1
|
Lo TC, Cheng JY, Lee CW, Hung JT, Lin CC, Yeh HF, Yang BC, Huang Y, Wu HM, Yu AL, Yu J. Priming of macrophage by glycosphingolipids from extracellular vesicles facilitates immune tolerance for embryo-maternal crosstalk. Dev Cell 2023; 58:2447-2459.e5. [PMID: 37989081 DOI: 10.1016/j.devcel.2023.09.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 06/26/2023] [Accepted: 09/29/2023] [Indexed: 11/23/2023]
Abstract
Glycosphingolipids (GSLs) display diverse functions during embryonic development. Here, we examined the GSL profiles of extracellular vesicles (EVs) secreted from human embryonic stem cells (hESCs) and investigated their functions in priming macrophages to enhance immune tolerance of embryo implantation. When peripheral blood mononuclear cells were incubated with ESC-secreted EVs, globo-series GSLs (GHCer, SSEA3Cer, and SSEA4Cer) were transferred via EVs into monocytes/macrophages. Incubation of monocytes during their differentiation into macrophages with either EVs or synthetic globo-series GSLs induced macrophages to exhibit phenotypic features that imitate immune receptivity, i.e., macrophage polarization, augmented phagocytic activity, suppression of T cell proliferation, and the increased trophoblast invasion. It was also demonstrated that decidual macrophages in first-trimester tissues expressed globo-series GSLs. These findings highlight the role of globo-series GSLs via transfer from EVs in priming macrophages to display decidual macrophage phenotypes, which may facilitate healthy pregnancy.
Collapse
Affiliation(s)
- Tzu-Chi Lo
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Jing-Yan Cheng
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chien-Wei Lee
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Jung-Tung Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chun-Cheng Lin
- Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsiao-Fong Yeh
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Bei-Chia Yang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yenlin Huang
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Anatomic Pathology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsien-Ming Wu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Department of Obstetrics and Gynecology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Alice L Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Pediatrics, University of California in San Diego, San Diego, California, USA
| | - John Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, School of Medicine, Chang Gung University, Taoyuan, Taiwan; Institute of Cellular and Organismic Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
2
|
Coler B, Cervantes O, Li M, Coler C, Li A, Shivakumar M, Every E, Schwartz D, Adams Waldorf KM. Common pathways targeted by viral hemorrhagic fever viruses to infect the placenta and increase the risk of stillbirth. Placenta 2023; 141:2-9. [PMID: 36939178 PMCID: PMC10102255 DOI: 10.1016/j.placenta.2022.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 09/19/2022] [Accepted: 10/02/2022] [Indexed: 01/06/2023]
Abstract
Viral hemorrhagic fevers (VHF) are endemic to Africa, South America and Asia and contribute to significant maternal and fetal morbidity and mortality. Viruses causing VHFs are typically zoonotic, spreading to humans through livestock, wildlife, or mosquito vectors. Some of the most lethal VHF viruses also impart a high-risk of stillbirth including ebolaviruses, Marburg virus (MARV), Lassa virus (LASV), and Rift Valley Fever Virus (RVFV). Large outbreaks and epidemics are common, though the impact on the mother, fetus and placenta is understudied from a public health, clinical and basic science perspective. Notably, these viruses utilize ubiquitous cellular surface entry receptors critical for normal placental function to enable viral invasion into multiple key cell types of the placenta and set the stage for maternal-fetal transmission and stillbirth. We employ insights from molecular virology and viral immunology to discuss how trophoblast expression of viral entry receptors for VHF viruses may increase the risk for viral transmission to the fetus and stillbirth. As the frequency of VHF outbreaks is expected to increase with worsening climate change, understanding the pathogenesis of VHF-related diseases in the placenta is paramount to predicting the impact of emerging viruses on the placenta and perinatal outcomes.
Collapse
Affiliation(s)
- Brahm Coler
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, USA
| | - Orlando Cervantes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA
| | - Miranda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Department of Biological Sciences, Columbia University, New York City, NY, USA
| | | | - Amanda Li
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Case Western Reserve, Cleveland, OH, USA
| | - Megana Shivakumar
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA
| | - Emma Every
- School of Medicine, University of Washington, Seattle, WA, USA
| | | | - Kristina M Adams Waldorf
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA.
| |
Collapse
|
3
|
Choi H, Yang SW, Joo JS, Park M, Jin Y, Kim JW, Lee SY, Lee SV, Yun TJ, Cho ML, Hwang HS, Kang YS. Sialylated IVIg binding to DC-SIGN + Hofbauer cells induces immune tolerance through the caveolin-1/NF-kB pathway and IL-10 secretion. Clin Immunol 2023; 246:109215. [PMID: 36581222 DOI: 10.1016/j.clim.2022.109215] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 11/15/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022]
Abstract
Although the use of IVIg has increased in various immune-driven diseases and even in pregnancy, the exact action mechanisms of IVIg are not fully understood. Dendritic cell-specific intercellular adhesion molecule-3 grabbing non-integrin (DC-SIGN) is a known receptor for α-2,6-sialylated IgG (sIVIg), which is responsible for the anti-inflammatory effect of IVIg. DC-SIGN is expressed on Hofbauer cells (HBCs) of the fetal villi of the placenta which act as an innate immune modulator at the maternal-fetal interface. Preeclampsia is a major complication in pregnancy and is related to IL-10, a cytokine with an important role in immune tolerance. DC-SIGN interaction with sIVIg in HBCs promoted IL-10 secretion through the activation of the caveolin-1/NF-κB pathway, especially in plasma lipid rafts. Consistent results were obtained for HBCs from patients with preeclampsia. Collectively, the stimulation of DC-SIGN+ HBCs with sIVIg enhanced immune tolerance in the feto-maternal environment, suggesting the therapeutic application of sIVIg to prevent preeclampsia.
Collapse
Affiliation(s)
- Hyeongjwa Choi
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Seung-Woo Yang
- Department of Obstetrics and Gynecology, Sang-Gye Paik Hospital, Inje University School of Medicine; Seoul 01757, Republic of Korea
| | - Jin-Soo Joo
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University; 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Min Park
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Yihua Jin
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Ji-Woon Kim
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea
| | - Seon-Yeong Lee
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Vin Lee
- Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University; 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Tae-Jin Yun
- Department of Pathology, New York University Grossman School of Medicine; New York, NY 10016, USA
| | - Mi-La Cho
- The Rheumatism Research Center, The Catholic University of Korea, Seoul, South Korea; Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, South Korea
| | - Han-Sung Hwang
- Division of Maternal and Fetal Medicine, Department of Obstetrics and Gynecology, Research Institute of Medical Science, Konkuk University School of Medicine; Seoul, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| | - Young-Sun Kang
- Department of KONKUK-KIST Biomedical Science & Technology, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea; Department of Veterinary Pharmacology and Toxicology, Veterinary Science Research Institute, College of Veterinary Medicine, Konkuk University; 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; KU Research Center for Zoonosis, Konkuk University; 1 Hwayang-dong, Gwangjin-gu, Seoul 143-701, Republic of Korea.
| |
Collapse
|
4
|
Arnold JN, Mitchell DA. Tinker, tailor, soldier, cell: the role of C-type lectins in the defense and promotion of disease. Protein Cell 2022; 14:4-16. [PMID: 36726757 PMCID: PMC9871964 DOI: 10.1093/procel/pwac012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/25/2022] [Indexed: 02/04/2023] Open
Abstract
C-type lectins (CTLs) represent a large family of soluble and membrane-bound proteins which bind calcium dependently via carbohydrate recognition domains (CRDs) to glycan residues presented on the surface of a variety of pathogens. The deconvolution of a cell's glycan code by CTLs underpins several important physiological processes in mammals such as pathogen neutralization and opsonization, leukocyte trafficking, and the inflammatory response. However, as our knowledge of CTLs has developed it has become apparent that the role of this innate immune family of proteins can be double-edged, where some pathogens have developed approaches to subvert and exploit CTL interactions to promote infection and sustain the pathological state. Equally, CTL interactions with host glycoproteins can contribute to inflammatory diseases such as arthritis and cancer whereby, in certain contexts, they exacerbate inflammation and drive malignant progression. This review discusses the 'dual agent' roles of some of the major mammalian CTLs in both resolving and promoting infection, inflammation and inflammatory disease and highlights opportunities and emerging approaches for their therapeutic modulation.
Collapse
|
5
|
Xiaocui L, Wei H, Yunlang C, Zhenzhen Z, Min A. CSF-1-induced DC-SIGN + macrophages are present in the ovarian endometriosis. Reprod Biol Endocrinol 2022; 20:48. [PMID: 35260161 PMCID: PMC8903642 DOI: 10.1186/s12958-022-00901-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 01/25/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Researchers have found that macrophages are the predominant cells in the peritoneal fluid (PF) of endometriosis patients. CSF-1 has been found to accumulate in the lesions and PF of endometriosis patients, and CSF-1 induces THP-1-derived macrophages to polarize toward a CD169+ DC-SIGN+ phenotype. Does the cytokine CSF-1 induce monocytes to differentiate into macrophages with a DC-SIGN+ phenotype in endometriosis? METHODS The level of CSF-1 in the endometrium of control subjects, and the eutopic, and ectopic endometrium of endometriosis patients was evaluated by real-time polymerase chain reaction (qRT-PCR) and was determined by enzyme-linked immunosorbent assay (ELISA) in the PF of control and endometriosis patients. CSF-1 expression was examined with a MILLIPLEX MAP Mouse Cytokine/Chemokine Magnetic Bead Panel. DC-SIGN+ macrophages were detected by immunohistochemical staining of tissues and flow cytometric analysis of the PF of control subjects (N = 25) and endometriosis (N = 35) patients. The phenotypes and biological activities of CSF-1 -induced macrophages were compared in an in vitro coculture system with peripheral blood lymphocytes from control subjects. RESULTS In this study, we found that the proportion of DC-SIGN+ CD169+ macrophages was higher in the abdominal immune microenvironment of endometriosis patients. CSF-1 was primarily secreted from ectopic lesions and peritoneum in mice with endometriosis. In addition, CSF-1 induced the polarization of macrophages toward a DC-SIGN+ CD169+ phenotype; this effect was abolished by the addition of an anti-CSF-1R antibody. CSF-1 induced the generation of DC-SIGN+ macrophages, leading to a depressed status of peripheral blood lymphocytes, including a high percentage of Treg cells and a low percentage of CD8+ T cells. Similarly, blockade with the anti-CSF-1R antibody abrogated this biological effect. CONCLUSIONS This is the first study on the role of DC-SIGN+ macrophages in the immune microenvironment of endometriosis. Further study of the mechanism and biological activities of CSF-1-induced DC-SIGN+ macrophages will enhance our understanding of the physiology of endometriosis.
