1
|
Su MT, Kumata S, Endo S, Okada Y, Takai T. LILRB4 promotes tumor metastasis by regulating MDSCs and inhibiting miR-1 family miRNAs. Oncoimmunology 2022; 11:2060907. [PMID: 35402083 PMCID: PMC8986222 DOI: 10.1080/2162402x.2022.2060907] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a population of immune suppressive cells that are involved in tumor-associated immunosuppression, and dominate tumor progression and metastasis. In this study, we report that the leukocyte immunoglobulin-like receptor subfamily B member 4 (LILRB4, murine ortholog gp49B) orchestrates the polarization of MDSCs to exhibit pro-tumor phenotypes. We found that gp49B deficiency inhibited tumor metastases of cancer cells, and reduced tumor-infiltration of monocytic MDSCs (M-MDSCs) in tumor-bearing mice. Gp49B−/− MDSCs inhibited pro-tumor immune responses, such as activation of Treg cells, promotion of cancer cell migration, and stimulation of tumor angiogenesis. Treatment of wild-type tumor-bearing mice with gp49B−/− M-MDSCs reduced cancer metastasis. Furthermore, gp49B knockout affected plasma exosome composition in terms of increased miR-1 family microRNAs (miRNAs) expression, which correlates with the upregulation of gp49B−/− MDSC-derived anti-tumor miRNAs. Collectively, our findings reveal that LILRB4/gp49B promotes MDSC-mediated tumor metastasis by regulating the M2-polarization of MDSCs and suppressing the secretion of miR-1 family miRNAs, which facilitate tumor migration and invasion. Abbreviations CTLA-4: cytotoxic T-lymphocyte-associated protein-4; FBS: fetal bovine serum; G-MDSCs: granulocytic-MDSCs; GP49B: glycoprotein 49B; HE: hematoxylin-eosin; ICI: immune checkpoint inhibitor; ITIM: immunoreceptor tyrosine-based inhibition motif; LILRB4: leukocyte immunoglobulin-like receptor B4; M-CSF: macrophage colony stimulating factor; MDSC: myeloid-derived suppressor cell; M-MDSC: monocytic MDSC; MMP-9: metallopeptidase-9; mAb: monoclonal antibody; PBS: phosphate-buffered saline; PCR: polymerase chain reaction; PD-1: programmed death-1; PD-L1: programmed death ligand-1; PMN-MDSC: polymorphonuclear-MDSC; qRT-PCR: quantitative reverse transcription PCR; TAM: tumor associated macrophage; TME: tumor microenvironment; TMM: trimmed mean of M value; VEGFA: vascular endothelial growth factor A
Collapse
Affiliation(s)
- Mei-Tzu Su
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Sakiko Kumata
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Shota Endo
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Yoshinori Okada
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Toshiyuki Takai
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
2
|
Abstract
Leukocyte immunoglobulin-like receptor B4 (LILRB4) is an inhibitory receptor in the LILR family mainly expressed on normal and malignant human cells of myeloid origin. By binding to ligands, LILRB4 is activated and subsequently recruits adaptors to cytoplasmic immunoreceptor tyrosine inhibitory motifs to initiate different signaling cascades, thus playing an important role in physiological and pathological conditions, including autoimmune diseases, microbial infections, and cancers. In normal myeloid cells, LILRB4 regulates intrinsic cell activation and differentiation. In disease-associated or malignant myeloid cells, LILRB4 is significantly correlated with disease severity or patient survival and suppresses T cells, thereby participating in the pathogenesis of various diseases. In summary, LILRB4 functions as an immune checkpoint on myeloid cells and may be a promising therapeutic target for various human immune diseases, especially for cancer immunotherapy.
Collapse
|
3
|
Peng H, Zou Y. The leukocyte immunoglobulin-like receptor gp49B1, coexpressed with c-Kit, modulates hematopoiesis and B cell leukemia development. Biochem Biophys Res Commun 2021; 565:72-78. [PMID: 34098314 DOI: 10.1016/j.bbrc.2021.05.090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 11/26/2022]
Abstract
A better understanding of cell-intrinsic factors involved in regulating stem cells and cancer cells will help advance stem cell applications and cancer cell treatment. Previously, we showed that leukocyte immunoglobulin-like receptor B2 (LILRB2) and its mouse ortholog, paired immunoglobulin-like receptor B (PIRB), promote blood stem cell and leukemia development. Another unique mouse paralog to PIRB called gp49B1 was also discovered. However, the roles of gp49B1 in hematopoietic stem cells and leukemia development are largely unknown. Here, we found that gp49B1 is expressed on LSK cells of mouse neonatal hematopoietic organs and is positively correlated with c-Kit expression. However, in noncompetitive and competitive repopulation assays, neonatal splenic gp49B1-positive and c-Kit-highly expressed LSK cells exhibited poor engraftment potential and lymphoid lineage bias. Moreover, in a mouse N-Myc-induced precursor B-acute lymphoblastic leukemia (pre-B ALL) model, we found that gp49B1 deficiency or low levels of c-Kit led to a delay in leukemia development. Together, our results suggest that gp49B1 expressed on hematopoietic progenitor cells supports hematopoietic and leukemia development.
Collapse
Affiliation(s)
- Huiyun Peng
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha City, Hunan Province, China.
| | - Yizhou Zou
- Department of Immunology, School of Basic Medical Science, Central South University, Changsha City, Hunan Province, China.
| |
Collapse
|
4
|
Deng M, Chen H, Liu X, Huang R, He Y, Yoo B, Xie J, John S, Zhang N, An Z, Zhang CC. Leukocyte immunoglobulin-like receptor subfamily B: therapeutic targets in cancer. Antib Ther 2021; 4:16-33. [PMID: 33928233 PMCID: PMC7944505 DOI: 10.1093/abt/tbab002] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
Inhibitory leukocyte immunoglobulin-like receptors (LILRBs 1–5) transduce signals via intracellular immunoreceptor tyrosine-based inhibitory motifs that recruit phosphatases to negatively regulate immune activation. The activation of LILRB signaling in immune cells may contribute to immune evasion. In addition, the expression and signaling of LILRBs in cancer cells especially in certain hematologic malignant cells directly support cancer development. Certain LILRBs thus have dual roles in cancer biology—as immune checkpoint molecules and tumor-supporting factors. Here, we review the expression, ligands, signaling, and functions of LILRBs, as well as therapeutic development targeting them. LILRBs may represent attractive targets for cancer treatment, and antagonizing LILRB signaling may prove to be effective anti-cancer strategies.
Collapse
Affiliation(s)
- Mi Deng
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Heyu Chen
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaoye Liu
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ryan Huang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yubo He
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Byounggyu Yoo
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jingjing Xie
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Samuel John
- Department of Pediatrics, Pediatric Hematology-Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Houston Health Science Center, Houston, TX 77030, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Houston Health Science Center, Houston, TX 77030, USA
| | - Cheng Cheng Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
5
|
Zhou H, Li N, Yuan Y, Jin YG, Wu Q, Yan L, Bian ZY, Deng W, Shen DF, Li H, Tang QZ. Leukocyte immunoglobulin-like receptor B4 protects against cardiac hypertrophy via SHP-2-dependent inhibition of the NF-κB pathway. J Mol Med (Berl) 2020; 98:691-705. [PMID: 32280997 DOI: 10.1007/s00109-020-01896-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/08/2020] [Accepted: 03/11/2020] [Indexed: 10/24/2022]
Abstract
Cardiac hypertrophy is a complex pathological process, and the molecular mechanisms underlying hypertrophic remodeling have not been clearly elucidated. Leukocyte immunoglobulin-like receptor B4 (lilrb4) is an inhibitory transmembrane protein that is necessary for the regulation of various cellular signaling pathways. To investigate whether lilrb4 plays a role in cardiac hypertrophy, we performed aortic banding in lilrb4 knockout mice, lilrb4 cardiac-specific transgenic mice, and their wild-type littermates. Cardiac hypertrophy was evaluated by echocardiographic, hemodynamic, pathological, and molecular analyses. We found that lilrb4 was expressed both in myocardial tissue and on cultured cardiomyocytes under basal conditions, but the expression was obviously decreased in mouse hearts following aortic banding and in cardiomyocytes treated with angiotensin II. Lilrb4 disruption aggravated cardiac hypertrophy, fibrosis, and dysfunction in response to pressure overload. Conversely, the cardiac overexpression of lilrb4 led to the opposite effects. Moreover, lilrb4 overexpression inhibited angiotensin II-induced cardiomyocyte hypertrophy in vitro. Mechanistically, we determined that the cardioprotective effect of lilrb4 was mediated through an interaction with SHP-2, the preservation of phosphorylated SHP-2, and the inhibition of the NF-κB pathway. In addition, SHP-2 knockdown in cardiomyocytes eliminated the inhibitory effects of lilrb4 on angiotensin II-induced hypertrophy and NF-κB activation. Our results suggest that lilrb4 protects against pathological cardiac hypertrophy via the SHP-2-dependent inhibition of the NF-κB pathway and may act as a potential therapeutic target for cardiac hypertrophy. KEY MESSAGES: Lilrb4 expression is decreased by hypertrophic stimuli. Lilrb4 protects against pathological cardiac hypertrophy. Lilrb4 interacts with SHP-2 and inhibits NF-κB pathway.
