059 and BAY-11-7082 treatments repressed Th1 and Th17 cell differentiation, and attenuated the effect of JKAP knockdown on these indices. In vivo, conditional CD4+ T-cell JKAP ablation increased Th1 and Th17 cell polarization in the spleen, lymph node, blood, and/or aortic root. Furthermore, CD4+ T-cell conditional JKAP ablation exaggerated atherosclerotic lesions in the aorta, elevated CD4+ cell infiltration and proinflammatory cytokines in the aortic root, and activated the ERK and NF-κB pathways in the aortic root. CONCLUSION JKAP ablation facilitates atherosclerosis progression by promoting Th1 and 17 polarization and inflammation through regulation of the ERK and NF-κB pathways.
Collapse
Affiliation(s)
- Xinjing Chen
- Department of Cardiology, Provincial Clinical Medical College of Fujian Medical University, Fujian Institute of Cardiovascular Disease, Fujian Provincial Hospital
| | - Mingcheng Fang
- Department of Cardiology, Provincial Clinical Medical College of Fujian Medical University, Fujian Institute of Cardiovascular Disease, Fujian Provincial Hospital
| | - Jingxuan Hong
- Department of Cardiology, Provincial Clinical Medical College of Fujian Medical University, Fujian Institute of Cardiovascular Disease, Fujian Provincial Hospital
| | - Yansong Guo
- Department of Cardiology, Provincial Clinical Medical College of Fujian Medical University, Fujian Institute of Cardiovascular Disease, Fujian Provincial Hospital
| |
Collapse
|
2
|
Lin HH, Chang CW, Liao YT, Yeh SD, Lin HP, Ho HM, Cheung CHY, Juan HF, Chen YR, Su YW, Chen LM, Tan TH, Lin WJ. DUSP22 inhibits lung tumorigenesis by suppression of EGFR/c-Met signaling. Cell Death Discov 2024; 10:285. [PMID: 38877005 PMCID: PMC11178881 DOI: 10.1038/s41420-024-02038-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 06/16/2024] Open
Abstract
DUSP22, an atypical dual-specificity phosphatase enzyme, plays a significant role in regulating multiple kinase signaling pathways by dephosphorylation. Our study demonstrated that decreased DUSP22 expression is associated with shorter disease-free survival, advanced TNM (tumor, lymph nodes, and metastasis), cancer stage, and higher tumor grade in lung adenocarcinoma (LUAD) patients. Exogenous DUSP22 expression reduces the colony-forming capacity of lung cancer cells and inhibits xenograft tumor growth primarily by targeting EGFR and suppressing its activity through dephosphorylation. Knockdown of DUSP22 using shRNA enhances EGFR dependency in HCC827 lung cancer cells and increases sensitivity to gefitinib, an EGFR inhibitor. Consistently, genetic deletion of DUSP22 enhances EGFRdel (exon 19 deletion)-driven lung tumorigenesis and elevates EGFR activity. Pharmacological inhibition of DUSP22 activates EGFR, ERK1/2, and upregulates downstream PD-L1 expression. Additionally, lentiviral deletion of DUSP22 by shRNA enhances lung cancer cell migration through EGFR/c-Met and PD-L1-dependent pathways. Gefitinib, an EGFR inhibitor, mechanistically suppresses migration induced by DUSP22 deletion and inhibits c-Met activity. Furthermore, cabozantinib, a c-Met inhibitor, reduces migration and attenuates EGFR activation caused by DUSP22 deletion. Collectively, our findings support the hypothesis that loss of DUSP22 function in lung cancer cells confers a survival advantage by augmenting EGFR signaling, leading to increased activation of downstream c-Met, ERK1/2, and PD-L1 axis, ultimately contributing to the progression of advanced lung cancer.
Collapse
Affiliation(s)
- Hsiao-Han Lin
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Cheng-Wei Chang
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Yu-Ting Liao
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Shauh-Der Yeh
- Department of Urology, Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Hsiu-Ping Lin
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Hui-Min Ho
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | | | - Hsueh-Fen Juan
- Department of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Yi-Rong Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Yu-Wen Su
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Li-Mei Chen
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan
| | - Wen-Jye Lin
- Immunology Research Center, National Health Research Institutes, Miaoli County, 35053, Taiwan.
| |
Collapse
|
3
|
Chen L, Su H, Tao Z, Liang C, Liu Z, Dong Y, Zheng P, Liu Y. DUSP22 Ameliorates Endothelial-to-Mesenchymal Transition in HUVECs through Smad2/3 and MAPK Signaling Pathways. Cardiovasc Ther 2024; 2024:5583961. [PMID: 38495810 PMCID: PMC10942825 DOI: 10.1155/2024/5583961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 03/19/2024] Open
Abstract
Endothelial-to-mesenchymal transition (EndMT) is the process by which endothelial cells lose their endothelial properties and acquire mesenchymal characteristics. Dual-specific protein phosphatase 22 (DUSP22) inactivates various protein kinases and transcription factors by dephosphorylating serine/threonine residues: hence, it plays a key role in many diseases. The aim of this study was to explore the functional role of DUSP22 in EndMT. In the transforming growth factor-β-induced EndMT model in human umbilical vein endothelial cells (HUVECs), we observed a downregulation of DUSP22 expression. This DUSP22 deficiency could aggravate EndMT. Conversely, the overexpression of DUSP22 could ameliorate EndMT. We used signaling pathway inhibitors to verify our results and found that DUSP22 could regulate EndMT through the smad2/3 and the mitogen-activated protein kinase (MAPK) signaling pathways. In summary, DUSP22 ameliorates EndMT in HUVECs in vitro through the smad2/3 and MAPK signaling pathways.
Collapse
Affiliation(s)
- Lu Chen
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongyu Su
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zekai Tao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Cui Liang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongzhao Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yiming Dong
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peipei Zheng
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuan Liu
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
4
|
Shih YC, Chen HF, Wu CY, Ciou YR, Wang CW, Chuang HC, Tan TH. The phosphatase DUSP22 inhibits UBR2-mediated K63-ubiquitination and activation of Lck downstream of TCR signalling. Nat Commun 2024; 15:532. [PMID: 38225265 PMCID: PMC10789758 DOI: 10.1038/s41467-024-44843-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 01/08/2024] [Indexed: 01/17/2024] Open
Abstract
DUSP22 is a dual-specificity phosphatase that inhibits T cell activation by inactivating the kinase Lck. Here we show that the E3 ubiquitin ligase UBR2 is a positive upstream regulator of Lck during T-cell activation. DUSP22 dephosphorylates UBR2 at specific Serine residues, leading to ubiquitin-mediated UBR2 degradation. UBR2 is also modified by the SCF E3 ubiquitin ligase complex via Lys48-linked ubiquitination at multiple Lysine residues. Single-cell RNA sequencing analysis and UBR2 loss of function experiments showed that UBR2 is a positive regulator of proinflammatory cytokine expression. Mechanistically, UBR2 induces Lys63-linked ubiquitination of Lck at Lys99 and Lys276 residues, followed by Lck Tyr394 phosphorylation and activation as part of TCR signalling. Inflammatory phenotypes induced by TCR-triggered Lck activation or knocking out DUSP22, are attenuated by genomic deletion of UBR2. UBR2-Lck interaction and Lck Lys63-linked ubiquitination are induced in the peripheral blood T cells of human SLE patients, which demonstrate the relevance of the UBR2-mediated regulation of inflammation to human pathology. In summary, we show here an important regulatory mechanism of T cell activation, which finetunes the balance between T cell response and aggravated inflammation.
Collapse
Affiliation(s)
- Ying-Chun Shih
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Hsueh-Fen Chen
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Chia-Ying Wu
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Yi-Ru Ciou
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Chia-Wen Wang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan.
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan.
| |
Collapse
|
5
|
Chen MH, Chuang HC, Yeh YC, Chou CT, Tan TH. Dual-specificity phosphatases 22-deficient T cells contribute to the pathogenesis of ankylosing spondylitis. BMC Med 2023; 21:46. [PMID: 36765305 PMCID: PMC9921195 DOI: 10.1186/s12916-023-02745-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 01/19/2023] [Indexed: 02/12/2023] Open
Abstract
BACKGROUND Dual-specificity phosphatases (DUSPs) can dephosphorylate both tyrosine and serine/threonine residues of their substrates and regulate T cell-mediated immunity and autoimmunity. The aim of this study was to investigate the potential roles of DUSPs in ankylosing spondylitis (AS). METHODS Sixty AS patients and 45 healthy controls were enrolled in this study. Associations of gene expression of 23 DUSPs in peripheral T cells with inflammatory cytokine gene expression and disease activity of AS were analyzed. Finally, we investigated whether the characteristics of AS are developed in DUSP-knockout mice. RESULTS The mRNA levels of DUSP4, DUSP5, DUSP6, DUSP7, and DUSP14 in peripheral T cells were significantly higher in AS group than those of healthy controls (all p < 0.05), while DUSP22 (also named JKAP) mRNA levels were significantly lower in AS group than healthy controls (p < 0.001). The mRNA levels of DUSP4, DUSP5, DUSP6, DUSP7, and DUSP14 in T cells were positively correlated with mRNA levels of tumor necrosis factor-α (TNF-α), whereas DUSP22 was inversely correlated (all p < 0.05). In addition, inverse correlations of DUSP22 gene expression in peripheral T cells with C-reactive protein, erythrocyte sedimentation rate, and Bath Ankylosing Spondylitis Disease Activity Index (BASDAI) were observed (all p < 0.05). More importantly, aged DUSP22 knockout mice spontaneously developed syndesmophyte formation, which was accompanied by an increase of TNF-α+, interleukin-17A+, and interferon-γ+ CD3+ T cells. CONCLUSIONS DUSP22 may play a crucial role in the pathogenesis and regulation of disease activity of AS.
Collapse
Affiliation(s)
- Ming-Han Chen
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital, Taipei, Taiwan.,Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Yi-Chen Yeh
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chung-Tei Chou
- Division of Allergy, Immunology & Rheumatology, Taipei Veterans General Hospital, Taipei, Taiwan.
