1
|
Shukla M, Sarkar RR. Differential cellular communication in tumor immune microenvironment during early and advanced stages of lung adenocarcinoma. Mol Genet Genomics 2024; 299:100. [PMID: 39460829 DOI: 10.1007/s00438-024-02193-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024]
Abstract
Heterogeneous behavior of each cell type and their cross-talks in tumor immune microenvironment (TIME) refers to tumor immunological heterogeneity that emerges during tumor progression and represents formidable challenges for effective anti-tumor immune response and promotes drug resistance. To comprehensively elucidate the heterogeneous behavior of individual cell types and their interactions across different stages of tumor development at system level, a computational framework was devised that integrates cell specific data from single-cell RNASeq into networks illustrating interactions among signaling and metabolic response genes within and between cells in TIME. This study identified stage specific novel markers which remodel the cross-talks, thereby facilitating immune stimulation. Particularly, multicellular knockout of metabolic gene APOE (Apolipoprotein E in mast cell, myeloid cell and fibroblast) combined with signaling gene CAV1 (Caveolin1 in endothelial and epithelial cells) resulted in the activation of T-cell mediated signaling pathways. Additionally, this knockout also initiated intervention of cytotoxic gene regulations during tumor immune cell interactions at the early stage of Lung Adenocarcinoma (LUAD). Furthermore, a unique interaction motif from multiple cells emerged significant in regulating the overall immune response at the advanced stage of LUAD. Most significantly, FCER1G (Fc Fragment of IgE Receptor Ig) was identified as the common regulator in activating the anti-tumor immune response at both stages. Predicted markers exhibited significant association with patient overall survival in patient specific dataset. This study uncovers the significance of signaling and metabolic interplay within TIME and discovers important targets to enhance anti-tumor immune response at each stage of tumor development.
Collapse
Affiliation(s)
- Mudita Shukla
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ram Rup Sarkar
- Chemical Engineering and Process Development Division, CSIR-National Chemical Laboratory, Pune, Maharashtra, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
2
|
Cui Y, Liao Y, Chen Y, Zhao X, Zhang Y, Wang H, Li L, Zhang X, Chen K, Jia M, Tian J, Ruan X, Shi Y, Yang P, Chen J. Low expression of selenoprotein S induces oxidative damage in cartilages. J Trace Elem Med Biol 2024; 85:127492. [PMID: 38964025 DOI: 10.1016/j.jtemb.2024.127492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024]
Abstract
Low levels of the indispensable trace element selenium (Se) can cause oxidative stress and disrupt environmental homeostasis in humans and animals. Selenoprotein S (Selenos), of which Se is a key component, is a member of the selenoprotein family involved in various biological processes. This study aimed to investigate whether low-level SELENOS gene expression can induce oxidative stress and decrease the antioxidative capacity of chondrocytes. Compared with control cells, SELENOS-knockdown ATDC5 cells showed substantially higher dihydroethidium, reactive oxygen species and malondialdehyde levels, and lower superoxide dismutase (SOD) expression. Knockout of the gene in C57BL/6 mice increased the 8-hydroxy-2-deoxyguanosine level considerably and decreased SOD expression in cartilages relative to the levels in wild-type mice. The results showed that the increased nuclear factor erythroid 2-related factor 2/heme oxygenase-1 signaling mediated by low-level SELENOS expression was involved in oxidative damage. The proliferative zone of the cartilage growth plate of SELENOS-knockout mice was shortened, suggesting cartilage differentiation dysfunction. In conclusion, this study confirmed that low-level Selenos expression plays a role in oxidative stress in cartilages.
Collapse
Affiliation(s)
- Yixin Cui
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Yucheng Liao
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Yonghui Chen
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Xu Zhao
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Yi Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Hui Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Lian Li
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Xinhe Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Kunpan Chen
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Mingzhao Jia
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Jing Tian
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Xingran Ruan
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Yawen Shi
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| | - Pinglin Yang
- Department of Orthopaedic Surgery, Second Affiliated Hospital of Medical School of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, People's Republic of China.
| | - Jinghong Chen
- School of Public Health, Xi'an Jiaotong University Health Science Center, Key Laboratory of Trace Elements and Endemic Diseases, National Health and Family Planning Commission, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, People's Republic of China.
