1
|
Liu J, Wang M, Tian X, Wu S, Peng H, Zhu Y, Liu Y. New insights into allergic rhinitis treatment: MSC nanovesicles targeting dendritic cells. J Nanobiotechnology 2024; 22:575. [PMID: 39294599 PMCID: PMC11411834 DOI: 10.1186/s12951-024-02748-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 08/02/2024] [Indexed: 09/20/2024] Open
Abstract
Allergic rhinitis (AR) is a condition with limited treatment options. This study investigates the potential use of mesenchymal stem cell (MSC) nanovesicles as a novel therapy for AR. Specifically, the study explores the underlying mechanisms of MSC nanovesicle therapy by targeting dendritic cells (DCs). The researchers fabricated DC-targeted P-D2-EVs nanovesicles and characterized their properties. Transcriptomic sequencing and single-cell sequencing analyses were performed to study the impact of P-D2-EVs on AR mice, identifying core genes involved in the treatment. In vitro cell experiments were conducted to validate the effects of P-D2-EVs on DC metabolism, Th2 differentiation, and ILC2 activation. The results showed that P-D2-EVs efficiently targeted DCs. Transcriptomic sequencing analysis revealed differential expression of 948 genes in nasal tissue DCs of mice treated with P-D2-EVs. Single-cell sequencing further revealed that P-D2-EVs had inhibitory effects on DC activation, Th2 differentiation, and ILC2 activation, with Fut1 identified as the core gene. Validation experiments demonstrated that P-D2-EVs improved IL10 metabolism in DCs by downregulating Fut1 expression, thereby suppressing Th2 differentiation and ILC2 activation. Animal experiments confirmed the inhibitory effects of P-D2-EVs and their ability to ameliorate AR symptoms in mice. The study suggests that P-D2-EVs reshape DC metabolism and suppress Th2 differentiation and ILC2 activation through the inhibition of the Fut1/ICAM1/P38 MAPK signaling pathway, providing a potential therapeutic approach for AR.
Collapse
Affiliation(s)
- Jianyu Liu
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Meiqun Wang
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Xiaoyan Tian
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Shuhong Wu
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Haisen Peng
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Yaqiong Zhu
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China
| | - Yuehui Liu
- Department of Otolaryngology Head and Neck Surgery, Jiangxi Medical College, Nanchang University, No. 1, Minde Road, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
2
|
Shilts J, Wright GJ. Mapping the Human Cell Surface Interactome: A Key to Decode Cell-to-Cell Communication. Annu Rev Biomed Data Sci 2024; 7:155-177. [PMID: 38723658 DOI: 10.1146/annurev-biodatasci-102523-103821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2024]
Abstract
Proteins on the surfaces of cells serve as physical connection points to bridge one cell with another, enabling direct communication between cells and cohesive structure. As biomedical research makes the leap from characterizing individual cells toward understanding the multicellular organization of the human body, the binding interactions between molecules on the surfaces of cells are foundational both for computational models and for clinical efforts to exploit these influential receptor pathways. To achieve this grander vision, we must assemble the full interactome of ways surface proteins can link together. This review investigates how close we are to knowing the human cell surface protein interactome. We summarize the current state of databases and systematic technologies to assemble surface protein interactomes, while highlighting substantial gaps that remain. We aim for this to serve as a road map for eventually building a more robust picture of the human cell surface protein interactome.
Collapse
Affiliation(s)
- Jarrod Shilts
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom;
- School of the Biological Sciences, University of Cambridge, Cambridge, United Kingdom;
| | - Gavin J Wright
- Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom;
| |
Collapse
|
3
|
Johannsen EB, Skakkebæk A, Kalucka JM, Fedder J, Gravholt CH, Just J. The testicular microvasculature in Klinefelter syndrome is immature with compromised integrity and characterized by excessive inflammatory cross-talk. Hum Reprod 2023; 38:2339-2349. [PMID: 37910660 PMCID: PMC10694403 DOI: 10.1093/humrep/dead224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/11/2023] [Indexed: 11/03/2023] Open
Abstract
STUDY QUESTION Does Klinefelter syndrome (KS) lead to a distinct gene expression pattern at single-cell level in the testes that could provide insight into the reported microvascular dysfunction in the testes? SUMMARY ANSWER A distinct gene expression pattern within microvascular-associated cells of males with KS suggests excessive endothelial cell (EC) activation, disorganized vessel formation, and the presence of immature vessels with compromised integrity. WHAT IS KNOWN ALREADY Recent studies show that males with KS exhibit microvascular dysfunction in their testes, which affects blood flow and is associated with lower circulating levels of testosterone. STUDY DESIGN, SIZE, DURATION A comparative cross-sectional study of males with KS (n = 6), non-obstructive azoospermia (NOA) (n = 5), cryptozoospermia (n = 3), and controls (n = 15) was carried out. PARTICIPANTS/MATERIALS, SETTING, METHODS We analyzed publicly available single-cell RNA sequencing data of testicular cells from males with KS, males with NOA, males with cryptozoospermia, and controls. The integration of these datasets allowed us to analyze gene expression profiles and communication patterns among the cell types within the testis and to identify capillary ECs to investigate changes at the microvascular level. MAIN RESULTS AND THE ROLE OF CHANCE Rooted in changes at the single-cell level, our study demonstrates a shift in gene expression forming the foundation for altered cellular communication, microvascular remodeling, and pro-inflammatory responses within the testes of males with KS. We identified genes that were dysregulated in capillary ECs from males with KS (Padj < 0.05). Specifically, the unique microvascular gene expression in males with KS indicated enhanced capillary EC activation and increased inflammatory cross-talk, leading to impaired vessel maturation and increased EC barrier permeability. LIMITATIONS, REASONS FOR CAUTION Our study is constrained by an unbalanced design, with varying sample sizes and number of cells within each group. We acknowledge the restricted access to clinical information. In addition, our findings were deduced from changes in gene expression, which limits us to infer potential biological consequences arising from these alterations. Furthermore, the absence of a pre-pubertal age group limits the generalizability of our findings and warrants further investigation. WIDER IMPLICATIONS OF THE FINDINGS This study offers novel insights into the testicular pathophysiology in KS and underscores the potential contribution of microvascular dysfunction to the hypogonadism and infertility observed in males with KS. While this study aims to better understand the microvascular dysfunction in KS, the precise connections to testosterone deficiency and testicular atrophy remain to be fully elucidated. STUDY FUNDING/COMPETING INTEREST(S) A.S. was supported by the Independent Research Fund Denmark (0134-00130B). C.H.G. was supported by Novo Nordisk Foundation (NNF15OC0016474, NNF20OC0060610), 'Fonden til lægevidenskabens fremme', the Familien Hede Nielsen foundation and the Independent Research Fund Denmark (0134-00406A). E.B.J. was supported by Aarhus University and E.B.J. and C.H.G by the Independent Research Fund Denmark (2096-00165A). J.M.K. was supported by Lundbeckfonden (R307-2018-3667), Carlsberg Fonden (CF19-0687), Novo Nordisk Fonden (0073440) and Steno Diabetes Center Aarhus (SDCA). The authors declare no conflicts of interest. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Emma B Johannsen
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Anne Skakkebæk
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus N, Denmark
| | - Joanna M Kalucka
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus N, Denmark
| | - Jens Fedder
- Centre of Andrology and Fertility Clinic, Odense University Hospital, Odense C, Denmark
- Research Unit of Gynaecology and Obstetrics, University of Southern Denmark, Odense C, Denmark
| | - Claus H Gravholt
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
- Department of Endocrinology, Aarhus University Hospital, Aarhus N, Denmark
| | - Jesper Just
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus N, Denmark
| |
Collapse
|
4
|
Xiang J, Devenport JM, Carter AJ, Staser KW, Kim MY, O' Neal J, Ritchey JK, Rettig MP, Gao F, Rettig G, Turk R, Lee BH, Cooper ML, DiPersio JF. An "off-the-shelf" CD2 universal CAR-T therapy for T-cell malignancies. Leukemia 2023; 37:2448-2456. [PMID: 37798328 PMCID: PMC10681896 DOI: 10.1038/s41375-023-02039-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/01/2023] [Accepted: 09/13/2023] [Indexed: 10/07/2023]
Abstract
T-cell malignancies are associated with frequent relapse and high morbidity, which is partly due to the lack of effective or targeted treatment options. To broaden the use of CAR-T cells in pan T-cell malignancies, we developed an allogeneic "universal" CD2-targeting CAR-T cell (UCART2), in which the CD2 antigen is deleted to prevent fratricide, and the T-cell receptor is removed to prevent GvHD. UCART2 demonstrated efficacy against T-ALL and CTCL and prolonged the survival of tumor-engrafted NSG mice in vivo. To evaluate the impact of CD2 on CAR-T function, we generated CD19 CAR-T cells (UCART19) with or without CD2 deletion, single-cell secretome analysis revealed that CD2 deletion in UCART19 reduced frequencies of the effector cytokines (Granzyme-B and IFN-γ). We also observed that UCART19ΔCD2 had reduced anti-tumor efficacy compared to UCART19 in a CD19+NALM6 xenograft model. Of note is that the reduced efficacy resulting from CD2 deletion was reversed when combined with rhIL-7-hyFc, a long-acting recombinant human interleukin-7. Treatment with rhIL-7-hyFc prolonged UCART2 persistence and increased survival in both the tumor re-challenge model and primary patient T-ALL model in vivo. Together, these data suggest that allogeneic fratricide-resistant UCART2, in combination with rhIL-7-hyFc, could be a suitable approach for treating T-cell malignancies.
Collapse
Affiliation(s)
- Jingyu Xiang
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Jessica M Devenport
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Alun J Carter
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Karl W Staser
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Division of Dermatology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Miriam Y Kim
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie O' Neal
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Julie K Ritchey
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael P Rettig
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | - Feng Gao
- Division of Public Health Sciences, Department of Surgery, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Rolf Turk
- Integrated DNA Technologies, Coralville, IA, USA
| | | | - Matthew L Cooper
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| | - John F DiPersio
- Division of Oncology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
5
|
Ji D, Jiang S, Zhang Q, Wang YK, Zhang J, Shen W, Li W, Liu R, Wang J, Mavis C, Gu JJ, Hu X. Early changes in soluble intracellular adhesion molecule-1 as prognostic biomarkers to immune checkpoint inhibitor. Clin Transl Sci 2023; 16:1396-1407. [PMID: 37317057 PMCID: PMC10432871 DOI: 10.1111/cts.13540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 04/09/2023] [Accepted: 04/27/2023] [Indexed: 06/16/2023] Open
Abstract
Serologic biomarker to predict clinical outcome is needed for immune checkpoint inhibitors (ICIs). We evaluated soluble intercellular adhesion molecules-1 (sICAM-1) as a predictor of response to ICIs treatment. Ninety-five patients with cancer treated with ICI were studied. The serum sICAM-1 levels of baseline, post two cycle therapy and end of therapy (EOT) were measured by enzyme-linked immunoassay. We randomly assigned the patients into the primary cohort (n = 47) and validation cohort (n = 48). Serum sICAM-1 post two cycle (277.7 ± 181.6 ng/mL) and EOT (403.9 ± 218.9 ng/mL) were significantly elevated compared to baseline (244.8 ± 153.8 ng/mL, p = 0.008 and p = 0.004, respectively). Early changes of sICAM-1 (ΔsICAM-1), deemed as sICAM-1 after two cycles minus baseline, were assessed. Following ICI treatments, responders had significantly lower ΔsICAM-1 compared with nonresponders in the primary cohort (p = 0.040) and the validation cohort (p = 0.026). High ΔsICAM-1 was strongly associated with inferior progression-free survival (PFS; (primary cohort: p = 0.001 and validation cohort: p = 0.002) and overall survival (OS; (primary cohort: p < 0.001 and validation cohort: p = 0.007). The ΔsICAM-1 remained independently associated with worse PFS and OS in the primary cohort and the validation cohort. Subgroup analysis indicated patients whose sICAM-1 significantly elevated had shorter PFS and OS in both anti-PD-1 and anti-PD-L1 treatment groups. Early change of serum sICAM-1 could be used to monitor and predict clinical benefit of ICI therapy in patients with solid cancer.