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan. .,Department of Pathology & Immunology, Baylor College of Medicine, Houston, Texas, USA.
| |
Collapse
|
6
|
Phillips RV, Wei L, Cardenas A, Hubbard AE, McHale CM, Vermeulen R, Wei H, Smith MT, Zhang L, Lan Q, Rothman N. Epigenome-wide association studies of occupational exposure to benzene and formaldehyde. Epigenetics 2022; 17:2259-2277. [PMID: 36017556 PMCID: PMC9665125 DOI: 10.1080/15592294.2022.2115604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 08/04/2022] [Accepted: 08/17/2022] [Indexed: 11/03/2022] Open
Abstract
Sufficient evidence supports a relationship between certain myeloid neoplasms and exposure to benzene or formaldehyde. DNA methylation could underlie benzene- and formaldehyde-induced health outcomes, but data in exposed human populations are limited. We conducted two cross-sectional epigenome-wide association studies (EWAS), one in workers exposed to benzene and another in workers exposed to formaldehyde. Using HumanMethylation450 BeadChips, we investigated differences in blood cell DNA methylation among 50 benzene-exposed subjects and 48 controls, and among 31 formaldehyde-exposed subjects and 40 controls. We performed CpG-level and regional-level analyses. In the benzene EWAS, we found genome-wide significant alterations, i.e., FWER-controlled P-values <0.05, in the mean and variance of methylation at 22 and 318 CpG sites, respectively, and in mean methylation of a large genomic region. Pathway analysis of genes corresponding to benzene-associated differential methylation sites revealed an impact on the AMPK signalling pathway. In formaldehyde-exposed subjects compared to controls, 9 CpGs in the DUSP22 gene promoter had genome-wide significant decreased methylation variability and a large region of the HOXA5 promoter with 44 CpGs was hypomethylated. Our findings suggest that DNA methylation may contribute to the pathogenesis of diseases related to benzene and formaldehyde exposure. Aberrant expression and methylation of HOXA5 previously has been shown to be clinically significant in myeloid leukaemias. The tumour suppressor gene DUSP22 is a potential biomarker of exposure to formaldehyde, and irregularities have been associated with multiple exposures and diseases.
Collapse
Affiliation(s)
- Rachael V. Phillips
- School of Public Health, University of California at Berkeley, Berkeley, CA, USA
| | - Linqing Wei
- School of Public Health, University of California at Berkeley, Berkeley, CA, USA
| | - Andres Cardenas
- School of Public Health, University of California at Berkeley, Berkeley, CA, USA
| | - Alan E. Hubbard
- School of Public Health, University of California at Berkeley, Berkeley, CA, USA
| | - Cliona M. McHale
- School of Public Health, University of California at Berkeley, Berkeley, CA, USA
| | - Roel Vermeulen
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Universiteit Utrecht (UU), Utrecht, The Netherlands
| | - Hu Wei
- Division of Cancer Epidemiology and Genetics, Occupational and Environmental Epidemiology Branch, NCI, NIH, DHHS, Bethesda, MD, USA
| | - Martyn T. Smith
- School of Public Health, University of California at Berkeley, Berkeley, CA, USA
| | - Luoping Zhang
- School of Public Health, University of California at Berkeley, Berkeley, CA, USA
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, Occupational and Environmental Epidemiology Branch, NCI, NIH, DHHS, Bethesda, MD, USA
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, Occupational and Environmental Epidemiology Branch, NCI, NIH, DHHS, Bethesda, MD, USA
| |
Collapse
|
7
|
Hepatocyte phosphatase DUSP22 mitigates NASH-HCC progression by targeting FAK. Nat Commun 2022; 13:5945. [PMID: 36209205 PMCID: PMC9547917 DOI: 10.1038/s41467-022-33493-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/21/2022] [Indexed: 11/08/2022] Open
Abstract
Nonalcoholic steatohepatitis (NASH), a common clinical disease, is becoming a leading cause of hepatocellular carcinoma (HCC). Dual specificity phosphatase 22 (DUSP22, also known as JKAP or JSP-1) expressed in numerous tissues plays essential biological functions in immune responses and tumor growth. However, the effects of DUSP22 on NASH still remain unknown. Here, we find a significant decrease of DUSP22 expression in human and murine fatty liver, which is mediated by reactive oxygen species (ROS) generation. Hepatic-specific DUSP22 deletion particularly exacerbates lipid deposition, inflammatory response and fibrosis in liver, facilitating NASH and non-alcoholic fatty liver disease (NAFLD)-associated HCC progression. In contrast, transgenic over-expression, lentivirus or adeno-associated virus (AAV)-mediated DUSP22 gene therapy substantially inhibit NASH-related phenotypes and HCC development in mice. We provide mechanistic evidence that DUSP22 directly interacts with focal adhesion kinase (FAK) and restrains its phosphorylation at Tyr397 (Y397) and Y576 + Y577 residues, subsequently prohibiting downstream activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and nuclear factor-κB (NF-κB) cascades. The binding of DUSP22 to FAK and the dephosphorylation of FAK are indispensable for DUSP22-meliorated NASH progression. Collectively, our findings identify DUSP22 as a key suppressor of NASH-HCC, and underscore the DUSP22-FAK axis as a promising therapeutic target for treatment of the disease.
Collapse
|
8
|
Wang C, Bai C, Mao C, Leng X, Wang F, Guo X. JNK pathway-associated phosphatase illustrates low expression and negative correlations with inflammation, disease activity, and T-helper 17 cells in inflammatory bowel disease children. J Clin Lab Anal 2022; 36:e24488. [PMID: 35908771 PMCID: PMC9459247 DOI: 10.1002/jcla.24488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 12/02/2022] Open
Abstract
Background C‐Jun N‐terminal kinase pathway‐associated phosphatase (JKAP) modulates the T cell receptor and mitogen‐activated protein kinase pathway‐mediated autoimmunity, thus participating in the pathogenesis of autoimmune diseases. This study aimed to explore the clinical implication of JKAP in inflammatory bowel disease (IBD) children. Methods C‐Jun N‐terminal kinase pathway‐associated phosphatase, tumor necrosis factor‐α (TNF‐α), interleukin‐23, interferon‐γ (T‐helper 1 secreted cytokine), and interleukin‐17A (T‐helper 17 secreted cytokine) in serum samples from 140 IBD children (including 60 Crohn's disease (CD) children and 80 ulcerative colitis (UC) children) were detected by ELISA. Meanwhile, JKAP from serum samples of 10 healthy controls (HCs) was also detected by ELISA. Results C‐Jun N‐terminal kinase pathway‐associated phosphatase was reduced in CD children (median (interquartile range (IQR)): 51.6 (36.8–69.5) pg/ml) and UC children (median (IQR): 57.5 (43.4–78.5) pg/ml) compared with HCs (median (IQR): 101.8 (70.0–143.2) pg/ml) (both p < 0.05). In CD children, JKAP was negatively correlated with C‐reactive protein (CRP) (p = 0.016) and erythrocyte sedimentation rate (ESR) (p = 0.029); while in UC children, JKAP was also negatively correlated with CRP (p = 0.006) and ESR (p = 0.022). Regarding the correlation of JKAP with disease activity, it presented negative correlations with PCDAI (p = 0.001) and PUCAI (p = 0.002). Besides, JKAP was negatively related to TNF‐α (both p < 0.05) but not interleukin‐23 (both p>0.05) in CD and UC children. Additionally, JKAP was not correlated with interferon‐γ in CD or UC children (both p>0.05), while negatively correlated with interleukin‐17A in CD and UC children (both p < 0.05). Conclusion C‐Jun N‐terminal kinase pathway‐associated phosphatase shows low expression and negative correlations with inflammation, disease activity, and T‐helper 17 cells in IBD children.
Collapse
Affiliation(s)
- Caixia Wang
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cui Bai
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chenggang Mao
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xuefei Leng
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fang Wang
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xingqing Guo
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Chou HC, Cheng CM, Yang CH, Lin TY, Liu YW, Tan TH, Chen YR. DUSP3 regulates phosphorylation-mediated degradation of occludin and is required for maintaining epithelial tight junction. J Biomed Sci 2022; 29:40. [PMID: 35705979 PMCID: PMC9199239 DOI: 10.1186/s12929-022-00826-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/08/2022] [Indexed: 12/03/2022] Open
Abstract
Background Tight junctions (TJ) are multi-protein complexes that hold epithelial cells together and form structural and functional barriers for maintaining proper biological activities. Dual specificity phosphatase 3 (DUSP3), a suppressor of multiple protein tyrosine (Tyr) kinases, is decreased in lung cancer tissues. Here we demonstrated the role of DUSP3 in regulation of epithelial TJ. Methods Barrier functions of TJ were examined in wild-type or DUSP3-deficient lung epithelial cells. Animal and clinical data were analyzed for the association between DUSP3 deficiency and lung cancer progression. Proximity ligation assay, immunoblotting, and phosphatase assay were performed to study the effect of DUSP3 on the TJ protein occludin (OCLN). Mutations of Tyr residues on OCLN showed the role of Tyr phosphorylation in regulating OCLN. Results Compared to those of the DUSP3-expressing cells, we found the expression and distribution of ZO-1, a TJ-anchoring molecule, were abnormal in DUSP3-deficient cells. OCLN had an increased phosphorylation level in DUSP3-deficient cells. We identified that OCLN is a direct substrate of DUSP3. DUSP3 regulated OCLN ubiquitination and degradation through decreasing OCLN tyrosine phosphorylation directly or through suppressing focal adhesion kinase, the OCLN kinase. Conclusion Our study revealed that DUSP3 is an important TJ regulatory protein and its decrease may be involved in progression of epithelial cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s12929-022-00826-x.