| |
Collapse
|
3
|
Magni S, Sawlekar R, Capelle CM, Tslaf V, Baron A, Zeng N, Mombaerts L, Yue Z, Yuan Y, Hefeng FQ, Gonçalves J. Inferring upstream regulatory genes of FOXP3 in human regulatory T cells from time-series transcriptomic data. NPJ Syst Biol Appl 2024; 10:59. [PMID: 38811598 PMCID: PMC11137136 DOI: 10.1038/s41540-024-00387-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/10/2024] [Indexed: 05/31/2024] Open
Abstract
The discovery of upstream regulatory genes of a gene of interest still remains challenging. Here we applied a scalable computational method to unbiasedly predict candidate regulatory genes of critical transcription factors by searching the whole genome. We illustrated our approach with a case study on the master regulator FOXP3 of human primary regulatory T cells (Tregs). While target genes of FOXP3 have been identified, its upstream regulatory machinery still remains elusive. Our methodology selected five top-ranked candidates that were tested via proof-of-concept experiments. Following knockdown, three out of five candidates showed significant effects on the mRNA expression of FOXP3 across multiple donors. This provides insights into the regulatory mechanisms modulating FOXP3 transcriptional expression in Tregs. Overall, at the genome level this represents a high level of accuracy in predicting upstream regulatory genes of key genes of interest.
Collapse
Affiliation(s)
- Stefano Magni
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Rucha Sawlekar
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
- Robotics and Artificial Intelligence, Department of Computer Science, Electrical and Space Engineering, Luleå University of Technology, Luleå, Sweden
| | - Christophe M Capelle
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Vera Tslaf
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
- Faculty of Science, Technology and Medicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
- Transversal Translational Medicine, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Alexandre Baron
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
| | - Ni Zeng
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg
| | - Laurent Mombaerts
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Zuogong Yue
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg
| | - Ye Yuan
- School of Artificial Intelligence and Automation, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Q Hefeng
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-Sur-Alzette, Luxembourg.
| | - Jorge Gonçalves
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, Luxembourg.
- Department of Plant Sciences, University of Cambridge, Cambridge, United Kingdom.
| |
Collapse
|
4
|
Yu SS, Du JL. Current views on selenoprotein S in the pathophysiological processes of diabetes-induced atherosclerosis: potential therapeutics and underlying biomarkers. Diabetol Metab Syndr 2024; 16:5. [PMID: 38172976 PMCID: PMC10763436 DOI: 10.1186/s13098-023-01247-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/24/2023] [Indexed: 01/05/2024] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) consistently ranks as the primary mortality factor among diabetic people. A thorough comprehension of the pathophysiological routes and processes activated by atherosclerosis (AS) caused by diabetes mellitus (DM), together with the recognition of new contributing factors, could lead to the discovery of crucial biomarkers and the development of innovative drugs against atherosclerosis. Selenoprotein S (SELENOS) has been implicated in the pathology and progression of numerous conditions, including diabetes, dyslipidemia, obesity, and insulin resistance (IR)-all recognized contributors to endothelial dysfunction (ED), a precursor event to diabetes-induced AS. Hepatic-specific deletion of SELENOS accelerated the onset and progression of obesity, impaired glucose tolerance and insulin sensitivity, and increased hepatic triglycerides (TG) and diacylglycerol (DAG) accumulation; SELENOS expression in subcutaneous and omental adipose tissue was elevated in obese human subjects, and act as a positive regulator for adipogenesis in 3T3-L1 preadipocytes; knockdown of SELENOS in Min6 β-cells induced β-cell apoptosis and reduced cell proliferation. SELENOS also participates in the early stages of AS, notably by enhancing endothelial function, curbing the expression of adhesion molecules, and lessening leukocyte recruitment-actions that collectively reduce the formation of foam cells. Furthermore, SELENOS forestalls the apoptosis of vascular smooth muscle cells (VSMCs) and macrophages, mitigates vascular calcification, and alleviates inflammation in macrophages and CD4+ T cells. These actions help stifle the creation of unstable plaque characterized by thinner fibrous caps, larger necrotic cores, heightened inflammation, and more extensive vascular calcification-features seen in advanced atherosclerotic lesion development. Additionally, serum SELENOS could function as a potential biomarker, and SELENOS single nucleotide polymorphisms (SNPs) rs4965814, rs28628459, and rs9806366, might be effective gene markers for atherosclerosis-related diseases in diabetes. This review accentuates the pathophysiological processes of atherosclerosis in diabetes and amasses current evidence on SELENOS's potential therapeutic benefits or as predictive biomarkers in the various stages of diabetes-induced atherosclerosis.
