1
|
A Morbillivirus Infection Shifts DC Maturation Toward a Tolerogenic Phenotype to Suppress T Cell Activation. J Virol 2022; 96:e0124022. [PMID: 36094317 PMCID: PMC9517701 DOI: 10.1128/jvi.01240-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Viruses have evolved numerous strategies to impair immunity so that they can replicate more efficiently. Among those, the immunosuppressive effects of morbillivirus infection can be particularly problematic, as they allow secondary infections to take hold in the host, worsening disease prognosis. In the present work, we hypothesized that the highly contagious morbillivirus peste des petits ruminants virus (PPRV) could target monocytes and dendritic cells (DC) to contribute to the immunosuppressive effects produced by the infection. Monocytes isolated from healthy sheep, a natural host of the disease, were able be infected by PPRV and this impaired the differentiation and phagocytic ability of immature monocyte-derived DC (MoDC). We also assessed PPRV capacity to infect differentiated MoDC. Ovine MoDC could be productively infected by PPRV, and this drastically reduced MoDC capacity to activate allogeneic T cell responses. Transcriptomic analysis of infected MoDC indicated that several tolerogenic DC signature genes were upregulated upon PPRV infection. Furthermore, PPRV-infected MoDC could impair the proliferative response of autologous CD4+ and CD8+ T cell to the mitogen concanavalin A (ConA), which indicated that DC targeting by the virus could promote immunosuppression. These results shed new light on the mechanisms employed by morbillivirus to suppress the host immune responses. IMPORTANCE Morbilliviruses pose a threat to global health given their high infectivity. The morbillivirus peste des petits ruminants virus (PPRV) severely affects small-ruminant-productivity and leads to important economic losses in communities that rely on these animals for subsistence. PPRV produces in the infected host a period of severe immunosuppression that opportunistic pathogens exploit, which worsens the course of the infection. The mechanisms of PPRV immunosuppression are not fully understood. In the present work, we demonstrate that PPRV can infect professional antigen-presenting cells called dendritic cells (DC) and disrupt their capacity to elicit an immune response. PPRV infection promoted a DC activation profile that favored the induction of tolerance instead of the activation of an antiviral immune response. These results shed new light on the mechanisms employed by morbilliviruses to suppress the immune responses.
Collapse
|
2
|
McDaniel MM, Meibers HE, Pasare C. Innate control of adaptive immunity and adaptive instruction of innate immunity: bi-directional flow of information. Curr Opin Immunol 2021; 73:25-33. [PMID: 34425435 PMCID: PMC8648974 DOI: 10.1016/j.coi.2021.07.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 07/14/2021] [Accepted: 07/28/2021] [Indexed: 12/21/2022]
Abstract
The ability of the innate and adaptive immune systems to communicate with each other is central to protective immune responses and maintenance of host health. Myeloid cells of the innate immune system are able to sense microbial ligands, perturbations in cellular homeostasis, and virulence factors, thereby allowing them to relay distinct pathogen-specific information to naïve T cells in the form of pathogen-derived peptides and a unique cytokine milieu. Once primed, effector T helper cells produce lineage-defining cytokines to help combat the original pathogen, and a subset of these cells persist as memory or effector-memory populations. These memory T cells then play a dual role in host protection by not only responding rapidly to reinfection, but by also directly instructing myeloid cells to express licensing cytokines. This means there is a bi-directional flow of information first from the innate to the adaptive immune system, and then from the adaptive back to innate immune system. Here, we focus on how signals, first from pathogens and then from primed effector and memory T cells, are integrated by myeloid cells and its consequences for protective immunity or systemic inflammation.
Collapse
Affiliation(s)
- Margaret M McDaniel
- Immunology Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States
| | - Hannah E Meibers
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45220, United States
| | - Chandrashekhar Pasare
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, United States; Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45220, United States.
| |
Collapse
|
3
|
Xie S, Liang Z, Yang X, Pan J, Yu D, Li T, Cao R. Japanese Encephalitis Virus NS2B-3 Protein Complex Promotes Cell Apoptosis and Viral Particle Release by Down-Regulating the Expression of AXL. Virol Sin 2021; 36:1503-1519. [PMID: 34487337 DOI: 10.1007/s12250-021-00442-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/12/2021] [Indexed: 12/22/2022] Open
Abstract
Japanese encephalitis virus (JEV) is a flavivirus transmitted by mosquitoes that causes severe encephalitis in humans and animals. It has been suggested that AXL, a transmembrane protein, can promote the replication of various flaviviruses, such as dengue (DENV), Zika (ZIKV), and West Nile (WNV) viruses. However, the effect of AXL on JEV infection has not yet been determined. In the present study, we demonstrate that AXL is down-regulated after JEV infection in the late stage. JEV NS2B-3 protein specifically interacted with AXL, and promoted AXL degradation through the ubiquitin-proteasome pathway. AXL-degradation increased cell apoptosis by disrupting phosphatidylinositol 3-kinase (PI3K)/Akt signal transduction. In addition, the degradation of AXL promoted JEV release to supernatant, whereas the virus in the cell lysates decreased. The supplementation of AXL ligand Gas6 inhibited the JEV-mediated degradation of AXL. Altogether, we discover a new function of NS2B-3 during the process of JEV replication, and provide a new insight into the interactions between JEV and cell hosts.
Collapse
Affiliation(s)
- Shengda Xie
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zhenjie Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xingmiao Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Junhui Pan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Du Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Tongtong Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Ruibing Cao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
4
|
Nan FL, Zheng W, Nan WL, Yu T, Xie CZ, Zhang H, Xu XH, Li CH, Ha Z, Zhang JY, Zhuang XY, Han JC, Wang W, Qian J, Zhao GY, Li ZX, Ge JY, Bu ZG, Zhang Y, Lu HJ, Jin NY. Newcastle Disease Virus Inhibits the Proliferation of T Cells Induced by Dendritic Cells In Vitro and In Vivo. Front Immunol 2021; 11:619829. [PMID: 33708193 PMCID: PMC7942023 DOI: 10.3389/fimmu.2020.619829] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/29/2020] [Indexed: 12/03/2022] Open
Abstract
Newcastle disease virus (NDV) infects poultry and antagonizes host immunity via several mechanisms. Dendritic cells (DCs) are characterized as specialized antigen presenting cells, bridging innate and adaptive immunity and regulating host resistance to viral invasion. However, there is little specific knowledge of the role of DCs in NDV infection. In this study, the representative NDV lentogenic strain LaSota was used to explore whether murine bone marrow derived DCs mature following infection. We examined surface molecule expression and cytokine release from DCs as well as proliferation and activation of T cells in vivo and in vitro in the context of NDV. The results demonstrated that infection with lentogenic strain LaSota induced a phenotypic maturation of immature DCs (imDCs), which actually led to curtailed T cell responses. Upon infection, the phenotypic maturation of DCs was reflected by markedly enhanced MHC and costimulatory molecule expression and secretion of proinflammatory cytokines. Nevertheless, NDV-infected DCs produced the anti-inflammatory cytokine IL-10 and attenuated T cell proliferation, inducing Th2-biased responses. Therefore, our study reveals a novel understanding that DCs are phenotypically mature but dysfunctional in priming T cell responses during NDV infection.
Collapse
Affiliation(s)
- Fu Long Nan
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Wei Zheng
- The 964Hospital of the PLA Joint Logistics, Changchun, China
| | - Wen Long Nan
- China Animal Health and Epidemiology Center, Qingdao, China
| | - Tong Yu
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China.,Agricultural College, Yanbian University, Yanji, China
| | - Chang Zhan Xie
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - He Zhang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Xiao Hong Xu
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, China
| | - Cheng Hui Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Zhuo Ha
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Jin Yong Zhang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Xin Yu Zhuang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Ji Cheng Han
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China.,Academician Workstation, Changchun University of Chinese Medicine, Changchun, China
| | - Wei Wang
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Jing Qian
- Institute of Veterinary Medicine, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Guan Yu Zhao
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Zhuo Xin Li
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Jin Ying Ge
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Zhi Gao Bu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Ying Zhang
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, China
| | - Hui Jun Lu
- Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| | - Ning Yi Jin
- College of Veterinary Medicine, College of Animal Science, Jilin University, Changchun, China.,Institute of Military Veterinary Medicine, Academy of Military Medical Sciences, Changchun, China
| |
Collapse
|
5
|
Sahoo M, M D, Thakor JC, Baloni S, Saxena S, Shrivastava S, Dhama K, Singh K, Singh R. Neuropathology mediated through caspase dependent extrinsic pathway in goat kids naturally infected with PPRV. Microb Pathog 2019; 140:103949. [PMID: 31875517 DOI: 10.1016/j.micpath.2019.103949] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 11/18/2019] [Accepted: 12/21/2019] [Indexed: 12/13/2022]
Abstract
Peste des petits ruminant (PPR), a highly contagious viral disease of small ruminants, is characterized by erosive stomatitis and pneumo-enteritis. However, its neurovirulence potential as observed with other morbilliviruses has not been fully investigated. The present study describes the neuropathological alterations induced by PPR virus through apoptotic pathway. A total number of 12 carcasses of local breed goat kids of either sex were received for postmortem examination. The clinical history was described as symptoms of mucopurulent nasal discharge, high to low grade fever, erosive stomatitis, dyspnoea and profuse watery diarrhoea followed by mortality of 35 goat kids within a week. The pathoanatomical lesions and immunohistochemical demonstration of PPRV antigen in lungs, intestine, spleen and lymph nodes confirmed PPR disease in goats. Grossly, five brain specimens showed moderate to severe leptomeningeal congestion during necropsy. Microscopically, brain sections showed leptomeningitis and nonsuppurative encephalitis characterized by vascular congestion, haemorrhages in the parenchyma, perivascular cuffing with mild to moderate mononuclear cells (mainly lymphocytes and few macrophages), focal to diffuse microgliosis, neuronal degeneration, satellitosis and neuronophagia. Immunolabelling of viral antigen was observed in the cytoplasm of neurons and glial cells. The RT-PCR amplification of N gene fragment also confirmed the presence of PPRV in the brain. The strong immunoreactivity of Caspase-3, Caspase-8 and comparatively lower expression of caspase-9 along with the absence of any reactivity for Apaf-1 antigen in the brain sections indicated the role of caspase dependent extrinsic pathway in inducing neuropathological changes. The presence of apoptotic neurons in the brain by TUNEL assay further confirmed the apoptosis and strong immunoreactivity of iNOS in neurons which suggested the generation of oxidative stress, that might have induced the apoptosis. The overall findings confirm the neurovirulence potential of PPR virus, via the extrinsic pathway of apoptosis, in natural cases of PPR disease in goat kids.