Collapse
Affiliation(s)
- Heng Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Ning Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Yuan Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Ya-Ge Jin
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Qingqing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Ling Yan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Zhou-Yan Bian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Wei Deng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Di-Fei Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China.,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China.,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, People's Republic of China. .,Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People's Republic of China. .,Cardiovascular Research Institute, Wuhan University, Wuhan, 430060, People's Republic of China.
| |
Collapse
|
6
|
Lu Y, Jiang Z, Dai H, Miao R, Shu J, Gu H, Liu X, Huang Z, Yang G, Chen AF, Yuan H, Li Y, Cai J. Hepatic leukocyte immunoglobulin-like receptor B4 (LILRB4) attenuates nonalcoholic fatty liver disease via SHP1-TRAF6 pathway. Hepatology 2018; 67:1303-1319. [PMID: 29091299 PMCID: PMC5900726 DOI: 10.1002/hep.29633] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 09/15/2017] [Accepted: 10/30/2017] [Indexed: 12/27/2022]
Abstract
UNLABELLED Nonalcoholic fatty liver disease (NAFLD) is an increasingly prevalent liver pathology characterized by hepatic steatosis and commonly accompanied by systematic inflammation and metabolic disorder. Despite an accumulating number of studies, no pharmacological strategy is available to treat this condition in the clinic. In this study, we applied extensive gain- and loss-of-function approaches to identify the key immune factor leukocyte immunoglobulin-like receptor B4 (LILRB4) as a negative regulator of NAFLD. The hepatocyte-specific knockout of LILRB4 (LILRB4-HKO) exacerbated high-fat diet-induced insulin resistance, glucose metabolic imbalance, hepatic lipid accumulation, and systematic inflammation in mice, whereas LILRB4 overexpression in hepatocytes showed a completely opposite phenotype relative to that of LILRB4-HKO mice when compared with their corresponding controls. Further investigations of molecular mechanisms demonstrated that LILRB4 recruits SHP1 to inhibit TRAF6 ubiquitination and subsequent inactivation of nuclear factor kappa B and mitogen-activated protein kinase cascades. From a therapeutic perspective, the overexpression of LILRB4 in a genetic model of NAFLD, ob/ob mice, largely reversed the inherent hepatic steatosis, inflammation, and metabolic disorder. CONCLUSION Targeting hepatic LILRB4 to improve its expression or activation represents a promising strategy for the treatment of NAFLD as well as related liver and metabolic diseases. (Hepatology 2018;67:1303-1319).
Collapse
Affiliation(s)
- Yao Lu
- Center of Clinical Pharmacology, Third Xiangya HospitalCentral South UniversityChangshaChina,Department of Clinical PharmacologySt. Thomas' Hospital, KCLLondonUK
| | - Zhou Jiang
- Institute of Model Animal of Wuhan UniversityWuhanChina
| | - Haijiang Dai
- Center of Clinical Pharmacology, Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Rujia Miao
- Health Management Center, Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Jingxian Shu
- Center of Clinical Pharmacology, Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Haotian Gu
- Department of Clinical PharmacologySt. Thomas' Hospital, KCLLondonUK
| | - Xing Liu
- Center of Clinical Pharmacology, Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Zhijun Huang
- Center of Clinical Pharmacology, Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Guoping Yang
- Center of Clinical Pharmacology, Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Alex F. Chen
- Center of Clinical Pharmacology, Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Hong Yuan
- Center of Clinical Pharmacology, Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Ying Li
- Health Management Center, Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Jingjing Cai
- Department of Cardiology, Third Xiangya HospitalCentral South UniversityChangshaChina
| |
Collapse
|
7
|
Pauls SD, Marshall AJ. Regulation of immune cell signaling by SHIP1: A phosphatase, scaffold protein, and potential therapeutic target. Eur J Immunol 2017; 47:932-945. [PMID: 28480512 DOI: 10.1002/eji.201646795] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/06/2017] [Accepted: 05/03/2017] [Indexed: 02/06/2023]
Abstract
The phosphoinositide phosphatase SHIP is a critical regulator of immune cell activation. Despite considerable study, the mechanisms controlling SHIP activity to ensure balanced cell activation remain incompletely understood. SHIP dampens BCR signaling in part through its association with the inhibitory coreceptor Fc gamma receptor IIB, and serves as an effector for other inhibitory receptors in various immune cell types. The established paradigm emphasizes SHIP's inhibitory receptor-dependent function in regulating phosphoinositide 3-kinase signaling by dephosphorylating the phosphoinositide PI(3,4,5)P3 ; however, substantial evidence indicates that SHIP can be activated independently of inhibitory receptors and can function as an intrinsic brake on activation signaling. Here, we integrate historical and recent reports addressing the regulation and function of SHIP in immune cells, which together indicate that SHIP acts as a multifunctional protein controlled by multiple regulatory inputs, and influences downstream signaling via both phosphatase-dependent and -independent means. We further summarize accumulated evidence regarding the functions of SHIP in B cells, T cells, NK cells, dendritic cells, mast cells, and macrophages, and data suggesting defective expression or activity of SHIP in autoimmune and malignant disorders. Lastly, we discuss the biological activities, therapeutic promise, and limitations of small molecule modulators of SHIP enzymatic activity.
Collapse
Affiliation(s)
- Samantha D Pauls
- Department of Immunology, University of Manitoba, Winnipeg, Canada
| | - Aaron J Marshall
- Department of Immunology, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
8
|
Kang X, Kim J, Deng M, John S, Chen H, Wu G, Phan H, Zhang CC. Inhibitory leukocyte immunoglobulin-like receptors: Immune checkpoint proteins and tumor sustaining factors. Cell Cycle 2016; 15:25-40. [PMID: 26636629 PMCID: PMC4825776 DOI: 10.1080/15384101.2015.1121324] [Citation(s) in RCA: 143] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Inhibitory leukocyte immunoglobulin-like receptors (LILRBs 1-5) transduce signals via intracellular immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that recruit protein tyrosine phosphatase non-receptor type 6 (PTPN6 or SHP-1), protein tyrosine phosphatase non-receptor type 11 (PTPN11 or SHP-2), or Src homology 2 domain-containing inositol phosphatase (SHIP), leading to negative regulation of immune cell activation. Certain of these receptors also play regulatory roles in neuronal activity and osteoclast development. The activation of LILRBs on immune cells by their ligands may contribute to immune evasion by tumors. Recent studies found that several members of LILRB family are expressed by tumor cells, notably hematopoietic cancer cells, and may directly regulate cancer development and relapse as well as the activity of cancer stem cells. LILRBs thus have dual concordant roles in tumor biology - as immune checkpoint molecules and as tumor-sustaining factors. Importantly, the study of knockout mice indicated that LILRBs do not affect hematopoiesis and normal development. Therefore LILRBs may represent ideal targets for tumor treatment. This review aims to summarize current knowledge on expression patterns, ligands, signaling, and functions of LILRB family members in the context of cancer development.
Collapse
Affiliation(s)
- Xunlei Kang
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Jaehyup Kim
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Mi Deng
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Samuel John
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Heyu Chen
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Guojin Wu
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Hiep Phan
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| | - Cheng Cheng Zhang
- a Department of Physiology , University of Texas Southwestern Medical Center , Dallas , TX , USA
| |
Collapse
|
9
|
Abstract
The immune system has evolved to defend the organism against an almost infinite number of pathogens in a locally confined and antigen-specific manner while at the same time preserving tolerance to harmless antigens and self. Regulatory T (Treg) cells essentially contribute to an immunoregulatory network preventing excessive immune responses and immunopathology. There is emerging evidence that Treg cells not only operate in secondary lymphoid tissue but also regulate immune responses directly at the site of inflammation. Hence, the classification of Treg cells might need to be further extended by Treg cell subsets that are functionally and phenotypically polarized by their residency. In this review, we discuss recent findings on these tissue-resident Treg cell subsets and how these cells may operate in a tissue- and context-dependent manner.
Collapse
|
10
|
Fukao S, Haniuda K, Nojima T, Takai T, Kitamura D. gp49B-mediated negative regulation of antibody production by memory and marginal zone B cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:635-44. [PMID: 24935931 DOI: 10.4049/jimmunol.1302772] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The rapid Ab responses observed after primary and secondary immunizations are mainly derived from marginal zone (MZ) and memory B cells, respectively, but it is largely unknown how these responses are negatively regulated. Several inhibitory receptors have been identified and their roles have been studied, but mainly on follicular B cells and much less so on MZ B, and never on memory B cells. gp49B is an Ig superfamily member that contains two ITIMs in its cytoplasmic tail, and it has been shown to negatively regulate mast cell, macrophage, and NK cell responses. In this study, we demonstrate that gp49B is preferentially expressed on memory and MZ B cells. We show that gp49B(-/-) mice produce more IgM after a primary immunization and more IgM and IgG1 after a secondary immunization than gp49B(+/+) mice in T cell-dependent immune responses. Memory and MZ B cells from gp49B(-/-) mice also produce more Abs upon in vitro stimulation with CD40 than those from gp49B(+/+) mice. The in vitro IgM production by MZ B cells from gp49B(+/+), but not gp49B(-/-), mice is suppressed by interaction with a putative gp49B ligand, the integrin αvβ3 heterodimer. In addition, gp49B(-/-) mice exhibited exaggerated IgE production in the memory recall response. These results suggest that plasma cell development from memory and MZ B cells, as well as subsequent Ab production, are suppressed via gp49B. In memory B cells, this suppression also prevents excessive IgE production, thus curtailing allergic diseases.