Collapse
Affiliation(s)
- Dongmei Ji
- Department of Head and Neck Tumors and Neuroendocrine TumorsFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Shiyu Jiang
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Department of LymphomaFudan University Shanghai Cancer CenterShanghaiChina
| | - Qunling Zhang
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Department of LymphomaFudan University Shanghai Cancer CenterShanghaiChina
| | - Y. Ken Wang
- Division of Management and EducationUniversity of Pittsburgh Bradford CampusBradfordPennsylvaniaUSA
| | - Jian Zhang
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Weina Shen
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Wenhua Li
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Department of Gastrointestinal Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Rujiao Liu
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Phase I Clinical Trial CenterFudan University Shanghai Cancer CenterShanghaiChina
| | - Jessica Wang
- Department of MedicineRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Cory Mavis
- Department of MedicineRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
- Department of ImmunologyRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Juan J. Gu
- Department of MedicineRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
- Department of ImmunologyRoswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| | - Xichun Hu
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
- Department of Breast cancer and Urological Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
| |
Collapse
|
6
|
Qin Y, Chen L, Fei Q, Shao X, Lv W, Yang J, Xu F, Shi J. Upregulation of CD226 on subsets of T cells and NK cells is associated with upregulated adhesion molecules and cytotoxic factors in patients with tuberculosis. Int Immunopharmacol 2023; 120:110360. [PMID: 37244120 DOI: 10.1016/j.intimp.2023.110360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/09/2023] [Accepted: 05/16/2023] [Indexed: 05/29/2023]
Abstract
Human T cells and natural killer (NK) cells are major effector cells of innate immunity exerting potential immune surveillance against tuberculosis infection. CD226 is an activating receptor playing vital roles in the functions of T cells and NK cells during HIV infection and tumorigenesis. However, CD226 is a less-studied activating receptor during Mycobacterium tuberculosis (Mtb) infection. In this study, we used peripheral blood from tuberculosis patients and healthy donors to evaluate CD226 immunoregulation functions from two independent cohorts using Flow cytometry. Here, we found that a subset of T cells and NK cells that constitutively express CD226 exhibit a distinct phenotype in TB patients. In fact, the proportions of CD226+ and CD226- cell subsets differ between healthy people and tuberculosis patients, and the expression of immune checkpoint molecules (TIGIT, NKG2A) and adhesion molecules (CD2, CD11a) in CD226+ and CD226- subsets of T cells and NK cells exhibits special regulatory roles. Furthermore, CD226+ subsets produced more IFN-γ and CD107a than CD226- subsets in tuberculosis patients. Our results imply that CD226 may be a potential predictor of disease progression and clinical efficacy in tuberculosis by mediating the cytotoxic capacity of T cells and NK cells.
Collapse
Affiliation(s)
- Yongwei Qin
- Department of Pathogen Biology, Medical College, Nantong University, No. 19 Qixiu Road, Nantong 226001, China
| | - Liangqiong Chen
- Department of Pathogen Biology, Medical College, Nantong University, No. 19 Qixiu Road, Nantong 226001, China; Affiliated Haian Hospital of Nantong University, Haian 226600, China
| | - Qiuwen Fei
- Department of Pathogen Biology, Medical College, Nantong University, No. 19 Qixiu Road, Nantong 226001, China
| | - Xiaoyi Shao
- Department of Pathogen Biology, Medical College, Nantong University, No. 19 Qixiu Road, Nantong 226001, China
| | - Wenxuan Lv
- Department of Pathogen Biology, Medical College, Nantong University, No. 19 Qixiu Road, Nantong 226001, China
| | - Junling Yang
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Nantong Clinical Medical Research Center of Cardiothoracic Disease, and Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China
| | - Feifan Xu
- Department of Pathogen Biology, Medical College, Nantong University, No. 19 Qixiu Road, Nantong 226001, China; Department of Clinical Laboratory, Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), Nantong, China.
| | - Jiahai Shi
- Nantong Key Laboratory of Translational Medicine in Cardiothoracic Diseases, Nantong Clinical Medical Research Center of Cardiothoracic Disease, and Institution of Translational Medicine in Cardiothoracic Diseases, Affiliated Hospital of Nantong University, Nantong, China.
| |
Collapse
|
7
|
Li B, Lu Y, Zhong MC, Qian J, Li R, Davidson D, Tang Z, Zhu K, Argenty J, de Peredo AG, Malissen B, Roncagalli R, Veillette A. Cis interactions between CD2 and its ligands on T cells are required for T cell activation. Sci Immunol 2022; 7:eabn6373. [PMID: 35930657 DOI: 10.1126/sciimmunol.abn6373] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
CD2 is largely described to promote T cell activation when engaged by its ligands, CD48 in mice and CD58 in humans, that are present on antigen-presenting cells (APCs). However, both CD48 and CD58 are also expressed on T cells. By generating new knockout mouse strains lacking CD2 or CD48 in the C57BL/6 background, we determined that whereas CD2 was necessary on T cells for T cell activation, its ligand CD48 was not required on APCs. Rather, CD48 was also needed on T cells. One exception was during cytotoxicity, which required CD48 on T cells and APCs. Fluorescence resonance energy transfer (FRET) studies in nonimmune cells provided evidence that cis interactions between CD2 and CD48 existed within individual cells. CD2-CD48 interactions on T cells enabled more robust T cell receptor (TCR) signals, including protein tyrosine phosphorylation. Using T cells from a CD2 knock-in mouse in which a tag was inserted at the carboxyl terminus of CD2, mass spectrometry analyses revealed that the role of CD2 in T cell activation correlated with its ability to interact with components of the TCR complex and the protein tyrosine kinase Lck. CD2-CD58 provided a similar function in human T cells. Thus, our data imply that T cell-intrinsic cis interactions of CD2 with its ligands are required for TCR signaling and T cell activation. Interactions with ligands on APCs contribute during cytotoxicity.
Collapse
Affiliation(s)
- Bin Li
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Molecular Biology Program, University of Montréal, Montréal, Québec H3T 1J4, Canada
| | - Yan Lu
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Ming-Chao Zhong
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Jin Qian
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Rui Li
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Dominique Davidson
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Zhenghai Tang
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada
| | - Kaiwen Zhu
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Jérémy Argenty
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - Anne Gonzalez de Peredo
- Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS UPS, Toulouse, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France.,Centre d'Immunophénomique, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - Romain Roncagalli
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université, INSERM, CNRS, 13288 Marseille, France
| | - André Veillette
- Laboratory of Molecular Oncology, Institut de recherches cliniques de Montréal (IRCM), Montréal, Québec H2W 1R7, Canada.,Molecular Biology Program, University of Montréal, Montréal, Québec H3T 1J4, Canada.,Department of Medicine, McGill University, Montréal, Québec H3G 1Y6, Canada
| |
Collapse
|
8
|
Engels R, Falk L, Albanese M, Keppler OT, Sewald X. LFA1 and ICAM1 are critical for fusion and spread of murine leukemia virus in vivo. Cell Rep 2022; 38:110279. [PMID: 35045303 DOI: 10.1016/j.celrep.2021.110279] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/18/2021] [Accepted: 12/23/2021] [Indexed: 11/25/2022] Open
Abstract
Murine leukemia virus (MLV)-presenting cells form stable intercellular contacts with target cells during infection of lymphoid tissue, indicating a role of cell-cell contacts in retrovirus dissemination. Whether host cell adhesion proteins are required for retrovirus spread in vivo remains unknown. Here, we demonstrate that the lymphocyte-function-associated-antigen-1 (LFA1) and its ligand intercellular-adhesion-molecule-1 (ICAM1) are important for cell-contact-dependent transmission of MLV between leukocytes. Infection experiments in LFA1- and ICAM1-deficient mice demonstrate a defect in MLV spread within lymph nodes. Co-culture of primary leukocytes reveals a specific requirement for ICAM1 on donor cells and LFA1 on target cells for cell-contact-dependent spread through trans- and cis-infection. Importantly, adoptive transfer experiments combined with a newly established MLV-fusion assay confirm that the directed LFA1-ICAM1 interaction is important for retrovirus fusion and transmission in vivo. Taken together, our data provide insights on how retroviruses exploit host proteins and the biology of cell-cell interactions for dissemination.
Collapse
Affiliation(s)
- Rebecca Engels
- LMU München, Max von Pettenkofer Institute & Gene Center, Virology, National Reference Center for Retroviruses, Munich, Germany
| | - Lisa Falk
- LMU München, Max von Pettenkofer Institute & Gene Center, Virology, National Reference Center for Retroviruses, Munich, Germany
| | - Manuel Albanese
- LMU München, Max von Pettenkofer Institute & Gene Center, Virology, National Reference Center for Retroviruses, Munich, Germany
| | - Oliver T Keppler
- LMU München, Max von Pettenkofer Institute & Gene Center, Virology, National Reference Center for Retroviruses, Munich, Germany
| | - Xaver Sewald
- LMU München, Max von Pettenkofer Institute & Gene Center, Virology, National Reference Center for Retroviruses, Munich, Germany.
| |
Collapse
|
9
|
Seyyar SA, Tıskaoğlu NS, Gürbostan G, Pekpak E, Sayglı O. Increased Endothelial Cell Density in Childhood Patients With Thalassemia Major. Eye Contact Lens 2021; 47:660-663. [PMID: 34173366 DOI: 10.1097/icl.0000000000000806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE We aimed to compare specular microscopy, biometric, refractive, and anterior segment topographic parameters in children with thalassemia major (TM) with normal children of the same age. MATERIAL METHOD This cross-sectional study included 55 eyes of 55 patients with TM and 63 eyes of 63 age-sex-matched healthy children; all patients with TM were receiving treatment with blood transfusions every 3 to 4 weeks and an oral iron chelator deferasirox (DFX) (ICL670) tablet. A comprehensive ophthalmic examination was performed, including refraction (spherical equivalent), best-corrected visual acuity, slit-lamp bio microscopy, specular examination, optical biometry, intraocular pressure, anterior segment topography, and fundus examination. RESULTS Endothelial cell density (ECD) was 3,138 in the TM group and 2,996 in the control group (P=0.003). The mean central corneal thickness was significantly thinner in the TM group (P=0.010). Flat keratometry (K1) was 43.5 D in the study group and 42.9 D in the control group (P=0.039). The mean anterior chamber depth (ACD) was shallower in the TM group (P=0.004); axial length (AL) was significantly shorter in the TM group (P=0.002). CONCLUSION This study shows that there are differences in ECD, AL, keratometry values, ACD, and anterior segment parameters of pediatric patients with TM compared with healthy controls.