Collapse
Affiliation(s)
- Monalisa Sahoo
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, India.
| | - Dinesh M
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, India
| | | | - Suraj Baloni
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, India
| | - Sonal Saxena
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, India
| | - Sameer Shrivastava
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, India
| | - Karampal Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, India
| | - Rajendra Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, India
| |
Collapse
|
6
|
Xu Q, Rangaswamy US, Wang W, Robbins SH, Harper J, Jin H, Cheng X. Evaluation of Newcastle disease virus mediated dendritic cell activation and cross-priming tumor-specific immune responses ex vivo. Int J Cancer 2019; 146:531-541. [PMID: 31584185 DOI: 10.1002/ijc.32694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/03/2019] [Accepted: 09/05/2019] [Indexed: 12/13/2022]
Abstract
We have developed an oncolytic Newcastle disease virus (NDV) that has potent in vitro and in vivo anti-tumor activities and attenuated pathogenicity in chickens. In this ex vivo study using the same recombinant NDV backbone with GFP transgene (NDV-GFP, designated as rNDV), we found that rNDV induces maturation of monocyte-derived immature dendritic cells (iDCs) by both direct and indirect mechanisms, which promote development of antigen-specific T cell responses. Addition of rNDV directly to iDCs culture induced DC maturation, as demonstrated by the increased expression of costimulatory and antigen-presenting molecules as well as the production of type I interferons (IFNs). rNDV infection of the HER-2 positive human breast cancer cell line (SKBR3) resulted in apoptotic cell death, release of proinflammatory cytokines, and danger-associated molecular pattern molecules (DAMPs) including high-mobility group protein B1 (HMGB1) and heat shock protein 70 (HSP70). Addition of rNDV-infected SKBR3 cells to iDC culture resulted in greatly enhanced upregulation of the maturation markers and release of type I IFNs by DCs than rNDV-infected DCs only. When co-cultured with autologous T cells, DCs pre-treated with rNDV-infected SKBR3 cells cross-primed T cells in an antigen-specific manner. Altogether, our data strongly support the potential of oncolytic NDV as efficient therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Qi Xu
- Microbial Sciences, AstraZeneca Ltd, South San Francisco, CA
| | | | - Weijia Wang
- Microbial Sciences, AstraZeneca Ltd, South San Francisco, CA
| | | | - James Harper
- Oncology R&D, AstraZeneca Ltd, Cambridge, United Kingdom
| | - Hong Jin
- Microbial Sciences, AstraZeneca Ltd, South San Francisco, CA
| | - Xing Cheng
- Microbial Sciences, AstraZeneca Ltd, South San Francisco, CA
| |
Collapse
|
7
|
Derakhshani S, Kurz A, Japtok L, Schumacher F, Pilgram L, Steinke M, Kleuser B, Sauer M, Schneider-Schaulies S, Avota E. Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium. Front Immunol 2019; 10:1294. [PMID: 31231395 PMCID: PMC6560165 DOI: 10.3389/fimmu.2019.01294] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/21/2019] [Indexed: 12/16/2022] Open
Abstract
Transmission of measles virus (MV) from dendritic to airway epithelial cells is considered as crucial to viral spread late in infection. Therefore, pathways and effectors governing this process are promising targets for intervention. To identify these, we established a 3D respiratory tract model where MV transmission by infected dendritic cells (DCs) relied on the presence of nectin-4 on H358 lung epithelial cells. Access to recipient cells is an important prerequisite for transmission, and we therefore analyzed migration of MV-exposed DC cultures within the model. Surprisingly, enhanced motility toward the epithelial layer was observed for MV-infected DCs as compared to their uninfected siblings. This occurred independently of factors released from H358 cells indicating that MV infection triggered cytoskeletal remodeling associated with DC polarization enforced velocity. Accordingly, the latter was also observed for MV-infected DCs in collagen matrices and was particularly sensitive to ROCK inhibition indicating infected DCs preferentially employed the amoeboid migration mode. This was also implicated by loss of podosomes and reduced filopodial activity both of which were retained in MV-exposed uninfected DCs. Evidently, sphingosine kinase (SphK) and sphingosine-1-phosphate (S1P) as produced in response to virus-infection in DCs contributed to enhanced velocity because this was abrogated upon inhibition of sphingosine kinase activity. These findings indicate that MV infection promotes a push-and-squeeze fast amoeboid migration mode via the SphK/S1P system characterized by loss of filopodia and podosome dissolution. Consequently, this enables rapid trafficking of virus toward epithelial cells during viral exit.
Collapse
Affiliation(s)
| | - Andreas Kurz
- Department for Biotechnology and Biophysics, University of Wuerzburg, Wuerzburg, Germany
| | - Lukasz Japtok
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Fabian Schumacher
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany.,Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Lisa Pilgram
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| | - Maria Steinke
- Fraunhofer Institute for Silicate Research ISC, Chair of Tissue Engineering and Regenerative Medicine, University Hospital Wuerzburg, University of Wuerzburg, Wuerzburg, Germany
| | - Burkhard Kleuser
- Department of Toxicology, Institute of Nutritional Science, University of Potsdam, Nuthetal, Germany
| | - Markus Sauer
- Department for Biotechnology and Biophysics, University of Wuerzburg, Wuerzburg, Germany
| | | | - Elita Avota
- Institute for Virology and Immunobiology, University of Wuerzburg, Wuerzburg, Germany
| |
Collapse
|
8
|
Son HA, Zhang L, Cuong BK, Van Tong H, Cuong LD, Hang NT, Nhung HTM, Yamamoto N, Toan NL. Combination of Vaccine-Strain Measles and Mumps Viruses Enhances Oncolytic Activity against Human Solid Malignancies. Cancer Invest 2018; 36:106-117. [PMID: 29485292 DOI: 10.1080/07357907.2018.1434539] [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] [Indexed: 01/11/2023]
Abstract
Oncolytic measles and mumps viruses (MeV, MuV) have a potential for anti-cancer treatment. We examined the anti-tumor activity of MeV, MuV, and MeV-MuV combination (MM) against human solid malignancies (HSM). MeV, MuV, and MM targeted and significantly killed various cancer cell lines of HSM but not normal cells. MM demonstrated a greater anti-tumor effect and prolonged survival in a human prostate cancer xenograft tumor model compared to MeV and MuV. MeV, MuV, and MM significantly induced the expression of immunogenic cell death markers and enhanced spleen-infiltrating immune cells. In conclusion, MM combination significantly improves the treatment of human solid malignancies.
Collapse
Affiliation(s)
- Ho Anh Son
- a Department of Pathophysiology , Vietnam Military Medical University , Hanoi , Vietnam
| | - LiFeng Zhang
- b Department of Microbiology , Yong Loo Lin School of Medicine , National University of Singapore , Singapore.,e Department of immunology , Guangxi University of Chinese Medicine , Nanning, Guangxi Zhuang autonomous region, China
| | - Bui Khac Cuong
- a Department of Pathophysiology , Vietnam Military Medical University , Hanoi , Vietnam
| | - Hoang Van Tong
- a Department of Pathophysiology , Vietnam Military Medical University , Hanoi , Vietnam.,c Institute of Biomedicine and Pharmacy , Vietnam Military Medical University , Hanoi , Vietnam
| | - Le Duy Cuong
- a Department of Pathophysiology , Vietnam Military Medical University , Hanoi , Vietnam
| | - Ngo Thu Hang
- a Department of Pathophysiology , Vietnam Military Medical University , Hanoi , Vietnam
| | | | - Naoki Yamamoto
- b Department of Microbiology , Yong Loo Lin School of Medicine , National University of Singapore , Singapore
| | - Nguyen Linh Toan
- a Department of Pathophysiology , Vietnam Military Medical University , Hanoi , Vietnam
| |
Collapse
|
9
|
George JA, Kim SB, Choi JY, Patil AM, Hossain FMA, Uyangaa E, Hur J, Park SY, Lee JH, Kim K, Eo SK. TLR2/MyD88 pathway-dependent regulation of dendritic cells by dengue virus promotes antibody-dependent enhancement via Th2-biased immunity. Oncotarget 2017; 8:106050-106070. [PMID: 29285314 PMCID: PMC5739701 DOI: 10.18632/oncotarget.22525] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 10/28/2017] [Indexed: 12/21/2022] Open
Abstract
Possible risk mediators in primary dengue virus (DenV) infection that favor secondary DenV infection to life-threatening dengue hemorrhagic fever (DHF) and shock syndrome (DSS) via antibody-dependent enhancement (ADE) have not yet been described. Here, DenV infection enhanced the expression of inflammatory mediators and activation molecules in dendritic cells (DCs) through TLR2/MyD88 pathway. TLR2 appeared to facilitate DenV infection in DCs that were less permissive than macrophages for viral replication. In experiments using separate evaluations of DenV-infected and uninfected bystander DCs, infected DCs showed impaired maturation accompanied with TLR2-dependent production of inflammatory cytokines, by which uninfected bystander DCs showed increased expression of co-stimulatory molecules. Differential phosphorylation of MAPK and STAT3 was also detected between DenV-infected and uninfected DCs. Furthermore, DenV infection stimulated Th2-polarized humoral and cellular immunity against foreign and DenV Ag via TLR2/MyD88 pathway, and DenV-infected DCs were revealed to facilitate Th2-biased immune responses in TLR2-dependent manner. TLR2/MyD88-mediated Th2-biased Ab responses to primary DenV infection increased the infectivity of secondary homotypic or heterotypic DenV via ADE. Collectively, these results indicate that TLR2/MyD88 pathway in DC-priming receptors can drive Th2-biased immune responses during primary DenV infection, which could favor secondary DenV infection to DHF/DSS via ADE.
