43 results on '"LAIR1"'
Search Results
2. Leukocyte-associated immunoglobulin-like receptor 1 (LAIR1) is reduced with preeclampsia and small for gestational aged fetuses.
- Author
-
Bartho, Lucy A., Walker, Susan P., Cannon, Ping, Nguyen, Tuong-Vi, Nguyen, Anna, Botha, Stefan M., Hannan, Natalie J., Tong, Stephen, and Kaitu'u-Lino, Tu'uhevaha J.
- Abstract
Leukocyte-associated immunoglobulin-like receptor 1 (LAIR1) is an inhibitory receptor expressed on immune cells. We evaluated LAIR1 in placentas from preeclamptic or small for gestational age (SGA) pregnancies, and placental explant model (1 % O 2 , IL6 and TNFα, or control). LAIR1 mRNA was reduced in placentas from preeclamptic (p < 0.0001, n = 78) and SGA (p < 0.0001, n = 32) pregnancies. LAIR1 protein expression was reduced in placentas from preeclampsia (p < 0.0001, n = 43) and SGA (p = 0.009, n = 10) pregnancies. Hypoxia (1 % O 2) reduced LAIR1 mRNA expression in placental explants (p = 0.008). These findings suggest hypoxia modulates LAIR1 expression in the placenta. • LAIR1 is reduced in placenta from preeclamptic and small for gestational age pregnancies. • Hypoxia reduced LAIR1 expression in placental explants. • IL6 and TNF-α did not change LAIR1 expression in placental explants. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. Blocking LAIR1 signaling in immune cells inhibits tumor development.
- Author
-
Jingjing Xie, Xun Gui, Mi Deng, Heyu Chen, Yuanzhi Chen, Xiaoye Liu, Zhiqiang Ku, Lingxiao Tan, Ryan Huang, Yubo He, Bruce Zhang, Cheryl Lewis, Kenian Chen, Lin Xu, Jian Xu, Tao Huang, Liao, X. Charlene, Ningyan Zhang, Zhiqiang An, and Cheng Cheng Zhang
- Subjects
KILLER cells ,MYELOID cells ,REGULATORY T cells ,IMMUNOLOGIC memory ,IMMUNE checkpoint proteins - Abstract
The current immune checkpoint blockade therapy has been successful in treating some cancers but not others. New molecular targets and therapeutic approaches of cancer immunology need to be identified. Leukocyte associated immunoglobulin like receptor 1 (LAIR1) is an immune inhibitory receptor expressing on most immune cell types. However, it remains a question whether we can specifically and actively block LAIR1 signaling to activate immune responses for cancer treatment. Here we report the development of specific antagonistic anti-LAIR1 monoclonal antibodies and studied the effects of LAIR1 blockade on the anti-tumor immune functions. The anti-LAIR1 antagonistic antibody stimulated the activities of T cells, natural killer cells, macrophages, and dendritic cells in vitro. The single-cell RNA sequencing analysis of intratumoral immune cells in syngeneic human LAIR1 transgenic mice treated with control or anti-LAIR1 antagonist antibodies indicates that LAIR1 signaling blockade increased the numbers of CD4 memory T cells and inflammatory macrophages, but decreased those of pro-tumor macrophages, regulatory T cells, and plasmacytoid dendritic cells. Importantly, the LAIR1 blockade by the antagonistic antibody inhibited the activity of immunosuppressive myeloid cells and reactivated T cells from cancer patients in vitro and impeded tumor metastasis in a humanized mouse model. Blocking LAIR1 signaling in immune cells represents a promising strategy for development of anti-cancer immunotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
4. LAIR1 promotes hepatocellular carcinoma cell metastasis and induces M2-macrophage infiltration through activating AKT-IKKβ-p65 axis.
- Author
-
Pan B, Shen S, Zhao J, Zhang Z, Ye D, Zhang X, Yao Y, Luo Y, Wang X, and Tang N
- Subjects
- Humans, I-kappa B Kinase metabolism, I-kappa B Kinase genetics, Transcription Factor RelA metabolism, Transcription Factor RelA genetics, Cell Line, Tumor, Tumor Microenvironment, Gene Expression Regulation, Neoplastic, Signal Transduction, Macrophages metabolism, Macrophages pathology, Cell Proliferation, Tumor-Associated Macrophages metabolism, Tumor-Associated Macrophages pathology, Tumor-Associated Macrophages immunology, Neoplasm Invasiveness, Carcinoma, Hepatocellular pathology, Carcinoma, Hepatocellular metabolism, Carcinoma, Hepatocellular genetics, Liver Neoplasms pathology, Liver Neoplasms metabolism, Liver Neoplasms genetics, Proto-Oncogene Proteins c-akt metabolism, Receptors, Immunologic metabolism, Receptors, Immunologic genetics, Cell Movement
- Abstract
LAIR1, a receptor found on immune cells, is capable of binding to collagen and is involved in immune-related diseases. However, the precise contribution of LAIR1 expressed on hepatocellular carcinoma (HCC) cells to tumor microenvironment is still unclear. In our study, bioinformatics analysis and immunofluorescence were employed to study the correlation between LAIR1 levels and clinical indicators. Transwell and scratch tests were used to evaluate how LAIR1 affected the migration and invasion of HCC cells. The chemotactic capacity and alternative activation of macrophages were investigated using RT-qPCR, transwell, and immunofluorescence. To investigate the molecular mechanisms, transcriptome sequencing analysis, Western blot, nucleus/cytoplasm fractionation, ELISA, and cytokine microarray were employed. We revealed a significant correlation between the presence of LAIR1 and an unfavorable outcome in HCC. We indicated that LAIR1 promoted migration and invasion of HCC cells through the AKT-IKKβ-p65 axis. Additionally, the alternative activation and infiltration of tumor-associated macrophages induced by LAIR1 were reliant on the upregulation of IL6 and CCL5 within this axis, respectively. In conclusion, blocking LAIR1 was found to be an effective approach in combating the cancerous advancement of HCC., (© 2024 Wiley Periodicals LLC.)
- Published
- 2024
- Full Text
- View/download PDF
5. Utility of leukocyte-associated immunoglobulin-like receptor-1 (CD305) in flow cytometric detection of minimal bone marrow involvement by B-cell non-Hodgkin lymphoma.
- Author
-
Singh A, Patil J, Ghogale SG, Deshpande N, Girase K, Shetye N, Rajpal S, Chatterjee G, Patkar N, Jain D, Epari S, Shet T, Gujral S, Subramanian PG, and Tembhare PR
- Subjects
- Humans, Male, Female, Aged, Middle Aged, Aged, 80 and over, Adult, Receptors, Immunologic metabolism, Glycoproteins, Flow Cytometry methods, Bone Marrow pathology, Bone Marrow metabolism, Lymphoma, B-Cell pathology, Lymphoma, B-Cell diagnosis, Lymphoma, B-Cell immunology, Immunophenotyping methods
- Abstract
Multicolor flow cytometry (MFC) is crucial in detecting occult or minimal bone marrow (BM) involvement by non-Hodgkin lymphomas (NHL), which may not be detected using trephine biopsy or imaging studies. Detection of low-level BM involvement can be challenging without definite immunophenotypic aberrancies. We studied the utility of CD305 in MFC detection of minimal BM involvement by B-NHL, especially in the absence of aberrancies by commonly used markers. The study included 1084 consecutive BM samples submitted for the staging of B-NHLs (excluding CLL) over two years. Samples were studied for morphological, immunophenotypic, and histopathological assessment. MFC studies were performed using 10-13 color MFC, including CD305-antibody (clone, DX26). Minimal BM involvement was defined with a cutoff of ≤10% lymphoma cells in viable cells on MFC assessment. Of 1084, 148 samples revealed overt morphological involvement by B-NHL and were excluded from analysis. BM samples of 172/936 patients were morphologically negative but revealed involvement using MFC independently. Corresponding trephine biopsy involvement was detected in only 79/172 (45.9%) patients. On MFC, 23/172 samples showed BM involvement with >10% lymphoma cells, and 149/172 (86.6%) samples revealed minimal involvement. In 54/149 (36.24%) samples, lymphoma cells were detected only with aberrant loss of CD305 expression. In 78 of the remaining 95 samples (82.1%), it provided an immunophenotypic aberrancy addition to other markers and supported the results. CD305 is a highly useful marker in the flow cytometric assessment of minimal BM involvement by B-NHL. MFC is a superior modality to trephine biopsy in detecting low-level BM involvement., (© 2024 The Author(s). Cytometry Part B: Clinical Cytometry published by Wiley Periodicals LLC on behalf of International Clinical Cytometry Society.)
- Published
- 2024
- Full Text
- View/download PDF
6. Structural basis of malarial parasite RIFIN-mediated immune escape against LAIR1
- Author
-
Yijia Xie, Xin Li, Yan Chai, Hao Song, Jianxun Qi, and George F. Gao
- Subjects
malaria ,Plasmodium falciparum ,RIFIN ,LAIR1 ,crystal structure ,immune escape ,Biology (General) ,QH301-705.5 - Abstract
Summary: Malaria infection by Plasmodium falciparum continues to pose a global threat to the human population. P. falciparum expresses variable erythrocyte surface antigens such as RIFINs. Public antibodies with LAIR1 insertion have been identified from malarial patients against a subset of RIFINs. In this study, we solve a LAIR1-binding RIFIN structure: the complex structures of two RIFINs bound to mutated or wild-type LAIR1 in two distinct patterns. Notably, the two RIFINs engage similar binding sites on LAIR1 with different angles, and the RIFIN-binding sites overlap with the collagen-binding site. Surprisingly, RIFINs use completely different binding sites to bind to LAIR1 or LILRB1, indicating the kaleidoscopic change of RIFINs. We then verify that RIFIN could induce LAIR1-mediated cell signaling, and LAIR1-containing antibodies could block the pathway. The findings of this study provide structural insights into the mechanism of the immune escape of P. falciparum and the endless arms race between parasite and host.
- Published
- 2021
- Full Text
- View/download PDF
7. Cancer immunotherapy by NC410, a LAIR-2 Fc protein blocking human LAIR-collagen interaction
- Author
-
M Ines Pascoal Ramos, Linjie Tian, Emma J de Ruiter, Chang Song, Ana Paucarmayta, Akashdip Singh, Eline Elshof, Saskia V Vijver, Jahangheer Shaik, Jason Bosiacki, Zachary Cusumano, Christina Jensen, Nicholas Willumsen, Morten A Karsdal, Linda Liu, Sol Langermann, Stefan Willems, Dallas Flies, and Linde Meyaard
- Subjects
tumor ,collagen ,LAIR1 ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Collagens are a primary component of the extracellular matrix and are functional ligands for the inhibitory immune receptor leukocyte-associated immunoglobulin-like receptor (LAIR)-1. LAIR-2 is a secreted protein that can act as a decoy receptor by binding collagen with higher affinity than LAIR-1. We propose that collagens promote immune evasion by interacting with LAIR-1 expressed on immune cells, and that LAIR-2 releases LAIR-1-mediated immune suppression. Analysis of public human datasets shows that collagens, LAIR-1 and LAIR-2 have unique and overlapping associations with survival in certain tumors. We designed a dimeric LAIR-2 with a functional IgG1 Fc tail, NC410, and showed that NC410 increases human T cell expansion and effector function in vivo in a mouse xenogeneic-graft versus-host disease model. In humanized mouse tumor models, NC410 reduces tumor growth that is dependent on T cells. Immunohistochemical analysis of human tumors shows that NC410 binds to collagen-rich areas where LAIR-1+ immune cells are localized. Our findings show that NC410 might be a novel strategy for cancer immunotherapy for immune-excluded tumors.
