1
|
Baig MS, Barmpoutsi S, Bharti S, Weigert A, Hirani N, Atre R, Khabiya R, Sharma R, Sarup S, Savai R. Adaptor molecules mediate negative regulation of macrophage inflammatory pathways: a closer look. Front Immunol 2024; 15:1355012. [PMID: 38482001 PMCID: PMC10933033 DOI: 10.3389/fimmu.2024.1355012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/22/2024] [Indexed: 04/13/2024] Open
Abstract
Macrophages play a central role in initiating, maintaining, and terminating inflammation. For that, macrophages respond to various external stimuli in changing environments through signaling pathways that are tightly regulated and interconnected. This process involves, among others, autoregulatory loops that activate and deactivate macrophages through various cytokines, stimulants, and other chemical mediators. Adaptor proteins play an indispensable role in facilitating various inflammatory signals. These proteins are dynamic and flexible modulators of immune cell signaling and act as molecular bridges between cell surface receptors and intracellular effector molecules. They are involved in regulating physiological inflammation and also contribute significantly to the development of chronic inflammatory processes. This is at least partly due to their involvement in the activation and deactivation of macrophages, leading to changes in the macrophages' activation/phenotype. This review provides a comprehensive overview of the 20 adaptor molecules and proteins that act as negative regulators of inflammation in macrophages and effectively suppress inflammatory signaling pathways. We emphasize the functional role of adaptors in signal transduction in macrophages and their influence on the phenotypic transition of macrophages from pro-inflammatory M1-like states to anti-inflammatory M2-like phenotypes. This endeavor mainly aims at highlighting and orchestrating the intricate dynamics of adaptor molecules by elucidating the associated key roles along with respective domains and opening avenues for therapeutic and investigative purposes in clinical practice.
Collapse
Affiliation(s)
- Mirza S. Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Spyridoula Barmpoutsi
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
| | - Shreya Bharti
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| | - Nik Hirani
- MRC Centre for Inflammation Research, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Rajat Atre
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rakhi Khabiya
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rahul Sharma
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Shivmuni Sarup
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rajkumar Savai
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, Germany
- Max Planck Institute for Heart and Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| |
Collapse
|
2
|
Zhang R, Wang D, Ruan GX, Wang R, Li Y, Chen W, Huang H, Wang J, Meng L, Zhu Z, Lei D, Xu S, Ou X. Spliceosome component PHD finger 5A is essential for early B lymphopoiesis. Development 2024; 151:dev202247. [PMID: 38095286 DOI: 10.1242/dev.202247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024]
Abstract
The spliceosome, a multi-megadalton ribonucleoprotein complex, is essential for pre-mRNA splicing in the nucleus and ensuring genomic stability. Its precise and dynamic assembly is pivotal for its function. Spliceosome malfunctions can lead to developmental abnormalities and potentially contribute to tumorigenesis. The specific role of the spliceosome in B cell development is poorly understood. Here, we reveal that the spliceosomal U2 snRNP component PHD finger protein 5A (Phf5a) is vital for early B cell development. Loss of Phf5a results in pronounced defects in B cell development, causing an arrest at the transition from pre-pro-B to early pro-B cell stage in the bone marrow of mutant mice. Phf5a-deficient B cells exhibit impaired immunoglobulin heavy (IgH) chain expression due to defective V-to-DJ gene rearrangement. Mechanistically, our findings suggest that Phf5a facilitates IgH gene rearrangement by regulating the activity of recombination-activating gene endonuclease and influencing chromatin interactions at the Igh locus.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Daoqin Wang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Gui-Xin Ruan
- Medical School, Taizhou University, Taizhou 318000, China
| | - Ruisi Wang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuxing Li
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wenjing Chen
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hengjun Huang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Jing Wang
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Limin Meng
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhijian Zhu
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Dengfeng Lei
- Department of Ophthalmology, Southern University of Science and Technology Hospital, Shenzhen 518055, China
| | - Shengli Xu
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Republic of Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 2 Medical Drive MD9, Singapore 117593, Republic of Singapore
| | - Xijun Ou
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
3
|
Li Y, Ruan GX, Chen W, Huang H, Zhang R, Wang J, Ouyang Y, Zhu Z, Meng L, Wang R, Huo J, Xu S, Ou X. The histone H2B ubiquitination regulator Wac is essential for plasma cell differentiation. FEBS Lett 2023; 597:1748-1760. [PMID: 37171241 DOI: 10.1002/1873-3468.14633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 05/13/2023]
Abstract
Naïve B cells become activated and differentiate into antibody-secreting plasma cells (PCs) when encountering antigens. Here, we reveal that the WW domain-containing adapter protein with coiled-coil (Wac), which is important for histone H2B ubiquitination (ubH2B), is essential for PC differentiation. We demonstrate that B cell-specific Wac knockout mice have severely compromised T cell-dependent and -independent antibody responses. PC differentiation is drastically compromised despite undisturbed germinal center B cell response in the mutant mice. We also observe a significant reduction in global ubH2B in Wac-deficient B cells, which is correlated with downregulated expression of some genes critical for cell metabolism. Thus, our findings demonstrate an essential role of Wac-mediated ubH2B in PC differentiation and shed light on the epigenetic mechanisms underlying this process.