Collapse
Affiliation(s)
- Li Xiaocui
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, P.R. China
| | - Hong Wei
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, P.R. China
| | - Cai Yunlang
- Department of Obstetrics and Gynecology, Medical School, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Zheng Zhenzhen
- Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, P.R. China
| | - An Min
- Department of Obstetrics and Gynecology, Medical School, Zhongda Hospital, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
6
|
Masomian M, Lalani S, Poh CL. Molecular Docking of SP40 Peptide towards Cellular Receptors for Enterovirus 71 (EV-A71). Molecules 2021; 26:molecules26216576. [PMID: 34770987 PMCID: PMC8587434 DOI: 10.3390/molecules26216576] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/13/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022] Open
Abstract
Enterovirus 71 (EV-A71) is one of the predominant etiological agents of hand, foot and mouth disease (HMFD), which can cause severe central nervous system infections in young children. There is no clinically approved vaccine or antiviral agent against HFMD. The SP40 peptide, derived from the VP1 capsid of EV-A71, was reported to be a promising antiviral peptide that targeted the host receptor(s) involved in viral attachment or entry. So far, the mechanism of action of SP40 peptide is unknown. In this study, interactions between ten reported cell receptors of EV-A71 and the antiviral SP40 peptide were evaluated through molecular docking simulations, followed by in vitro receptor blocking with specific antibodies. The preferable binding region of each receptor to SP40 was predicted by global docking using HPEPDOCK and the cell receptor-SP40 peptide complexes were refined using FlexPepDock. Local molecular docking using GOLD (Genetic Optimization for Ligand Docking) showed that the SP40 peptide had the highest binding score to nucleolin followed by annexin A2, SCARB2 and human tryptophanyl-tRNA synthetase. The average GoldScore for 5 top-scoring models of human cyclophilin, fibronectin, human galectin, DC-SIGN and vimentin were almost similar. Analysis of the nucleolin-SP40 peptide complex showed that SP40 peptide binds to the RNA binding domains (RBDs) of nucleolin. Furthermore, receptor blocking by specific monoclonal antibody was performed for seven cell receptors of EV-A71 and the results showed that the blocking of nucleolin by anti-nucleolin alone conferred a 93% reduction in viral infectivity. Maximum viral inhibition (99.5%) occurred when SCARB2 was concurrently blocked with anti-SCARB2 and the SP40 peptide. This is the first report to reveal the mechanism of action of SP40 peptide in silico through molecular docking analysis. This study provides information on the possible binding site of SP40 peptide to EV-A71 cellular receptors. Such information could be useful to further validate the interaction of the SP40 peptide with nucleolin by site-directed mutagenesis of the nucleolin binding site.
Collapse
Affiliation(s)
- Malihe Masomian
- Correspondence: (M.M.); (C.L.P.); Tel.: +603-74918622 (ext. 7603) (M.M.); +603-74918622 (ext. 7338) (C.L.P.)
| | | | - Chit Laa Poh
- Correspondence: (M.M.); (C.L.P.); Tel.: +603-74918622 (ext. 7603) (M.M.); +603-74918622 (ext. 7338) (C.L.P.)
| |
Collapse
|
7
|
Hasan SS, Dey D, Singh S, Martin M. The Structural Biology of Eastern Equine Encephalitis Virus, an Emerging Viral Threat. Pathogens 2021; 10:pathogens10080973. [PMID: 34451437 PMCID: PMC8400090 DOI: 10.3390/pathogens10080973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/21/2021] [Accepted: 07/28/2021] [Indexed: 11/16/2022] Open
Abstract
Alphaviruses are arboviruses that cause arthritis and encephalitis in humans. Eastern Equine Encephalitis Virus (EEEV) is a mosquito-transmitted alphavirus that is implicated in severe encephalitis in humans with high mortality. However, limited insights are available into the fundamental biology of EEEV and residue-level details of its interactions with host proteins. In recent years, outbreaks of EEEV have been reported mainly in the United States, raising concerns about public safety. This review article summarizes recent advances in the structural biology of EEEV based mainly on single-particle cryogenic electron microscopy (cryoEM) structures. Together with functional analyses of EEEV and related alphaviruses, these structural investigations provide clues to how EEEV interacts with host proteins, which may open avenues for the development of therapeutics.
Collapse
Affiliation(s)
- S. Saif Hasan
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
- Center for Biomolecular Therapeutics, University of Maryland School of Medicine, 9600 Gudelsky Drive, Rockville, MD 20850, USA
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, University of Maryland Medical Center, 22. S. Greene St., Baltimore, MD 21201, USA
- Correspondence:
| | - Debajit Dey
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| | - Suruchi Singh
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| | - Matthew Martin
- Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 108 N. Greene Street, Baltimore, MD 21201, USA; (D.D.); (S.S.); (M.M.)
| |
Collapse
|
8
|
Amraei R, Yin W, Napoleon MA, Suder EL, Berrigan J, Zhao Q, Olejnik J, Chandler KB, Xia C, Feldman J, Hauser BM, Caradonna TM, Schmidt AG, Gummuluru S, Mühlberger E, Chitalia V, Costello CE, Rahimi N. CD209L/L-SIGN and CD209/DC-SIGN Act as Receptors for SARS-CoV-2. ACS CENTRAL SCIENCE 2021; 7:1156-1165. [PMID: 34341769 PMCID: PMC8265543 DOI: 10.1021/acscentsci.0c01537] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Indexed: 05/17/2023]
Abstract
As the COVID-19 pandemic continues to spread, investigating the processes underlying the interactions between SARS-CoV-2 and its hosts is of high importance. Here, we report the identification of CD209L/L-SIGN and the related protein CD209/DC-SIGN as receptors capable of mediating SARS-CoV-2 entry into human cells. Immunofluorescence staining of human tissues revealed prominent expression of CD209L in the lung and kidney epithelia and endothelia. Multiple biochemical assays using a purified recombinant SARS-CoV-2 spike receptor-binding domain (S-RBD) or S1 encompassing both N termal domain and RBD and ectopically expressed CD209L and CD209 revealed that CD209L and CD209 interact with S-RBD. CD209L contains two N-glycosylation sequons, at sites N92 and N361, but we determined that only site N92 is occupied. Removal of the N-glycosylation at this site enhances the binding of S-RBD with CD209L. CD209L also interacts with ACE2, suggesting a role for heterodimerization of CD209L and ACE2 in SARS-CoV-2 entry and infection in cell types where both are present. Furthermore, we demonstrate that human endothelial cells are permissive to SARS-CoV-2 infection, and interference with CD209L activity by a knockdown strategy or with soluble CD209L inhibits virus entry. Our observations demonstrate that CD209L and CD209 serve as alternative receptors for SARS-CoV-2 in disease-relevant cell types, including the vascular system. This property is particularly important in tissues where ACE2 has low expression or is absent and may have implications for antiviral drug development.
Collapse
Affiliation(s)
- Razie Amraei
- Department
of Pathology, School of Medicine, Boston
University Medical Campus, Boston, Massachusetts 02118, United States
| | - Wenqing Yin
- Renal
Section, Department of Medicine, Boston
University Medical Center, Boston, Massachusetts 02118, United States
| | - Marc A. Napoleon
- Renal
Section, Department of Medicine, Boston
University Medical Center, Boston, Massachusetts 02118, United States
| | - Ellen L. Suder
- Department
of Microbiology, Boston University School
of Medicine, Boston, Massachusetts 02118, United States
- National
Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, Massachusetts 02118, United States
| | - Jacob Berrigan
- Department
of Microbiology, Boston University School
of Medicine, Boston, Massachusetts 02118, United States
| | - Qing Zhao
- Department
of Pathology, School of Medicine, Boston
University Medical Campus, Boston, Massachusetts 02118, United States
| | - Judith Olejnik
- Department
of Microbiology, Boston University School
of Medicine, Boston, Massachusetts 02118, United States
- National
Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, Massachusetts 02118, United States
| | - Kevin Brown Chandler
- Center
for Biomedical Mass Spectrometry, Boston
University School of Medicine, Boston, Massachusetts 02118, United States
| | - Chaoshuang Xia
- Center
for Biomedical Mass Spectrometry, Boston
University School of Medicine, Boston, Massachusetts 02118, United States
| | - Jared Feldman
- Ragon Institute
of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, United States
| | - Blake M. Hauser
- Ragon Institute
of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, United States
| | - Timothy M. Caradonna
- Ragon Institute
of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, United States
| | - Aaron G. Schmidt
- Ragon Institute
of MGH, MIT, and Harvard, Cambridge, Massachusetts 02139, United States
- Department
of Microbiology, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Suryaram Gummuluru
- Department
of Microbiology, Boston University School
of Medicine, Boston, Massachusetts 02118, United States
| | - Elke Mühlberger
- Department
of Microbiology, Boston University School
of Medicine, Boston, Massachusetts 02118, United States
- National
Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, Massachusetts 02118, United States
| | - Vipul Chitalia
- Renal
Section, Department of Medicine, Boston
University Medical Center, Boston, Massachusetts 02118, United States
| | - Catherine E. Costello
- Center
for Biomedical Mass Spectrometry, Boston
University School of Medicine, Boston, Massachusetts 02118, United States
| | - Nader Rahimi
- Department
of Pathology, School of Medicine, Boston
University Medical Campus, Boston, Massachusetts 02118, United States
| |
Collapse
|
9
|
Amraei R, Yin W, Napoleon MA, Suder EL, Berrigan J, Zhao Q, Olejnik J, Chandler KB, Xia C, Feldman J, Hauser BM, Caradonna TM, Schmidt AG, Gummuluru S, Muhlberger E, Chitalia V, Costello CE, Rahimi N. CD209L/L-SIGN and CD209/DC-SIGN act as receptors for SARS-CoV-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2020.06.22.165803. [PMID: 32607506 PMCID: PMC7325172 DOI: 10.1101/2020.06.22.165803] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
As the COVID-19 pandemic continues to spread, investigating the processes underlying the interactions between SARS-CoV-2 and its hosts is of high importance. Here, we report the identification of CD209L/L-SIGN and the related protein CD209/DC-SIGN as receptors capable of mediating SARS-CoV-2 entry into human cells. Immunofluorescence staining of human tissues revealed prominent expression of CD209L in the lung and kidney epithelium and endothelium. Multiple biochemical assays using a purified recombinant SARS-CoV-2 spike receptor binding domain (S-RBD) or S1 encompassing both NTB and RBD and ectopically expressed CD209L and CD209 revealed that CD209L and CD209 interact with S-RBD. CD209L contains two N-glycosylation sequons, at sites N92 and N361, but we determined that only site N92 is occupied. Removal of the N-glycosylation at this site enhances the binding of S-RBD with CD209L. CD209L also interacts with ACE2, suggesting a role for heterodimerization of CD209L and ACE2 in SARS-CoV-2 entry and infection in cell types where both are present. Furthermore, we demonstrate that human endothelial cells are permissive to SARS-CoV-2 infection and interference with CD209L activity by knockdown strategy or with soluble CD209L inhibits virus entry. Our observations demonstrate that CD209L and CD209 serve as alternative receptors for SARS-CoV-2 in disease-relevant cell types, including the vascular system. This property is particularly important in tissues where ACE2 has low expression or is absent, and may have implications for antiviral drug development.