Collapse
Affiliation(s)
- Saori Fukao
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba 278-0022, Japan; and
| | - Kei Haniuda
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba 278-0022, Japan; and
| | - Takuya Nojima
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba 278-0022, Japan; and
| | - Toshiyuki Takai
- Department of Experimental Immunology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Miyagi 980-8575, Japan
| | - Daisuke Kitamura
- Division of Molecular Biology, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba 278-0022, Japan; and
| |
Collapse
|
11
|
Nakata K, Suzuki Y, Inoue T, Ra C, Yakura H, Mizuno K. Deficiency of SHP1 leads to sustained and increased ERK activation in mast cells, thereby inhibiting IL-3-dependent proliferation and cell death. Mol Immunol 2010; 48:472-80. [PMID: 21044800 DOI: 10.1016/j.molimm.2010.10.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 10/01/2010] [Accepted: 10/06/2010] [Indexed: 10/18/2022]
Abstract
SHP-1 plays an important role for the regulation of signaling from various hematopoietic cell receptors. In this study, we examined IL-3-induced cell proliferation and IL-3 depletion-induced apoptosis in bone marrow-derived mast cells (BMMC) established from motheaten (me) that lack SHP-1 expression, viable motheaten (me(v)) expressing phosphatase-deficient SHP-1, and wild-type (WT) mice. When BMMC were stimulated with IL-3, increased ERK activation was evident in resting state and sustained in me-BMMC relative to WT-BMMC. ERK is known to be involved in the regulation of cell proliferation and apoptosis in some cells. In accordance with sustained ERK activation, apoptosis was decreased in me- and me(v)-BMMC compared with WT-BMMC. In contrast to the predicted role of ERK as a pro-survival molecule, IL-3-induced cell proliferation was much lower in me- and me(v)-BMMC than WT-BMMC. Stimulation with lower concentration of IL-3 or addition of PD98059, a MEK inhibitor, to the culture resulted in the suppression of decreased apoptosis and cell proliferation in me- and me(v)-BMMC. Collectively, these results suggest that SHP-1 positively regulates IL-3-dependent mast cell proliferation and apoptosis by inhibiting ERK activity through its phosphatase activity. Furthermore, our results indicate that ERK would act as a negative regulator for cell proliferation and induce apoptosis when its activity is highly increased.
Collapse
Affiliation(s)
- Kazuko Nakata
- Department of Immunology and Signal Transduction, Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Science, Fuchu, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
12
|
Inoue T, Suzuki Y, Mizuno K, Nakata K, Yoshimaru T, Ra C. SHP-1 exhibits a pro-apoptotic function in antigen-stimulated mast cells: Positive regulation of mitochondrial death pathways and negative regulation of survival signaling pathways. Mol Immunol 2009; 47:222-32. [DOI: 10.1016/j.molimm.2009.09.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 09/24/2009] [Indexed: 11/16/2022]
|
13
|
Kasai S, Inui M, Nakamura K, Kakizaki Y, Endo S, Nakamura A, Ito S, Takai T. A novel regulatory role of gp49B on dendritic cells in T-cell priming. Eur J Immunol 2008; 38:2426-37. [PMID: 18792399 DOI: 10.1002/eji.200737550] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Dendritic cells (DC) play pivotal roles in the induction and regulation of both innate and acquired immunity. DC express several cell-surface immune inhibitory receptors. However, little is known about their potential immunoregulatory functions in the context of T-cell activation. Here we report that murine gp49B, a member of the immunoglobulin superfamily, harboring immunoreceptor tyrosine-based inhibitory motifs, is expressed on DC and downregulates cellular activity to prevent the excessive activation of T cells in vitro and in vivo. Bone marrow-derived DC (BMDC) from newly generated gp49B-deficient (gp49B(-/-)) mice induced enhanced proliferation and IL-2 release of antigen-specific CD4(+) and CD8(+) T cells compared with BMDC from wild-type mice, in a cell-cell contact manner. The enhanced proliferation by gp49B(-/-) BMDC was also observed in allogeneic CD4(+) and CD8(+) T cells. Moreover, the transfer of allogeneic BALB/c splenocytes into C57BL/6 gp49B(-/-) mice induced severe acute graft-versus-host disease with an augmented upregulation of CD86 on CD11c(+) splenic gp49B(-/-) DC, while transfer of C57BL/6 gp49B(-/-) splenocytes into BALB/c mice did not, suggesting the exacerbation of the disease was due, at least in part, to augmented activation of recipient gp49B(-/-) DC. These findings demonstrate a novel regulatory role of gp49B in the function of DC.
Collapse
Affiliation(s)
- Satoshi Kasai
- Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Dissen E, Fossum S, Hoelsbrekken SE, Saether PC. NK cell receptors in rodents and cattle. Semin Immunol 2008; 20:369-75. [PMID: 18977671 DOI: 10.1016/j.smim.2008.09.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2008] [Accepted: 09/04/2008] [Indexed: 11/17/2022]
Abstract
Natural killer (NK) cells discriminate between normal syngeneic cells and infected, neoplastic or MHC-disparate allogeneic cells. The reactivity of NK cells appears to be regulated by a balance between activating receptors that recognize non-self or altered self, and inhibitory receptors recognizing normal, self-encoded MHC class I molecules. Subfamilies of NK receptors undergo rapid evolution, and appear to co-evolve with the MHC. We here review present views on the evolution and function of NK cell receptors, with an emphasis on knowledge gained in cattle and rodents.
Collapse
Affiliation(s)
- Erik Dissen
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, PO Box 1105 Blindern, N-0317 Oslo, Norway.
| | | | | | | |
Collapse
|
15
|
Nakata K, Yoshimaru T, Suzuki Y, Inoue T, Ra C, Yakura H, Mizuno K. Positive and negative regulation of high affinity IgE receptor signaling by Src homology region 2 domain-containing phosphatase 1. THE JOURNAL OF IMMUNOLOGY 2008; 181:5414-24. [PMID: 18832698 DOI: 10.4049/jimmunol.181.8.5414] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Src homology region 2 domain-containing phosphatase 1 (SHP-1), a cytoplasmic protein tyrosine phosphatase, plays an important role for the regulation of signaling from various hematopoietic cell receptors. Although SHP-1 is shown to be a negative signal modulator in mast cells, its precise molecular mechanisms are not well defined. To elucidate how SHP-1 regulates mast cell signaling, we established bone marrow-derived mast cells from SHP-1-deficient motheaten and wild-type mice and analyzed downstream signals induced by cross-linking of high affinity IgE receptor, Fc epsilonRI. Upon Fc epsilonRI ligation, motheaten-derived bone marrow-derived mast cells showed enhanced tyrosine phosphorylation of Src homology region 2 domain-containing leukocyte protein of 76 kDa (SLP-76) and linker for activation of T cells, activation of mitogen-activated protein kinases and gene transcription and production of cytokine. Because the activity of Syk, responsible for the phosphorylation of SLP-76 and linker for activation of T cells, is comparable irrespective of SHP-1, both molecules might be substrates of SHP-1 in mast cells. Interestingly, the absence of SHP-1 expression disrupted the association between SLP-76 and phospholipase Cgamma, which resulted in the decreased phospholipase Cgamma phosphorylation, calcium mobilization, and degranulation. Collectively, these results suggest that SHP-1 regulates Fc epsilonRI-induced downstream signaling events both negatively and positively by functioning as a protein tyrosine phosphatase and as an adaptor protein contributing to the formation of signaling complex, respectively.
Collapse
Affiliation(s)
- Kazuko Nakata
- Department of Immunology and Signal Transduction, Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Science, Fuchu, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
16
|
Mori Y, Tsuji S, Inui M, Sakamoto Y, Endo S, Ito Y, Fujimura S, Koga T, Nakamura A, Takayanagi H, Itoi E, Takai T. Inhibitory Immunoglobulin-Like Receptors LILRB and PIR-B Negatively Regulate Osteoclast Development. THE JOURNAL OF IMMUNOLOGY 2008; 181:4742-51. [DOI: 10.4049/jimmunol.181.7.4742] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
17
|
An essential function for beta-arrestin 2 in the inhibitory signaling of natural killer cells. Nat Immunol 2008; 9:898-907. [PMID: 18604210 DOI: 10.1038/ni.1635] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Accepted: 06/17/2008] [Indexed: 11/09/2022]
Abstract
The inhibitory signaling of natural killer (NK) cells is crucial in the regulation of innate immune responses. Here we show that the association of KIR2DL1, an inhibitory receptor of NK cells, with beta-arrestin 2 mediated recruitment of the tyrosine phosphatases SHP-1 and SHP-2 to KIR2DL1 and facilitated 'downstream' inhibitory signaling. Consequently, the cytotoxicity of NK cells was higher in beta-arrestin 2-deficient mice but was inhibited in beta-arrestin 2-transgenic mice. Moreover, beta-arrestin 2-deficient mice were less susceptible than wild-type mice to mouse cytomegalovirus infection, an effect that was abolished by depletion of NK cells. Our findings identify a previously unknown mechanism by which the inhibitory signaling in NK cells is regulated.