Collapse
Affiliation(s)
- Shan-Shan Yu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
- Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, 116011, Liaoning, China
| | - Jian-Ling Du
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
- Dalian Key Laboratory of Prevention and Treatment of Metabolic Diseases and the Vascular Complications, Dalian, 116011, Liaoning, China.
| |
Collapse
|
5
|
Atalay Şahar E, Ballar Kirmizibayrak P. Differential Expression and Function of SVIP in Breast Cancer Cell Lines and In Silico Analysis of Its Expression and Prognostic Potential in Human Breast Cancer. Cells 2023; 12:1362. [PMID: 37408196 DOI: 10.3390/cells12101362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 07/07/2023] Open
Abstract
The heterogeneity of cancer strongly suggests the need to explore additional pathways to target. As cancer cells have increased proteotoxic stress, targeting proteotoxic stress-related pathways such as endoplasmic reticulum stress is attracting attention as a new anticancer treatment. One of the downstream responses to endoplasmic reticulum stress is endoplasmic reticulum-associated degradation (ERAD), a major degradation pathway that facilitates proteasome-dependent degradation of unfolded or misfolded proteins. Recently, SVIP (small VCP/97-interacting protein), an endogenous ERAD inhibitor, has been implicated in cancer progression, especially in glioma, prostate, and head and neck cancers. Here, the data of several RNA-sequencing (RNA-seq) and gene array studies were combined to evaluate the SVIP gene expression analysis on a variety of cancers, with a particular focus on breast cancer. The mRNA level of SVIP was found to be significantly higher in primary breast tumors and correlated well with its promoter methylation status and genetic alterations. Strikingly, the SVIP protein level was found to be low despite increased mRNA levels in breast tumors compared to normal tissues. On the other hand, the immunoblotting analysis showed that the expression of SVIP protein was significantly higher in breast cancer cell lines compared to non-tumorigenic epithelial cell lines, while most of the key proteins of gp78-mediated ERAD did not exhibit such an expression pattern, except for Hrd1. Silencing of SVIP enhanced the proliferation of p53 wt MCF-7 and ZR-75-1 cells but not p53 mutant T47D and SK-BR-3 cells; however, it increased the migration ability of both types of cell lines. Importantly, our data suggest that SVIP may increase p53 protein levels in MCF7 cells by inhibiting Hrd1-mediated p53 degradation. Overall, our data reveal the differential expression and function of SVIP on breast cancer cell lines together with in silico data analysis.
Collapse
Affiliation(s)
- Esra Atalay Şahar
- Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, Izmir 35100, Turkey
| | - Petek Ballar Kirmizibayrak
- Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, Izmir 35100, Turkey
- Department of Biochemistry, Faculty of Pharmacy, Ege University, Izmir 35100, Turkey
| |
Collapse
|
6
|
Ghelichkhani F, Gonzalez FA, Kapitonova MA, Rozovsky S. Selenoprotein S Interacts with the Replication and Transcription Complex of SARS-CoV-2 by Binding nsp7. J Mol Biol 2023; 435:168008. [PMID: 36773692 PMCID: PMC9911985 DOI: 10.1016/j.jmb.2023.168008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 12/05/2022] [Accepted: 02/03/2023] [Indexed: 02/12/2023]
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) replicates and evades detection using ER membranes and their associated protein machinery. Among these hijacked human proteins is selenoprotein S (selenos). This selenoprotein takes part in the protein quality control, signaling, and the regulation of cytokine secretion. While the role of selenos in the viral life cycle is not yet known, it has been reported to interact with SARS-CoV-2 nonstructural protein 7 (nsp7), a viral protein essential for the replication of the virus. We set to study whether selenos and nsp7 interact directly and if they can still bind when nsp7 is bound to the replication and transcription complex of the virus. Using biochemical assays, we show that selenos binds directly to nsp7. In addition, we found that selenos can bind to nsp7 when it is in a complex with the coronavirus's minimal replication and transcription complex, comprised of nsp7, nsp8, and the RNA-dependent RNA polymerase nsp12. In addition, through crosslinking experiments, we mapped the interaction sites of selenos and nsp7 in the replication complex and showed that the hydrophobic segment of selenos is essential for binding to nsp7. This arrangement leaves an extended helix and the intrinsically disordered segment of selenos-including the reactive selenocysteine-exposed and free to potentially recruit additional proteins to the replication and transcription complex.