Collapse
Affiliation(s)
- Hsiao-Chin Chou
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Chun-Mei Cheng
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Chi-Hwa Yang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Tzu-Yin Lin
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Ya-Wen Liu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan.,Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Yi-Rong Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, 35053, Taiwan.
| |
Collapse
|
10
|
Gao W, Gao L, Yang F, Li Z. Circulating JNK pathway-associated phosphatase: A novel biomarker correlates with Th17 cells, acute exacerbation risk, and severity in chronic obstructive pulmonary disease patients. J Clin Lab Anal 2021; 36:e24153. [PMID: 34918391 PMCID: PMC8761399 DOI: 10.1002/jcla.24153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022] Open
Abstract
Background JNK pathway‐associated phosphatase (JKAP) involves in the regulation of inflammation, immunity, and lung injury. The current study aimed to investigate correlation of JKAP with Th1, Th17 cells, acute exacerbation risk, and disease severity in chronic obstructive pulmonary disease (COPD) patients. Methods Totally, 45 stable COPD (SCOPD) patients, 45 acute exacerbation COPD (AECOPD) patients, and 45 controls were enrolled. Serum was collected for JKAP, interferon‐gamma (IFN‐γ) (Th1 cytokine), and interleukin 17 (IL‐17) (Th17 cytokine) detection. Besides, peripheral blood mononuclear cell from COPD patients was collected for evaluating Th1 and Th17 cells. Results JKAP was highest in controls followed by SCOPD patients and lowest in AECOPD patients (median: 105.673 vs. 75.374 vs. 41.807 pg/ml, p < 0.001). Meanwhile, receiver operating characteristic (ROC) curves revealed that JKAP differentiated the AECOPD patients from the controls (area under curve (AUC): 0.910 (95% confidence interval (CI): 0.849–0.970)) and AECOPD patients from SCOPD patients (AUC: 0.726 (95% CI: 0.622–0.830)). Moreover, JKAP positively correlated with FEV1 (%predicted) in AECOPD patients (r = 0.347 p = 0.019). Additionally, JKAP was negatively correlated with the GOLD stage in AECOPD patients (r = −0.344, p = 0.021) and SCOPD patients (r = −0.357, p = 0.016). Whereas, JKAP was not associated with other clinical features (all p > 0.05). Besides, JKAP was negatively linked with Th17 cells (r = −0.378, p = 0.010), IFN‐γ (r = −0.358, p = 0.016), IL‐17 (r = −0.414, p = 0.005) in AECOPD patients and Th17 cells (r = −0.342, p = 0.022), IL‐17 (r = −0.299, p = 0.046) in SCOPD patients. Conclusion Downregulated JKAP correlates with Th17 cells, higher acute exacerbation risk, and severity in COPD patients, indicating its underlying potency as a biomarker for COPD.
Collapse
Affiliation(s)
- Wei Gao
- Department of Respiratory Medicine, Capital Medical University School of Rehabilitation Medicine, China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, China
| | - Lianjun Gao
- Department of Respiratory Medicine, Capital Medical University School of Rehabilitation Medicine, China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, China
| | - Feng Yang
- Department of Respiratory Medicine, Capital Medical University School of Rehabilitation Medicine, China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, China
| | - Zongjun Li
- Department of Respiratory Medicine, Capital Medical University School of Rehabilitation Medicine, China Rehabilitation Research Center, Beijing Bo'ai Hospital, Beijing, China
| |
Collapse
|
11
|
Qian C, Chen J, Xu X, Liu Q, Gu M, Lu S, Bai H, Wang Q, Xue M. Measurement of synovium and serum dual specificity phosphatase 22 level: Their inter-correlation and potency as biomarkers in rheumatoid arthritis. J Clin Lab Anal 2021; 36:e24111. [PMID: 34811816 PMCID: PMC8761394 DOI: 10.1002/jcla.24111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/17/2021] [Accepted: 10/31/2021] [Indexed: 11/06/2022] Open
Abstract
Background Dual specificity phosphatase 22 (DUSP22), also named as Jun N‐terminal kinase pathway associated phosphatase recently, is reported to be closely engaged in immune and inflammation regulation. This study aimed to investigate the interaction between synovium DUSP22 and serum DUSP22 levels and to explore their correlation with rheumatoid arthritis (RA) risk, inflammation, and disease activity. Methods Synovium and serum samples from 42 RA patients with knee involvement underwent arthroscopy, and 20 knee trauma patients were collected. Besides, serum samples from 40 healthy controls were also obtained. Synovium DUSP22 expression was detected by reverse transcription quantitative polymerase chain reaction, while serum DUSP22 level was detected by enzyme‐linked immunosorbent assay. Results Synovium DUSP22 level was greatly decreased in RA patients compared to trauma controls (p < 0.001), and it was negatively correlated with tender joint count (TJC) (r = −0.318, p = 0.040), C‐reactive protein (CRP) (r = −0.330, p = 0.033), and Lysholm score (r = −0.423, p = 0.005) in RA patients. Serum DUSP22 level was lowest in RA patients, followed by trauma controls, then highest in healthy controls (p < 0.001). Serum DUSP22 level was negatively associated with TJC (r = −0.438, p = 0.004), swollen joint count (SJC) (r = −0.372, p = 0.015), CRP (r = −0.391, p = 0.011), and disease activity score in 28 joints (DAS28ESR) score (r = −0.406, p = 0.008), and it increased after treatment (p = 0.001) in RA patients. In addition, serum DUSP22 level positively related to synovium DUSP22 level in RA patients (r = 0.394, p = 0.010). Conclusion Synovium and serum DUSP22 are intercorrelated and insufficiently expressed in RA patients; meanwhile, their deficiency correlates with increased systemic inflammation, disease activity, and joint dysfunction.
Collapse
Affiliation(s)
- Chen Qian
- Department of Clinical Laboratory, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Jie Chen
- Department of Orthopedics, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaopeng Xu
- Department of Clinical Laboratory, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Qingyang Liu
- Department of Clinical Laboratory, Wuxi 9th People's Hospital, Wuxi 9th Affiliated Hospital of Soochow University, Wuxi, China
| | - Minhong Gu
- Department of Clinical Laboratory, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Sheng Lu
- Department of Clinical Laboratory, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Hongxia Bai
- Department of Clinical Laboratory, Xishan People's Hospital of Wuxi City, Wuxi, China
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi 9th People's Hospital, Wuxi 9th Affiliated Hospital of Soochow University, Wuxi, China
| | - Mingyu Xue
- Department of Clinical Orthopaedics, Wuxi 9th People's Hospital, Wuxi 9th Affiliated Hospital of Soochow University, Wuxi, China
| |
Collapse
|
12
|
Downregulation of Snail by DUSP1 Impairs Cell Migration and Invasion through the Inactivation of JNK and ERK and Is Useful as a Predictive Factor in the Prognosis of Prostate Cancer. Cancers (Basel) 2021; 13:cancers13051158. [PMID: 33800291 PMCID: PMC7962644 DOI: 10.3390/cancers13051158] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 01/06/2023] Open
Abstract
Dual specificity phosphatase 1 (DUSP1) is crucial in prostate cancer (PC), since its expression is downregulated in advanced carcinomas. Here, we investigated DUSP1 effects on the expression of mesenchymal marker Snail, cell migration and invasion, analyzing the underlying mechanisms mediated by mitogen-activated protein kinases (MAPKs) inhibition. To this purpose, we used different PC cells overexpressing or lacking DUSP1 or incubated with MAPKs inhibitors. Moreover, we addressed the correlation of DUSP1 expression with Snail and activated MAPKs levels in samples from patients diagnosed with benign hyperplasia or prostate carcinoma, studying its implication in tumor prognosis and survival. We found that DUSP1 downregulates Snail expression and impairs migration and invasion in PC cells. Similar results were obtained following the inhibition of c-Jun N-terminal kinase (JNK) and extracellular-signal-regulated kinase (ERK). In clinical samples, we evidenced an inverse correlation between DUSP1 expression and Snail levels, which are further associated with JNK and ERK activation. Consequently, the pattern DUSP1high/activated JNKlow/activated ERKlow/Snaillow is associated with an overall extended survival of PC patients. In summary, the ratio between DUSP1 and Snail expression, with additional JNK and ERK activity measurement, may serve as a potential biomarker to predict the clinical outcome of PC patients. Furthermore, DUSP1 induction or inhibition of JNK and ERK pathways could be useful to treat PC.
Collapse
|
13
|
Sun L, Tu J, Chen X, Dai M, Xia X, Liu C, Zhou Y. JNK pathway-associated phosphatase associates with rheumatoid arthritis risk, disease activity, and its longitudinal elevation relates to etanercept treatment response. J Clin Lab Anal 2021; 35:e23709. [PMID: 33547838 PMCID: PMC8059732 DOI: 10.1002/jcla.23709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/22/2020] [Accepted: 12/28/2020] [Indexed: 12/29/2022] Open
Abstract
Background This study aimed to investigate the relationship of serum JNK pathway‐associated phosphatase (JKAP) expression with rheumatoid arthritis (RA) risk and clinical features, also to explore the longitudinal change of JKAP during etanercept treatment and its relationship with etanercept treatment response in RA patients. Methods A total of 87 RA patients and 44 healthy controls (HCs) were enrolled; then, their JKAP expression in serum was determined by enzyme‐linked immunosorbent assay (ELISA). Among 87 RA patients, 42 cases further received the 24‐week etanercept treatment; then, their JKAP level in serum (detected by ELISA) and clinical response (evaluated by disease activity score in 28 joints (DAS28) score) were evaluated at week 4 (W4), week 12 (W12), and week 24 (W24) after initiation of etanercept treatment. Results JKAP expression was decreased in RA patients compared to HCs, which disclosed a good predictive value for RA risk. JKAP expression was negatively associated with tender joint count, swollen joint count, erythrocyte sedimentation rate, C‐reactive protein, and DAS28 in RA patients, respectively. For RA patients who received 24‐week etanercept treatment, their clinical response rate was 0.0%, 33.3%, 50.0%, and 69% at W0, W4, W12, and W24, respectively. Importantly, JKAP was gradually increased during etanercept treatment, whose longitudinal elevation positively related to etanercept treatment response in RA patients. Conclusion Circulating JKAP links with decreased RA risk and mild disease activity, whose longitudinal elevation positively relates to etanercept treatment response.
Collapse
Affiliation(s)
- Li Sun
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianxin Tu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaowei Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Meijie Dai
- Department of Laboratory Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaoru Xia
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Cailong Liu
- Department of Orthopaedic Sports Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yan Zhou
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
14
|
Thompson EM, Stoker AW. A Review of DUSP26: Structure, Regulation and Relevance in Human Disease. Int J Mol Sci 2021; 22:ijms22020776. [PMID: 33466673 PMCID: PMC7828806 DOI: 10.3390/ijms22020776] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/05/2021] [Accepted: 01/08/2021] [Indexed: 01/10/2023] Open
Abstract
Dual specificity phosphatases (DUSPs) play a crucial role in the regulation of intracellular signalling pathways, which in turn influence a broad range of physiological processes. DUSP malfunction is increasingly observed in a broad range of human diseases due to deregulation of key pathways, most notably the MAP kinase (MAPK) cascades. Dual specificity phosphatase 26 (DUSP26) is an atypical DUSP with a range of physiological substrates including the MAPKs. The residues that govern DUSP26 substrate specificity are yet to be determined; however, recent evidence suggests that interactions with a binding partner may be required for DUSP26 catalytic activity. DUSP26 is heavily implicated in cancer where, akin to other DUSPs, it displays both tumour-suppressive and -promoting properties, depending on the context. Here we review DUSP26 by evaluating its transcriptional patterns, protein crystallographic structure and substrate binding, as well as its physiological role(s) and binding partners, its role in human disease and the development of DUSP26 inhibitors.
