1
|
Li X, Chen Z, Ye W, Yu J, Zhang X, Li Y, Niu Y, Ran S, Wang S, Luo Z, Zhao J, Hao Y, Zong J, Xia C, Xia J, Wu J. High-throughput CRISPR technology: a novel horizon for solid organ transplantation. Front Immunol 2024; 14:1295523. [PMID: 38239344 PMCID: PMC10794540 DOI: 10.3389/fimmu.2023.1295523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 12/12/2023] [Indexed: 01/22/2024] Open
Abstract
Organ transplantation is the gold standard therapy for end-stage organ failure. However, the shortage of available grafts and long-term graft dysfunction remain the primary barriers to organ transplantation. Exploring approaches to solve these issues is urgent, and CRISPR/Cas9-based transcriptome editing provides one potential solution. Furthermore, combining CRISPR/Cas9-based gene editing with an ex vivo organ perfusion system would enable pre-implantation transcriptome editing of grafts. How to determine effective intervention targets becomes a new problem. Fortunately, the advent of high-throughput CRISPR screening has dramatically accelerated the effective targets. This review summarizes the current advancements, utilization, and workflow of CRISPR screening in various immune and non-immune cells. It also discusses the ongoing applications of CRISPR/Cas-based gene editing in transplantation and the prospective applications of CRISPR screening in solid organ transplantation.
Collapse
Affiliation(s)
- Xiaohan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weicong Ye
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jizhang Yu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Zhang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuan Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqing Niu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuan Ran
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zilong Luo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiulu Zhao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanglin Hao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junjie Zong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengkun Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahong Xia
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission (NHC) Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Jie Wu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, National Health Commission (NHC) Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
2
|
Chang T, Yang J, Deng H, Chen D, Yang X, Tang ZH. Depletion and Dysfunction of Dendritic Cells: Understanding SARS-CoV-2 Infection. Front Immunol 2022; 13:843342. [PMID: 35265087 PMCID: PMC8898834 DOI: 10.3389/fimmu.2022.843342] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 01/31/2022] [Indexed: 12/15/2022] Open
Abstract
Uncontrolled severe acute respiratory syndrome-coronavirus (SARS-CoV)-2 infection is closely related to disorders of the innate immune and delayed adaptive immune systems. Dendritic cells (DCs) “bridge” innate immunity and adaptive immunity. DCs have important roles in defending against SARS-CoV-2 infection. In this review, we summarize the latest research concerning the role of DCs in SARS-CoV-2 infection. We focus on the complex interplay between DCs and SARS-CoV-2: pyroptosis-induced activation; activation of the renin–angiotensin–aldosterone system; and activation of dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin. We also discuss the decline in DC number, the impaired antigen-presentation capability, and the reduced production of type-I interferon of DCs in severe SARS-CoV-2 infection. In addition, we discuss the potential mechanisms for pathological activation of DCs to understand the pattern of SARS-CoV-2 infection. Lastly, we provide a brief overview of novel vaccination and immunotherapy strategies based on DC targeting to overcome SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Teding Chang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji, China
| | - Jingzhi Yang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji, China
| | - Hai Deng
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji, China
| | - Deng Chen
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji, China
| | - XiangPing Yang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhao-Hui Tang
- Division of Trauma & Surgical Critical Care, Department of Surgery, Tongji Hospital, Tongji, China
| |
Collapse
|
3
|
Chen Y, Huang J, Xu C. Lipopolysaccharide-induced DC-SIGN/TLR4 crosstalk activates NLRP3 inflammasomes via MyD88-independent signaling in gastric epithelial cells. Exp Cell Res 2020; 396:112292. [PMID: 32961144 DOI: 10.1016/j.yexcr.2020.112292] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/03/2020] [Accepted: 09/15/2020] [Indexed: 12/28/2022]
Abstract
Abnormal pattern recognition receptor (PRR) signaling plays an important role in gastric mucosal damage caused by stomach microbiota; however, the underlying molecular mechanisms remain obscure. Here, we show that DC-SIGN, a surface phenotype marker of dendritic cells, is overexpressed in gastric epithelial cells facing LPS stimulation. NLRP3 expression in gastric epithelial cells are significantly increased and related to the degree of LPS stimulation. Furthermore, DC-SIGN could interact with TLR4, promote NLRP3 and related genes expression via MyD88-independent signaling pathway and regulate the secretion of IL-1β and IL-18 in gastric epithelial cells. The results of flow cytometry analysis show that DC-SIGN primarily mediates Th1 differentiation when co-cultured with gastric epithelial cells. These results reveal that LPS-induced DC-SIGN expression modulates NLRP3 inflammasomes formation via MyD88-independent TLR4 signaling in gastric epithelial cell, and induces a Th1-predominant host immune response,these findings may indicate a new function of DC-SIGN in non-immune cells, and elucidate the diversity role of gastric epithelial cells in mechanism of immune damage caused by microbial flora.
Collapse
Affiliation(s)
- Yufan Chen
- Department of Pediatric Neurosurgery, Children's Hospital of Fudan University, 399 Wan Yuan Road, Shanghai, 201102, China
| | - Jiebin Huang
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Rd. II, Shanghai, 200025, China
| | - Chundi Xu
- Department of Pediatrics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Rd. II, Shanghai, 200025, China.
| |
Collapse
|
4
|
De Laere M, Berneman ZN, Cools N. To the Brain and Back: Migratory Paths of Dendritic Cells in Multiple Sclerosis. J Neuropathol Exp Neurol 2019; 77:178-192. [PMID: 29342287 PMCID: PMC5901086 DOI: 10.1093/jnen/nlx114] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Migration of dendritic cells (DC) to the central nervous system (CNS) is a critical event in the pathogenesis of multiple sclerosis (MS). While up until now, research has mainly focused on the transmigration of DC through the blood-brain barrier, experimental evidence points out that also the choroid plexus and meningeal vessels represent important gateways to the CNS, especially in early disease stages. On the other hand, DC can exit the CNS to maintain immunological tolerance to patterns expressed in the CNS, a process that is perturbed in MS. Targeting trafficking of immune cells, including DC, to the CNS has demonstrated to be a successful strategy to treat MS. However, this approach is known to compromise protective immune surveillance of the brain. Unravelling the migratory paths of regulatory and pathogenic DC within the CNS may ultimately lead to the design of new therapeutic strategies able to selectively interfere with the recruitment of pathogenic DC to the CNS, while leaving host protective mechanisms intact.
Collapse
Affiliation(s)
- Maxime De Laere
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp
| |
Collapse
|
5
|
Inflammation research sails through the sea of immunology to reach immunometabolism. Int Immunopharmacol 2019; 73:128-145. [PMID: 31096130 DOI: 10.1016/j.intimp.2019.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/18/2019] [Accepted: 05/01/2019] [Indexed: 02/08/2023]
Abstract
Inflammation occurs as a result of acute trauma, invasion of the host by different pathogens, pathogen-associated molecular patterns (PAMPs) or chronic cellular stress generating damage-associated molecular patterns (DAMPs). Thus inflammation may occur under both sterile inflammatory conditions including certain cancers, autoimmune or autoinflammatory diseases (Rheumatic arthritis (RA)) and infectious diseases including sepsis, pneumonia-associated acute lung inflammation (ALI) or acute respiratory distress syndrome (ARDS). The pathogenesis of inflammation involves dysregulation of an otherwise protective immune response comprising of various innate and adaptive immune cells and humoral (cytokines and chemokines) mediators secreted by these immune cells upon the activation of signaling mechanisms regulated by the activation of different pattern recognition receptors (PRRs). However, the pro-inflammatory and anti-inflammatory action of these immune cells is determined by the metabolic stage of the immune cells. The metabolic process of immune cells is called immunometabolism and its shift determined by inflammatory stimuli is called immunometabolic reprogramming. The article focuses on the involvement of various immune cells generating the inflammation, their interaction, immunometabolic reprogramming, and the therapeutic targeting of the immunometabolism to manage inflammation.
Collapse
|
6
|
Wang X, Jiang Y, Yuan M, Chen C, Wang K, Zhang Q, Zuo Y, Ren S. Overexpression of dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin-related protein in cervical cancer and correlation with squamous cell carcinoma antigen. Oncol Lett 2017; 14:2813-2821. [PMID: 28927040 PMCID: PMC5588121 DOI: 10.3892/ol.2017.6508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 01/17/2017] [Indexed: 01/23/2023] Open
Abstract
Dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin-related protein (DC-SIGNR) is a type II transmembrane protein that has been reported to bind to various pathogens and participate in immunoregulation and tumorigenesis. However, further research is required to investigate whether the level of DC-SIGNR and cervical cancer are associated. The present study aimed to explore the clinical diagnostic significance of DC-SIGNR in cervical cancer. Immunohistochemical staining of DC-SIGNR was performed in samples from 25 patients with early stage cervical cancer, 14 patients with cervical intraepithelial neoplasia (CIN) and cervical polyp samples from 15 individuals. DC-SIGNR expression in cervical cancer tissue was significantly higher compared with that in CIN and cervical polyp tissue (P=0.0184 and P=0.0236, respectively). However, there was no significant difference in DC-SIGNR expression between CIN and cervical polyp tissue (P=0.8103). Additionally, the serum DC-SIGNR levels in 84 cervical cancer patients and 69 healthy female individuals were measured using an ELISA. Serum (s)DC-SIGNR levels were significantly higher in cervical cancer patients compared with healthy female individuals (P<0.0001). A sDC-SIGNR level of 93.7 ng/ml was revealed by receiver operating characteristic curve analysis to predict the presence of cervical cancer with 69.57% sensitivity and 66.67% specificity (area under the curve, 0.6989; P<0.0001). Levels of sDC-SIGNR in cervical cancer patients were also correlated with serum levels of squamous cell carcinoma antigen (r=0.2583; P=0.0348). The results of the present study demonstrate that DC-SIGNR is overexpressed in cervical cancer tissue, and suggest that DC-SIGNR could serve as a biomarker for the early diagnosis of cervical cancer. Nevertheless, further studies are required to demonstrate what role DC-SIGNR serves in cervical cancer.