Collapse
Affiliation(s)
- Junu Aleyas George
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Seong Bum Kim
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Jin Young Choi
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Ajit Mahadev Patil
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Ferdaus Mohd Altaf Hossain
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Erdenebelig Uyangaa
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Jin Hur
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Sang-Youel Park
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| | - John-Hwa Lee
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Seong Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Chonbuk National University, Iksan 54596, Republic of Korea
| |
Collapse
|
10
|
Does Zika virus infection induce prolonged remissions in children with idiopathic nephrotic syndrome? Pediatr Nephrol 2017; 32:897-900. [PMID: 28175986 DOI: 10.1007/s00467-017-3588-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2016] [Revised: 01/03/2017] [Accepted: 01/04/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Zika is an emerging mosquito-borne flavivirus. We report two pediatric patients diagnosed with idiopathic nephrotic syndrome who achieved complete remission of the disease after suffering Zika virus (ZIKV) infection. CASE DIAGNOSIS/TREATMENT The first patient was a young girl aged 2.5 years with steroid-dependent nephrotic syndrome who was subsequently diagnosed with ZIKV infection. Following the infection, the steroid dose could be reduced until complete withdrawal. The patient persists in complete remission. The second patient was a steroid-resistant boy aged 7 years who was scheduled for a renal biopsy when he was diagnosed with ZIKV infection. A week after the recovery phase of the acute rash, proteinuria was noted to be gradually falling. Today, 12 months later, he is in complete remission of the disease. CONCLUSIONS We are aware that the improvement observed in our two patients after ZIKV infection may be be random. However, it is also possible that future studies will discover that ZIKV infection has some effect on the cellular immune system similar to that of measles infection.
Collapse
|
11
|
Oreshkova N, Wichgers Schreur PJ, Spel L, Vloet RPM, Moormann RJM, Boes M, Kortekaas J. Nonspreading Rift Valley Fever Virus Infection of Human Dendritic Cells Results in Downregulation of CD83 and Full Maturation of Bystander Cells. PLoS One 2015; 10:e0142670. [PMID: 26575844 PMCID: PMC4648518 DOI: 10.1371/journal.pone.0142670] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 10/26/2015] [Indexed: 01/08/2023] Open
Abstract
Vaccines based on nonspreading Rift Valley fever virus (NSR) induce strong humoral and robust cellular immune responses with pronounced Th1 polarisation. The present work was aimed to gain insight into the molecular basis of NSR-mediated immunity. Recent studies have demonstrated that wild-type Rift Valley fever virus efficiently targets and replicates in dendritic cells (DCs). We found that NSR infection of cultured human DCs results in maturation of DCs, characterized by surface upregulation of CD40, CD80, CD86, MHC-I and MHC-II and secretion of the proinflammatory cytokines IFN-β, IL-6 and TNF. Interestingly, expression of the most prominent marker of DC maturation, CD83, was consistently downregulated at 24 hours post infection. Remarkably, NSR infection also completely abrogated CD83 upregulation by LPS. Downregulation of CD83 was not associated with reduced mRNA levels or impaired CD83 mRNA transport from the nucleus and could not be prevented by inhibition of the proteasome or endocytic degradation pathways, suggesting that suppression occurs at the translational level. In contrast to infected cells, bystander DCs displayed full maturation as evidenced by upregulation of CD83. Our results indicate that bystander DCs play an important role in NSR-mediated immunity.
Collapse
Affiliation(s)
- Nadia Oreshkova
- Department of Virology, Central Veterinary Institute, part of Wageningen University and Research Centre, Lelystad, The Netherlands
- Department of Infectious Diseases and Immunology, Virology Division, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Paul J. Wichgers Schreur
- Department of Virology, Central Veterinary Institute, part of Wageningen University and Research Centre, Lelystad, The Netherlands
| | - Lotte Spel
- Department of Pediatric Immunology and Laboratory of Translational Immunology, University Medical Centre Utrecht/Wilhelmina Children’s Hospital, Utrecht, The Netherlands
| | - Rianka P. M. Vloet
- Department of Virology, Central Veterinary Institute, part of Wageningen University and Research Centre, Lelystad, The Netherlands
| | - Rob J. M. Moormann
- Department of Virology, Central Veterinary Institute, part of Wageningen University and Research Centre, Lelystad, The Netherlands
- Department of Infectious Diseases and Immunology, Virology Division, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Marianne Boes
- Department of Pediatric Immunology and Laboratory of Translational Immunology, University Medical Centre Utrecht/Wilhelmina Children’s Hospital, Utrecht, The Netherlands
| | - Jeroen Kortekaas
- Department of Virology, Central Veterinary Institute, part of Wageningen University and Research Centre, Lelystad, The Netherlands
| |
Collapse
|
12
|
Kubicka-Sierszen A, Grzegorczyk JŁ. The influence of infectious factors on dendritic cell apoptosis. Arch Med Sci 2015; 11:1044-51. [PMID: 26528349 PMCID: PMC4624750 DOI: 10.5114/aoms.2015.54860] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/04/2013] [Accepted: 10/02/2013] [Indexed: 12/23/2022] Open
Abstract
Pathogens can have a negative influence on dendritic cells (DCs), causing their apoptosis, which prevents active presentation of foreign antigens. It results in a state of immunosuppression which makes the body susceptible to secondary infections. Infected immature DCs have lower expression of co-stimulatory and adhesion molecules, reduced ability to secrete cytokines and an inhibited maturation process and are incapable of effective antigen presentation and activation of T-lymphocytes. In some cases, the ability of DCs to undergo rapid apoptosis is important for the body defense, which is probably because of DCs' ability to cross-present and cooperate with other cells. Apoptotic bodies released from the infected DCs are phagocytosed by other DCs, which then stimulate the effector cells and present antigens more efficiently than infected cells. The aim of this article is to review how the DCs respond to viral and bacterial factors and which biochemical mechanisms are responsible for their apoptosis.
Collapse
Affiliation(s)
- Agata Kubicka-Sierszen
- Department of Microbiology and Laboratory Medical Immunology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Janina Ł Grzegorczyk
- Department of Microbiology and Laboratory Medical Immunology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
13
|
Oh IS, Park SH. Immune-mediated Liver Injury in Hepatitis B Virus Infection. Immune Netw 2015; 15:191-8. [PMID: 26330805 PMCID: PMC4553257 DOI: 10.4110/in.2015.15.4.191] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Revised: 07/26/2015] [Accepted: 08/02/2015] [Indexed: 12/29/2022] Open
Abstract
Hepatitis B virus (HBV) is responsible for approximately 350 million chronic infections worldwide and is a leading cause of broad-spectrum liver diseases such as hepatitis, cirrhosis and liver cancer. Although it has been well established that adaptive immunity plays a critical role in viral clearance, the pathogenetic mechanisms that cause liver damage during acute and chronic HBV infection remain largely known. This review describes our current knowledge of the immune-mediated pathogenesis of HBV infection and the role of immune cells in the liver injury during hepatitis B.
Collapse
Affiliation(s)
- In Soo Oh
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Korea. ; Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul 06973, Korea
| | - Su-Hyung Park
- Laboratory of Translational Immunology and Vaccinology, Graduate School of Medical Science and Engineering, KAIST, Daejeon 34141, Korea
| |
Collapse
|
14
|
|
15
|
Zhang LF, Tan DQC, Jeyasekharan AD, Hsieh WS, Ho AS, Ichiyama K, Ye M, Pang B, Ohba K, Liu X, de Mel S, Cuong BK, Chng WJ, Ryo A, Suzuki Y, Yeoh KG, Toan NL, Yamamoto N. Combination of vaccine-strain measles and mumps virus synergistically kills a wide range of human hematological cancer cells: Special focus on acute myeloid leukemia. Cancer Lett 2014; 354:272-80. [PMID: 25193462 DOI: 10.1016/j.canlet.2014.08.034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 08/14/2014] [Accepted: 08/19/2014] [Indexed: 11/28/2022]
Abstract
Through combining vaccine-derived measles and mumps viruses (MM), we efficiently targeted a wide range of hematopoietic cancer cell lines. MM synergistically killed many cell lines including acute myeloid leukemia (AML) cell lines. Further investigation suggested that enhanced oncolytic effect of MM was due to increased apoptosis induction. In an U937 xenograft AML mouse model, MM displayed greater tumor suppression and prolonged survival. Furthermore, MM efficiently killed blasts from 16 out of 20 AML patients and elicited more efficient killing effect on 11 patients when co-administered with Ara-C. Our results demonstrate that MM is a promising therapeutic candidate for hematological malignancies.
Collapse
Affiliation(s)
- Li Feng Zhang
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Darren Qian Cheng Tan
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - Wen Son Hsieh
- Department of Otolaryngology - Head and Neck Surgery, National University Health System, Singapore; Cancer Science Institute, National University of Singapore, Singapore
| | - Anh Son Ho
- Department of Pathophysiology, Vietnam Military Medical University, Vietnam
| | - Koji Ichiyama
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Min Ye
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Brendan Pang
- Department of Pathology, National University Health System and National University of Singapore, Singapore
| | - Kenji Ohba
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Xin Liu
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Sanjay de Mel
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Bui Khac Cuong
- Department of Pathophysiology, Vietnam Military Medical University, Vietnam
| | - Wee Joo Chng
- Cancer Science Institute, National University of Singapore, Singapore; Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Akihide Ryo
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Youichi Suzuki
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Khay Guan Yeoh
- Department of Microbiology, Yokohama City University School of Medicine, Yokohama, Japan
| | - Nguyen Linh Toan
- Department of Pathophysiology, Vietnam Military Medical University, Vietnam
| | - Naoki Yamamoto
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
16
|
Loss of the death receptor CD95 (Fas) expression by dendritic cells protects from a chronic viral infection. Proc Natl Acad Sci U S A 2014; 111:8559-64. [PMID: 24912151 DOI: 10.1073/pnas.1401750111] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chronic viral infections incapacitate adaptive immune responses by "exhausting" virus-specific T cells, inducing their deletion and reducing productive T-cell memory. Viral infection rapidly induces death receptor CD95 (Fas) expression by dendritic cells (DCs), making them susceptible to elimination by the immune response. Lymphocytic choriomeningitis virus (LCMV) clone 13, which normally establishes a chronic infection, is rapidly cleared in C57Black6/J mice with conditional deletion of Fas in DCs. The immune response to LCMV is characterized by an extended survival of virus-specific effector T cells. Moreover, transfer of Fas-negative DCs from noninfected mice to preinfected animals results in either complete clearance of the virus or a significant reduction of viral titers. Thus, DC-specific Fas expression plays a role in regulation of antiviral responses and suggests a strategy for stimulation of T cells in chronically infected animals and humans to achieve the clearance of persistent viruses.