Collapse
Affiliation(s)
- Sevim Ayca Seyyar
- Ophthalmology Department (S.A.S.), Kocaeli Derince Education and Research Hospital; Ophthalmology Department (N.S.T.), Ersin Arslan Education and Research Hospital; and Ophthalmology Department (G.G., E.P., O.S.), Gaziantep University Hospital
| | | | | | | | | |
Collapse
|
10
|
Coburn J, Garcia B, Hu LT, Jewett MW, Kraiczy P, Norris SJ, Skare J. Lyme Disease Pathogenesis. Curr Issues Mol Biol 2020; 42:473-518. [PMID: 33353871 DOI: 10.21775/cimb.042.473] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Lyme disease Borrelia are obligately parasitic, tick- transmitted, invasive, persistent bacterial pathogens that cause disease in humans and non-reservoir vertebrates primarily through the induction of inflammation. During transmission from the infected tick, the bacteria undergo significant changes in gene expression, resulting in adaptation to the mammalian environment. The organisms multiply and spread locally and induce inflammatory responses that, in humans, result in clinical signs and symptoms. Borrelia virulence involves a multiplicity of mechanisms for dissemination and colonization of multiple tissues and evasion of host immune responses. Most of the tissue damage, which is seen in non-reservoir hosts, appears to result from host inflammatory reactions, despite the low numbers of bacteria in affected sites. This host response to the Lyme disease Borrelia can cause neurologic, cardiovascular, arthritic, and dermatologic manifestations during the disseminated and persistent stages of infection. The mechanisms by which a paucity of organisms (in comparison to many other infectious diseases) can cause varied and in some cases profound inflammation and symptoms remains mysterious but are the subjects of diverse ongoing investigations. In this review, we provide an overview of virulence mechanisms and determinants for which roles have been demonstrated in vivo, primarily in mouse models of infection.
Collapse
Affiliation(s)
- Jenifer Coburn
- Center For Infectious Disease Research, Medical College of Wisconsin, 8701 Watertown Plank Rd., TBRC C3980, Milwaukee, WI 53226, USA
| | - Brandon Garcia
- Department of Microbiology and Immunology, East Carolina University, Brody School of Medicine, Greenville, NC 27858, USA
| | - Linden T Hu
- Department of Molecular Biology and Microbiology, Vice Dean of Research, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA 02111, USA
| | - Mollie W Jewett
- Immunity and Pathogenesis Division Head, Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, 6900 Lake Nona Blvd. Orlando, FL 32827, USA
| | - Peter Kraiczy
- Institute of Medical Microbiology and Infection Control, University Hospital Frankfurt, Goethe University Frankfurt, Paul-Ehrlich-Str. 40, 60596 Frankfurt, Germany
| | - Steven J Norris
- Department of Pathology and Laboratory Medicine, University of Texas Medical School at Houston, P.O. Box 20708, Houston, TX 77225, USA
| | - Jon Skare
- Professor and Associate Head, Texas A and M University, 8447 Riverside Pkwy, Bryan, TX 77807, USA
| |
Collapse
|
11
|
Van Laethem F, Saba I, Lu J, Bhattacharya A, Tai X, Guinter TI, Engelhardt B, Alag A, Rojano M, Ashe JM, Hanada KI, Yang JC, Sun PD, Singer A. Novel MHC-Independent αβTCRs Specific for CD48, CD102, and CD155 Self-Proteins and Their Selection in the Thymus. Front Immunol 2020; 11:1216. [PMID: 32612609 PMCID: PMC7308553 DOI: 10.3389/fimmu.2020.01216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/15/2020] [Indexed: 11/13/2022] Open
Abstract
MHC-independent αβTCRs (TCRs) recognize conformational epitopes on native self-proteins and arise in mice lacking both MHC and CD4/CD8 coreceptor proteins. Although naturally generated in the thymus, these TCRs resemble re-engineered therapeutic chimeric antigen receptor (CAR) T cells in their specificity for MHC-independent ligands. Here we identify naturally arising MHC-independent TCRs reactive to three native self-proteins (CD48, CD102, and CD155) involved in cell adhesion. We report that naturally arising MHC-independent TCRs require high affinity TCR-ligand engagements in the thymus to signal positive selection and that high affinity positive selection generates a peripheral TCR repertoire with limited diversity and increased self-reactivity. We conclude that the affinity of TCR-ligand engagements required to signal positive selection in the thymus inversely determines the diversity and self-tolerance of the mature TCR repertoire that is selected.
Collapse
Affiliation(s)
- François Van Laethem
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Ingrid Saba
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Jinghua Lu
- Structural Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, MD, United States
| | - Abhisek Bhattacharya
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Xuguang Tai
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Terry I Guinter
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Britta Engelhardt
- Theodor Kocher Institute, Faculty of Bern, Universität Bern, Bern, Switzerland
| | - Amala Alag
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Mirelle Rojano
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Jennifer M Ashe
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Ken-Ichi Hanada
- Surgery Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - James C Yang
- Surgery Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| | - Peter D Sun
- Structural Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, MD, United States
| | - Alfred Singer
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
12
|
Panova V, Attig J, Young GR, Stoye JP, Kassiotis G. Antibody-induced internalisation of retroviral envelope glycoproteins is a signal initiation event. PLoS Pathog 2020; 16:e1008605. [PMID: 32453763 PMCID: PMC7274472 DOI: 10.1371/journal.ppat.1008605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/05/2020] [Accepted: 05/05/2020] [Indexed: 12/31/2022] Open
Abstract
As obligate parasites, viruses highjack, modify and repurpose the cellular machinery for their own replication. Viral proteins have, therefore, evolved biological functions, such as signalling potential, that alter host cell physiology in ways that are still incompletely understood. Retroviral envelope glycoproteins interact with several host proteins, extracellularly with their cellular receptor and anti-envelope antibodies, and intracellularly with proteins of the cytoskeleton or sorting, endocytosis and recirculation pathways. Here, we examined the impact of endogenous retroviral envelope glycoprotein expression and interaction with host proteins, particularly antibodies, on the cell, independently of retroviral infection. We found that in the commonly used C57BL/6 substrains of mice, where murine leukaemia virus (MLV) envelope glycoproteins are expressed by several endogenous MLV proviruses, the highest expressed MLV envelope glycoprotein is under the control of an immune-responsive cellular promoter, thus linking MLV envelope glycoprotein expression with immune activation. We further showed that antibody ligation induces extensive internalisation from the plasma membrane into endocytic compartments of MLV envelope glycoproteins, which are not normally subject to constitutive endocytosis. Importantly, antibody binding and internalisation of MLV envelope glycoproteins initiates signalling cascades in envelope-expressing murine lymphocytic cell lines, leading to cellular activation. Similar effects were observed by MLV envelope glycoprotein ligation by its cellular receptor mCAT-1, and by overexpression in human lymphocytic cells, where it required an intact tyrosine-based YXXΦ motif in the envelope glycoprotein cytoplasmic tail. Together, these results suggest that signalling potential is a general property of retroviral envelope glycoproteins and, therefore, a target for intervention. The outcome of viral infection depends on the balance between host immunity and the ability of the virus to avoid, evade or subvert it. The envelope glycoproteins of diverse viruses, including retroviruses, are displayed on the surface of virions and of infected cells and thus constitute the major target of the host antibody response. Antibody responses are elicited not only against infectious viruses we acquire during our life-history, but also against the numerous retroviral envelopes encoded by our genome and acquired during our species’ life-history. In turn, viruses have evolved ways to reduce exposure of their envelope glycoproteins to the host immune system, including constitutive endocytosis or antibody-induced internalisation. Using murine leukaemia viruses as models of infectious and endogenous retroviruses, we show that antibody binding to retroviral envelopes induces extensive internalisation of the envelope-antibody complex and initiates signalling cascades, ultimately leading to transcriptional activation of envelope glycoprotein-expressing lymphocytes. We further show that expression of endogenous retroviral envelopes is coupled to physiological lymphocyte activation, integrating them with the immune response. These findings reveal an unexpected layer of interaction between the host antibody response and retroviral envelope glycoproteins, which could be considered immune receptors.
Collapse
Affiliation(s)
- Veera Panova
- Retroviral Immunology, The Francis Crick Institute, United Kingdom
| | - Jan Attig
- Retroviral Immunology, The Francis Crick Institute, United Kingdom
| | - George R. Young
- Retrovirus-Host Interactions, The Francis Crick Institute, London, United Kingdom
| | - Jonathan P. Stoye
- Retrovirus-Host Interactions, The Francis Crick Institute, London, United Kingdom
- Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, United Kingdom
- Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Poggi A, Benelli R, Venè R, Costa D, Ferrari N, Tosetti F, Zocchi MR. Human Gut-Associated Natural Killer Cells in Health and Disease. Front Immunol 2019; 10:961. [PMID: 31130953 PMCID: PMC6509241 DOI: 10.3389/fimmu.2019.00961] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 04/15/2019] [Indexed: 12/14/2022] Open
Abstract
It is well established that natural killer (NK) cells are involved in both innate and adaptive immunity. Indeed, they can recognize molecules induced at the cell surface by stress signals and virus infections. The functions of NK cells in the gut are much more complex. Gut NK cells are not precisely organized in lymphoid aggregates but rather scattered in the epithelium or in the stroma, where they come in contact with a multitude of antigens derived from commensal or pathogenic microorganisms in addition to components of microbiota. Furthermore, NK cells in the bowel interact with several cell types, including epithelial cells, fibroblasts, macrophages, dendritic cells, and T lymphocytes, and contribute to the maintenance of immune homeostasis and development of efficient immune responses. NK cells have a key role in the response to intestinal bacterial infections, primarily through production of IFNγ, which can stimulate recruitment of additional NK cells from peripheral blood leading to amplification of the anti-bacterial immune response. Additionally, NK cells can have a role in the pathogenesis of gut autoimmune inflammatory bowel diseases (IBDs), such as Crohn's Disease and Ulcerative Colitis. These diseases are considered relevant to the generation of gastrointestinal malignancies. Indeed, the role of gut-associated NK cells in the immune response to bowel cancers is known. Thus, in the gut immune system, NK cells play a dual role, participating in both physiological and pathogenic processes. In this review, we will analyze the known functions of NK cells in the gut mucosa both in health and disease, focusing on the cross-talk among bowel microenvironment, epithelial barrier integrity, microbiota, and NK cells.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Roberto Benelli
- Immunology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Roberta Venè
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Delfina Costa
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nicoletta Ferrari
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesca Tosetti
- Molecular Oncology and Angiogenesis Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
14
|
Corradetti B, Taraballi F, Martinez JO, Minardi S, Basu N, Bauza G, Evangelopoulos M, Powell S, Corbo C, Tasciotti E. Hyaluronic acid coatings as a simple and efficient approach to improve MSC homing toward the site of inflammation. Sci Rep 2017; 7:7991. [PMID: 28801676 PMCID: PMC5554184 DOI: 10.1038/s41598-017-08687-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 07/13/2017] [Indexed: 12/13/2022] Open
Abstract
A major challenge in regenerative medicine is to improve therapeutic cells' delivery and targeting using an efficient and simple protocol. Mesenchymal stem cells (MSC) are currently employed for the treatment of inflammatory-based diseases, due to their powerful immunosoppressive potential. Here we report a simple and versatile method to transiently overexpress the hyaluronic acid (HA) receptor, CD44, on MSC membranes, to improve their homing potential towards an inflammatory site without affecting their behavior. The effect of HA-coatings on murine MSC was functionally determined both, in vitro and in vivo as a consequence of the transient CD44 overexpression induced by HA. Data obtained from the in vitro migration assay demonstrated a two-fold increase in the migratory potential of HA-treated MSC compared to untreated cells. In an LPS-induced inflamed ear murine model, HA-treated MSC demonstrated a significantly higher inflammatory targeting as observed at 72 hrs as compared to untreated cells. This increased accumulation for HA-treated MSC yielded a substantial reduction in inflammation as demonstrated by the decrease in the expression of pro-inflammatory markers and by the induction of a pro-regenerative environment.
