1
|
Deobagkar-Lele M, Crawford G, Crockford TL, Back J, Hodgson R, Bhandari A, Bull KR, Cornall RJ. B cells require DOCK8 to elicit and integrate T cell help when antigen is limiting. Sci Immunol 2024; 9:eadd4874. [PMID: 39121196 PMCID: PMC7616390 DOI: 10.1126/sciimmunol.add4874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/01/2023] [Accepted: 07/12/2024] [Indexed: 08/11/2024]
Abstract
Dedicator of cytokinesis 8 (DOCK8) immunodeficiency syndrome is characterized by a failure of the germinal center response, a process involving the proliferation and positive selection of antigen-specific B cells. Here, we describe how DOCK8-deficient B cells are blocked at a light-zone checkpoint in the germinal centers of immunized mice, where they are unable to respond to T cell-dependent survival and selection signals and consequently differentiate into plasma cells or memory B cells. Although DOCK8-deficient B cells can acquire and present antigen to initiate activation of cognate T cells, integrin up-regulation, B cell-T cell conjugate formation, and costimulation are insufficient for sustained B cell and T cell activation when antigen availability is limited. Our findings provide an explanation for the failure of the humoral response in DOCK8 immunodeficiency syndrome and insight into how the level of available antigen modulates B cell-T cell cross-talk to fine-tune humoral immune responses and immunological memory.
Collapse
Affiliation(s)
- Mukta Deobagkar-Lele
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford
| | - Greg Crawford
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford
| | - Tanya L. Crockford
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford
| | - Jennifer Back
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford
| | - Rose Hodgson
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford
| | - Aneesha Bhandari
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford
| | - Katherine R Bull
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford
- CAMS-Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford
- Oxford Kidney Unit, Oxford University Hospitals Trust, Oxford
| | - Richard J. Cornall
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford
- CAMS-Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford
| |
Collapse
|
2
|
Vervoordeldonk MYL, Hengeveld PJ, Levin MD, Langerak AW. B cell receptor signaling proteins as biomarkers for progression of CLL requiring first-line therapy. Leuk Lymphoma 2024; 65:1031-1043. [PMID: 38619476 DOI: 10.1080/10428194.2024.2341151] [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: 02/18/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
The molecular landscape of chronic lymphocytic leukemia (CLL) has been extensively characterized, and various potent prognostic biomarkers were discovered. The genetic composition of the B-cell receptor (BCR) immunoglobulin (IG) was shown to be especially powerful for discerning indolent from aggressive disease at diagnosis. Classification based on the IG heavy chain variable gene (IGHV) somatic hypermutation status is routinely applied. Additionally, BCR IGH stereotypy has been implicated to improve risk stratification, through characterization of subsets with consistent clinical profiles. Despite these advances, it remains challenging to predict when CLL progresses to requiring first-line therapy, thus emphasizing the need for further refinement of prognostic indicators. Signaling pathways downstream of the BCR are essential in CLL pathogenesis, and dysregulated components within these pathways impact disease progression. Considering not only genomics but the entirety of factors shaping BCR signaling activity, this review offers insights in the disease for better prognostic assessment of CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/genetics
- Signal Transduction
- Disease Progression
- Biomarkers, Tumor/genetics
- Prognosis
Collapse
Affiliation(s)
- Mischa Y L Vervoordeldonk
- Department of Immunology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Paul J Hengeveld
- Department of Immunology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Mark-David Levin
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, The Netherlands
| | - Anton W Langerak
- Department of Immunology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
3
|
Deborah EA, Nabekura T, Shibuya K, Shibuya A. THEMIS2 Impairs Antitumor Activity of NK Cells by Suppressing Activating NK Receptor Signaling. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1819-1828. [PMID: 38619282 DOI: 10.4049/jimmunol.2300771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 03/19/2024] [Indexed: 04/16/2024]
Abstract
NK cells are cytotoxic innate lymphocytes that play a critical role in antitumor immunity. NK cells recognize target cells by using a repertoire of activating NK receptors and exert the effector functions. Although the magnitude of activation signals through activating NK receptors controls NK cell function, it has not been fully understood how these activating signals are modulated in NK cells. In this study, we found that a scaffold protein, THEMIS2, inhibits activating NK receptor signaling. Overexpression of THEMIS2 attenuated the effector function of human NK cells, whereas knockdown of THEMIS2 enhanced it. Mechanistically, THEMIS2 binds to GRB2 and phosphorylated SHP-1 and SHP-2 at the proximity of activating NK receptors DNAM-1 and NKG2D. Knockdown of THEMIS2 in primary human NK cells promoted the effector functions. Furthermore, Themis2-deficient mice showed low metastatic burden in an NK cell-dependent manner. These findings demonstrate that THEMIS2 has an inhibitory role in the antitumor activity of NK cells, suggesting that THEMIS2 might be a potential therapeutic target for NK cell-mediated cancer immunotherapy.