Collapse
|
18
|
Ozawa T, Nakata K, Mizuno K, Yakura H. Negative autoregulation of Src homology region 2-domain-containing phosphatase-1 in rat basophilic leukemia-2H3 cells. Int Immunol 2007; 19:1049-61. [PMID: 17675340 DOI: 10.1093/intimm/dxm070] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Src homology region 2-domain-containing phosphatase-1 (SHP-1) plays an important role in the regulation of signaling from various receptors in hematopoietic cells. In mast cells, SHP-1 has been shown to negatively regulate the initial signaling triggered by high-affinity receptor for IgE (FcepsilonRI) and positively regulate downstream outputs. To clarify the molecular mechanisms of SHP-1 in mast cells, we determined substrates for SHP-1 by using the substrate-trapping approach. When phosphatase-inactive SHP-1 was over-expressed in rat basophilic leukemia (RBL)-2H3 cells, tyrosine phosphorylation of a 68-kDa protein was enhanced before and after FcepsilonRI aggregation. Immunoprecipitation and western blot analyses revealed that this protein is SHP-1, either endogenous or ectopically expressed. FcepsilonRI-induced activation of Lyn and Syk was comparable between cells expressing wild-type (wt) and phosphatase-inactive SHP-1. In vitro phosphatase assay and combined transfection, immunoprecipitation and immunoblot analyses showed that tyrosine 536 of SHP-1 was potent phosphorylation site and that SHP-1 could dephosphorylate this site that had been phosphorylated by Lyn. Furthermore, the phosphatase activity of SHP-1 immunoprecipitated from cells expressing a phosphatase-inactive SHP-1 was increased compared with that from vector-transfected or wt SHP-1-expressing cells. Finally, expression of phosphatase-inactive SHP-1 resulted in decreased activation of mitogen-activated protein kinases and suppressed transcription of cytokine genes, whereas wt SHP-1 enhanced these processes. Taken collectively, these results suggest that SHP-1 may be a physiological substrate of SHP-1 in RBL-2H3 cells and that dephosphorylation of SHP-1 leads to a decrease in its catalytic activity and an enhancement of downstream signaling. A negative autoregulatory circuit of SHP-1 may contribute to mast cell regulation.
Collapse
Affiliation(s)
- Tomoko Ozawa
- Department of Immunology and Signal Transduction, Tokyo Metropolitan Institute for Neuroscience, Tokyo Metropolitan Organization for Medical Science, 2-6 Musashidai, Fuchu, Tokyo 183-8526, Japan
| | | | | | | |
Collapse
|
19
|
Katz HR. Inhibition of pathologic inflammation by leukocyte Ig-like receptor B4 and related inhibitory receptors. Immunol Rev 2007; 217:222-30. [PMID: 17498062 DOI: 10.1111/j.1600-065x.2007.00522.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Leukocyte immunoglobulin (Ig)-like receptor B4 (LILRB4)(previously termed gp49B1) is a member of the Ig superfamily expressed constitutively on the surface of mast cells, neutrophils, and macrophages. LILRB4 inhibits IgE-dependent activation of mast cells in vitro through its two immunoreceptor tyrosine-based inhibitory motifs (ITIMs) that recruit the src homology domain type-2-containing tyrosine phosphatase 1 into the cell membrane. Accordingly, Lilrb4(-/-) mice exhibit greater incidence and severity of IgE- and mast cell-dependent anaphylactic inflammation compared with mice that express LILRB4. In addition, mast cell-dependent inflammation induced by the interaction of stem cell factor (SCF) with its receptor Kit is also more severe in Lilrb4(-/-) mice, indicating that the counterregulatory function of LILRB4 extends beyond inflammation induced by Fc receptors, which signal through ITIMs, to responses initiated through a receptor tyrosine kinase. Indeed, pathologic inflammatory responses induced by activation of neutrophils with lipopolysaccharide (LPS) alone or with tissue-specific autoantibodies are greatly exacerbated in Lilrb4(-/-) mice. The rapid upregulation of LILRB4 expression on neutrophils in Lilrb4(+/+) mice in response to LPS suggests it is an innate counterregulatory response designed to reduce pathologic inflammation. Nevertheless, LILRB4 also serves a similar purpose for inflammation induced by the humoral adaptive immune response that is manifested through effector cells bearing Fc receptors.
Collapse
Affiliation(s)
- Howard R Katz
- Department of Medicine, Harvard Medical School, Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Abstract
The type I Fc epsilon receptor (Fc epsilon RI) is one of the better understood members of its class and is central to the immunological activation of mast cells and basophils, the key players in immunoglobulin E (IgE)-dependent immediate hypersensitivity. This review provides background information on several distinct regulatory mechanisms controlling this receptor's stimulus-response coupling network. First, we review the current understanding of this network's operation, and then we focus on the inhibitory regulatory mechanisms. In particular, we discuss the different known cytosolic molecules (e.g. kinases, phosphatases, and adapters) as well as cell membrane proteins involved in negatively regulating the Fc epsilon RI-induced secretory responses. Knowledge of this field is developing at a fast rate, as new proteins endowed with regulatory functions are still being discovered. Our understanding of the complex networks by which these proteins exert regulation is limited. Although the scope of this review does not include addressing several important biochemical and biophysical aspects of the regulatory mechanisms, it does provide general insights into a central field in immunology.
Collapse
Affiliation(s)
- Jakub Abramson
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
21
|
Abstract
The immune system must effectively regulate the balance between beneficial and detrimental inflammation. This process is achieved in part through cell surface receptors that rapidly integrate activating and inhibitory signals. The inhibitory members of the leukocyte Ig-like receptor (LILR) family, termed LILRBs, are broadly distributed among cell populations in the immune system and potently counterregulate cell activation induced by stimuli of innate and adaptive immune responses. Studies in mice and humans indicate that LILRBs appreciably downregulate harmful inflammatory responses induced by microbial, allergic, and cytotoxic mechanisms. Hence, the LILRBs likely play significant roles in regulating the incidence and severity of many inflammatory diseases, making them potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Howard R Katz
- Division of Rheumatology, Immunology, and Allergy, Harvard Medical School/Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
22
|
Nutku E, Hudson SA, Bochner BS. Mechanism of Siglec-8-induced human eosinophil apoptosis: role of caspases and mitochondrial injury. Biochem Biophys Res Commun 2005; 336:918-24. [PMID: 16157303 DOI: 10.1016/j.bbrc.2005.08.202] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Accepted: 08/25/2005] [Indexed: 11/16/2022]
Abstract
Sialic acid binding immunoglobulin like lectin (Siglec)-8 crosslinking with specific antibodies causes human eosinophil apoptosis. Mechanisms by which Siglec-8 crosslinking induces apoptosis are not known. Peripheral blood eosinophils were examined for caspase, mitochondria and reactive oxygen species (ROS) involvement after incubating the cells with anti-Siglec-8 crosslinking Abs or control Abs, in the presence or absence of selective inhibitors. Siglec-8 crosslinking induced rapid cleavage of caspase-3, caspase-8, and caspase-9 in eosinophils. Selective caspase-8 and/or caspase-9 inhibitors inhibited this apoptosis. Siglec-8 crosslinking on eosinophils increased dissipation of mitochondrial membrane potential upstream of caspase activation. Rotenone and antimycin, inhibitors of mitochondrial respiratory chain components, completely inhibited apoptosis. Additional experiments with an inhibitor of ROS, diphenyleneiodonium, demonstrated that ROS was also essential for Siglec-8-mediated apoptosis and preceded Siglec-8-mediated mitochondrial dissipation. These experiments show that Siglec-8-induced apoptosis occurs through the sequential production of ROS, followed by induction of mitochondrial injury and caspase cleavage.
Collapse
Affiliation(s)
- Esra Nutku
- Department of Medicine, Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | |
Collapse
|
23
|
Hoelsbrekken SE, Fossum S, Dissen E. Molecular cloning of LILRC1 and LILRC2 in the mouse and the rat, two novel immunoglobulin-like receptors encoded by the leukocyte receptor gene complex. Immunogenetics 2005; 57:479-86. [PMID: 16041585 DOI: 10.1007/s00251-005-0014-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2005] [Accepted: 06/13/2005] [Indexed: 10/25/2022]
Abstract
We report the molecular cloning of two novel single-member receptor families with homology to LILR/CD85, PIR, and gp49: LILRC1 in the rat and the mouse, and LILRC2 in the rat. LILRC1 and LILRC2 both have two extracellular Ig-like domains and a cytoplasmic tail devoid of any known signaling motifs. The transmembrane regions of LILRC1 and LILRC2 contain an arginine residue, a common feature in receptors that associate with activating adaptor proteins. Rat and mouse LILRC1 are orthologs sharing 81.5% amino acid identity. LILRC2 represents a distinct receptor family, 47.9% identical to LILRC1. No murine LILRC2 ortholog was detected in genome or expressed sequence tag sequence databases. By radiation hybrid mapping, the rat Lilrc1 and Lilrc2 loci were localized to the leukocyte receptor gene complex (LRC) on chromosome 1, and the mouse Lilrc1 locus was mapped to the LRC on chromosome 7. Moreover, the mouse and rat Lilrc1 loci were localized to similar positions within the LRC. As shown by RT-PCR, rat LILRC1 was expressed by B cells, neutrophils, and a macrophage cell line. Transcription of LILRC2 was detected in T cells, B cells, neutrophils, and macrophages.