Collapse
Affiliation(s)
- Xiangdong Wang
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Yangmei Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Menglang Yuan
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Chunlin Chen
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Keyong Wang
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Qianshi Zhang
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| | - Yunfei Zuo
- Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Shuangyi Ren
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116023, P.R. China
| |
Collapse
|
7
|
Dodagatta-Marri E, Mitchell DA, Pandit H, Sonawani A, Murugaiah V, Idicula-Thomas S, Nal B, Al-Mozaini MM, Kaur A, Madan T, Kishore U. Protein-Protein Interaction between Surfactant Protein D and DC-SIGN via C-Type Lectin Domain Can Suppress HIV-1 Transfer. Front Immunol 2017; 8:834. [PMID: 28824609 PMCID: PMC5534670 DOI: 10.3389/fimmu.2017.00834] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 07/03/2017] [Indexed: 01/01/2023] Open
Abstract
Surfactant protein D (SP-D) is a soluble C-type lectin, belonging to the collectin (collagen-containing calcium-dependent lectin) family, which acts as an innate immune pattern recognition molecule in the lungs at other mucosal surfaces. Immune regulation and surfactant homeostasis are salient functions of SP-D. SP-D can bind to a range of viral, bacterial, and fungal pathogens and trigger clearance mechanisms. SP-D binds to gp120, the envelope protein expressed on HIV-1, through its C-type lectin or carbohydrate recognition domain. This is of importance since SP-D is secreted by human mucosal epithelial cells and is present in the female reproductive tract, including vagina. Another C-type lectin, dendritic cell (DC)-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), present on the surface of the DCs, also binds to HIV-1 gp120 and facilitates viral transfer to the lymphoid tissues. DCs are also present at the site of HIV-1 entry, embedded in vaginal or rectal mucosa. In the present study, we report a direct protein-protein interaction between recombinant forms of SP-D (rfhSP-D) and DC-SIGN via their C-type lectin domains. Both SP-D and DC-SIGN competed for binding to immobilized HIV-1 gp120. Pre-incubation of human embryonic kidney cells expressing surface DC-SIGN with rfhSP-D significantly inhibited the HIV-1 transfer to activated peripheral blood mononuclear cells. In silico analysis revealed that SP-D and gp120 may occupy same sites on DC-SIGN, which may explain the reduced transfer of HIV-1. In summary, we demonstrate, for the first time, that DC-SIGN is a novel binding partner of SP-D, and this interaction can modulate HIV-1 capture and transfer to CD4+ T cells. In addition, the present study also reveals a novel and distinct mechanism of host defense by SP-D against HIV-1.
Collapse
Affiliation(s)
- Eswari Dodagatta-Marri
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Daniel A Mitchell
- Clinical Sciences Research Laboratories, Warwick Medical School, University Hospital Coventry and Warwickshire Campus, Coventry, United Kingdom
| | - Hrishikesh Pandit
- Department of Innate Immunity, National Institute for Research in Reproductive Health, Indian Council of Medical Research, Mumbai, India
| | - Archana Sonawani
- Department of Innate Immunity, National Institute for Research in Reproductive Health, Indian Council of Medical Research, Mumbai, India
| | - Valarmathy Murugaiah
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Susan Idicula-Thomas
- Department of Innate Immunity, National Institute for Research in Reproductive Health, Indian Council of Medical Research, Mumbai, India
| | - Béatrice Nal
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom.,Institute of Environment, Health and Societies, Brunel University London, Uxbridge, United Kingdom
| | - Maha M Al-Mozaini
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Anuvinder Kaur
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Taruna Madan
- Department of Innate Immunity, National Institute for Research in Reproductive Health, Indian Council of Medical Research, Mumbai, India
| | - Uday Kishore
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| |
Collapse
|
8
|
Abstract
Tuberculosis remains one of the greatest threats to human health. The causative bacterium, Mycobacterium tuberculosis, is acquired by the respiratory route. It is exquisitely adapted to humans and is a prototypic intracellular pathogen of macrophages, with alveolar macrophages being the primary conduit of infection and disease. However, M. tuberculosis bacilli interact with and are affected by several soluble and cellular components of the innate immune system which dictate the outcome of primary infection, most commonly a latently infected healthy human host, in whom the bacteria are held in check by the host immune response within the confines of tissue granuloma, the host histopathologic hallmark. Such individuals can develop active TB later in life with impairment in the immune system. In contrast, in a minority of infected individuals, the early host immune response fails to control bacterial growth, and progressive granulomatous disease develops, facilitating spread of the bacilli via infectious aerosols. The molecular details of the M. tuberculosis-host innate immune system interaction continue to be elucidated, particularly those occurring within the lung. However, it is clear that a number of complex processes are involved at the different stages of infection that may benefit either the bacterium or the host. In this article, we describe a contemporary view of the molecular events underlying the interaction between M. tuberculosis and a variety of cellular and soluble components and processes of the innate immune system.
Collapse
|
9
|
Johansson P, Wang T, Collet B, Corripio-Miyar Y, Monte MM, Secombes CJ, Zou J. Identification and expression modulation of a C-type lectin domain family 4 homologue that is highly expressed in monocytes/macrophages in rainbow trout (Oncorhynchus mykiss). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2016; 54:55-65. [PMID: 26279216 DOI: 10.1016/j.dci.2015.08.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 08/08/2015] [Accepted: 08/09/2015] [Indexed: 06/04/2023]
Abstract
The C-type lectin domain containing (CLEC) receptors including CD209 are expressed in vivo by monocytes, monocyte-derived macrophages and dendritic cells and by in vitro generated monocyte-derived cells. This paper reports the cloning and sequencing of a lectin molecule, CLEC4T1, in rainbow trout that is a homologue of the CLEC4 family. The expression pattern of the CLEC4T1 was investigated in vivo after infection with a bacterial pathogen and in cultured macrophages after modulation with microbial mimics. Trout CLEC4T1 was highly expressed in spleen and head kidney following infection with Yersinia ruckeri. Expression could also be induced in macrophage cultures by LPS but not by Poly I:C, and suggests that the regulation of CLEC4T1 expression in trout varies according to the nature of the stimulant. A polyclonal CLEC4T1 antibody was generated and validated by Western blotting for use in evaluation of CLEC4T1(+) cells by flow cytometry analysis. Freshly isolated adherent trout head kidney cultures, potentially containing macrophages and dendritic cell precursors, showed an increase of CLEC4T1(+) cells (assessed by flow cytometry) upon stimulation with recombinant interleukin-4/13A. The results suggest that CLEC4T1 is a useful marker for further characterisation of monocyte derived antigen presenting cells in fish.
Collapse
Affiliation(s)
- Petronella Johansson
- Scottish Fish Immunology Research Centre (SFIRC), School of Biological Sciences, University of Aberdeen, Zoology Building, Tillydrone Avenue, Aberdeen, AB24 2TZ, Scotland, UK; Marine Scotland - Science, Marine Laboratory, Victoria Road, Aberdeen, AB11 9DB, Scotland, UK
| | - Tiehui Wang
- Scottish Fish Immunology Research Centre (SFIRC), School of Biological Sciences, University of Aberdeen, Zoology Building, Tillydrone Avenue, Aberdeen, AB24 2TZ, Scotland, UK
| | - Bertrand Collet
- Marine Scotland - Science, Marine Laboratory, Victoria Road, Aberdeen, AB11 9DB, Scotland, UK
| | - Yolanda Corripio-Miyar
- Scottish Fish Immunology Research Centre (SFIRC), School of Biological Sciences, University of Aberdeen, Zoology Building, Tillydrone Avenue, Aberdeen, AB24 2TZ, Scotland, UK
| | - Milena M Monte
- Scottish Fish Immunology Research Centre (SFIRC), School of Biological Sciences, University of Aberdeen, Zoology Building, Tillydrone Avenue, Aberdeen, AB24 2TZ, Scotland, UK
| | - Christopher J Secombes
- Scottish Fish Immunology Research Centre (SFIRC), School of Biological Sciences, University of Aberdeen, Zoology Building, Tillydrone Avenue, Aberdeen, AB24 2TZ, Scotland, UK.
| | - Jun Zou
- Scottish Fish Immunology Research Centre (SFIRC), School of Biological Sciences, University of Aberdeen, Zoology Building, Tillydrone Avenue, Aberdeen, AB24 2TZ, Scotland, UK
| |
Collapse
|
10
|
Ding D, Chen W, Zhang C, Chen Z, Jiang Y, Yang Z, Jiang X, Zuo Y, Ren S. Low expression of dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin in non-Hodgkin lymphoma and a significant correlation with β2-microglobulin. Med Oncol 2014; 31:202. [PMID: 25182705 DOI: 10.1007/s12032-014-0202-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Accepted: 08/22/2014] [Indexed: 12/22/2022]
Abstract
Dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN), a member of the C-type lectin superfamily, has been reported to bind to various pathogens and several tumor cells and to participate in immunoregulation. It is still unclear whether there is a significant association between the level of DC-SIGN and non-Hodgkin lymphoma (NHL). To investigate the clinical diagnostic significance of DC-SIGN in NHL, we conducted a study with 52 NHL patients and 104 healthy individuals. Enzyme-linked immunosorbent assay and tissue microarray technology were utilized for the analysis. The serum sDC-SIGN levels in the NHL patients were significantly lower than those in the healthy controls (P=0.0019). A cutoff value of 1.499 µg/ml for sDC-SIGN predicted the presence of NHL with 78.85% sensitivity and 53.85% specificity [area under the curve (AUC)=0.6531, P=0.0019]. The serum sDC-SIGN levels in NHL patients were also significantly correlated with β2-microglobulin (P=0.0062). Moreover, tissue microarray analysis demonstrated that the expression of DC-SIGN in the lymph nodes or tissues of 96 NHL patients was significantly lower than that in 18 normal lymph nodes (P<0.0001). However, the expression of DC-SIGN in NHL displayed no significant correlation with the expression of CD20 or CD79a. In conclusion, DC-SIGN may be a promising biological molecule for clinical research on NHL, whereas the underlying roles need to be investigated in additional studies.
Collapse
Affiliation(s)
- Dongbing Ding
- Department of General Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, 116023, China
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Macrophages in tuberculosis: friend or foe. Semin Immunopathol 2013; 35:563-83. [PMID: 23864058 DOI: 10.1007/s00281-013-0388-2] [Citation(s) in RCA: 197] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/20/2013] [Indexed: 12/14/2022]
Abstract
Tuberculosis (TB) remains one of the greatest threats to human health. The causative bacterium, Mycobacterium tuberculosis (Mtb), is acquired by the respiratory route. It is exquisitely human adapted and a prototypic intracellular pathogen of macrophages, with alveolar macrophages (AMs) being the primary conduit of infection and disease. The outcome of primary infection is most often a latently infected healthy human host, in whom the bacteria are held in check by the host immune response. Such individuals can develop active TB later in life with impairment in the immune system. In contrast, in a minority of infected individuals, the host immune response fails to control the growth of bacilli, and progressive granulomatous disease develops, facilitating spread of the bacilli via infectious aerosols coughed out into the environment and inhaled by new hosts. The molecular details of the Mtb-macrophage interaction continue to be elucidated. However, it is clear that a number of complex processes are involved at the different stages of infection that may benefit either the bacterium or the host. Macrophages demonstrate tremendous phenotypic heterogeneity and functional plasticity which, depending on the site and stage of infection, facilitate the diverse outcomes. Moreover, host responses vary depending on the specific characteristics of the infecting Mtb strain. In this chapter, we describe a contemporary view of the behavior of AMs and their interaction with various Mtb strains in generating unique immunologic lung-specific responses.