- Published
- 2021
- Full Text
- View/download PDF
8. A novel potential target of IL‐35‐regulated JAK/STAT signaling pathway in lupus nephritis
- Author
-
Zhe Cai, Song Zhang, Ping Wu, Qi Ren, Ping Wei, Ming Hong, Yu Feng, Chun Kwok Wong, Hong Tang, and Huasong Zeng
- Subjects
IL‐35 ,JAK/STAT signaling pathway ,JSLE‐LN ,LAIR1 ,mesangial calls ,Medicine (General) ,R5-920 - Abstract
Abstract Background In this study, we have investigated the potential regulatory mechanisms of IL‐35 to relieve lupus nephritis (LN) through regulating Janus kinase (JAK)/signal transducers and activators of transcription (STAT) signaling pathway in mesangial cells. Results Among 105 significant differentially expressed proteins (DEPs) between juvenile systemic lupus erythematosus (JSLE) patients with LN and healthy controls, LAIR1, PDGFRβ, VTN, EPHB4, and EPHA4 were downregulated in JSLE‐LN. They consist of an interactive network with PTPN11 and FN1, which involved in IL‐35‐related JAK/STAT signaling pathway. Besides, urinary LAIR1 was significantly correlated with JSLE‐LN clinical parameters such as SLEDAI‐2K, %CD19+ B, and %CD3+ T cells. Through bioinformatics analysis of co‐immunoprecipitation with mass spectrometry results, including GO, KEGG, and STRING, five genes interacted with Lair1 were upregulated by IL‐35, but only Myh10 was downregulated. Therefore, we presumed an interactive network among these DEPs, JAK/STAT, and IL‐35. Moreover, the downregulated phosphorylated (p)‐STAT3, p‐p38 MAPK, and p‐ERK, and the upregulated p‐JAK2/p‐STAT1/4 in IL‐35 overexpressed mesangial cells, and RNA‐sequencing results validated the potential regulatory mechanisms of IL‐35 in alleviating JSLE‐LN disease. Moreover, the relieved histopathological features of nephritis including urine protein and leukocyte scores, a decreased %CD90+αSMA+ mesangial cells and pro‐inflammatory cytokines, the inactivated JAK/STAT signals and the significant upregulated Tregs in spleen, thymus and peripheral blood were validated in Tregs and IL‐35 overexpression plasmid‐treated lupus mice. Conclusions Our study provided a reference proteomic map of urinary biomarkers for JSLE‐LN and elucidated evidence that IL‐35 may regulate the interactive network of LAIR1‐PTPN11‐JAK‐STAT‐FN1 to affect JAK/STAT and MAPK signaling pathways to alleviate inflammation in JSLE‐LN. This finding may provide a further prospective mechanism for JSLE‐LN clinical treatment.
- Published
- 2021
- Full Text
- View/download PDF
9. LAIR1-mediated resistance of hepatocellular carcinoma cells to T cells through a GSK-3β/β-catenin/MYC/PD-L1 pathway.
- Author
-
Pan, Banglun, Ke, Xiaoling, Qiu, Jiacheng, Ye, Dongjie, Zhang, Zhu, Zhang, Xiaoxia, Luo, Yue, Yao, Yuxin, Wu, Xiaoxuan, Wang, Xiaoqian, and Tang, Nanhong
- Subjects
- *
PROGRAMMED cell death 1 receptors , *T cells , *HEPATOCELLULAR carcinoma , *T-cell exhaustion , *IMMUNE checkpoint proteins - Abstract
An increasing number of studies have reported the involvement of oncogenes in the regulation of the immune system. LAIR1 is an immunosuppressive molecule and its role in immune-related diseases has been mainly reported. To date, it is unclear whether LAIR1 in tumor cells is involved in immune regulation. Therefore, the aim of this study was to investigate the role of LAIR1 in the immune microenvironment of hepatocellular carcinoma (HCC) to seek the novel therapeutic discoveries. Tumor Immune Dysfunction and Exclusion database was used to predict the response of LAIR1 expression to immune checkpoint blockade. CD8+ T cells were co-cultured with HCC cells, and the killing efficiency of leukocytes on HCC cells was detected by flow cytometry. Flow cytometry was also used to detect the expression of inhibitory receptors. In addition, Western blot, immunofluorescence, and nucleus/cytoplasm fractionation experiments were performed to explore the molecular mechanisms by which LAIR1 created a suppressive tumor microenvironment. LAIR1 expression in HCC was associated with worse immune prognosis and T-cell dysfunction. HCC cells overexpressing LAIR1 co-cultured with CD8+ T cells induced exhaustion of latter. Mechanism studies indicated that LAIR1 in HCC cells up-regulated the phosphorylation of β-catenin by inducing the phosphorylation of GSK-3β, leading to the impairment of the expression and the nuclear localization signal of β-catenin. Low β-catenin expression and nuclear localization signal inhibited MYC-mediated PD-L1 expression. Therefore, PD-L1 up-regulated by LAIR1 caused the exhaustion of infiltrating CD8+ T cells in HCC, which aggravated the malignant progression of HCC. LAIR1 increased PD-L1 expression through the GSK-3β/β-catenin/MYC/PD-L1 pathway and promoted immune evasion of HCC cells. Targeted inhibition of LAIR1 helped to enhance the immune killing effect of CD8+ T cells in HCC. • LAIR1 in HCC leads to impaired anti-tumor activity function of CD8+ T cells, which is not conducive to immunotherapy of HCC. • LAIR1 facilitates the exhaustion of CD8+ T cells by inducing the expression of PD-L1 in HCC cells. • LAIR1 enhances PD-L1 expression via the GSK-3β/β-catenin/MYC/PD-L1 pathway, thereby promoting immune evasion in HCC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. IL-7-induced phosphorylation of the adaptor Crk-like and other targets.
- Author
-
Aiello, Francesca B., Guszczynski, Tad, Li, Wenqing, Hixon, Julie A., Jiang, Qiong, Hodge, Deborah L., Massignan, Tania, Di Lisio, Chiara, Merchant, Anand, Procopio, Antonio D., Bonetto, Valentina, and Durum, Scott K.
- Subjects
- *
INTERLEUKIN-7 , *T cell differentiation , *ELECTROPHORESIS , *TYROSINE , *PHOSPHORYLATION , *IMMUNOBLOTTING - Abstract
IL-7 is required for T cell differentiation and mature T cell homeostasis and promotes pro-B cell proliferation and survival. Tyrosine phosphorylation plays a central role in IL-7 signaling. We identified by two-dimensional electrophoresis followed by anti-phosphotyrosine immunoblotting and mass spectrometry sixteen tyrosine phosphorylated proteins from the IL-7-dependent cell line D1. IL-7 stimulation induced the phosphorylation of the proteins STI1, ATIC and hnRNPH, involved in pathways related to survival, proliferation and gene expression, respectively, and increased the phosphorylation of CrkL, a member of a family of adaptors including the highly homologous Crk isoforms CrkII and CrkI, important in multiple signaling pathways. We observed an increased phosphorylation of CrkL in murine pro-B cells and in murine and human T cells. In addition, IL-7 increased the association of CrkL with the transcription factor Stat5, essential for IL-7 pro-survival activity. The selective tyrosine kinase inhibitor Imatinib. counteracted the IL-7 pro-survival effect in D1 cells and decreased CrkL phosphorylation. These data suggested that CrkL could play a pro-survival role in IL-7-mediated signaling. We observed that pro-B cells also expressed, in addition to CrkL, the Crk isoforms CrkII and CrkI and therefore utilized pro-B cells conditionally deficient in all three to evaluate the role of these proteins. The observation that the IL-7 pro-survival effect was reduced in Crk/CrkL conditionally-deficient pro-B cells further pointed to a pro-survival role of these adaptors. To further evaluate the role of these proteins, gene expression studies were performed in Crk/CrkL conditionally-deficient pro-B cells. IL-7 decreased the transcription of the receptor LAIR1, which inhibits B cell proliferation, in a Crk/CrkL-dependent manner, suggesting that the Crk family of proteins may promote pro-B cell proliferation. Our data contribute to the understanding of IL-7 signaling and suggest the involvement of Crk family proteins in pathways promoting survival and proliferation. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
11. The structure of a LAIR1-containing human antibody reveals a novel mechanism of antigen recognition
- Author
-
Fu-Lien Hsieh and Matthew K Higgins
- Subjects
antibody structure ,LAIR1 ,novel antigen recognition ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Antibodies are critical components of the human adaptive immune system, providing versatile scaffolds to display diverse antigen-binding surfaces. Nevertheless, most antibodies have similar architectures, with the variable immunoglobulin domains of the heavy and light chain each providing three hypervariable loops, which are varied to generate diversity. The recent identification of a novel class of antibody in humans from malaria endemic regions of Africa was therefore surprising as one hypervariable loop contains the entire collagen-binding domain of human LAIR1. Here, we present the structure of the Fab fragment of such an antibody. We show that its antigen-binding site has adopted an architecture that positions LAIR1, while itself being occluded. This therefore represents a novel means of antigen recognition, in which the Fab fragment of an antibody acts as an adaptor, linking a human protein insert with antigen-binding potential to the constant antibody regions which mediate immune cell recruitment.