Collapse
Affiliation(s)
- Yuxing Li
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Gui-Xin Ruan
- Medical School, Taizhou University, Zhejiang, China
| | - Wenjing Chen
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Hengjun Huang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Rui Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jing Wang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yu Ouyang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Zhijian Zhu
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Limin Meng
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Ruisi Wang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jianxin Huo
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore City, Singapore
| | - Shengli Xu
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore City, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xijun Ou
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
4
|
Wang YF, Hu YQ, Hu YN, Bai YC, Wang H, Zhang Q. Expression and clinical significance of DOK3 in renal clear cell carcinoma. J Int Med Res 2023; 51:3000605231174974. [PMID: 37235715 DOI: 10.1177/03000605231174974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2023] Open
Abstract
OBJECTIVES Docking Protein 3 (DOK3) is an adapter protein that has been implicated in various cellular processes relevant to diseases, such as cancer. In this study, we aimed to evaluate the role of DOK3 in kidney renal clear cell carcinoma (KIRC) by examining how its expression levels are correlated with patient characteristics and prognosis. METHODS We analyzed KIRC-related data from The Cancer Genome Atlas and used several bioinformatics tools, such as LinkedOmics and Oncomine, to evaluate DOK3 mRNA expression in KIRC. DOK3 protein expression was examined in 150 clinical KIRC samples and 100 non-cancerous renal tissues with immunohistochemistry assays. The prognostic value of DOK3 mRNA expression on patient overall survival was analyzed retrospectively using Kaplan-Meier survival and Cox regression analyses. RESULTS DOK3 mRNA expression was notably higher in KIRC samples compared with normal tissues. Significant correlations were found between DOK3 mRNA expression levels and tumor size, lymph node metastasis, distant metastasis, and pathological grade using the bioinformatics data. This was confirmed at the protein level with immunohistochemistry data. Survival analyses indicated that elevated DOK3 expression is linked to a lower overall survival rate in KIRC patients. CONCLUSIONS DOK3 is a potential biomarker for determining KIRC patient clinical prognosis.
Collapse
Affiliation(s)
- Yi-Fan Wang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Bengbu Medical College, Bengbu, China
| | - Yu-Qi Hu
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu-Ning Hu
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yu-Chen Bai
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Heng Wang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Qi Zhang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
5
|
Huang H, Li Y, Zhang G, Ruan GX, Zhu Z, Chen W, Zou J, Zhang R, Wang J, Ouyang Y, Xu S, Ou X. The RNA-binding protein hnRNP F is required for the germinal center B cell response. Nat Commun 2023; 14:1731. [PMID: 36997512 PMCID: PMC10063658 DOI: 10.1038/s41467-023-37308-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 03/10/2023] [Indexed: 04/01/2023] Open
Abstract
The T cell-dependent (TD) antibody response involves the generation of high affinity, immunoglobulin heavy chain class-switched antibodies that are generated through germinal center (GC) response. This process is controlled by coordinated transcriptional and post-transcriptional gene regulatory mechanisms. RNA-binding proteins (RBPs) have emerged as critical players in post-transcriptional gene regulation. Here we demonstrate that B cell-specific deletion of RBP hnRNP F leads to diminished production of class-switched antibodies with high affinities in response to a TD antigen challenge. B cells deficient in hnRNP F are characterized by defective proliferation and c-Myc upregulation upon antigenic stimulation. Mechanistically, hnRNP F directly binds to the G-tracts of Cd40 pre-mRNA to promote the inclusion of Cd40 exon 6 that encodes its transmembrane domain, thus enabling appropriate CD40 cell surface expression. Furthermore, we find that hnRNP A1 and A2B1 can bind to the same region of Cd40 pre-mRNA but suppress exon 6 inclusion, suggesting that these hnRNPs and hnRNP F might antagonize each-other's effects on Cd40 splicing. In summary, our study uncovers an important posttranscriptional mechanism regulating the GC response.
Collapse
Affiliation(s)
- Hengjun Huang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yuxing Li
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Gaopu Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Gui-Xin Ruan
- Medical School, Taizhou University, Taizhou, 318000, China
| | - Zhijian Zhu
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wenjing Chen
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jia Zou
- Department of Computer Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Rui Zhang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jing Wang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yu Ouyang
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Shengli Xu
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, 138648, Singapore.