Collapse
|
17
|
Samrat SK, Li W, Singh S, Kumar R, Agrawal B. Alternate reading frame protein (F protein) of hepatitis C virus: paradoxical effects of activation and apoptosis on human dendritic cells lead to stimulation of T cells. PLoS One 2014; 9:e86567. [PMID: 24475147 PMCID: PMC3903568 DOI: 10.1371/journal.pone.0086567] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 12/11/2013] [Indexed: 12/24/2022] Open
Abstract
Hepatitis C virus (HCV) leads to chronic infection in the majority of infected individuals due to lack, failure, or inefficiency of generated adaptive immune responses. In a minority of patients, acute infection is followed by viral clearance. The immune correlates of viral clearance are not clear yet but have been extensively investigated, suggesting that multispecific and multifunctional cellular immunity is involved. The generation of cellular immunity is highly dependent upon how antigen presenting cells (APCs) process and present various viral antigens. Various structural and non-structural HCV proteins derived from the open reading frame (ORF) have been implicated in modulation of dendritic cells (DCs) and APCs. Besides the major ORF proteins, the HCV core region also encodes an alternate reading frame protein (ARFP or F), whose function in viral pathogenesis is not clear. In the current studies, we sought to determine the role of HCV-derived ARFP in modulating dendritic cells and stimulation of T cell responses. Recombinant adenovirus vectors containing F or core protein derived from HCV (genotype 1a) were prepared and used to endogenously express these proteins in dendritic cells. We made an intriguing observation that endogenous expression of F protein in human DCs leads to contrasting effects on activation and apoptosis of DCs, allowing activated DCs to efficiently internalize apoptotic DCs. These in turn result in efficient ability of DCs to process and present antigen and to prime and stimulate F protein derived peptide-specific T cells from HCV-naive individuals. Taken together, our findings suggest important aspects of F protein in modulating DC function and stimulating T cell responses in humans.
Collapse
Affiliation(s)
- Subodh Kumar Samrat
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Wen Li
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shakti Singh
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Rakesh Kumar
- Department of Laboratory Medicine and Pathology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- * E-mail:
| |
Collapse
|
18
|
Yi C, Liu X, Liu Y, Lu S, Qi Y. Hemagglutinin protein of measles virus induces apoptosis of HeLa cells via both extrinsic and intrinsic pathways. Can J Microbiol 2013; 59:814-24. [PMID: 24313454 DOI: 10.1139/cjm-2013-0544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In this study, we investigated the potential for different components of the measles virus (MV) to induce apoptosis of HeLa cells and explored the apoptotic molecular mechanisms. After testing the 2 envelope glycoproteins hemagglutinin (H) and fusion (F), we found that MV H alone was sufficient to induce the apoptosis of HeLa cells, whereas MV F did not. MV F also had no influence on MV-H-mediated apoptosis. MV H could induce cellular apoptosis in HeLa cells through its interaction with the cellular receptor CD46 via both the TRAIL-mediated extrinsic pathway and the mitochondria-controlled intrinsic pathway, and that cross talk between these 2 pathways occurred during the process. These findings extend the functions of MV envelope glycoproteins in the pathogenesis of MV infection and suggest that MV H may be a potential therapeutic in the treatment of some cancers.
Collapse
Affiliation(s)
- Changhua Yi
- a State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, People's Republic of China
| | | | | | | | | |
Collapse
|
19
|
Gupta M, Lo MK, Spiropoulou CF. Activation and cell death in human dendritic cells infected with Nipah virus. Virology 2013; 441:49-56. [PMID: 23587249 DOI: 10.1016/j.virol.2013.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 11/13/2012] [Accepted: 03/04/2013] [Indexed: 01/23/2023]
Abstract
Nipah virus (NiV) is a highly pathogenic paramyxovirus that causes pulmonary disease and encephalitis in humans with 40-70% fatality. Interactions between NiV and the human immune system remain poorly understood. Here, we demonstrate the effects of NiV infection on DC and T cell function. Using an in vitro system, we found that NiV infects and replicates at low levels in DCs and induces the expression of TNF-α, IL-1α, IL-1β, IL-8, and IP-10. NiV infection activates DCs, and upregulates the expression of CD40, CD80, and CD86. Also have reduced levels of bcl2 and high levels of active caspase 3, suggesting the induction of apoptosis. DCs infected by NiV are unable to efficiently prime CD4 and CD8 T cells, but instead induce apoptosis in T cells. Interestingly, DCs treated with inactivated NiV also show signs of apoptosis. These findings indicate that NiV infected DCs could play an important role in NiV pathogenesis.
Collapse
Affiliation(s)
- Manisha Gupta
- Viral Special Pathogens Branch, NCEZID, DHCPP, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA.
| | | | | |
Collapse
|
20
|
Zhang SC, Cai WS, Zhang Y, Jiang KL, Zhang KR, Wang WL. Engineered measles virus Edmonston strain used as a novel oncolytic viral system against human neuroblastoma through a CD46 and nectin 4-independent pathway. Cancer Lett 2012; 325:227-37. [PMID: 22796607 DOI: 10.1016/j.canlet.2012.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 07/04/2012] [Accepted: 07/04/2012] [Indexed: 10/28/2022]
Abstract
Neuroblastoma (NB) is the most common extracranial solid tumor in children. In this study, we investigated the potential antitumor capability of the engineered Edmonston strain of the carcinoembryonic antigen-expressing measles virus (MV-CEA) against human NB. The infection of a variety of NB cell lines, including SK-N-SH, SMS-KCNR, and primary NB cells, resulted in significant cytopathic effects. None of the NB cell lines showed an overexpression of the measles virus receptor CD46 and nectin 4, but the cell lines did support robust viral replication. The efficacy of this approach was examined in murine SK-N-SH xenograft models. Flow cytometry and TUNEL assays indicated an apoptotic mechanism of cell death. In summary, MV-CEA has potent therapeutic efficacy against NB mediated by a CD46- and nectin 4-independent pathway.
Collapse
Affiliation(s)
- Shu-Cheng Zhang
- Department of Pediatric Surgery, Major Laboratory of the Chinese Health Ministry for Congenital Malformations, Shengjing Hospital of China Medical University, Heping District, Shenyang, PR China
| | | | | | | | | | | |
Collapse
|
21
|
Coughlin MM, Bellini WJ, Rota PA. Contribution of dendritic cells to measles virus induced immunosuppression. Rev Med Virol 2012; 23:126-38. [DOI: 10.1002/rmv.1735] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 10/29/2012] [Accepted: 10/30/2012] [Indexed: 12/19/2022]
Affiliation(s)
- Melissa M. Coughlin
- Centers for Disease Control and Prevention, Measles, Mumps, Rubella and Herpesvirus Laboratory Branch; Atlanta GA USA
| | - William J. Bellini
- Centers for Disease Control and Prevention, Measles, Mumps, Rubella and Herpesvirus Laboratory Branch; Atlanta GA USA
| | - Paul A. Rota
- Centers for Disease Control and Prevention, Measles, Mumps, Rubella and Herpesvirus Laboratory Branch; Atlanta GA USA
| |
Collapse
|
22
|
Abstract
Dengue viruses and other members of the Flaviviridae family are emerging human pathogens. Dengue is transmitted to humans by Aedes aegypti female mosquitoes. Following infection through the bite, cells of the hematopoietic lineage, like dendritic cells, are the first targets of dengue virus infection. Dendritic cells (DCs) are key antigen presenting cells, sensing pathogens, processing and presenting the antigens to T lymphocytes, and triggering an adaptive immune response. Infection of DCs by dengue virus may induce apoptosis, impairing their ability to present antigens to T cells, and thereby contributing to dengue pathogenesis. This review focuses on general mechanisms by which dengue virus triggers apoptosis, and possible influence of DC-apoptosis on dengue disease severity.
Collapse
|
23
|
Zhang SC, Wang WL, Cai WS, Jiang KL, Yuan ZW. Engineered measles virus Edmonston strain used as a novel oncolytic viral system against human hepatoblastoma. BMC Cancer 2012; 12:427. [PMID: 23009685 PMCID: PMC3488522 DOI: 10.1186/1471-2407-12-427] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 09/23/2012] [Indexed: 12/26/2022] Open
Abstract
Background Hepatoblastoma (HB) is the most common primary, malignant pediatric liver tumor in children. The treatment results for affected children have markedly improved in recent decades. However, the prognosis for high-risk patients who have extrahepatic extensions, invasion of the large hepatic veins, distant metastases and very high alpha-fetoprotein (AFP) serum levels remains poor. There is an urgent need for the development of novel therapeutic approaches. Methods An attenuated strain of measles virus, derived from the Edmonston vaccine lineage, was genetically engineered to produce carcinoembryonic antigen (CEA). We investigated the antitumor potential of this novel viral agent against human HB both in vitro and in vivo. Results Infection of the Hep2G and HUH6 HB cell lines, at multiplicities of infection (MOIs) ranging from 0.01 to 1, resulted in a significant cytopathic effect consisting of extensive syncytia formation and massive cell death at 72–96 h after infection. Both of the HB lines overexpressed the measles virus receptor CD46 and supported robust viral replication, which correlated with CEA production. The efficacy of this approach in vivo was examined in murine Hep2G xenograft models. Flow cytometry assays indicated an apoptotic mechanism of cell death. Intratumoral administration of MV-CEA resulted in statistically significant delay of tumor growth and prolongation of survival. Conclusions The engineered measles virus Edmonston strain MV-CEA has potent therapeutic efficacy against HB cell lines and xenografts. Trackable measles virus derivatives merit further exploration in HB treatment.