Collapse
Affiliation(s)
- Farid Ghelichkhani
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Fabio A Gonzalez
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Mariia A Kapitonova
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA
| | - Sharon Rozovsky
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA.
| |
Collapse
|
7
|
Ghelichkhani F, Gonzalez FA, Kapitonova MA, Schaefer-Ramadan S, Liu J, Cheng R, Rozovsky S. Selenoprotein S: A versatile disordered protein. Arch Biochem Biophys 2022; 731:109427. [PMID: 36241082 PMCID: PMC10026367 DOI: 10.1016/j.abb.2022.109427] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/16/2022]
Abstract
Selenoprotein S (selenos) is a small, intrinsically disordered membrane protein that is associated with various cellular functions, such as inflammatory processes, cellular stress response, protein quality control, and signaling pathways. It is primarily known for its contribution to the ER-associated degradation (ERAD) pathway, which governs the extraction of misfolded proteins or misassembled protein complexes from the ER to the cytosol for degradation by the proteasome. However, selenos's other cellular roles in signaling are equally vital, including the control of transcription factors and cytokine levels. Consequently, genetic polymorphisms of selenos are associated with increased risk for diabetes, dyslipidemia, and cardiovascular diseases, while high expression levels correlate with poor prognosis in several cancers. Its inhibitory role in cytokine secretion is also exploited by viruses. Since selenos binds multiple protein complexes, however, its specific contributions to various cellular pathways and diseases have been difficult to establish. Thus, the precise cellular functions of selenos and their interconnectivity have only recently begun to emerge. This review aims to summarize recent insights into the structure, interactome, and cellular roles of selenos.
Collapse
Affiliation(s)
- Farid Ghelichkhani
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, USA
| | - Fabio A Gonzalez
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, USA
| | - Mariia A Kapitonova
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, USA
| | | | - Jun Liu
- Enlaza Therapeutics, 11099 N. Torrey Pines Rd, suite 290, La Jolla, CA, 92037, USA
| | - Rujin Cheng
- NGM Biopharmaceuticals, Inc., 333 Oyster Point Blvd, South San Francisco, CA, 94080, USA
| | - Sharon Rozovsky
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, USA.
| |
Collapse
|
8
|
Wang T, Dang N, Tang G, Li Z, Li X, Shi B, Xu Z, Li L, Yang X, Xu C, Ye K. Integrating bulk and single-cell RNA sequencing reveals cellular heterogeneity and immune infiltration in hepatocellular carcinoma. Mol Oncol 2022; 16:2195-2213. [PMID: 35124891 PMCID: PMC9168757 DOI: 10.1002/1878-0261.13190] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/07/2021] [Accepted: 02/04/2022] [Indexed: 11/09/2022] Open
Abstract
Efficacy of immunotherapy in hepatocellular carcinoma (HCC) is blocked by its high degree of inter- and intra-tumor heterogeneity and immunosuppressive tumor microenvironment. However, the correlation between tumor heterogeneity and immunosuppressive microenvironment in HCC has not been well addressed. Here, we endeavored to dissect inter- and intra-tumor heterogeneity in HCC and uncover how they contribute to the immunosuppressive microenvironment. We performed consensus molecular subtyping with non-negative matrix factorization (NMF) clustering to stratify the inter-heterogeneity profile of HCC tumors. We grouped HCC tumors from the Cancer Genome Atlas (TCGA) patients into three subtypes (S1, S2 and S3), where S1 was characterized as a 'hot tumor' profile with high expression level of T cell genes and rate of immune scores. S2 was characterized as a 'cold tumor' profile with the highest tumor purity score, and S3 as an 'immunosuppressed tumor' profile with the poorest prognosis and a high expression level of immunosuppressive genes such as cytotoxic T-lymphocyte-associated protein-4, TIGIT, and PDCD1. Moreover, we combined weighted gene co-expression network analysis and single-cell regulatory network inference and clustering (SCENIC) in the single-cell dataset of the S3-like subtype (CS3) and identified a transcription factor, BATF, which could upregulate immunosuppressive genes. Finally, we identified a cell interaction network in which a myeloid-derived suppressor cell-like macrophage subtype could promote the formation of immunosuppressive T-cells.