- Published
- 2017
- Full Text
- View/download PDF
12. LAIR1, an ITIM-Containing Receptor Involved in Immune Disorders and in Hematological Neoplasms.
- Author
-
Van Laethem, François, Donaty, Lucie, Tchernonog, Emmanuelle, Lacheretz-Szablewski, Vanessa, Russello, Jennifer, Buthiau, Delphine, Almeras, Marion, Moreaux, Jérôme, and Bret, Caroline
- Subjects
- *
BLOOD diseases , *IMMUNOLOGIC diseases , *LYMPHOCYTE subsets , *B cells , *CELL physiology , *KILLER cell receptors , *KILLER cells - Abstract
Leukocyte-associated immunoglobulin (Ig)-like receptor 1 (LAIR1, CD305) belongs to the family of immune-inhibitory receptors and is widely expressed on hematopoietic mature cells, particularly on immune cells. Four different types of ligands of LAIR1 have been described, including collagens, suggesting a potential immune-regulatory function on the extracellular matrix. By modulating cytokine secretion and cellular functions, LAIR1 displays distinct patterns of expression among NK cell and T/B lymphocyte subsets during their differentiation and cellular activation and plays a major negative immunoregulatory role. Beyond its implications in physiology, the activity of LAIR1 can be inappropriately involved in various autoimmune or inflammatory disorders and has been implicated in cancer physiopathology, including hematological neoplasms. Its action as an inhibitory receptor can result in the dysregulation of immune cellular responses and in immune escape within the tumor microenvironment. Furthermore, when expressed by tumor cells, LAIR1 can modulate their proliferation or invasion properties, with contradictory pro- or anti-tumoral effects depending on tumor type. In this review, we will focus on its role in normal physiological conditions, as well as during pathological situations, including hematological malignancies. We will also discuss potential therapeutic strategies targeting LAIR1 for the treatment of various autoimmune diseases and cancer settings. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
13. Plasmodium falciparum RIFIN is a novel ligand for inhibitory immune receptor LILRB2
- Author
-
Kyoko Shida, Fumiji Saito, Sawako Itagaki, Kouyuki Hirayasu, Masako Kohyama, Wataru Nakai, Akihito Sakoguchi, Sumiko Matsuoka, Toshihiro Horii, Shiroh Iwanaga, Hisashi Arase, and Tadahiro Suenaga
- Subjects
0301 basic medicine ,Erythrocytes ,Plasmodium falciparum ,Protozoan Proteins ,Biophysics ,Human leukocyte antigen ,Immune receptor ,Ligands ,Biochemistry ,LILRB1 ,03 medical and health sciences ,0302 clinical medicine ,Protein Domains ,LILRB2 ,parasitic diseases ,Animals ,Humans ,Malaria, Falciparum ,Receptors, Immunologic ,Receptor ,Molecular Biology ,Mice, Inbred BALB C ,Membrane Glycoproteins ,biology ,LAIR1 ,Membrane Proteins ,Cell Biology ,Acquired immune system ,biology.organism_classification ,Cell biology ,HEK293 Cells ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Protein Binding - Abstract
Plasmodium falciparum causes the most severe form of malaria. Acquired immunity against P. falciparum provides insufficient protection even after repeated infections. Therefore, P. falciparum parasites might exploit inhibitory receptors for immune evasion. P. falciparum RIFINs are products of a multigene family consisting of 150–200 genes. Previously, we demonstrated that some RIFINs downregulate the immune response through the leukocyte immunoglobulin-like receptor (LILR) family inhibitory receptor, LILRB1, and leukocyte-associated immunoglobulin-like receptor 1, LAIR1. In this study, we further analyzed the expression of inhibitory receptor ligands on P. falciparum-infected erythrocytes and found that P. falciparum-infected erythrocytes expressed ligands for another LILR family inhibitory receptor, LILRB2, that recognizes HLA class I molecules as a host ligand. Furthermore, we identified that a specific RIFIN was a ligand for LILRB2 by using a newly developed RIFIN expression library. In addition, the domain 3 of LILRB2 was involved in RIFIN binding, whereas the domains 1 and 2 of LILRB2 were involved in the binding to HLA class I molecules. These results suggest that inhibitory receptor LILRB2 is also targeted by RIFIN for immune evasion of P. falciparum similar to LILRB1 and LAIR1.
- Published
- 2021
- Full Text
- View/download PDF
14. Structural basis of malaria RIFIN binding by LILRB1-containing antibodies
- Author
-
Roger Geiger, Joshua Tan, Federica Sallusto, Baoshan Zhang, Kai Xu, Yiwei Chen, Peter D. Crompton, Yaroslav Tsybovsky, Peter D. Kwong, Luca Piccoli, Mathilde Foglierini, Antonio Lanzavecchia, Jason Gorman, Boubacar Traore, Claudia Daubenberger, Chiara Silacci-Fregni, and Wenjie Jin
- Subjects
Adult ,Models, Molecular ,0301 basic medicine ,Adolescent ,Plasmodium falciparum ,030106 microbiology ,Antigens, Protozoan ,Mali ,Antibodies ,Article ,Cohort Studies ,LILRB1 ,Young Adult ,03 medical and health sciences ,chemistry.chemical_compound ,Leukocyte Immunoglobulin-like Receptor B1 ,Protein Domains ,Antigen ,Antibody Specificity ,medicine ,Humans ,Amino Acid Sequence ,Child ,B cell ,Multidisciplinary ,biology ,LAIR1 ,Cryoelectron Microscopy ,Infant ,biology.organism_classification ,Molecular biology ,030104 developmental biology ,medicine.anatomical_structure ,Immunoglobulin class switching ,chemistry ,Child, Preschool ,biology.protein ,Binding Sites, Antibody ,Antibody ,DNA - Abstract
Some Plasmodium falciparum repetitive interspersed families of polypeptides (RIFINs)—variant surface antigens that are expressed on infected erythrocytes1—bind to the inhibitory receptor LAIR1, and insertion of DNA that encodes LAIR1 into immunoglobulin genes generates RIFIN-specific antibodies2,3. Here we address the general relevance of this finding by searching for antibodies that incorporate LILRB1, another inhibitory receptor that binds to β2 microglobulin and RIFINs through their apical domains4,5. By screening plasma from a cohort of donors from Mali, we identified individuals with LILRB1-containing antibodies. B cell clones isolated from three donors showed large DNA insertions in the switch region that encodes non-apical LILRB1 extracellular domain 3 and 4 (D3D4) or D3 alone in the variable–constant (VH–CH1) elbow. Through mass spectrometry and binding assays, we identified a large set of RIFINs that bind to LILRB1 D3. Crystal and cryo-electron microscopy structures of a RIFIN in complex with either LILRB1 D3D4 or a D3D4-containing antibody Fab revealed a mode of RIFIN–LILRB1 D3 interaction that is similar to that of RIFIN–LAIR1. The Fab showed an unconventional triangular architecture with the inserted LILRB1 domains opening up the VH–CH1 elbow without affecting VH–VL or CH1–CL pairing. Collectively, these findings show that RIFINs bind to LILRB1 through D3 and illustrate, with a naturally selected example, the general principle of creating novel antibodies by inserting receptor domains into the VH–CH1 elbow. Plasmodium antigens called RIFINs bind to specific antibodies that incorporate the inhibitory receptor LILRB1 through its D3 domain, illustrating the principle of receptor-containing antibodies.
- Published
- 2021
- Full Text
- View/download PDF
15. Collagen architecture and signaling orchestrate cancer development.
- Author
-
Su H and Karin M
- Subjects
- Humans, Collagen metabolism, Signal Transduction, Tumor Microenvironment, Neoplasms pathology
- Abstract
The tumor microenvironment (TME) controls tumor progression and maintenance. Accordingly, tumor-centric cancer treatment must adjust to being more holistic and TME-centric. Collagens are the most abundant TME proteins, and their dynamic remodeling profoundly affects both TME architecture and tumor development. Recent evidence shows that in addition to being structural elements, collagens are an important source of nutrients and decisive growth controlling and immunoregulatory signals. This review focuses on macropinocytosis-dependent collagen support of cancer cell metabolism and the role of collagen fiber remodeling and trimer heterogeneity in control of tumor bioenergetics, growth, progression, and response to therapy. If properly translated, these basic advances may alter the future of cancer treatment., Competing Interests: Declaration of interests None are declared by the authors., (Copyright © 2023 Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
16. Of myeloid cells and fibroblasts—A love story
- Author
-
Elvira Mass and Nikola Makdissi
- Subjects
LAIR1 ,Immunology ,Cancer ,Fibroblasts ,Biology ,medicine.disease ,Monocytes ,Pathophysiology ,Extracellular Matrix ,Collagen receptor ,Cell biology ,Extracellular matrix ,Crosstalk (biology) ,Infectious Diseases ,Immunity ,medicine ,Immunology and Allergy ,Myeloid Cells ,Collagen ,Tissue homeostasis - Abstract
The interaction between myeloid cells and the extracellular matrix is important for tissue homeostasis and pathophysiology. In this issue of Immunity, Keerthivasan et al. reveal crosstalk dependent on the collagen receptor LAIR1 that regulates the dynamics of monocytes and macrophages during steady-state and cancer.
- Published
- 2021
- Full Text
- View/download PDF
17. Structural basis of malarial parasite RIFIN-mediated immune escape against LAIR1
- Author
-
Xin Li, Yijia Xie, Hao Song, George F. Gao, Jianxun Qi, and Yan Chai
- Subjects
Cell signaling ,crystal structure ,Erythrocytes ,Protein Conformation ,QH301-705.5 ,Population ,Plasmodium falciparum ,Protozoan Proteins ,malaria ,Antibodies, Protozoan ,Antigens, Protozoan ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,LAIR1 ,Protein Domains ,Antigen ,Humans ,Parasite hosting ,Malaria, Falciparum ,Receptors, Immunologic ,Binding site ,Biology (General) ,education ,Immune Evasion ,Genetics ,education.field_of_study ,Binding Sites ,biology ,immune escape ,Membrane Proteins ,biology.organism_classification ,Mutation ,biology.protein ,RIFIN ,Antibody ,Protein Binding ,Signal Transduction - Abstract
Summary Malaria infection by Plasmodium falciparum continues to pose a global threat to the human population. P. falciparum expresses variable erythrocyte surface antigens such as RIFINs. Public antibodies with LAIR1 insertion have been identified from malarial patients against a subset of RIFINs. In this study, we solve a LAIR1-binding RIFIN structure: the complex structures of two RIFINs bound to mutated or wild-type LAIR1 in two distinct patterns. Notably, the two RIFINs engage similar binding sites on LAIR1 with different angles, and the RIFIN-binding sites overlap with the collagen-binding site. Surprisingly, RIFINs use completely different binding sites to bind to LAIR1 or LILRB1, indicating the kaleidoscopic change of RIFINs. We then verify that RIFIN could induce LAIR1-mediated cell signaling, and LAIR1-containing antibodies could block the pathway. The findings of this study provide structural insights into the mechanism of the immune escape of P. falciparum and the endless arms race between parasite and host.
- Published
- 2021
18. Elevated lymphocyte specific protein 1 expression is involved in the regulation of leukocyte migration and immunosuppressive microenvironment in glioblastoma
- Author
-
Guang-Yu Li, Cunyi Zou, Jing‐yuan Cao, Peng Cheng, Anhua Wu, Wen Cheng, Qing Guo, Guo-li Wang, Lu-yang Zhang, Chen Zhu, and Gefei Guan
- Subjects
Aging ,Leukocyte migration ,LSP1 ,Regulatory T cell ,medicine.medical_treatment ,Cell ,Biology ,migration ,Immunomodulation ,Risk Factors ,T-Lymphocyte Subsets ,Cell Line, Tumor ,Glioma ,Tumor Microenvironment ,medicine ,Humans ,Neoplasm Staging ,Tumor microenvironment ,immunosuppression ,Macrophages ,LAIR1 ,Microfilament Proteins ,glioblastoma ,Immunosuppression ,Cell Biology ,Prognosis ,medicine.disease ,microenvironment ,Isocitrate Dehydrogenase ,Gene Expression Regulation, Neoplastic ,Chemotaxis, Leukocyte ,medicine.anatomical_structure ,biology.protein ,Cancer research ,Neoplasm Grading ,Antibody ,Biomarkers ,Research Paper - Abstract
Immune cell infiltration mediates therapeutic response to immune therapies. The investigation on the genes regulating leukocyte migration may help us to understand the mechanisms regulating immune cell infiltration in tumor microenvironment. Here, we collected the data from Chinese Glioma Genome Atlas (CGGA) and The Cancer Genome Atlas (TCGA) to analyze the expression of leukocyte migration related genes in glioblastoma (GBM). Lymphocyte specific protein 1 (LSP1) was identified as the only gene in this family which not only has an elevated expression, but also serve as an independent predictive factor for progressive malignancy in glioma. We further confirmed these results in clinical glioma samples by quantitative PCR, immunofluorescence, immunohistochemistry, and western blot. Moreover, LSP1 expression was closely related to the response to radio- and chemotherapy in GBM, and positively correlated with immunosuppressive cell populations, including M2 macrophages, neutrophil, and regulatory T cell. Additionally, elevated LSP-1 expression enhanced the expression of immunosuppression related genes like programmed cell death 1 (PD1) and leukocyte associated immunoglobulin like receptor 1 (LAIR1) in macrophages. LSP1 also promoted the migration of macrophages. Together, our study suggests a novel role of LSP1 contributing to immunosuppressive microenvironment in GBM and serving as a potential therapeutic target for it.