Collapse
|
15
|
Structural Insights into the Active Site Formation of DUSP22 in N-loop-containing Protein Tyrosine Phosphatases. Int J Mol Sci 2020; 21:ijms21207515. [PMID: 33053837 PMCID: PMC7589817 DOI: 10.3390/ijms21207515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/07/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023] Open
Abstract
Cysteine-based protein tyrosine phosphatases (Cys-based PTPs) perform dephosphorylation to regulate signaling pathways in cellular responses. The hydrogen bonding network in their active site plays an important conformational role and supports the phosphatase activity. Nearly half of dual-specificity phosphatases (DUSPs) use three conserved residues, including aspartate in the D-loop, serine in the P-loop, and asparagine in the N-loop, to form the hydrogen bonding network, the D-, P-, N-triloop interaction (DPN-triloop interaction). In this study, DUSP22 is used to investigate the importance of the DPN-triloop interaction in active site formation. Alanine mutations and somatic mutations of the conserved residues, D57, S93, and N128 substantially decrease catalytic efficiency (kcat/KM) by more than 102-fold. Structural studies by NMR and crystallography reveal that each residue can perturb the three loops and induce conformational changes, indicating that the hydrogen bonding network aligns the residues in the correct positions for substrate interaction and catalysis. Studying the DPN-triloop interaction reveals the mechanism maintaining phosphatase activity in N-loop-containing PTPs and provides a foundation for further investigation of active site formation in different members of this protein class.
Collapse
|
16
|
Li M, Wang L, Shi DC, Foo JN, Zhong Z, Khor CC, Lanzani C, Citterio L, Salvi E, Yin PR, Bei JX, Wang L, Liao YH, Chen J, Chen QK, Xu G, Jiang GR, Wan JX, Chen MH, Chen N, Zhang H, Zeng YX, Liu ZH, Liu JJ, Yu XQ. Genome-Wide Meta-Analysis Identifies Three Novel Susceptibility Loci and Reveals Ethnic Heterogeneity of Genetic Susceptibility for IgA Nephropathy. J Am Soc Nephrol 2020; 31:2949-2963. [PMID: 32912934 DOI: 10.1681/asn.2019080799] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 07/21/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Eighteen known susceptibility loci for IgAN account for only a small proportion of IgAN risk. METHODS Genome-wide meta-analysis was performed in 2628 patients and 11,563 controls of Chinese ancestry, and a replication analysis was conducted in 6879 patients and 9019 controls of Chinese descent and 1039 patients and 1289 controls of European ancestry. The data were used to assess the association of susceptibility loci with clinical phenotypes for IgAN, and to investigate genetic heterogeneity of IgAN susceptibility between the two populations. Imputation-based analysis of the MHC/HLA region extended the scrutiny. RESULTS Identification of three novel loci (rs6427389 on 1q23.1 [P=8.18×10-9, OR=1.132], rs6942325 on 6p25.3 [P=1.62×10-11, OR=1.165], and rs2240335 on 1p36.13 [P=5.10×10-9, OR=1.114]), implicates FCRL3, DUSP22.IRF4, and PADI4 as susceptibility genes for IgAN. Rs2240335 is associated with the expression level of PADI4, and rs6427389 is in high linkage disequilibrium with rs11264799, which showed a strong expression quantitative trail loci effect on FCRL3. Of the 24 confirmed risk SNPs, six showed significant heterogeneity of genetic effects and DEFA showed clear evidence of allelic heterogeneity between the populations. Imputation-based analysis of the MHC region revealed significant associations at three HLA polymorphisms (HLA allele DPB1*02, AA_DRB1_140_32657458_T, and AA_DQA1_34_32717152) and two SNPs (rs9275464 and rs2295119). CONCLUSIONS A meta-analysis of GWAS data revealed three novel genetic risk loci for IgAN, and three HLA polymorphisms and two SNPs within the MHC region, and demonstrated the genetic heterogeneity of seven loci out of 24 confirmed risk SNPs. These variants may explain susceptibility differences between Chinese and European populations.
Collapse
Affiliation(s)
- Ming Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China .,National Health Commission Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Ling Wang
- Human Genetics, Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Dian-Chun Shi
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.,Guangdong Provincial People's Hospital, Guangzhou, China
| | - Jia-Nee Foo
- Human Genetics, Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Zhong Zhong
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Chiea-Chuen Khor
- Human Genetics, Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore
| | - Chiara Lanzani
- Genomics of Renal Diseases and Hypertension Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Lorena Citterio
- Genomics of Renal Diseases and Hypertension Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Erika Salvi
- Neurology Unit, IRCCS Neurology Institute "Carlo Besta," Milan, Italy
| | - Pei-Ran Yin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,National Health Commission Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Jin-Xin Bei
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Li Wang
- Department of Nephrology, Sichuan Provincial People's Hospital, Chengdu, China
| | - Yun-Hua Liao
- Department of Nephrology, The First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Jian Chen
- Department of Nephrology, Fuzhou General Hospital of Nanjing Military Command, Fuzhou, China
| | - Qin-Kai Chen
- Department of Nephrology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gang Xu
- Department of Nephrology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Geng-Ru Jiang
- Department of Nephrology, XinHua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Xin Wan
- Department of Nephrology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Meng-Hua Chen
- Department of Nephrology, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Nan Chen
- Department of Nephrology, RuiJin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Zhang
- Renal Division, Peking University First Hospital, Peking University, Institute of Nephrology, Beijing, China
| | - Yi-Xin Zeng
- Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China.,State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Zhi-Hong Liu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jian-Jun Liu
- Human Genetics, Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore .,Guangdong Provincial People's Hospital, Guangzhou, China.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xue-Qing Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China .,National Health Commission Key Laboratory of Nephrology (Sun Yat-sen University), Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China.,Guangdong Provincial People's Hospital, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
17
|
Yang CY, Chuang HC, Tsai CY, Xiao YZ, Yang JY, Huang RH, Shih YC, Tan TH. DUSP11 Attenuates Lipopolysaccharide-Induced Macrophage Activation by Targeting TAK1. THE JOURNAL OF IMMUNOLOGY 2020; 205:1644-1652. [PMID: 32796023 DOI: 10.4049/jimmunol.2000334] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/19/2020] [Indexed: 01/23/2023]
Abstract
Dual-specificity phosphatase 11 (DUSP11, also named as PIR1) is a member of the atypical DUSP protein tyrosine phosphatase family. DUSP11 is only known to be an RNA phosphatase that regulates noncoding RNA stability. To date, the role of DUSP11 in immune cell signaling and immune responses remains unknown. In this study, we generated and characterized the immune cell functions of DUSP11-deficient mice. We identified TGF-β-activated kinase 1 (TAK1) as a DUSP11-targeted protein. DUSP11 interacted directly with TAK1, and the DUSP11-TAK1 interaction was enhanced by LPS stimulation in bone marrow-derived macrophages. DUSP11 deficiency enhanced the LPS-induced TAK1 phosphorylation and cytokine production in bone marrow-derived macrophages. Furthermore, DUSP11-deficient mice were more susceptible to LPS-induced endotoxic shock. The LPS-induced serum levels of IL-1β, TNF-α, and IL-6 were significantly elevated in DUSP11-deficient mice compared with those of wild-type mice. The data indicate that DUSP11 inhibits LPS-induced macrophage activation by targeting TAK1.
Collapse
Affiliation(s)
- Chia-Yu Yang
- Immunology Research Center, National Health Research Institutes, 35053 Zhunan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, 33302 Tao-Yuan, Taiwan; and
| | - Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, 35053 Zhunan, Taiwan
| | - Ching-Yi Tsai
- Immunology Research Center, National Health Research Institutes, 35053 Zhunan, Taiwan
| | - Yu-Zhi Xiao
- Immunology Research Center, National Health Research Institutes, 35053 Zhunan, Taiwan
| | - Jhih-Yu Yang
- Immunology Research Center, National Health Research Institutes, 35053 Zhunan, Taiwan
| | - Rou-Huei Huang
- Immunology Research Center, National Health Research Institutes, 35053 Zhunan, Taiwan
| | - Ying-Chun Shih
- Immunology Research Center, National Health Research Institutes, 35053 Zhunan, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, 35053 Zhunan, Taiwan; .,Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
18
|
Abstract
Anaplastic large cell lymphomas are a rare subtype of peripheral/mature T-cell lymphomas which are clinically, pathologically and genetically heterogeneous. Both ALK-positive (ALK+) and ALK-negative (ALK-) ALCL are composed of large lymphoid cells with abundant cytoplasm and pleomorphic features with horseshoe-shaped and reniform nuclei. ALK+ ALCL were considered as a definite entity in the 2008 World Health Organization classification of hematopoietic and lymphoid tissues. ALK-ALCL was included as a provisional entity in the WHO 2008 edition and in the most recent 2017 edition, it is now considered a distinct entity that includes cytogenetic subsets that appear to have prognostic implications (e.g. 6p25 rearrangements at IRF4/DUSP22 locus). ALK+ ALCLs are distinct in epidemiology and pathogenetic origin and should be distinguished from ALK-ALCL, cutaneous ALCL and breast implant associated ALCL which have distinct clinical course and pathogenetic features. Breast implant-associated ALCL is now recognized as a new provisional entity distinct from other ALK-ALCL; notably that it is a noninvasive disease associated with excellent outcome. In this article, we will provide an overview of the salient themes relevant to the pathology and genetic mechanisms in ALCL.