Collapse
|
24
|
Bryceson YT, Torgersen KM, Inngjerdingen M, Berg SF, Hoelsbrekken SE, Fossum S, Dissen E. The rat orthologue to the inhibitory receptor gp49B is expressed by neutrophils and monocytes, but not by NK cells or mast cells. Eur J Immunol 2005; 35:1230-9. [PMID: 15756648 DOI: 10.1002/eji.200324593] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Mouse gp49B is a member of the leukocyte immunoglobulin-like receptor family. It is constitutively expressed by mast cells and certain myeloid cells, and expression can be induced on natural killer (NK) cells and T cells. We have cloned several rat cDNA, 78% identical to mouse gp49B at the amino acid level, that represent the rat orthologue to mouse gp49B. A mouse monoclonal antibody (WEN29) against rat gp49B was generated. By flow cytometry and Northern blot analysis, gp49B was found to be expressed by neutrophils and monocytes, but not NK cells (primary or IL-2-activated), T cells (resting or concanavalin A-stimulated) or peritoneal mast cells. Following pervanadate treatment, the tyrosine phosphatase SHP-1 was co-immunoprecipitated with gp49B in the macrophage cell line R2. In glutathione S-transferase pull-down experiments, the cytoplasmic tail of rat gp49B associated with the SH2 domains of both SHP-1 and SHP-2, dependent on intact and phosphorylated immunoreceptor tyrosine-based inhibition motifs (ITIM). Compared to mouse, the cytoplasmic domain of rat gp49B contains a third ITIM-like sequence (YLYASV) that was phosphorylated by several Src family tyrosine kinases, enhanced the phosphorylation of other ITIM, and bound to the SH2 domains of SHP-2, suggesting a role in the recruitment of downstream phosphatases.
Collapse
Affiliation(s)
- Yenan T Bryceson
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
25
|
Nutku E, Aizawa H, Hudson SA, Bochner BS. Ligation of Siglec-8: a selective mechanism for induction of human eosinophil apoptosis. Blood 2003; 101:5014-20. [PMID: 12609831 DOI: 10.1182/blood-2002-10-3058] [Citation(s) in RCA: 237] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sialic acid binding immunoglobulin-like lectin 8 (Siglec-8), which exists in 2 isoforms including one possessing cytoplasmic tyrosine motifs, is expressed only on human eosinophils, basophils, and mast cells. Until now, its function was unknown. Here we define a novel function of Siglec-8 on eosinophils. Siglec-8 cross-linking with antibodies rapidly generated caspase-3-like activity and reduced eosinophil viability through induction of apoptosis. The pancaspase inhibitor benzyloxycarbonyl (Cbz)-Val-Ala-Asp-(Ome)-fluoromethyl ketone (zVAD-FMK) completely blocked this response, implicating caspases in Siglec-8 cross-linking-induced apoptosis. Eosinophil survival-promoting cytokines such as interleukin 5 (IL-5) and granulocyte-macrophage colony-stimulating factor (GM-CSF) failed to block apoptosis and instead enhanced the sensitivity of eosinophils to undergo apoptosis in response to Siglec-8 antibody. Siglec-8 activation may provide a useful therapeutic approach to reduce numbers of eosinophils (and perhaps basophils and mast cells) in disease states where these cells are important.
Collapse
Affiliation(s)
- Esra Nutku
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD 21224, USA
| | | | | | | |
Collapse
|
26
|
Minoo P, Chughtai N, Campiglio M, Stein-Gerlach M, Lebrun JJ, Ullrich A, Ali S. The adaptor function of SHP-2 downstream of the prolactin receptor is required for the recruitment of p29, a substrate of SHP-2. Cell Signal 2003; 15:319-26. [PMID: 12531430 DOI: 10.1016/s0898-6568(02)00122-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
SHP-2, a cytosolic protein tyrosine phosphatase with two SH2 domains and multiple tyrosine phosphorylation sites, contributes to signal transduction as an enzyme and/or adaptor molecule. Here we demonstrate that prolactin (PRL) stimulation of the PRL-responsive Nb2 cells, a rat lymphoma cell line, and T47D cells, a human breast cancer cell line, lead to the complex formation of SHP-2 and growth factor receptor-bound protein-2 (grb2). Using transient co-overexpression studies of the prolactin receptor (PRLR) and several tyrosine to phenylalanine mutants of SHP-2, we show that grb2 associates with SHP-2 through the C-terminal tyrosine residues of SHP-2, Y(546) and Y(584). Furthermore, in this study, we found a highly phosphorylated, 29-kDa protein (p29), a substrate of SHP-2. The recruitment of p29 to SHP-2 requires the carboxy-terminal tyrosine residues of SHP-2 (Y(546) and Y(584)). Together, our results indicate that SHP-2 may function as an adaptor molecule downstream of the PRLR and highlight a new recruitment mechanism of SHP-2 substrates.
Collapse
Affiliation(s)
- Parham Minoo
- Division of Hematology, Department of Medicine, Molecular Oncology Group, H5-81, Royal Victoria Hospital, McGill University, 687 Pine Avenue West, Montreal, Quebec, Canada H3A 1A1
| | | | | | | | | | | | | |
Collapse
|
27
|
Welch AY, Kasahara M, Spain LM. Identification of the mouse killer immunoglobulin-like receptor-like (Kirl) gene family mapping to chromosome X. Immunogenetics 2003; 54:782-90. [PMID: 12618911 DOI: 10.1007/s00251-002-0529-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2002] [Accepted: 11/13/2002] [Indexed: 10/25/2022]
Abstract
Natural killer (NK) inhibitory receptors, which recognize major histocompatability complex (MHC) proteins in humans, are known as killer Ig-like receptors (KIRs) and are encoded by a multi-gene immunoglobulin (Ig) superfamily. In a screen for genes differentially expressed in the mouse thymus, we discovered the first close rodent homologue of the NK receptor KIR family, which we named KIR- Like (Kirl). KIRL1 shares 40% amino acid identity with primate KIR family members, with the majority of the homology contained within the Ig-like ectodomains. KIRL1 is more similar to the KIRs than to any other known member of the Ig domain-containing leukocyte receptor superfamily. This highly significant homology suggests that the KIR family did not arise independently in primates, as has been previously suggested, but rather evolved from a primordial gene already present in the common rodent/primate ancestor. KIRL1 lacks the cytoplasmic protein motifs that mediate inhibition in KIRs (immunoregulatory tyrosine inhibiting motif, ITIM); KIRL1 also lacks the transmembrane activation signature (a conserved K residue involved in association with the immunoregulatory tyrosine activating motif-containing DAP12 molecule) found in some KIRs. Nevertheless, we hypothesize that Kirl1 is functional, for the following reasons: (1) Kirl1 mRNA is expressed at high levels in immature thymocytes; (2) Kirl1 is regulated during thymocyte development; (3) KIRL1 protein is detected in thymus. We also show that the mouse genome contains a closely related, transcribed gene, which we name Kirl2. Kirl2 encodes a KIR-like molecule with three Ig-like domains and also lacks tyrosine-based immunoregulatory motifs in its cytoplasmic region.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Chromosome Mapping
- DNA, Complementary/genetics
- Gene Expression
- Killer Cells, Natural/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Molecular Sequence Data
- Multigene Family
- Phylogeny
- RNA/genetics
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, KIR
- Receptors, KIR3DL1
- Sequence Homology, Amino Acid
- Sequence Homology, Nucleic Acid
- X Chromosome/genetics
Collapse
Affiliation(s)
- Alice Y Welch
- Immunology Department, Holland Laboratory for Biomedical Research, American Red Cross, 15601 Crabbs Branch Way, Rockville, MD 20855, USA
| | | | | |
Collapse
|
28
|
Abstract
We present a hypothesis regarding the mode of induction of the inhibitory phosphatases SHP-1 and SHIP in hematopoietic cells. One mode is a general one in which the phosphatase regulates but does not abort signal transduction and biology. Regulator phosphatases are induced by directly or indirectly engaging the amino acid motifs present in the activating receptor, and act to control the biochemical and biological output. The other mode of induction is a specific one, which critically involves paired co-clustering of activating and inhibitory receptors. Phosphatases working in this way act only under conditions of paired co-clustering of activating and inhibitory receptors, and directly bind amino acid motifs present in the inhibitory receptor. However, this mode of induction is apparently more efficient, as cellular activation is completely aborted. This review presents several examples of each mode of inhibition and speculates on their mechanisms.
Collapse
Affiliation(s)
- K M Coggeshall
- The Oklahoma Medical Research Foundation, Program in Immunobiology, 825 N.E. 13th St., Oklahoma City, OK 73104, USA.
| | | | | |
Collapse
|
29
|
Abstract
gp49B1 is a member of the immunoglobulin (Ig) superfamily expressed on the surface of mast cells, macrophages, and activated natural killer cells. gp49B1 inhibits FcepsilonRI-induced activation of mast cells in vitro by virtue of two immunoreceptor, tyrosine-based inhibitory motifs that recruit the SHP-1 tyrosine phosphatase to the plasma membrane. We created gp49B1 null mice by targeted gene disruption, and found that IgE-dependent mast cell activation is augmented in these animals. Moreover, the ensuing anaphylactic reactions and inflammation are enhanced in the absence of gp49B1. Thus, gp49B1 innately counter-regulates mast cell activation mediated by Ig generated through the adaptive immune response in vivo.