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
| | - Xijun Ou
- School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
6
|
Jin K, Qiu S, Chen B, Zhang Z, Zhang C, Zhou X, Yang L, Ai J, Wei Q. DOK3 promotes proliferation and inhibits apoptosis of prostate cancer via the NF-κB signaling pathway. Chin Med J (Engl) 2023; 136:423-432. [PMID: 36867541 PMCID: PMC10106266 DOI: 10.1097/cm9.0000000000002251] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND DOK3 (Downstream of kinase 3) is involved primarily with immune cell infiltration. Recent research reported the role of DOK3 in tumor progression, with opposite effects in lung cancer and gliomas; however, its role in prostate cancer (PCa) remains elusive. This study aimed to explore the role of DOK3 in PCa and to determine the mechanisms involved. METHODS To investigate the functions and mechanisms of DOK3 in PCa, we performed bioinformatic and biofunctional analyses. Samples from patients with PCa were collected from West China Hospital, and 46 were selected for the final correlation analysis. A lentivirus-based short hairpin ribonucleic acid (shRNA) carrier was established for silencing DOK3. A series of experiments involving the cell counting kit-8, bromodeoxyuridine, and flow cytometry assays were performed to identify cell proliferation and apoptosis. Changes in biomarkers from the nuclear factor kappa B (NF-κB) signaling pathway were detected to verify the relationship between DOK3 and the NF-κB pathway. A subcutaneous xenograft mouse model was performed to examine phenotypes after knocking down DOK3 in vivo . Rescue experiments with DOK3 knockdown and NF-κB pathway activation were designed to verify regulating effects. RESULTS DOK3 was up-regulated in PCa cell lines and tissues. In addition, a high level of DOK3 was predictive of higher pathological stages and worse prognoses. Similar results were observed with PCa patient samples. After silencing DOK3 in PCa cell lines 22RV1 and PC3, cell proliferation was significantly inhibited while apoptosis was promoted. Gene set enrichment analysis revealed that DOK3 function was enriched in the NF-κB pathway. Mechanism experiments determined that knockdown of DOK3 suppressed activation of the NF-κB pathway, increased the expressions of B-cell lymphoma-2 like 11 (BIM) and B-cell lymphoma-2 associated X (BAX), and decreased the expression of phosphorylated-P65 and X-linked inhibitor of apoptosis (XIAP). In the rescue experiments, pharmacological activation of NF-κB by tumor necrosis factor-α (TNF-α) partially recovered cell proliferation after the knockdown of DOK3. CONCLUSION Our findings suggest that overexpression of DOK3 promotes PCa progression by activating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Kun Jin
- Department of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shi Qiu
- Center of Biomedical Big Data, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Bo Chen
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zilong Zhang
- Department of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chichen Zhang
- Department of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xianghong Zhou
- Department of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lu Yang
- Department of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jianzhong Ai
- Department of Urology and Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Qiang Wei
- Department of Urology and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
7
|
Li Y, Ruan GX, Chen W, Huang H, Zhang R, Wang J, Li Y, Xu S, Ou X. RNA-Editing Enzyme ADAR1 p150 Isoform Is Critical for Germinal Center B Cell Response. THE JOURNAL OF IMMUNOLOGY 2022; 209:1071-1082. [DOI: 10.4049/jimmunol.2200149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/11/2022] [Indexed: 01/04/2023]
Abstract
Abstract
Adenosine deaminase acting on RNA (ADAR)1 is the principal enzyme for adenosine-to-inosine editing, an RNA modification–avoiding cytosolic nucleic acid sensor’s activation triggered by endogenous dsRNAs. Two ADAR1 isoforms exist in mammals, a longer IFN-inducible and mainly cytoplasm-localized p150 isoform and a shorter constitutively expressed and primarily nucleus-localized p110 isoform. Studies of ADAR1 mutant mice have demonstrated that ADAR1 is essential for multiple physiological processes, including embryonic development, innate immune response, and B and T lymphocyte development. However, it remained unknown whether ADAR1 plays a role in the humoral immune response. In this study, we conditionally delete Adar1 in activated B cells and show that ADAR1-deficient mice have a defective T cell–dependent Ab response and diminished germinal center (GC) B cells. Using various double mutant mice concurrently deficient in ADAR1 and different downstream dsRNA sensors, we demonstrate that ADAR1 regulates the GC response by preventing hyperactivation of the melanoma differentiation-associated protein 5 (MDA5) but not the protein kinase R or RNase L pathway. We also show that p150 is exclusively responsible for ADAR1’s function in the GC response, and the p110 isoform cannot substitute for the p150’s role, even when p110 is constitutively expressed in the cytoplasm. We further demonstrated that the dsRNA-binding but not the RNA-editing activity is required for ADAR1’s function in the GC response. Thus, our data suggest that the ADAR1 p150 isoform plays a crucial role in regulating the GC B cell response.
Collapse
Affiliation(s)
- Yuxing Li
- *Harbin Institute of Technology, Harbin, China
- †Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Gui-Xin Ruan
- †Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Wenjing Chen
- †Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Hengjun Huang
- †Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Rui Zhang
- †Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jing Wang
- †Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yan Li
- †Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Shengli Xu
- ‡Singapore Immunology Network, Agency for Science, Technology and Research, Singapore; and
- §Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xijun Ou
- †Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
8
|
Huang H, Zhang G, Ruan GX, Li Y, Chen W, Zou J, Zhang R, Wang J, Ji SJ, Xu S, Ou X. Mettl14-Mediated m6A Modification Is Essential for Germinal Center B Cell Response. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:1924-1936. [PMID: 35365563 DOI: 10.4049/jimmunol.2101071] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 02/07/2022] [Indexed: 12/25/2022]
Abstract
The germinal center (GC) response is essential for generating memory B and long-lived Ab-secreting plasma cells during the T cell-dependent immune response. In the GC, signals via the BCR and CD40 collaboratively promote the proliferation and positive selection of GC B cells expressing BCRs with high affinities for specific Ags. Although a complex gene transcriptional regulatory network is known to control the GC response, it remains elusive how the positive selection of GC B cells is modulated posttranscriptionally. In this study, we show that methyltransferase like 14 (Mettl14)-mediated methylation of adenosines at the position N 6 of mRNA (N 6-methyladenosine [m6A]) is essential for the GC B cell response in mice. Ablation of Mettl14 in B cells leads to compromised GC B cell proliferation and a defective Ab response. Interestingly, we unravel that Mettl14-mediated m6A regulates the expression of genes critical for positive selection and cell cycle regulation of GC B cells in a Ythdf2-dependent but Myc-independent manner. Furthermore, our study reveals that Mettl14-mediated m6A modification promotes mRNA decay of negative immune regulators, such as Lax1 and Tipe2, to upregulate genes requisite for GC B cell positive selection and proliferation. Thus, our findings suggest that Mettl14-mediated m6A modification plays an essential role in the GC B cell response.