Collapse
|
12
|
The role of lectins in allergic sensitization and allergic disease. J Allergy Clin Immunol 2013; 132:27-36. [DOI: 10.1016/j.jaci.2013.02.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Revised: 01/03/2013] [Accepted: 02/01/2013] [Indexed: 01/05/2023]
|
13
|
Bakdash G, Sittig SP, van Dijk T, Figdor CG, de Vries IJM. The nature of activatory and tolerogenic dendritic cell-derived signal II. Front Immunol 2013; 4:53. [PMID: 23450201 PMCID: PMC3584294 DOI: 10.3389/fimmu.2013.00053] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 02/11/2013] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DCs) are central in maintaining the intricate balance between immunity and tolerance by orchestrating adaptive immune responses. Being the most potent antigen presenting cells, DCs are capable of educating naïve T cells into a wide variety of effector cells ranging from immunogenic CD4+ T helper cells and cytotoxic CD8+ T cells to tolerogenic regulatory T cells. This education is based on three fundamental signals. Signal I, which is mediated by antigen/major histocompatibility complexes binding to antigen-specific T cell receptors, guarantees antigen specificity. The co-stimulatory signal II, mediated by B7 family molecules, is crucial for the expansion of the antigen-specific T cells. The final step is T cell polarization by signal III, which is conveyed by DC-derived cytokines and determines the effector functions of the emerging T cell. Although co-stimulation is widely recognized to result from the engagement of T cell-derived CD28 with DC-expressed B7 molecules (CD80/CD86), other co-stimulatory pathways have been identified. These pathways can be divided into two groups based on their impact on primed T cells. Whereas pathways delivering activatory signals to T cells are termed co-stimulatory pathways, pathways delivering tolerogenic signals to T cells are termed co-inhibitory pathways. In this review, we discuss how the nature of DC-derived signal II determines the quality of ensuing T cell responses and eventually promoting either immunity or tolerance. A thorough understanding of this process is instrumental in determining the underlying mechanism of disorders demonstrating distorted immunity/tolerance balance, and would help innovating new therapeutic approaches for such disorders.
Collapse
Affiliation(s)
- Ghaith Bakdash
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre Nijmegen, Netherlands
| | | | | | | | | |
Collapse
|
14
|
Ma WY, Zhuang L, Qi QC, Sun Q. Expression of dendritic cell lysosome-associated membrane protein and dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin in condyloma acuminatum lesions. J Int Med Res 2013; 41:138-45. [PMID: 23569139 DOI: 10.1177/0300060513476991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE Retrospective, observational study to explore the role of dendritic cells (DCs) in condyloma acuminatum lesions (genital warts) and their relationship with duration of the disease. METHODS Condyloma acuminatum lesion samples were collected from male patients with the condition and compared with normal foreskin samples from male volunteers. Cellular locations of dendritic cell lysosome-associated membrane protein (DC-LAMP) and dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin (DC-SIGN) were detected using immunohistochemistry. Levels of both proteins were determined using Western blot analysis; levels of their corresponding mRNAs were measured using reverse transcription-polymerase chain reaction. RESULTS The mRNA and protein levels of DC-LAMP and DC-SIGN were both significantly higher in condyloma acuminatum lesions (n = 30 samples) compared with normal skin samples (n = 13). Levels of DC-LAMP and DC-SIGN protein and duration of disease were inversely correlated. CONCLUSIONS DC-LAMP and DC-SIGN may be involved in the pathogenesis of condyloma acuminatum. Their levels were inversely correlated with the duration of disease, suggesting that DCs might be involved in human papillomavirus clearance.
Collapse
Affiliation(s)
- Wei-Yuan Ma
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan, Shandong Province, China
| | | | | | | |
Collapse
|
15
|
Sun J, Luo Y, Tian Z, Gu L, Xia SC, Yu Y. Expression of ERBB3 binding protein 1 (EBP1) in salivary adenoid cystic carcinoma and its clinicopathological relevance. BMC Cancer 2012; 12:499. [PMID: 23110497 PMCID: PMC3499390 DOI: 10.1186/1471-2407-12-499] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 10/18/2012] [Indexed: 11/23/2022] Open
Abstract
Background ERBB3 binding protein 1 (EBP1) gene transfer into human salivary adenoid cystic carcinoma cells has been shown to significantly inhibit cell proliferation and reduce tumor metastasis in mouse models. In the current study, to evaluate if EBP1 is a novel biomarker capable of identifying patients at higher risk of disease progression and recurrence, we examined the EBP1 expression profile in adenoid cystic carcinoma (ACC) patients and analyzed its clinicopathological relevance. To understand the underlying anti-metastatic mechanism, we investigated if EBP1 regulates invasion-related molecules. Methods We performed immunohistochemical analysis on 132 primary adenoid cystic carcinoma and adjacent non-cancerous tissues using commercial EBP1, MMP9, E-cadherin and ICAM-1 antibodies. Results were correlated to clinicopathological parameters, long-term survival and invasion-related molecules by statistical analysis. Cell motility and invasiveness of vector or wild-type EBP1-transfected ACC-M cell lines were evaluated using wound healing and Boyden chamber assays. MMP9, E-cadherin and ICAM-1 proteins in these cell lines were detected using western blot assay. Results The expression of EBP1 was significantly higher in non-cancerous adjacent tissues compared with corresponding cancer tissues. The intensity and percentage of cells that reacted with EBP1 antibodies were significantly higher in cases with tubular pattern than those with solid pattern (P<0.0001). We also found adenoid cystic carcinoma with local lymphatic metastasis had significantly lower EBP1 expression than ACC with no local lymphatic node metastasis (P<0.0001). Similar findings were observed in ACC with lung metastasis compared with cases with no lung metastasis (P<0.0001), in particular, in cases with perineural invasion compared with cases with no perineural invasion (P<0.0001). Furthermore, a decrease in EBP1 expression was positively associated with a reduction in overall survival of ACC patients. Of note, EBP1 inhibits migration and invasiveness of ACC cells by upregulating E-cadherin but downregulating MMP9. In clinical adenoid cystic carcinoma patients, higher EBP1 expression was positively correlated with E-cadherin levels (P<0.001) but negatively correlated with MMP9 expression (P=0.0002). Conclusions EBP1 expression is reduced in adenoid cystic carcinoma, indicating unfavorable prognosis of ACC patients. Its regulation of MMP9 and E-cadherin protein levels suggests a critical therapeutic potential.
Collapse
Affiliation(s)
- Jian Sun
- Department of Stomatology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | | | | | | | | | | |
Collapse
|
16
|
Raghavan S, Alagarasu K, Selvaraj P. Immunogenetics of HIV and HIV associated tuberculosis. Tuberculosis (Edinb) 2012; 92:18-30. [PMID: 21943869 DOI: 10.1016/j.tube.2011.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 08/08/2011] [Indexed: 11/28/2022]
Affiliation(s)
- S Raghavan
- Department of Immunology, Tuberculosis Research Centre (ICMR), Mayor V.R. Ramanathan Road, Chetput, Chennai 600031, India
| | | | | |
Collapse
|
17
|
Abstract
In the immune system, C-type lectins and CTLDs have been shown to act both as adhesion and as pathogen recognition receptors. The Dendritic cell-specific ICAM-3 grabbing non-integrin (DC-SIGN) and its homologs in human and mouse represent an important C-type lectin family. DC-SIGN contains a lectin domain that recognizes in a Ca2+-dependent manner carbohydrates such as mannose-containing structures present on glycoproteins such as ICAM-2 and ICAM-3. DC-SIGN is a prototype C-type lectin organized in microdomains, which have their role as pathogen recognition receptors in sensing microbes. Although the integrin LFA-1 is a counter-receptor for both ICAM-2 and ICAM-3 on DC, DC-SIGN is the high affinity adhesion receptor for ICAM-2/-3. While cell–cell contact is a primary function of selectins, collectins are specialized in recognition of pathogens. Interestingly, DC-SIGN is a cell adhesion receptor as well as a pathogen recognition receptor. As adhesion receptor, DC-SIGN mediates the contact between dendritic cells (DCs) and T lymphocytes, by binding to ICAM-3, and mediates rolling of DCs on endothelium, by interacting with ICAM-2. As pathogen receptor, DC-SIGN recognizes a variety of microorganisms, including viruses, bacteria, fungi and several parasites (Cambi et al. 2005). The natural ligands of DC-SIGN consist of mannose oligosaccharides or fucose-containing Lewis-type determinants. In this chapter, we shall focus on the structure and functions of DC-SIGN and related CTLDs in the recognition of pathogens, the molecular and structural determinants that regulate the interaction with pathogen-associated molecular patterns. The heterogeneity of carbohydrate residues exposed on cellular proteins and pathogens regulates specific binding of DC-expressed C-type lectins that contribute to the diversity of immune responses created by DCs (van Kooyk et al. 2003a; Cambi et al. 2005).
Collapse
|
18
|
Liao CF, Wang SF, Lin YT, Ho DD, Chen YMA. Identification of the DC-SIGN-interactive domains on the envelope glycoprotein of HIV-1 CRF07_BC. AIDS Res Hum Retroviruses 2011; 27:831-9. [PMID: 21142800 DOI: 10.1089/aid.2010.0215] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
DC-SIGN, a C-type lection expressed on dendritic cells, enhances HIV-1 infection in cis and in trans. HIV-1 circulating recombinant form (CRF) 07_BC viruses have been the predominant strain found among injection drug users in southern China and Taiwan. The goal of this study was to map the DC-SIGN-interactive domain on the gp120 of CRF07_BC. Pseudotyped viruses containing single (N233Q, N275Q, N330Q, N351Q, N355Q, N381Q, and N387Q), double (N233Q + N275Q, N233Q + N351Q, N275Q + N351Q), or triple (N233Q + N275Q + N351Q) N-glycan mutant gp120 were generated. Capture assays showed that the DC-SIGN-binding capacity of pseudoviruses with N275Q or N351Q decreased significantly. Rabbit antisera against synthetic peptides covering the N275 (R72 antiserum) or N351 (R77 antiserum) region blocked the interaction between wild-type gp120 and DC-SIGN in the capture assay. Furthermore, pseudotype viruses containing gp120 from five different CRF07_BC isolates were generated and R72 and R77 antisera blocked their interactions with DC-SIGN (80% for R72 and 40% for R77, respectively) in the capture assays. In conclusion, the N275 and N351 glycan sites on the CRF07_BC gp120 play an important role in mediating the interaction between gp120 and DC-SIGN. This information is valuable for developing both therapeutic and preventive agents for HIV-1 infection.