Collapse
Affiliation(s)
- Howard R Katz
- Division of Rheumatology, Immunology and Allergy, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Vanhaesebroeck B, Leevers SJ, Ahmadi K, Timms J, Katso R, Driscoll PC, Woscholski R, Parker PJ, Waterfield MD. Synthesis and function of 3-phosphorylated inositol lipids. Annu Rev Biochem 2002; 70:535-602. [PMID: 11395417 DOI: 10.1146/annurev.biochem.70.1.535] [Citation(s) in RCA: 1218] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The 3-phosphorylated inositol lipids fulfill roles as second messengers by interacting with the lipid binding domains of a variety of cellular proteins. Such interactions can affect the subcellular localization and aggregation of target proteins, and through allosteric effects, their activity. Generation of 3-phosphoinositides has been documented to influence diverse cellular pathways and hence alter a spectrum of fundamental cellular activities. This review is focused on the 3-phosphoinositide lipids, the synthesis of which is acutely triggered by extracellular stimuli, the enzymes responsible for their synthesis and metabolism, and their cell biological roles. Much knowledge has recently been gained through structural insights into the lipid kinases, their interaction with inhibitors, and the way their 3-phosphoinositide products interact with protein targets. This field is now moving toward a genetic dissection of 3-phosphoinositide action in a variety of model organisms. Such approaches will reveal the true role of the 3-phosphoinositides at the organismal level in health and disease.
Collapse
Affiliation(s)
- B Vanhaesebroeck
- Ludwig Institute for Cancer Research, Riding House Street, London W1W 7BS.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Multiple lines of experimental data indicate that SHIP1 is an important negative regulator of the immune system. SHIP1 has been demonstrated to control survival and proliferation, as well as differentiation. In the cases of some inhibitory receptors, such as Fc gamma RIIB1, the molecular mechanisms of control by SHIP1 are established. For other receptors, particularly activating receptors where SHIP1 appears to set activation thresholds, the mechanisms remain to be discovered. Further study on SHIP and other SHIP family members could be critical for our understanding the negative regulation in multiple hematopoietic lineages and the immune system as a whole.
Collapse
Affiliation(s)
- Michael E March
- Beirne B. Carter Center for Immunology Research and the Department of Microbiology, University of Virginia, Charlottesville, VA 22908, USA
| | | |
Collapse
|
32
|
Clark GJ, Cooper B, Fitzpatrick S, Green BJ, Hart DN. The gene encoding the immunoregulatory signaling molecule CMRF-35A localized to human chromosome 17 in close proximity to other members of the CMRF-35 family. TISSUE ANTIGENS 2001; 57:415-23. [PMID: 11556966 DOI: 10.1034/j.1399-0039.2001.057005415.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The immunoregulatory signaling (IRS) family includes several molecules, which play major roles in the regulation of the immune response. The CMRF-35A and CMRF-35H molecules are two new members of the IRS family of molecules, that are found on a wide variety of haemopoietic lineages. The extracellular functional interactions of these molecules is presently unknown, although CMRF-35H can initiate an inhibitory signal and is internalized when cross-linked. In this paper, we described the gene structure for the CMRF-35A gene and its localization to human chromosome 17. The gene consists of four exons spanning approximately 4.5 kb. Exon 1 encodes the 5' untranslated region and leader sequence, exon 2 encodes the immunoglobulin (Ig)-like domain, exon 3 encodes the membrane proximal region and exon 4 encodes the transmembrane region, the cytoplasmic tail and the 3' untranslated region. A region in the 5' flanking sequence of the CMRF-35A gene, that promoted expression of a reporter gene was identified. The genes for the CMRF-35A and CMRF-35H molecules are closely linked on chromosome 17. Similarity between the Ig-like exons and the preceding intron of the two genes suggests exon duplication was involved in their evolution. We also identified a further member of the CMRF-35 family, the CMRF-35J pseudogene. This gene appears to have arisen by gene duplication of the CMRF-35A gene. These three loci - the CMRF-35A, CMRF-35J and CMRF-35H genes-form a new complex of IRS genes on chromosome 17.
Collapse
Affiliation(s)
- G J Clark
- Mater Medical Research Institute, Aubigny Place, South Brisbane, Queensland, Australia.
| | | | | | | | | |
Collapse
|
33
|
Damen JE, Ware MD, Kalesnikoff J, Hughes MR, Krystal G. SHIP's C-terminus is essential for its hydrolysis of PIP3 and inhibition of mast cell degranulation. Blood 2001; 97:1343-51. [PMID: 11222379 DOI: 10.1182/blood.v97.5.1343] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The SH2-containing inositol-5'-phosphatase, SHIP, restrains bone marrow-derived mast cell (BMMC) degranulation, at least in part, by hydrolyzing phosphatidylinositol (PI)-3-kinase generated PI-3,4,5-P(3) (PIP3) to PI-3,4-P(2). To determine which domains within SHIP influence its ability to hydrolyze PIP3, bone marrow from SHIP(-/-) mice was retrovirally infected with various SHIP constructs. Introduction of wild-type SHIP into SHIP(-/-) BMMCs reverted the Steel factor (SF)-induced increases in PIP3, calcium entry, and degranulation to those observed in SHIP(+/+) BMMCs. A 5'-phosphatase dead SHIP, however, could not revert the SHIP(-/-) response, whereas a SHIP mutant in which the 2 NPXY motifs were converted to NPXFs (2NPXF) could partially revert the SHIP(-/-) response. SF stimulation of BMMCs expressing the 2NPXF, which could not bind Shc, led to the same level of mitogen-activated protein kinase (MAPK) phosphorylation as that seen in BMMCs expressing the other constructs. Surprisingly, C-terminally truncated forms of SHIP, lacking different amounts of the proline rich C-terminus, could not revert the SHIP(-/-) response at all. These results suggest that the C-terminus plays a critical role in enabling SHIP to hydrolyze PIP(3) and inhibit BMMC degranulation.
Collapse
Affiliation(s)
- J E Damen
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | | | | | | | | |
Collapse
|
34
|
Bruhns P, Vely F, Malbec O, Fridman WH, Vivier E, Daeron M. Molecular basis of the recruitment of the SH2 domain-containing inositol 5-phosphatases SHIP1 and SHIP2 by fcgamma RIIB. J Biol Chem 2000; 275:37357-64. [PMID: 11016922 DOI: 10.1074/jbc.m003518200] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
FcgammaRIIB are single-chain low affinity receptors for IgG that negatively regulate immunoreceptor tyrosine-based activation motif-dependent cell activation. They bear one immunoreceptor tyrosine-based inhibition motif (ITIM) that becomes tyrosyl-phosphorylated upon coaggregation of FcgammaRIIB with immunoreceptor tyrosine-based activation motif-bearing receptors and that recruits SH2 domain-containing inositol 5-phosphatases (SHIPs) in vivo. Synthetic FcgammaRIIB ITIM phosphopeptides, however, also bind SH2 domain-containing protein-tyrosine phosphatases (SHPs) in vitro. To identify SHIP-binding sites, we exchanged residues between the FcgammaRIIB ITIM and the N-terminal ITIM of a killer cell Ig-like receptor that does not bind SHIPs. Loss of function and gain of function substitutions identified the Y+2 leucine, in the FcgammaRIIB ITIM, as determining the binding of both SHIP1 and SHIP2, but not the binding of SHP-1 or SHP-2. Conversely, the Y-2 isoleucine that determines the in vitro binding of SHP-1 and SHP-2 affected neither the binding nor the recruitment of SHIP1 or SHIP2. One hydrophobic residue, in the ITIM of FcgammaRIIB therefore determines the affinity for SHIPs. This residue is symmetrical to the hydrophobic residue that determines the affinity of all ITIMs for SHPs. It defines a SHIP-binding site, distinct from a SHP-binding site, that enables FcgammaRIIB to recruit SHIP1 and SHIP2 and that is preferentially used in vivo.