Collapse
Affiliation(s)
- Shu-Cheng Zhang
- Department of Pediatric Surgery, Major Laboratory of Chinese Health Ministry for Congenital Malformations, Shengjing Hospital of China Medical University, 36 Sanhao Street Heping District, Shenyang 110004, P.R. China.
| | | | | | | | | |
Collapse
|
24
|
A human coronavirus responsible for the common cold massively kills dendritic cells but not monocytes. J Virol 2012; 86:7577-87. [PMID: 22553325 DOI: 10.1128/jvi.00269-12] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Human coronaviruses are associated with upper respiratory tract infections that occasionally spread to the lungs and other organs. Although airway epithelial cells represent an important target for infection, the respiratory epithelium is also composed of an elaborate network of dendritic cells (DCs) that are essential sentinels of the immune system, sensing pathogens and presenting foreign antigens to T lymphocytes. In this report, we show that in vitro infection by human coronavirus 229E (HCoV-229E) induces massive cytopathic effects in DCs, including the formation of large syncytia and cell death within only few hours. In contrast, monocytes are much more resistant to infection and cytopathic effects despite similar expression levels of CD13, the membrane receptor for HCoV-229E. While the differentiation of monocytes into DCs in the presence of granulocyte-macrophage colony-stimulating factor and interleukin-4 requires 5 days, only 24 h are sufficient for these cytokines to sensitize monocytes to cell death and cytopathic effects when infected by HCoV-229E. Cell death induced by HCoV-229E is independent of TRAIL, FasL, tumor necrosis factor alpha, and caspase activity, indicating that viral replication is directly responsible for the observed cytopathic effects. The consequence of DC death at the early stage of HCoV-229E infection may have an impact on the early control of viral dissemination and on the establishment of long-lasting immune memory, since people can be reinfected multiple times by HCoV-229E.
Collapse
|
25
|
Abstract
Measles is a highly contagious disease caused by measles virus and is one of the most devastating infectious diseases of man--measles was responsible for millions of deaths annually worldwide before the introduction of the measles vaccines. Remarkable progress in reducing the number of people dying from measles has been made through measles vaccination, with an estimated 164,000 deaths attributed to measles in 2008. This achievement attests to the enormous importance of measles vaccination to public health. However, this progress is threatened by failure to maintain high levels of measles vaccine coverage. Recent measles outbreaks in sub-Saharan Africa, Europe, and the USA show the ease with which measles virus can re-enter communities if high levels of population immunity are not sustained. The major challenges for continued measles control and eventual eradication will be logistical, financial, and the garnering of sufficient political will. These challenges need to be met to ensure that future generations of children do not die of measles.
Collapse
Affiliation(s)
- William J Moss
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA.
| | | |
Collapse
|
26
|
Protective potential of MMR vaccine against complete Freund’s adjuvant-induced inflammation in rats. Inflammopharmacology 2011; 19:343-8. [DOI: 10.1007/s10787-011-0094-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 09/22/2011] [Indexed: 10/15/2022]
|
27
|
Del Puerto HL, Martins AS, Braz GF, Alves F, Heinemann MB, Rajão DS, Araújo FC, Martins SF, Nascimento DR, Leite RC, Vasconcelos AC. Vero cells infected with the Lederle strain of canine distemper virus have increased Fas receptor signaling expression at 15 h post-infection. GENETICS AND MOLECULAR RESEARCH 2011; 10:2527-33. [PMID: 22009866 DOI: 10.4238/2011.october.18.3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
We evaluated the expression of the Fas receptor gene in Vero cells infected with the Lederle vaccine strain of canine distemper virus using RT-PCR. Vero cells were plated, and after being grown for 24 h in MEM with 5% FBS, 80-90% confluent monolayer cultures were infected with the virus. The cells were harvested at 3, 6, 9, and 15 h post-infection. Uninfected Vero cells were used as a control. Total RNA was isolated from Vero cells using 1 mL Trizol(®) LS, and RT was performed using 2 μg total RNA. Primer pairs for RT-PCR amplification for the canine distemper virus nucleocapsid gene, the S26 reference gene, and the Vero rFas gene were used to analyze expression in Vero cells. RT-PCR results revealed virus activity at 3, 6, 9, and 15 h in the virus-infected Vero cells. The S26 housekeeping gene was amplified in virus infected and control samples. However, expression of the cell death receptor Fas was detected in Vero cells only at 15 h post-infection. We suggest that the Lederle vaccine induces apoptosis by Fas receptor signaling, possibly through caspase-8 signaling rather than through mitochondrial signaling in the infected cells.
Collapse
Affiliation(s)
- H L Del Puerto
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brasil.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Laforge M, Campillo-Gimenez L, Monceaux V, Cumont MC, Hurtrel B, Corbeil J, Zaunders J, Elbim C, Estaquier J. HIV/SIV infection primes monocytes and dendritic cells for apoptosis. PLoS Pathog 2011; 7:e1002087. [PMID: 21731488 PMCID: PMC3121878 DOI: 10.1371/journal.ppat.1002087] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Accepted: 04/13/2011] [Indexed: 12/13/2022] Open
Abstract
Subversion or exacerbation of antigen-presenting cells (APC) death modulates host/pathogen equilibrium. We demonstrated during in vitro differentiation of monocyte-derived macrophages and monocyte-derived dendritic cells (DCs) that HIV sensitizes the cells to undergo apoptosis in response to TRAIL and FasL, respectively. In addition, we found that HIV-1 increased the levels of pro-apoptotic Bax and Bak molecules and decreased the levels of anti-apoptotic Mcl-1 and FLIP proteins. To assess the relevance of these observations in the context of an experimental model of HIV infection, we investigated the death of APC during pathogenic SIV-infection in rhesus macaques (RMs). We demonstrated increased apoptosis, during the acute phase, of both peripheral blood DCs and monocytes (CD14+) from SIV+RMs, associated with a dysregulation in the balance of pro- and anti-apoptotic molecules. Caspase-inhibitor and death receptors antagonists prevented apoptosis of APCs from SIV+RMs. Furthermore, increased levels of FasL in the sera of pathogenic SIV+RMs were detected, compared to non-pathogenic SIV infection of African green monkey. We suggest that inappropriate apoptosis of antigen-presenting cells may contribute to dysregulation of cellular immunity early in the process of HIV/SIV infection. Antigen-presenting cells (APCs) are critical for both innate and adaptive immunity. They have a profound impact on the hosts' ability to combat microbes. Dysfunction and premature death by apoptosis of APCs may contribute to an abnormal immune response unable to clear pathogens. Circulating blood monocytes exhibit developmental plasticity, with the capability of differentiating into either macrophages or dendritic cells (DCs), and they represent important cellular targets for HIV-1. We report that HIV infection renders monocytes/macrophages and DCs in vitro more prone to undergo apoptosis and this heightened susceptibility is associated with changes in the expression of anti- and pro-apoptotic molecules. Our results show that during the acute phase of SIV-infection of rhesus macaques, monocytes and DCs are more prone to die by apoptosis. They express lower levels of Mcl-1 and FLIP proteins, two anti-apoptotic molecules, but higher expression of the active form of Bax and Bak, the gatekeepers of the mitochondria, major sensor of the apoptotic machinery. Because the early events are important in the pathogenesis of this disease, early death of APCs should play a major role leading to the defective immune response. Strategies aimed at preventing death of APCs could be beneficial in helping the immune response to fight HIV-1.
Collapse
Affiliation(s)
| | | | - Valérie Monceaux
- Unité de Physiopathologie des Infections Lentivirales, Institut Pasteur, Paris, France
| | | | - Bruno Hurtrel
- Unité de Physiopathologie des Infections Lentivirales, Institut Pasteur, Paris, France
| | - Jacques Corbeil
- Université Laval, Centre de Recherche en Infectiologie, Québec, Canada
| | - John Zaunders
- St Vincent's Centre for Applied Medical Research, St Vincent's Hospital, Darlinghurst, Australia
| | - Carole Elbim
- INSERM U955, Faculté Créteil Henri Mondor, Créteil, France
- Université Paris Descartes, UMR S 872, Paris, France
| | - Jérôme Estaquier
- INSERM U955, Faculté Créteil Henri Mondor, Créteil, France
- Unité de Physiopathologie des Infections Lentivirales, Institut Pasteur, Paris, France
- Université Laval, Centre de Recherche en Infectiologie, Québec, Canada
- Assistance Publique-Hôpitaux de Paris, Hôpital Henri Mondor, Créteil, France
- * E-mail:
| |
Collapse
|
29
|
Interplay between modified vaccinia virus Ankara and dendritic cells: phenotypic and functional maturation of bystander dendritic cells. J Virol 2011; 85:5532-45. [PMID: 21411535 DOI: 10.1128/jvi.02267-10] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Modified vaccinia virus Ankara (MVA) is an attenuated poxvirus strain, currently under evaluation as a vaccine vector in various clinical settings. It has been reported that human dendritic cells (DCs) mature after infection with MVA, but reports on the functionality of DCs have so far been controversial. In this work, we studied the phenotype and functionality of MVA-infected DCs. As previously reported, we found that human monocyte-derived DCs upregulated CD86 and HLA-DR in response to MVA infection. Moreover, infected DCs produced a broad array of chemokines and cytokines and were able to activate and induce gamma interferon (IFN-γ) production both in CD4(+) and in CD8(+) allogeneic T cells and in specific autologous peripheral blood lymphocytes (PBLs). Analysis of DC maturation following infection with a recombinant green fluorescent protein (GFP)-expressing MVA revealed that upregulation of CD86 expression was mainly observed in GFP(neg) (bystander) cells. While GFP(pos) (infected) DCs produced tumor necrosis factor alpha (TNF-α), they were unable to produce CXCL10 and were less efficient at inducing IFN-γ production in CEF-specific autologous PBLs. Maturation of bystander DCs could be achieved by incubation with supernatant from infected cultures or with apoptotic infected cells. Type I IFNs were partially responsible for the induction of CXCL10 on bystander DCs. Our findings demonstrate for the first time that, in MVA-infected DC cultures, the leading role with respect to functionality and maturation characteristics is achieved by the bystander DCs.