Collapse
Affiliation(s)
- Elfira Amalia Deborah
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Doctoral Program in Medical Science, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Tsukasa Nabekura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kazuko Shibuya
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Shibuya
- Department of Immunology, Institute of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
4
|
Nabekura T, Deborah EA, Tahara S, Arai Y, Love PE, Kako K, Fukamizu A, Muratani M, Shibuya A. Themis2 regulates natural killer cell memory function and formation. Nat Commun 2023; 14:7200. [PMID: 37938555 PMCID: PMC10632368 DOI: 10.1038/s41467-023-42578-8] [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: 05/01/2023] [Accepted: 10/16/2023] [Indexed: 11/09/2023] Open
Abstract
Immunological memory is a hallmark of the adaptive immune system. Although natural killer (NK) cells are innate immune cells important for the immediate host defence, they can differentiate into memory NK cells. The molecular mechanisms controlling this differentiation are yet to be fully elucidated. Here we identify the scaffold protein Themis2 as a critical regulator of memory NK cell differentiation and function. Themis2-deficient NK cells expressing Ly49H, an activating NK receptor for the mouse cytomegalovirus (MCMV) antigen m157, show enhanced differentiation into memory NK cells and augment host protection against MCMV infection. Themis2 inhibits the effector function of NK cells after stimulation of Ly49H and multiple activating NK receptors, though not specific to memory NK cells. Mechanistically, Themis2 suppresses Ly49H signalling by attenuating ZAP70/Syk phosphorylation, and it also translocates to the nucleus, where it promotes Zfp740-mediated repression to regulate the persistence of memory NK cells. Zfp740 deficiency increases the number of memory NK cells and enhances the effector function of memory NK cells, which further supports the relevance of the Themis2-Zfp740 pathway. In conclusion, our study shows that Themis2 quantitatively and qualitatively regulates NK cell memory formation.
Collapse
Affiliation(s)
- Tsukasa Nabekura
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8575, Japan.
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan.
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Ibaraki, 305-8575, Japan.
| | - Elfira Amalia Deborah
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Saeko Tahara
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- College of Medicine, School of Medicine and Health Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
- Bioinformatics Laboratory, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Yuya Arai
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- Bioinformatics Laboratory, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
- College of Biological Sciences, School of Life and Environmental Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Paul E Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Koichiro Kako
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8575, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Akiyoshi Fukamizu
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Masafumi Muratani
- Department of Genome Biology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan
| | - Akira Shibuya
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8575, Japan.
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Ibaraki, 305-8575, Japan.
- R&D Center for Innovative Drug Discovery, University of Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
5
|
Hengeveld PJ, Kolijn PM, Demmers JA, Doff W, Dubois JM, Rijken M, Assmann JL, van der Straten L, Boiten HJ, Gussinklo KJ, Valk PJ, Faber LM, Westerweel PE, Kater AP, Levin MD, Langerak AW. High-throughput Proteomics Identifies THEMIS2 as Independent Biomarker of Treatment-free Survival in Untreated CLL. Hemasphere 2023; 7:e951. [PMID: 37731707 PMCID: PMC10508458 DOI: 10.1097/hs9.0000000000000951] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/31/2023] [Indexed: 09/22/2023] Open
Abstract
It remains challenging in chronic lymphocytic leukemia (CLL) to distinguish between patients with favorable and unfavorable time-to-first treatment (TTFT). Additionally, the downstream protein correlates of well-known molecular features of CLL are not always clear. To address this, we selected 40 CLL patients with TTFT ≤24 months and compared their B cell intracellular protein expression with 40 age- and sex-matched CLL patients with TTFT >24 months using mass spectrometry. In total, 3268 proteins were quantified in the cohort. Immunoglobulin heavy-chain variable (IGHV) mutational status and trisomy 12 were most impactful on the CLL proteome. Comparing cases to controls, 5 proteins were significantly upregulated, whereas 3 proteins were significantly downregulated. Of these, only THEMIS2, a signaling protein acting downstream of the B cell receptor, was significantly associated with TTFT, independently of IGHV and TP53 mutational status (hazard ratio, 2.49 [95% confidence interval, 1.62-3.84]; P < 0.001). This association was validated on the mRNA and protein level by quantitative polymerase chain reaction and ELISA, respectively. Analysis of 2 independently generated RNA sequencing and mass spectrometry datasets confirmed the association between THEMIS2 expression and clinical outcome. In conclusion, we present a comprehensive characterization of the proteome of untreated CLL and identify THEMIS2 expression as a putative biomarker of TTFT.
Collapse
Affiliation(s)
- Paul J. Hengeveld
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, the Netherlands
| | | | | | - Wouter Doff
- Proteomics Center, Erasmus MC, Rotterdam, the Netherlands
| | - Julie M.N. Dubois
- Department of Hematology and Experimental Immunology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands
| | - Melissa Rijken
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, the Netherlands
| | | | - Lina van der Straten
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, the Netherlands
| | - Henk Jan Boiten
- Department of Immunology, Erasmus MC, Rotterdam, the Netherlands
| | - Kirsten J. Gussinklo
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, the Netherlands
| | - Peter J.M. Valk
- Department of Hematology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, the Netherlands
| | - Laura M. Faber