Collapse
Affiliation(s)
- Hengjun Huang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Gaopu Zhang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Gui-Xin Ruan
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Yuxing Li
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Wenjing Chen
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jia Zou
- Department of Computer Science and Engineering, College of Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Rui Zhang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Jing Wang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China
| | - Sheng-Jian Ji
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China;
| | - Shengli Xu
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore; and.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xijun Ou
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China;
| |
Collapse
|
9
|
Ruan GX, Li Y, Chen W, Huang H, Zhang R, Chen C, Lam KP, Xu S, Ou X. The spliceosome component Usp39 controls B cell development by regulating immunoglobulin gene rearrangement. Cell Rep 2022; 38:110338. [PMID: 35139388 DOI: 10.1016/j.celrep.2022.110338] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 11/18/2021] [Accepted: 01/13/2022] [Indexed: 12/17/2022] Open
Abstract
The spliceosome is a large ribonucleoprotein complex responsible for pre-mRNA splicing and genome stability maintenance. Disruption of the spliceosome activity may lead to developmental disorders and tumorigenesis. However, the physiological role that the spliceosome plays in B cell development and function is still poorly defined. Here, we demonstrate that ubiquitin-specific peptidase 39 (Usp39), a spliceosome component of the U4/U6.U5 tri-snRNP complex, is essential for B cell development. Ablation of Usp39 in B cell lineage blocks pre-pro-B to pro-B cell transition in the bone marrow, leading to a profound reduction of mature B cells in the periphery. We show that Usp39 specifically regulates immunoglobulin gene rearrangement in a spliceosome-dependent manner, which involves modulating chromatin interactions at the Igh locus. Moreover, our results indicate that Usp39 deletion reduces the pre-malignant B cells in Eμ-Myc transgenic mice and significantly improves their survival.
Collapse
Affiliation(s)
- Gui-Xin Ruan
- Harbin Institute of Technology, Harbin 150001, China; Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yuxing Li
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Wenjing Chen
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Hengjun Huang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Rui Zhang
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Changxu Chen
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Kong-Peng Lam
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore; Departments of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Shengli Xu
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Xijun Ou
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China.
| |
Collapse
|
10
|
Yang YY, Ye L, Chen J, Qiu Y, Yin YL, Li P. Dok3 is involved in cisplatin-induced acute kidney injury via regulation of inflammation and apoptosis. Biochem Biophys Res Commun 2021; 569:132-138. [PMID: 34245977 DOI: 10.1016/j.bbrc.2021.06.097] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 06/20/2021] [Accepted: 06/30/2021] [Indexed: 11/17/2022]
Abstract
Cisplatin-induced acute kidney injury (AKI) is associated with high morbidity and mortality worldwide, but the underlying mechanisms are not fully understood. Downstream-of-kinase 3 (Dok3), a member of the Dok family of adaptor proteins plays a critical role in inflammatory response and immune regulation; however, the role of Dok3 in cisplatin-induced AKI remains unclear. This study explored the effect and potential molecular mechanisms of Dok3 in cisplatin-induced AKI using Dok3 knockout (Dok3-/-) and control mice (129S) with or without administration of a single intraperitoneal injection of cisplatin. Apoptosis was assessed by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, lactate dehydrogenase (LDH) release, and Hoechst staining. Inflammatory factors were measured using ELISA kits. Protein and gene expression levels were measured by western blot analysis and real-time PCR, respectively. The results showed that Dok3 was expressed in renal tubular epithelial cells. Dok3 expression was decreased in kidneys of mice treated with cisplatin and cisplatin-treated HK2 cells. Dok3-/- mice showed lower creatinine levels and NGAL expression, and increased survival rates compared to 129S mice. Cisplatin-induced production of TNF-α and IL-6, and renal tubular cell apoptosis was attenuated in Dok3-/- mice. In vitro experiments demonstrated that HK2 cells overexpressing Dok3 exhibited exacerbated cisplatin-induced apoptosis and production of TNF-α and IL-6. These findings demonstrate that Dok3 regulates cisplatin-induced AKI by regulating apoptosis and inflammation.