Collapse
Affiliation(s)
- Che-Feng Liao
- AIDS Prevention and Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - Sheng-Fan Wang
- AIDS Prevention and Research Center, National Yang-Ming University, Taipei, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei, Taiwan
- Institude of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yu-Ting Lin
- AIDS Prevention and Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
| | - David D. Ho
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, New York
| | - Yi-Ming Arthur Chen
- AIDS Prevention and Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Public Health, National Yang-Ming University, Taipei, Taiwan
- Department and Institute of Microbiology and Immunology, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
19
|
Weber W, Weber E, Geisse S, Memmert K. Optimisation of protein expression and establishment of the Wave Bioreactor for Baculovirus/insect cell culture. Cytotechnology 2011; 38:77-85. [PMID: 19003089 DOI: 10.1023/a:1021102015070] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
As the interest of research is beginning to shift from genomicsto proteomics the number of proteins to be expressed is rapidlyincreasing. To do so, well-established, high-level expressionsystems and rapid, cost-effective production means are needed. For addressing the latter, a novel cultivation system for recombinant cells, the Wave Bioreactortrade mark has recently becomeavailable. We describe the set-up and the optimisation of parameters essential for successful operation and growth of insect cells to high cell densities in the Wave Bioreactor. According to our experience, the Cellbagtrade mark system comparesvery favorably to conventional cultivation vessels such as bioreactors and roller cultures with respect to simplicity ofoperation and cost. Additionally, we developed a rapid and simple protocol for assessing expression and production conditions for the Baculovirus/insect cell system applicable to many different genes/proteins. Important parameters like MOI,TOI, peak cell density (PCD) and expression levels are determinedin pre-experiments on small scale to achieve optimal expressionof a given protein. These conditions are subsequently transformedand applied to large scale cultures grown in nutrient-supplemented medium in the Wave Bioreactor.
Collapse
Affiliation(s)
- Wilfried Weber
- Core Technologies/Biomolecules Production, Novartis Pharma AG, Research, Building WSJ-506.101, CH-4002, Basel, Switzerland
| | | | | | | |
Collapse
|
20
|
Avota E, Gulbins E, Schneider-Schaulies S. DC-SIGN mediated sphingomyelinase-activation and ceramide generation is essential for enhancement of viral uptake in dendritic cells. PLoS Pathog 2011; 7:e1001290. [PMID: 21379338 PMCID: PMC3040670 DOI: 10.1371/journal.ppat.1001290] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 01/12/2011] [Indexed: 11/23/2022] Open
Abstract
As pattern recognition receptor on dendritic cells (DCs), DC-SIGN binds carbohydrate structures on its pathogen ligands and essentially determines host pathogen interactions because it both skews T cell responses and enhances pathogen uptake for cis infection and/or T cell trans-infection. How these processes are initiated at the plasma membrane level is poorly understood. We now show that DC-SIGN ligation on DCs by antibodies, mannan or measles virus (MV) causes rapid activation of neutral and acid sphingomyelinases followed by accumulation of ceramides in the outer membrane leaflet. SMase activation is important in promoting DC-SIGN signaling, but also for enhancement of MV uptake into DCs. DC-SIGN-dependent SMase activation induces efficient, transient recruitment of CD150, which functions both as MV uptake receptor and microbial sensor, from an intracellular Lamp-1+ storage compartment shared with acid sphingomyelinase (ASM) within a few minutes. Subsequently, CD150 is displayed at the cell surface and co-clusters with DC-SIGN. Thus, DC-SIGN ligation initiates SMase-dependent formation of ceramide-enriched membrane microdomains which promote vertical segregation of CD150 from intracellular storage compartments along with ASM. Given the ability to promote receptor and signalosome co-segration into (or exclusion from) ceramide enriched microdomains which provide a favorable environment for membrane fusion, DC-SIGN-dependent SMase activation may be of general importance for modes and efficiency of pathogen uptake into DCs, and their routing to specific compartments, but also for modulating T cell responses. Dendritic cells (DCs) bear receptors specialized on recognition of patterns specific to pathogens (such as carbohydrates), which can either promote functional activation of these cells (such as TLRs), which renders them capable of efficiently presenting antigens to T cells, or, as DC-SIGN, endocytic uptake as essential for loading MHC molecules. Viruses such as HIV and measles virus (MV) exploit DC-SIGN for both their uptake into DCs and modulation of TLR signaling, yet how this is mechanistically exerted is poorly understood. We now show that DC-SIGN activates sphingomyelinases (SMases) which convert their sphingomyelin substrate into ceramides, thereby catalysing the formation of membrane platforms able to recruit and concentrate receptors and associated signaling components. We found DC-SIGN-dependent SMase activation as essential for DC-SIGN and thereby modulation of TLR signaling, but also for enhancement of MV uptake. This is mediated by a fast, transient recruitment of its entry receptor, CD150, from an intracellular storage compartment to the cell surface where it co-clusters in ceramide enriched platforms with DC-SIGN. The ability to segregate viral receptors into (or exclude them from) membrane microdomains, which, based on their biophysical properties, facilitate membrane fusion, proposes DC-SIGN-mediated SMAse activation as a central regulator of pathogen uptake into DCs.
Collapse
MESH Headings
- Antigen Presentation
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Blotting, Western
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cells, Cultured
- Ceramides/metabolism
- Dendritic Cells/metabolism
- Dendritic Cells/virology
- Flow Cytometry
- Humans
- Immunoprecipitation
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Measles
- Measles virus/genetics
- Measles virus/growth & development
- Measles virus/immunology
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Virus/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signaling Lymphocytic Activation Molecule Family Member 1
- Sphingomyelin Phosphodiesterase/metabolism
- T-Lymphocytes/virology
Collapse
Affiliation(s)
- Elita Avota
- Institute for Virology and Immunobiology, University of Würzburg, Wuerzburg, Germany
| | - Erich Gulbins
- Department of Molecular Medicine, University of Essen, Essen, Germany
| | | |
Collapse
|
21
|
Rojas D, Krishnan R. IFN-γ generates maturation-arrested dendritic cells that induce T cell hyporesponsiveness independent of Foxp3+ T-regulatory cell generation. Immunol Lett 2010; 132:31-7. [DOI: 10.1016/j.imlet.2010.05.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Revised: 05/13/2010] [Accepted: 05/14/2010] [Indexed: 12/13/2022]
|
22
|
Abstract
Dendritic cell-specific ICAM-3-grabbing nonintegrin (DC-SIGN). DC-SIGN is a C-type lectin receptor that recognizes N-linked high-mannose oligosaccharides and branched fucosylated structures. It is now clear that the biological role of DC-SIGN is two-fold. It is primarily expressed by dendritic cells and mediates important functions necessary for the induction of successful immune responses that are essential for the clearance of microbial infections, such as the capture, destruction, and presentation of microbial pathogens to induce successful immune responses. Yet, on the other hand, pathogens may also exploit DC-SIGN to modulate DC functioning thereby skewing the immune response and promoting their own survival. This chapter presents an overview of the structure of DC-SIGN and its expression pattern among immune cells. The current state of knowledge of DC-SIGN-carbohydrate interactions is discussed and how these interactions influence dendritic cell functioning is examined. The molecular aspects that underlie the selectivity of DC-SIGN for mannose-and fucose-containing carbohydrates are detailed. Furthermore, the chapter discusses the role of DC-SIGN in dendritic cell biology and how certain bacterial pathogens exploit DC-SIGN to escape immune surveillance.
Collapse
|
23
|
Sierra-Filardi E, Estecha A, Samaniego R, Fernández-Ruiz E, Colmenares M, Sánchez-Mateos P, Steinman RM, Granelli-Piperno A, Corbí AL. Epitope mapping on the dendritic cell-specific ICAM-3-grabbing non-integrin (DC-SIGN) pathogen-attachment factor. Mol Immunol 2010; 47:840-8. [DOI: 10.1016/j.molimm.2009.09.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2009] [Revised: 09/20/2009] [Accepted: 09/30/2009] [Indexed: 12/26/2022]
|
24
|
Liu Z, Guo B, Lopez RD. Expression of intercellular adhesion molecule (ICAM)-1 or ICAM-2 is critical in determining sensitivity of pancreatic cancer cells to cytolysis by human gammadelta-T cells: implications in the design of gammadelta-T-cell-based immunotherapies for pancreatic cancer. J Gastroenterol Hepatol 2009; 24:900-11. [PMID: 19175829 DOI: 10.1111/j.1440-1746.2008.05668.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS gammadelta-T cells can recognize and kill malignant cells, particularly those of epithelial origin, through mechanisms which do not require the recognition of tumor-specific antigens (innate immune response). This natural ability of gammadelta-T cells to kill tumor cells in a tumor antigen-independent manner provides a strong rationale for developing clinical trials designed to exploit the innate antitumor properties of gammadelta-T cells. METHODS In vitro studies were carried out to asses the sensitivity of pancreatic cancer cells (MIA PaCa2, BxPC-3, PANC-1) to killing by ex vivo expanded human gammadelta-T cells. RESULTS The capacity of gammadelta-T cells to bind to as well as to kill pancreatic cancer cells correlated with the degree of surface expression of key intercellular adhesion molecules (ICAM) present on pancreatic cancer cells. Moreover, pancreatic cancer cells expressing neither ICAM-1 nor ICAM-2 were bound poorly by gammadelta-T cells and were found to be resistant to gammadelta-T-cell killing. However, upon transfection of resistant cells with ICAM-1 or ICAM-2, gammadelta-T cells were then able to bind to and subsequently kill these cells. CONCLUSION In vitro, the expression of ICAM-1 or ICAM-2 on human pancreatic cancer cells is critically important in determining the extent to which these cells are sensitive to killing by human gammadelta-T cells. Accordingly, in ongoing and future clinical studies using gammadelta-T cells for the treatment of a variety of epithelial-derived solid tumors-including pancreatic cancer-interventions intended to modulate ICAM expression on tumor cells may become important adjuncts to gammadelta-T-cell-based immunotherapies.
Collapse
Affiliation(s)
- Zhiyong Liu
- Division of Hematology and Oncology, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | |
Collapse
|
25
|
Huang Y, Dryman B, Li W, Meng X. Porcine DC-SIGN: molecular cloning, gene structure, tissue distribution and binding characteristics. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2009; 33:464-480. [PMID: 18951915 PMCID: PMC7103218 DOI: 10.1016/j.dci.2008.09.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 09/14/2008] [Accepted: 09/16/2008] [Indexed: 05/27/2023]
Abstract
DC-SIGN, a human C-type lectin, is involved in the transmission of many enveloped viruses. Here we report the cloning and characterization of the cDNA and gene encoding porcine DC-SIGN (pDC-SIGN). The full-length pDC-SIGN cDNA encodes a type II transmembrane protein of 240 amino acids. Phylogenetic analysis revealed that pDC-SIGN, together with bovine, canis and equine DC-SIGN, are more closely related to mouse SIGNR7 and SIGNR8 than to human DC-SIGN. pDC-SIGN has the same gene structure as bovine, canis DC-SIGN and mouse SIGNR8 with eight exons. pDC-SIGN mRNA expression was detected in pig spleen, thymus, lymph node, lung, bone marrow and muscles. pDC-SIGN protein was found to express on the surface of monocyte-derived macrophages and dendritic cells, alveolar macrophages, lymph node sinusoidal macrophage-like, dendritic-like and endothelial cells but not of monocytes, peripheral blood lymphocytes or lymph node lymphocytes. A BHK cell line stably expressing pDC-SIGN binds to human ICAM-3 and ICAM-2 immunoadhesins in a calcium-dependent manner, and enhances the transmission of porcine reproductive and respiratory syndrome virus (PRRSV) to target cells in trans. The results will help better understand the biological role(s) of DC-SIGN family in innate immunity during the evolutionary process.