- Department of Hematology, Red Cross Hospital, Beverwijk, the Netherlands
| | - Peter E. Westerweel
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, the Netherlands
| | - Arnon P. Kater
- Department of Hematology and Experimental Immunology, Cancer Center Amsterdam, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands
| | - Mark-David Levin
- Department of Internal Medicine, Albert Schweitzer Hospital, Dordrecht, the Netherlands
| | | |
Collapse
|
6
|
Guo Y, Zhang Y, Yu J, Dong Y, Chen Z, Zhu C, Hong X, Xie Z, Zhang M, Wang S, Liang Y, He X, Ju W, Chen M. Novel ceRNA network construction associated with programmed cell death in acute rejection of heart allograft in mice. Front Immunol 2023; 14:1184409. [PMID: 37753085 PMCID: PMC10518384 DOI: 10.3389/fimmu.2023.1184409] [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: 03/30/2023] [Accepted: 08/28/2023] [Indexed: 09/28/2023] Open
Abstract
Background T cell-mediated acute rejection(AR) after heart transplantation(HT) ultimately results in graft failure and is a common indication for secondary transplantation. It's a serious threat to heart transplant recipients. This study aimed to explore the novel lncRNA-miRNA-mRNA networks that contributed to AR in a mouse heart transplantation model. Methods The donor heart from Babl/C mice was transplanted to C57BL/6 mice with heterotopic implantation to the abdominal cavity. The control group was syngeneic heart transplantation with the same kind of mice donor. The whole-transcriptome sequencing was performed to obtain differentially expressed mRNAs (DEmRNAs), miRNAs (DEmiRNAs) and lncRNAs (DElncRNAs) in mouse heart allograft. The biological functions of ceRNA networks was analyzed by GO and KEGG enrichment. Differentially expressed ceRNA involved in programmed cell death were further verified with qRT-PCR testing. Results Lots of DEmRNAs, DEmiRNAs and DElncRNAs were identified in acute rejection and control after heart transplantation, including up-regulated 4754 DEmRNAs, 1634 DElncRNAs, 182 DEmiRNAs, and down-regulated 4365 DEmRNAs, 1761 DElncRNAs, 132 DEmiRNAs. Based on the ceRNA theory, lncRNA-miRNA-mRNA regulatory networks were constructed in allograft acute rejection response. The functional enrichment analysis indicate that the down-regulated mRNAs are mainly involved in cardiac muscle cell contraction, potassium channel activity, etc. and the up-regulated mRNAs are mainly involved in T cell differentiation and mononuclear cell migration, etc. The KEGG pathway enrichment analysis showed that the down-regulated DEmRNAs were mainly enriched in adrenergic signaling, axon guidance, calcium signaling pathway, etc. The up-regulated DEmRNAs were enriched in the adhesion function, chemokine signaling pathway, apoptosis, etc. Four lncRNA-mediated ceRNA regulatory pathways, Pvt1/miR-30c-5p/Pdgfc, 1700071M16Rik/miR-145a-3p/Pdgfc, 1700071M16Rik/miR-145a-3p/Tox, 1700071M16Rik/miR-145a-3p/Themis2, were finally validated. In addition, increased expression of PVT1, 1700071M16Rik, Tox and Themis2 may be considered as potential diagnostic gene biomarkers in AR. Conclusion We speculated that Pvt1/miR-30c-5p/Pdgfc, 1700071M16Rik/miR-145a-3p/Pdgfc, 1700071M16Rik/miR-145a-3p/Tox and 1700071M16Rik/miR-145a-3p/Themis2 interaction pairs may serve as potential biomarkers in AR after HT.
Collapse
Affiliation(s)
- Yiwen Guo
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Yixi Zhang
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jia Yu
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Yuqi Dong
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Zhitao Chen
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Chuchen Zhu
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Xitao Hong
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Zhonghao Xie
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Min Zhang
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Shuai Wang
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Yichen Liang
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Xiaoshun He
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Weiqiang Ju
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| | - Maogen Chen
- The First Affiliated Hospital, Sun Yat-Sen University, Organ Transplant Centre, Guangzhou, China
- Guangdong Provincial Key Laboratory of Organ Donation and Transplant Immunology, Guangzhou, China
- Guangdong Provincial International Cooperation Base of Science and Technology (Organ Transplantation), Guangzhou, China
| |
Collapse
|
7
|
Choi S, Lee J, Hatzihristidis T, Gaud G, Dutta A, Arya A, Clubb LM, Stamos DB, Markovics A, Mikecz K, Love PE. THEMIS increases TCR signaling in CD4 +CD8 + thymocytes by inhibiting the activity of the tyrosine phosphatase SHP1. Sci Signal 2023; 16:eade1274. [PMID: 37159521 PMCID: PMC10410529 DOI: 10.1126/scisignal.ade1274] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 04/20/2023] [Indexed: 05/11/2023]
Abstract
The T cell lineage-restricted protein THEMIS plays a critical role in T cell development at the positive selection stage. In the SHP1 activation model, THEMIS is proposed to enhance the activity of the tyrosine phosphatase SHP1 (encoded by Ptpn6), thereby dampening T cell antigen receptor (TCR) signaling and preventing the inappropriate negative selection of CD4+CD8+ thymocytes by positively selecting ligands. In contrast, in the SHP1 inhibition model, THEMIS is proposed to suppress SHP1 activity, rendering CD4+CD8+ thymocytes more sensitive to TCR signaling initiated by low-affinity ligands to promote positive selection. We sought to resolve the controversy regarding the molecular function of THEMIS. We found that the defect in positive selection in Themis-/- thymocytes was ameliorated by pharmacologic inhibition of SHP1 or by deletion of Ptpn6 and was exacerbated by SHP1 overexpression. Moreover, overexpression of SHP1 phenocopied the Themis-/- developmental defect, whereas deletion of Ptpn6, Ptpn11 (encoding SHP2), or both did not result in a phenotype resembling that of Themis deficiency. Last, we found that thymocyte negative selection was not enhanced but was instead impaired in the absence of THEMIS. Together, these results provide evidence favoring the SHP1 inhibition model, supporting a mechanism whereby THEMIS functions to enhance the sensitivity of CD4+CD8+ thymocytes to TCR signaling, enabling positive selection by low-affinity, self-ligand-TCR interactions.