Collapse
Affiliation(s)
- Yong-Yu Yang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, 410011, China; Hunan Provincial Engineering Research Center of Translational Medical and Innovative Drug, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
| | - Ling Ye
- Department of Geriatrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jing Chen
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yue Qiu
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, 453003, China
| | - Ya-Ling Yin
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, 453003, China; School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, 453003, China; Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang, 453003, China; Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang, 453003, China.
| |
Collapse
|
11
|
Chen S, Duan Y, Wu Y, Yang D, An J. A Novel Integrated Metabolism-Immunity Gene Expression Model Predicts the Prognosis of Lung Adenocarcinoma Patients. Front Pharmacol 2021; 12:728368. [PMID: 34393804 PMCID: PMC8361602 DOI: 10.3389/fphar.2021.728368] [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: 06/21/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Although multiple metabolic pathways are involved in the initiation, progression, and therapy of lung adenocarcinoma (LUAD), the tumor microenvironment (TME) for immune cell infiltration that is regulated by metabolic enzymes has not yet been characterized. Methods: 517 LUAD samples and 59 non-tumor samples were obtained from The Cancer Genome Atlas (TCGA) database as the training cohort. Kaplan-Meier analysis and Univariate Cox analysis were applied to screen the candidate metabolic enzymes for their role in relation to survival rate in LUAD patients. A prognostic metabolic enzyme signature, termed the metabolic gene risk score (MGRS), was established based on multivariate Cox proportional hazards regression analysis and was verified in an independent test cohort, GSE31210. In addition, we analyzed the immune cell infiltration characteristics in patients grouped by their Risk Score. Furthermore, the prognostic value of these four enzymes was verified in another independent cohort by immunohistochemistry and an optimized model of the metabolic-immune protein risk score (MIPRS) was constructed. Results: The MGRS model comprising 4 genes (TYMS, NME4, LDHA, and SMOX) was developed to classify patients into high-risk and low-risk groups. Patients with a high-risk score had a poor prognosis and exhibited activated carbon and nucleotide metabolism, both of which were associated with changes to TME immune cell infiltration characteristics. In addition, the optimized MIPRS model showed more accurate predictive power in prognosis of LUAD. Conclusion: Our study revealed an integrated metabolic enzyme signature as a reliable prognostic tool to accurately predict the prognosis of LUAD.
Collapse
Affiliation(s)
- Songming Chen
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha, China.,Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
| | - Yumei Duan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Yanhao Wu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Desong Yang
- Thoracic Surgery Department II, Hunan Cancer Hospital & the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jian An
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Key Clinical Specialty, Branch of National Clinical Research Center for Respiratory Disease, Xiangya Hospital, Central South University, Changsha, China.,Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Clinical Research Center for Respiratory Diseases, Changsha, China
| |
Collapse
|
12
|
Gao WS, Qu YJ, Huai J, Wei H, Zhang Y, Yue SW. DOK3 is involved in microglial cell activation in neuropathic pain by interacting with GPR84. Aging (Albany NY) 2020; 13:389-410. [PMID: 33281117 PMCID: PMC7835011 DOI: 10.18632/aging.202144] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 09/09/2020] [Indexed: 01/12/2023]
Abstract
Adaptor molecule downstream of kinase-3 (DOK3) is a vital regulator of innate immune responses in macrophages and B cells, and G-protein-coupled receptor 84 (GPR84) is significant in mediating the biosynthesis and maintenance of inflammatory mediators that are induced by neuropathic pain in microglia. In the present study, we determined the role of DOK3 in activating microglia-induced neuropathic pain and investigated the underlying mechanisms associated with GPR84. We found that knockdown of DOK3 in microglial cells dramatically reduced the levels of inflammatory factors, and we uncovered a physical association between DOK3 and GPR84 in the induction of inflammatory responses. We also observed that neuropathic pain and inflammatory responses induced by chronic constriction injury (CCI) of the sciatic nerve or intrathecal injection of a GPR84 agonist were compromised in DOK3-/- mice in vivo. Finally, enforced expression of DOK3 provoked inflammatory responses, and administration of pregabalin relieved neuropathic pain via inhibition of DOK3 expression. In conclusion, DOK3 induced neuropathic pain in mice by interacting with GPR84 in microglia. We hypothesize that targeting the adaptor protein DOK3 may open new avenues for pharmaceutical approaches to the alleviation of neuropathic pain in the spinal cord.
Collapse
Affiliation(s)
- Wen-Shuang Gao
- Rehabilitation Center, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu-Juan Qu
- Rehabilitation Center, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Juan Huai
- Rehabilitation Center, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui Wei
- Rehabilitation Center, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yang Zhang
- Rehabilitation Center, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shou-Wei Yue
- Rehabilitation Center, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
13
|
Various Aspects of Calcium Signaling in the Regulation of Apoptosis, Autophagy, Cell Proliferation, and Cancer. Int J Mol Sci 2020; 21:ijms21218323. [PMID: 33171939 PMCID: PMC7664196 DOI: 10.3390/ijms21218323] [Citation(s) in RCA: 203] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/11/2022] Open
Abstract
Calcium (Ca2+) is a major second messenger in cells and is essential for the fate and survival of all higher organisms. Different Ca2+ channels, pumps, or exchangers regulate variations in the duration and levels of intracellular Ca2+, which may be transient or sustained. These changes are then decoded by an elaborate toolkit of Ca2+-sensors, which translate Ca2+ signal to intracellular operational cell machinery, thereby regulating numerous Ca2+-dependent physiological processes. Alterations to Ca2+ homoeostasis and signaling are often deleterious and are associated with certain pathological states, including cancer. Altered Ca2+ transmission has been implicated in a variety of processes fundamental for the uncontrolled proliferation and invasiveness of tumor cells and other processes important for cancer progression, such as the development of resistance to cancer therapies. Here, we review what is known about Ca2+ signaling and how this fundamental second messenger regulates life and death decisions in the context of cancer, with particular attention directed to cell proliferation, apoptosis, and autophagy. We also explore the intersections of Ca2+ and the therapeutic targeting of cancer cells, summarizing the therapeutic opportunities for Ca2+ signal modulators to improve the effectiveness of current anticancer therapies.