Collapse
Affiliation(s)
- Vasiliki Leventaki
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Siddharth Bhattacharyya
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA United States
| | - Megan S Lim
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA United States.
| |
Collapse
|
19
|
Chuang HC, Tan TH. MAP4K Family Kinases and DUSP Family Phosphatases in T-Cell Signaling and Systemic Lupus Erythematosus. Cells 2019; 8:cells8111433. [PMID: 31766293 PMCID: PMC6912701 DOI: 10.3390/cells8111433] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/08/2019] [Accepted: 11/11/2019] [Indexed: 12/21/2022] Open
Abstract
T cells play a critical role in the pathogenesis of systemic lupus erythematosus (SLE), which is a severe autoimmune disease. In the past 60 years, only one new therapeutic agent with limited efficacy has been approved for SLE treatment; therefore, the development of early diagnostic biomarkers and therapeutic targets for SLE is desirable. Mitogen-activated protein kinase kinase kinase kinases (MAP4Ks) and dual-specificity phosphatases (DUSPs) are regulators of MAP kinases. Several MAP4Ks and DUSPs are involved in T-cell signaling and autoimmune responses. HPK1 (MAP4K1), DUSP22 (JKAP), and DUSP14 are negative regulators of T-cell activation. Consistently, HPK1 and DUSP22 are downregulated in the T cells of human SLE patients. In contrast, MAP4K3 (GLK) is a positive regulator of T-cell signaling and T-cell-mediated immune responses. MAP4K3 overexpression-induced RORγt–AhR complex specifically controls interleukin 17A (IL-17A) production in T cells, leading to autoimmune responses. Consistently, MAP4K3 and the RORγt–AhR complex are overexpressed in the T cells of human SLE patients, as are DUSP4 and DUSP23. In addition, DUSPs are also involved in either human autoimmune diseases (DUSP2, DUSP7, DUSP10, and DUSP12) or T-cell activation (DUSP1, DUSP5, and DUSP14). In this review, we summarize the MAP4Ks and DUSPs that are potential biomarkers and/or therapeutic targets for SLE.
Collapse
|
20
|
Lin HP, Ho HM, Chang CW, Yeh SD, Su YW, Tan TH, Lin WJ. DUSP22 suppresses prostate cancer proliferation by targeting the EGFR-AR axis. FASEB J 2019; 33:14653-14667. [PMID: 31693867 DOI: 10.1096/fj.201802558rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Dual-specificity phosphatases (DUSPs) regulate the activity of various downstream kinases through serine or threonine or tyrosine dephosphorylation. Loss of function and aberrant expression of DUSPs has been implicated in cancer progression and poor survival, yet the function of DUSP22 in prostate cancer (PCa) cells is not clear. Gene Expression Omnibus and cBioPortal microarray database analyses showed that DUSP22 expression was lower in PCa tissues than normal prostate tissues, and altered DUSP22 expression was associated with shorter progression-free and disease-free survival of patients with PCa. Exogenous DUSP22 expression in LNCaP, PC3, and C4-2B PCa cells inhibited cellular proliferation and colony formation, supporting a growth inhibitory role for DUSP22 in PCa cells. DUSP22 expression significantly attenuated epidermal growth factor (EGF) receptor (EGFR) and its downstream ERK1/2 signaling by dephosphorylation. However, DUSP22 failed to suppress the growth of CWR22Rv1 and DU145 cells with elevated phosphorylated (p-)ERK1/2 levels. A serine-to-alanine mutation at position 58, a potential ERK1/2-targeted phosphorylation site in DUSP22, was sufficient to suppress growth of CWR22Rv1 cells with elevated p-ERK1/2 levels, suggesting a mutually antagonistic relationship between DUSP22 and ERK1/2 dependent on phosphorylation status. We showed that DUSP22 can suppress prostate-specific antigen gene expression through phosphatase-dependent pathways, suggesting that DUSP22 is an important regulator of the androgen receptor (AR) in PCa cells. Mechanistically, DUSP22 can interact with AR as a regulatory partner and interfere with EGF-induced AR phosphorylation at Tyr534, suggesting that DUSP22 serves as a crucial suppressor of both EGFR and AR-dependent signaling in PCa cells via dephosphorylation. Our findings indicate that loss of function of DUSP22 in PCa cells leads to aberrant activation of both EGFR-ERKs and AR signaling and ultimately progression of PCa, supporting the potential for novel therapeutic design of harnessing DUSP22 in the treatment of PCa.-Lin, H.-P., Ho, H.-M., Chang, C.-W., Yeh, S.-D., Su, Y.-W., Tan, T.-H., Lin, W.-J. DUSP22 suppresses prostate cancer proliferation by targeting the EGFR-AR axis.
Collapse
Affiliation(s)
- Hsiu-Ping Lin
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Hui-Min Ho
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Cheng-Wei Chang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Shauh-Der Yeh
- Department of Urology, Graduate Institute of Clinical Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wen Su
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| | - Wen-Jye Lin
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan; and
| |
Collapse
|
21
|
Shi X, Yang W, Wang N, Zhu J. Circulating JNK pathway-associated phosphatase level correlates with decreased risk, activity, inflammation level and reduced clinical response to tumor necrosis factor-α inhibitor in Crohn disease patients. Medicine (Baltimore) 2019; 98:e16622. [PMID: 31415355 PMCID: PMC6831387 DOI: 10.1097/md.0000000000016622] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE This study aimed to investigate the correlation of serum Jun-amino-terminal kinase (JNK) pathway-associated phosphatase (JKAP) level with disease risk, severity, inflammation, and treatment response to tumor necrosis factor (TNF)-α inhibitor in Crohn disease (CD) patients. METHOD Ninety-six active CD patients and 90 healthy controls (HCs) were consecutively enrolled. Serum JKAP level of participants was determined via enzyme-linked immunosorbent assay (ELISA). In CD patients, C-reactive protein (CRP), erythrocyte sedimentation rate, Crohn disease activity index (CDAI), and inflammatory cytokine levels (determined by ELISA) were recorded. All CD patients underwent infliximab (IFX) treatment for 12 weeks, then treatment response (defined as decrement of CDAI ≥70) was assessed at week 12 (W12). RESULTS Serum JKAP level in CD patients was lower compared to HCs, and it disclosed a good predictive value for decreased CD risk; meanwhile, it was negatively correlated with CRP level, CDAI score, TNF-α, interleukin (IL)-6, and IL-17 levels in CD patients. Sixty-eight (70.8%) patients achieved treatment response to IFX at W12, and JKAP level was increased at W12 compared to baseline. Interestingly, baseline JKAP level in response patients was decreased compared to nonresponse patients, and it exhibited a good predictive value for decreased treatment response to IFX, multivariate logistic regression revealed that JKAP was an independent factor for predicting reduced IFX response. CONCLUSION Circulating JKAP expression correlates with decreased disease risk, activity, and inflammation level, and it could be served as a novel biomarker for predicting reduced clinical response to TNF-α inhibitor in CD patients.
Collapse
Affiliation(s)
- Xue Shi
- Department of Anesthesiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan
| | - Wei Yang
- Department of Gastroenterology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang
| | - Nian Wang
- Division of Gastroenterology and Hepatology, Wuhan No.1 Hospital, Wuhan Hospital of Traditional Chinese and Western Hospital
| | - Junyi Zhu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Yang CY, Chiu LL, Chang CC, Chuang HC, Tan TH. Induction of DUSP14 ubiquitination by PRMT5-mediated arginine methylation. FASEB J 2018; 32:fj201800244RR. [PMID: 29920217 PMCID: PMC6219832 DOI: 10.1096/fj.201800244rr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/04/2018] [Indexed: 12/24/2022]
Abstract
Dual-specificity phosphatase (DUSP)14 (also known as MAP-kinase phosphatase 6) inhibits T-cell receptor (TCR) signaling and T-cell-mediated immune responses by inactivation of the TGF-β activated kinase 1 binding protein (TAB1)-TGF-β activated kinase 1 (TAK1) complex and ERK. DUSP14 phosphatase activity is induced by the E3 ligase TNF receptor associated factor (TRAF)2-mediated Lys63-linked ubiquitination. Here we report an interaction between DUSP14 and protein arginine methyltransferase (PRMT)5 by proximity ligation assay; similarly, DUSP14 directly interacted with TAB1 but not TAK1. DUSP14 is methylated by PRMT5 at arginine 17, 38, and 45 residues. The DUSP14 triple-methylation mutant was impaired in PRMT5-mediated arginine methylation, TRAF2-mediated lysine ubiquitination, and DUSP14 phosphatase activity. Consistently, DUSP14 methylation, TRAF2 binding, and DUSP14 ubiquitination were attenuated by PRMT5 short hairpin RNA knockdown. Furthermore, DUSP14 was inducibly interacted with PRMT5 and was methylated during TCR signaling in T cells. Together, these findings reveal a novel regulatory mechanism of DUSP14 by which PRMT5-mediated arginine methylation may sequentially stimulate TRAF2-mediated DUSP14 ubiquitination and phosphatase activity, leading to inhibition of TCR signaling.-Yang, C.-Y., Chiu, L.-L., Chang, C.-C., Chuang, H.-C., Tan, T.-H. Induction of DUSP14 ubiquitination by PRMT5-mediated arginine methylation.
Collapse
Affiliation(s)
- Chia-Yu Yang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Li-Li Chiu
- Department of Medical Education and Research, Taichung Veterans General Hospital, Taichung, Taiwan; and
| | - Chih-Chi Chang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Huai-Chia Chuang
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
| | - Tse-Hua Tan
- Immunology Research Center, National Health Research Institutes, Zhunan, Taiwan
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
23
|
Downregulation of the phosphatase JKAP/DUSP22 in T cells as a potential new biomarker of systemic lupus erythematosus nephritis. Oncotarget 2018; 7:57593-57605. [PMID: 27557500 PMCID: PMC5295375 DOI: 10.18632/oncotarget.11419] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 08/13/2016] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease that is characterized by systemic inflammation and multiple organ failures. Dysregulation of T cells plays a critical role in SLE pathogenesis. Our previous study indicates that JKAP (also named DUSP22) inhibits T-cell activation and that JKAP knockout mice develop spontaneous autoimmunity; therefore, we investigated whether JKAP downregulation is involved in SLE patients. JKAP protein levels in purified T cells were examined by immunoblotting using blood samples from 43 SLE patients and 32 healthy controls. SLE patients showed significantly decreased JKAP protein levels in peripheral blood T cells compared to healthy controls. JKAP protein levels in peripheral blood T cells were inversely correlated with SLE disease activity index (SLEDAI) and anti-dsDNA antibody levels. JKAP downregulation in T cells was highly correlated with daily urinary protein amounts and with poor renal outcome in lupus nephritis patients. Notably, the diagnostic power of JKAP downregulation in T cells for active lupus nephritis was higher than those of serum anti-dsDNA antibody, C3, and C4 levels. Moreover, T-cell-specific transgenic mice expressing a dominant-negative JKAP mutant developed spontaneous autoimmune nephritis. Furthermore, JKAP-deficient T cells overproduced complement components, soluble ICAM-1, and soluble VCAM-1 in the kidney; these cytokines have been reported to be involved in lupus nephritis. Taken together, JKAP downregulation in T cells is a novel diagnostic and prognostic biomarker for SLE nephritis.