Collapse
Affiliation(s)
- Seeyoung Choi
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Jan Lee
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Teri Hatzihristidis
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Guillaume Gaud
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Avik Dutta
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Awadhesh Arya
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
- Shock, Trauma & Anesthesiology Research (STAR) Center, University of Maryland, School of Medicine, Baltimore, MD 21201, USA
| | - Lauren M. Clubb
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Daniel B. Stamos
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - Adrienn Markovics
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Katalin Mikecz
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL 60612, USA
| | - Paul E. Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| |
Collapse
|
8
|
Garcia-Vilanova A, Olmo-Fontánez AM, Moliva JI, Allué-Guardia A, Singh H, Merritt RE, Maselli DJ, Peters JI, Restrepo BI, Wang Y, Schlesinger LS, Turner J, Weintraub ST, Torrelles JB. The Aging Human Lung Mucosa: A Proteomics Study. J Gerontol A Biol Sci Med Sci 2022; 77:1969-1974. [PMID: 35460553 PMCID: PMC9536443 DOI: 10.1093/gerona/glac091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Indexed: 11/13/2022] Open
Abstract
The older adult population, estimated to double by 2050, is at increased risk of respiratory infections and other pulmonary diseases. Biochemical changes in the lung alveolar lining fluid (ALF) and in alveolar compartment cells can alter local immune responses as we age, generating opportunities for invading pathogens to establish successful infections. Indeed, the lung alveolar space of older adults is a pro-inflammatory, pro-oxidative, dysregulated environment that remains understudied. We performed an exploratory, quantitative proteomic profiling of the soluble proteins present in ALF, developing insight into molecular fingerprints, pathways, and regulatory networks that characterize the alveolar space in old age, comparing it to that of younger individuals. We identified 457 proteins that were significantly differentially expressed in older adult ALF, including increased production of matrix metalloproteinases, markers of cellular senescence, antimicrobials, and proteins of neutrophilic granule origin, among others, suggesting that neutrophils in the lungs of older adults could be potential contributors to the dysregulated alveolar environment with increasing age. Finally, we describe a hypothetical regulatory network mediated by the serum response factor that could explain the neutrophilic profile observed in the older adult population.
Collapse
Affiliation(s)
- Andreu Garcia-Vilanova
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Angélica M Olmo-Fontánez
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
- Integrated Biomedical Sciences Program, The University of Texas Health Science Center, San Antonio, Texas, USA
| | - Juan I Moliva
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Anna Allué-Guardia
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Harjinder Singh
- Division of Pulmonary and Critical Care Medicine, School of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| | - Robert E Merritt
- Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Diego J Maselli
- Division of Pulmonary and Critical Care Medicine, School of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| | - Jay I Peters
- Division of Pulmonary and Critical Care Medicine, School of Medicine, UT Health San Antonio, San Antonio, Texas, USA
| | | | - Yufeng Wang
- Department of Molecular Microbiology and Immunology, South Texas Center for Emerging Infectious Diseases, UTSA, San Antonio, Texas, USA
| | - Larry S Schlesinger
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Joanne Turner
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Susan T Weintraub
- Department of Biochemistry and Structural Biology, UT Health San Antonio, San Antonio, Texas, USA
| | - Jordi B Torrelles
- Population Health and Host Pathogen Interactions Programs, Texas Biomedical Research Institute, San Antonio, Texas, USA
| |
Collapse
|
9
|
Li K, Ran B, Wang Y, Liu L, Li W. PLCγ2 impacts microglia-related effectors revealing variants and pathways important in Alzheimer’s disease. Front Cell Dev Biol 2022; 10:999061. [PMID: 36147734 PMCID: PMC9485805 DOI: 10.3389/fcell.2022.999061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Alzheimer’s disease (AD) is an irreversible neurodegenerative disease mainly characterized by memory loss and cognitive decline. The etiology of AD is complex and remains incompletely understood. In recent years, genome-wide association studies (GWAS) have increasingly highlighted the central role of microglia in AD pathology. As a trans-membrane receptor specifically present on the microglia in the central nervous system, phosphatidylinositol-specific phospholipase C gamma 2 (PLCγ2) plays an important role in neuroinflammation. GWAS data and corresponding pathological research have explored the effects of PLCG2 variants on amyloid burden and tau pathologies that underline AD. The link between PLCγ2 and other AD-related effectors in human and mouse microglia has also been established, placing PLCγ2 downstream of the triggering receptor expressed on myeloid cells 2 (TREM2), toll-like receptor 4 (TLR4), Bruton’s tyrosine kinase (BTK), and colony-stimulating factor 1 receptor (CSF1R). Because the research on PLCγ2’s role in AD is still in its early stages, few articles have been published, therefore in this paper, we integrate the relevant research published to date, review the structural features, expression patterns, and related pathways of PLCγ2, and summarize the recent studies on important PLCG2 variants related to AD. Furthermore, the possibility and challenge of using PLCγ2 to develop therapeutic drugs for AD are also discussed.
Collapse
|
10
|
Ozerov M, Noreikiene K, Kahar S, Huss M, Huusko A, Kõiv T, Sepp M, López M, Gårdmark A, Gross R, Vasemägi A. Whole-genome sequencing illuminates multifaceted targets of selection to humic substances in Eurasian perch. Mol Ecol 2022; 31:2367-2383. [PMID: 35202502 PMCID: PMC9314028 DOI: 10.1111/mec.16409] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 02/10/2022] [Accepted: 02/17/2022] [Indexed: 11/30/2022]
Abstract
Extreme environments are inhospitable to the majority of species, but some organisms are able to survive in such hostile conditions due to evolutionary adaptations. For example, modern bony fishes have colonized various aquatic environments, including perpetually dark, hypoxic, hypersaline and toxic habitats. Eurasian perch (Perca fluviatilis) is among the few fish species of northern latitudes that is able to live in very acidic humic lakes. Such lakes represent almost "nocturnal" environments; they contain high levels of dissolved organic matter, which in addition to creating a challenging visual environment, also affects a large number of other habitat parameters and biotic interactions. To reveal the genomic targets of humic-associated selection, we performed whole-genome sequencing of perch originating from 16 humic and 16 clear-water lakes in northern Europe. We identified over 800,000 single nucleotide polymorphisms, of which >10,000 were identified as potential candidates under selection (associated with >3000 genes) using multiple outlier approaches. Our findings suggest that adaptation to the humic environment may involve hundreds of regions scattered across the genome. Putative signals of adaptation were detected in genes and gene families with diverse functions, including organism development and ion transportation. The observed excess of variants under selection in regulatory regions highlights the importance of adaptive evolution via regulatory elements, rather than via protein sequence modification. Our study demonstrates the power of whole-genome analysis to illuminate the multifaceted nature of humic adaptation and provides the foundation for further investigation of causal mutations underlying phenotypic traits of ecological and evolutionary importance.