Collapse
Affiliation(s)
- Bruna Corradetti
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, 60131, Ancona, Italy
| | - Francesca Taraballi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Orthopaedic & Sports Medicine, The Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Jonathan O Martinez
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Silvia Minardi
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Nupur Basu
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Guillermo Bauza
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Centre for NanoHealth, Swansea University Medical School, Swansea University Bay, Singleton Park, SA2 8PP, Wales, UK
| | - Michael Evangelopoulos
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Sebastian Powell
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Claudia Corbo
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Ennio Tasciotti
- Center for Biomimetic Medicine, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Centre for NanoHealth, Swansea University Medical School, Swansea University Bay, Singleton Park, SA2 8PP, Wales, UK.
- Department of Orthopaedic & Sports Medicine, The Houston Methodist Hospital, Houston, TX, 77030, USA.
| |
Collapse
|
15
|
Grosche L, Draßner C, Mühl-Zürbes P, Kamm L, Le-Trilling VTK, Trilling M, Steinkasserer A, Heilingloh CS. Human Cytomegalovirus-Induced Degradation of CYTIP Modulates Dendritic Cell Adhesion and Migration. Front Immunol 2017; 8:461. [PMID: 28484459 PMCID: PMC5399032 DOI: 10.3389/fimmu.2017.00461] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 04/04/2017] [Indexed: 11/24/2022] Open
Abstract
As potent antigen-presenting cells, dendritic cells (DCs) are essential for the initiation of effective antiviral immune responses. Viruses and especially herpesviruses, which are able to establish lifelong persistence, exploit several immune evasion mechanisms targeting DC biology. Our group has previously shown that the α-herpesvirus herpes simplex virus type 1 inhibits mature DC (mDC) migration by inducing adhesion via degrading the cellular protein CYTIP (cytohesin-1 interacting protein), an important negative regulator of β2-integrin activity. In the present study, we extended our analysis to the β-herpesvirus human cytomegalovirus (HCMV), to investigate whether other herpesviridae also induce such modulations. Indeed, HCMV impairs mDC transwell migration capability following a CCL19-chemokine gradient, despite equivalent expression levels of the cognate chemokine receptor CCR7 at the corresponding time points post-infection. Remarkably, HCMV infection potently induced β2-integrin activity on mDCs. Furthermore, directly HCMV-infected mDCs, exhibiting viral gene expression, strongly adhere to fibronectin and ICAM-1, in contrast to mDCs lacking infection or viral gene expression. Interestingly, HCMV-positive mDCs display a proteasome-dependent degradation of CYTIP. Contrasting the migration toward CCL19, elevated expression levels of the chemokine receptor CXCR4 in HCMV-infected mDCs were associated with functional CXCL12-chemotaxis under the herein used conditions. In summary, our results show that HCMV shapes mDC adhesion to compromise migration toward CCL19, but retaining CXCL12 responsiveness. Thus, we hypothesize that a preferred migration pattern toward the bone marrow, but not to secondary lymphoid organs, could ultimately cause a failure in the induction of potent antiviral immune responses.
Collapse
Affiliation(s)
- Linda Grosche
- Department of Immunomodulation, University Hospital Erlangen, Erlangen, Germany
| | - Christina Draßner
- Department of Immunomodulation, University Hospital Erlangen, Erlangen, Germany
| | - Petra Mühl-Zürbes
- Department of Immunomodulation, University Hospital Erlangen, Erlangen, Germany
| | - Lisa Kamm
- Department of Immunomodulation, University Hospital Erlangen, Erlangen, Germany
| | | | - Mirko Trilling
- Institute for Virology, University Hospital Essen, Essen, Germany
| | | | | |
Collapse
|
16
|
Kolegraff K, Bostik P, Ansari AA. Characterization and Role of Lentivirus-Associated Host Proteins. Exp Biol Med (Maywood) 2016; 231:252-63. [PMID: 16514170 DOI: 10.1177/153537020623100303] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Enveloped viruses obtain their envelopes during the process of budding from infected cells. During this process, however, these viruses acquire parts of the host cell membranes and host cell-derived proteins as integral parts of their mature envelopes. These host-derived components of viral envelopes may subsequently exhibit various effects on the life cycle of the virus; virus cell interactions, especially host response to virus-incorporated self-proteins; and the pathogenesis of the disease induced by these viruses. Although it was known for some time that various viruses incorporate host cell-derived proteins, the issue of the role of these proteins has received increased attention, specifically in connection with human immunodeficiency virus (HIV) infection and development of acquired immunodeficiency syndrome (AIDS) in humans. The aim of this review is to summarize our current knowledge of the analysis and role of host-derived proteins associated with enveloped viruses, with emphasis on the potential role of these proteins in the pathogenesis of AIDS. Clearly, differences in the clinical outcome of those nonhuman primates infected with simian immunodeficiency virus (SIV) that are disease resistant compared with SIV-infected species that are disease susceptible provide a unique opportunity to determine whether differences in the incorporation of distinct sets of host proteins play a role with distinct clinical outcomes.
Collapse
Affiliation(s)
- Keli Kolegraff
- Department of Pathology and Laboratory Medicine, Emory University, WMB Room 2309, 101 Woodruff Circle, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
17
|
Yoo HY, Song YH, Foo M, Seo E, Hwang DS, Seo JH. Recombinant mussel proximal thread matrix protein promotes osteoblast cell adhesion and proliferation. BMC Biotechnol 2016; 16:16. [PMID: 26879700 PMCID: PMC4754843 DOI: 10.1186/s12896-016-0247-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 02/09/2016] [Indexed: 12/01/2022] Open
Abstract
Background von Willebrand factor (VWF) is a key load bearing domain for mamalian cell adhesion by binding various macromolecular ligands in extracellular matrix such as, collagens, elastin, and glycosaminoglycans. Interestingly, vWF like domains are also commonly found in load bearing systems of marine organisms such as in underwater adhesive of mussel and sea star, and nacre of marine abalone, and play a critical load bearing function. Recently, Proximal Thread Matrix Protein1 (PTMP1) in mussel composed of two vWF type A like domains has characterized and it is known to bind both mussel collagens and mammalian collagens. Results Here, we cloned and mass produced a recombinant PTMP1 from E. coli system after switching all the minor codons to the major codons of E. coli. Recombinant PTMP1 has an ability to enhance mouse osteoblast cell adhesion, spreading, and cell proliferation. In addition, PTMP1 showed vWF-like properties as promoting collagen expression as well as binding to collagen type I, subsequently enhanced cell viability. Consequently, we found that recombinant PTMP1 acts as a vWF domain by mediating cell adhesion, spreading, proliferation, and formation of actin cytoskeleton. Conclusions This study suggests that both mammalian cell adhesion and marine underwater adhesion exploits a strong vWF-collagen interaction for successful wet adhesion. In addition, vWF like domains containing proteins including PTMP1 have a great potential for tissue engineering and the development of biomedical adhesives as a component for extra-cellular matrix.
Collapse
Affiliation(s)
- Hee Young Yoo
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 790-784, Korea
| | - Young Hoon Song
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 712-749, Korea
| | - Mathias Foo
- School of Engineering, University of Warwick, Coventry, CV4 7AL, UK
| | - Eunseok Seo
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 790-784, Korea
| | - Dong Soo Hwang
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, 790-784, Korea. .,School of Environmental Science and Engineering, Pohang University of Science and Technology, Pohang, 790-784, Korea.
| | - Jeong Hyun Seo
- School of Chemical Engineering, Yeungnam University, Gyeongsan, 712-749, Korea.
| |
Collapse
|
18
|
Ostuni MA, Guellec J, Hermand P, Durand P, Combadière C, Pincet F, Deterre P. CX3CL1, a chemokine finely tuned to adhesion: critical roles of the stalk glycosylation and the membrane domain. Biol Open 2014; 3:1173-82. [PMID: 25395671 PMCID: PMC4265755 DOI: 10.1242/bio.20149845] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The multi-domain CX3CL1 transmembrane chemokine triggers leukocyte adherence without rolling and migration by presenting its chemokine domain (CD) to its receptor CX3CR1. Through the combination of functional adhesion assays with structural analysis using FRAP, we investigated the functional role of the other domains of CX3CL1, i.e., its mucin stalk, transmembrane domain, and cytosolic domain. Our results indicate that the CX3CL1 molecular structure is finely adapted to capture CX3CR1 in circulating cells and that each domain has a specific purpose: the mucin stalk is stiffened by its high glycosylation to present the CD away from the membrane, the transmembrane domain generates the permanent aggregation of an adequate amount of monomers to guarantee adhesion and prevent rolling, and the cytosolic domain ensures adhesive robustness by interacting with the cytoskeleton. We propose a model in which quasi-immobile CX3CL1 bundles are organized to quickly generate adhesive patches with sufficiently high strength to capture CX3CR1+ leukocytes but with sufficiently low strength to allow their patrolling behavior.