Collapse
|
14
|
Loh JT, Teo JKH, Lim HH, Lam KP. Emerging Roles of Downstream of Kinase 3 in Cell Signaling. Front Immunol 2020; 11:566192. [PMID: 33133079 PMCID: PMC7550416 DOI: 10.3389/fimmu.2020.566192] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 09/10/2020] [Indexed: 11/18/2022] Open
Abstract
Downstream of kinase (Dok) 3 is a member of the Dok family of adaptor proteins known to regulate signaling pathways downstream of various immunoreceptors. As Dok-3 lacks intrinsic catalytic activity, it functions primarily as a molecular scaffold to facilitate the nucleation of protein complexes in a regulated manner and hence, achieve specificity in directing signaling cascades. Since its discovery, considerable progress has been made toward defining the role of Dok-3 in limiting B cell-receptor signaling. Nonetheless, Dok-3 has since been implicated in the signaling of Toll-like and C-type lectin receptors. Emerging data further demonstrate that Dok-3 can act both as an activator and inhibitor, in lymphoid and non-lymphoid cell types, suggesting Dok-3 involvement in a plethora of signal transduction pathways. In this review, we will focus on the structure and expression profile of Dok-3 and highlight its role during signal transduction in B cells, innate cells as well as in bone and lung tissues.
Collapse
Affiliation(s)
- Jia Tong Loh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Joey Kay Hui Teo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - Hong-Hwa Lim
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Kong-Peng Lam
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore, Singapore.,Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,School of Biological Sciences, College of Science, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
15
|
Liu X, Chen F, Li W. Elevated expression of DOK3 indicates high suppressive immune cell infiltration and unfavorable prognosis of gliomas. Int Immunopharmacol 2020; 83:106400. [PMID: 32193105 DOI: 10.1016/j.intimp.2020.106400] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/07/2020] [Accepted: 03/10/2020] [Indexed: 02/07/2023]
Abstract
Docking protein 3 has been implicated in immune response, including interferon-β production in macrophage and plasma cell differentiation. And its importance in lung adenocarcinoma has been reported. However, studies about its role in gliomas are rare. In this study, we explored the clinical and prognostic characteristics of DOK3 expression in 921 glioma samples. Kaplan-Meier survival analysis and Cox regression analysis verified the independent unfavorable prognostic value and high prognostic accuracy of DOK3 expression for overall survival. Functional analysis with Database for Annotation, Visualization and Integrated Discovery (DAVID) and Gene Set Enrichment Analysis (GSEA) implied the involvement of DOK3 in immune related responses. Immune cell infiltration analysis with online tools, CIBERSORT and EPIC, showed that samples with higher DOK3 expression were infiltrated with much more macrophages. DOK3 was also found to be strongly positively correlated with marker genes of tumor-associated macrophages and M2 macrophages, not M1. Results of immunohistochemical staining also demonstrated that samples with higher DOK3 expression level were infiltrated with more microglia/macrophages and immunosuppressive M2 macrophages. In summary, our results demonstrated the correlation between high DOK3 expression level and malignant progression of gliomas, and the possible involvement of DOK3 in immunosuppressive responses in gliomas.
Collapse
Affiliation(s)
- Xiu Liu
- Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, PR China
| | - Feng Chen
- Department of Neuro-Oncology, Neurosurgery Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, PR China.
| | - Wenbin Li
- Department of Neuro-Oncology, Neurosurgery Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, PR China.