Collapse
Affiliation(s)
- Razie Amraei
- Department of Pathology, School of Medicine, Boston University Medical Campus, Boston, MA 02118
| | - Wenqing Yin
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA
| | - Marc A. Napoleon
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA
| | - Ellen L. Suder
- Department of Microbiology, Boston University School of Medicine, Boston, MA
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA
| | - Jacob Berrigan
- Department of Microbiology, Boston University School of Medicine, Boston, MA
| | - Qing Zhao
- Department of Pathology, School of Medicine, Boston University Medical Campus, Boston, MA 02118
| | - Judith Olejnik
- Department of Microbiology, Boston University School of Medicine, Boston, MA
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA
| | - Kevin Brown Chandler
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, MA 02118
| | - Chaoshuang Xia
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, MA 02118
| | - Jared Feldman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139
| | - Blake M. Hauser
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139
| | | | - Aaron G. Schmidt
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139
- Department of Microbiology, Harvard Medical School, Boston, MA 02115
| | - Suryaram Gummuluru
- Department of Microbiology, Boston University School of Medicine, Boston, MA
| | - Elke Muhlberger
- Department of Microbiology, Boston University School of Medicine, Boston, MA
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA
| | - Vipul Chitalia
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, MA
| | - Catherine E. Costello
- Center for Biomedical Mass Spectrometry, Boston University School of Medicine, Boston, MA 02118
| | - Nader Rahimi
- Department of Pathology, School of Medicine, Boston University Medical Campus, Boston, MA 02118
| |
Collapse
|
10
|
Abstract
The innate immune system is comprised of both cellular and humoral players that recognise and eradicate invading pathogens. Therefore, the interplay between retroviruses and innate immunity has emerged as an important component of viral pathogenesis. HIV-1 infection in humans that results in hematologic abnormalities and immune suppression is well represented by changes in the CD4/CD8 T cell ratio and consequent cell death causing CD4 lymphopenia. The innate immune responses by mucosal barriers such as complement, DCs, macrophages, and NK cells as well as cytokine/chemokine profiles attain great importance in acute HIV-1 infection, and thus, prevent mucosal capture and transmission of HIV-1. Conversely, HIV-1 has evolved to overcome innate immune responses through RNA-mediated rapid mutations, pathogen-associated molecular patterns (PAMPs) modification, down-regulation of NK cell activity and complement receptors, resulting in increased secretion of inflammatory factors. Consequently, epithelial tissues lining up female reproductive tract express innate immune sensors including anti-microbial peptides responsible for forming primary barriers and have displayed an effective potent anti-HIV activity during phase I/II clinical trials.
Collapse
|
11
|
Mezouar S, Katsogiannou M, Ben Amara A, Bretelle F, Mege JL. Placental macrophages: Origin, heterogeneity, function and role in pregnancy-associated infections. Placenta 2020; 103:94-103. [PMID: 33120051 PMCID: PMC7568513 DOI: 10.1016/j.placenta.2020.10.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/22/2020] [Accepted: 10/12/2020] [Indexed: 12/15/2022]
Abstract
Placental macrophages are a heterogenous population of immune cells present throughout pregnancy. They are essential for maintenance of the homeostatic placenta environment and host defense against infections. The characterization of placental macrophages as well as their activation have been limited for a long time by the lack of convenient tools. The emergence of unbiased methods makes it possible to reappraise the study of placental macrophages. In this review, we discuss the diversity and the functions of placental macrophages to better understand their dysfunctions during placental infections.
Collapse
Affiliation(s)
- Soraya Mezouar
- Aix-Marseille Univ, IRD, AP-HM, MEPHI, Marseille, France; IHU - Mediterranean Infection, Marseille, France.
| | - Maria Katsogiannou
- Hôpital Saint Joseph, Department of Obstetrics and Gynecology, FR-13008, Marseille, France
| | - Amira Ben Amara
- Aix-Marseille Univ, IRD, AP-HM, MEPHI, Marseille, France; IHU - Mediterranean Infection, Marseille, France
| | - Florence Bretelle
- Aix-Marseille Univ, IRD, AP-HM, MEPHI, Marseille, France; IHU - Mediterranean Infection, Marseille, France; AP-HM, Gynecology Department, Marseille, France
| | - Jean-Louis Mege
- Aix-Marseille Univ, IRD, AP-HM, MEPHI, Marseille, France; IHU - Mediterranean Infection, Marseille, France; AP-HM, UF Immunology, Marseille, France.
| |
Collapse
|
12
|
Naqvi KF, Endsley JJ. Myeloid C-Type Lectin Receptors in Tuberculosis and HIV Immunity: Insights Into Co-infection? Front Cell Infect Microbiol 2020; 10:263. [PMID: 32582566 PMCID: PMC7283559 DOI: 10.3389/fcimb.2020.00263] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 05/04/2020] [Indexed: 12/11/2022] Open
Abstract
C-type lectin receptors (CLRs) are carbohydrate binding pattern recognition receptors (PRRs) which play a central role in host recognition of pathogenic microorganisms. Signaling through CLRs displayed on antigen presenting cells dictates important innate and adaptive immune responses. Several pathogens have evolved mechanisms to exploit the receptors or signaling pathways of the CLR system to gain entry or propagate in host cells. CLR responses to high priority pathogens such as Mycobacterium tuberculosis (Mtb), HIV, Ebola, and others are described and considered potential avenues for therapeutic intervention. Mtb and HIV are the leading causes of death due to infectious disease and have a synergistic relationship that further promotes aggressive disease in co-infected persons. Immune recognition through CLRs and other PRRs are important determinants of disease outcomes for both TB and HIV. Investigations of CLR responses to Mtb and HIV, to date, have primarily focused on single infection outcomes and do not account for the potential effects of co-infection. This review will focus on CLRs recognition of Mtb and HIV motifs. We will describe their respective roles in protective immunity and immune evasion or exploitation, as well as their potential as genetic determinants of disease susceptibility, and as avenues for development of therapeutic interventions. The potential convergence of CLR-driven responses of the innate and adaptive immune systems in the setting of Mtb and HIV co-infection will further be discussed relevant to disease pathogenesis and development of clinical interventions.
Collapse
Affiliation(s)
- Kubra F Naqvi
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| | - Janice J Endsley
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
13
|
Hendrix P, Tang Z, Silasi M, Racicot KE, Mor G, Abrahams VM, Guller S. Herpesvirus-infected Hofbauer cells activate endothelial cells through an IL-1β-dependent mechanism. Placenta 2020; 91:59-65. [PMID: 32174308 DOI: 10.1016/j.placenta.2020.01.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/10/2019] [Accepted: 01/22/2020] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Placental viral infections are associated with fetal inflammation and adverse pregnancy outcomes. However, there have been limited studies on how placental macrophages in the villous and adjacent fetal umbilical endothelial cells respond to a viral insult. This study aimed to evaluate the communication between Hofbauer cells (HBCs) and human umbilical vein endothelial cells (HUVECs) during a viral infection. METHODS HBCs were either uninfected or infected with the γ-herpesvirus, MHV-68, and the conditioned medium (CM) collected. HUVECs were exposed to HBC CM and the levels of the pro-neutrophilic response markers: IL-8; E-selectin; intercellular adhesion molecule 1 (ICAM-1); and vascular adhesion molecule 1 (VCAM-1) measured by ELISA and qPCR. The role of HBC-derived IL-1β was investigated using an IL-1β blocking antibody (Ab) or IL-1 receptor antagonist (IL-1Ra). RESULTS MHV-68 infection of HBCs induced a significant increase in IL-1β secretion. CM from infected HBCs induced HUVEC expression of IL-8, E-selectin, VCAM-1, ICAM-1 mRNA, and secretion of IL-8. The HUVEC response to the CM of MHV-infected HBCs was inhibited by a neutralizing IL-1β Ab and by IL-1Ra. DISCUSSION Virally-induced HBC IL-1β activates HUVECs to generate a pro-neutrophilic response. This novel cell-cell communication pathway may play an important role in the genesis of fetal inflammation associated with placental viral infection.
Collapse
Affiliation(s)
- Paul Hendrix
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Zhonghua Tang
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Michelle Silasi
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Karen E Racicot
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing, MI, USA
| | - Gil Mor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Vikki M Abrahams
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA.
| | - Seth Guller
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
14
|
DC-SIGN expression in Hofbauer cells may play an important role in immune tolerance in fetal chorionic villi during the development of preeclampsia. J Reprod Immunol 2017; 124:30-37. [PMID: 29049918 DOI: 10.1016/j.jri.2017.09.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 06/19/2017] [Accepted: 09/27/2017] [Indexed: 01/15/2023]
Abstract
Immune tolerance at feto-maternal interfaces is a complex phenomenon. Although maternal decidual macrophages are well-known immune cells, little is known about fetal-derived macrophages (Hofbauer cells) within chorionic villi. Preeclampsia (PE) is a major cause of maternal mortality in the field of obstetrics, and the innate immunological role of maternal decidual macrophages is well known. In this study, we assessed the differential phenotypes and marker expression in fetal macrophages, known as dendritic cell-specific ICAM-grabbing non-integrin (DC-SIGN)-positive Hofbauer cells. We compared Hofbauer cell properties between normal and PE placenta chorionic villi and performed sequential staining of DC-SIGN, CD14, and CD68 to evaluate the existence of Hofbauer cells. Furthermore, to evaluate the immunological function of these cells, we stained the cells for CD163, a marker of immunoregulatory type 2 (M2) macrophages. Additionally, we examined the expression of the immunosuppressive cytokine interleukin (IL)-10, which is known to be produced by M2 macrophages. DC-SIGN+/CD14+, DC-SIGN+/CD68+, and CD163+/DC-SIGN+ cells were quantified based on photomicrographs. The results showed that CD14, CD163, DC-SIGN, and IL-10 levels were significantly downregulated in PE compared with normal. Additionally, CD163+/DC-SIGN+ Hofbauer cells were significantly less frequent in PE than in normal. DC-SIGN Hofbauer cells produced IL-10 at lower levels in the PE than in the normal. Thus, we speculate that fetal-derived Hofbauer cells may play an important role in normal pregnancy with immunosuppressive effects based on their M2 macrophage characteristics to maintain immune tolerance during pregnancy. Additionally, in PE, these functions were defective, supporting the roles of these macrophages in PE development.