Collapse
Affiliation(s)
- Mariano A Ostuni
- INSERM, U 1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Sorbonne Universités, UPMC Université Paris 06, UMRS CR7, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Present address: INSERM, U 1134, Biologie Intégrée du Globule Rouge; Université Paris Diderot; Institut National de la Transfusion Sanguine, 6 rue Alexandre Cabanel, 75015, Paris, France
| | - Julie Guellec
- INSERM, U 1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Sorbonne Universités, UPMC Université Paris 06, UMRS CR7, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France
| | - Patricia Hermand
- INSERM, U 1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Sorbonne Universités, UPMC Université Paris 06, UMRS CR7, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France
| | - Pauline Durand
- Sorbonne Universités, UPMC Université Paris 06, UMR 94550 ENS Laboratoire de Physique Statistique, F-75005, Paris, France
| | - Christophe Combadière
- INSERM, U 1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Sorbonne Universités, UPMC Université Paris 06, UMRS CR7, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France
| | - Frédéric Pincet
- Sorbonne Universités, UPMC Université Paris 06, UMR 94550 ENS Laboratoire de Physique Statistique, F-75005, Paris, France
| | - Philippe Deterre
- INSERM, U 1135, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France Sorbonne Universités, UPMC Université Paris 06, UMRS CR7, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses, F-75013, Paris, France
| |
Collapse
|
19
|
Okanishi H, Kagawa Y, Watari T. Expression of selectins and P-selectin glycoprotein ligand-1 in dogs with lymphocytic–plasmacytic enteritis. Vet Immunol Immunopathol 2014; 161:42-8. [DOI: 10.1016/j.vetimm.2014.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/20/2014] [Accepted: 06/26/2014] [Indexed: 12/22/2022]
|
20
|
|
21
|
Poggi A, Zocchi MR. NK cell autoreactivity and autoimmune diseases. Front Immunol 2014; 5:27. [PMID: 24550913 PMCID: PMC3912987 DOI: 10.3389/fimmu.2014.00027] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 01/17/2014] [Indexed: 01/14/2023] Open
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.
Collapse
Affiliation(s)
- Alessandro Poggi
- Molecular Oncology and Angiogenesis Unit, IRCCS AOU San Martino-IST , Genoa , Italy
| | - Maria Raffaella Zocchi
- Division of Immunology, Transplants and Infectious Diseases, Scientific Institute San Raffaele , Milan , Italy
| |
Collapse
|
22
|
Schäkel K, Döbel T, Bosselmann I. Future treatment options for atopic dermatitis – Small molecules and beyond. J Dermatol Sci 2014; 73:91-100. [DOI: 10.1016/j.jdermsci.2013.11.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Revised: 11/12/2013] [Accepted: 11/19/2013] [Indexed: 01/10/2023]
|
23
|
|
24
|
Rodriguez-Plata MT, Puigdomènech I, Izquierdo-Useros N, Puertas MC, Carrillo J, Erkizia I, Clotet B, Blanco J, Martinez-Picado J. The infectious synapse formed between mature dendritic cells and CD4(+) T cells is independent of the presence of the HIV-1 envelope glycoprotein. Retrovirology 2013; 10:42. [PMID: 23590845 PMCID: PMC3640963 DOI: 10.1186/1742-4690-10-42] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 04/05/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Since cell-mediated infection of human immunodeficiency virus type 1 (HIV-1) is more efficient than cell-free infection, cell-to-cell propagation plays a crucial role in the pathogenesis of HIV-1 infection. Transmission of HIV-1 is enabled by two types of cellular contacts, namely, virological synapses between productively infected cells and uninfected target cells and infectious synapses between uninfected dendritic cells (DC) harboring HIV-1 and uninfected target cells. While virological synapses are driven by expression of the viral envelope glycoprotein on the cell surface, little is known about the role of envelope glycoprotein during contact between DC and T cells. We explored the contribution of HIV-1 envelope glycoprotein, adhesion molecules, and antigen recognition in the formation of conjugates comprising mature DC (mDC) and CD4(+) T cells in order to further evaluate their role in mDC-mediated HIV-1 transmission at the immunological synapse. RESULTS Unlike virological synapse, HIV-1 did not modulate the formation of cell conjugates comprising mDC harboring HIV-1 and non-activated primary CD4(+) T cells. Disruption of interactions between ICAM-1 and LFA-1, however, resulted in a 60% decrease in mDC-CD4(+) T-cell conjugate formation and, consequently, in a significant reduction of mDC-mediated HIV-1 transmission to non-activated primary CD4(+) T cells (p < 0.05). Antigen recognition or sustained MHC-TcR interaction did not enhance conjugate formation, but significantly boosted productive mDC-mediated transmission of HIV-1 (p < 0.05) by increasing T-cell activation and proliferation. CONCLUSIONS Formation of the infectious synapse is independent of the presence of the HIV-1 envelope glycoprotein, although it does require an interaction between ICAM-1 and LFA-1. This interaction is the main driving force behind the formation of mDC-CD4(+) T-cell conjugates and enables transmission of HIV-1 to CD4(+) T cells. Moreover, antigen recognition boosts HIV-1 replication without affecting the frequency of cellular conjugates. Our results suggest a determinant role for immune activation driven by mDC-CD4(+) T-cell contacts in viral dissemination and that this activation likely contributes to the pathogenesis of HIV-1 infection.
Collapse
Affiliation(s)
- Maria T Rodriguez-Plata
- AIDS Research Institute IrsiCaixa, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, 08916, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Sehgal M, Khan ZK, Talal AH, Jain P. Dendritic Cells in HIV-1 and HCV Infection: Can They Help Win the Battle? Virology (Auckl) 2013; 4:1-25. [PMID: 25512691 PMCID: PMC4222345 DOI: 10.4137/vrt.s11046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Persistent infections with human immunodeficiency virus type 1 (HIV-1) and hepatitis C virus (HCV) are a major cause of morbidity and mortality worldwide. As sentinels of our immune system, dendritic cells (DCs) play a central role in initiating and regulating a potent antiviral immune response. Recent advances in our understanding of the role of DCs during HIV-1 and HCV infection have provided crucial insights into the mechanisms employed by these viruses to impair DC functions in order to evade an effective immune response against them. Modulation of the immunological synapse between DC and T-cell, as well as dysregulation of the crosstalk between DCs and natural killer (NK) cells, are emerging as two crucial mechanisms. This review focuses on understanding the interaction of HIV-1 and HCV with DCs not only to understand the immunopathogenesis of chronic HIV-1 and HCV infection, but also to explore the possibilities of DC-based immunotherapeutic approaches against them. Host genetic makeup is known to play major roles in infection outcome and rate of disease progression, as well as response to anti-viral therapy in both HIV-1 and HCV-infected individuals. Therefore, we highlight the genetic variations that can potentially affect DC functions, especially in the setting of chronic viral infection. Altogether, we address if DCs’ potential as critical effectors of antiviral immune response could indeed be utilized to combat chronic infection with HIV-1 and HCV.
Collapse
Affiliation(s)
- Mohit Sehgal
- Department of Microbiology and Immunology, and the Drexel Institute for Biotechnology and Virology Research, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Zafar K Khan
- Department of Microbiology and Immunology, and the Drexel Institute for Biotechnology and Virology Research, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Andrew H Talal
- Center for the Study of Hepatitis C, Weill Cornell Medical College, New York, NY
| | - Pooja Jain
- Department of Microbiology and Immunology, and the Drexel Institute for Biotechnology and Virology Research, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
26
|
Epstein-Barr virus deubiquitinase downregulates TRAF6-mediated NF-κB signaling during productive replication. J Virol 2013; 87:4060-70. [PMID: 23365429 DOI: 10.1128/jvi.02020-12] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Epstein-Barr virus (EBV), a human oncogenic herpesvirus that establishes a lifelong latent infection in the host, occasionally enters lytic infection to produce progeny viruses. The EBV oncogene latent membrane protein 1 (LMP1), which is expressed in both latent and lytic infection, constitutively activates the canonical NF-κB (p65) pathway. Such LMP1-mediated NF-κB activation is necessary for proliferation of latently infected cells and inhibition of viral lytic cycle progression. Actually, canonical NF-κB target gene expression was suppressed upon the onset of lytic infection. TRAF6, which is activated by conjugation of polyubiquitin chains, associates with LMP1 to mediate NF-κB signal transduction. We have found that EBV-encoded BPLF1 interacts with and deubiquitinates TRAF6 to inhibit NF-κB signaling during lytic infection. HEK293 cells with BPLF1-deficient recombinant EBV exhibited poor viral DNA replication compared with the wild type. Furthermore, exogenous expression of BPLF1 or p65 knockdown in cells restored DNA replication of BPLF1-deficient viruses, indicating that EBV BPLF1 deubiquitinates TRAF6 to inhibit NF-κB signal transduction, leading to promotion of viral lytic DNA replication.
Collapse
|
27
|
Jung WC, Jang YJ, Kim JH, Park SS, Park SH, Kim SJ, Mok YJ, Kim CS. Expression of intercellular adhesion molecule-1 and e-selectin in gastric cancer and their clinical significance. J Gastric Cancer 2012; 12:140-8. [PMID: 23094225 PMCID: PMC3473220 DOI: 10.5230/jgc.2012.12.3.140] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 08/09/2012] [Accepted: 08/10/2012] [Indexed: 01/25/2023] Open
Abstract
Purpose Among cell adhesion molecules, serum levels of intercellular adhesion molecule-1 and E-selectin are known to be correlated with the metastatic potential of gastric cancer. In the present study, the authors investigated the expression of intercellular adhesion molecule-1 and E-selectin in gastric cancer tissues and cultured gastric cancer cells, and examined their clinical value in gastric cancer. Materials and Methods The protein was extracted from gastric cancer tissues and cultured gastric cancer cells (MKN-28 and Kato-III) and the expression of intercellular adhesion molecule-1 and E-selectin was examined by western blotting. The clinical significance of intercellular adhesion molecule-1 and E-selectin was explored, using immunohistochemical staining of specimens from 157 gastric cancer patients. Results In western blot analysis, the expressions of intercellular adhesion molecule-1 in gastric cancer tissues and cultured gastric cancer cells were increased, however, E-selectin in gastric cancer tissues and cells were not increased. Among 157 gastric cancer patients, 79 patients (50%) were intercellular adhesion molecule-1 positive and had larger tumor size, an increased depth of tumor invasion, lymph node metastasis and perineural invasion. The intercellular adhesion molecule-1 positive group showed a higher incidence of tumor recurrence (40.5%), and a poorer 3-year survival than the negative group (54.9 vs. 85.9%, respectively). Conclusions Intercellular adhesion molecule-1 is overexpressed in gastric cancer tissues and cultured gastric cancer cells, whereas E-selectin is not overexpressed. Increased expression of intercellular adhesion molecule-1 in gastric cancer could be related to the aggressive nature of the tumor, and has a poor prognostic effect on gastric cancer.