Collapse
Affiliation(s)
- P Bruhns
- Laboratoire d'Immunologie Cellulaire et Clinique, INSERM U255, Institut Curie, 75005 Paris, France
| | | | | | | | | | | |
Collapse
|
35
|
Lee KH, Ono M, Inui M, Yuasa T, Takai T. Stimulatory function of gp49A, a murine Ig-like receptor, in rat basophilic leukemia cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:4970-7. [PMID: 11046024 DOI: 10.4049/jimmunol.165.9.4970] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Murine gp49, a 49-kDa type I transmembrane glycoprotein, is a member of the Ig-like receptors expressed on the surface of cells involved in natural immunity such as mast cells, NK cells, and macrophages. The two major subtypes, gp49A and gp49B, are encoded by two different genes adjacent to each other. gp49B contains an immunoreceptor tyrosine-based inhibitory motif in its cytoplasmic region and is known to function as an inhibitory molecule. In contrast, gp49A does not harbor any specific motif for signal transduction, nor has its physiological role been determined. Here we report on the stimulatory nature of gp49A by analyzing biochemical characteristics of chimeric molecules consisting of an ectodomain of Fc receptor and a C-terminal half of gp49A, namely the pretransmembrane, transmembrane, and cytoplasmic portions, expressed on the rat basophilic leukemia mast cell line. Cross-linking of the chimeric receptors evoked cytoplasmic calcium mobilization, PGD(2) release, and transcription of IL-3 and IL-4 genes, but did not elicit degranulation of the cells. The chimeric molecule could be expressed as a singlet and a homodimeric form on the cell surface. A pretransmembrane cysteine residue of gp49A was necessary for dimer formation. Dimerization was be necessary for their incorporation into glycolipid-enriched membrane fraction (GEM) upon cross-linking stimuli. The calcium mobilization response was inhibited by treatment of cells with methyl-beta-cyclodextrin, an inhibitor of GEM formation. Together with these results, it was strongly suggested that gp49A could be expressed as a homodimer and elicit activation signals that lead to calcium mobilization, eicosanoid production, and cytokine gene transcription through its incorporation into GEM.
Collapse
MESH Headings
- Animals
- Antigens, Surface/genetics
- Antigens, Surface/metabolism
- Antigens, Surface/physiology
- Calcium Signaling/genetics
- Calcium Signaling/immunology
- Cell Degranulation/genetics
- Cell Degranulation/immunology
- Cysteine/genetics
- Cysteine/metabolism
- Cytokines/genetics
- Dimerization
- Leukemia, Basophilic, Acute/immunology
- Leukemia, Basophilic, Acute/metabolism
- Mast Cells/immunology
- Mast Cells/metabolism
- Mast-Cell Sarcoma/immunology
- Mast-Cell Sarcoma/metabolism
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Membrane Glycoproteins/physiology
- Membrane Microdomains/genetics
- Membrane Microdomains/metabolism
- Membrane Microdomains/physiology
- Mice
- Prostaglandin D2/metabolism
- RNA, Messenger/metabolism
- Rats
- Receptor Aggregation/genetics
- Receptor Aggregation/immunology
- Receptors, IgG/genetics
- Receptors, IgG/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/physiology
- Recombinant Fusion Proteins/metabolism
- Recombinant Fusion Proteins/physiology
- Transcription, Genetic/immunology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- K H Lee
- Department of Experimental Immunology and Core Research for Evolutionary Science and Technology Program, Japan Science and Technology Corp, Institute of Development, Aging and Cancer, Tohoku University, Seiryo, Sendai, Japan
| | | | | | | | | |
Collapse
|
36
|
Beebe KD, Wang P, Arabaci G, Pei D. Determination of the binding specificity of the SH2 domains of protein tyrosine phosphatase SHP-1 through the screening of a combinatorial phosphotyrosyl peptide library. Biochemistry 2000; 39:13251-60. [PMID: 11052678 DOI: 10.1021/bi0014397] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A method for the rapid identification of high-affinity ligands to Src homology-2 (SH2) domains is reported. A phosphotyrosyl (pY) peptide library containing completely randomized residues at positions -2 to +3 relative to the pY was synthesized on TentaGel resin, with a unique peptide sequence on each resin bead (total 2.5 x 10(6) different sequences). The library was screened against the biotinylated N- and C-terminal SH2 domains of protein tyrosine phosphatase SHP-1, and the beads that carry high-affinity ligands of the SH2 domains were identified using an enzyme-linked assay involving a streptavidin-alkaline phosphatase conjugate. Peptide ladder sequencing of the selected beads using matrix-assisted laser desorption ionization mass spectrometry revealed consensus sequences for both SH2 domains. The N-terminal SH2 domain strongly selects for peptides with a leucine at the -2 position; at the C-terminal side of the pY residue, it can recognize two distinct classes of peptides with consensus sequences of LXpY(M/F)X(F/M) and LXpYAXL (X = any amino acid), respectively. The C-terminal SH2 domain exhibits almost exclusive selectivity for peptides of the consensus sequence, (V/I/L)XpYAX(L/V). Several representative sequences selected from the library were individually synthesized and tested for binding to the SH2 domains by surface plasmon resonance and for their ability to stimulate the catalytic activity of SHP-1. Both experiments have demonstrated that the selected peptides are capable of binding to the SH2 domains with dissociation constants (K(D)) in the low micromolar range.
Collapse
Affiliation(s)
- K D Beebe
- Ohio State Biochemistry Program and Department of Chemistry, The Ohio State University, 100 West 18th Avenue, Columbus, Ohio 43210, USA
| | | | | | | |
Collapse
|
37
|
Rojo S, Stebbins CC, Peterson ME, Dombrowicz D, Wagtmann N, Long EO. Natural killer cells and mast cells from gp49B null mutant mice are functional. Mol Cell Biol 2000; 20:7178-82. [PMID: 10982834 PMCID: PMC86271 DOI: 10.1128/mcb.20.19.7178-7182.2000] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Immune responses are controlled by a combination of positive and negative cellular signals. Effector cells in the immune system express inhibitory receptors that serve to limit effector cell expansion and to protect the host from autoreactivity. gp49B is a receptor of unknown function that is expressed on activated mast cells and natural killer (NK) cells and whose cytoplasmic tail endows it with inhibitory potential. To gain insight into the function of gp49B in mice, we disrupted the gp49B gene by homologous recombination. gp49B(0) mice were born at expected ratios, were healthy and fertile, and displayed normal long-term survival rates. gp49B(0) mice showed no defect in NK or mast cell development. Furthermore, NK and mast cells from the gp49B(0) mice showed activation properties in vitro similar to those of cells isolated from wild-type mice. Therefore, gp49B is not critical for the development, expansion, and maturation of mast cells and NK cells in vivo. The healthy status of gp49B(0) mice makes them suitable for testing the role of gp49B in immune responses to infectious agents.
Collapse
Affiliation(s)
- S Rojo
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA
| | | | | | | | | | | |
Collapse
|
38
|
Ott VL, Cambier JC. Activating and inhibitory signaling in mast cells: new opportunities for therapeutic intervention? J Allergy Clin Immunol 2000; 106:429-40. [PMID: 10984360 DOI: 10.1067/mai.2000.109428] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Immune responses are tightly controlled by the activities of both activating and inhibitory signals. At the cellular level, these signals are generated through engagement of membrane-associated receptors and coreceptors. The high-affinity IgE receptor FcepsilonRI is expressed on mast cells and basophils and, on cross-linking by multivalent antigen (allergen), stimulates the release of inflammatory mediators that induce acute allergic responses. Activation signals mediated by a variety of immune receptors (eg, B-cell receptor, T-cell receptor, and FcepsilonRI) are subject to negative regulation by a growing family of structurally and functionally related inhibitory receptors. Recent studies indicate that mast cells express multiple inhibitory receptors that may regulate FcepsilonRI-induced mast cell activation through similar mechanisms. The ability of inhibitory receptors to attenuate IgE-mediated allergic responses implicates them as potential targets for therapeutic intervention in the treatment of atopic disease. Indeed, coaggregation of activating and inhibitory receptors has been suggested as one possible mechanism to explain the beneficial effects of specific immunotherapy in the treatment of allergy. In this review we summarize the current knowledge of inhibitory receptors expressed in mast cells and the mechanisms through which they regulate mast cell function.
Collapse
Affiliation(s)
- V L Ott
- Department of Immunology, University of Colorado Health Sciences Center and, National Jewish Medical and Research Center, Denver, CO, USA
| | | |
Collapse
|
39
|
Abstract
In the last 10 years we have come to appreciate the central role that phosphatidylinositol (PI)-3,4,5-P(3)plays in regulating a vast array of biological responses to extracellular signals. The level of this phospholipid is low in resting cells but increases rapidly in response to growth factor/cytokine-stimulated plasma membrane recruitment and activation of PI-3-kinase. Within the last 3 years three enzymes, SHIP, SHIP2 and PTEN, that play key roles in regulating the level of PI-3,4,5-P(3)have been cloned. In this review I have attempted to summarize our current knowledge regarding these three intriguing phosphatases.