Collapse
|
15
|
Mitchell DA, Zhang Q, Voorhaar L, Haddleton DM, Herath S, Gleinich AS, Randeva HS, Crispin M, Lehnert H, Wallis R, Patterson S, Becer CR. Manipulation of cytokine secretion in human dendritic cells using glycopolymers with picomolar affinity for DC-SIGN. Chem Sci 2017; 8:6974-6980. [PMID: 29147524 PMCID: PMC5642150 DOI: 10.1039/c7sc01515a] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/11/2017] [Indexed: 12/25/2022] Open
Abstract
The human C-type lectin DC-SIGN (CD209) is a significant receptor on the surface of dendritic cells (DCs) - crucial components of host defense that bridge the innate and adaptive immune systems. A range of linear glycopolymers, constructed via controlled radical polymerization techniques have been shown to interact with DC-SIGN with affinities in the physiologically active range. However, these first generation glycopolymers possess limited structural definition and their effects on DCs were not known. Here we report the development of star-shaped mannose glycopolymers with the aim of targeting the clustered domain arrangement of DC-SIGN and these were shown to bind with picomolar affinity. Increased secretion of IL-10 with simultaneous decrease in secreted IL-12p70 occurred in activated DCs incubated with star-shaped glycopolymers - a cytokine secretion pattern characteristic of wound-healing tissue environments. Incorporating stellar architecture into glycopolymer design could be key to developing selective and very high-affinity therapeutic materials with distinct immunomodulatory and tissue repair potential.
Collapse
Affiliation(s)
- Daniel A Mitchell
- Clinical Sciences Research Laboratories , University of Warwick , Coventry CV2 2DX , United Kingdom . .,University Hospital Coventry , Warwickshire NHS Trust , Coventry CV2 2DX , United Kingdom
| | - Qiang Zhang
- Department of Chemistry , University of Warwick , Coventry CV4 7AL , United Kingdom
| | - Lenny Voorhaar
- Department of Chemistry , University of Warwick , Coventry CV4 7AL , United Kingdom
| | - David M Haddleton
- Department of Chemistry , University of Warwick , Coventry CV4 7AL , United Kingdom
| | - Shan Herath
- Chelsea & Westminster Hospital , Imperial College School of Medicine , London SW10 9NH , United Kingdom
| | - Anne S Gleinich
- Clinical Sciences Research Laboratories , University of Warwick , Coventry CV2 2DX , United Kingdom .
| | - Harpal S Randeva
- Clinical Sciences Research Laboratories , University of Warwick , Coventry CV2 2DX , United Kingdom . .,University Hospital Coventry , Warwickshire NHS Trust , Coventry CV2 2DX , United Kingdom
| | - Max Crispin
- Glycobiology Institute , University of Oxford , Oxford OX1 3QU , United Kingdom
| | | | - Russell Wallis
- Department of Biochemistry , University of Leicester , Leicester LE1 9HN , United Kingdom
| | - Steven Patterson
- Chelsea & Westminster Hospital , Imperial College School of Medicine , London SW10 9NH , United Kingdom
| | - C Remzi Becer
- School of Engineering and Materials Science , Queen Mary University , London E1 4NS , United Kingdom .
| |
Collapse
|
16
|
Dodagatta-Marri E, Mitchell DA, Pandit H, Sonawani A, Murugaiah V, Idicula-Thomas S, Nal B, Al-Mozaini MM, Kaur A, Madan T, Kishore U. Protein-Protein Interaction between Surfactant Protein D and DC-SIGN via C-Type Lectin Domain Can Suppress HIV-1 Transfer. Front Immunol 2017; 8:834. [PMID: 28824609 PMCID: PMC5534670 DOI: 10.3389/fimmu.2017.00834] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 07/03/2017] [Indexed: 01/01/2023] Open
Abstract
Surfactant protein D (SP-D) is a soluble C-type lectin, belonging to the collectin (collagen-containing calcium-dependent lectin) family, which acts as an innate immune pattern recognition molecule in the lungs at other mucosal surfaces. Immune regulation and surfactant homeostasis are salient functions of SP-D. SP-D can bind to a range of viral, bacterial, and fungal pathogens and trigger clearance mechanisms. SP-D binds to gp120, the envelope protein expressed on HIV-1, through its C-type lectin or carbohydrate recognition domain. This is of importance since SP-D is secreted by human mucosal epithelial cells and is present in the female reproductive tract, including vagina. Another C-type lectin, dendritic cell (DC)-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), present on the surface of the DCs, also binds to HIV-1 gp120 and facilitates viral transfer to the lymphoid tissues. DCs are also present at the site of HIV-1 entry, embedded in vaginal or rectal mucosa. In the present study, we report a direct protein-protein interaction between recombinant forms of SP-D (rfhSP-D) and DC-SIGN via their C-type lectin domains. Both SP-D and DC-SIGN competed for binding to immobilized HIV-1 gp120. Pre-incubation of human embryonic kidney cells expressing surface DC-SIGN with rfhSP-D significantly inhibited the HIV-1 transfer to activated peripheral blood mononuclear cells. In silico analysis revealed that SP-D and gp120 may occupy same sites on DC-SIGN, which may explain the reduced transfer of HIV-1. In summary, we demonstrate, for the first time, that DC-SIGN is a novel binding partner of SP-D, and this interaction can modulate HIV-1 capture and transfer to CD4+ T cells. In addition, the present study also reveals a novel and distinct mechanism of host defense by SP-D against HIV-1.
Collapse
Affiliation(s)
- Eswari Dodagatta-Marri
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Daniel A Mitchell
- Clinical Sciences Research Laboratories, Warwick Medical School, University Hospital Coventry and Warwickshire Campus, Coventry, United Kingdom
| | - Hrishikesh Pandit
- Department of Innate Immunity, National Institute for Research in Reproductive Health, Indian Council of Medical Research, Mumbai, India
| | - Archana Sonawani
- Department of Innate Immunity, National Institute for Research in Reproductive Health, Indian Council of Medical Research, Mumbai, India
| | - Valarmathy Murugaiah
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Susan Idicula-Thomas
- Department of Innate Immunity, National Institute for Research in Reproductive Health, Indian Council of Medical Research, Mumbai, India
| | - Béatrice Nal
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom.,Institute of Environment, Health and Societies, Brunel University London, Uxbridge, United Kingdom
| | - Maha M Al-Mozaini
- Department of Infection and Immunity, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Anuvinder Kaur
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| | - Taruna Madan
- Department of Innate Immunity, National Institute for Research in Reproductive Health, Indian Council of Medical Research, Mumbai, India
| | - Uday Kishore
- Department of Life Sciences, College of Health and Life Sciences, Brunel University London, Uxbridge, United Kingdom
| |
Collapse
|
17
|
Gavegnano C, Bassit LC, Cox BD, Hsiao HM, Johnson EL, Suthar M, Chakraborty R, Schinazi RF. Jak Inhibitors Modulate Production of Replication-Competent Zika Virus in Human Hofbauer, Trophoblasts, and Neuroblastoma cells. Pathog Immun 2017; 2:199-218. [PMID: 28776046 PMCID: PMC5538373 DOI: 10.20411/pai.v2i2.190] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Zika Virus (ZIKV) is a flavivirus that has been implicated in causing brain deformations, birth defects, and microcephaly in fetuses, and associated with Guillain-Barre syndrome. Mechanisms responsible for transmission of ZIKV across the placenta to the fetus are incompletely understood. Herein, we define key events modulating infection in clinically relevant cells, including primary placental macrophages (human Hofbauer cells; HC), trophoblasts, and neuroblastoma cells. Consistent with previous findings, HC and trophoblasts are permissive to ZIKV infection. Decrease of interferon signaling by Jak ½ inhibition (using ruxolitinib) significantly increased ZIKV replication in HC, trophoblasts, and neuroblasts. Enhanced ZIKV production in ruxolitinib-treated HC was associated with increased expression of HLA-DR and DC-SIGN. Nucleoside analogs blocked ruxolitinib-mediated production of extracellular virus. Although low-level ZIKV infection occurred in untreated HC and trophoblasts, replicating virions were incapable of infecting naive Vero cells. These deficient virions from untreated HC have “thin-coats” suggesting an immature structure. Blocking Jak ½ signaling (with ruxolitinib) restored replication competence as virions produced under these conditions confer cytopathic effects to naive Vero cells. These data demonstrate that Jak-STAT signaling directly impacts the ability of primary placental cells to produce replication-competent virus and is a key determinant in the production of mature virions in clinically relevant cells, including HC and trophoblasts. Design of targeted agents to prevent ZIKV replication in the placenta should consider Jak ½ signaling, the impact of its block on ZIKV infection, and subsequent transmission to the fetus.
Collapse
Affiliation(s)
- Christina Gavegnano
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Leda C Bassit
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Bryan D Cox
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Hui-Mien Hsiao
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Erica L Johnson
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Mehul Suthar
- Emory Vaccine Center, Yerkes National Primate Center, Emory University, Atlanta, Georgia
| | - Rana Chakraborty
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| | - Raymond F Schinazi
- Center for AIDS Research, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University, Atlanta, Georgia
| |
Collapse
|
18
|
Abstract
Pregnancy complications such as preterm birth, miscarriage, maternal and/or neonatal morbidities, and mortality can be manifestations of underlying placental pathology. Hofbauer cells refer to a heterogeneous population of fetal macrophages that reside within the functional unit of the placenta known as the chorionic villus. Hofbauer cells can be detected within the connective tissue matrix of the placenta as early as 4 weeks post-conception and are present throughout pregnancy. These cells are implicated in a wide array of functions important for a successful pregnancy including placental morphogenesis, immune regulation, control of stromal water content, and the transfer of ions and serum proteins across the maternal-fetal barrier. Derangements in Hofbauer cell homeostasis are associated with placental pathologies involving infection, inflammation, and inadequate placental development. Despite a growing body of evidence that these cells are important, our knowledge about Hofbauer cell function in both normal and dysfunctional pregnancy is rudimentary. The goal of this chapter is to provide an overview of what is known about Hofbauer cell origins and their potential roles in normal and complicated pregnancy. We also review established and emerging methodologies available for the study of Hofbauer cells during in vitro and in vivo conditions.