Collapse
|
24
|
Mélard P, Idrissi Y, Andrique L, Poglio S, Prochazkova-Carlotti M, Berhouet S, Boucher C, Laharanne E, Chevret E, Pham-Ledard A, De Souza Góes AC, Guyonnet-Duperat V, Bibeyran A, Moreau-Gaudry F, Vergier B, Beylot-Barry M, Merlio JP, Cappellen D. Molecular alterations and tumor suppressive function of the DUSP22 (Dual Specificity Phosphatase 22) gene in peripheral T-cell lymphoma subtypes. Oncotarget 2018; 7:68734-68748. [PMID: 27626696 PMCID: PMC5356586 DOI: 10.18632/oncotarget.11930] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/31/2016] [Indexed: 12/27/2022] Open
Abstract
Monoallelic 6p25.3 rearrangements associated with DUSP22 (Dual Specificity Phosphatase 22) gene silencing have been reported in CD30+ peripheral T-cell lymphomas (PTCL), mostly with anaplastic morphology and of cutaneous origin. However, the mechanism of second allele silencing and the putative tumor suppressor function of DUSP22 have not been investigated so far. Here, we show that the presence, in most individuals, of an inactive paralog hampers genetic and epigenetic evaluation of the DUSP22 gene. Identification of DUSP22-specific single-nucleotide polymorphisms haplotypes and fluorescence in situ hybridization and epigenetic characterization of the paralog status led us to develop a comprehensive strategy enabling reliable identification of DUSP22 alterations. We showed that one cutaneous anaplastic large T-cell lymphomas (cALCL) case with monoallelic 6p25.3 rearrangement and DUSP22 silencing harbored exon 1 somatic mutations associated with second allele inactivation. Another cALCL case carried an intron 1 somatic splice site mutation with predicted deleterious exon skipping effect. Other tested PTCL cases with 6p25.3 rearrangement exhibited neither mutation nor deletion nor methylation accounting for silencing of the non-rearranged DUSP22 allele, thus inactivated by a so far unknown mechanism. We also characterized the expression status of four DUSP22 splice variants and found that they are all silenced in cALCL cases with 6p25.3 breakpoints. We finally showed that restoring expression of the physiologically predominant isoform in DUSP22-deficient malignant T cells inhibits cellular expansion by stimulating apoptosis and impairs soft agar clonogenicity and tumorigenicity. This study therefore shows that DUSP22 behaves as a tumor suppressor gene in PTCL.
Collapse
Affiliation(s)
- Pierre Mélard
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France.,Service de Pathologie, Centre Hospitalier Universitaire de Bordeaux, Hôpital Haut-Lévêque, F-33604 Pessac, France
| | - Yamina Idrissi
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France
| | - Laetitia Andrique
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France.,Service de Biologie des Tumeurs-Tumorothèque, Centre Hospitalier Universitaire de Bordeaux, Hôpital Haut-Lévêque, F-33604 Pessac, France
| | - Sandrine Poglio
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France
| | - Martina Prochazkova-Carlotti
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France
| | - Sabine Berhouet
- Service de Biologie des Tumeurs-Tumorothèque, Centre Hospitalier Universitaire de Bordeaux, Hôpital Haut-Lévêque, F-33604 Pessac, France
| | - Cécile Boucher
- Service de Biologie des Tumeurs-Tumorothèque, Centre Hospitalier Universitaire de Bordeaux, Hôpital Haut-Lévêque, F-33604 Pessac, France
| | - Elodie Laharanne
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France.,Service de Biologie des Tumeurs-Tumorothèque, Centre Hospitalier Universitaire de Bordeaux, Hôpital Haut-Lévêque, F-33604 Pessac, France
| | - Edith Chevret
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France
| | - Anne Pham-Ledard
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France.,Service de Dermatologie, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, F-33000 Bordeaux, France
| | - Andréa Carla De Souza Góes
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France.,Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, CEP 20550-013 Rio de Janeiro, Brazil
| | - Véronique Guyonnet-Duperat
- Plateforme de Vectorologie, Unité Mixte de Services (UMS TBM-Core), Centre National de la Recherche Scientifique (CNRS)- Institut National de la Santé et de la Recherche Médicale (Inserm)-Universitaire de Bordeaux, F-33076 Bordeaux, France
| | - Alice Bibeyran
- Plateforme de Vectorologie, Unité Mixte de Services (UMS TBM-Core), Centre National de la Recherche Scientifique (CNRS)- Institut National de la Santé et de la Recherche Médicale (Inserm)-Universitaire de Bordeaux, F-33076 Bordeaux, France
| | - François Moreau-Gaudry
- Plateforme de Vectorologie, Unité Mixte de Services (UMS TBM-Core), Centre National de la Recherche Scientifique (CNRS)- Institut National de la Santé et de la Recherche Médicale (Inserm)-Universitaire de Bordeaux, F-33076 Bordeaux, France.,Biothérapies des Maladies Génétiques et Cancers, Institut National de la Santé et de la Recherche Médicale (Inserm), U1035, Universitaire de Bordeaux, F-33076 Bordeaux, France
| | - Béatrice Vergier
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France.,Service de Pathologie, Centre Hospitalier Universitaire de Bordeaux, Hôpital Haut-Lévêque, F-33604 Pessac, France
| | - Marie Beylot-Barry
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France.,Service de Dermatologie, Centre Hospitalier Universitaire de Bordeaux, Hôpital Saint-André, F-33000 Bordeaux, France
| | - Jean-Philippe Merlio
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France.,Service de Biologie des Tumeurs-Tumorothèque, Centre Hospitalier Universitaire de Bordeaux, Hôpital Haut-Lévêque, F-33604 Pessac, France
| | - David Cappellen
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1053, Universitaire de Bordeaux, F-33076 Bordeaux, France.,Service de Biologie des Tumeurs-Tumorothèque, Centre Hospitalier Universitaire de Bordeaux, Hôpital Haut-Lévêque, F-33604 Pessac, France
| |
Collapse
|
25
|
Hamada N, Mizuno M, Tomita H, Iwamoto I, Hara A, Nagata KI. Expression analyses of Dusp22 (Dual-specificity phosphatase 22) in mouse tissues. Med Mol Morphol 2017; 51:111-117. [PMID: 29282540 DOI: 10.1007/s00795-017-0178-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 12/21/2017] [Indexed: 12/25/2022]
Abstract
Dusp22 (dual-specificity phosphatase 22) is considered to regulate various cellular processes through the regulation of protein dephosphorylation. In this study, we prepared a specific antibody against Dusp22, anti-Dusp22, and carried out expression analyses with mouse tissues and cultured cell lines. Western blotting analyses demonstrated a tissue-dependent expression profile of Dusp22 in the adult mouse, and strongly suggested the presence of isoforms with larger molecular masses. In fibroblast NIH3T3 cells, while both endogenous and Myc-tagged Dusp22 was diffusely distributed in the cytoplasm, Myc-Dusp22 was partially colocalized with actin cytoskeleton. From the obtained results, anti-Dusp22 was found to be a useful tool for biochemical and cell biological analyses of Dusp22.
Collapse
Affiliation(s)
- Nanako Hamada
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya, Kasugai, 480-0392, Japan
| | - Makoto Mizuno
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya, Kasugai, 480-0392, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Ikuko Iwamoto
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya, Kasugai, 480-0392, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, 501-1194, Japan
| | - Koh-Ichi Nagata
- Department of Molecular Neurobiology, Institute for Developmental Research, Aichi Human Service Center, 713-8 Kamiya, Kasugai, 480-0392, Japan.
| |
Collapse
|
26
|
Meeusen B, Janssens V. Tumor suppressive protein phosphatases in human cancer: Emerging targets for therapeutic intervention and tumor stratification. Int J Biochem Cell Biol 2017; 96:98-134. [PMID: 29031806 DOI: 10.1016/j.biocel.2017.10.002] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 02/06/2023]
Abstract
Aberrant protein phosphorylation is one of the hallmarks of cancer cells, and in many cases a prerequisite to sustain tumor development and progression. Like protein kinases, protein phosphatases are key regulators of cell signaling. However, their contribution to aberrant signaling in cancer cells is overall less well appreciated, and therefore, their clinical potential remains largely unexploited. In this review, we provide an overview of tumor suppressive protein phosphatases in human cancer. Along their mechanisms of inactivation in defined cancer contexts, we give an overview of their functional roles in diverse signaling pathways that contribute to their tumor suppressive abilities. Finally, we discuss their emerging roles as predictive or prognostic markers, their potential as synthetic lethality targets, and the current feasibility of their reactivation with pharmacologic compounds as promising new cancer therapies. We conclude that their inclusion in clinical practice has obvious potential to significantly improve therapeutic outcome in various ways, and should now definitely be pushed forward.