Collapse
Affiliation(s)
- Woo-Chul Jung
- Department of Surgery, Korea University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Gelam honey has a protective effect against lipopolysaccharide (LPS)-induced organ failure. Int J Mol Sci 2012; 13:6370-6381. [PMID: 22754370 PMCID: PMC3382820 DOI: 10.3390/ijms13056370] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Revised: 05/04/2012] [Accepted: 05/07/2012] [Indexed: 12/03/2022] Open
Abstract
Gelam honey exerts anti-inflammatory and antioxidant activities and is thought to have potent effects in reducing infections and healing wounds. The aim of this study was to investigate the effects of intravenously-injected Gelam honey in protecting organs from lethal doses of lipopolysaccharide (LPS). Six groups of rabbits (N = 6) were used in this study. Two groups acted as controls and received only saline and no LPS injections. For the test groups, 1 mL honey (500 mg/kg in saline) was intravenously injected into two groups (treated), while saline (1 mL) was injected into the other two groups (untreated); after 1 h, all four test groups were intravenously-injected with LPS (0.5 mg/kg). Eight hours after the LPS injection, blood and organs were collected from three groups (one from each treatment stream) and blood parameters were measured and biochemical tests, histopathology, and myeloperoxidase assessment were performed. For survival rate tests, rabbits from the remaining three groups were monitored over a 2-week period. Treatment with honey showed protective effects on organs through the improvement of organ blood parameters, reduced infiltration of neutrophils, and decreased myeloperoxidase activity. Honey-treated rabbits also showed reduced mortality after LPS injection compared with untreated rabbits. Honey may have a therapeutic effect in protecting organs during inflammatory diseases.
Collapse
|
29
|
Grace PM, Rolan PE, Hutchinson MR. Peripheral immune contributions to the maintenance of central glial activation underlying neuropathic pain. Brain Behav Immun 2011; 25:1322-32. [PMID: 21496480 DOI: 10.1016/j.bbi.2011.04.003] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 04/04/2011] [Accepted: 04/04/2011] [Indexed: 10/18/2022] Open
Abstract
Recent evidence implicates an adaptive immune response in the central nervous system (CNS) mechanisms of neuropathic pain. This review identifies how neuropathic pain alters CNS immune privilege to facilitate T cell infiltration. Once in the CNS, T cells may interact with the local antigen presenting cells, microglia, via the major histocompatibility complex and the costimulatory molecules CD40 and B7. In this way, T cells may contribute to the maintenance of neuropathic pain through pro-inflammatory interactions with microglia and by facilitating the activation of astrocytes in the spinal dorsal horn. Based on the evidence presented in this review, we suggest that this bidirectional, pro-inflammatory system of neurons, glia and T cells in neuropathic pain should be renamed the pentapartite synapse, and identifies the latest member as a potential disease-modifying therapeutic target.
Collapse
Affiliation(s)
- Peter M Grace
- Discipline of Pharmacology, School of Medical Sciences, University of Adelaide, Adelaide, SA 5005, Australia.
| | | | | |
Collapse
|
30
|
Gutenkunst RN, Coombs D, Starr T, Dustin ML, Goldstein B. A biophysical model of cell adhesion mediated by immunoadhesin drugs and antibodies. PLoS One 2011; 6:e19701. [PMID: 21629715 PMCID: PMC3100730 DOI: 10.1371/journal.pone.0019701] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Accepted: 04/07/2011] [Indexed: 12/29/2022] Open
Abstract
A promising direction in drug development is to exploit the ability of natural killer cells to kill antibody-labeled target cells. Monoclonal antibodies and drugs designed to elicit this effect typically bind cell-surface epitopes that are overexpressed on target cells but also present on other cells. Thus it is important to understand adhesion of cells by antibodies and similar molecules. We present an equilibrium model of such adhesion, incorporating heterogeneity in target cell epitope density, nonspecific adhesion forces, and epitope immobility. We compare with experiments on the adhesion of Jurkat T cells to bilayers containing the relevant natural killer cell receptor, with adhesion mediated by the drug alefacept. We show that a model in which all target cell epitopes are mobile and available is inconsistent with the data, suggesting that more complex mechanisms are at work. We hypothesize that the immobile epitope fraction may change with cell adhesion, and we find that such a model is more consistent with the data, although discrepancies remain. We also quantitatively describe the parameter space in which binding occurs. Our model elaborates substantially on previous work, and our results offer guidance for the refinement of therapeutic immunoadhesins. Furthermore, our comparison with data from Jurkat T cells also points toward mechanisms relating epitope immobility to cell adhesion.
Collapse
Affiliation(s)
- Ryan N. Gutenkunst
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
- * E-mail:
| | - Daniel Coombs
- Department of Mathematics and Institute of Applied Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Toby Starr
- Department of Pathology, New York University School of Medicine and Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, New York, New York, United States of America
| | - Michael L. Dustin
- Department of Pathology, New York University School of Medicine and Program in Molecular Pathogenesis, Skirball Institute of Biomolecular Medicine, New York, New York, United States of America
| | - Byron Goldstein
- Theoretical Biology and Biophysics Group, Theoretical Division, Los Alamos National Laboratory, Los Alamos, New Mexico, United States of America
| |
Collapse
|
31
|
Grace PM, Hutchinson MR, Bishop A, Somogyi AA, Mayrhofer G, Rolan PE. Adoptive transfer of peripheral immune cells potentiates allodynia in a graded chronic constriction injury model of neuropathic pain. Brain Behav Immun 2011; 25:503-13. [PMID: 21134441 DOI: 10.1016/j.bbi.2010.11.018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 11/30/2010] [Accepted: 11/30/2010] [Indexed: 10/18/2022] Open
Abstract
Recent evidence demonstrates that peripheral immune cells contribute to the nociceptive hypersensitivity associated with neuropathic pain by infiltrating the central nervous system (CNS). We have recently developed a rat model of graded chronic constriction injury (CCI) by varying the exposure of the sciatic nerve and control non-nerve tissue to surgical placement of chromic gut. We demonstrate that splenocytes can contribute significantly to CCI-induced allodynia, as adoptive transfer of these cells from high pain donors to low pain recipients potentiates allodynia (P<0.001). The phenomenon was replicated with peripheral blood mononuclear cells (P<0.001). Adoptive transfer of allodynia was not achieved in sham recipients, indicating that peripheral immune cells are only capable of potentiating existing allodynia, rather than establishing allodynia. As adoptively transferred cells were found by flow cytometry to migrate to the spleen (P<0.05) and potentiation of allodynia was prevented in splenectomised low pain recipients, adoptive transfer of high pain splenocytes may induce the migration of host-derived immune cells from the spleen to the CNS as observed by flow cytometry (P<0.05). Importantly, intrathecal transfer of CD45(+) cells prepared from spinal cords of high pain donors into low pain recipients led to potentiated allodynia (P<0.001), confirming that infiltrating immune cells are not passive bystanders, but actively contribute to nociceptive hypersensitivity in the lumbar spinal cord.
Collapse
Affiliation(s)
- Peter M Grace
- Discipline of Pharmacology, School of Medical Sciences, University of Adelaide, Adelaide 5005, Australia.
| | | | | | | | | | | |
Collapse
|
32
|
Romero M, Monzón CG, Clemente G, Salcedo M, Bañares R, Alvarez E, Diego A, Santos L, Otero RM. Modulation of ICAM-1 tissue expression in patients with liver transplantation (LT) and acute rejection (AR) after glucocorticoid treatment. Transpl Int 2011. [DOI: 10.1111/j.1432-2277.2000.tb02083.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
33
|
Fernández-Messina L, Ashiru O, Agüera-González S, Reyburn HT, Valés-Gómez M. The human NKG2D ligand ULBP2 can be expressed at the cell surface with or without a GPI anchor and both forms can activate NK cells. J Cell Sci 2011; 124:321-7. [PMID: 21224393 DOI: 10.1242/jcs.076042] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The activating immune receptor NKG2D binds to several stress-induced ligands that are structurally different. MHC-class-I-related chain (MIC) A/B molecules have a transmembrane domain, whereas most UL16 binding proteins (ULBPs) are glycosylphosphatidylinositol (GPI)-linked molecules. The significance of this variability in membrane anchors is unclear. Here, we demonstrate that ULBP2, but not ULBP1 or ULBP3, can reach the cell surface without the GPI modification. Several proteins are expressed at the cell surface as both transmembrane and GPI-linked molecules, either via alternative splicing or by the expression of linked genes. However, to our knowledge, ULBP2 is the first single mammalian cDNA that can be expressed as either a transmembrane or a GPI-anchored protein. The rate of maturation and the levels of cell surface expression of the non-GPI-linked form were lower than those of the GPI-linked ULBP2. Nonetheless, non-GPI ULBP2 was recognised by NKG2D and triggered NK cell cytotoxicity. These data show that differences in membrane attachment by NKG2D ligands are more important for regulation of their surface expression than for cytotoxic recognition by NKG2D and emphasise that detailed characterisation of the cell biology of individual NKG2D ligands will be necessary to allow targeted modulation of this system.
Collapse
Affiliation(s)
- Lola Fernández-Messina
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge CB21QP, UK
| | | | | | | | | |
Collapse
|
34
|
del Rio ML, Bernhardt G, Rodriguez-Barbosa JI, Förster R. Development and functional specialization of CD103+ dendritic cells. Immunol Rev 2010; 234:268-81. [PMID: 20193025 DOI: 10.1111/j.0105-2896.2009.00874.x] [Citation(s) in RCA: 206] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
CD103 (alpha(E)) integrin expression distinguishes a population of dendritic cells (DCs) that can be found in many if not all lymphoid and non-lymphoid organs. CD103(+) DCs display distinct functional activities. Migratory CD103(+) DCs derived from skin, lung, and intestine efficiently present exogenous antigens in their corresponding draining lymph nodes to specific CD8(+) T cells through a mechanism known as cross-presentation. On the T cells they prime, intestinal CD103(+) DCs can drive the induction of the chemokine receptor CCR9 and alpha(4)beta(7) integrin, both known as gut-homing receptors. CD103(+) DCs also contribute to control inflammatory responses and intestinal homeostasis by fostering the conversion of naive T cells into induced Foxp3(+) regulatory T cells, a mechanism that relies on transforming growth factor-beta and retinoic acid signaling. This review discusses recent findings that identify murine CD103(+) DCs as important regulators of the immune response.
Collapse
|
35
|
Single lysophosphatidylcholine components exhibit adjuvant activities in vitro and in vivo. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2010; 17:429-38. [PMID: 20071492 DOI: 10.1128/cvi.00420-09] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Improving vaccine immunogenicity by developing new adjuvant formulations has long been a goal of vaccinologists. It has previously been shown that a natural mix of lysophosphatidylcholine (LPC) from chicken eggs promotes mature dendritic cell (DC) generation in vitro and primes antigen-specific immune responses in mice. In the present study, we dissected the adjuvant potentials of five individual LPC components found in the chicken egg mixture. In vitro analyses of the impact of the individual components on the maturation of human DCs were performed by means of phenotypic analysis, chemokine secretion analysis, and analysis of the ability of mature DC to stimulate T lymphocytes. Two components, C16:0-LPC and C18:0-LPC, were identified to be capable of the upregulation of expression of CD86, HLA-DR, and CD40 on in vitro-cultured monocyte-derived DCs from healthy donors. Both induced the release of chemokines to high concentrations (macrophage inflammatory protein 1, monocyte chemoattractant protein 1) or moderate concentrations (interleukin-8 [IL-8], gamma interferon-inducible protein 10). In addition, C16:0-LPC engaged naïve T cells to produce gamma interferon. This suggests that C16:0-LPC and C18:0-LPC have the capacity to promote, at least in vitro, a Th1-oriented response. The intravenous injection of C16:0-LPC or C18:0-LPC into mice resulted in the detectable secretion of IL-6 and IL-5 in sera. Both LPC components were tested for their capacities to act as adjuvants for two selected immunogens: the hepatitis B virus surface antigen and the hepatitis C virus NS3 helicase. The secretion of specific IgG1 was observed with either or both C16:0-LPC and C18:0-LPC, depending on the immunogen tested, and was observed at an efficiency comparable to that of alum. These data identify C16:0-LPC and C18:0-LPC as the active components of the LPC natural mixture. Although discrepancies between the results of the in vitro and in vivo analyses existed, studies with animals suggest that these components can trigger significant and specific humoral-mediated immunity.