Collapse
Affiliation(s)
- Mikhail Ozerov
- Department of Aquatic ResourcesInstitute of Freshwater ResearchSwedish University of Agricultural SciencesDrottningholmSweden
- Department of BiologyUniversity of TurkuTurkuFinland
- Biodiversity UnitUniversity of TurkuTurkuFinland
| | - Kristina Noreikiene
- Chair of AquacultureInstitute of Veterinary Medicine and Animal SciencesEstonian University of Life SciencesTartuEstonia
| | - Siim Kahar
- Chair of AquacultureInstitute of Veterinary Medicine and Animal SciencesEstonian University of Life SciencesTartuEstonia
| | - Magnus Huss
- Department of Aquatic ResourcesSwedish University of Agricultural SciencesÖregrundSweden
| | - Ari Huusko
- Natural resources Institute Finland (Luke)PaltamoFinland
| | - Toomas Kõiv
- Chair of Hydrobiology and FisheryInstitute of Agricultural and Environmental SciencesEstonian University of Life SciencesTartuEstonia
| | - Margot Sepp
- Chair of Hydrobiology and FisheryInstitute of Agricultural and Environmental SciencesEstonian University of Life SciencesTartuEstonia
| | - María‐Eugenia López
- Department of Aquatic ResourcesInstitute of Freshwater ResearchSwedish University of Agricultural SciencesDrottningholmSweden
| | - Anna Gårdmark
- Department of Aquatic ResourcesSwedish University of Agricultural SciencesÖregrundSweden
| | - Riho Gross
- Chair of AquacultureInstitute of Veterinary Medicine and Animal SciencesEstonian University of Life SciencesTartuEstonia
| | - Anti Vasemägi
- Department of Aquatic ResourcesInstitute of Freshwater ResearchSwedish University of Agricultural SciencesDrottningholmSweden
- Chair of AquacultureInstitute of Veterinary Medicine and Animal SciencesEstonian University of Life SciencesTartuEstonia
| |
Collapse
|
11
|
Xing X, Xia Q, Gong B, Shen Z, Zhang Y. Identification of Tissue-Specific Expressed Hub Genes and Potential Drugs in Rheumatoid Arthritis Using Bioinformatics Analysis. Front Genet 2022; 13:855557. [PMID: 35368701 PMCID: PMC8971206 DOI: 10.3389/fgene.2022.855557] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/18/2022] [Indexed: 12/29/2022] Open
Abstract
Background: Rheumatoid arthritis (RA) is a common autoimmune disease characterized by progressive, destructive polyarthritis. However, the cause and underlying molecular events of RA are not clear. Here, we applied integrated bioinformatics to identify tissue-specific expressed hub genes involved in RA and reveal potential targeted drugs. Methods: Three expression profiles of human microarray datasets involving fibroblast-like synoviocytes (FLS) were downloaded from the Gene Expression Omnibus (GEO) database, the differentially expressed mRNAs (DEGs), miRNAs (DEMs), and lncRNAs (DELs) between normal and RA synovial samples were screened using GEO2R tool. BioGPS was used to identified tissue-specific expressed genes. Functional and pathway enrichment analyses were performed for common DEGs using the DAVID database, and the protein-protein interaction (PPI) network of common DEGs was constructed to recognize hub genes by the STRING database. Based on receiver operating characteristic (ROC) curve, we further investigated the prognostic values of tissue-specific expressed hub genes in RA patients. Connectivity Map (CMap) was run to identify novel anti-RA potential drugs. The DEM–DEG pairs and ceRNA network containing key DEMs were established by Cytoscape. Results: We obtain a total of 418 DEGs, 23 DEMs and 49 DELs. 64 DEGs were verified as tissue-specific expressed genes, most derive from the hematologic/immune system (20/64, 31.25%). GO term and KEGG pathway enrichment analysis showed that DEGs focused primarily on immune-related biological process and NF-κB pathway. 10 hub genes were generated via using MCODE plugin. Among them, SPAG5, CUX2, and THEMIS2 were identified as tissue-specific expressed hub genes, these 3 tissue-specific expressed hub genes have superior diagnostic value in the RA samples compared with osteoarthritis (OA) samples. 5 compounds (troleandomycin, levodopa, trichostatin A, LY-294002, and levamisole) rank among the top five in connectivity score. In addition, 5 miRNAs were identified to be key DEMs, the lncRNA–miRNA–mRNA network with five key DEMs was formed. The networks containing tissue-specific expressed hub genes are as follows: ARAP1-AS2/miR-20b-3p/TRIM3, ARAP1-AS2/miR-30c-3p/FRZB. Conclusion: This study indicates that screening for identify tissue-specific expressed hub genes and ceRNA network in RA using integrated bioinformatics analyses could help us understand the mechanism of development of RA. Besides, SPAG5 and THEMIS2 might be candidate biomarkers for diagnosis of RA. LY-294002, trichostatin A, and troleandomycin may be potential drugs for RA.