- Published
- 2020
- Full Text
- View/download PDF
19. Expression analysis of LILRB3, LAIR1 and LXRA genes in porcine monocytes stimulated with Salmonella LPS
- Author
-
Kosuke Jyozaki, Yasuhiko Wada, Yuki Nishiyama, Eiko Kohara, Momoko Matsumoto, and Misato Ohara
- Subjects
Salmonella ,LILRB3 ,LAIR1 ,Expression analysis ,medicine ,Biology ,medicine.disease_cause ,Gene ,Molecular biology - Published
- 2020
- Full Text
- View/download PDF
20. Molecular basis of reduced LAIR1 expression in childhood severe malarial anaemia: Implications for leukocyte inhibitory signalling
- Author
-
Qiuying Cheng, John M. Ong'echa, Angela O. Achieng, Bernard Guyah, Collins Ouma, Christophe G. Lambert, and Douglas J Perkins
- Subjects
Male ,0301 basic medicine ,Research paper ,medicine.medical_treatment ,Interleukin-1beta ,PBMCs, Peripheral blood mononuclear cells ,LAIR2, Leukocyte-associated immunoglobulin like receptor-2 ,PfHz, Plasmodium falciparum haemozoin ,LAIR1, Leukocyte-associated immunoglobulin like receptor-1 ,SHP-1, SH2 domain-containing tyrosine phosphatase-1 ,Pathogenesis ,chemistry.chemical_compound ,0302 clinical medicine ,PCN, Pigment containing neutrophils ,TNF-α, Tumor necrosis factor alpha ,Malaria, Falciparum ,Receptors, Immunologic ,050207 economics ,Receptor ,Leukocyte-associated immunoglobulin like receptor-1 ,Leukocyte-associated immunoglobulin like receptor-2 ,P. falciparum, Plasmodium falciparum ,050208 finance ,CLL, Chronic lymphocytic leukaemia ,ITIM, immuno-tyrosine inhibition motifs ,biology ,Protein Tyrosine Phosphatase, Non-Receptor Type 6 ,05 social sciences ,Anemia ,General Medicine ,SMA ,Severe malarial anaemia ,3. Good health ,HFRS, haemorrhagic fever with renal syndrome ,Child, Preschool ,HIV-1, Human immunodeficiency virus 1 ,030220 oncology & carcinogenesis ,IL-6, Interleukin 6 ,Female ,Tumor necrosis factor alpha ,Antibody ,Signal Transduction ,C1q, Complement component 1q ,Hemeproteins ,SHP-2, SH2 domain-containing tyrosine phosphatase-2 ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Immune system ,Phagocytosis ,SLE, Systemic lupus erythematosus ,Complement component 1q ,In vivo ,0502 economics and business ,medicine ,Humans ,SMA, severe malarial anaemia ,Interleukin 6 ,Plasmodium falciparum malaria ,Interleukin-6 ,Tumor Necrosis Factor-alpha ,business.industry ,Complement C1q ,LAIR1 ,Plasmodium falciparum haemozoin ,Antagonist ,Infant ,PCM, Pigment containing monocytes ,NF-κB ,Immunotherapy ,AML, Acute myeloid leukaemia ,030104 developmental biology ,chemistry ,NF-κB, Nuclear factor-kappa beta ,Immunology ,Leukocytes, Mononuclear ,IL-1β, Interleukin 1 beta ,biology.protein ,PCR, Polymerase chain reaction ,business ,Biomedical sciences - Abstract
Background: Leukocyte-associated immunoglobulin like receptor-1 (LAIR1) is a transmembrane inhibitory receptor that influences susceptibility to a myriad of inflammatory diseases. Our recent investigations of severe malarial anemia (SMA) pathogenesis in Kenyan children discovered that novel LAIR1 genetic variants associated with decreased LAIR1 enhanced the longitudinal risk of SMA and all-cause mortality. Methods: To further characterize LAIR1 expression and identify novel molecular mechanisms, at least in part, responsible for reduced LAIR1 in severe malaria, we conducted a series of experiments in samples from children with malaria, followed by in vitro investigations driven by the in vivo findings. Findings: Kenyan children with SMA had elevated circulating levels of soluble LAIR1 (sLAIR1) relative to non-SMA (1.69-fold P
- Published
- 2019
- Full Text
- View/download PDF
21. Structural basis of LAIR1 targeting by polymorphic Plasmodium RIFINs
- Author
-
Peter D. Kwong, Shuishu Wang, Raffaello Verardi, Jason Gorman, Chen-Hsiang Shen, Gwo-Yu Chuang, Kai Xu, Baoshan Zhang, Yongping Yang, Yiran Wang, Tyler Stephens, Kevin Liu, Yaroslav Tsybovsky, S. Katie Farney, Tongqing Zhou, Antonio Lanzavecchia, Luca Piccoli, and Yiwei Chen
- Subjects
Sequence analysis ,Science ,Plasmodium falciparum ,Protozoan Proteins ,Antibodies, Protozoan ,General Physics and Astronomy ,Antigens, Protozoan ,Crystallography, X-Ray ,medicine.disease_cause ,Plasmodium ,Article ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Protein Domains ,Antigen ,parasitic diseases ,medicine ,Humans ,Malaria, Falciparum ,Receptors, Immunologic ,Receptor ,X-ray crystallography ,030304 developmental biology ,Genetics ,0303 health sciences ,Mutation ,Multidisciplinary ,biology ,LAIR1 ,Membrane Proteins ,General Chemistry ,biology.organism_classification ,Antigenic Variation ,Malaria ,biology.protein ,Antibody ,030217 neurology & neurosurgery - Abstract
RIFIN, a large family of Plasmodium variant surface antigens, plays a crucial role in malaria pathogenesis by mediating immune suppression through activation of inhibitory receptors such as LAIR1, and antibodies with LAIR1 inserts have been identified that bind infected erythrocytes through RIFIN. However, details of RIFIN-mediated LAIR1 recognition and receptor activation have been unclear. Here, we use negative-stain EM to define the architecture of LAIR1-inserted antibodies and determine crystal structures of RIFIN-variable 2 (V2) domain in complex with a LAIR1 domain. These structures reveal the LAIR1-binding region of RIFIN to be hydrophobic and membrane-distal, to exhibit extensive structural diversity, and to interact with RIFIN-V2 in a one-to-one fashion. Through structural and sequence analysis of various LAIR1 constructs, we identify essential elements of RIFIN-binding on LAIR1. Furthermore, a structure-derived LAIR1-binding sequence signature ascertained >20 LAIR1-binding RIFINs, including some from P. falciparum field strains and Plasmodium species infecting gorillas and chimpanzees., Nature Communications, 12, ISSN:2041-1723
- Published
- 2021
- Full Text
- View/download PDF
22. Tissue-specific transcriptional profiling of plasmacytoid dendritic cells reveals a hyperactivated state in chronic SIV infection
- Author
-
Simon M. Barratt-Boyes, Steven E. Bosinger, Gregory K. Tharp, Chi N. Chan, Justin L. Harper, Amit A. Upadhyay, Elizabeth R. Wonderlich, Kirti A. Karunakaran, Guido Silvestri, Kiran Gill, Diane G. Carnathan, Reem A. Dawoud, Ernestine A. Mahar, Jacob D. Estes, Sydney A. Nelson, Vijayakumar Velu, Michelle Y.H. Lee, Barbara Cervasi, Hasse Walum, Kyndal L. Goss, Christine Grech, and Mirko Paiardini
- Subjects
RNA viruses ,Physiology ,Cell ,Simian Acquired Immunodeficiency Syndrome ,Monkeys ,Pathology and Laboratory Medicine ,Transcriptome ,White Blood Cells ,Spectrum Analysis Techniques ,Immunodeficiency Viruses ,Animal Cells ,Medicine and Health Sciences ,Cytotoxic T cell ,RNA-Seq ,Biology (General) ,Mammals ,0303 health sciences ,education.field_of_study ,T Cells ,Eukaryota ,virus diseases ,hemic and immune systems ,Flow Cytometry ,Body Fluids ,Blood ,medicine.anatomical_structure ,SIV ,Medical Microbiology ,Spectrophotometry ,Viral Pathogens ,Viruses ,Vertebrates ,Infectious diseases ,Cytophotometry ,Pathogens ,Cellular Types ,Anatomy ,Macaque ,Research Article ,HIV infections ,Primates ,Medical conditions ,QH301-705.5 ,Immune Cells ,T cell ,Immunology ,Population ,Viral diseases ,Biology ,Research and Analysis Methods ,Microbiology ,Lymphatic System ,Cercocebus atys ,03 medical and health sciences ,TIGIT ,Virology ,Retroviruses ,Old World monkeys ,Genetics ,medicine ,Animals ,Molecular Biology Techniques ,education ,Microbial Pathogens ,Molecular Biology ,030304 developmental biology ,Blood Cells ,030306 microbiology ,Gene Expression Profiling ,LAIR1 ,Lentivirus ,Organisms ,Biology and Life Sciences ,Cell Biology ,Dendritic Cells ,RC581-607 ,Macaca mulatta ,Chronic infection ,Amniotes ,Parasitology ,Lymph Nodes ,Immunologic diseases. Allergy ,Zoology ,Cloning - Abstract
HIV associated immune activation (IA) is associated with increased morbidity in people living with HIV (PLWH) on antiretroviral therapy, and remains a barrier for strategies aimed at reducing the HIV reservoir. The underlying mechanisms of IA have not been definitively elucidated, however, persistent production of Type I IFNs and expression of ISGs is considered to be one of the primary factors. Plasmacytoid DCs (pDCs) are a major producer of Type I IFN during viral infections, and are highly immunomodulatory in acute HIV and SIV infection, however their role in chronic HIV/SIV infection has not been firmly established. Here, we performed a detailed transcriptomic characterization of pDCs in chronic SIV infection in rhesus macaques, and in sooty mangabeys, a natural host non-human primate (NHP) species that undergoes non-pathogenic SIV infection. We also investigated the immunostimulatory capacity of lymph node homing pDCs in chronic SIV infection by contrasting gene expression of pDCs isolated from lymph nodes with those from blood. We observed that pDCs in LNs, but not blood, produced high levels of IFNα transcripts, and upregulated gene expression programs consistent with T cell activation and exhaustion. We apply a novel strategy to catalogue uncharacterized surface molecules on pDCs, and identified the lymphoid exhaustion markers TIGIT and LAIR1 as highly expressed in SIV infection. pDCs from SIV-infected sooty mangabeys lacked the activation profile of ISG signatures observed in infected macaques. These data demonstrate that pDCs are a primary producer of Type I IFN in chronic SIV infection. Further, this study demonstrated that pDCs trafficking to LNs persist in a highly activated state well into chronic infection. Collectively, these data identify pDCs as a highly immunomodulatory cell population in chronic SIV infection, and a putative therapeutic target to reduce immune activation., Author summary For people living with HIV (PLWH), persistent immune activation is an obstacle to optimal health. In this study, we investigate the immunostimulatory potential of plasmacytoid dendritic cells in chronic SIV infection using comparative RNA-Seq. We observed that pDCs from SIV-infected rhesus macaques have highly activated profiles relative to uninfected animals; in contrast, pDCs from SIV-infected natural host sooty mangabeys had expression profiles similar to cells from uninfected animals. In chronically infected RMs, pDCs from lymph nodes maintained activation profiles elevated at levels even higher than those in the blood. Further, transcripts for the immunostimulatory cytokine family IFNA were readily detected in LN homing pDCs, but not those from blood. These data confirm pDCs as a major producer of Type I IFN in chronic SIV infection, and identify them as a target for immunotherapy.