Collapse
|
36
|
Smeed JA, Watkins CA, Gossner AG, Hopkins J. Expression profiling reveals differences in immuno-inflammatory gene expression between the two disease forms of sheep paratuberculosis. Vet Immunol Immunopathol 2009; 135:218-25. [PMID: 20053460 DOI: 10.1016/j.vetimm.2009.11.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Revised: 10/26/2009] [Accepted: 11/30/2009] [Indexed: 01/22/2023]
Abstract
Paratuberculosis is a chronic enteropathy of ruminants caused by Mycobacterium avium subspecies paratuberculosis (MAP); infection of sheep results in two disease forms - paucibacillary (tuberculoid) and multibacillary (lepromatous) associated with the differential polarization of the immune response. In addition the majority of MAP-infected animals show no pathology and remain asymptomatic. Microarray and real-time RT-qPCR analyses were used to compare gene expression in ileum from sheep with the two disease forms and asymptomatic sheep, to further understand the molecular basis of the pathologies. Microarrays identified 36 genes with fold-change of >1.5 and P< or = 0.05 in at least one comparison; eight candidates were chosen for RT-qPCR validation. Sequence analysis of two candidates, CXCR4 and IGFBP6, identified three SNPs in each; five were found in all three forms of disease and showed no significant relationship to pathological type. The IGFBP6 G(3743) A SNP was not detected in asymptomatic sheep. The data show that the two forms of disease are associated with distinct molecular profiles highlighted by the differential expression of chemokine and chemokine receptor transcripts, the protein products of which might be implicated in the different cell infiltrates of the pathologies. The cells within the lesions also show evidence of abnormal activation; they express high levels of cytokine transcripts but have reduced expression levels of transcripts for T cell receptor associated molecules.
Collapse
Affiliation(s)
- J A Smeed
- The Roslin Institute, Royal (Dick) School of Veterinary Studies, University of Edinburgh, Summerhall, Edinburgh EH9 1QH, UK
| | | | | | | |
Collapse
|
37
|
Trabattoni D, Saresella M, Pacei M, Marventano I, Mendozzi L, Rovaris M, Caputo D, Borelli M, Clerici M. Costimulatory Pathways in Multiple Sclerosis: Distinctive Expression of PD-1 and PD-L1 in Patients with Different Patterns of Disease. THE JOURNAL OF IMMUNOLOGY 2009; 183:4984-93. [DOI: 10.4049/jimmunol.0901038] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
38
|
Tanaka, Yoshihiro Ohashi, Yasushi K A. The Herbal Medicine Shoseiryu-to inhibits Allergen-induced Synthesis of Tumour Necrosis Factor Alpha by Peripheral Blood Mononuclear Cells in Patients with Perennial Allergic Rhinitis. Acta Otolaryngol 2009. [DOI: 10.1080/00016489850182837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
39
|
Maio M, Del Vecchio L. Expression and Functional Role of CD54/Intercellular Adhesion Molecule-1 (ICAM-1) on Human Blood Cells. Leuk Lymphoma 2009; 8:23-33. [PMID: 1362919 DOI: 10.3109/10428199209049814] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CD54/Intercellular Adhesion Molecule-1 (ICAM-1) is a cell adhesion molecule largely distributed among normal and neoplastic tissues. Through the binding to its ligand(s) CD54 plays a key role in cell to cell interactions leading to the immune response. Recently, CD54 expression has been investigated on hematopoietic cells: the antigen is predominantly expressed in the early stages of normal hematopoiesis and during the activation of blood cells. As regards to hematological malignancies, CD54 is strongly expressed on neoplastic cells from "stem cell derived" neoplasms. In AML, CD54 expression is related with other differentiation-linked molecules such as CD34 and HLA-DR and is significantly correlated with FAB morphological classification. In lymphoproliferative disorders, a high CD54 expression is associated with germinal centre lymphomas. This review summarizes our current understanding of CD54 with emphasis on recent advances and reference to unresolved issues such as its prognostic role in the clinical outcome of oncohematological diseases.
Collapse
Affiliation(s)
- M Maio
- Division of Experimental Oncology 2, Immunology Section, C.R.O., Aviano, Italy
| | | |
Collapse
|
40
|
Intercellular adhesion molecule 1 (ICAM-1), but not ICAM-2 and -3, is important for dendritic cell-mediated human immunodeficiency virus type 1 transmission. J Virol 2009; 83:4195-204. [PMID: 19211748 DOI: 10.1128/jvi.00006-09] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dendritic cells (DCs) play a critical role in cell-to-cell-mediated transmission of human immunodeficiency virus type 1 (HIV-1). Interactions between intercellular adhesion molecules (ICAMs) and their ligands facilitate DC-T-cell contact. The interaction between ICAM-1 on DCs and leukocyte function-associated molecule 1 (LFA-1) on CD4(+) T cells has been proposed to be important for DC-mediated HIV-1 transmission. Given that DCs and T cells express multiple ICAMs and binding ligands, the relative importance of ICAMs in DC-mediated HIV-1 transmission remains to be defined. Here, we examine the role of ICAM-1, -2, and -3 in DC-mediated HIV-1 transmission to various types of target cells including primary CD4(+) T cells. The expression levels of ICAMs and their ligands on immature and mature DCs and various types of HIV-1 target cells were measured by flow cytometry. Blocking ICAM-1 in DCs with specific monoclonal antibodies and small interfering RNA impaired DC-mediated HIV-1 transmission. DC-mediated viral transmission was significantly inhibited when both ICAM-1 on DCs and LFA-1 on CD4(+) T cells were blocked. However, blockade of ICAM-1 on target cells did not significantly inhibit DC-mediated HIV-1 transmission. Ectopic expression and antibody blocking suggest that DC-mediated HIV-1 transmission to primary CD4(+) T cells is independent of ICAM-2 and ICAM-3. Taken together, our data clarified the role of ICAMs in DC-mediated HIV-1 transmission to CD4(+) T cells.
Collapse
|
41
|
Owens T, Bechmann I, Engelhardt B. Perivascular spaces and the two steps to neuroinflammation. J Neuropathol Exp Neurol 2009; 67:1113-21. [PMID: 19018243 DOI: 10.1097/nen.0b013e31818f9ca8] [Citation(s) in RCA: 247] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Immune cells enter the central nervous system (CNS) from the circulation under normal conditions for immunosurveillance and in inflammatory neurologic diseases. This review describes the distinct anatomic features of the CNS vasculature that permit it to maintain parenchymal homeostasis and which necessitate specific mechanisms for neuroinflammation to occur. We review the historical evolution of the concept of the blood-brain barrier and discuss distinctions between diffusion/transport of solutes and migration of cells from the blood to CNS parenchyma. The former is regulated at the level of capillaries, whereas the latter takes place in postcapillary venules. We summarize evidence that entry of immune cells into the CNS parenchyma in inflammatory conditions involves 2 differently regulated steps: transmigration of the vascular wall into the perivascular space and progression across the glia limitans into the parenchyma.
Collapse
Affiliation(s)
- Trevor Owens
- Medical Biotechnology Center, University of Southern Denmark, Odense C, Denmark.
| | | | | |
Collapse
|
42
|
Gaddi E, Laucella S, Balbaryski J, Cantisano C, Barboni G, Candi M, Giraudi V. Prognostic Value of Soluble Intercellular Adhesion Molecule-1 (s-ICAM-1) in HIV-Infected Children. Scand J Immunol 2008. [DOI: 10.1111/j.1365-3083.2000.00820.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
43
|
Taub DD, Schaffer E. Biological responses to chemokine superfamily members. CURRENT PROTOCOLS IN IMMUNOLOGY 2008; Chapter 6:6.12.1-6.12.32. [PMID: 18432804 DOI: 10.1002/0471142735.im0612s38] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
A detailed summary of all the chemokine subfamily members including alternative names, receptor-ligand pairs, receptor cellular distribution, and leukocyte responsiveness is presented here along with assays to measure the biological activities of chemokines on each of the major leukocyte subsets (i.e., microchemotaxis chambers, fluorescence-based assays, use of nitrocellulose filters and membranes coated with extracellular matrix proteins, cellular adhesion assays, mobilization of intracellular free calcium, actin polymerization, degranulation assays, and the use of chemokine receptor-specific antibodies in flow cytometric analysis and radiolabeled chemokine binding assays).
Collapse
|
44
|
Moul DK, Routhouska SB, Robinson MR, Korman NJ. Alefacept for moderate to severe atopic dermatitis: a pilot study in adults. J Am Acad Dermatol 2008; 58:984-9. [PMID: 18395294 DOI: 10.1016/j.jaad.2008.02.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2007] [Revised: 01/29/2008] [Accepted: 02/12/2008] [Indexed: 11/18/2022]
Abstract
BACKGROUND Atopic dermatitis is a common inflammatory skin condition with acute and chronic phases showing a prevalence of memory T cells. Alefacept is a fully human LFA-3/IgG1 fusion protein that inhibits T-cell activation and selectively reduces memory T cells, which may prove to be effective in the treatment of atopic dermatitis. OBJECTIVE We sought to evaluate clinical response of alefacept intramuscular (IM) injection for 16 weeks in adults with atopic dermatitis. METHODS This was an open-label study of a 16-week treatment regimen of alefacept IM injection in adults with moderate to severe inflammatory atopic dermatitis. Patients received alefacept (30 mg IM) weekly for the first 8 weeks. At week 9, patients who did not achieve a 50% reduction in their Eczema Area Severity Index (EASI) score continued on alefacept (30 mg IM) weekly; those patients with a 50% reduction in their EASI (EASI 50) score or higher had their weekly dose decreased (15 mg IM) for the remaining 8 weeks. RESULTS Nine patients with moderate to severe atopic dermatitis were enrolled and treated. At the primary end point, week 18, 1 patient achieved EASI 50 score and 1 patient achieved EASI 90 score; 4 patients had a decrease in EASI score of less than 50%, 1 patient had an increase in EASI score, and 2 patients withdrew early before the primary end point because of worsening disease. A Physician Global Assessment score of mild was achieved in 2 patients and 1 patient achieved a Physician Global Assessment score of almost clear. Minimal pruritus was reported by 3 patients and 1 patient reported no pruritus. The 16-week course of alefacept was well tolerated. LIMITATIONS The study was inherently limited by its small sample size, concomitant use of antihistamines, and open-label design, which increases the likelihood of observer and self-assessment bias. CONCLUSION The treatment regimen of alefacept for 16 weeks was well tolerated by our patients. Although, in this study, only 2 of the 9 patients with atopic dermatitis responded to treatment with alefacept, the study was inherently limited by the small sample size. Additional studies with a larger sample size, continued weekly use, or concomitant use of ultraviolet-B light therapy may be warranted to evaluate the possibility of alefacept as a therapy for patients with chronic atopic dermatitis.