Collapse
Affiliation(s)
| | | | | | - X.J. Meng
- Corresponding author. Tel.: +1 540 231 6912; fax: +1 540 231 3426.
| |
Collapse
|
26
|
Dendritic Cell Adhesion to Cerebral Endothelium: Role of Endothelial Cell Adhesion Molecules and Their Ligands. J Neuropathol Exp Neurol 2009; 68:300-13. [DOI: 10.1097/nen.0b013e31819a8dd1] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
27
|
Surface alpha 2-3- and alpha 2-6-sialylation of human monocytes and derived dendritic cells and its influence on endocytosis. Glycoconj J 2007; 25:259-68. [PMID: 18080182 DOI: 10.1007/s10719-007-9092-6] [Citation(s) in RCA: 263] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2007] [Revised: 08/24/2007] [Accepted: 11/15/2007] [Indexed: 10/22/2022]
Abstract
Several glycoconjugates are involved in the immune response. Sialic acid is frequently the glycan terminal sugar and it may modulate immune interactions. Dendritic cells (DCs) are antigen-presenting cells with high endocytic capacity and a central role in immune regulation. On this basis, DCs derived from monocytes (mo-DC) are utilised in immunotherapy, though many features are ignored and their use is still limited. We analyzed the surface sialylated glycans expressed during human mo-DC generation. This was monitored by lectin binding and analysis of sialyltransferases (ST) at the mRNA level and by specific enzymatic assays. We showed that alpha 2-3-sialylated O-glycans and alpha 2-6- and alpha 2-3-sialylated N-glycans are present in monocytes and their expression increases during mo-DC differentiation. Three main ST genes are committed with this rearrangement: ST6Gal1 is specifically involved in the augmented alpha 2-6-sialylated N-glycans; ST3Gal1 contributes for the alpha2-3-sialylation of O-glycans, particularly T antigens; and ST3Gal4 may contribute for the increased alpha2-3-sialylated N-glycans. Upon mo-DC maturation, ST6Gal1 and ST3Gal4 are downregulated and ST3Gal1 is altered in a stimulus-dependent manner. We also observed that removing surface sialic acid of immature mo-DC by neuraminidase significantly decreased its endocytic capacity, while it increased in monocytes. Our results indicate the STs expression modulates the increased expression of surface sialylated structures during mo-DC generation, which is probably related with changes in cell mechanisms. The ST downregulation after mo-DC maturation probably results in a decreased sialylation or sialylated glycoconjugates involved in the endocytosis, contributing to the downregulation of one or more antigen-uptake mechanisms specific of mo-DC.
Collapse
|
28
|
Serrano-Gómez D, Sierra-Filardi E, Martínez-Nuñez RT, Caparrós E, Delgado R, Muñoz-Fernández MA, Abad MA, Jimenez-Barbero J, Leal M, Corbí AL. Structural requirements for multimerization of the pathogen receptor dendritic cell-specific ICAM3-grabbing non-integrin (CD209) on the cell surface. J Biol Chem 2007; 283:3889-903. [PMID: 18073208 DOI: 10.1074/jbc.m706004200] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The myeloid C-type lectin dendritic cell-specific ICAM3-grabbing non-integrin (DC-SIGN, CD209) recognizes oligosaccharide ligands on clinically relevant pathogens (HIV, Mycobacterium, and Aspergillus). Alternative splicing and genomic polymorphism generate DC-SIGN mRNA variants, which have been detected at sites of pathogen entrance and transmission. We present evidence that DC-SIGN neck variants are expressed on dendritic and myeloid cells at the RNA and protein levels. Structural analysis revealed that multimerization of DC-SIGN within a cellular context depends on the lectin domain and the number and arrangement of the repeats within the neck region, whose glycosylation negatively affects oligomer formation. Naturally occurring DC-SIGN neck variants differ in multimerization competence in the cell membrane, exhibit altered sugar binding ability, and retain pathogen-interacting capacity, implying that pathogen-induced cluster formation predominates over the basal multimerization capability. Analysis of DC-SIGN neck polymorphisms indicated that the number of allelic variants is higher than previously thought and that multimerization of the prototypic molecule is modulated in the presence of allelic variants with a different neck structure. Our results demonstrate that the presence of allelic variants or a high level of expression of neck domain splicing isoforms might influence the presence and stability of DC-SIGN multimers on the cell surface, thus providing a molecular explanation for the correlation between DC-SIGN polymorphisms and altered susceptibility to HIV-1 and other pathogens.
Collapse
Affiliation(s)
- Diego Serrano-Gómez
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Ramiro de Maeztu 9, Madrid 28040, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Chan KYK, Ching JCY, Xu MS, Cheung ANY, Yip SP, Yam LYC, Lai ST, Chu CM, Wong ATY, Song YQ, Huang FP, Liu W, Chung PH, Leung GM, Chow EYD, Chan EYT, Chan JCK, Ngan H, Tam P, Chan LC, Sham P, Chan VSF, Peiris M, Lin SCL, Khoo US. Association of ICAM3 genetic variant with severe acute respiratory syndrome. J Infect Dis 2007; 196:271-80. [PMID: 17570115 PMCID: PMC7202406 DOI: 10.1086/518892] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2005] [Accepted: 02/16/2007] [Indexed: 12/12/2022] Open
Abstract
Genetic polymorphisms have been demonstrated to be associated with vulnerability to human infection. ICAM3, an intercellular adhesion molecule important for T cell activation, and FCER2 (CD23), an immune response gene, both located on chromosome 19p13.3 were investigated for host genetic susceptibility and association with clinical outcome. A case-control study based on 817 patients with confirmed severe acute respiratory syndrome (SARS), 307 health care worker control subjects, 290 outpatient control subjects, and 309 household control subjects unaffected by SARS from Hong Kong was conducted to test for genetic association. No significant association to susceptibility to SARS-CoV infection was found for the FCER2 and the ICAM3 single nucleotide polymorphisms. However, patients with SARS homozygous for ICAM3 Gly143 showed significant association with higher lactate dehydrogenase levels (P=.0067; odds ratio [OR], 4.31 [95% confidence interval [CI], 1.37–13.56]) and lower total white blood cell counts (P=.022; OR, 0.30 [95% CI, 0.10–0.89]) on admission. These findings support the role of ICAM3 in the immunopathogenesis of SARS.
Collapse
Affiliation(s)
- Kelvin Y. K. Chan
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Johannes C. Y. Ching
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - M. S. Xu
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Annie N. Y. Cheung
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Shea-Ping Yip
- Department of Health Technology and Informatics, Hong Kong Polytechnic UniversityHong Kong
| | | | | | | | | | - You-Qiang Song
- Department of Biochemistry, Hong Kong Jockey Club Clinical Research CentreHong Kong
- Genome Research Center, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Fang-Ping Huang
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Wei Liu
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | | | - G. M. Leung
- Department of Community Medicine, Li Ka Shing Faculty of Medicine, University of Hong KongHong Kong
| | | | - Eric Y. T. Chan
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Jane C. K. Chan
- Hospital Authority Severe Acute Respiratory Syndrome Collaborative Group, Hong Kong Hospital Authority Head OfficeHong Kong
| | - Hextan Ngan
- Department of Obstetrics and Gynecology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Paul Tam
- Department of Surgery, Hong Kong Jockey Club Clinical Research CentreHong Kong
- Genome Research Center, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Li-Chong Chan
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Pak Sham
- Department of Psychiatry, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Vera S. F. Chan
- Department of Biosurgery and Surgical Technology, Imperial College LondonLondon, United Kingdom
| | - Malik Peiris
- Department of Microbiology, Hong Kong Jockey Club Clinical Research CentreHong Kong
| | - Steve C. L. Lin
- Department of Biosurgery and Surgical Technology, Imperial College LondonLondon, United Kingdom
| | - Ui-Soon Khoo
- Department of Pathology, Hong Kong Jockey Club Clinical Research CentreHong Kong
- Reprints or correspondence: Dr. Ui-Soon Khoo, Rm. 324, 3/F, University Pathology Bldg., Dept. of Pathology, University of Hong Kong, Queen Mary Hospital, Pokfulam Rd., Hong Kong ()
| |
Collapse
|
30
|
Valdivia-Arenas M, Amer A, Henning L, Wewers M, Schlesinger L. Lung infections and innate host defense. ACTA ACUST UNITED AC 2007; 4:73-81. [PMID: 18592001 DOI: 10.1016/j.ddmec.2007.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Ma Valdivia-Arenas
- Center for Microbial Interface Biology, Dorothy M. Davis Heart and Lung Research Institute, Divisions of Infectious Diseases and Pulmonary, Allergy, Critical Care & Sleep Medicine, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210
| | | | | | | | | |
Collapse
|
31
|
Lau AH, Thomson AW, Colvin BL. Chronic ethanol exposure affects in vivo migration of hepatic dendritic cells to secondary lymphoid tissue. Hum Immunol 2007; 68:577-85. [PMID: 17584579 DOI: 10.1016/j.humimm.2007.03.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2006] [Revised: 02/09/2007] [Accepted: 03/15/2007] [Indexed: 12/24/2022]
Abstract
The mechanisms by which chronic ethanol (EtOH) consumption results in an immune-compromised state have not been fully elucidated. No studies to date have ascertained whether EtOH affects the migratory capacity of dendritic cells (DC), potent immune regulators. We hypothesized that EtOH exposure might affect hepatic and splenic DC trafficking to secondary lymphoid tissues and the resulting immune response. Hepatic DC from EtOH-treated animals migrated in greater numbers to draining lymphoid tissue than controls, whereas spleen DC were unaffected. Moreover, hepatic EtOH-exposed (E) DC induced more vigorous priming of allogeneic T cells in vivo compared with splenic EDC or controls. Altered hepatic EDC migration was independent of either CCR7 or CD11a expression, with no striking changes in surface expression of other adhesion molecules analyzed. The modified trafficking to secondary lymphoid tissue observed for hepatic EDC may play a role in the altered immune response to microbial pathogens in chronic alcohol users.