- Published
- 2021
23. Antibody Display of cell surface receptor Tetraspanin12 and SARS-CoV-2 spike protein
- Author
-
Fu-Lien Hsieh and Tao-Hsin Chang
- Subjects
chemistry.chemical_classification ,Enzyme ,biology ,chemistry ,Cell surface receptor ,LAIR1 ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,biology.protein ,Extracellular ,Spike Protein ,Antibody ,Receptor ,Cell biology - Abstract
In previous work, Hsieh and Higgins presented a novel structure of antibodies identified from malaria-exposed individuals, in which the extracellular immunoglobulin (Ig)-like domain of leukocyte-associated immunoglobulin-like receptor 1 (LAIR1) is presented on the third complementarity determining regions (CDR3) of the Ig heavy chain. Here we develop an Antibody Display technology based on this LAIR1-containing antibody, by grafting proteins of interest (POI) onto the heavy chain CDR3 while retaining the biological properties of the POI. As a proof of principle, we displayed the second extracellular domain of Tetraspanin12 (Tspan12EC2) and the receptor-binding domain (RBD) of SARS-CoV-2 spike protein on the heavy chain CDR3. Our data revealed that Antibody Display Tspan12EC2 bound to Norrie Disease Protein (Norrin) and Antibody Display SARS-CoV-2 RBD bound to angiotensin-converting enzyme 2 (ACE2) and neutralizing nanobodies. Collectively, Antibody Display technology offers the general strategy of designing novel antibodies by grafting POI onto the CDR3.
- Published
- 2021
- Full Text
- View/download PDF
24. A novel potential target of IL‐35‐regulated JAK/STAT signaling pathway in lupus nephritis
- Author
-
Yu Feng, Huasong Zeng, Ping Wei, Chun-Kwok Wong, Zhe Cai, Qi Ren, Hong Tang, Song Zhang, Ping Wu, and Ming Hong
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Male ,Lupus nephritis ,Medicine (miscellaneous) ,IL‐35 ,CD19 ,stat ,03 medical and health sciences ,Mice ,LAIR1 ,0302 clinical medicine ,medicine ,Animals ,Humans ,Child ,Research Articles ,Janus Kinases ,JAK/STAT signaling pathway ,Mice, Inbred BALB C ,lcsh:R5-920 ,Systemic lupus erythematosus ,biology ,business.industry ,Interleukins ,JAK-STAT signaling pathway ,medicine.disease ,Lupus Nephritis ,JSLE‐LN ,Disease Models, Animal ,STAT Transcription Factors ,030104 developmental biology ,030220 oncology & carcinogenesis ,Child, Preschool ,Cancer research ,biology.protein ,Molecular Medicine ,Female ,Signal transduction ,mesangial calls ,Janus kinase ,business ,lcsh:Medicine (General) ,Biomarkers ,Research Article ,Signal Transduction - Abstract
Background In this study, we have investigated the potential regulatory mechanisms of IL‐35 to relieve lupus nephritis (LN) through regulating Janus kinase (JAK)/signal transducers and activators of transcription (STAT) signaling pathway in mesangial cells. Results Among 105 significant differentially expressed proteins (DEPs) between juvenile systemic lupus erythematosus (JSLE) patients with LN and healthy controls, LAIR1, PDGFRβ, VTN, EPHB4, and EPHA4 were downregulated in JSLE‐LN. They consist of an interactive network with PTPN11 and FN1, which involved in IL‐35‐related JAK/STAT signaling pathway. Besides, urinary LAIR1 was significantly correlated with JSLE‐LN clinical parameters such as SLEDAI‐2K, %CD19+ B, and %CD3+ T cells. Through bioinformatics analysis of co‐immunoprecipitation with mass spectrometry results, including GO, KEGG, and STRING, five genes interacted with Lair1 were upregulated by IL‐35, but only Myh10 was downregulated. Therefore, we presumed an interactive network among these DEPs, JAK/STAT, and IL‐35. Moreover, the downregulated phosphorylated (p)‐STAT3, p‐p38 MAPK, and p‐ERK, and the upregulated p‐JAK2/p‐STAT1/4 in IL‐35 overexpressed mesangial cells, and RNA‐sequencing results validated the potential regulatory mechanisms of IL‐35 in alleviating JSLE‐LN disease. Moreover, the relieved histopathological features of nephritis including urine protein and leukocyte scores, a decreased %CD90+αSMA+ mesangial cells and pro‐inflammatory cytokines, the inactivated JAK/STAT signals and the significant upregulated Tregs in spleen, thymus and peripheral blood were validated in Tregs and IL‐35 overexpression plasmid‐treated lupus mice. Conclusions Our study provided a reference proteomic map of urinary biomarkers for JSLE‐LN and elucidated evidence that IL‐35 may regulate the interactive network of LAIR1‐PTPN11‐JAK‐STAT‐FN1 to affect JAK/STAT and MAPK signaling pathways to alleviate inflammation in JSLE‐LN. This finding may provide a further prospective mechanism for JSLE‐LN clinical treatment., A reference proteomic map of urinary biomarkers contributes to the diagnosis of JSLE‐LN. IL‐35 may interact with LAIR1‐PTPN11‐JAK‐STAT‐FN1 to affect JAK/STAT and MAPK signaling pathways to alleviate inflammation in vivo and in vitro. LAIR1 could be a novel potential target of IL‐35‐regulated JAK/STAT signaling pathway in JSLE‐LN.
- Published
- 2021
25. Cancer immunotherapy by NC410, a LAIR-2 Fc protein blocking human LAIR-collagen interaction
- Author
-
Ana Paucarmayta, Emma J de Ruiter, Dallas Flies, Nicholas Willumsen, Akashdip Singh, Linjie Tian, Linda Liu, Saskia V. Vijver, Morten A. Karsdal, M. Inês Pascoal Ramos, Linde Meyaard, Chang Song, Stefan M. Willems, Sol Langermann, Jason Bosiacki, Jahangheer Shaik, Eline Elshof, Christina Jensen, and Zachary Cusumano
- Subjects
0301 basic medicine ,collagen ,tumor ,Mouse ,QH301-705.5 ,Science ,medicine.medical_treatment ,T cell ,Recombinant Fusion Proteins ,Immune receptor ,General Biochemistry, Genetics and Molecular Biology ,Extracellular matrix ,03 medical and health sciences ,Mice ,LAIR1 ,0302 clinical medicine ,Immune system ,Antineoplastic Agents, Immunological ,Cancer immunotherapy ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,Biology (General) ,Receptors, Immunologic ,Receptor ,Cancer Biology ,General Immunology and Microbiology ,Chemistry ,General Neuroscience ,Computational Biology ,General Medicine ,Xenograft Model Antitumor Assays ,Cell biology ,Immunoglobulin Fc Fragments ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunoglobulin G ,Humanized mouse ,Medicine ,Immunotherapy ,Research Article - Abstract
Collagens are a primary component of the extracellular matrix and are functional ligands for the inhibitory immune receptor leukocyte-associated immunoglobulin-like receptor (LAIR)-1. LAIR-2 is a secreted protein that can act as a decoy receptor by binding collagen with higher affinity than LAIR-1. We propose that collagens promote immune evasion by interacting with LAIR-1 expressed on immune cells, and that LAIR-2 releases LAIR-1-mediated immune suppression. Analysis of public human datasets shows that collagens, LAIR-1 and LAIR-2 have unique and overlapping associations with survival in certain tumors. We designed a dimeric LAIR-2 with a functional IgG1 Fc tail, NC410, and showed that NC410 increases human T cell expansion and effector function in vivo in a mouse xenogeneic-graft versus-host disease model. In humanized mouse tumor models, NC410 reduces tumor growth that is dependent on T cells. Immunohistochemical analysis of human tumors shows that NC410 binds to collagen-rich areas where LAIR-1+ immune cells are localized. Our findings show that NC410 might be a novel strategy for cancer immunotherapy for immune-excluded tumors.
- Published
- 2020
26. Leukocyte-Associated Ig-like Receptor 1 Inhibits Th1 Responses but Is Required for Natural and Induced Monocyte-Dependent Th17 Responses
- Author
-
Jeremy A. Sullivan, Ewa Jankowska-Gan, Jose R. Torrealba, Vrushali V. Agashe, David S. Wilkes, William J. Burlingham, Melissa R. Keller, Melanie Dart, Drew A. Roenneburg, Marco Colonna, Lynn D. Haynes, and John F. Kernien
- Subjects
0301 basic medicine ,Chemistry ,Cell growth ,Monocyte ,LAIR1 ,Immunology ,Cell ,Collagen receptor ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,medicine.anatomical_structure ,Immunity ,medicine ,Immunology and Allergy ,Receptor ,030215 immunology - Abstract
Leukocyte-associated Ig-like receptor 1 (LAIR1) is an ITIM-bearing collagen receptor expressed by leukocytes and is implicated in immune suppression. However, using a divalent soluble LAIR1/Fc recombinant protein to block interaction of cell surface LAIR1 with matrix collagen, we found that whereas Th1 responses were enhanced as predicted, Th17 responses were strongly inhibited. Indeed, LAIR1 on both T cells and monocytes was required for optimal Th17 responses to collagen type (Col)V. For pre-existing “natural” Th17 response to ColV, the LAIR1 requirement was absolute, whereas adaptive Th17 and Th1/17 immune responses in both mice and humans were profoundly reduced in the absence of LAIR1. Furthermore, the addition of C1q, a natural LAIR1 ligand, decreased Th1 responses in a dose-dependent manner, but it had no effect on Th17 responses. In IL-17–dependent murine organ transplant models of chronic rejection, LAIR1+/+ but not LAIR1−/− littermates mounted strong fibroproliferative responses. Surface LAIR1 expression was higher on human Th17 cells as compared with Th1 cells, ruling out a receptor deficiency that could account for the differences. We conclude that LAIR1 ligation by its natural ligands favors Th17 cell development, allowing for preferential activity of these cells in collagen-rich environments. The emergence of cryptic self-antigens such as the LAIR1 ligand ColV during ischemia/reperfusion injury and early acute rejection, as well as the tendency of macrophages/monocytes to accumulate in the allograft during chronic rejection, favors Th17 over Th1 development, posing a risk to long-term graft survival.