Collapse
Affiliation(s)
- Bob Meeusen
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium
| | - Veerle Janssens
- Laboratory of Protein Phosphorylation & Proteomics, Dept. of Cellular & Molecular Medicine, Faculty of Medicine, KU Leuven & Leuven Cancer Institute (LKI), KU Leuven, Belgium.
| |
Collapse
|
27
|
Ribeiro IP, Marques F, Barroso L, Rodrigues J, Caramelo F, Melo JB, Carreira IM. Genomic profile of oral squamous cell carcinomas with an adjacent leukoplakia or with an erythroleukoplakia that evolved after the treatment of primary tumor: A report of two cases. Mol Med Rep 2017; 16:6780-6786. [PMID: 28901451 PMCID: PMC5865835 DOI: 10.3892/mmr.2017.7428] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/23/2017] [Indexed: 12/21/2022] Open
Abstract
Oral leukoplakia and erythroleukoplakia are common oral potentially malignant disorders diagnosed in the oral cavity. The specific outcome of these lesions remains to be elucidated, as their malignant transformation rate exhibits great variation. The ability to predict which of those potentially malignant lesions are likely to progress to cancer would be vital to guide their future clinical management. The present study reported two patients with tongue squamous cell carcinoma: Case study 1 was diagnosed with a simultaneous leukoplakia and case study 2 developed an erythroleukoplakia following the primary tumor treatment. Whole genome copy number alterations were analyzed using array comparative genomic hybridization. The present study determined more genomic imbalances in the tissues from leukoplakia and erythroleukoplakia compared with their respective tumors. The present study also identified in tumor and potentially malignant lesions common alterations of chromosomal regions and genes, including FBXL5, UGT2B15, UGT2B28, KANSL1, GSTT1 and DUSP22, being some of these typical aberrations described in oral cancer and others are linked to chemoradioresistance. Several putative genes associated with hallmarks of malignancy that may have an important role in predicting the progression of leukoplakia and erythroleukoplakia to squamous cell carcinoma, namely gains in BNIPL, MCL1, STAG2, CSPP1 and ZNRF3 genes were also identified.
Collapse
Affiliation(s)
- Ilda P Ribeiro
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, 3000‑354 Coimbra, Portugal
| | - Francisco Marques
- Center of Investigation on Environment Genetics and Oncobiology, Faculty of Medicine, University of Coimbra, 3000‑354 Coimbra, Portugal
| | - Leonor Barroso
- Maxillofacial Surgery Department, Coimbra Hospital and University Centre, EPE, 3000‑075 Coimbra, Portugal
| | - Joana Rodrigues
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, 3000‑354 Coimbra, Portugal
| | - Francisco Caramelo
- Laboratory of Biostatistics and Medical Informatics, Faculty of Medicine, University of Coimbra, 3000‑354 Coimbra, Portugal
| | - Joana B Melo
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, 3000‑354 Coimbra, Portugal
| | - Isabel M Carreira
- Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, 3000‑354 Coimbra, Portugal
| |
Collapse
|
28
|
Shear force-based genetic screen reveals negative regulators of cell adhesion and protrusive activity. Proc Natl Acad Sci U S A 2017; 114:E7727-E7736. [PMID: 28847951 DOI: 10.1073/pnas.1616600114] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The model organism Dictyostelium discoideum has greatly facilitated our understanding of the signal transduction and cytoskeletal pathways that govern cell motility. Cell-substrate adhesion is downstream of many migratory and chemotaxis signaling events. Dictyostelium cells lacking the tumor suppressor PTEN show strongly impaired migratory activity and adhere strongly to their substrates. We reasoned that other regulators of migration could be obtained through a screen for overly adhesive mutants. A screen of restriction enzyme-mediated integration mutagenized cells yielded numerous mutants with the desired phenotypes, and the insertion sites in 18 of the strains were mapped. These regulators of adhesion and motility mutants have increased adhesion and decreased motility. Characterization of seven strains demonstrated decreased directed migration, flatness, increased filamentous actin-based protrusions, and increased signal transduction network activity. Many of the genes share homology to human genes and demonstrate the diverse array of cellular networks that function in adhesion and migration.
Collapse
|
29
|
Deficiency in VHR/DUSP3, a suppressor of focal adhesion kinase, reveals its role in regulating cell adhesion and migration. Oncogene 2017; 36:6509-6517. [DOI: 10.1038/onc.2017.255] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/24/2022]
|
30
|
Arruga F, Gizdic B, Bologna C, Cignetto S, Buonincontri R, Serra S, Vaisitti T, Gizzi K, Vitale N, Garaffo G, Mereu E, Diop F, Neri F, Incarnato D, Coscia M, Allan J, Piva R, Oliviero S, Furman RR, Rossi D, Gaidano G, Deaglio S. Mutations in NOTCH1 PEST domain orchestrate CCL19-driven homing of chronic lymphocytic leukemia cells by modulating the tumor suppressor gene DUSP22. Leukemia 2016; 31:1882-1893. [DOI: 10.1038/leu.2016.383] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 11/03/2016] [Accepted: 11/28/2016] [Indexed: 12/26/2022]
|
31
|
JNK Signaling: Regulation and Functions Based on Complex Protein-Protein Partnerships. Microbiol Mol Biol Rev 2016; 80:793-835. [PMID: 27466283 DOI: 10.1128/mmbr.00043-14] [Citation(s) in RCA: 346] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs), as members of the mitogen-activated protein kinase (MAPK) family, mediate eukaryotic cell responses to a wide range of abiotic and biotic stress insults. JNKs also regulate important physiological processes, including neuronal functions, immunological actions, and embryonic development, via their impact on gene expression, cytoskeletal protein dynamics, and cell death/survival pathways. Although the JNK pathway has been under study for >20 years, its complexity is still perplexing, with multiple protein partners of JNKs underlying the diversity of actions. Here we review the current knowledge of JNK structure and isoforms as well as the partnerships of JNKs with a range of intracellular proteins. Many of these proteins are direct substrates of the JNKs. We analyzed almost 100 of these target proteins in detail within a framework of their classification based on their regulation by JNKs. Examples of these JNK substrates include a diverse assortment of nuclear transcription factors (Jun, ATF2, Myc, Elk1), cytoplasmic proteins involved in cytoskeleton regulation (DCX, Tau, WDR62) or vesicular transport (JIP1, JIP3), cell membrane receptors (BMPR2), and mitochondrial proteins (Mcl1, Bim). In addition, because upstream signaling components impact JNK activity, we critically assessed the involvement of signaling scaffolds and the roles of feedback mechanisms in the JNK pathway. Despite a clarification of many regulatory events in JNK-dependent signaling during the past decade, many other structural and mechanistic insights are just beginning to be revealed. These advances open new opportunities to understand the role of JNK signaling in diverse physiological and pathophysiological states.
Collapse
|
32
|
Ou D, Wang Q, Huang Y, Zeng D, Wei T, Ding L, Li X, Zheng Q, Jin Y. Co-culture with neonatal cardiomyocytes enhances the proliferation of iPSC-derived cardiomyocytes via FAK/JNK signaling. BMC DEVELOPMENTAL BIOLOGY 2016; 16:11. [PMID: 27141946 PMCID: PMC4855360 DOI: 10.1186/s12861-016-0112-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 04/26/2016] [Indexed: 01/26/2023]
Abstract
Background We previously reported that the pluripotent stem cells can differentiate into cardiomyocytes (CMs) by co-culture with neonatal CMs (NCMs) in vitro. However, the involving mechanism is not clear. Methods Mouse induced pluripotent stem cells (iPSCs) were cultured in hanging drops to form embryoid bodies (EBs) and to induce myocardial differentiation. Co-culture of EBs and NCMs was established in a transwell insert system, while EBs grown alone in the wells were used as controls. Results Co-culture with NCMs markedly increased the generation of functional CMs from iPSCs. The focal adhesion kinase (FAK) phosphorylation, and c-Jun N-terminal kinase (JNK) phosphorylation in co-culture were higher than that in EBs grown alone. Treating FAK small interfering RNA (FAK siRNA) or specific inhibitor for JNK (SP600125) to iPSCs significantly reduced the phosphorylation of JNK and the expressions of Mef2c and Bcl-2. The expressions of cTnT and MLC-2V were also decreased. Our results revealed that co-culture with NCMs significantly enhance the differentiation ability of iPSCs by increasing Mef2c and Bcl-2 expressions concomitantly with a marked augment on cell proliferation through JNK signaling pathways. Conclusions These findings indicated that co-culture of EBs with NCMs induces genes expressed in a mature pattern and stimulates the proliferation of iPSC-derived CMs (iPS-CMs) by activating FAK/JNK signaling. Electronic supplementary material The online version of this article (doi:10.1186/s12861-016-0112-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dongbo Ou
- State Key Laboratory of Military Stomatology, Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, 1st Kang-fu Road, Xi'an, 710032, Shaanxi, China.,Department of Cardiology, NO. 422 Hospital of PLA, Zhanjiang, 524005, Guangdong, China.,Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Qi Wang
- Department of Cardiology, NO. 422 Hospital of PLA, Zhanjiang, 524005, Guangdong, China
| | - Yanjin Huang
- Department of Cardiology, NO. 422 Hospital of PLA, Zhanjiang, 524005, Guangdong, China
| | - Di Zeng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Ting Wei
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Lu Ding
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Xiaoli Li
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China
| | - Qiangsun Zheng
- Department of Cardiology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710038, Shaanxi, China.
| | - Yan Jin
- State Key Laboratory of Military Stomatology, Research and Development Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, 1st Kang-fu Road, Xi'an, 710032, Shaanxi, China. .,Research and Development Center for Tissue Engineering, Fourth Military Medical University, 1st Kang-fu Road, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
33
|
Lu D, Liu L, Ji X, Gao Y, Chen X, Liu Y, Liu Y, Zhao X, Li Y, Li Y, Jin Y, Zhang Y, McNutt MA, Yin Y. The phosphatase DUSP2 controls the activity of the transcription activator STAT3 and regulates TH17 differentiation. Nat Immunol 2015; 16:1263-73. [PMID: 26479789 DOI: 10.1038/ni.3278] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 08/24/2015] [Indexed: 02/08/2023]
Abstract
Deregulation of the TH17 subset of helper T cells is closely linked with immunological disorders and inflammatory diseases. However, the mechanism by which TH17 cells are regulated remains elusive. Here we found that the phosphatase DUSP2 (PAC1) negatively regulated the development of TH17 cells. DUSP2 was directly associated with the signal transducer and transcription activator STAT3 and attenuated its activity through dephosphorylation of STAT3 at Tyr705 and Ser727. DUSP2-deficient mice exhibited severe susceptibility to experimental colitis, with enhanced differentiation of TH17 cells and secretion of proinflammatory cytokines. In clinical patients with ulcerative colitis, DUSP2 was downregulated by DNA methylation and was not induced during T cell activation. Our data demonstrate that DUSP2 is a true STAT3 phosphatase that modulates the development of TH17 cells in the autoimmune response and inflammation.