| |
Collapse
|
16
|
Xie Y, Ding J, Cui X, Wu M, Huang C, Zhang R, Wang J, Li X, Cen S, Zhou J. Screening of kinase inhibitors downregulating PD-L1 expression via on/in cell quantitative immunoblots. Eur J Pharm Sci 2020; 142:105088. [DOI: 10.1016/j.ejps.2019.105088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/02/2019] [Accepted: 09/21/2019] [Indexed: 01/05/2023]
|
17
|
Li T, Li B, Sara A, Ay C, Leung WY, Zhang Y, Dong Y, Liang Q, Zhang X, Weidner P, Gutting T, Behrens HM, Röcken C, Sung JJ, Ebert MP, Yu J, Burgermeister E. Docking protein-1 promotes inflammatory macrophage signaling in gastric cancer. Oncoimmunology 2019; 8:e1649961. [PMID: 31646096 DOI: 10.1080/2162402x.2019.1649961] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 07/23/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023] Open
Abstract
Docking protein-1 (DOK1) is a tumor suppressor frequently lost in malignant cells, however, it retains the ability to control activities of immune receptors in adjacent stroma cells of the tumor microenvironment. We therefore hypothesized that addressing DOK1 may be useful for cancer immunotherapy. DOK1 mRNA and DOK1 protein expression were downregulated in tumor cells of gastric cancer patients (n = 249). Conversely, its expression was up-regulated in cases positive for Epstein Barr Virus (EBV+) together with genes related to macrophage biology and targets of clinical immunotherapy such as programmed-cell-death-ligand-1 (PD-L1). Notably, high DOK1 positivity in stroma cells conferred poor prognosis in patients and correlated with high levels of inducible nitric oxide synthase in CD68+ tumor-associated macrophages. In macrophages derived from human monocytic leukemia cell lines, DOK1 (i) was inducible by agonists of the anti-diabetic transcription factor peroxisome proliferator-activated receptor-gamma (PPARγ), (ii) increased polarization towards an inflammatory phenotype, (iii) augmented nuclear factor-κB-dependent transcription of pro-inflammatory cytokines and (iv) reduced PD-L1 expression. These properties empowered DOK1+ macrophages to decrease the viability of human gastric cancer cells in contact-dependent co-cultures. DOK1 also reduced PD-L1 expression in human primary blood monocytes. Our data propose that the drugability of DOK1 may be exploited to reprogram myeloid cells and enforce the innate immune response against EBV+ human gastric cancer.
Collapse
Affiliation(s)
- Tong Li
- Institute of Digestive Disease and The Dept. of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Dept. of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Beifang Li
- Dept. of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Asgharpour Sara
- Dept. of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christine Ay
- Dept. of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Wing Yan Leung
- Institute of Digestive Disease and The Dept. of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yanquan Zhang
- Institute of Digestive Disease and The Dept. of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yujuan Dong
- Institute of Digestive Disease and The Dept. of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Qiaoyi Liang
- Institute of Digestive Disease and The Dept. of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- Institute of Digestive Disease and The Dept. of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Philip Weidner
- Dept. of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Tobias Gutting
- Dept. of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | | | - Christoph Röcken
- Dept. of Pathology, Christian-Albrechts University, Kiel, Germany
| | - Joseph Jy Sung
- Institute of Digestive Disease and The Dept. of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Matthias P Ebert
- Dept. of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jun Yu
- Institute of Digestive Disease and The Dept. of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Elke Burgermeister
- Dept. of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
18
|
Loh JT, Xu S, Huo JX, Kim SSY, Wang Y, Lam KP. Dok3-protein phosphatase 1 interaction attenuates Card9 signaling and neutrophil-dependent antifungal immunity. J Clin Invest 2019; 129:2717-2729. [PMID: 31180338 DOI: 10.1172/jci126341] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 04/25/2019] [Indexed: 12/29/2022] Open
Abstract
Invasive fungal infection is a serious health threat with high morbidity and mortality. Current antifungal drugs only demonstrate partial success in improving prognosis. Furthermore, mechanisms regulating host defense against fungal pathogens remain elusive. Here, we report that the downstream of kinase 3 (Dok3) adaptor negatively regulates antifungal immunity in neutrophils. Our data revealed that Dok3 deficiency increased phagocytosis, proinflammatory cytokine production, and netosis in neutrophils, thereby enhancing mutant mouse survival against systemic infection with a lethal dose of the pathogenic fungus Candida albicans. Biochemically, Dok3 recruited protein phosphatase 1 (PP1) to dephosphorylate Card9, an essential player in innate antifungal defense, to dampen downstream NF-κB and JNK activation and immune responses. Thus, Dok3 suppresses Card9 signaling, and disrupting Dok3-Card9 interaction or inhibiting PP1 activity represents therapeutic opportunities to develop drugs to combat candidaemia.
Collapse
Affiliation(s)
- Jia Tong Loh
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| | - Shengli Xu
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jian Xin Huo
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| | - Susana Soo-Yeon Kim
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore
| | - Yue Wang
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Biochemistry and
| | - Kong-Peng Lam
- Bioprocessing Technology Institute, Agency for Science, Technology and Research, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,School of Biological Sciences, College of Science, Nanyang Technological University, Singapore
| |
Collapse
|
19
|
Ho CM, Chen HL, Hu RH, Lee PH. Harnessing immunotherapy for liver recipients with hepatocellular carcinoma: a review from a transplant oncology perspective. Ther Adv Med Oncol 2019; 11:1758835919843463. [PMID: 31065295 PMCID: PMC6487770 DOI: 10.1177/1758835919843463] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/18/2019] [Indexed: 12/14/2022] Open
Abstract
Without stringent criteria, liver transplantation for hepatocellular carcinoma (HCC) can lead to high cancer recurrence and poor prognosis in the current treatment context. Checkpoint inhibitors can lead to long survival by targeting coinhibitory pathways and promoting T-cell activity; thus, they have great potential for cancer immunotherapy. Therapeutic modulation of cosignaling pathways may shift paradigms from surgical prevention of recurrence to oncological intervention. Herein, we review the available evidence from a therapeutic perspective and focus on immune microenvironment perturbation by immunosuppressants and checkpoint inhibitors. Partial and reversible interleukin-2 signaling blockade is the mainstream strategy of immunosuppression for graft protection. Programmed cell death protein 1 (PD-1) is abundantly expressed on human liver allograft-infiltrating T-cells, which proliferate considerably after programmed death-ligand 1 (PD-L1) blockade. Clinically, checkpoint inhibitors are used in heart, liver, and kidney recipients with various cancers. Rejection can occur after checkpoint inhibitor administration through acute T-cell-mediated, antibody-mediated, or chronic allograft rejection mechanisms. Nevertheless, liver recipients may demonstrate favorable responses to treatment for HCC recurrence without rejection. Pharmacodynamically, substantial degrees of receptor occupancy can be achieved with lower doses, with favorable clinical outcomes. Manipulation of the immune microenvironment is a therapeutic niche that balances seemingly conflicting anticancer and graft protection needs. Additional translational and clinical studies emphasizing the comparative effectiveness of signaling networks within the immune microenvironment and conducting overall assessment of the immune microenvironment may aid in creating a therapeutic window and benefiting future liver recipients with HCC recurrence.