- Published
- 2018
- Full Text
- View/download PDF
27. LAIR1
- Author
-
Schwab, Manfred, editor
- Published
- 2011
- Full Text
- View/download PDF
28. Blocking LAIR1 signaling in immune cells inhibits tumor development.
- Author
-
Xie J, Gui X, Deng M, Chen H, Chen Y, Liu X, Ku Z, Tan L, Huang R, He Y, Zhang B, Lewis C, Chen K, Xu L, Xu J, Huang T, Liao XC, Zhang N, An Z, and Zhang CC
- Subjects
- Animals, Antibodies, Monoclonal therapeutic use, Humans, Immunotherapy, Mice, T-Lymphocytes, Regulatory, Immune Checkpoint Inhibitors, Neoplasms
- Abstract
The current immune checkpoint blockade therapy has been successful in treating some cancers but not others. New molecular targets and therapeutic approaches of cancer immunology need to be identified. Leukocyte associated immunoglobulin like receptor 1 (LAIR1) is an immune inhibitory receptor expressing on most immune cell types. However, it remains a question whether we can specifically and actively block LAIR1 signaling to activate immune responses for cancer treatment. Here we report the development of specific antagonistic anti-LAIR1 monoclonal antibodies and studied the effects of LAIR1 blockade on the anti-tumor immune functions. The anti-LAIR1 antagonistic antibody stimulated the activities of T cells, natural killer cells, macrophages, and dendritic cells in vitro . The single-cell RNA sequencing analysis of intratumoral immune cells in syngeneic human LAIR1 transgenic mice treated with control or anti-LAIR1 antagonist antibodies indicates that LAIR1 signaling blockade increased the numbers of CD4 memory T cells and inflammatory macrophages, but decreased those of pro-tumor macrophages, regulatory T cells, and plasmacytoid dendritic cells. Importantly, the LAIR1 blockade by the antagonistic antibody inhibited the activity of immunosuppressive myeloid cells and reactivated T cells from cancer patients in vitro and impeded tumor metastasis in a humanized mouse model. Blocking LAIR1 signaling in immune cells represents a promising strategy for development of anti-cancer immunotherapy., Competing Interests: JJX, XG, XCL, NZ, ZA, CCZ, and XCL were inventors of a patent application covering anti-LAIR1 antibodies and their uses. NZ, ZA, and CCZ hold equity in and have Sponsored Research Agreements with Immune-Onc Therapeutics, Inc. XCL and TH are employees and hold equities of Immune-Onc Therapeutics, Inc. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Xie, Gui, Deng, Chen, Chen, Liu, Ku, Tan, Huang, He, Zhang, Lewis, Chen, Xu, Xu, Huang, Liao, Zhang, An and Zhang.)
- Published
- 2022
- Full Text
- View/download PDF
29. LAIR1
- Author
-
Schwab, Manfred, editor
- Published
- 2009
- Full Text
- View/download PDF
30. LAIR2 localizes specifically to sites of extravillous trophoblast invasion.
- Author
-
Founds, S.A., Fallert-Junecko, B., Reinhart, T.A., Conley, Y.P., and Parks, W.T.
- Subjects
TROPHOBLAST ,GENE expression ,CHORIONIC villus sampling ,IMMUNOGLOBULINS ,PLACENTA ,IMMUNOHISTOCHEMISTRY ,IN situ hybridization ,PREECLAMPSIA - Abstract
Abstract: Purpose: A global gene expression microarray analysis of surplus chorionic villus sampling (CVS) tissues identified leukocyte-associated immunoglobulin-like receptor 2 (LAIR2) as down-regulated in the first trimester of pregnancies destined for preeclampsia. Neither the localization nor the function of LAIR2 has been examined in the placenta. Localization studies were conducted in placental tissues to determine the precise sites of LAIR2 mRNA production and protein binding. Results: Quantitative real time polymerase chain reaction (qRT-PCR) indicated LAIR2 expression in CVS, but none in breast, lymph node, kidney, skin, uterus, or third trimester placentas. In situ hybridization (ISH) revealed a highly restricted LAIR2 localization. LAIR2 mRNA was found only in the more distal portions of trophoblast anchoring cell columns, adjacent to the invading extravillous trophoblast (EVT). Immunohistochemistry (IHC) detected intracellular LAIR2 staining in these same cells. Extracellular staining of this soluble receptor was found in the acellular material between invasive EVT cells distal to the anchoring cell columns. Conclusions: ISH and IHC staining for LAIR2 detected specific, highly localized expression at the leading edge of EVT anchoring cell columns in first trimester placentas. This staining likely identifies the site of production for this soluble receptor. Following secretion, the receptor appears to bind extracellular material among the invasive EVT. The precise restriction of this protein only to the sites of EVT invasion strongly suggests that it functions to regulate this invasion. The decreased LAIR2 expression noted in first trimester placentas that ultimately developed preeclampsia further suggests that alterations in LAIR2 may play an etiologic role in preeclampsia. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
31. Immune evasion of Plasmodium falciparum by RIFIN via inhibitory receptors
- Author
-
Thomas Lavstsen, Shiroh Iwanaga, Junichi Takagi, Toshihiro Horii, John Lusingu, Takafumi Tsuboi, Sawako Itagaki, Marco Colonna, Hisashi Arase, Takao Arimori, Nirianne Marie Q. Palacpac, Fumiji Saito, Tadahiro Suenaga, Eizo Takashima, Christian W. Wang, Masako Kohyama, Takeshi Satoh, Kouyuki Hirayasu, and Kyoko Shida
- Subjects
0301 basic medicine ,Erythrocytes ,Plasmodium falciparum ,Protozoan Proteins ,CHO Cells ,Biology ,Ligands ,LILRB1 ,03 medical and health sciences ,Cricetulus ,Leukocyte Immunoglobulin-like Receptor B1 ,0302 clinical medicine ,Immune system ,parasitic diseases ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Malaria, Falciparum ,Receptors, Immunologic ,Receptor ,Gene ,Immune Evasion ,B-Lymphocytes ,Multidisciplinary ,LAIR1 ,Membrane Proteins ,medicine.disease ,biology.organism_classification ,Acquired immune system ,Virology ,3. Good health ,Killer Cells, Natural ,HEK293 Cells ,030104 developmental biology ,Sample Size ,030220 oncology & carcinogenesis ,Malaria - Abstract
Malaria is among the most serious infectious diseases affecting humans, accounting for approximately half a million deaths each year. Plasmodium falciparum causes most life-threatening cases of malaria. Acquired immunity to malaria is inefficient, even after repeated exposure to P. falciparum, but the immune regulatory mechanisms used by P. falciparum remain largely unknown. Here we show that P. falciparum uses immune inhibitory receptors to achieve immune evasion. RIFIN proteins are products of a polymorphic multigene family comprising approximately 150-200 genes per parasite genome that are expressed on the surface of infected erythrocytes. We found that a subset of RIFINs binds to either leucocyte immunoglobulin-like receptor B1 (LILRB1) or leucocyte-associated immunoglobulin-like receptor 1 (LAIR1). LILRB1-binding RIFINs inhibit activation of LILRB1-expressing B cells and natural killer (NK) cells. Furthermore, P. falciparum-infected erythrocytes isolated from patients with severe malaria were more likely to interact with LILRB1 than erythrocytes from patients with non-severe malaria, although an extended study with larger sample sizes is required to confirm this finding. Our results suggest that P. falciparum has acquired multiple RIFINs to evade the host immune system by targeting immune inhibitory receptors.
- Published
- 2017
- Full Text
- View/download PDF
32. Structural basis for RIFIN-mediated activation of LILRB1 in malaria
- Author
-
Adam J. Reid, Matthew K. Higgins, James H. Felce, Akihito Sakoguchi, Hisashi Arase, Michael L. Dustin, Thomas E. Harrison, and Alexander M. Mørch
- Subjects
0301 basic medicine ,Models, Molecular ,Plasmodium ,Erythrocytes ,Plasmodium falciparum ,Lipid Bilayers ,Protozoan Proteins ,Cell Communication ,Major histocompatibility complex ,Ligands ,Lymphocyte Activation ,Article ,Immunological synapse ,03 medical and health sciences ,Immune system ,Leukocyte Immunoglobulin-like Receptor B1 ,Antigens, CD ,MHC class I ,Animals ,Humans ,Amino Acid Sequence ,Malaria, Falciparum ,Multidisciplinary ,Binding Sites ,030102 biochemistry & molecular biology ,biology ,Perforin ,LAIR1 ,Intracellular parasite ,Histocompatibility Antigens Class I ,Molecular Mimicry ,Antibody-Dependent Cell Cytotoxicity ,Membrane Proteins ,biology.organism_classification ,Malaria ,Cell biology ,Killer Cells, Natural ,030104 developmental biology ,Mutation ,biology.protein ,Signal Transduction - Abstract
The Plasmodium species that cause malaria are obligate intracellular parasites, and disease symptoms occur when these parasites replicate in human blood. Despite the risk of immune detection, the parasite delivers proteins that bind to host receptors on the cell surfaces of infected erythrocytes. In the causative parasite of the most deadly form of malaria in humans, Plasmodium falciparum, RIFINs form the largest family of surface proteins displayed by erythrocytes1. Some RIFINs can bind to inhibitory immune receptors, and these RIFINs act as targets for unusual antibodies that contain a LAIR1 ectodomain2–4 or as ligands for LILRB15. RIFINs stimulate the activation of and signalling by LILRB15, which could potentially lead to the dampening of human immune responses. Here, to understand how RIFINs activate LILRB1-mediated signalling, we determine the structure of a RIFIN bound to LILRB1. We show that this RIFIN mimics the natural activating ligand of LILRB1, MHC class I, in its LILRB1-binding mode. A single mutation in the RIFIN disrupts the complex, blocks LILRB1 binding of all tested RIFINs and abolishes signalling in a reporter assay. In a supported lipid bilayer system, which mimics the activation of natural killer (NK) cells by antibody-dependent cell-mediated cytotoxicity, both RIFIN and MHC are recruited to the immunological synapse of NK cells and reduce the activation of NK cells, as measured by the mobilization of perforin. Therefore, LILRB1-binding RIFINs mimic the binding mode of the natural ligand of LILRB1 and suppress the function of NK cells. The structure of a RIFIN–LILRB1 complex reveals that a subset of RIFINs of Plasmodium falciparum mimics the binding mode of the natural ligand of human LILRB1 and suppress the function of natural killer cells in humans.