Collapse
Affiliation(s)
- Xuewu Xing
- Department of Orthopaedics, Tianjin First Central Hospital, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Qun Xia
- Department of Orthopaedics, Tianjin First Central Hospital, Tianjin, China
| | - Baoqi Gong
- Department of Rheumatology, Tianjin First Central Hospital, Tianjin, China
| | - Zhongyang Shen
- Department of Transplant Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Yingze Zhang
- School of Medicine, Nankai University, Tianjin, China
- Department of Orthopaedic Surgery of Hebei Province, Third Hospital of Hebei Medical University, Shijiazhuang, China
- Chinese Academy of Engineering, Beijing, China
- *Correspondence: Yingze Zhang,
| |
Collapse
|
12
|
Huang WC, Yen JH, Sung YW, Tung SL, Chen PM, Chu PY, Shih YC, Chi HC, Huang YC, Huang SJ, Wang LH. Novel function of THEMIS2 in the enhancement of cancer stemness and chemoresistance by releasing PTP1B from MET. Oncogene 2022; 41:997-1010. [PMID: 34974522 PMCID: PMC8837547 DOI: 10.1038/s41388-021-02136-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/16/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022]
Abstract
Triple negative breast cancer (TNBC) possesses poor prognosis mainly due to lack of effective endocrine or targeted therapies, aggressive nature and high rate of chemoresistance. Cancer stem cells (CSCs) are considered to play critical roles in cancer recurrence and chemoresistance. THEMIS2 was identified as the sole common elevated gene in three triple negative breast cancer (TNBC) and two ovarian CSC lines. We discovered an intrinsic signaling scaffold function of THEMIS2, which acts as a novel regulator of cancer stemness in promoting multiple cancer stemness properties including sphere formation, stemness markers expression, chemoresistance and tumorigenicity with low numbers of cancer cells implantation. For the first time, we demonstrated that THEMIS2 specifically enhanced MET activating phosphorylation by suppressing the association of protein-tyrosine phosphatases 1B (PTP1B) with p-MET and MET, which accounted mainly for THEMIS2-mediated effect on cancer stemness and chemoresistance. Increased THEMIS2 expression was associated with poor survival in TNBC patients and in patients from our breast cancer cohort. We found that non-cytotoxic dosages of cryptotanshinone (CPT) could potently inhibit cancer stemness, chemoresistance and tumorigenicity by suppressing expression of THEMIS2. Notably, stable overexpression of THEMIS2 is associated with enhanced sensitivity toward Capmatinib and CPT treatment. Expression levels of THEMIS2 and p-MET protein were positively correlated in the 465 breast cancer specimens. Our study revealed the novel oncogenic role of THEMIS2 and its underlying mechanism via suppressing PTP1B association with MET and thus leading to its activation. Our findings suggest that THEMIS2 could be a biomarker for MET targeted therapy and also provide a potential clinical application using low dosages of CPT for treatment of THEMIS2 positive TNBC.
Collapse
Affiliation(s)
- Wei-Chieh Huang
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Jia-Hau Yen
- Research Cancer Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Wen Sung
- Department of Obstetrics and Gynecologics, China Medical University Hospital, Taichung City, Taiwan
| | - Shiao-Lin Tung
- Department of Hematology and Oncology, Ton-Yen General Hospital, Hsinchu County, Taiwan
- Department of Nursing, Hsin Sheng Junior College of Medical Care and Management, Taoyuan City, Taiwan
| | - Po-Ming Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Pei-Yi Chu
- Department of Pathology, Show Chwan Memorial Hospital, Chung-Shang Road, Changhua County, Taiwan
| | - Ya-Chi Shih
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Hsiang-Cheng Chi
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Yi-Ching Huang
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Shih-Jei Huang
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Lu-Hai Wang
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan.
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
13
|
Jackson JT, Mulazzani E, Nutt SL, Masters SL. The role of PLCγ2 in immunological disorders, cancer, and neurodegeneration. J Biol Chem 2021; 297:100905. [PMID: 34157287 PMCID: PMC8318911 DOI: 10.1016/j.jbc.2021.100905] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 06/15/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Phosphatidylinositol-specific phospholipase Cγ2 (PLCγ2) is a critical signaling molecule activated downstream from a variety of cell surface receptors that contain an intracellular immunoreceptor tyrosine-based activation motif. These receptors recruit kinases such as Syk, BTK, and BLNK to phosphorylate and activate PLCγ2, which then generates 1D-myo-inositol 1,4,5-trisphosphate and diacylglycerol. These well-known second messengers are required for diverse membrane functionality including cellular proliferation, endocytosis, and calcium flux. As a result, PLCγ2 dysfunction is associated with a variety of diseases including cancer, neurodegeneration, and immune disorders. The diverse pathologies associated with PLCγ2 are exemplified by distinct genetic variants. Inherited mutations at this locus cause PLCγ2-associated antibody deficiency and immune dysregulation, in some cases with autoinflammation. Acquired mutations at this locus, which often arise as a result of BTK inhibition to treat chronic lymphocytic leukemia, result in constitutive downstream signaling and lymphocyte proliferation. Finally, a third group of PLCγ2 variants actually has a protective effect in a variety of neurodegenerative disorders, presumably by increased uptake and degradation of deleterious neurological aggregates. Therefore, manipulating PLCγ2 activity either up or down could have therapeutic benefit; however, we require a better understanding of the signaling pathways propagated by these variants before such clinical utility can be realized. Here, we review the signaling roles of PLCγ2 in hematopoietic cells to help understand the effect of mutations driving immune disorders and cancer and extrapolate from this to roles which may relate to protection against neurodegeneration.
Collapse
Affiliation(s)
- Jacob T Jackson
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Elisabeth Mulazzani
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Stephen L Nutt
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Seth L Masters
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia; Immunology Laboratory, Guangzhou Institute of Paediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou, Guangdong, China.