Collapse
Affiliation(s)
- Leticia Reyes
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI, 53706, USA.
| | - Bryce Wolfe
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Thaddeus Golos
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
19
|
Small non-coding RNAs transfer through mammalian placenta and directly regulate fetal gene expression. Protein Cell 2016; 6:391-396. [PMID: 25963995 PMCID: PMC4444809 DOI: 10.1007/s13238-015-0156-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
20
|
The evolution of HIV-1 interactions with coreceptors and mannose C-type lectin receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 129:109-40. [PMID: 25595802 DOI: 10.1016/bs.pmbts.2014.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The phenotype of human immunodeficiency virus type 1 (HIV-1) commonly evolves between and within infected individuals, at virus transmission, and during disease progression. This evolution includes altered interactions between the virus and its coreceptors, i.e., chemokine receptors, as well as mannose C-type lectin receptors (CLRs). Transmitted/founder viruses are predominantly restricted to CCR5, whereas the subsequent intrapatient evolution of HIV-1 coreceptor use during progressive disease can be subdivided into two distinct pathways. Accordingly, the CCR5-restricted virus population is either gradually replaced by virus variants able to use CXCR4 or evolves toward an altered, more flexible use of CCR5. Despite a strong dependency on these coreceptors for host cell entry, HIV-1 also interacts with other cell surface molecules during target cell attachment, including the CLRs. The virus interaction with the CLRs may result either in the efficient transfer of virus to CD4(+) T cells or in the degradation of the virus in endosomal compartments. The determinants of the diverse outcomes depend on which CLR is engaged and also on the glycan makeup of the envelope glycoproteins, which may evolve with the strength of the immune pressure during the disease course. With the current clinical introduction of CCR5 antagonists and the development of additional entry inhibitors, knowledge on the evolution and baseline characteristics of HIV-1 interactions with coreceptor and CLR interactions may play important roles for individualized and optimized treatment strategies. This review summarizes our current understanding of the evolution of HIV-1 interactions with these receptors.
Collapse
|
21
|
Abstract
The C-type lectins DC-SIGN, DC-SIGNR and LSECtin are encoded by the lectin gene cluster on chromosome 19p13.3 and perform cell-adhesion and pathogen recognition functions on dendritic cells, liver cells and lymph node sinusoidal endothelial cells. DC-SIGN and DC-SIGNR share similar overall gene and protein molecule structures, and they exhibit high affinity for high-mannose carbohydrates. LSECtin, a Ca2+-dependent C-type lectin, interacts with mannose, NAcGlc and fucose. These lectins allow pathogen recognition (e.g., viruses, bacteria and allergens) and cell adhesion for dendritic and endothelial cells in different tissues, which may enhance the infection and facilitate the spread of those pathogens. A better understanding of these lectins may yield information about how pathogens are captured by particular cells and how they spread in different tissues. These studies would provide more detail about the physiopathological mechanisms of viral and bacterial infections and may also lead to new strategies to treat or prevent infections.
Collapse
Affiliation(s)
- Feng Zhang
- 1Department of Clinical Biochemistry, College of Laboratory Diagnostic Medicine, Dalian Medical University, Dalian, China
| | | | | |
Collapse
|
22
|
Abstract
A number of advances in recent years have significantly furthered our understanding of filovirus attachment and cellular tropism. For example, several cell-surface molecules have been identified as attachment factors with the potential to facilitate the in vivo targeting of particular cell types such as macrophages and hepatic cells. Furthermore, our knowledge of internalization and subsequent events during filovirus entry has also been widened, adding new variations to the paradigms for viral entry established for HIV and influenza. In particular, host cell factors such as endosomal proteases and the intracellular receptor Niemann-Pick C1 are now known to play a vital role in activating the membrane fusion potential of filovirus glycoproteins.
Collapse
Affiliation(s)
- Stefan Pöhlmann
- grid.10423.340000000095299877Institute for Virology, Hannover Medical School, Hannover, Germany ,grid.418215.b0000000085027018German Primate Center, Göttingen, Germany
| | - Graham Simmons
- grid.266102.10000000122976811Blood Systems Research Institute, and Department of Laboratory Medicine, University of California San Francisco, San Francisco, California USA
| |
Collapse
|
23
|
Garcia-Vallejo JJ, van Kooyk Y. The physiological role of DC-SIGN: a tale of mice and men. Trends Immunol 2013; 34:482-6. [PMID: 23608151 DOI: 10.1016/j.it.2013.03.001] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 03/08/2013] [Accepted: 03/10/2013] [Indexed: 02/01/2023]
Abstract
The innate immune receptor DC-SIGN (dendritic cell-specific intercellular adhesion molecule-3 grabbing non-integrin) was discovered over a decade ago and was initially identified as a pattern recognition receptor. In addition to its ability to recognize a broad range of pathogen-derived ligands and self-glycoproteins, DC-SIGN also mediates intercellular adhesion, as well as antigen uptake and signaling, which is a functional hallmark of dendritic cells (DCs). Most research on DC-SIGN has relied on in vitro studies. The in vivo function of DC-SIGN is difficult to address, in part because there are eight genetic homologs in mice with no clear DC-SIGN ortholog. Here, we summarize the functions attributed to DC-SIGN based on in vitro data and discuss the limitations of available mouse models to uncover the physiological role of this receptor in vivo.
Collapse
Affiliation(s)
- Juan J Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, VU University Medical Center, Amsterdam, 1081BT, The Netherlands
| | | |
Collapse
|
24
|
Borggren M, Navér L, Casper C, Ehrnst A, Jansson M. R5 human immunodeficiency virus type 1 with efficient DC-SIGN use is not selected for early after birth in vertically infected children. J Gen Virol 2013; 94:767-773. [DOI: 10.1099/vir.0.043620-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The binding of human immunodeficiency virus (HIV) to C-type lectin receptors may result in either enhanced trans-infection of T-cells or virus degradation. We have investigated the efficacy of HIV-1 utilization of DC-SIGN, a C-type lectin receptor, in the setting of intrauterine or intrapartum mother-to-child transmission (MTCT). Viruses isolated from HIV-1-infected mothers at delivery and from their vertically infected children both shortly after birth and later during the progression of the disease were analysed for their use of DC-SIGN, binding and ability to trans-infect. DC-SIGN use of a child’s earlier virus isolate tended to be reduced as compared with that of the corresponding maternal isolate. Furthermore, the children’s later isolate displayed enhanced DC-SIGN utilization compared with that of the corresponding earlier virus. These results were also supported in head-to-head competition assays and suggest that HIV-1 variants displaying efficient DC-SIGN use are not selected for during intrauterine or intrapartum MTCT. However, viruses with increased DC-SIGN use may evolve later in paediatric HIV-1 infections.
Collapse
Affiliation(s)
- Marie Borggren
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Lars Navér
- Department of Clinical Science, Interventions and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatrics, Karolinska University Hospital, Stockholm, Sweden
| | - Charlotte Casper
- INSERM U1043, Hospital Purpan, Paul Sabatier University, Toulouse, France
| | - Anneka Ehrnst
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Marianne Jansson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| |
Collapse
|
25
|
Dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin mediates HIV-1 infection of and transmission by M2a-polarized macrophages in vitro. AIDS 2013; 27:707-16. [PMID: 23211775 DOI: 10.1097/qad.0b013e32835cfc82] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To assess in-vitro effects of monocyte-derived macrophage (MDM) polarization into M1 and M2a cells on HIV-1 replication and transmission and obtain new insights into the potential importance of macrophage polarization in vivo. DESIGN Human peripheral blood monocytes were differentiated into MDM for 7 days. Control and MDM polarized into M1 or M2a cells were exposed to different strains of HIV-1 and assessed for their ability to bind and transmit virus to CD4 T lymphocytes. METHODS MDM were incubated with either tumour necrosis factor-alpha (TNF-α) along with interferon-gamma (IFN-γ) or with interleukin-4 (IL-4) for 18 h to obtain M1 or M2a cells, respectively. Expression of cell surface antigens, including CD4 and dendritic cell-specific intercellular adhesion molecule-3 grabbing nonintegrin (DC-SIGN), was evaluated by flow cytometry. C-C chemokine receptor type 5 (CCR5)-dependent (R5) HIV-1 binding, DNA synthesis and viral replication were assessed in the presence or absence of anti-DC-SIGN blocking mAbs. Transmission of C-X-C chemokine receptor type 4 (CXCR4)-dependent (X4) and R5 HIV-1 from MDM to IL-2 activated CD4 T cells was also investigated. RESULTS DC-SIGN was strongly upregulated on M2a-MDM and downregulated on M1-MDM compared with control MDM. DC-SIGN facilitated HIV-1 entry and DNA synthesis in M2a-MDM, compensating for their low levels of CD4 cell expression. M2a-MDM efficiently transmitted both R5 and X4 HIV-1 to CD4 T cells in a DC-SIGN-dependent manner. CONCLUSION DC-SIGN facilitates HIV-1 infection of M2a-MDM, and HIV-1 transfer from M2a-MDM to CD4 T cells. M2a-polarized tissue macrophages may play an important role in the capture and spread of HIV-1 in mucosal tissues and placenta.
Collapse
|
26
|
Abstract
The immune cells that reside at the interface between the placenta and uterus are thought to play many important roles in pregnancy. Recent work has revealed that the composition and function of these cells are locally controlled by the specialized uterine stroma (the decidua) that surrounds the implanted conceptus. Here, I discuss how key immune cell types (natural killer cells, macrophages, dendritic cells, and T cells) are either enriched or excluded from the decidua, how their function is regulated within the decidua, and how they variously contribute to pregnancy success or failure. The discussion emphasizes the relationship between human and mouse studies. Deeper understanding of the immunology of the maternal-fetal interface promises to yield significant insight into the pathogenesis of many human pregnancy complications, including preeclampsia, intrauterine growth restriction, spontaneous abortion, preterm birth, and congenital infection.