Collapse
Affiliation(s)
- Dan Lu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Liang Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xin Ji
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yanan Gao
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xi Chen
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yu Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yang Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xuyang Zhao
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yan Li
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yunqiao Li
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yan Jin
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yu Zhang
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Michael A McNutt
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,Department of Pathology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China.,Peking-Tsinghua Center for Life Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
34
|
Decreased expression of dual specificity phosphatase 22 in colorectal cancer and its potential prognostic relevance for stage IV CRC patients. Tumour Biol 2015; 36:8531-5. [PMID: 26032091 DOI: 10.1007/s13277-015-3588-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Accepted: 05/19/2015] [Indexed: 02/06/2023] Open
Abstract
Dual specificity phosphatase 22 (DUSP22) is a novel dual specificity phosphatase that has been demonstrated to be a cancer suppressor gene associated with numerous biological and pathological processes. However, little is known of DUSP22 expression profiling in colorectal cancer and its prognostic value. Our study aims to investigate the role of DUSP22 expression in the prognosis of colorectal cancer. We detected the mRNA expression in 92 paired primary colorectal cancer tissues and the corresponding adjacent normal tissues by using QuantiGenePlex assay. The Friedman test was used to determine the statistical difference of gene expression. Kaplan-Meier survival analysis was performed. Mann-Whitney test and Kruskal-Wallis test were used to conduct data analyses to determine the prognostic value. Statistical significance was set at P < 0.05. In 74 of 92 cases, DUSP22 mRNA was reduced in primary colorectal cancer tissues, compared to the adjacent normal tissues. The mRNA levels of DUSP22 were significantly lower in colorectal cancer tissues than in adjacent normal tissues (0.0290 vs. 0.0658; P < 0.001). Low expression of DUSP22 correlated significantly with large tumor size (P = 0.013). No association was observed between DUSP22 mRNA expression and differentiation, histopathological type, tumor invasion, lymph node metastases, metastases, TNM stage, and Duke's phase (all P > 0.05). Kaplan-Meier analysis indicated that DUSP22 expression had no significant relationship with overall survival in all patients (P > 0.05). Interestingly, low expression level of DUSP22 in stage IV patients had a poor survival measures with a marginal P value (P = 0.07). Reduced DUSP22 expression was found in colorectal cancer specimens. Low expression level of DUSP22 in stage IV patients had a poor survival outcome. Further study is required for the investigation of the role of DUSP22 in colorectal cancer.
Collapse
|
35
|
WANG DONG, HAN SHENG, PENG RUI, JIAO CHENYU, WANG XING, HAN ZEGUANG, LI XIANGCHENG. DUSP28 contributes to human hepatocellular carcinoma via regulation of the p38 MAPK signaling. Int J Oncol 2014; 45:2596-604. [DOI: 10.3892/ijo.2014.2653] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 08/30/2014] [Indexed: 11/06/2022] Open
|
36
|
The phosphatase JKAP/DUSP22 inhibits T-cell receptor signalling and autoimmunity by inactivating Lck. Nat Commun 2014; 5:3618. [PMID: 24714587 DOI: 10.1038/ncomms4618] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 03/11/2014] [Indexed: 12/21/2022] Open
Abstract
JNK pathway-associated phosphatase (JKAP, also known as DUSP22 or JSP-1) is a JNK activator. The in vivo role of JKAP in immune regulation remains unclear. Here we report that JKAP directly inactivates Lck by dephosphorylating tyrosine-394 residue during T-cell receptor (TCR) signalling. JKAP-knockout T cells display enhanced cell proliferation and cytokine production. JKAP-knockout mice show enhanced T-cell-mediated immune responses and are more susceptible to experimental autoimmune encephalomyelitis (EAE). In addition, the recipient mice that are adoptively transferred with JKAP-knockout T cells show exacerbated EAE symptoms. Aged JKAP-knockout mice spontaneously develop inflammation and autoimmunity. Thus, our results indicate that JKAP is an important phosphatase that inactivates Lck in the TCR signalling turn-off stage, leading to suppression of T-cell-mediated immunity and autoimmunity.
Collapse
|
37
|
You H, Lei P, Andreadis ST. JNK is a novel regulator of intercellular adhesion. Tissue Barriers 2013; 1:e26845. [PMID: 24868495 PMCID: PMC3942331 DOI: 10.4161/tisb.26845] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 10/16/2013] [Accepted: 10/16/2013] [Indexed: 12/11/2022] Open
Abstract
c-Jun N-terminal Kinase (JNK) is a family of protein kinases, which are activated by stress stimuli such as inflammation, heat stress and osmotic stress, and regulate diverse cellular processes including proliferation, survival and apoptosis. In this review, we focus on a recently discovered function of JNK as a regulator of intercellular adhesion. We summarize the existing knowledge regarding the role of JNK during the formation of cell-cell junctions. The potential mechanisms and implications for processes requiring dynamic formation and dissolution of cell-cell junctions including wound healing, migration, cancer metastasis and stem cell differentiation are also discussed.
Collapse
Affiliation(s)
- Hui You
- Bioengineering Laboratory; Department of Chemical and Biological Engineering; University at Buffalo; The State University of New York; Amherst, NY USA
| | - Pedro Lei
- Bioengineering Laboratory; Department of Chemical and Biological Engineering; University at Buffalo; The State University of New York; Amherst, NY USA
| | - Stelios T Andreadis
- Bioengineering Laboratory; Department of Chemical and Biological Engineering; University at Buffalo; The State University of New York; Amherst, NY USA ; Department of Biomedical Engineering; University at Buffalo; The State University of New York; Amherst, NY USA ; Center for Excellence in Bioinformatics and Life Sciences; University at Buffalo; The State University of New York; Amherst, NY USA
| |
Collapse
|
38
|
Sassa T. The Role of Human-Specific Gene Duplications During Brain Development and Evolution. J Neurogenet 2013; 27:86-96. [DOI: 10.3109/01677063.2013.789512] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
39
|
Abstract
Phosphatases are important regulators of intracellular signaling events, and their functions have been implicated in many biological processes. Dual-specificity phosphatases (DUSPs), whose family currently contains 25 members, are phosphatases that can dephosphorylate both tyrosine and serine/threonine residues of their substrates. The archetypical DUSP, DUSP1/MKP1, was initially discovered to regulate the activities of MAP kinases by dephosphorylating the TXY motif in the kinase domain. However, although DUSPs were discovered more than a decade ago, only in the past few years have their various functions begun to be described. DUSPs can be categorized based on the presence or absence of a MAP kinase-interacting domain into typical DUSPs and atypical DUSPs, respectively. In this review, we discuss the current understanding of how the activities of typical DUSPs are regulated and how typical DUSPs can regulate the functions of their targets. We also summarize recent findings from several in vivo DUSP-deficient mouse models that studied the involvement of DUSPs during the development and functioning of T cells. Finally, we discuss briefly the potential roles of DUSPs in the regulation of non-MAP kinase targets, as well as in the modulation of tumorigenesis.
Collapse
Affiliation(s)
- Ching-Yu Huang
- Immunology Research Center, National Health Research Institutes, Zhunan, Miaoli County, 35053, Taiwan.
| | | |
Collapse
|
40
|
Tsai SY, Huang YL, Yang WH, Tang CH. Hepatocyte growth factor-induced BMP-2 expression is mediated by c-Met receptor, FAK, JNK, Runx2, and p300 pathways in human osteoblasts. Int Immunopharmacol 2012; 13:156-62. [PMID: 22504529 DOI: 10.1016/j.intimp.2012.03.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Revised: 03/16/2012] [Accepted: 03/29/2012] [Indexed: 12/24/2022]
Abstract
Hepatocyte growth factor (HGF) has been demonstrated to stimulate osteoblast proliferation and participated bone remodeling. Bone morphogenetic protein-2 (BMP-2) is a crucial mediator in bone formation during fracture healing. However, the effects of HGF in BMP-2 expression in human osteoblasts are large unknown. Here we found that HGF induced BMP-2 expression in human osteoblasts dose-dependently. HGF-mediated BMP-2 production was attenuated by c-Met inhibitor or siRNA. Pretreatment with FAK inhibitor or JNK inhibitor (SP600125) also blocked the potentiating action of HGF. Stimulation of osteoblasts with HGF enhanced FAK phosphorylation, JNK phosphorylation, and RunX2 translocation from cytosol to the nucleus. HGF-mediated Runx2 binding to BMP-2 promoter was inhibited by c-Met inhibitor, FAK inhibitor, and SP600125. The binding of Runx2 to the BMP-2 promoter, as well as the recruitment of p300 and the enhancement of histones H3 and H4 acetylation on the BMP-2 promoter was enhanced by HGF. Our results suggest that HGF increased BMP-2 production in human osteoblasts via the c-Met receptor/FAK/JNK/Runx2 and p300 signaling pathways.
Collapse
Affiliation(s)
- Shu-Yao Tsai
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | | | | | | |
Collapse
|
41
|
Wang JY, Yeh CL, Chou HC, Yang CH, Fu YN, Chen YT, Cheng HW, Huang CYF, Liu HP, Huang SF, Chen YR. Vaccinia H1-related phosphatase is a phosphatase of ErbB receptors and is down-regulated in non-small cell lung cancer. J Biol Chem 2011; 286:10177-84. [PMID: 21262974 DOI: 10.1074/jbc.m110.163295] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vaccinia H1-related phosphatase (VHR) is classified as a dual specificity phosphatase. Unlike typical dual specificity phosphatases, VHR lacks the MAPK-binding domain and shows poor activity against MAPKs. We found that EGF receptor (EGFR) was a direct substrate of VHR and that overexpression of VHR down-regulated EGFR phosphorylation, particularly at Tyr-992 residue. Expression of VHR inhibited the activation of phospholipase Cγ and protein kinase C, both downstream effectors of Tyr-992 phosphorylation of EGFR. Decreasing VHR expression by RNA interference caused higher EGFR phosphorylation at Tyr-992. In addition to EGFR, VHR also directly dephosphorylated ErbB2. Consistent with these results, suppression of VHR augmented the foci formation ability of H1299 non-small cell lung cancer (NSCLC) cells, whereas overexpression of VHR suppressed cell growth in both two- and three-dimensional cultures. Expression of VHR also suppressed tumor formation in a mouse xenograft model. Furthermore, VHR expression was significantly lower in NSCLC tissues in comparison to that in normal lung tissues. Collectively, this study shows that down-regulation of VHR expression enhances the signaling of ErbB receptors and may be involved in NSCLC pathogenesis.
Collapse
Affiliation(s)
- Jiz-Yuh Wang
- Division of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan 35053
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|