| |
Collapse
|
14
|
Src Family Protein Kinase Controls the Fate of B Cells in Autoimmune Diseases. Inflammation 2020; 44:423-433. [PMID: 33037966 DOI: 10.1007/s10753-020-01355-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/07/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023]
Abstract
There are more than 80 kinds of autoimmune diseases known at present, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), systemic sclerosis (SSc), inflammatory bowel disease (IBD), as well as other disorders. Autoimmune diseases have a characteristic of immune responses directly attacking own tissues, leading to systematic inflammation and subsequent tissue damage. B cells play a vital role in the development of autoimmune diseases and differentiate into plasma cells or memory B cells to secrete high-affinity antibody or provide long-lasting function. Drugs targeting B cells show good therapeutic effects for the treatment of autoimmune diseases, such as rituximab (anti-CD20 antibody). Src family protein kinases (SFKs) are believed to play important roles in a variety of cellular functions such as growth, proliferation, and differentiation of B cell via B cell antigen receptor (BCR). Lck/Yes-related novel protein tyrosine kinase (LYN), BLK (B lymphocyte kinase), and Fyn are three different kinds of SFKs mainly expressed in B cells. LYN has a dual role in the BCR signal. On the one hand, positive signals are beneficial to the development and maturation of B cells. On the other hand, LYN can also inhibit excessively activated B cells. BLK is involved in the proliferation, differentiation, and immune tolerance of B lymphocytes, and further affects the function of B cells, which may lead to autoreactive or regulatory cellular responses, increasing the risk of autoimmune diseases. Fyn may affect the development of autoimmune disorders via the differentiation of B cells in the early stage of B cell development. This article reviews the recent advances of SFKs in B lymphocytes in autoimmune diseases.
Collapse
|
15
|
Miles AM, Huson HJ. Time- and population-dependent genetic patterns underlie bovine milk somatic cell count. J Dairy Sci 2020; 103:8292-8304. [PMID: 32622601 DOI: 10.3168/jds.2020-18322] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022]
Abstract
The objective of this study was to determine whether genetic regulation of bovine milk somatic cell count (SCC) varied throughout the course of an individual lactation and to identify quantitative trait loci (QTL) that may differentiate populations of chronically mastitic and robustly healthy cows. Milk SCC has long been a proxy for clinical mastitis diagnosis in management and genetic improvement strategies to control the disease. Cows (n = 471) were genotyped on the Illumina BovineHD 777K BeadChip (Illumina Inc., San Diego, CA), and composite milk samples were collected for SCC at 0-1 d in milk (DIM), 3-5 DIM, 10-14 DIM, 90-110 DIM, and 210-230 DIM, with each time span representing key physiological transitions for the cow. Median lactation somatic cell score (SCS) and area under the SCS curve were calculated from farm test data. A total of 8 genome-wide associations were performed and 167 SNP spanning the genome were significantly associated (false discovery rate <0.05). Of these associated regions, 27 of 48 associated QTL were novel for clinical mastitis or SCC. The linkage disequilibrium block surrounding the associated QTL or a 1-Mb window in the absence of linkage disequilibrium was interrogated for candidate genes, and many of those identified were related to multiple arms of the immune system, including toll-like receptor signaling, macrophage activation, B-cell maturation, T-cell recruitment, and the complement pathway. These genes included EXOC4, BAMBI, ITSN2, IL34, FCN3, CD8A, and CD8B. In addition, we identified populations of robustly healthy (SCS ≤4 from 10-14 DIM until study end), chronically mastitic (SCS >4 from 10-14 DIM until study end), and average cows with fluctuating SCS, and calculated fixation indices to identify regions of the genome differentiating these 3 populations. A total of 12 SNP were identified that showed moderate allelic differentiation (Wright's F statistic, FST ≥ 0.4) between the "chronic," "healthy," and "average" populations of cows. Candidate genes in the region surrounding differentiated QTL were related to cell signaling and immune response, such as JAKMIP1 and MADCAM1. The wide range of significantly associated QTL spanning the genome and the diversity of gene functions reinforces that mastitis is a complex trait and suggests that selection based on lactation stage-specific SCS rather than a generalized score may lead to greater success in breeding mastitis-resistant cows.
Collapse
Affiliation(s)
- Asha M Miles
- Department of Animal Science, Cornell University, Ithaca, NY 14853
| | - Heather J Huson
- Department of Animal Science, Cornell University, Ithaca, NY 14853.
| |
Collapse
|
16
|
|
17
|
Chen J, Li N, Yin Y, Zheng N, Min M, Lin B, Zhang L, Long X, Zhang Y, Cai Z, Zhai S, Qin J, Wang X. Methyltransferase Nsd2 Ensures Germinal Center Selection by Promoting Adhesive Interactions between B Cells and Follicular Dendritic Cells. Cell Rep 2019; 25:3393-3404.e6. [PMID: 30566865 DOI: 10.1016/j.celrep.2018.11.096] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/23/2018] [Accepted: 11/28/2018] [Indexed: 01/23/2023] Open
Abstract
Antibody affinity maturation, which is an antigen-based selection process for B cells, occurs in germinal centers (GCs). GCB cells must efficiently recognize, acquire, and present antigens from follicular dendritic cells (FDCs) to receive positive selection signals from T helper cells. Previous studies showed that GCB cells undergo adhesive interactions with FDCs, but the regulatory mechanisms underlying the cell adhesions and their functional relevance remain unclear. Here, we identified H3K36me2 methyltransferase Nsd2 as a critical regulator of GCB cell-FDC adhesion. Nsd2 deletion modestly reduced GC responses but strongly impaired B cell affinity maturation. Mechanistically, Nsd2 directly regulated expression of multiple actin polymerization-related genes in GCB cells. Nsd2 loss reduced B cell adhesion to FDC-expressed adhesion molecules, thus affecting both B cell receptor (BCR) signaling and antigen acquisition. Overall, Nsd2 coordinates GCB positive selection by enhancing both BCR signaling and T cell help.