Collapse
Affiliation(s)
- Adrian Erlebacher
- Department of Pathology and NYU Cancer Institute, NYU School of Medicine, NYU Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
27
|
Johnson EL, Chakraborty R. Placental Hofbauer cells limit HIV-1 replication and potentially offset mother to child transmission (MTCT) by induction of immunoregulatory cytokines. Retrovirology 2012; 9:101. [PMID: 23217137 PMCID: PMC3524025 DOI: 10.1186/1742-4690-9-101] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 11/16/2012] [Indexed: 12/26/2022] Open
Abstract
Background Despite readily detectable levels of the HIV-1 (co)-receptors CD4, CCR5 and DC-SIGN on placental macrophages (Hofbauer Cells [HCs]), the rate of HIV-1 infection in utero in the absence of interventions is only 7% of exposed infants. Here, we examine the replication kinetics of human HCs to the primary isolate HIV-1BaL. We also determined the infectivity of HIV-1-exposed HCs by co-culturing with isolated cord and peripheral blood mononuclear cells [CBMCs, PBMCs]. To understand the limiting nature of HCs to HIV-1 replication, we examined the effect of endogenously secreted cytokines on replication kinetics. Results HCs have reduced ability to replicate HIV-1 in vitro (p < 0.01) and to transmit virus to CBMCs and PBMCs (p < 0.001 for both) compared to standard infections of MDMs. HCs were shown to release HIV-1 particles at levels comparable to MDMs, however exhibit significant decreases in viral transcription (gag and env), which may account for lower levels of HIV-1 replication. Un-stimulated HCs constitutively express significantly higher levels of regulatory cytokines, IL-10 and TGF-β, compared to MDMs (p < 0.01), which may contribute to immunoregulatory predominance at the placenta and possibly account for down-regulation of HIV-1 replication and infectivity by HCs. We further demonstrate that these regulatory cytokines inhibit HIV-1 replication within HCs in vitro. Conclusion HCs have reduced ability to replicate and disseminate R5-tropic HIV-1BaLin vitro and potentially offset mother to child transmission (MTCT) of HIV-1 by the induction of immunoregulatory cytokines. Despite the potential for migration and infectivity, HCs are not present in the neighboring fetal circulation. These results implicate HCs as important mediators of protection at the feto-maternal interface during ongoing HIV-1 exposure.
Collapse
Affiliation(s)
- Erica L Johnson
- Department of Pediatrics and Children's Healthcare of Atlanta, Emory University, Atlanta, GA 30322, USA
| | | |
Collapse
|
28
|
da Silva RC, Segat L, Zanin V, Arraes LC, Crovella S. Polymorphisms in DC-SIGN and L-SIGN genes are associated with HIV-1 vertical transmission in a Northeastern Brazilian population. Hum Immunol 2012; 73:1159-65. [DOI: 10.1016/j.humimm.2012.07.338] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Revised: 07/11/2012] [Accepted: 07/30/2012] [Indexed: 10/28/2022]
|
29
|
Boily-Larouche G, Milev MP, Zijenah LS, Labbé AC, Zannou DM, Humphrey JH, Ward BJ, Poudrier J, Mouland AJ, Cohen ÉA, Roger M. Naturally-occurring genetic variants in human DC-SIGN increase HIV-1 capture, cell-transfer and risk of mother-to-child transmission. PLoS One 2012; 7:e40706. [PMID: 22808239 PMCID: PMC3393705 DOI: 10.1371/journal.pone.0040706] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 06/12/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Mother-to-child transmission (MTCT) is the main cause of HIV-1 infection in children worldwide. Dendritic cell-specific ICAM-3 grabbing-nonintegrin (DC-SIGN, also known as CD209) is an HIV-1 receptor that enhances its transmission to T cells and is expressed on placental macrophages. METHODS AND FINDINGS We have investigated the association between DC-SIGN genetic variants and risk of MTCT of HIV-1 among Zimbabwean infants and characterized the impact of the associated mutations on DC-SIGN expression and interaction with HIV-1. DC-SIGN promoter (p-336C and p-201A) and exon 4 (198Q and 242V) variants were all significantly associated with increased risk of intrauterine (IU) HIV-1 infection. Promoter variants decreased DC-SIGN expression both in vitro and in placental CD163(+) macrophages (Hofbauer cells) of HIV-1 unexposed infants but not of HIV-1 exposed infants. The exon 4 protein-modifying mutations increased HIV-1 capture and transmission to T cells in vitro. CONCLUSION This study provides compelling evidence to support an important role of DC-SIGN in IU HIV-1 infection.
Collapse
Affiliation(s)
- Geneviève Boily-Larouche
- Laboratoire d’Immunogénétique, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada
- Département de Microbiologie et Immunologie, Université de Montréal, Montréal, Canada
| | - Miroslav P. Milev
- Department of Medicine, McGill University, The Lady Davis Institute for Medical Research and McGill AIDS Center, Montreal, Canada
| | - Lynn S. Zijenah
- Department of Immunology, University of Zimbabwe College of Health Sciences, Harare, Zimbabwe
| | - Annie-Claude Labbé
- Département de Microbiologie et Immunologie, Université de Montréal, Montréal, Canada
- Département de Microbiologie de l’hôpital Maisonneuve-Rosemont, Montréal, Canada
| | - Djimon M. Zannou
- Centre National Hospitalier Universitaire, Université d’Abomey Calavi, Cotonou, Bénin
| | - Jean H. Humphrey
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Brian J. Ward
- Research Institute of the McGill University Health Centre, Montreal, Canada
| | - Johanne Poudrier
- Laboratoire d’Immunogénétique, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada
- Département de Microbiologie et Immunologie, Université de Montréal, Montréal, Canada
| | - Andrew J. Mouland
- Department of Medicine, McGill University, The Lady Davis Institute for Medical Research and McGill AIDS Center, Montreal, Canada
| | - Éric A. Cohen
- Département de Microbiologie et Immunologie, Université de Montréal, Montréal, Canada
- Institut de Recherches Cliniques de Montréal, Montréal, Canada
| | - Michel Roger
- Laboratoire d’Immunogénétique, Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), Montréal, Canada
- Département de Microbiologie et Immunologie, Université de Montréal, Montréal, Canada
- * E-mail:
| |
Collapse
|
30
|
Abstract
The human immune system is under constant challenge from many viruses, some of which the body is successfully able to clear. Other viruses have evolved to escape the host immune responses and thus persist, leading to the development of chronic diseases. Dendritic cells are professional antigen-presenting cells that play a major role in both innate and adaptive immunity against different pathogens. This review focuses on the interaction of different chronic viruses with dendritic cells and the viruses' ability to exploit this critical cell type to their advantage so as to establish persistence within the host.
Collapse
Affiliation(s)
- Saifur Rahman
- Department of Microbiology and Immunology, Drexel Institute for Biotechnology and Virology Research, Drexel University College of Medicine, 3805 Old Easton Road, Doylestown, PA 18902, USA
| | | | | |
Collapse
|
31
|
Cook JA. Associations between use of crack cocaine and HIV-1 disease progression: research findings and implications for mother-to-infant transmission. Life Sci 2011; 88:931-9. [PMID: 21219914 PMCID: PMC3100443 DOI: 10.1016/j.lfs.2011.01.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2010] [Revised: 10/08/2010] [Accepted: 12/23/2010] [Indexed: 10/18/2022]
Abstract
Recent in vitro and in vivo research has suggested that cocaine has a direct effect on the pathogenesis of AIDS. These findings are confirmed by epidemiological studies linking the use of injected, inhaled, and smoked (crack) cocaine and indicators of HIV disease progression, even among adherent users of highly active antiretroviral therapy. Recent studies of vertical HIV transmission suggest that cocaine use may play a role in mother-to-child infection via alteration of maternal immune responses, enhanced viral replication in maternal immune cells, or alterations in the immune systems of neonates or infants. The purpose of this article is to review research conducted over the past several decades on associations between use of cocaine and HIV disease progression, especially among HIV+ women, and to explore its potential relevance for understanding mother-to-infant transmission of HIV.
Collapse
Affiliation(s)
- Judith A Cook
- Department of Psychiatry, University of Illinois at Chicago, United States.
| |
Collapse
|
32
|
Purohit V, Rapaka RS, Schnur P, Shurtleff D. Potential impact of drugs of abuse on mother-to-child transmission (MTCT) of HIV in the era of highly active antiretroviral therapy (HAART). Life Sci 2011; 88:909-16. [DOI: 10.1016/j.lfs.2011.03.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2011] [Accepted: 03/19/2011] [Indexed: 11/16/2022]
|
33
|
Joerink M, Rindsjö E, van Riel B, Alm J, Papadogiannakis N. Placental macrophage (Hofbauer cell) polarization is independent of maternal allergen-sensitization and presence of chorioamnionitis. Placenta 2011; 32:380-5. [PMID: 21419483 DOI: 10.1016/j.placenta.2011.02.003] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Revised: 02/21/2011] [Accepted: 02/22/2011] [Indexed: 12/19/2022]
Abstract
BACKGROUND Macrophages can polarize in which M1/classically activated and M2/alternatively activated macrophages are considered to be the extremes. M1 macrophages are involved in inflammatory reactions, while M2 macrophages are suggested to be involved in homeostasis, parasite killing, tumor promotion, tissue remodeling and in allergic reactions. We hypothesized that polarization of placental macrophages (Hofbauer cells) is influenced by the allergen-sensitization status of the mother and/or the presence of chorioamnionitis, a placental inflammation. This Hofbauer cell polarization might be associated to the intrauterine environment and influence the risk of allergy development for the child. Therefore we aimed to determine the polarization status of Hofbauer cells in health and disease. METHODS We determined the expression of CD68, CX3CR1, IL-7R, DC-SIGN/CD209 and CD163 in placentas of sensitized versus non-sensitized mothers (n = 17), and placentas with or without histological chorioamnionitis (n = 10) by means of immunohistochemical analysis and quantitative real-time PCR (qPCR). RESULTS Protein expression of the M1 markers (CX3CR1, IL-7R and CCR7) could not be detected in any of the analyzed samples while the M2 markers (DC-SIGN, CD163 and mannose receptor/CD206) were readily detected. Significant differences between non-sensitized versus sensitized mothers and uncomplicated versus chorioamnionitis complicated pregnancies were not detected at protein or at mRNA expression level. CONCLUSIONS These results suggest that Hofbauer cells have an M2 phenotype, and that their polarization is not affected by maternal allergen-sensitization or by presence of chorioamnionitis.
Collapse
Affiliation(s)
- M Joerink
- Department of Medicine Solna, Clinical Allergy Research Unit, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
34
|
Tirado-González I, Muñoz-Fernández R, Blanco O, Leno-Durán E, Abadía-Molina AC, Olivares EG. Reduced proportion of decidual DC-SIGN+ cells in human spontaneous abortion. Placenta 2011; 31:1019-22. [PMID: 20934749 DOI: 10.1016/j.placenta.2010.09.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2010] [Revised: 09/10/2010] [Accepted: 09/14/2010] [Indexed: 11/30/2022]
Abstract
Recent studies showed that some functions of decidual dendritic cells appear to be essential for pregnancy. In humans, decidual dendritic cells are identifiable by their expression of DC-SIGN. We compared the subpopulations of human decidual DC-SIGN+ cells from first-trimester normal pregnancies and spontaneous abortions by flow cytometry. In normal decidua, DC-SIGN+ cells expressed antigens associated with immature myeloid dendritic cells. In samples from spontaneous abortions, we detected decidual DC-SIGN+ cells with an antigen phenotype equivalent to that of DC-SIGN+ cells from normal pregnancies, but at a significantly lower proportion (P < 0.01). Our results support the hypothesis that dendritic cells play a role in normal or pathological human pregnancy outcomes.