Collapse
MESH Headings
- Alcohol-Induced Disorders/immunology
- Animals
- Antibodies, Monoclonal
- CD11 Antigens/biosynthesis
- CD11 Antigens/immunology
- Cell Adhesion Molecules/biosynthesis
- Cell Adhesion Molecules/immunology
- Cell Movement/drug effects
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/physiology
- Ethanol/administration & dosage
- Ethanol/toxicity
- Lectins, C-Type/biosynthesis
- Lectins, C-Type/immunology
- Liver/drug effects
- Liver/immunology
- Lymphoid Tissue/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Oligodeoxyribonucleotides/immunology
- Receptors, CCR7
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/immunology
- Receptors, Chemokine/biosynthesis
- Receptors, Chemokine/immunology
- Spleen/immunology
- Spleen/metabolism
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Audrey H Lau
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | |
Collapse
|
32
|
Altin JG, Parish CR. Liposomal vaccines--targeting the delivery of antigen. Methods 2007; 40:39-52. [PMID: 16997712 DOI: 10.1016/j.ymeth.2006.05.027] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Accepted: 05/05/2006] [Indexed: 01/05/2023] Open
Abstract
Vaccines that can prime the adaptive immune system for a quick and effective response against a pathogen or tumor cells, require the generation of antigen (Ag)-specific memory T and B cells. The unique ability of dendritic cells (DCs) to activate naïve T cells, implies a key role for DCs in this process. The generation of tumor-specific CD8(+) cytotoxic T cells (CTLs) is dependent on both T cell stimulation with Ag (peptide-MHC-complexes) and costimulation. Interestingly, tumor cells that lack expression of T cell costimulatory molecules become highly immunogenic when transfected to express such molecules on their surface. Adoptive immunotherapy with Ag-pulsed DCs also is a strategy showing promise as a treatment for cancer. The use of such cell-based vaccines, however, is cumbersome and expensive to use clinically, and/or may carry risks due to genetic manipulations. Liposomes are particulate vesicular lipid structures that can incorporate Ag, immunomodulatory factors and targeting molecules, and hence can serve as potent vaccines. Similarly, Ag-containing plasma membrane vesicles (PMV) derived from tumor cells can be modified to incorporate a T cell costimulatory molecule to provide both TCR stimulation, and costimulation. PMVs also can be modified to contain IFN-gamma and molecules for targeting DCs, permitting delivery of both Ag and a DC maturation signal for initiating an effective immune response. Our results show that use of such agents as vaccines can induce potent anti-tumor immune responses and immunotherapeutic effects in tumor models, and provide a strategy for the development of effective vaccines and immunotherapies for cancer and infectious diseases.
Collapse
Affiliation(s)
- Joseph G Altin
- School of Biochemistry and Molecular Biology, Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia.
| | | |
Collapse
|
33
|
Hänel K, Willbold D. SARS-CoV accessory protein 7a directly interacts with human LFA-1. Biol Chem 2007; 388:1325-32. [DOI: 10.1515/bc.2007.157] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
34
|
Butler M, Morel AS, Jordan WJ, Eren E, Hue S, Shrimpton RE, Ritter MA. Altered expression and endocytic function of CD205 in human dendritic cells, and detection of a CD205-DCL-1 fusion protein upon dendritic cell maturation. Immunology 2006; 120:362-71. [PMID: 17163964 PMCID: PMC2265885 DOI: 10.1111/j.1365-2567.2006.02512.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
CD205 (DEC-205) is a member of the macrophage mannose receptor family of C-type lectins. These molecules are known to mediate a wide variety of biological functions including the capture and internalization of ligands for subsequent processing and presentation by dendritic cells. Although its ligands await identification, the endocytic properties of CD205 make it an ideal target for those wishing to design vaccines and targeted immunotherapies. We present a detailed analysis of CD205 expression, distribution and endocytosis in human monocyte-derived dendritic cells undergoing lipopolysaccharide-induced maturation. Unlike other members of the macrophage mannose receptor family, CD205 was up-regulated upon dendritic cell maturation. This increase was a result of de novo synthesis as well as a redistribution of molecules from endocytic compartments to the cell surface. Furthermore, the endocytic capacity of CD205 was abrogated and small amounts of the recently identified CD205-DCL-1 fusion protein were detected in mature DC. Our results suggest that CD205 has two distinct functions -- one as an endocytic receptor on immature dendritic cells and a second as a non-endocytic molecule on mature dendritic cells -- and further highlight its potential as an immuno-modulatory target for vaccine and immunotherapy development.
Collapse
MESH Headings
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Cell Differentiation/immunology
- Cells, Cultured
- Dendritic Cells/immunology
- Down-Regulation/immunology
- Endocytosis/immunology
- Humans
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Leukocytes, Mononuclear/immunology
- Lipopolysaccharides/immunology
- Mannose Receptor
- Mannose-Binding Lectins/metabolism
- Minor Histocompatibility Antigens
- Monocytes/immunology
- Oncogene Proteins, Fusion/metabolism
- Polymerase Chain Reaction/methods
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Receptors, Mitogen/metabolism
- Translocation, Genetic/immunology
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Matt Butler
- Department of Immunology, Imperial College London, Faculty of Medicine, Hammersmith Campus, London, UK.
| | | | | | | | | | | | | |
Collapse
|
35
|
Chan R, Gilbert M, Thompson KM, Marsh HN, Epstein DM, Pendergrast PS. Co-expression of anti-NFkappaB RNA aptamers and siRNAs leads to maximal suppression of NFkappaB activity in mammalian cells. Nucleic Acids Res 2006; 34:e36. [PMID: 16517938 PMCID: PMC1390692 DOI: 10.1093/nar/gnj028] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The specific down-regulation of gene expression in cells is a powerful method for elucidating a gene's function. A common method for suppressing gene expression is the elimination of mRNA by RNAi or antisense. Alternatively, oligonucleotide-derived aptamers have been used as protein-directed agents for the specific knock-down of both intracellular and extracellular protein activity. Protein-directed methods offer the advantage of more closely mimicking small molecule therapeutics' mechanism of activity. Furthermore, protein-directed methods may synergize with RNA-directed methods since the two methods attack gene expression at different levels. Here we have knocked down a well-characterized intracellular protein's activity, NFκB, by expressing either aptamers or small interfering RNAs (siRNAs). Both methods can diminish NFκB's activity to similar levels (from 29 to 64%). Interestingly, expression of both aptamers and siRNAs simultaneously, suppressed NFκB activity better than either method alone (up to 90%). These results demonstrate that the expression of intracellular aptamers is a viable alternative to siRNA knock-down. Furthermore, for the first time, we show that the use of aptamers and siRNA together can be the most effective way to achieve maximal knock-down of protein activity.
Collapse
|
36
|
Altin JG, van Broekhoven CL, Parish CR. Targeting dendritic cells with antigen-containing liposomes: antitumour immunity. Expert Opin Biol Ther 2005; 4:1735-47. [PMID: 15500402 DOI: 10.1517/14712598.4.11.1735] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Dendritic cells (DCs) are antigen-presenting cells that play an important role in the body's immune defence against cancer. Strategies using antigen-primed DCs as tumour vaccines show promise in patients, but the approach is cumbersome to use clinically. Soluble tumour antigens can be targeted to DCs in vivo, but this often induces antigenic tolerance rather than immunity. Liposomes are vesicular lipid structures with adjuvant-like properties. Importantly, liposomes can encapsulate antigen and immunomodulatory factors, thus serving as potent delivery vehicles. Different strategies are being explored to target liposomal antigens to DCs in vivo. One approach has employed single-chain antibody fragments to the DC surface molecules CD11c and DEC-205, attached to the vesicle surface by metal-chelating linkage, to target liposomal membranes containing antigen and either interferon-gamma or lipopolysaccharide to DCs. Such membranes induce dramatic antitumour responses and immunotherapeutic effects when used as a vaccine in the murine tumour model B16-OVA melanoma. Liposomal targeting of antigen and maturation signals directly to DCs in vivo, therefore, represents a much simpler strategy for cancer immunotherapy than antigen loading DCs ex vivo.
Collapse
Affiliation(s)
- Joseph G Altin
- The Australian National University, School of Biochemistry and Molecular Biology, Faculty of Science, Canberra, ACT 0200, Australia.
| | | | | |
Collapse
|
37
|
Hänel K, Stangler T, Stoldt M, Willbold D. Solution structure of the X4 protein coded by the SARS related coronavirus reveals an immunoglobulin like fold and suggests a binding activity to integrin I domains. J Biomed Sci 2005; 13:281-93. [PMID: 16328780 PMCID: PMC7089389 DOI: 10.1007/s11373-005-9043-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2005] [Accepted: 10/11/2005] [Indexed: 02/08/2023] Open
Abstract
The SARS related Coronavirus genome contains a variety of novel accessory genes. One of these, called ORF7a or ORF8, code for a protein, known as 7a, U122 or X4. We set out to determine the three-dimensional structure of the soluble ectodomain of this type-I transmembrane protein by nuclear magnetic resonance spectroscopy. The fold of the protein is the first member of a further variation of the immunoglobulin like beta-sandwich fold. Because X4 does not reveal significant sequence homologies to proteins in the data bases, we carried out a structure based similarity search for proteins with known function. High structural similarity to Dl domains of ICAM-1 and ICAM-2, and common features in amino acid sequence between X4 and ICAM-1, suggest X4 to possess binding activity for the alpha(L) integrin I domain of LFA-1. Further, based on this structure based prediction, potential functions of X4 in virus replication and pathogenesis are discussed.
Collapse
Affiliation(s)
- Karen Hänel
- Forschungszentrum Jülich, Institut für Biologische Informationsverarbeitung (IBI-2), 52425 Jülich, Germany
- Heinrich-Heine-Universität, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| | - Thomas Stangler
- Forschungszentrum Jülich, Institut für Biologische Informationsverarbeitung (IBI-2), 52425 Jülich, Germany
- Heinrich-Heine-Universität, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| | - Matthias Stoldt
- Forschungszentrum Jülich, Institut für Biologische Informationsverarbeitung (IBI-2), 52425 Jülich, Germany
- Heinrich-Heine-Universität, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| | - Dieter Willbold
- Forschungszentrum Jülich, Institut für Biologische Informationsverarbeitung (IBI-2), 52425 Jülich, Germany
- Heinrich-Heine-Universität, Institut für Physikalische Biologie and BMFZ, 40225 Düsseldorf, Germany
| |
Collapse
|
38
|
Martinez O, Brackenridge S, El-Idrissi MEA, Prabhakar BS. DC-SIGN, but not sDC-SIGN, can modulate IL-2 production from PMA- and anti-CD3-stimulated primary human CD4 T cells. Int Immunol 2005; 17:769-78. [PMID: 15944195 DOI: 10.1093/intimm/dxh258] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Dendritic cell (DC)-specific intercellular cell adhesion molecule-3 (ICAM-3)-grabbing non-integrin (DC-SIGN) is expressed on the surface of DCs and specialized macrophages and can support T cell proliferation. Antibody-mediated co-ligation of CD3 and ICAM-3, the ligand for both DC-SIGN and leukocyte function-associated antigen-1, leads to T cell activation. Therefore, we tested to see whether DC-SIGN or a splice variant of dendritic cell-specific intercellular cell adhesion molecule-3-grabbing non-integrin (sDC-SIGN) can co-stimulate primary human T cells. The sDC-SIGN lacking the transmembrane domain encoded by exon 3 localizes to the cytoplasm of cells and is not secreted. Both B7 and DC-SIGN co-stimulated phorbol myristate acetate-stimulated CD4+ cells as compared with controls. However, unlike B7, both DC-SIGN and sDC-SIGN failed to co-stimulate CD4+ T cells treated with sub-optimal amounts of anti-CD3 (2 microg ml(-1)) as defined by a lack of CD69 and CD25 up-regulation, cell division and cytokine secretion. Instead, DC-SIGN, and not sDC-SIGN, induced a small but consistent down-regulation of IL-2 production by these CD4+ T cells. In contrast, DC-SIGN in the presence of 30 mug ml(-1) of anti-CD3 modestly up-regulated cytokine production as compared with control. These results suggest that DC-SIGN can differentially modulate T cell stimulation.