Collapse
Affiliation(s)
- Cheng-Maw Ho
- Department of Surgery, National Taiwan University Hospital and College of Medicine, 7 Chung-Shan South Road, Taipei 100, Taiwan
| | - Hui-Ling Chen
- Hepatitis Research Center, National Taiwan University Hospital, Taipei, Taiwan
| | - Rey-Heng Hu
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Huang Lee
- Department of Surgery, National Taiwan University Hospital and College of Medicine, Taipei College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
20
|
Transcription factor YY1 is essential for iNKT cell development. Cell Mol Immunol 2018; 16:547-556. [PMID: 29500401 DOI: 10.1038/s41423-018-0002-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/09/2018] [Accepted: 01/09/2018] [Indexed: 12/21/2022] Open
Abstract
Invariant natural killer T (iNKT) cells develop from CD4+CD8+ double-positive (DP) thymocytes and express an invariant Vα14-Jα18 T-cell receptor (TCR) α-chain. Generation of these cells requires the prolonged survival of DP thymocytes to allow for Vα14-Jα18 gene rearrangements and strong TCR signaling to induce the expression of the iNKT lineage-specific transcription factor PLZF. Here, we report that the transcription factor Yin Yang 1 (YY1) is essential for iNKT cell formation. Thymocytes lacking YY1 displayed a block in iNKT cell development at the earliest progenitor stage. YY1-deficient thymocytes underwent normal Vα14-Jα18 gene rearrangements, but exhibited impaired cell survival. Deletion of the apoptotic protein BIM failed to rescue the defect in iNKT cell generation. Chromatin immunoprecipitation and deep-sequencing experiments demonstrated that YY1 directly binds and activates the promoter of the Plzf gene. Thus, YY1 plays essential roles in iNKT cell development by coordinately regulating cell survival and PLZF expression.
Collapse
|
21
|
PD-1 and its ligands are important immune checkpoints in cancer. Oncotarget 2018; 8:2171-2186. [PMID: 27974689 PMCID: PMC5356790 DOI: 10.18632/oncotarget.13895] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 11/21/2016] [Indexed: 12/31/2022] Open
Abstract
Checkpoint programmed death-1 (PD-1)/programmed cell death ligands (PD-Ls) have been identified as negative immunoregulatory molecules that promote immune evasion of tumor cells. The interaction of PD-1 and PD-Ls inhibits the function of T cells and tumor-infiltrating lymphocytes (TIL) while increasing the function of immunosuppressive regulatory T cells (Tregs). This condition causes the tumor cells to evade immune response. Thus, the blockade of PD-1/PD-L1 enhances anti-tumor immunity by reducing the number and/or the suppressive activity of Tregs and by restoring the activity of effector T cells. Furthermore, some monoclonal antibodies blockading PD-1/PD-Ls axis have achieved good effect and received Food and Drug Administration approval. The role of PD-1/PD-Ls in tumors has been well studied, but little is known on the mechanism by which PD-1 blocks T-cell activation. In this study, we provide a brief overview on the discovery and regulatory mechanism of PD-1 and PD-L1 dysregulation in tumors, as well as the function and signaling pathway of PD-1 and its ligands; their roles in tumor evasion and clinical treatment were also studied.
Collapse
|
22
|
A functional genomics predictive network model identifies regulators of inflammatory bowel disease. Nat Genet 2017; 49:1437-1449. [PMID: 28892060 PMCID: PMC5660607 DOI: 10.1038/ng.3947] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Accepted: 08/11/2017] [Indexed: 02/07/2023]
Abstract
A major challenge in inflammatory bowel disease (IBD) is the integration of diverse IBD data sets to construct predictive models of IBD. We present a predictive model of the immune component of IBD that informs causal relationships among loci previously linked to IBD through genome-wide association studies (GWAS) using functional and regulatory annotations that relate to the cells, tissues, and pathophysiology of IBD. Our model consists of individual networks constructed using molecular data generated from intestinal samples isolated from three populations of patients with IBD at different stages of disease. We performed key driver analysis to identify genes predicted to modulate network regulatory states associated with IBD, prioritizing and prospectively validating 12 of the top key drivers experimentally. This validated key driver set not only introduces new regulators of processes central to IBD but also provides the integrated circuits of genetic, molecular, and clinical traits that can be directly queried to interrogate and refine the regulatory framework defining IBD.
Collapse
|