Collapse
Affiliation(s)
- I Tirado-González
- Instituto de Biopatología y Medicina Regenerativa, Centro de Investigación Biomédica, Universidad de Granada, 18100 Armilla, Granada, Spain
| | | | | | | | | | | |
Collapse
|
35
|
Role of DC-SIGN and L-SIGN receptors in HIV-1 vertical transmission. Hum Immunol 2011; 72:305-11. [PMID: 21277928 PMCID: PMC7115691 DOI: 10.1016/j.humimm.2011.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Revised: 12/23/2010] [Accepted: 01/13/2011] [Indexed: 12/28/2022]
Abstract
The innate immune system acts in the first line of host defense against pathogens. One of the mechanisms used involves the early recognition and uptake of microbes by host professional phagocytes, through pattern recognition receptors (PRRs). These PRRs bind to conserved microbial ligands expressed by pathogens and initiate both innate and adaptative immune responses. Some PRRs located on the surface of dendritic cells (DCs) and other cells seem to play an important role in human immunodeficiency virus type 1 (HIV-1) transmission. Dendritic cell-specific intercellular adhesion molecule-3 grabbing non-integrin, CD209 (DC-SIGN) and its homolog, DC-SIGN-related (DC-SIGNR or L-SIGN) receptors are PPRs able to bind the HIV-1 gp120 envelope protein and, because alterations in their expression patterns also occur, they might play a role in both horizontal and vertical transmission as well as in disseminating the virus within the host. This review aims to explore the involvement of the DC-SIGN and L-SIGN receptors in HIV-1 transmission from mother to child.
Collapse
|
36
|
Cheong C, Matos I, Choi JH, Schauer JD, Dandamudi DB, Shrestha E, Makeyeva JA, Li X, Li P, Steinman RM, Park CG. New monoclonal anti-mouse DC-SIGN antibodies reactive with acetone-fixed cells. J Immunol Methods 2010; 360:66-75. [PMID: 20558171 PMCID: PMC2924951 DOI: 10.1016/j.jim.2010.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2010] [Accepted: 06/07/2010] [Indexed: 10/19/2022]
Abstract
Mouse DC-SIGN CD209a is a type II transmembrane protein, one of a family of C-type lectin genes syntenic and homologous to human DC-SIGN. Current anti-mouse DC-SIGN monoclonal antibodies (MAbs) are unable to react with DC-SIGN in acetone-fixed cells, limiting the chance to visualize DC-SIGN in tissue sections. We first produced rabbit polyclonal PAb-DSCYT14 against a 14-aa peptide in the cytosolic domain of mouse DC-SIGN, and it specifically detected DC-SIGN and not the related lectins, SIGN-R1 and SIGN-R3 expressed in transfected CHO cells. MAbs were generated by immunizing rats and DC-SIGN knockout mice with the extracellular region of mouse DC-SIGN. Five rat IgG2a or IgM MAbs, named BMD10, 11, 24, 25, and 30, were selected and each MAb specifically detected DC-SIGN by FACS and Western blots, although BMD25 was cross-reactive to SIGN-R1. Two mouse IgG2c MAbs MMD2 and MMD3 interestingly bound mouse DC-SIGN but at 10 fold higher levels than the rat MAbs. When the binding epitopes of the new BMD and two other commercial rat anti-DC-SIGN MAbs, 5H10 and LWC06, were examined by competition assays, the epitopes of BMD11, 24, and LWC06 were identical or closely overlapping while BMD10, 30, and 5H10 were shown to bind different epitopes. MMD2 and MMD3 epitopes were on a 3rd noncompeting region of mouse DC-SIGN. DC-SIGN expressed on the cell surface was sensitive to collagenase treatment, as monitored by polyclonal and MAb. These new reagents should be helpful to probe the biology of DC-SIGN in vivo.
Collapse
MESH Headings
- Acetone/metabolism
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, CD/metabolism
- CHO Cells
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/immunology
- Cell Adhesion Molecules/metabolism
- Cricetinae
- Cricetulus
- Cross Reactions/immunology
- Epitope Mapping
- Epitopes/metabolism
- Female
- Hybridomas
- Immunization
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Lectins, C-Type/metabolism
- Mice
- Mice, Knockout
- Protein Engineering
- Protein Structure, Tertiary/genetics
- Rabbits
- Rats
- Rats, Inbred WF
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Recombinant Fusion Proteins/genetics
- Tissue Fixation
Collapse
Affiliation(s)
- Cheolho Cheong
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Ines Matos
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Jae-Hoon Choi
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Joseph D. Schauer
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Durga Bhavani Dandamudi
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Elina Shrestha
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Jessy A. Makeyeva
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Xiaojun Li
- Department of Biochemistry and Biophysics, Texas A&M University, 2128 TAMU, College Station, TX 77843-2128
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, 2128 TAMU, College Station, TX 77843-2128
| | - Ralph M. Steinman
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Chae Gyu Park
- Laboratory of Cellular Physiology and Immunology and Chris Browne Center for Immunology and Immune Diseases, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| |
Collapse
|
37
|
Joubert BR, Lange EM, Franceschini N, Mwapasa V, North KE, Meshnick SR. A whole genome association study of mother-to-child transmission of HIV in Malawi. Genome Med 2010; 2:17. [PMID: 20487506 PMCID: PMC2873795 DOI: 10.1186/gm138] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Revised: 09/16/2009] [Accepted: 03/01/2010] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND More than 300,000 children are newly infected with HIV each year, predominantly through mother-to-child transmission (HIV MTCT). Identification of host genetic traits associated with transmission may more clearly explain the mechanisms of HIV MTCT and further the development of a vaccine to protect infants from infection. Associations between transmission and a selection of genes or single nucleotide polymorphisms (SNP)s may give an incomplete picture of HIV MTCT etiology. Thus, this study employed a genome-wide association approach to identify novel variants associated with HIV MTCT. METHODS We conducted a nested case-control study of HIV MTCT using infants of HIV(+) mothers, drawn from a cohort study of malaria and HIV in pregnancy in Blantyre, Malawi. Whole genome scans (650,000 SNPs genotyped using Illumina genotyping assays) were obtained for each infant. Logistic regression was used to evaluate the association between each SNP and HIV MTCT. RESULTS Genotype results were available for 100 HIV(+) infants (at birth, 6, or 12 weeks) and 126 HIV(-) infants (at birth, 6, and 12 weeks). We identified 9 SNPs within 6 genes with a P-value < 5 × 10(-5) associated with the risk of transmission, in either unadjusted or adjusted by maternal HIV viral load analyses. Carriers of the rs8069770 variant allele were associated with a lower risk of HIV MTCT (odds ratio = 0.27, 95% confidence interval = 0.14, 0.51), where rs8069770 is located within HS3ST3A1, a gene involved in heparan sulfate biosynthesis. Interesting associations for SNPs located within or near genes involved in pregnancy and development, innate immunological response, or HIV protein interactions were also observed. CONCLUSIONS This study used a genome-wide approach to identify novel variants associated with the risk of HIV MTCT in order to gain new insights into HIV MTCT etiology. Replication of this work using a larger sample size will help us to differentiate true positive findings.
Collapse
Affiliation(s)
- Bonnie R Joubert
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Ethan M Lange
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA ; Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA ; Carolina Center for Genome Sciences, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Victor Mwapasa
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA ; Carolina Center for Genome Sciences, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Steven R Meshnick
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | | |
Collapse
|
38
|
Saïdi H, Carbonneil C, Magri G, Eslahpazir J, Sekaly RP, Bélec L. Differential modulation of CCR5-tropic human immunodeficiency virus–1 transfer from macrophages towards T cells under interleukin-4/interleukin-13 microenvironment. Hum Immunol 2010; 71:1-13. [DOI: 10.1016/j.humimm.2009.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2008] [Revised: 08/13/2009] [Accepted: 08/18/2009] [Indexed: 12/16/2022]
|
39
|
Abstract
Dendritic cell-specific ICAM-3-grabbing nonintegrin (DC-SIGN). DC-SIGN is a C-type lectin receptor that recognizes N-linked high-mannose oligosaccharides and branched fucosylated structures. It is now clear that the biological role of DC-SIGN is two-fold. It is primarily expressed by dendritic cells and mediates important functions necessary for the induction of successful immune responses that are essential for the clearance of microbial infections, such as the capture, destruction, and presentation of microbial pathogens to induce successful immune responses. Yet, on the other hand, pathogens may also exploit DC-SIGN to modulate DC functioning thereby skewing the immune response and promoting their own survival. This chapter presents an overview of the structure of DC-SIGN and its expression pattern among immune cells. The current state of knowledge of DC-SIGN-carbohydrate interactions is discussed and how these interactions influence dendritic cell functioning is examined. The molecular aspects that underlie the selectivity of DC-SIGN for mannose-and fucose-containing carbohydrates are detailed. Furthermore, the chapter discusses the role of DC-SIGN in dendritic cell biology and how certain bacterial pathogens exploit DC-SIGN to escape immune surveillance.
Collapse
|
40
|
Sierra-Filardi E, Estecha A, Samaniego R, Fernández-Ruiz E, Colmenares M, Sánchez-Mateos P, Steinman RM, Granelli-Piperno A, Corbí AL. Epitope mapping on the dendritic cell-specific ICAM-3-grabbing non-integrin (DC-SIGN) pathogen-attachment factor. Mol Immunol 2010; 47:840-8. [DOI: 10.1016/j.molimm.2009.09.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2009] [Revised: 09/20/2009] [Accepted: 09/30/2009] [Indexed: 12/26/2022]
|
41
|
Jain P, Manuel SL, Khan ZK, Ahuja J, Quann K, Wigdahl B. DC-SIGN mediates cell-free infection and transmission of human T-cell lymphotropic virus type 1 by dendritic cells. J Virol 2009; 83:10908-21. [PMID: 19692463 PMCID: PMC2772783 DOI: 10.1128/jvi.01054-09] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2009] [Accepted: 08/12/2009] [Indexed: 12/20/2022] Open
Abstract
Despite the susceptibility of dendritic cells (DCs) to human T-cell lymphotropic virus type 1 (HTLV-1) infection and the defined role of these cells in disease pathogenesis, the mechanisms of viral binding to DCs have not been fully delineated. Recently, a glucose transporter, GLUT-1, heparan sulfate proteoglycans (HSPGs), and neuropilin-1 (NRP-1) were demonstrated to facilitate HTLV-1 entry into T cells. DCs express their own array of antigen receptors, the most important being the DC-specific intercellular adhesion molecule-3 (ICAM-3)-grabbing nonintegrin (DC-SIGN) with respect to retrovirus binding. Consequently, the role of DC-SIGN and other HTLV-1 attachment factors was analyzed in viral binding, transmission, and productive infection using monocyte-derived DCs (MDDCs), blood myeloid DCs, and B-cell lines expressing DC-SIGN. The relative expression of DC-SIGN, GLUT-1, HSPGs, and NRP-1 first was examined on both DCs and B-cell lines. Although the inhibition of these molecules reduced viral binding, HTLV-1 transmission from DCs to T cells was mediated primarily by DC-SIGN. DC-SIGN also was shown to play a role in the infection of MDDCs as well as model B-cell lines. The HTLV-1 infection of MDDCs also was achieved in blood myeloid DCs following the enhancement of virus-induced interleukin-4 production and subsequent DC-SIGN expression in this cell population. This study represents the first comprehensive analysis of potential HTLV-1 receptors on DCs and strongly suggests that DC-SIGN plays a critical role in HTLV-1 binding, transmission, and infection, thereby providing an attractive target for the development of antiretroviral therapeutics and microbicides.