Collapse
|
30
|
Abstract
Measles is an important cause of child mortality that has a seemingly paradoxical interaction with the immune system. In most individuals, the immune response is successful in eventually clearing measles virus (MV) infection and in establishing life-long immunity. However, infection is also associated with persistence of viral RNA and several weeks of immune suppression, including loss of delayed type hypersensitivity responses and increased susceptibility to secondary infections. The initial T-cell response includes CD8+ and T-helper 1 CD4+ T cells important for control of infectious virus. As viral RNA persists, there is a shift to a T-helper 2 CD4+ T-cell response that likely promotes B-cell maturation and durable antibody responses but may suppress macrophage activation and T-helper 1 responses to new infections. Suppression of mitogen-induced lymphocyte proliferation can be induced by lymphocyte infection with MV or by lymphocyte exposure to a complex of the hemagglutinin and fusion surface glycoproteins without infection. Dendritic cells (DCs) are susceptible to infection and can transmit infection to lymphocytes. MV-infected DCs are unable to stimulate a mixed lymphocyte reaction and can induce lymphocyte unresponsiveness through expression of MV glycoproteins. Thus, multiple factors may contribute both to measles-induced immune suppression and to the establishment of durable protective immunity.
Collapse
Affiliation(s)
- Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
31
|
Meera S, Madhuri T, Manisha G, Ramesh P. Irreversible loss of pDCs by apoptosis during early HIV infection may be a critical determinant of immune dysfunction. Viral Immunol 2010; 23:241-9. [PMID: 20565289 DOI: 10.1089/vim.2009.0112] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The dendritic cell subsets myeloid dendritic cells (mDCs) and plasmacytoid dendritic cells (pDCs) play an important role in HIV pathogenesis. While pDCs play a major role in the innate immune response, mDCs are important for induction of the antigen-specific immune response. We studied pDCs and mDCs at different stages of HIV infection, and found that there were decreased percentages of pDCs and mDCs in the advanced stage of the disease (p < 0.0001), and that slow progressors did not show as great a decrease as more healthy individuals. Persons who had acquired infection within the last year showed a normal mDC percentage but a lower pDC percentage (p = 0.0092) than healthy individuals (0.16%). pDC percentages in those with late-stage disease did not revert to normal after successful antiretroviral therapy (ART), whereas mDC percentages reverted to levels comparable to those seen in the healthy population (0.08% pre-ART to 0.18% post-ART; p < 0.0001). The pDC population had high levels of apoptotic markers in those with recent (p = 0.0025) and advanced (p = 0.0012) HIV infection, with no difference in their migratory capacity from controls and slow progressors, indicating that apoptosis is the major mechanism of declining pDC numbers in the circulation. mDCs showed increased levels of apoptotic markers (p = 0.0012), as well as migration (p = 0.03), in those with advanced-stage disease compared to controls, suggesting that both migration and apoptosis contribute to the decline seen in mDCs in the circulation. The irreversible loss of pDCs due to apoptosis seen early in HIV infection may be responsible for an impaired innate anti-HIV immune response. However, the presence of functionally-competent pDCs in slow progressors implies that the loss of pDCs early in infection may be critical to control of HIV infection through innate immune mechanisms, and may influence the progression of disease.
Collapse
Affiliation(s)
- Singh Meera
- National AIDS Research Institute, Pune, Maharashtra, India
| | | | | | | |
Collapse
|
32
|
Torrentes-Carvalho A, Azeredo EL, Reis SRI, Miranda AS, Gandini M, Barbosa LS, Kubelka CF. Dengue-2 infection and the induction of apoptosis in human primary monocytes. Mem Inst Oswaldo Cruz 2010; 104:1091-9. [PMID: 20140369 DOI: 10.1590/s0074-02762009000800005] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2009] [Accepted: 10/19/2009] [Indexed: 11/22/2022] Open
Abstract
Monocytes/macrophages are important targets for dengue virus (DENV) replication; they induce inflammatory mediators and are sources of viral dissemination in the initial phase of the disease. Apoptosis is an active process of cellular destruction genetically regulated, in which a complex enzymatic pathway is activated and may be trigged by many viral infections. Since the mechanisms of apoptotic induction in DENV-infected target cells are not yet defined, we investigated the virus-cell interaction using a model of primary human monocyte infection with DENV-2 with the aim of identifying apoptotic markers. Cultures analyzed by flow cytometry and confocal microscopy yielded DENV antigen positive cells with rates that peaked at the second day post infection (p.i.), decayed afterwards and produced the apoptosis-related cytokines TNF-alpha and IL-10. Phosphatidylserine, an early marker for apoptosis, was increased at the cell surface and the Fas death receptor was upregulated at the second day p.i. at significantly higher rates in DENV infected cell cultures than controls. However, no detectable changes were observed in the expression of the anti-apoptotic protein Bcl-2 in infected cultures. Our data support virus modulation of extrinsic apoptotic factors in the in vitro model of human monocyte DENV-2 infection. DENV may be interfering in activation and death mechanisms by inducing apoptosis in target cells.
Collapse
|
33
|
Abstract
Measles virus (MV) causes transient severe immunosuppression in patients, which may lead to secondary viral and bacterial infections, largely accounting for measles-related morbidity and mortality. MV is known to infect immune cells by using the human signaling lymphocyte activation molecule (SLAM; also called CD150) as a cellular receptor, but the mechanism by which MV causes immunosuppression is not well understood. We show that MV infection of SLAM knock-in mice, in which the V domain of mouse SLAM was replaced by the V domain of human SLAM, crossed with alpha/beta-interferon receptor knockout mice, reproduced many immunological alterations observed in human patients. These included lymphopenia, inhibition of T-cell proliferation and antibody production, increased production of the Th2 cytokine interleukin-4 (IL-4) and the immunosuppressive cytokine IL-10, and suppression of contact hypersensitivity. Gross redistribution of lymphocytes among lymphoid tissues was not apparent in infected mice, nor was an increase of regulatory T cells. The numbers of lymphocytes in lymph nodes remained almost unchanged after MV infection, despite enhanced apoptosis, suggesting that lymph nodes were replenished with lymphocytes from the peripheral blood, which may have contributed to the observed lymphopenia in the spleen. Blocking of IL-10 by use of an anti-IL-10 receptor antibody ameliorated suppression of contact hypersensitivity in infected mice. These results indicate that SLAM knock-in mice lacking the expression of the alpha/beta-interferon receptor serve as a useful small animal model with which to elucidate MV-induced immunosuppression.
Collapse
|
34
|
Hahm B. Hostile communication of measles virus with host innate immunity and dendritic cells. Curr Top Microbiol Immunol 2009; 330:271-87. [PMID: 19203114 DOI: 10.1007/978-3-540-70617-5_13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Following measles virus (MV) infection, host innate immune responses promptly operate to purge the virus. Detection of alerting measles viral components or replication intermediates by pattern-recognizing host machinery of Toll-like receptors and RNA helicases triggers signaling to synthesize array of anti-viral and immunoregulatory molecules, including type I interferon (IFN). Diverse subtypes of dendritic cells (DCs) play pivotal roles in both host innate immunity on the primary MV-infected site and initiating adaptive immune responses on secondary lymphoid tissues. Responding to the predictable host immune responses, MV appears to have devised multiple strategies to evade, suppress, or even utilize host innate immunity and DC responses. This review focuses on versatile actions of MV-induced type I IFNs causing beneficial or deleterious influence on host immunity and the interplay between MV and heterogeneous DCs at distinct locations.
Collapse
Affiliation(s)
- B Hahm
- Department of Surgery, Center for Cellular and Molecular Immunology, University of Missouri-Columbia School of Medicine, One Hospital Dr., Columbia, MO 65212, USA.
| |
Collapse
|
35
|
Lay MDH, Zhang L, Ribeiro RM, Mueller SN, Belz GT, Davenport MP. Kinetics of major histocompatibility class I antigen presentation in acute infection. THE JOURNAL OF IMMUNOLOGY 2009; 182:902-11. [PMID: 19124733 DOI: 10.4049/jimmunol.182.2.902] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Ag presentation within the regional lymph node is crucial for the initiation of CD8+ T cell responses following viral infection. The magnitude and quality of the CD8+ T cell response are regulated by the interplay between the size of the APC population and duration of Ag presentation. To understand how these parameters are finely regulated during an immune response, we have investigated the dynamics of Ag presentation in influenza A virus and HSV-1 infection. In both infections, APC production was calculated to occur over the first few days of infection, after which there was slow exponential decay over a period of up to 2 wk. This production rate is most likely determined by the Ag availability and recruitment and/or maturation rate of dendritic cells. APC production was found to closely parallel lymph node cell recruitment in both infections. This was greatest in the first 6 h of infection for HSV and over the second and third day for influenza. In HSV infection, the peak production also coincides with peak viral levels. By contrast, in influenza infection, APC production ceased between the third and fourth day despite the presence of high levels of virus until 5 days after infection. These analyses demonstrate that two quite different self-limiting infections generate the APC necessary to drive T cell responses early in infection at different rates. Understanding how such contrasting kinetics of Ag presentation impacts on the growth and size of developing protective T cell populations has important implications for the design of vaccines and immunotherapies.
Collapse
Affiliation(s)
- Matthew D H Lay
- Complex Systems in Biology Group, Centre for Vascular Research, University of New South Wales, Kensington, Australia
| | | | | | | | | | | |
Collapse
|
36
|
Pfaller CK, Conzelmann KK. Measles virus V protein is a decoy substrate for IkappaB kinase alpha and prevents Toll-like receptor 7/9-mediated interferon induction. J Virol 2008; 82:12365-73. [PMID: 18922877 PMCID: PMC2593327 DOI: 10.1128/jvi.01321-08] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Accepted: 10/03/2008] [Indexed: 12/25/2022] Open
Abstract
The central role of plasmacytoid dendritic cells (pDC) in activating host immune responses stems from their high capacity to express alpha interferon (IFN-alpha) after stimulation of Toll-like receptors 7 and 9 (TLR7 and -9). This involves the adapter MyD88 and the kinases interleukin-1 receptor-associated kinase 1 (IRAK1), IRAK4, and IkappaB kinase alpha (IKKalpha), which activates IFN regulatory factor 7 (IRF7) and is independent of the canonical kinases TBK1 and IKKepsilon. We have recently shown that the immunosuppressive measles virus (MV) abolishes TLR7/9/MyD88-dependent IFN induction in human pDC (Schlender et al., J. Virol. 79:5507-5515, 2005), but the molecular mechanisms remained elusive. Here, we have reconstituted the pathway in cell lines and identified IKKalpha and IRF7 as specific targets of the MV V protein (MV-V). Binding of MV-V to IKKalpha resulted in phosphorylation of V on the expense of IRF7 phosphorylation by IKKalpha in vitro and in living cells. This corroborates the role of IKKalpha as the kinase phosphorylating IRF7. MV-V in addition bound to IRF7 and to phosphomimetic IRF7 and inhibited IRF7 transcriptional activity. Binding to both IKKalpha and IRF7 required the 68-amino-acid unique C-terminal domain of V. Inhibition of TLR/MyD88-dependent IFN induction by MV-V is unique among paramyxovirus V proteins and should contribute to the unique immunosuppressive phenotype of measles. The mechanisms employed by MV-V inspire strategies to interfere with immunopathological TLR/MyD88 signaling.