- Published
- 2019
33. The structure of a LAIR1-containing human antibody reveals a novel mechanism of antigen recognition
- Author
-
Fu-Lien Hsieh and Matthew K Higgins
- Subjects
Models, Molecular ,Binding Sites ,QH301-705.5 ,Protein Conformation ,Science ,Immunology ,Short Report ,Crystallography, X-Ray ,Antibodies ,LAIR1 ,Immunoglobulin Fab Fragments ,antibody structure ,None ,Medicine ,Humans ,Biology (General) ,Antigens ,Receptors, Immunologic ,novel antigen recognition ,Protein Binding - Abstract
Antibodies are critical components of the human adaptive immune system, providing versatile scaffolds to display diverse antigen-binding surfaces. Nevertheless, most antibodies have similar architectures, with the variable immunoglobulin domains of the heavy and light chain each providing three hypervariable loops, which are varied to generate diversity. The recent identification of a novel class of antibody in humans from malaria endemic regions of Africa was therefore surprising as one hypervariable loop contains the entire collagen-binding domain of human LAIR1. Here, we present the structure of the Fab fragment of such an antibody. We show that its antigen-binding site has adopted an architecture that positions LAIR1, while itself being occluded. This therefore represents a novel means of antigen recognition, in which the Fab fragment of an antibody acts as an adaptor, linking a human protein insert with antigen-binding potential to the constant antibody regions which mediate immune cell recruitment. DOI: http://dx.doi.org/10.7554/eLife.27311.001, eLife digest When bacteria, viruses or parasites invade the human body, the immune system responds by producing proteins called antibodies. Antibodies recognize and bind to molecules (known as antigens) on the surface of the invaders. This binding can either neutralize the invader directly or trigger signals that cause other parts of the immune system to destroy it. Our blood contains a huge range of different antibody molecules that each bind to a different antigen. This is despite most human antibodies having the same basic shape and structure. Six loops, known as complementarity determining regions (CDRs), emerge from the surface of the antibody to form the surface that recognizes the antigen. However, variations in the structure of the loops alter this surface enough to allow different antibodies to recognize completely different molecules. In 2016, a new class of antibodies was identified. Unlike previously identified antibodies, these molecules had an entire human protein, called LAIR1, inserted into one of their CDR loops. Members of this group of antibodies bind to a molecule, known as a RIFIN, that is found on the surface of human red blood cells that are infected with the parasite that causes malaria. How do LAIR1-containing antibodies bind to their RIFIN targets? Hsieh and Higgins investigated this question by using a technique called X-ray crystallography to determine the structure of the antibody. This revealed that instead of binding directly to an antigen, all of the six CDR loops in the LAIR1-containing antibody bind to the LAIR1 insert. By doing so, LAIR1 is oriented in a manner that enables it to bind to the RIFIN molecule from the parasite. This is the first known example of an antibody that recruits another protein to bind to an antigen rather than binding directly to the pathogen itself. A future challenge will be to see if other antibodies exist that use this mechanism and whether it can be employed to design new therapeutic antibodies. DOI: http://dx.doi.org/10.7554/eLife.27311.002
- Published
- 2019
34. Structural basis of malarial parasite RIFIN-mediated immune escape against LAIR1.
- Author
-
Xie, Yijia, Li, Xin, Chai, Yan, Song, Hao, Qi, Jianxun, and Gao, George F.
- Abstract
Malaria infection by Plasmodium falciparum continues to pose a global threat to the human population. P. falciparum expresses variable erythrocyte surface antigens such as RIFINs. Public antibodies with LAIR1 insertion have been identified from malarial patients against a subset of RIFINs. In this study, we solve a LAIR1-binding RIFIN structure: the complex structures of two RIFINs bound to mutated or wild-type LAIR1 in two distinct patterns. Notably, the two RIFINs engage similar binding sites on LAIR1 with different angles, and the RIFIN-binding sites overlap with the collagen-binding site. Surprisingly, RIFINs use completely different binding sites to bind to LAIR1 or LILRB1, indicating the kaleidoscopic change of RIFINs. We then verify that RIFIN could induce LAIR1-mediated cell signaling, and LAIR1-containing antibodies could block the pathway. The findings of this study provide structural insights into the mechanism of the immune escape of P. falciparum and the endless arms race between parasite and host. [Display omitted] • Variable region of RIFINs reveals a primarily α-helical structure with loops • Two RIFINs bind to similar sites of LAIR1 with different angles • RIFIN can induce LAIR1-mediated cell signaling • Antibodies with a mutated LAIR1 insert can block the RIFIN-LAIR1 interaction Plasmodium falciparum expresses RIFINs for pathogenesis, and antibodies with mutated LAIR1 insertions have been identified from malarial patients against RIFINs. Xie et al. report the mechanisms behind the immune escape of P. falciparum and host immune responses by the RIFIN-LAIR1 interaction. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
35. Homeostatic functions of monocytes and interstitial lung macrophages are regulated via collagen domain-binding receptor LAIR1.
- Author
-
Keerthivasan, Shilpa, Şenbabaoğlu, Yasin, Martinez-Martin, Nadia, Husain, Bushra, Verschueren, Erik, Wong, Anne, Yang, Yeqing Angela, Sun, Yonglian, Pham, Victoria, Hinkle, Trent, Oei, Yoko, Madireddi, Shravan, Corpuz, Racquel, Tam, Lucinda, Carlisle, Samantha, Roose-Girma, Merone, Modrusan, Zora, Ye, Zhengmao, Koerber, James T., and Turley, Shannon J.
- Subjects
- *
MYELOID cells , *MONOCYTES , *MACROPHAGES , *CELL physiology , *BONE marrow , *COLLAGEN - Abstract
Myeloid cells encounter stromal cells and their matrix determinants on a continual basis during their residence in any given organ. Here, we examined the impact of the collagen receptor LAIR1 on myeloid cell homeostasis and function. LAIR1 was highly expressed in the myeloid lineage and enriched in non-classical monocytes. Proteomic definition of the LAIR1 interactome identified stromal factor Colec12 as a high-affinity LAIR1 ligand. Proteomic profiling of LAIR1 signaling triggered by Collagen1 and Colec12 highlighted pathways associated with survival, proliferation, and differentiation. Lair1 −/− mice had reduced frequencies of Ly6C− monocytes, which were associated with altered proliferation and apoptosis of non-classical monocytes from bone marrow and altered heterogeneity of interstitial macrophages in lung. Myeloid-specific LAIR1 deficiency promoted metastatic growth in a melanoma model and LAIR1 expression associated with improved clinical outcomes in human metastatic melanoma. Thus, monocytes and macrophages rely on LAIR1 sensing of stromal determinants for fitness and function, with relevance in homeostasis and disease. [Display omitted] • LAIR1 controls the homeostasis of monocytes and interstitial macrophages in lung • The stromal protein, Colec12, is a high-affinity binding partner of LAIR1 • LAIR1 loss in murine myeloid cells augments CSF1R expression and lung metastasis • LAIR1-expressing myeloid cells correlate with good prognosis in metastatic melanoma Keerthivasan et al. show that the homeostasis of monocytes and lung interstitial macrophages is regulated by LAIR1 in vivo. Via high-affinity binding partners Collagen1 and Colec12, LAIR1 maintains crucial signaling pathways associated with survival, proliferation, and differentiation in myeloid cells. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
36. The ITIM-containing receptor LAIR1 is essential for acute myeloid leukaemia development
- Author
-
Xiangshu Xiao, Xin Han, M. James You, Robert H. Collins, Baijun Dong, Cheng Cheng Zhang, Jeffrey W. Tyner, Yuqi Fan, Changhao Cui, Mi Deng, Xunlei Kang, John E. Coligan, Fuchun Xie, and Zhigang Lu
- Subjects
Adult ,Male ,Time Factors ,Cell Survival ,Amino Acid Motifs ,Phosphatase ,Transfection ,CREB ,Article ,Young Adult ,Cell Line, Tumor ,hemic and lymphatic diseases ,Tumor Cells, Cultured ,Animals ,Humans ,Receptors, Immunologic ,Cyclic AMP Response Element-Binding Protein ,Receptor ,Transcription factor ,Aged ,Cell Proliferation ,Mice, Knockout ,biology ,Kinase ,Protein Tyrosine Phosphatase, Non-Receptor Type 6 ,LAIR1 ,Cell Biology ,Middle Aged ,Hedgehog signaling pathway ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,Leukemia, Myeloid, Acute ,Calcium-Calmodulin-Dependent Protein Kinase Type 1 ,Neoplastic Stem Cells ,biology.protein ,Cancer research ,Female ,RNA Interference ,Stem cell ,Signal Transduction - Abstract
Conventional strategies are not particularly successful in the treatment of leukaemia, and identification of signalling pathways crucial to the activity of leukaemia stem cells will provide targets for the development of new therapies. Here we report that certain receptors containing the immunoreceptor tyrosine-based inhibition motif (ITIM) are crucial for the development of acute myeloid leukaemia (AML). Inhibition of expression of the ITIM-containing receptor LAIR1 does not affect normal haematopoiesis but abolishes leukaemia development. LAIR1 induces activation of SHP-1, which acts as a phosphatase-independent signalling adaptor to recruit CAMK1 for activation of downstream CREB in AML cells. The LAIR1-SHP-1-CAMK1-CREB pathway sustains the survival and self-renewal of AML stem cells. Intervention in the signalling initiated by ITIM-containing receptors such as LAIR1 may result in successful treatment of AML.
- Published
- 2015
- Full Text
- View/download PDF
37. Author response: The structure of a LAIR1-containing human antibody reveals a novel mechanism of antigen recognition
- Author
-
Matthew K. Higgins and Fu-Lien Hsieh
- Subjects
biology ,Chemistry ,Mechanism (biology) ,LAIR1 ,biology.protein ,Antibody ,Antigen recognition ,Cell biology - Published
- 2017
- Full Text
- View/download PDF
38. LAIR2-expressing extravillous trophoblasts associate with maternal spiral arterioles undergoing physiologic conversion
- Author
-
T.A. Reinhart, Sandra A. Founds, B. Fallert-Junecko, and W.T. Parks
- Subjects
Adult ,Placenta ,Neovascularization, Physiologic ,Chorionic villus sampling ,In situ hybridization ,Biology ,Andrology ,Pregnancy ,Decidua ,medicine ,Humans ,Regeneration ,Receptors, Immunologic ,Receptor ,In Situ Hybridization ,reproductive and urinary physiology ,medicine.diagnostic_test ,LAIR1 ,Gene Expression Regulation, Developmental ,Obstetrics and Gynecology ,Trophoblast ,Cell Differentiation ,Trophoblasts ,Arterioles ,medicine.anatomical_structure ,Reproductive Medicine ,embryonic structures ,Immunology ,Immunohistochemistry ,Female ,Developmental Biology - Abstract
Leukocyte-associated immunoglobulin-like receptor 2 (LAIR2) was identified on a global gene expression microarray analysis of surplus chorionic villus sampling (CVS) tissues as down-regulated in the first trimester of preeclampsia pregnancies. LAIR2 is the soluble receptor counterpart to LAIR1, an inhibitory receptor found on multiple immune cell subsets. In situ and immunohistochemical studies have previously shown that placental expression of LAIR2 expression is highly restricted, confined to the more distal portions of extravillous trophoblast (EVT) cell columns. This study examines LAIR2 expression in deeper layers of trophoblasts in the placental implantation site, maternal decidua and maternal spiral arterioles. Immunohistochemical staining detected LAIR2 expression on a subset of EVT within the implantation site. This trophoblast included the invasive EVT infiltrating the maternal decidual vessels and the EVT forming the endovascular trophoblastic plugs. More specifically, LAIR2-positive EVT showed a striking predilection for maternal decidual arterioles and the immediately surrounding decidua. Moreover, the appearance of EVT expressing LAIR2 in these areas was contemporaneous with the process of spiral arteriole remodeling. Based on these findings, we suggest that LAIR2-expressing EVT may play an important role in the remodeling of maternal spiral arterioles.