Collapse
Affiliation(s)
- Danielle K Moul
- Department of Dermatology, University Hospitals Case Medical Center, Cleveland, Ohio 44106, USA
| | | | | | | |
Collapse
|
45
|
Kim YJ, Hwang SY, Hwang JS, Lee JW, Oh ES, Han IO. C6 glioma cell insoluble matrix components enhance interferon-gamma-stimulated inducible nitric-oxide synthase/nitric oxide production in BV2 microglial cells. J Biol Chem 2007; 283:2526-33. [PMID: 17981810 DOI: 10.1074/jbc.m610219200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Microglia are the primary central nervous system immune effector cells. Microglial activation is linked to interactions with extracellular cytokines and the extracellular matrix (ECM). Astrocytomas are characterized by their diffuse nature, which is regulated by insoluble ECM components produced by the tumor cells that are largely absent from normal central nervous system tissue. The present study examined the influence of astrocytoma (C6 rat glioma) insoluble matrix components on interferon-gamma (IFN-gamma)-mediated inducible nitric-oxide synthase (iNOS) induction in microglial cells. We found that IFN-gamma-stimulated iNOS induction and nitric oxide release was greater in microglia cultured on C6 glioma cell-derived matrices compared with microglia cultured on primary rat astrocyte-derived matrices. Culture of microglia on C6 glioma cell-derived matrices also led to activation of STAT1, augmentation of IFN-gamma-induced STAT-3 activation, and an increase in IFN-gamma-activated site (GAS)-luciferase reporter activity. In addition, culture of microglia on C6 glioma cell-derived matrices activated NF-kappaB DNA binding activity and transcriptional activity. The results suggest that insoluble matrix components derived from malignant glioma cells can regulate microglia activation. These factors may include ECM components, such as fibronectin, collagen, laminin, vitronectin, and other nondiffusible compounds, and laminin seems to a critical regulator of this process. Microglia activation and subsequent brain inflammation may influence tumor growth, treatment, and metastasis. Better understanding of the regulation of microglial activation by astrocytoma-derived insoluble matrix components may be important in the development of immune-based treatment strategies against malignant brain tumors.
Collapse
Affiliation(s)
- Yoon-Jung Kim
- Department of Physiology and Biophysics, Inha University, College of Medicine, 253 Yonghyun-Dong, Nam-Ku, Incheon, Republic of Korea
| | | | | | | | | | | |
Collapse
|
46
|
Hawkins BJ, Solt LA, Chowdhury I, Kazi AS, Abid MR, Aird WC, May MJ, Foskett JK, Madesh M. G protein-coupled receptor Ca2+-linked mitochondrial reactive oxygen species are essential for endothelial/leukocyte adherence. Mol Cell Biol 2007; 27:7582-93. [PMID: 17724077 PMCID: PMC2169045 DOI: 10.1128/mcb.00493-07] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Receptor-mediated signaling is commonly associated with multiple functions, including the production of reactive oxygen species. However, whether mitochondrion-derived superoxide (mROS) contributes directly to physiological signaling is controversial. Here we demonstrate a previously unknown mechanism in which physiologic Ca(2+)-evoked mROS production plays a pivotal role in endothelial cell (EC) activation and leukocyte firm adhesion. G protein-coupled receptor (GPCR) and tyrosine kinase-mediated inositol 1,4,5-trisphosphate-dependent mitochondrial Ca(2+) uptake resulted in NADPH oxidase-independent mROS production. However, GPCR-linked mROS production did not alter mitochondrial function or trigger cell death but rather contributed to activation of NF-kappaB and leukocyte adhesion via the EC induction of intercellular adhesion molecule 1. Dismutation of mROS by manganese superoxide dismutase overexpression and a cell-permeative superoxide dismutase mimetic ablated NF-kappaB transcriptional activity and facilitated leukocyte detachment from the endothelium under simulated circulation following GPCR- but not cytokine-induced activation. These results demonstrate that mROS is the downstream effector molecule that translates receptor-mediated Ca(2+) signals into proinflammatory signaling and leukocyte/EC firm adhesion.
Collapse
Affiliation(s)
- Brian J Hawkins
- Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA 19104-6068, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chamian F, Lin SL, Lee E, Kikuchi T, Gilleaudeau P, Sullivan-Whalen M, Cardinale I, Khatcherian A, Novitskaya I, Wittkowski KM, Krueger JG, Lowes MA. Alefacept (anti-CD2) causes a selective reduction in circulating effector memory T cells (Tem) and relative preservation of central memory T cells (Tcm) in psoriasis. J Transl Med 2007; 5:27. [PMID: 17555598 PMCID: PMC1906741 DOI: 10.1186/1479-5876-5-27] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Accepted: 06/07/2007] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alefacept (anti-CD2) biological therapy selectively targets effector memory T cells (Tem) in psoriasis vulgaris, a model Type 1 autoimmune disease. METHODS Circulating leukocytes were phenotyped in patients receiving alefacept for moderate to severe psoriasis. RESULTS In all patients, this treatment caused a preferential decrease in effector memory T cells (CCR7- CD45RA-) (mean 63% reduction) for both CD4+ and CD8+ Tem, while central memory T cells (Tcm) (CCR7+CD45RA-) were less affected, and naïve T cells (CCR7+CD45RA+) were relatively spared. Circulating CD8+ effector T cells and Type 1 T cells (IFN-gamma-producing) were also significantly reduced. CONCLUSION Alefacept causes a selective reduction in circulating effector memory T cells (Tem) and relative preservation of central memory T cells (Tcm) in psoriasis.
Collapse
Affiliation(s)
- Francesca Chamian
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Shao-Lee Lin
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Edmund Lee
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Toyoko Kikuchi
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Patricia Gilleaudeau
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Mary Sullivan-Whalen
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Irma Cardinale
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Artemis Khatcherian
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Inna Novitskaya
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Knut M Wittkowski
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| | - Michelle A Lowes
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, USA
| |
Collapse
|
48
|
Greenstein JI. Current concepts of the cellular and molecular pathophysiology of multiple sclerosis. Dev Neurobiol 2007; 67:1248-65. [PMID: 17514718 DOI: 10.1002/dneu.20387] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Multiple sclerosis (MS) is the most common demyelinating disease. It poses many challenges both clinically and scientifically. Progress made in understanding the genetics, immunology, and neurobiology of MS to date has positioned the field for further breakthroughs both in understanding the etiology and pathogenesis as well as the development of rationally based therapeutics. This review will cover fundamental aspects of the clinical and pathologic features of MS. Identified genetic markers will be considered as well as the evolving understanding of immunologic and neurobiological aspects of the disease. The development of immune therapy based on this knowledge is already apparent and it is likely that neuroprotective therapies will evolve to complement immune modulation in treating the disease.
Collapse
|
49
|
Bonnekoh B, Böckelmann R, Pommer AJ, Malykh Y, Philipsen L, Gollnick H. The CD11a Binding Site of Efalizumab in Psoriatic Skin Tissue as Analyzed by Multi-Epitope Ligand Cartography Robot Technology. Skin Pharmacol Physiol 2006; 20:96-111. [PMID: 17167274 DOI: 10.1159/000097982] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Accepted: 10/04/2006] [Indexed: 11/19/2022]
Abstract
Efalizumab (Raptiva) is an immunomodulating recombinant humanized IgG1 monoclonal antibody that binds to CD11a, the alpha-subunit of leukocyte function antigen-1 (LFA-1). By blocking the binding of LFA-1 to ICAM-1, efalizumab inhibits the adhesion of leukocytes to other cell types and interferes with the migration of T lymphocytes to sites of inflammation (including psoriatic skin plaques). Analysis of the response in patients treated with efalizumab to date shows that distinct groups of responders and nonresponders to the drug exist. It would therefore be of great practical value to be able to predict which patients are most likely to respond to treatment, by identifying key parameters in the mechanism of action of efalizumab. Detailed investigation and detection of multiple epitopes in microcompartments of skin tissue has until recently been restricted by the available technology. However, the newly developed technique of Multi-Epitope Ligand Cartography (MELC) robot technology combines proteomics and biomathematical tools to visualize protein networks at the cellular and subcellular levels in situ, and to decipher cell functions. The MELC technique, which is outlined in this paper, was used to help characterize the binding of efalizumab to affected and unaffected psoriatic skin as compared to normal control skin under ex vivomodel conditions. Efalizumab was labeled with fluorescein isothiocyanate and integrated into a MELC library of more than 40 antibodies. These antibodies were selected for their potential to detect epitopes which may be indicative of (a) various cell types, (b) structural components of the extracellular matrix, or (c) the processes of cell proliferation, activation and adhesion. Efalizumab bound to CD11a in affected psoriatic skin by a factor 15x and 32x higher than in unaffected psoriatic skin and normal control skin, respectively. CD11a and the efalizumab binding site were primarily expressed in the extravascular dermis, whereas CD54 (ICAM-1) as its ligand was most prevalent in the dermal vessels. T lymphocytes (for which the markers were CD3, CD8, CD4, and CD45R0) were the major cellular targets of efalizumab. In contrast, NK cells were only a minor target of efalizumab. Our study demonstrated that efalizumab represents a treatment for psoriasis that primarily targets memory CD4+ and CD8+ T cells and has a high specificity for psoriatic disease activity. Moreover, we hereby introduce the novel principle of a biological drug-binding biochip assay being especially useful for the future monitoring of psoriatic skin lesions under efalizumab treatment conditions.
Collapse
Affiliation(s)
- B Bonnekoh
- Clinic for Dermatology and Venereology, Otto-von-Guericke-University, Magdeburg, Germany.
| | | | | | | | | | | |
Collapse
|
50
|
Abstract
Since Medawar's initial contemplations in 1953 on the mechanisms of immune evasion allowing for the survival of the allogeneic conceptus in an immunologically competent mother, physicians and immunologists alike have struggled to understand the immunological paradox of pregnancy. Ultimately, our attempts to define the immunology of normal pregnancy have broadened our appreciation of the myriad mechanisms at play that enable the promotion of implantation and maintenance of pregnancy. In this review, we summarise what is known regarding the immunology of normal pregnancy, with special emphasis on the relation to common disorders of pregnancy.
Collapse
Affiliation(s)
- Kjersti M Aagaard-Tillery
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Utah Health Sciences, 30 North 1900 East, SOM 2B200, Salt Lake City, UT 84132, USA.
| | | | | |
Collapse
|