Collapse
Affiliation(s)
- Christian K Pfaller
- Max von Pettenkofer-Institute & Gene Center, Feodor-Lynen-Str. 25, D-81377 Munich, Germany
| | | |
Collapse
|
37
|
Dendritic cells are preferentially targeted among hematolymphocytes by Modified Vaccinia Virus Ankara and play a key role in the induction of virus-specific T cell responses in vivo. BMC Immunol 2008; 9:15. [PMID: 18412969 PMCID: PMC2359732 DOI: 10.1186/1471-2172-9-15] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2007] [Accepted: 04/15/2008] [Indexed: 11/30/2022] Open
Abstract
Background Modified Vaccinia Ankara (MVA) is a highly attenuated strain of vaccinia virus (VV) that has lost approximately 15% of the VV genome, along with the ability to replicate in most mammalian cells. It has demonstrated impressive safety and immunogenicity profile in both preclinical and clinical studies, and is being actively explored as a promising vaccine vector for a number of infectious diseases and malignancies. However, little is known about how MVA interacts with the host immune system constituents, especially dendritic cells (DCs), to induce strong immune responses despite its inability to replicate in vivo. Using in vitro and in vivo murine models, we systematically investigated the susceptibility of murine DCs to MVA infection, and the immunological consequences of the infection. Results Our data demonstrate that MVA preferentially infects professional antigen presenting cells, especially DCs, among all the subsets of hematolymphoid cells. In contrast to the reported blockage of DC maturation and function upon VV infection, DCs infected by MVA undergo phenotypic maturation and produce innate cytokine IFN-α within 18 h of infection. Substantial apoptosis of MVA-infected DCs occurs after 12 h following infection and the apoptotic DCs are readily phagocytosed by uninfected DCs. Using MHC class I – deficient mice, we showed that both direct and cross-presentation of viral Ags are likely to be involved in generating viral-specific CD8+ T cell responses. Finally, DC depletion abrogated the T cell activation in vivo. Conclusion We present the first in vivo evidence that among hematolymphoid cells, DCs are the most susceptible targets for MVA infection, and DC-mediated Ag presentation is required for the induction of MVA-specific immune responses. These results provide important information concerning the mechanisms by which strong immune responses are elicited to MVA-encoded antigens and may inform efforts to further improve the immunogenicity of this already promising vaccine vector.
Collapse
|
38
|
Chapter 4 Receptor Interactions, Tropism, and Mechanisms Involved in Morbillivirus‐Induced Immunomodulation. Adv Virus Res 2008; 71:173-205. [DOI: 10.1016/s0065-3527(08)00004-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
39
|
Liu C, Sarkaria JN, Petell CA, Paraskevakou G, Zollman PJ, Schroeder M, Carlson B, Decker PA, Wu W, James CD, Russell SJ, Galanis E. Combination of Measles Virus Virotherapy and Radiation Therapy Has Synergistic Activity in the Treatment of Glioblastoma Multiforme. Clin Cancer Res 2007; 13:7155-65. [DOI: 10.1158/1078-0432.ccr-07-1306] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
Abstract
Dendritic cells (DCs) act not only as sentinels for detection of, but also as target cells for viruses, and this can be important for viral transport and spread. All subsets of DCs are equipped with a battery of receptors recognizing virus‐associated molecular signatures, and recognition of those launches a maturation programme that results in substantial alterations of morphology, motility and the DCs' interactive properties with the extracellular matrix and scanning T cells. In addition to being sensed, viruses are internalized into DCs and, for the major proportion, processed into peptides that are subsequently presented by major histocompatibility complex (MHC) molecules. Transmission of virus to T cells can occur after completion of their replication cycle if the intracellular milieu of the DC permits that. Alternatively, viruses can remain protected from degradation following entrapment by pattern recognition receptors in intracellular compartments, also referred to as virosomes, which translocate towards the DC/T cell interface. Most likely, transfer of virus to T cells occurs in these junctions, referred to as infectious synapses. In addition to promoting DC maturation, many viruses are able to downmodulate DC development and functions in order to evade immune recognition or to induce a generalized immunosuppression.
Collapse
Affiliation(s)
- Christine Pohl
- University of Wuerzburg, Institute for Virology and Immunobiology, Versbacher Str. 7, D-97078 Wuerzburg, Germany
| | | | | |
Collapse
|
41
|
Abstract
Measles remains a leading vaccine-preventable cause of child mortality worldwide, particularly in sub-Saharan Africa where almost half of the estimated 454,000 measles deaths in 2004 occurred. However, great progress in measles control has been made in resource-poor countries through accelerated measles-control efforts. The global elimination of measles has been debated since measles vaccines were first licensed in the 1960's, and this debate is likely to be renewed if polio virus is eradicated. This review discusses the pathogenesis of measles and the likelihood of the worldwide elimination of this disease.
Collapse
Affiliation(s)
- William J. Moss
- Department of Epidemiology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, 21205 Maryland USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, 21205 Maryland USA
| | - Diane E. Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, 21205 Maryland USA
| |
Collapse
|
42
|
Nashida Y, Kumamoto T, Azuma E, Hirayama M, Araki M, Yamada H, Dida F, Iwamoto S, Tamaki S, Ido M, Ihara T, Komada Y. Development of a dendritic cell vaccine against measles for patients following hematopoietic cell transplantation. Transplantation 2006; 82:1104-7. [PMID: 17060861 DOI: 10.1097/01.tp.0000232695.91370.96] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Most patients who have undergone hematopoietic cell transplantation (HCT) lose specific immunity to measles. However, due to its immunosuppressive potential, it has been recommended that a live attenuated measles vaccination be administered two years following HCT. Measles virus (MV) glycoproteins including hemagglutinin (HA) are expressed on MV-infected dendritic cells (DCs), and they impair efficient antigen presentation between the DC and T cell. We produced a DC-based vaccine against MV by loading DCs with MV-infected autologous DCs. MV in the infected DCs was inactivated using ultraviolet-B. The DC-based vaccine neither expressed HA nor inhibited allogeneic T cell proliferation, while it induced the production of interferon-gamma (IFN-gamma) by autologous CD4 and CD8 naive T cells ex vivo. Importantly, the vaccine derived from patients who had undergone HCT also efficiently induced IFN-gamma producing cells. These findings indicate that our DC-based MV vaccine induces MV-specific immunity even in post-HCT patients without causing immunosuppression.
Collapse
Affiliation(s)
- Yuji Nashida
- Department of Pediatrics and Cell Transplantation, Mie University School of Medicine, Mie, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Measles virus-dendritic cell interaction via SLAM inhibits innate immunity: selective signaling through TLR4 but not other TLRs mediates suppression of IL-12 synthesis. Virology 2006; 358:251-7. [PMID: 17070884 DOI: 10.1016/j.virol.2006.10.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2006] [Revised: 08/23/2006] [Accepted: 10/04/2006] [Indexed: 10/24/2022]
Abstract
Two hallmarks of measles virus (MV) infection are the ability of the virus to cause immunosuppression and the resultant enhanced susceptibility of the infected host to microbial insults. We investigated the effect of MV infection on the ability of dendritic cells (DCs) to induce IL-12 via toll-like receptor (TLR) signaling. When infected with MV, transgenic mice which expressed human SLAM receptor on their DCs were defective in the selective synthesis of IL-12 in DCs in response to stimulation of TLR4 signaling, but not to engagements of TLR2, 3, 7 or 9. MV suppressed TLR4-mediated IL-12 induction in DCs even in the presence of co-stimulation with another ligand for TLR2, 3, 7, or 9. While MV V and C proteins were not responsible for IL-12 inhibition, interaction of MV hemagglutinin with human SLAM facilitated the suppression. These results suggest that MV, by altering DC function, renders them unresponsive to secondary pathogens via TLR4.
Collapse
|
44
|
Kajita M, Katayama H, Murata T, Kai C, Hori M, Ozaki H. Canine distemper virus induces apoptosis through caspase-3 and -8 activation in vero cells. ACTA ACUST UNITED AC 2006; 53:273-7. [PMID: 16907958 DOI: 10.1111/j.1439-0450.2006.00963.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We investigated the signal-transduction pathway of canine distemper virus-Onderstepoort (CDV-Ond) vaccine strain-mediated apoptosis in Vero cells. Canine distemper virus-Onderstepoort at a multiplicity of infection (MOI) of 0.1 induced DNA fragmentation 48 h after infection. Immunofluorescence staining revealed that 57% +/- 4% of the CDV-N-protein-positive cells were terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive, and all TUNEL-positive cells were CDV-N-protein-positive, indicating that CDV-Ond induced apoptosis only in the infected cells. We also found that CDV-Ond infection induced activation of caspase-3 and caspase-8. In the semi-quantitative reverse transcription-polymerase chain reaction assay for apoptosis-related genes, the expression of mRNA of the death receptor, Fas, was also increased in CDV-Ond-infected cells. In contrast, the expressions of Bcl-2 and Bax, regulators for intrinsic apoptotic signaling through the mitochondria, did not change. These results suggest that CDV-Ond induced apoptosis by activating caspase-3, possibly through caspase-8 signaling rather than through p53/Bax-mediated, mitochondrial signaling in the infected cells.