- Published
- 2013
- Full Text
- View/download PDF
39. LAIR2 localizes specifically to sites of extravillous trophoblast invasion
- Author
-
B. Fallert-Junecko, Yvette P. Conley, Sandra A. Founds, T.A. Reinhart, and W.T. Parks
- Subjects
In situ hybridization ,Biology ,Pre-Eclampsia ,Pregnancy ,Fetal membrane ,Placenta ,medicine ,Extracellular ,Humans ,Receptors, Immunologic ,Receptor ,In Situ Hybridization ,Reverse Transcriptase Polymerase Chain Reaction ,LAIR1 ,Obstetrics and Gynecology ,Trophoblast ,Immunohistochemistry ,Molecular biology ,Trophoblasts ,Pregnancy Trimester, First ,medicine.anatomical_structure ,Reproductive Medicine ,embryonic structures ,Immunology ,RNA ,Female ,Chorionic Villi ,Developmental Biology - Abstract
Purpose A global gene expression microarray analysis of surplus chorionic villus sampling (CVS) tissues identified leukocyte-associated immunoglobulin-like receptor 2 (LAIR2) as down-regulated in the first trimester of pregnancies destined for preeclampsia. Neither the localization nor the function of LAIR2 has been examined in the placenta. Localization studies were conducted in placental tissues to determine the precise sites of LAIR2 mRNA production and protein binding. Results Quantitative real time polymerase chain reaction (qRT-PCR) indicated LAIR2 expression in CVS, but none in breast, lymph node, kidney, skin, uterus, or third trimester placentas. In situ hybridization (ISH) revealed a highly restricted LAIR2 localization. LAIR2 mRNA was found only in the more distal portions of trophoblast anchoring cell columns, adjacent to the invading extravillous trophoblast (EVT). Immunohistochemistry (IHC) detected intracellular LAIR2 staining in these same cells. Extracellular staining of this soluble receptor was found in the acellular material between invasive EVT cells distal to the anchoring cell columns. Conclusions ISH and IHC staining for LAIR2 detected specific, highly localized expression at the leading edge of EVT anchoring cell columns in first trimester placentas. This staining likely identifies the site of production for this soluble receptor. Following secretion, the receptor appears to bind extracellular material among the invasive EVT. The precise restriction of this protein only to the sites of EVT invasion strongly suggests that it functions to regulate this invasion. The decreased LAIR2 expression noted in first trimester placentas that ultimately developed preeclampsia further suggests that alterations in LAIR2 may play an etiologic role in preeclampsia.
- Published
- 2010
- Full Text
- View/download PDF
40. NK Cell Autoreactivity and Autoimmune Diseases
- Author
-
Maria Raffaella Zocchi and Alessandro Poggi
- Subjects
lcsh:Immunologic diseases. Allergy ,Innate immune system ,DNAM1 ,Janus kinase 3 ,Innate lymphoid cell ,Immunology ,autoimmunity ,Review Article ,NK cells ,Biology ,Acquired immune system ,NKG2D ,Interleukin 21 ,LAIR1 ,regulatory NK cells ,Interleukin 12 ,Immunology and Allergy ,lcsh:RC581-607 ,mesenchymal stromal cells ,Tissue homeostasis ,autoreactivity - Abstract
Increasing evidences have pointed out the relevance of natural killer (NK) cells in organ-specific and systemic autoimmune diseases. NK cells bear a plethora of activating and inhibiting receptors that can play a role in regulating reactivity with autologous cells. The activating receptors recognize natural ligands up-regulated on virus-infected or stressed or neoplastic cells. Of note, several autoimmune diseases are thought to be linked to viral infections as one of the first event in inducing autoimmunity. Also, it is conceivable that autoimmunity can be triggered when a dysregulation of innate immunity occurs, activating T and B lymphocytes to react with self-components. This would imply that NK cells can play a regulatory role during adaptive immunity; indeed, innate lymphoid cells (ILCs), comprising the classical CD56(+) NK cells, have a role in maintaining or alternating tissue homeostasis secreting protective and/or pro-inflammatory cytokines. In addition, NK cells display activating receptors involved in natural cytotoxicity and the activating isoforms of receptors for HLA class I that can interact with healthy host cells and induce damage without any evidence of viral infection or neoplastic-induced alteration. In this context, the interrelationship among ILC, extracellular-matrix components, and mesenchymal stromal cells can be considered a key point for the control of homeostasis. Herein, we summarize evidences for a role of NK cells in autoimmune diseases and will give a point of view of the interplay between NK cells and self-cells in triggering autoimmunity.
- Published
- 2014
41. Concerted effect of lymphopenia, viraemia and T-cell activation on Fas expression of peripheral B cells in HIV-1-infected patients
- Author
-
Bence Rethi, Stefano Sammicheli, Sylvie Amu, Simone Pensieroso, Pham Hong Thang, Bo Hejdeman, Francesca Chiodi, and Danika Schepis
- Subjects
Adult ,CD4-Positive T-Lymphocytes ,Male ,medicine.medical_specialty ,T cell ,Immunology ,Programmed Cell Death 1 Receptor ,Apoptosis ,HIV Infections ,Lymphocyte Activation ,Flow cytometry ,03 medical and health sciences ,0302 clinical medicine ,Downregulation and upregulation ,Internal medicine ,Lymphopenia ,Immunology and Allergy ,Medicine ,Humans ,030212 general & internal medicine ,Viremia ,fas Receptor ,Receptors, Immunologic ,Receptor ,030304 developmental biology ,0303 health sciences ,B-Lymphocytes ,medicine.diagnostic_test ,business.industry ,LAIR1 ,Middle Aged ,Flow Cytometry ,3. Good health ,Peripheral ,CD4 Lymphocyte Count ,Infectious Diseases ,Endocrinology ,medicine.anatomical_structure ,HIV-1 ,Female ,business ,Immunologic Memory ,Homeostasis - Abstract
Objective Decreased memory B-cell maintenance during HIV-1 infection has been associated with the viraemia-induced accumulation of activated memory B cells, sensitive to Fas-mediated apoptosis. We aimed at clarifying whether other B-cell subsets might also be affected by an increased Fas expression in HIV-1-infected patients, and we studied the possible contribution of viraemia, lymphopenia or T-cell activation in Fas upregulation on B cells. We analysed whether Fas upregulation might have collaborative effects with the dysregulation of other B-cell modulatory molecules, leukocyte-associated immunoglobulin-like receptor 1 (LAIR1) and programmed cell death protein 1 (PD-1), on B-cell homeostasis. Design Fas, LAIR1 and PD-1 were analysed on B-cell subpopulations in HIV-1-infected patients who were treatment naive, nonlymphopenic; antiretroviral therapy (ART)-treated, nonlymphopenic; or ART-treated, lymphopenic or in noninfected controls. Methods Flow cytometry was used to study B-cell subsets and Milliplex for serum cytokines. Results Fas expression increased on all B-cell subpopulations of viraemic or lymphopenic individuals. The decreased ratio of resting memory B cells and their increased Fas expression were not normalized by ART. Cytokines associated with T-cell activation might influence Fas expression on the naive and transitional B cells. LAIR1 expression decreased in all HIV-1-infected patients, but only on memory B cells, whereas PD-1 increased on resting memory B cells in viraemic patients. Conclusion Fas is regulated by the concerted action of viraemia, lymphopenia and T-cell activation during HIV-1 infection, and Fas expression is altered on all peripheral B-cell subsets. Resting memory B-cell homeostasis shows the highest sensitivity to HIV-1-induced perturbations.
- Published
- 2012
42. NK cell autoreactivity and autoimmune diseases.
- Author
-
Poggi A and Zocchi MR
- Abstract
Increasing evidences have pointed out the relevance of natural killer (NK) cells in organ-specific and systemic autoimmune diseases. NK cells bear a plethora of activating and inhibiting receptors that can play a role in regulating reactivity with autologous cells. The activating receptors recognize natural ligands up-regulated on virus-infected or stressed or neoplastic cells. Of note, several autoimmune diseases are thought to be linked to viral infections as one of the first event in inducing autoimmunity. Also, it is conceivable that autoimmunity can be triggered when a dysregulation of innate immunity occurs, activating T and B lymphocytes to react with self-components. This would imply that NK cells can play a regulatory role during adaptive immunity; indeed, innate lymphoid cells (ILCs), comprising the classical CD56(+) NK cells, have a role in maintaining or alternating tissue homeostasis secreting protective and/or pro-inflammatory cytokines. In addition, NK cells display activating receptors involved in natural cytotoxicity and the activating isoforms of receptors for HLA class I that can interact with healthy host cells and induce damage without any evidence of viral infection or neoplastic-induced alteration. In this context, the interrelationship among ILC, extracellular-matrix components, and mesenchymal stromal cells can be considered a key point for the control of homeostasis. Herein, we summarize evidences for a role of NK cells in autoimmune diseases and will give a point of view of the interplay between NK cells and self-cells in triggering autoimmunity.
- Published
- 2014
- Full Text
- View/download PDF
43. [Untitled]
- Subjects
0301 basic medicine ,030102 biochemistry & molecular biology ,General Immunology and Microbiology ,General Neuroscience ,LAIR1 ,General Medicine ,Complementarity determining region ,Biology ,Virology ,Primary and secondary antibodies ,General Biochemistry, Genetics and Molecular Biology ,Insert (molecular biology) ,3. Good health ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,Immune system ,Antigen ,biology.protein ,Antibody ,Pathogen - Abstract
When bacteria, viruses or parasites invade the human body, the immune system responds by producing proteins called antibodies. Antibodies recognize and bind to molecules (known as antigens) on the surface of the invaders. This binding can either neutralize the invader directly or trigger signals that cause other parts of the immune system to destroy it. Our blood contains a huge range of different antibody molecules that each bind to a different antigen. This is despite most human antibodies having the same basic shape and structure. Six loops, known as complementarity determining regions (CDRs), emerge from the surface of the antibody to form the surface that recognizes the antigen. However, variations in the structure of the loops alter this surface enough to allow different antibodies to recognize completely different molecules. In 2016, a new class of antibodies was identified. Unlike previously identified antibodies, these molecules had an entire human protein, called LAIR1, inserted into one of their CDR loops. Members of this group of antibodies bind to a molecule, known as a RIFIN, that is found on the surface of human red blood cells that are infected with the parasite that causes malaria. How do LAIR1-containing antibodies bind to their RIFIN targets? Hsieh and Higgins investigated this question by using a technique called X-ray crystallography to determine the structure of the antibody. This revealed that instead of binding directly to an antigen, all of the six CDR loops in the LAIR1-containing antibody bind to the LAIR1 insert. By doing so, LAIR1 is oriented in a manner that enables it to bind to the RIFIN molecule from the parasite. This is the first known example of an antibody that recruits another protein to bind to an antigen rather than binding directly to the pathogen itself. A future challenge will be to see if other antibodies exist that use this mechanism and whether it can be employed to design new therapeutic antibodies.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.