Collapse
Affiliation(s)
- Jingjing Chen
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Ni Li
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yuye Yin
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Nan Zheng
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Min Min
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Bichun Lin
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Le Zhang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Xuehui Long
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Yang Zhang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Zhenming Cai
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Sulan Zhai
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China
| | - Jun Qin
- CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| | - Xiaoming Wang
- Department of Immunology, State Key Laboratory of Reproductive Medicine, Nanjing Medical University, 101 Longmian Road, Nanjing, Jiangsu 211166, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Nutrition and Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China.
| |
Collapse
|
18
|
Abadie K, Pease NA, Wither MJ, Kueh HY. Order by chance: origins and benefits of stochasticity in immune cell fate control. CURRENT OPINION IN SYSTEMS BIOLOGY 2019; 18:95-103. [PMID: 33791444 PMCID: PMC8009491 DOI: 10.1016/j.coisb.2019.10.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
To protect against diverse challenges, the immune system must continuously generate an arsenal of specialized cell types, each of which can mount a myriad of effector responses upon detection of potential threats. To do so, it must generate multiple differentiated cell populations with defined sizes and proportions, often from rare starting precursor cells. Here, we discuss the emerging view that inherently probabilistic mechanisms, involving rare, rate-limiting regulatory events in single cells, control fate decisions and population sizes and fractions during immune development and function. We first review growing evidence that key fate control points are gated by stochastic signaling and gene regulatory events that occur infrequently over decision-making timescales, such that initially homogeneous cells can adopt variable outcomes in response to uniform signals. We next discuss how such stochastic control can provide functional capabilities that are harder to achieve with deterministic control strategies, and may be central to robust immune system function.
Collapse
Affiliation(s)
| | - Nicholas A Pease
- Department of Bioengineering, University of Washington
- Molecular and Cellular Biology Program, University of Washington
| | | | - Hao Yuan Kueh
- Department of Bioengineering, University of Washington
| |
Collapse
|
19
|
Choi S, Cornall R, Lesourne R, Love PE. THEMIS: Two Models, Different Thresholds. Trends Immunol 2017; 38:622-632. [PMID: 28697966 DOI: 10.1016/j.it.2017.06.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/13/2017] [Accepted: 06/14/2017] [Indexed: 11/17/2022]
Abstract
THEMIS, a recently identified T-lineage-restricted protein, is the founding member of a large metazoan protein family. Gene inactivation studies have revealed a critical requirement for THEMIS during thymocyte positive selection, implicating THEMIS in signaling downstream of the T cell antigen receptor (TCR), but the mechanistic underpinnings of THEMIS function have remained elusive. A previous model posited that THEMIS prevents thymocytes from inappropriately crossing the positive/negative selection threshold by dampening TCR signaling. However, new data suggest an alternative model where THEMIS enhances TCR signaling enabling thymocytes to reach the threshold for positive selection, avoiding death by neglect. We review the data supporting each model and conclude that the preponderance of evidence favors an enhancing function for THEMIS in TCR signaling.
Collapse
Affiliation(s)
- Seeyoung Choi
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard Cornall
- MRC Human Immunology Unit, Weatherall Institute for Molecular Medicine, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Renaud Lesourne
- Centre de Physiopathologie de Toulouse Purpan, Toulouse, France; Institut National de la Santé et de la Recherche Médicale, U1043, Centre National de la Recherche Scientifique, U5282, and Université de Toulouse, Université Paul Sabatier, Toulouse F-31300, France
| | - Paul E Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
20
|
Guan Q, Ezzati P, Spicer V, Krokhin O, Wall D, Wilkins JA. Interferon γ induced compositional changes in human bone marrow derived mesenchymal stem/stromal cells. Clin Proteomics 2017; 14:26. [PMID: 28694743 PMCID: PMC5501357 DOI: 10.1186/s12014-017-9161-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/28/2017] [Indexed: 12/18/2022] Open
Abstract
Background Mesenchymal stem/stromal cells (MSC) display a range of immunoregulatory properties which can be enhanced by the exposure to cytokines such interferon γ (IFN-γ). However the compositional changes associated with the ‘licensing’ of these cells have not been clearly defined. The present study was undertaken to provide a detailed comparative proteomic analysis of the compositional changes that occur in human bone marrow derived MSC following 20 h treatment with IFN-γ. Methods 2D LC MSMS analysis of control and IFN-γ treated cells from 5 different healthy donors provided confident identification of more than 8400 proteins. Results In total 210 proteins were shown to be significantly altered in their expression levels (≥|2SD|) following IFN-γ treatment. The changes for several of these proteins were confirmed by flow cytometry. STRING analysis determined that approximately 30% of the altered proteins physically interacted in described interferon mediated processes. Comparison of the list of proteins that were identified as changed in the proteomic analysis with data for the same proteins in the Interferome DB indicated that ~35% of these proteins have not been reported to be IFN-γ responsive in a range of cell types. Conclusions This data provides an in depth analysis of the proteome of basal and IFN-γ treated human mesenchymal stem cells and it identifies a number of novel proteins that may contribute to the immunoregulatory capacity if IFN-γ licensed cells. Electronic supplementary material The online version of this article (doi:10.1186/s12014-017-9161-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qingdong Guan
- Manitoba Centre for Advanced Cell and Tissue Therapy, Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB Canada.,Cellular Therapy Laboratory, CancerCare Manitoba, Winnipeg, MB Canada
| | - Peyman Ezzati
- Manitoba Centre for Proteomics and Systems Biology, Section of Biomedical Proteomics, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba and Health Sciences Centre, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB R3E 3P4 Canada
| | - Victor Spicer
- Manitoba Centre for Proteomics and Systems Biology, Section of Biomedical Proteomics, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba and Health Sciences Centre, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB R3E 3P4 Canada
| | - Oleg Krokhin
- Manitoba Centre for Proteomics and Systems Biology, Section of Biomedical Proteomics, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba and Health Sciences Centre, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB R3E 3P4 Canada
| | - Donna Wall
- Division of Haematology/Oncology, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON Canada
| | - John A Wilkins
- Manitoba Centre for Proteomics and Systems Biology, Section of Biomedical Proteomics, Department of Internal Medicine, Rady Faculty of Health Sciences, University of Manitoba and Health Sciences Centre, 799 John Buhler Research Centre, 715 McDermot Ave, Winnipeg, MB R3E 3P4 Canada
| |
Collapse
|
21
|
|