Collapse
Affiliation(s)
- Tingjie Wang
- School of Automation Science and EngineeringFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityChina
| | - Ningxin Dang
- Genome InstituteThe First Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | - Guangbo Tang
- School of Life Science and TechnologyXi’an Jiaotong UniversityXi’an, ShaanxiChina
| | - Zihang Li
- School of Life Science and TechnologyXi’an Jiaotong UniversityXi’an, ShaanxiChina
| | - Xiujuan Li
- School of Automation Science and EngineeringFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityChina
| | - Bingyin Shi
- Department of EndocrinologyThe First Affiliated Hospital of Xi’an Jiaotong UniversityChina
| | - Zhong Xu
- Guizhou Provincial People's HospitalGuiyangChina
| | - Lei Li
- School of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xiaofei Yang
- Genome InstituteThe First Affiliated Hospital of Xi’an Jiaotong UniversityChina
- School of Computer Science and TechnologyFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityChina
| | - Chuanrui Xu
- School of PharmacyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Kai Ye
- School of Automation Science and EngineeringFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityChina
- Genome InstituteThe First Affiliated Hospital of Xi’an Jiaotong UniversityChina
- School of Life Science and TechnologyXi’an Jiaotong UniversityXi’an, ShaanxiChina
- Faculty of ScienceLeiden UniversityThe Netherlands
- MOE Key Lab for Intelligent Networks & Networks SecurityFaculty of Electronic and Information EngineeringXi’an Jiaotong UniversityChina
| |
Collapse
|
9
|
Capelle CM, Chen A, Zeng N, Baron A, Grzyb K, Arns T, Skupin A, Ollert M, Hefeng FQ. Stress hormone signaling inhibits Th1 polarization in a CD4 T-cell-intrinsic manner via mTORC1 and the circadian gene PER1. Immunology 2022; 165:428-444. [PMID: 35143696 PMCID: PMC9426625 DOI: 10.1111/imm.13448] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/18/2022] [Accepted: 01/24/2022] [Indexed: 11/30/2022] Open
Abstract
Stress hormones are believed to skew the CD4 T‐cell differentiation towards a Th2 response via a T‐cell‐extrinsic mechanism. Using isolated primary human naïve and memory CD4 T cells, here we show that both adrenergic‐ and glucocorticoid‐mediated stress signalling pathways play a CD4 naïve T‐cell‐intrinsic role in regulating the Th1/Th2 differentiation balance. Both stress hormones reduced the Th1 programme and cytokine production by inhibiting mTORC1 signalling via two parallel mechanisms. Stress hormone signalling inhibited mTORC1 in naïve CD4 T cells (1) by affecting the PI3K/AKT pathway and (2) by regulating the expression of the circadian rhythm gene, period circadian regulator 1 (PER1). Both stress hormones induced the expression of PER1, which inhibited mTORC1 signalling, thus reducing Th1 differentiation. This previously unrecognized cell‐autonomous mechanism connects stress hormone signalling with CD4 T‐cell differentiation via mTORC1 and a specific circadian clock gene, namely PER1.