Collapse
Affiliation(s)
- G Krystal
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC, Canada V5Z 1L3.
| |
Collapse
|
40
|
Colonna M, Nakajima H, Cella M. A family of inhibitory and activating Ig-like receptors that modulate function of lymphoid and myeloid cells. Semin Immunol 2000; 12:121-7. [PMID: 10764620 DOI: 10.1006/smim.2000.0214] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- M Colonna
- Basel Institute for Immunology, 487 Grenzacherstrasse, Basel, CH-4005, Switzerland
| | | | | |
Collapse
|
41
|
Rohrschneider LR, Fuller JF, Wolf I, Liu Y, Lucas DM. Structure, function, and biology of SHIP proteins. Genes Dev 2000. [DOI: 10.1101/gad.14.5.505] [Citation(s) in RCA: 118] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
42
|
Zhao R, Zhao ZJ. Dissecting the interaction of SHP-2 with PZR, an immunoglobulin family protein containing immunoreceptor tyrosine-based inhibitory motifs. J Biol Chem 2000; 275:5453-9. [PMID: 10681522 DOI: 10.1074/jbc.275.8.5453] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tyrosine phosphorylation of membrane proteins plays a crucial role in cell signaling by recruiting Src homology 2 (SH2) domain-containing signaling molecules. Recently, we have isolated a transmembrane protein designated PZR that specifically binds tyrosine phosphatase SHP-2, which has two SH2 domains (Zhao, Z. J., and Zhao, R. (1998) J. Biol. Chem. 273, 29367-29372). PZR belongs to the immunoglobulin superfamily. Its intracellular segment contains four putative sites of tyrosine phosphorylation. By site-specific mutagenesis, we found that the tyrosine 241 and 263 embedded in the consensus immunoreceptor tyrosine-based inhibitory motifs VIYAQL and VVYADI, respectively, accounted for the entire tyrosine phosphorylation of PZR. The interaction between PZR and SHP-2 requires involvement of both tyrosyl residues of the former and both SH2 domains of the latter, since its was disrupted by mutating a single tyrosyl residue or an SH2 domain. Overexpression of catalytically inactive but not active forms of SHP-2 bearing intact SH2 domains in cells caused hyperphosphorylation of PZR. In vitro, tyrosine-phosphorylated PZR was efficiently dephosphorylated by the full-length form of SHP-2 but not by its SH2 domain-truncated form. Together, the data indicate that PZR serves not only as a specific anchor protein of SHP-2 on the plasma membrane but also as a physiological substrate of the enzyme. The coexisting binding and dephosphorylation of PZR by SHP-2 may function to terminate signal transduction initiated by PZR and SHP-2 and to set a threshold for the signal transduction to be initiated.
Collapse
Affiliation(s)
- R Zhao
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232-6305, USA
| | | |
Collapse
|
43
|
Mousseau DD, Banville D, L'Abbé D, Bouchard P, Shen SH. PILRalpha, a novel immunoreceptor tyrosine-based inhibitory motif-bearing protein, recruits SHP-1 upon tyrosine phosphorylation and is paired with the truncated counterpart PILRbeta. J Biol Chem 2000; 275:4467-74. [PMID: 10660620 DOI: 10.1074/jbc.275.6.4467] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SHP-1-mediated dephosphorylation of protein tyrosine residues is central to the regulation of several cell signaling pathways, the specificity of which is dictated by the intrinsic affinity of SH2 domains for the flanking sequences of phosphotyrosine residues. By using a modified yeast two-hybrid system and SHP-1 as bait, we have cloned a human cDNA, PILRalpha, encoding a 303-amino acid immunoglobulin-like transmembrane receptor bearing two cytoplasmic tyrosines positioned within an immunoreceptor tyrosine-based inhibitory motif. Substrate trapping in combination with pervanadate treatment of 293T cells confirms that PILRalpha associates with SHP-1 in vivo upon tyrosine phosphorylation. Mutation of the tyrosine residues in PILRalpha indicates the pivotal role of the Tyr-269 residue in recruiting SHP-1. Surface plasmon resonance analysis further suggests that the association between PILRalpha-Tyr-269 and SHP-1 is mediated primarily via the amino-terminal SH2 domain of the latter. Polymerase chain reaction amplification of cDNA in combination with genomic sequence analysis revealed a second gene, PILRbeta, coding for a putative activating receptor as suggested by a truncated cytoplasmic tail and a charged lysine residue in its transmembrane region. The PILRalpha and PILRbeta genes are localized to chromosome 7 which is in contrast with the mapping of known members of the inhibitory receptor superfamily.
Collapse
Affiliation(s)
- D D Mousseau
- Mammalian Cell Genetics, National Research Council-Biotechnology Research Institute, Montreal, Quebec H4P 2R2
| | | | | | | | | |
Collapse
|
44
|
Xie ZH, Zhang J, Siraganian RP. Positive regulation of c-Jun N-terminal kinase and TNF-alpha production but not histamine release by SHP-1 in RBL-2H3 mast cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:1521-8. [PMID: 10640770 DOI: 10.4049/jimmunol.164.3.1521] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The SH2-containing protein tyrosine phosphatase1 (SHP-1) is important for signaling from immune receptors. To investigate the role of SHP-1 in mast cells we overexpressed the wild-type and the phosphatase-inactive forms of SHP-1 in rat basophilic leukemia 2H3 (RBL-2H3) mast cell line. The phosphatase-inactive SHP-1 (C453S or D419A) retains its ability to bind tyrosine phosphorylated substrates and thereby competes with the endogenous wild-type enzyme. Overexpression of wild-type SHP-1 decreased the FcepsilonRI aggregation-induced tyrosine phosphorylation of the beta and gamma subunits of the receptor whereas the dominant negative SHP-1 enhanced phosphorylation. There were also similar changes in the tyrosine phosphorylation of Syk. However, receptor-induced histamine release in the cells expressing either wild-type or dominant negative SHP-1 was similar to that in the parental control cells. In contrast, compared with the parental RBL-2H3 cells, FcepsilonRI-induced c-Jun N-terminal kinase phosphorylation and the level of TNF-alpha mRNA was increased in the cells overexpressing wild-type SHP-1 whereas the dominant negative SHP-1 had the opposite effect. The substrate-trapping mutant SHP1/D419A identified pp25 and pp30 as two major potential substrates of SHP-1 in RBL-2H3 cells. Therefore, SHP-1 may play a role in allergy and inflammation by regulating mast cell cytokine production.
Collapse
Affiliation(s)
- Z H Xie
- Receptors and Signal Transduction Section, OIIB, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
45
|
Mustelin T, Brockdorff J, Rudbeck L, Gjörloff-Wingren A, Han S, Wang X, Tailor P, Saxena M. The next wave: protein tyrosine phosphatases enter T cell antigen receptor signalling. Cell Signal 1999; 11:637-50. [PMID: 10530872 DOI: 10.1016/s0898-6568(99)00016-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Recent years have seen an exponentially increasing interest in the molecular mechanisms of signal transduction. Much of the focus has been on protein tyrosine kinase-mediated signalling, while the study of protein tyrosine phosphatases has lagged behind. We predict that the phosphatases will become a "hot topic" in the field within the next few years. This review summarizes the current state-of-the-art in our understanding of the structure, regulation and role of protein tyrosine phosphatases in T lymphocyte activation.
Collapse
Affiliation(s)
- T Mustelin
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, San Diego, CA 92121, USA.
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Daëron M, Vivier E. Biology of immunoreceptor tyrosine-based inhibition motif-bearing molecules. Curr Top Microbiol Immunol 1999; 244:1-12. [PMID: 10453645 DOI: 10.1007/978-3-642-58537-1_1] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- M Daëron
- Laboratoire d'Immunologie Cellulaire et Clinique, INSERM U.255, Institut Curie, Paris, France
| | | |
Collapse
|
47
|
Wagtmann N. gp49: an Ig-like receptor with inhibitory properties on mast cells and natural killer cells. Curr Top Microbiol Immunol 1999; 244:107-13. [PMID: 10453653 DOI: 10.1007/978-3-642-58537-1_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Affiliation(s)
- N Wagtmann
- Centre d'Immunologie, INSERM/CNRS de Marseille-Luminy, France
| |
Collapse
|
48
|
Affiliation(s)
- S Bolland
- Rockefeller University, New York, New York 10021, USA
| | | |
Collapse
|
49
|
Abstract
Major histocompatibility complex class I-specific inhibitory receptors on natural killer cells prevent the lysis of healthy autologous cells. The outcome of this negative signal is not anergy or apoptosis of natural killer cells but a transient abortion of activation signals. The natural killer inhibitory receptors fulfill this function by recruiting the tyrosine phosphatase SHP-1 through a cytoplasmic immunoreceptor tyrosine-based inhibition motif. This immunoreceptor tyrosine-based inhibition motif has become the hallmark of a growing family of receptors with inhibitory potential, which are expressed in various cell types such as monocytes, macrophages, dendritic cells, leukocytes, and mast cells. Most of the natural killer inhibitory receptors and two members of a monocyte inhibitory-receptor family bind major histocompatibility complex class I molecules. Ligands for many of the other receptors have yet to be identified. The inhibitory-receptor superfamily appears to regulate many types of immune responses by blocking cellular activation signals.
Collapse
Affiliation(s)
- E O Long
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland 20852, USA.
| |
Collapse
|
50
|
Huber M, Helgason CD, Damen JE, Scheid M, Duronio V, Liu L, Ware MD, Humphries RK, Krystal G. The role of SHIP in growth factor induced signalling. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 1999; 71:423-34. [PMID: 10354708 DOI: 10.1016/s0079-6107(98)00049-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The recently cloned, hemopoietic-specific, src homology 2 (SH2)-containing inositol phosphatase, SHIP, is rapidly gaining prominence as a potential regulator of all phosphatidylinositol (PI)-3 kinase mediated events since it has been shown both in vitro and in vivo to hydrolyze the 5' phosphate from phosphatidylinositol-3,4,5-trisphosphate (PI-3,4,5-P3). Thus SHIP, and its more widely expressed counterpart, SHIP2, could play a central role in determining PI-3,4,5-P3 and PI-3,4-P2 levels in many cell types. To explore the in vivo function of SHIP further we recently generated a SHIP knock out mouse and in this review we discuss experiments carried out with bone marrow derived mast cells (BMMCs) from these animals.
Collapse
Affiliation(s)
- M Huber
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|