Collapse
Affiliation(s)
- M Kajita
- Department of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Müller N, Avota E, Schneider-Schaulies J, Harms H, Krohne G, Schneider-Schaulies S. Measles virus contact with T cells impedes cytoskeletal remodeling associated with spreading, polarization, and CD3 clustering. Traffic 2006; 7:849-58. [PMID: 16787397 DOI: 10.1111/j.1600-0854.2006.00426.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
CD3/CD28-induced activation of the PI3/Akt kinase pathway and proliferation is impaired in T cells after contact with the measles virus (MV) glycoprotein (gp) complex. We now show that this signal also impairs actin cytoskeletal remodeling in T cells, which loose their ability to adhere and to promote microvilli formation. MV exposure results in an almost complete collapse of membrane protrusions associated with reduced phosphorylation levels of cofilin and ezrin/radixin/moesin (ERM) proteins. Consistent with their inability to activate Cdc42 and Rac1 in response to the ligation of CD3/CD28, T cells exposed to MV fail to acquire a morphology consistent with spreading and lamellopodia formation. In spite of these impairments of cytoskeleton-driven morphological alterations, these cells are recruited into conjugates with dendritic cells as efficiently as control T cells. The signal elicited by MV, however, prevents T cells to polarize as documented by a failure to redistribute the microtubule organizing center toward the synapse. Moreover, CD3 cannot be efficiently clustered and redistributed to the central region of the immunological synapse. Thus, by inducing microvillar collapse and interfering with cytoskeletal remodeling, MV signaling disturbs the ability of T cells to adhere, spread, and cluster receptors essential for sustained T-cell activation.
Collapse
Affiliation(s)
- Nora Müller
- Institute for Virology and Immunobiology, University of Würzburg, Versbacher Str. 7, D-97078 Würzburg, Germany
| | | | | | | | | | | |
Collapse
|
46
|
Schneider-Schaulies S, Dittmer U. Silencing T cells or T-cell silencing: concepts in virus-induced immunosuppression. J Gen Virol 2006; 87:1423-1438. [PMID: 16690907 DOI: 10.1099/vir.0.81713-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The ability to evade or suppress the host's immune response is a property of many viruses, indicating that this provides an advantage for the pathogen to spread efficiently or even to establish a persistent infection. The type and complexity of its genome and cell tropism but also its preferred type of host interaction are important parameters which define the strategy of a given virus to modulate the immune system in an optimal manner. Because they take a central position in any antiviral defence, the activation and function of T cells are the predominant target of many viral immunosuppressive regimens. In this review, two different strategies whereby this could be achieved are summarized. Retroviruses can infect professional antigen-presenting cells and impair their maturation and functional properties. This coincides with differentiation and expansion of silencing T cells referred to as regulatory T cells with suppressive activity, mainly to CD8+ effector T cells. The second concept, outlined for measles virus, is a direct, contact-mediated silencing of T cells which acquire a transient paralytic state.
Collapse
Affiliation(s)
| | - Ulf Dittmer
- Institut für Virologie des Universitätsklinikums Essen, D-45122 Essen, Germany
| |
Collapse
|
47
|
de Witte L, Abt M, Schneider-Schaulies S, van Kooyk Y, Geijtenbeek TBH. Measles virus targets DC-SIGN to enhance dendritic cell infection. J Virol 2006; 80:3477-86. [PMID: 16537615 PMCID: PMC1440360 DOI: 10.1128/jvi.80.7.3477-3486.2006] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Dendritic cells (DCs) are involved in the pathogenesis of measles virus (MV) infection by inducing immune suppression and possibly spreading the virus from the respiratory tract to lymphatic tissues. It is becoming evident that DC function can be modulated by the involvement of different receptors in pathogen interaction. Therefore, we have investigated the relative contributions of different MV-specific receptors on DCs to MV uptake into and infection of these cells. DCs express the MV receptors CD46 and CD150, and we demonstrate that the C-type lectin DC-specific intercellular adhesion molecule 3-grabbing nonintegrin (DC-SIGN) is a novel receptor for laboratory-adapted and wild-type MV strains. The ligands for DC-SIGN are both MV glycoproteins F and H. In contrast to CD46 and CD150, DC-SIGN does not support MV entry, since DC-SIGN does not confer susceptibility when stably expressed in CHO cells. However, DC-SIGN is important for the infection of immature DCs with MV, since both attachment and infection of immature DCs with MV are blocked in the presence of DC-SIGN inhibitors. Our data demonstrate that DC-SIGN is crucial as an attachment receptor to enhance CD46/CD150-mediated infection of DCs in cis. Moreover, MV might not only target DC-SIGN to infect DCs but may also use DC-SIGN for viral transmission and immune suppression.
Collapse
Affiliation(s)
- Lot de Witte
- Department of Molecular Cell Biology and Immunology, V U University Medical Center, van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
48
|
Kerdiles YM, Sellin CI, Druelle J, Horvat B. Immunosuppression caused by measles virus: role of viral proteins. Rev Med Virol 2006; 16:49-63. [PMID: 16237742 DOI: 10.1002/rmv.486] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Measles virus (MV) causes transient but profound immunosuppression resulting in increased susceptibility to secondary bacterial and viral infections. Due to the development of these opportunistic infections, measles remains the leading vaccine-preventable cause of child death worldwide. Different immune abnormalities have been associated with measles, including disappearance of delayed-type hypersensitivity reactions, impaired lymphocyte and antigen-presenting cell functions, down-regulation of pro-inflammatory interleukin 12 production and altered interferon alpha/beta signalling pathways. Several MV proteins have been suggested to hinder immune functions: hemagglutinin, fusion protein, nucleoprotein and the non-structural V and C proteins. This review will focus on the novel functions attributed to MV proteins in the immunosuppression associated with measles. Here, we highlight new advances in the field, emphasising the interaction between MV proteins and their cellular targets, in particular the cell membrane receptors, CD46, CD150, TLR2 and FcgammaRII in the induction of immunological abnormalities associated with measles.
Collapse
Affiliation(s)
- Yann M Kerdiles
- INSERM U404, IFR 128, Biosciences Lyon-Gerland, 21 Ave. Tony Garnier, 69365 Lyon, France
| | | | | | | |
Collapse
|
49
|
Zilliox MJ, Parmigiani G, Griffin DE. Gene expression patterns in dendritic cells infected with measles virus compared with other pathogens. Proc Natl Acad Sci U S A 2006; 103:3363-8. [PMID: 16492729 PMCID: PMC1413941 DOI: 10.1073/pnas.0511345103] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Gene expression patterns supply insight into complex biological networks that provide the organization in which viruses and host cells interact. Measles virus (MV) is an important human pathogen that induces transient immunosuppression followed by life-long immunity in infected individuals. Dendritic cells (DCs) are potent antigen-presenting cells that initiate the immune response to pathogens and are postulated to play a role in MV-induced immunosuppression. To better understand the interaction of MV with DCs, we examined the gene expression changes that occur over the first 24 h after infection and compared these changes to those induced by other viral, bacterial, and fungal pathogens. There were 1,553 significantly regulated genes with nearly 60% of them down-regulated. MV-infected DCs up-regulated a core of genes associated with maturation of antigen-presenting function and migration to lymph nodes but also included genes for IFN-regulatory factors 1 and 7, 2'5' oligoadenylate synthetase, Mx, and TNF superfamily proteins 2, 7, 9, and 10 (TNF-related apoptosis-inducing ligand). MV induced genes for IFNs, ILs, chemokines, antiviral proteins, histones, and metallothioneins, many of which were also induced by influenza virus, whereas genes for protein synthesis and oxidative phosphorylation were down-regulated. Unique to MV were the induction of genes for a broad array of IFN-alphas and the failure to up-regulate dsRNA-dependent protein kinase. These results provide a modular view of common and unique DC responses after infection and suggest mechanisms by which MV may modulate the immune response.
Collapse
Affiliation(s)
- Michael J. Zilliox
- *The W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, and
| | - Giovanni Parmigiani
- Departments of Oncology, Biostatistics, and Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Diane E. Griffin
- *The W. Harry Feinstone Department of Molecular Microbiology and Immunology, The Johns Hopkins Bloomberg School of Public Health, and
| |
Collapse
|
50
|
Réthi B, Gogolák P, Szatmari I, Veres A, Erdôs E, Nagy L, Rajnavölgyi E, Terhorst C, Lányi A. SLAM/SLAM interactions inhibit CD40-induced production of inflammatory cytokines in monocyte-derived dendritic cells. Blood 2005; 107:2821-9. [PMID: 16317102 PMCID: PMC1895370 DOI: 10.1182/blood-2005-06-2265] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Signaling lymphocyte activation molecule (SLAM, CD150, or SLAMF1) is a self-ligand receptor on the surface of activated T- and B-lymphocytes, macrophages, and dendritic cells (DCs). Here we examine the effect of SLAM/SLAM interactions on CD40L-induced CD40 signaling pathways in human DCs. CD40L-expressing L929 cells induced DCs to produce interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-alpha), and IL-12, which was strongly inhibited by coexpression of SLAM on the surface of the L929 cells. Similarly, transfection of DCs with SLAM strongly reduced CD40L-induced IL-12 production. Furthermore, the negative effect of SLAM/SLAM interactions on CD40L-induced DC activation was also detected in the presence of lipopolysaccharide (LPS). LPS-induced IL-12 secretion, however, was not inhibited by SLAM engagement. CD40L-activated DCs affected by exposure to SLAM/SLAM engagement were impaired in their ability to induce differentiation of naive T lymphocytes into interferon-gamma (IFN-gamma)-producing T-helper 1 (Th1) effector cells. These inhibitory effects were not the result of a general unresponsiveness of DCs to CD40L, as SLAM/SLAM interactions did not prevent CD40L-induced up-regulation of CD83, CD86, or human leukocyte antigen (HLA)-DQ on the surface of DCs. Taken together, the results indicate that SLAM/SLAM interactions inhibit CD40-induced signal transduction in monocyte-derived dendritic cells, an effect that was not detectable in earlier studies using anti-SLAM monoclonal antibodies.
Collapse
Affiliation(s)
- Bence Réthi
- Institute of Immunology, University of Debrecen Medical and Health Science Center, 98 Nagyerdei krt, H-4012 Debrecen, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|