Collapse
Affiliation(s)
- Christophe M Capelle
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg.,Faculty of Science, Technology and Communication, University of Luxembourg, 2, avenue de Université, L-4365, Esch-sur-Alzette, Luxembourg
| | - Anna Chen
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg
| | - Ni Zeng
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg.,Faculty of Science, Technology and Communication, University of Luxembourg, 2, avenue de Université, L-4365, Esch-sur-Alzette, Luxembourg
| | - Alexandre Baron
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6, avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Thais Arns
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6, avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 6, avenue du Swing, L-4367, Belvaux, Luxembourg
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Center, Odense Research Center for Anaphylaxis (ORCA), University of Southern Denmark, Odense, 5000 C, Denmark
| | - Feng Q Hefeng
- Department of Infection and Immunity, Luxembourg Institute of Health (LIH), 29, rue Henri Koch, L-4354, Esch-sur-Alzette, Luxembourg.,Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, D-45122, Essen, Germany
| |
Collapse
|
10
|
Zeng N, Capelle CM, Baron A, Kobayashi T, Cire S, Tslaf V, Leonard C, Coowar D, Koseki H, Westendorf AM, Buer J, Brenner D, Krüger R, Balling R, Ollert M, Hefeng FQ. DJ-1 depletion prevents immunoaging in T-cell compartments. EMBO Rep 2022; 23:e53302. [PMID: 35037711 PMCID: PMC8892345 DOI: 10.15252/embr.202153302] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 12/09/2021] [Accepted: 12/16/2021] [Indexed: 12/22/2022] Open
Abstract
Decline in immune function during aging increases susceptibility to different aging-related diseases. However, the underlying molecular mechanisms, especially the genetic factors contributing to imbalance of naïve/memory T-cell subpopulations, still remain largely elusive. Here, we show that loss of DJ-1 encoded by PARK7/DJ-1, causing early-onset familial Parkinson's disease (PD), unexpectedly diminished signs of immunoaging in T-cell compartments of both human and mice. Compared with two gender-matched unaffected siblings of similar ages, the index PD patient with DJ-1 deficiency showed a decline in many critical immunoaging features, including almost doubled non-senescent T cells. The observation was further consolidated by the results in 45-week-old DJ-1 knockout mice. Our data demonstrated that DJ-1 regulates several immunoaging features via hematopoietic-intrinsic and naïve-CD8-intrinsic mechanisms. Mechanistically, DJ-1 depletion reduced oxidative phosphorylation (OXPHOS) and impaired TCR sensitivity in naïve CD8 T cells at a young age, accumulatively leading to a reduced aging process in T-cell compartments in older mice. Our finding suggests an unrecognized critical role of DJ-1 in regulating immunoaging, discovering a potent target to interfere with immunoaging- and aging-associated diseases.
Collapse
Affiliation(s)
- Ni Zeng
- Department of Infection and ImmunityLuxembourg Institute of Health (LIH)Esch‐sur‐AlzetteLuxembourg
- Faculty of Science, Technology and MedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Christophe M Capelle
- Department of Infection and ImmunityLuxembourg Institute of Health (LIH)Esch‐sur‐AlzetteLuxembourg
- Faculty of Science, Technology and MedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
| | - Alexandre Baron
- Department of Infection and ImmunityLuxembourg Institute of Health (LIH)Esch‐sur‐AlzetteLuxembourg
| | - Takumi Kobayashi
- Department of Infection and ImmunityLuxembourg Institute of Health (LIH)Esch‐sur‐AlzetteLuxembourg
| | - Severine Cire
- Department of Infection and ImmunityLuxembourg Institute of Health (LIH)Esch‐sur‐AlzetteLuxembourg
| | - Vera Tslaf
- Department of Infection and ImmunityLuxembourg Institute of Health (LIH)Esch‐sur‐AlzetteLuxembourg
- Faculty of Science, Technology and MedicineUniversity of LuxembourgEsch‐sur‐AlzetteLuxembourg
- Transversal Translational MedicineLuxembourg Institute of Health (LIH)StrassenLuxembourg
| | - Cathy Leonard
- Department of Infection and ImmunityLuxembourg Institute of Health (LIH)Esch‐sur‐AlzetteLuxembourg
| | - Djalil Coowar
- Luxembourg Centre for Systems Biomedicine (LCSB)University of LuxembourgBelvauxLuxembourg
| | - Haruhiko Koseki
- Laboratory for Developmental GeneticsRIKEN Center for Integrative Medical SciencesYokohamaJapan
- AMED‐CRESTJapanese Agency for Medical Research and DevelopmentYokohamaJapan
| | - Astrid M Westendorf
- Institute of Medical MicrobiologyUniversity Hospital EssenUniversity Duisburg‐EssenEssenGermany
| | - Jan Buer
- Institute of Medical MicrobiologyUniversity Hospital EssenUniversity Duisburg‐EssenEssenGermany
| | - Dirk Brenner
- Department of Infection and ImmunityLuxembourg Institute of Health (LIH)Esch‐sur‐AlzetteLuxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB)University of LuxembourgBelvauxLuxembourg