Collapse
MESH Headings
- Alternative Splicing
- Animals
- Antibodies
- Antigens, CD/analysis
- CD4-Positive T-Lymphocytes/immunology
- CHO Cells/metabolism
- Cell Adhesion Molecules/biosynthesis
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Cells, Cultured
- Cloning, Molecular
- Cricetinae
- Cricetulus
- Genes, Reporter
- Humans
- Interferon-gamma/biosynthesis
- Interleukin-2/biosynthesis
- Lectins, C-Type/biosynthesis
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lymphocyte Activation
- Lymphocyte Subsets/immunology
- Protein Isoforms/biosynthesis
- Protein Isoforms/genetics
- Protein Isoforms/immunology
- Receptors, Cell Surface/biosynthesis
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- Transfection
Collapse
Affiliation(s)
- Osvaldo Martinez
- Department of Microbiology and Immunology, University of Illinois, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
39
|
Boes M, Cuvillier A, Ploegh H. Membrane specializations and endosome maturation in dendritic cells and B cells. Trends Cell Biol 2004; 14:175-83. [PMID: 15066635 DOI: 10.1016/j.tcb.2004.02.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Interest in the cell biology of antigen presentation is centered on dendritic cells (DCs) as initiators of the immune response. The ability to examine primary antigen-presenting cells, as opposed to cell lines, has opened a new window for study of antigen processing and peptide acquisition by Class II major histocompatibility complex (MHC) products, especially where intracellular trafficking of peptide-Class-II complexes is concerned. Here, we review the dynamics of Class II MHC-positive intracellular structures in dendritic cells as well as B cells. We focus on the generation of multivesicular bodies, where Class II MHC products acquire antigenic peptide, on the endosomal transport of peptide-loaded Class II MHC to the cell surface and on the importance of Class II MHC localization in membrane microdomains.
Collapse
Affiliation(s)
- Marianne Boes
- Department of Pathology, Harvard Medical School, The New Research Building, Room 836, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | | | | |
Collapse
|
40
|
Abstract
DC-SIGN, a dendritic Cell-specific adhesion receptor and a type II transmembrane mannose-binding C-type lectin, is very important in the function of DC, both in mediating naive T cell interactions through ICAM-3 and as a rolling receptor that mediates the DC-specific ICAM-2-dependent migration processes. It can be used by viral and bacterial pathogens including Human Immunodeficiency Virus (HIV), HCV, Ebola Virus, CMV and Mycobacterium tuberculosis to facilitate infection. Both DC-SIGN and DC-SIGNR can act either in cis, by concentrating virus on target cells, or in trans, by transmission of bound virus to a target cell expressing appropropriate entry receptors. Recent work showed that DC-SIGN are high-affinity binding receptors for HCV. Besides playing a role in entry into DC, HCV E2 interaction with DC-SIGN might also be detrimental for the interaction of DC with T cells during antigen presentation. The clinical strategies that target DC-SIGN may be successful in restricting HCV dissemination and pathogenesis as well as directing the migration of DCs to manipulate appropriate immune responses in autoimmunity and tumorigenic situations.
Collapse
Affiliation(s)
- Zhi-Hua Feng
- The Center of Diagnosis and Treatment for Infectious Diseases of PLA, Tangdu Hospital, Fourth Military Medical University, Xi'an 710038, Shaanxi Province, China.
| | | | | | | | | |
Collapse
|
41
|
Geijtenbeek TBH, van Vliet SJ, Engering A, 't Hart BA, van Kooyk Y. Self- and nonself-recognition by C-type lectins on dendritic cells. Annu Rev Immunol 2004; 22:33-54. [PMID: 15032573 DOI: 10.1146/annurev.immunol.22.012703.104558] [Citation(s) in RCA: 364] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DCs) are highly efficient antigen-presenting cells (APCs) that collect antigen in body tissues and transport them to draining lymph nodes. Antigenic peptides are loaded onto major histocompatibility complex (MHC) molecules for presentation to naive T cells, resulting in the induction of cellular and humoral immune responses. DCs take up antigen through phagocytosis, pinocytosis, and endocytosis via different groups of receptor families, such as Fc receptors for antigen-antibody complexes, C-type lectin receptors (CLRs) for glycoproteins, and pattern recognition receptors, such as Toll-like receptors (TLRs), for microbial antigens. Uptake of antigen by CLRs leads to presentation of antigens on MHC class I and II molecules. DCs are well equipped to distinguish between self- and nonself-antigens by the variable expression of cell-surface receptors such as CLRs and TLRs. In the steady state, DCs are not immunologically quiescent but use their antigen-handling capacities to maintain peripheral tolerance. DCs are continuously sampling and presenting self- and harmless environmental proteins to silence immune activation. Uptake of self-components in the intestine and airways are good examples of sites where continuous presentation of self- and foreign antigens occurs without immune activation. In contrast, efficient antigen-specific immune activation occurs upon encounter of DCs with nonself-pathogens. Recognition of pathogens by DCs triggers specific receptors such as TLRs that result in DC maturation and subsequently immune activation. Here we discuss the concept that cross talk between TLRs and CLRs, differentially expressed by subsets of DCs, accounts for the different pathways to peripheral tolerance, such as deletion and suppression, and immune activation.
Collapse
Affiliation(s)
- Teunis B H Geijtenbeek
- Department of Molecular Cell Biology and Immunology, Vrije Universiteit Medical Center Amsterdam, 1081 BT Amsterdam, Netherlands
| | | | | | | | | |
Collapse
|
42
|
Larregina AT, Morelli AE, Tkacheva O, Erdos G, Donahue C, Watkins SC, Thomson AW, Falo LD. Highly efficient expression of transgenic proteins by naked DNA-transfected dendritic cells through terminal differentiation. Blood 2004; 103:811-9. [PMID: 14551149 DOI: 10.1182/blood-2003-02-0524] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AbstractDendritic cells (DCs) play a key role in the induction and control of immunity. Genetic engineering of DCs is a promising approach for the development of a broad range of immunomodulatory strategies, for purposes ranging from genetic immunization to tolerance induction. The development of DC-based immunotherapies is limited by the inability to efficiently transfect DCs using naked DNA. Here we demonstrate that after plasmid DNA delivery, the transgene expression level controlled by the human immediate-early cytomegalovirus promoter (hIE-CMVp) is higher in mature DCs than in immature DCs and is further increased after terminal differentiation of DCs by agonist anti-CD40 monoclonal antibody (mAb) or after DC interaction with CD4+ T cells. CD40 signaling of DCs resulted in nuclear translocation of the transcription factors nuclear factor-κB (NF-κB), activator of protein-1 (AP-1), and cyclic adenosine monophosphate (cAMP)–responsive element, necessary for the activation of hIE-CMVp. Transgene expression by DCs diminished after the inhibition of these transcription factors or the blockade of adhesion molecules involved in the DC–T-cell synapse. Importantly, CD40 signaling of DCs results in the highly efficient expression and presentation of transgenic antigens and the induction of “in vivo” cytotoxic T-cell (CTL) responses specific for transgenic antigen peptides, demonstrating the functional potential of genetically engineered DCs.
Collapse
|
43
|
Shimaoka T, Nakayama T, Fukumoto N, Kume N, Takahashi S, Yamaguchi J, Minami M, Hayashida K, Kita T, Ohsumi J, Yoshie O, Yonehara S. Cell surface-anchored SR-PSOX/CXC chemokine ligand 16 mediates firm adhesion of CXC chemokine receptor 6-expressing cells. J Leukoc Biol 2003; 75:267-74. [PMID: 14634054 DOI: 10.1189/jlb.1003465] [Citation(s) in RCA: 184] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Direct contacts between dendritic cells (DCs) and T cells or natural killer T (NKT) cells play important roles in primary and secondary immune responses. SR-PSOX/CXC chemokine ligand 16 (CXCL16), which is selectively expressed on DCs and macrophages, is a scavenger receptor for oxidized low-density lipoprotein and also the chemokine ligand for a G protein-coupled receptor CXC chemokine receptor 6 (CXCR6), expressed on activated T cells and NKT cells. SR-PSOX/CXCL16 is the second transmembrane-type chemokine with a chemokine domain fused to a mucin-like stalk, a structure very similar to that of fractalkine (FNK). Here, we demonstrate that SR-PSOX/CXCL16 functions as a cell adhesion molecule for cells expressing CXCR6 in the same manner that FNK functions as a cell adhesion molecule for cells expressing CX(3)C chemokine receptor 1 (CX(3)CR1) without requiring CX(3)CR1-mediated signal transduction or integrin activation. The chemokine domain of SR-PSOX/CXCL16 mediated the adhesion of CXCR6-expressing cells, which was not impaired by treatment with pertussis toxin, a Galphai protein blocker, which inhibited chemotaxis of CXCR6-expressing cells induced by SR-PSOX/CXCL16. Furthermore, the adhesion activity was up-regulated by treatment of SR-PSOX/CXCL16-expressing cells with a metalloprotease inhibitor, which increased surface expression levels of SR-PSOX/CXCL16. Thus, SR-PSOX/CXCL16 is a unique molecule that not only attracts T cells and NKT cells toward DCs but also supports their firm adhesion to DCs.
Collapse
MESH Headings
- Animals
- Cell Adhesion
- Cell Adhesion Molecules
- Chemokine CXCL16
- Chemokine CXCL6
- Chemokines, CXC/metabolism
- Chemokines, CXC/physiology
- Enzyme Inhibitors/pharmacology
- Humans
- Membrane Proteins/metabolism
- Membrane Proteins/physiology
- Metalloproteases/antagonists & inhibitors
- Mice
- Pertussis Toxin/pharmacology
- Protein Binding
- Protein Structure, Tertiary
- Receptors, CXCR
- Receptors, CXCR6
- Receptors, Chemokine
- Receptors, Cytokine
- Receptors, G-Protein-Coupled
- Receptors, Immunologic
- Receptors, Scavenger
- Receptors, Virus
Collapse
Affiliation(s)
- Takeshi Shimaoka
- Kyoto University, Shogoin Kawahara-cho 53, Sakyo-ku, Kyoto 606-8507, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mohty M, Gaugler B. Dendritic cells: interfaces with immunobiology and medicine. A report from the Keystone Symposia Meeting held in Keystone, 3-8 March 2003. Leukemia 2003; 17:1753-8. [PMID: 12970774 DOI: 10.1038/sj.leu.2403049] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2003] [Accepted: 04/30/2003] [Indexed: 01/07/2023]
Abstract
Dendritic cells (DCs) are the most potent antigen-presenting cells in vitro and in vivo. They play a key role in the initiation of the immune response and are considered promising targets for immunotherapy. The recent DC Keystone Symposia, organized by Ralph M Steinman, Anne O'Garra and Jacques Banchereau, was held on 3-8 March 2003 in Keystone (CO, USA). This multidisciplinary meeting developed various areas related to the DC biology including: (i) DC and the control of immunity vs tolerance; (ii) DC maturation and manipulation for immunotherapy in vivo; and (iii) antigen capture by DC in situ and interactions with microbial pathogens. The aim of this report is to present some of the highlights developed during the meeting and debated among the DC community.