| | - Rejko Krüger
- Transversal Translational MedicineLuxembourg Institute of Health (LIH)StrassenLuxembourg
- Luxembourg Centre for Systems Biomedicine (LCSB)University of LuxembourgBelvauxLuxembourg
- Centre Hospitalier de Luxembourg (CHL)LuxembourgLuxembourg
| | - Rudi Balling
- Luxembourg Centre for Systems Biomedicine (LCSB)University of LuxembourgBelvauxLuxembourg
- Present address:
Institute of Molecular PsychiatryUniversity of BonnBonnGermany
| | - Markus Ollert
- Department of Infection and ImmunityLuxembourg Institute of Health (LIH)Esch‐sur‐AlzetteLuxembourg
- Department of Dermatology and Allergy CenterOdense Research Center for Anaphylaxis (ORCA)University of Southern DenmarkOdenseDenmark
| | - Feng Q Hefeng
- Department of Infection and ImmunityLuxembourg Institute of Health (LIH)Esch‐sur‐AlzetteLuxembourg
- Institute of Medical MicrobiologyUniversity Hospital EssenUniversity Duisburg‐EssenEssenGermany
| |
Collapse
|
11
|
Cheng R, Liu J, Daithankar V, Rozovsky S. Applying selenocysteine-mediated expressed protein ligation to prepare the membrane enzyme selenoprotein S. Methods Enzymol 2022; 662:159-185. [PMID: 35101209 DOI: 10.1016/bs.mie.2021.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The intrinsically disordered membrane-bound selenoprotein s (selenos) takes part in the protein quality control pathway, vesicle trafficking, and NF-kB signaling. The reactive selenocysteine (Sec) at the penultimate position is responsible for its enzymatic activity. We report the preparation of the soluble segment as well as the full-length selenos using expressed protein ligation. This chapter discusses the practical considerations of expressed protein ligation using selenopeptides and describes our optimized procedure for the semi-synthesis of selenos.
Collapse
Affiliation(s)
- Rujin Cheng
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, United States
| | - Jun Liu
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, United States; Department of Pharmaceutical Chemistry, University of California, San Francisco, CA, United States
| | - Vidyadhar Daithankar
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, United States; Syva formulation, Siemens Healthineers, Newark, DE, United States
| | - Sharon Rozovsky
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, United States.
| |
Collapse
|
12
|
Schomburg L. Selenium Deficiency Due to Diet, Pregnancy, Severe Illness, or COVID-19-A Preventable Trigger for Autoimmune Disease. Int J Mol Sci 2021; 22:8532. [PMID: 34445238 PMCID: PMC8395178 DOI: 10.3390/ijms22168532] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/30/2021] [Accepted: 08/06/2021] [Indexed: 12/11/2022] Open
Abstract
The trace element selenium (Se) is an essential part of the human diet; moreover, increased health risks have been observed with Se deficiency. A sufficiently high Se status is a prerequisite for adequate immune response, and preventable endemic diseases are known from areas with Se deficiency. Biomarkers of Se status decline strongly in pregnancy, severe illness, or COVID-19, reaching critically low concentrations. Notably, these conditions are associated with an increased risk for autoimmune disease (AID). Positive effects on the immune system are observed with Se supplementation in pregnancy, autoimmune thyroid disease, and recovery from severe illness. However, some studies reported null results; the database is small, and randomized trials are sparse. The current need for research on the link between AID and Se deficiency is particularly obvious for rheumatoid arthritis and type 1 diabetes mellitus. Despite these gaps in knowledge, it seems timely to realize that severe Se deficiency may trigger AID in susceptible subjects. Improved dietary choices or supplemental Se are efficient ways to avoid severe Se deficiency, thereby decreasing AID risk and improving disease course. A personalized approach is needed in clinics and during therapy, while population-wide measures should be considered for areas with habitual low Se intake. Finland has been adding Se to its food chain for more than 35 years-a wise and commendable decision, according to today's knowledge. It is unfortunate that the health risks of Se deficiency are often neglected, while possible side effects of Se supplementation are exaggerated, leading to disregard for this safe and promising preventive and adjuvant treatment options. This is especially true in the follow-up situations of pregnancy, severe illness, or COVID-19, where massive Se deficiencies have developed and are associated with AID risk, long-lasting health impairments, and slow recovery.
Collapse
Affiliation(s)
- Lutz Schomburg
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institut für Experimentelle Endokrinologie, Cardiovascular-Metabolic-Renal (CMR)-Research Center, Hessische Straße 3-4, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|