Collapse
Affiliation(s)
- M Mohty
- Laboratoire d'Immunologie des Tumeurs, Institut Paoli-Calmettes, Marseille, France
| | | |
Collapse
|
45
|
Geijtenbeek TBH, van Kooyk Y. DC-SIGN: a novel HIV receptor on DCs that mediates HIV-1 transmission. Curr Top Microbiol Immunol 2003; 276:31-54. [PMID: 12797442 DOI: 10.1007/978-3-662-06508-2_2] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The dendritic cell (DC)-specific HIV-1 receptor DC-SIGN plays a key-role in the dissemination of HIV-1 by DCs. DC-SIGN captures HIV-1 at sites of entry, enabling its transport to lymphoid tissues, where DC-SIGN efficiently transmits low amounts of HIV-1 to T cells. The expression pattern of DC-SIGN in mucosal tissue, lymph nodes, placenta and blood suggests a function for DC-SIGN in both horizontal and vertical transmission of HIV-1. Moreover, the efficiency of DC-SIGN+ blood DC to transmit HIV-1 to T cells supports a role in HIV-1 transmission via blood. To date, DC-SIGN represents a novel class of HIV-1 receptor, because it does not allow viral infection but binds HIV-1 and enhances its infection of T cells in trans. Its unique function is further underscored by its restricted expression on DCs. Although DC-SIGN is a C-type lectin with an affinity for carbohydrates exemplified by its interaction with its immunological ligand ICAM-3, recent evidence demonstrates that glycosylation of gp120 is not necessary for its interaction with DC-SIGN. Moreover, mutational analysis demonstrates that the HIV-1 gp120 binding site in DC-SIGN is different from that of ICAM-3. Besides its role in DC-mediated adhesion processes, DC-SIGN also functions as an antigen receptor that captures and internalises antigens for presentation by DC. Strikingly, HIV-1 circumvents processing after binding DC-SIGN and remains infectious for several days after capture. A better understanding of the action of this novel HIV receptor in initial viral infection and subsequent transmission will provide a basis for the design of drugs that inhibit or alter interactions of DC-SIGN with gp120, interfering with HIV-1 dissemination and that may have a therapeutic value in both immunological diseases and/or HIV-1 infections.
Collapse
Affiliation(s)
- T B H Geijtenbeek
- Department of Molecular Cell Biology, Vrije Universiteit Medical Center Amsterdam, van de Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | | |
Collapse
|
46
|
Geijtenbeek TBH, van Kooyk Y. Pathogens target DC-SIGN to influence their fate DC-SIGN functions as a pathogen receptor with broad specificity. APMIS 2003; 111:698-714. [PMID: 12974773 DOI: 10.1034/j.1600-0463.2003.11107803.x] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Dendritic cells (DC) are vital in the defense against pathogens. To sense pathogens DC express pathogen recognition receptors such as toll-like receptors (TLR) and C-type lectins that recognize different fragments of pathogens, and subsequently activate or present pathogen fragments to T cells. It is now becoming evident that some pathogens subvert DC functions to escape immune surveillance. HIV-1 targets the DC-specific C-type lectin DC-SIGN to hijack DC for viral dissemination. HIV-1 binding to DC-SIGN protects HIV-1 from antigen processing and facilitates its transport to lymphoid tissues, where DC-SIGN promotes HIV-1 infection of T cells. Recent studies demonstrate that DC-SIGN is a more universal pathogen receptor that also recognizes Ebola, cytomegalovirus and mycobacteria. Mycobacterium tuberculosis targets DC-SIGN by a mechanism that is distinct from that of HIV-1, leading to inhibition of the immunostimulatory function of DC and pathogen survival. Thus, a better understanding of DC-SIGN-pathogen interactions and their effects on DC function is necessary to combat infections.
Collapse
Affiliation(s)
- Teunis B H Geijtenbeek
- Department of Molecular Cell Biology, Vrije Universiteit Medical Center Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
47
|
Kabashima K, Murata T, Tanaka H, Matsuoka T, Sakata D, Yoshida N, Katagiri K, Kinashi T, Tanaka T, Miyasaka M, Nagai H, Ushikubi F, Narumiya S. Thromboxane A2 modulates interaction of dendritic cells and T cells and regulates acquired immunity. Nat Immunol 2003; 4:694-701. [PMID: 12778172 DOI: 10.1038/ni943] [Citation(s) in RCA: 156] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2003] [Accepted: 05/02/2003] [Indexed: 11/09/2022]
Abstract
Physical interaction of T cells and dendritic cells (DCs) is essential for T cell proliferation and differentiation, but it has been unclear how this interaction is regulated physiologically. Here we show that DCs produce thromboxane A2 (TXA2), whereas naive T cells express the thromboxane receptor (TP). In vitro, a TP agonist enhances random cell movement (chemokinesis) of naive but not memory T cells, impairs DC-T cell adhesion, and inhibits DC-dependent proliferation of T cells. In vivo, immune responses to foreign antigens are enhanced in TP-deficient mice, which also develop marked lymphadenopathy with age. Similar immune responses were seen in wild-type mice treated with a TP antagonist during the sensitization period. Thus, TXA2-TP signaling modulates acquired immunity by negatively regulating DC-T cell interactions.
Collapse
Affiliation(s)
- Kenji Kabashima
- Department of Pharmacology, Kyoto University Faculty of Medicine, Kyoto 606-8501, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
te Velde AA, van Kooyk Y, Braat H, Hommes DW, Dellemijn TAM, Slors JFM, van Deventer SJH, Vyth-Dreese FA. Increased expression of DC-SIGN+IL-12+IL-18+ and CD83+IL-12-IL-18- dendritic cell populations in the colonic mucosa of patients with Crohn's disease. Eur J Immunol 2003; 33:143-51. [PMID: 12594843 DOI: 10.1002/immu.200390017] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Dentritic cells (DC) as antigen-presenting cells are most likely responsible for regulation of abnormal T cell activation in Crohn's disease (CD), a chronic inflammatory bowel disease. We have analyzed the expression of activation and maturation markers on DC in the colon mucosa from patients with CD compared with normal colon, using immunohistochemical techniques. We found two distinct populations of DC present in CD patients: a DC-specific ICAM-3 grabbing non-integrin (DC-SIGN)(+) population that was present scattered throughout the mucosa, and a CD83(+) population that was present in aggregated lymphoid nodules and as single cells in the lamina propria. In normal colon the number of DC-SIGN(+) DC was lower and CD83(+) DC were detected only in very few solitary lymphoid nodules. Co-expression of activation markers and cytokine synthesis was analyzed with three-color confocal laser scanning microscopy analysis. CD80 expression was enhanced on the majority of DC-SIGN(+) DC in CD patients, whereas only a proportion of the CD83(+) DC co-expressed CD80 in CD as well as in normal tissue. Surprisingly, IL-12 and IL-18 were only detected in DC-SIGN(+) DC and not in CD83(+) DC. A similar pattern of cytokine production was observed in normal colon albeit to a much lesser extent. The characteristics of these in-situ-differentiated DC markedly differ from the in-vitro-generated DC that simultaneously express DC-SIGN, CD83 and cytokines.
Collapse
Affiliation(s)
- Anje A te Velde
- Laboratory of Experimental Internal Medicine, Academic Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
de Vos AF, van Meurs M, Brok HP, Boven LA, Hintzen RQ, van der Valk P, Ravid R, Rensing S, Boon L, 't Hart BA, Laman JD. Transfer of central nervous system autoantigens and presentation in secondary lymphoid organs. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5415-23. [PMID: 12421916 DOI: 10.4049/jimmunol.169.10.5415] [Citation(s) in RCA: 202] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cells are thought to regulate tolerance induction vs immunization by transferring Ags and peripheral signals to draining lymph nodes (LN). However, whether myelin Ag transfer and presentation in LN occurs during demyelinating brain disease is unknown. In this study, we demonstrate redistribution of autoantigens from brain lesions to cervical LN in monkey experimental autoimmune encephalomyelitis (EAE) and in multiple sclerosis (MS). Immunohistochemical analysis revealed significantly more cells containing myelin Ags in cervical LN of monkeys with EAE compared with those of healthy control monkeys. Myelin Ags were observed in cells expressing dendritic cell/macrophage-specific markers, MHC class II, and costimulatory molecules. Moreover, these cells were directly juxtaposed to T cells, suggesting that cognate interactions between myelin-containing APC and T cells are taking place in brain-draining LN. Indeed, myelin Ag-reactive T cells were observed in cervical LN from marmosets and rhesus monkeys. Importantly, these findings were paralleled by our findings in human tissue. We observed significantly more myelin Ag-containing cells in LN of individuals with MS compared with those of control individuals. These cells expressed APC markers, as observed in marmosets and rhesus monkeys. These findings suggest that during MS and EAE, modulation of T cell reactivity against brain-derived Ags also takes place in cervical LN and not necessarily inside the brain. A major implication is that novel therapeutic strategies may be targeted to peripheral events, thereby circumventing the blood-brain barrier.
Collapse
Affiliation(s)
- Alex F de Vos
- Department of Immunology, Erasmus Medical Centre, 3000 DR Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Gruber A, Chalmers AS, Popov S, Ruprecht RM. Functional aspects of binding of monoclonal antibody DCN46 to DC-SIGN on dendritic cells. Immunol Lett 2002; 84:103-8. [PMID: 12270546 DOI: 10.1016/s0165-2478(02)00144-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Dendritic cell (DC)-specific ICAM-3 grabbing nonintegrin (DC-SIGN) is a DC-specific antigen that plays an important role in the induction of primary immune responses as well as during HIV infection. In the present study, we analyzed the effect of binding of monoclonal antibody DCN46 to DC surface, expressed DC-SIGN on DC function. DC-SIGN antibody treated, immature DC were able to differentiate into mature DC and had the same capacity as untreated DC to induce primary and secondary immune responses. In combination with flow cytometric cell sorting, DC-SIGN antibody treatment of DC yielded highly pure and functional DC. Given the apparent lack of functional effect of monoclonal antibody DCN46 on DC, the latter antibody may prove useful for research on and clinical use of highly pure and functional DC.
Collapse
Affiliation(s)
- Andreas Gruber
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, 44 Binney Street, JFB-809, Boston, MA 02115-6084, USA.
| | | | | | | |
Collapse
|