Collapse
Affiliation(s)
- Pooja Jain
- Department of Microbiology and Immunology, Center for Molecular Virology and Neuroimmunology, Center for Cancer Biology, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Sharrón L. Manuel
- Department of Microbiology and Immunology, Center for Molecular Virology and Neuroimmunology, Center for Cancer Biology, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Zafar K. Khan
- Department of Microbiology and Immunology, Center for Molecular Virology and Neuroimmunology, Center for Cancer Biology, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Jaya Ahuja
- Department of Microbiology and Immunology, Center for Molecular Virology and Neuroimmunology, Center for Cancer Biology, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Kevin Quann
- Department of Microbiology and Immunology, Center for Molecular Virology and Neuroimmunology, Center for Cancer Biology, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Brian Wigdahl
- Department of Microbiology and Immunology, Center for Molecular Virology and Neuroimmunology, Center for Cancer Biology, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| |
Collapse
|
42
|
Renaud SJ, Graham CH. The Role of Macrophages in Utero-placental Interactions During Normal and Pathological Pregnancy. Immunol Invest 2009; 37:535-64. [DOI: 10.1080/08820130802191375] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
43
|
Huang Y, Dryman B, Li W, Meng X. Porcine DC-SIGN: molecular cloning, gene structure, tissue distribution and binding characteristics. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2009; 33:464-480. [PMID: 18951915 PMCID: PMC7103218 DOI: 10.1016/j.dci.2008.09.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2008] [Revised: 09/14/2008] [Accepted: 09/16/2008] [Indexed: 05/27/2023]
Abstract
DC-SIGN, a human C-type lectin, is involved in the transmission of many enveloped viruses. Here we report the cloning and characterization of the cDNA and gene encoding porcine DC-SIGN (pDC-SIGN). The full-length pDC-SIGN cDNA encodes a type II transmembrane protein of 240 amino acids. Phylogenetic analysis revealed that pDC-SIGN, together with bovine, canis and equine DC-SIGN, are more closely related to mouse SIGNR7 and SIGNR8 than to human DC-SIGN. pDC-SIGN has the same gene structure as bovine, canis DC-SIGN and mouse SIGNR8 with eight exons. pDC-SIGN mRNA expression was detected in pig spleen, thymus, lymph node, lung, bone marrow and muscles. pDC-SIGN protein was found to express on the surface of monocyte-derived macrophages and dendritic cells, alveolar macrophages, lymph node sinusoidal macrophage-like, dendritic-like and endothelial cells but not of monocytes, peripheral blood lymphocytes or lymph node lymphocytes. A BHK cell line stably expressing pDC-SIGN binds to human ICAM-3 and ICAM-2 immunoadhesins in a calcium-dependent manner, and enhances the transmission of porcine reproductive and respiratory syndrome virus (PRRSV) to target cells in trans. The results will help better understand the biological role(s) of DC-SIGN family in innate immunity during the evolutionary process.
Collapse
Affiliation(s)
| | | | | | - X.J. Meng
- Corresponding author. Tel.: +1 540 231 6912; fax: +1 540 231 3426.
| |
Collapse
|
44
|
Sun P, Fernandez S, Marovich MA, Palmer DR, Celluzzi CM, Boonnak K, Liang Z, Subramanian H, Porter KR, Sun W, Burgess TH. Functional characterization of ex vivo blood myeloid and plasmacytoid dendritic cells after infection with dengue virus. Virology 2008; 383:207-15. [PMID: 19013627 DOI: 10.1016/j.virol.2008.10.022] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Revised: 09/17/2008] [Accepted: 10/10/2008] [Indexed: 10/21/2022]
Abstract
Myeloid and plasmacytoid dendritic cells (mDC and pDC) are naturally distinctive subsets. We exposed both subsets to dengue virus (DV) in vitro and investigated their functional characteristics. High levels of DV replication in mDC were found to correlate with DC-SIGN expression. Production of inflammatory cytokines by mDC increased gradually after DV-infection, which was dependent on DV replication. Co-stimulatory markers were upregulated on mDC upon DV-infection. On the contrary, lower levels of DV-replication were observed in pDC, but the cytokine production in pDC was quicker and stronger. This cytokine response was not dependent on viral replication, but dependent on cell endosomal activity and TLR7, and could be also induced by purified DV genome RNA. These results clearly suggested functional differences between mDC and pDC in response to DV infection. Additionally, the TLR7-mediated recognition of DV RNA may be involved in pDC functional activation.
Collapse
Affiliation(s)
- Peifang Sun
- Naval Medical Research Center, Silver Spring, MD 20910, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Chaudhary O, Rajsekar K, Ahmed I, Verma R, Bala M, Bhasin R, Luthra K. Polymorphic variants in DC-SIGN, DC-SIGNR and SDF-1 in high risk seronegative and HIV-1 patients in Northern Asian Indians. J Clin Virol 2008; 43:196-201. [PMID: 18775666 DOI: 10.1016/j.jcv.2008.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2008] [Revised: 06/05/2008] [Accepted: 06/06/2008] [Indexed: 11/16/2022]
Abstract
A single nucleotide polymorphism (SNP) in SDF-1, the natural ligand for the HIV-1 coreceptor CXCR4, is implicated to have protective effects against HIV-1 infection. Dendritic cells are the first to encounter HIV-1 at mucosal sites and virus binding occurs via receptors known as DC-SIGN. Variations in the number of repeats in the neck region of DC-SIGN and DC-SIGNR are reported to possibly influence host susceptibility to HIV-1 infection. We examined the SNP of SDF1-3'A by PCR-restriction fragment length polymorphism (RFLP) and repeat region polymorphisms in DC-SIGN and DC SIGNR by PCR in healthy HIV seronegative individuals, high risk STD patients seronegative for HIV, and HIV-1 seropositive patients from northern India. The detected polymorphisms were confirmed by cloning and sequencing. The genotypic frequency of SDF1-3'A/SDF1-3'A in the 100 HIV-seronegative healthy individuals, 150 HIV seronegative STD patients, and 100 HIV-1 seropositive patients were 4%, 18% and 7%, respectively. A significantly higher frequency of SDF1-3'A/SDF1-3'A was observed in high risk STD patients as compared to HIV seropositive (p=0.014) and healthy HIV-1 seronegative tested individuals (p=0.001), suggesting a protective role of SDF1-3'A in HIV-1 infection. DC-SIGN polymorphism was rare and genotype 7/7 was predominant in all groups studied. DC-SIGNR was highly polymorphic and 11 genotypes were observed among the different study groups. The precise role of the polymorphic variants of DC-SIGNR needs to be elucidated in the population.
Collapse
Affiliation(s)
- Omkar Chaudhary
- Department of Biochemistry, Room No. 3002, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India.
| | | | | | | | | | | | | |
Collapse
|
46
|
Khoo US, Chan KYK, Chan VSF, Lin CLS. DC-SIGN and L-SIGN: the SIGNs for infection. J Mol Med (Berl) 2008; 86:861-74. [PMID: 18458800 PMCID: PMC7079906 DOI: 10.1007/s00109-008-0350-2] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2007] [Revised: 03/01/2008] [Accepted: 03/05/2008] [Indexed: 12/16/2022]
Abstract
Two closely related trans-membrane C-type lectins dendritic cell-specific intracellular adhesion molecules (ICAM)-3 grabbing non-integrin (DC-SIGN or CD209) and liver/lymph node-specific ICAM-3 grabbing non-integrin (L-SIGN also known as DC-SIGNR, CD209L or CLEC4M) directly recognize a wide range of micro-organisms of major impact on public health. Both genes have long been considered to share similar overall structure and ligand-binding characteristics. This review presents more recent biochemical and structural studies, which show that they have distinct ligand-binding properties and different physiological functions. Of importance in both these genes is the presence of an extra-cellular domain consisting of an extended neck region encoded by tandem repeats that support the carbohydrate-recognition domain, which plays a crucial role in influencing the pathogen-binding properties of these receptors. The notable difference between these two genes is in this extra-cellular domain. Whilst the tandem-neck-repeat region remains relatively constant size for DC-SIGN, there is considerable polymorphism for L-SIGN. Homo-oligomerization of the neck region of L-SIGN has been shown to be important for high-affinity ligand binding, and heterozygous expression of the polymorphic variants of L-SIGN in which neck lengths differ could thus affect ligand-binding affinity. Functional studies on the effect of this tandem-neck-repeat region on pathogen-binding, as well as genetic association studies for various infectious diseases and among different populations, are discussed. Worldwide demographic data of the tandem-neck-repeat region showing distinct differences in the neck-region allele and genotype distribution among different ethnic groups are presented. These findings support the neck region as an excellent candidate acting as a functional target for selective pressures exerted by pathogens.
Collapse
Affiliation(s)
- Ui-Soon Khoo
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, University Pathology Building, Hong Kong, SAR, China.
| | | | | | | |
Collapse
|
47
|
Yearley JH, Kanagy S, Anderson DC, Dalecki K, Pauley DR, Suwyn C, Donahoe RM, McClure HM, O'Neil SP. Tissue-specific reduction in DC-SIGN expression correlates with progression of pathogenic simian immunodeficiency virus infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2008; 32:1510-1521. [PMID: 18606180 DOI: 10.1016/j.dci.2008.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Revised: 05/21/2008] [Accepted: 06/06/2008] [Indexed: 05/26/2023]
Abstract
Studies were undertaken to determine whether previously described reductions in splenic DC-SIGN expression in simian acquired immune deficiency syndrome (AIDS) are limited to pathogenic simian immunodeficiency virus (SIV) infection. DC-SIGN expression was evaluated by immunohistochemistry in lymphoid tissues from AIDS-susceptible Asian macaque monkeys as compared with AIDS-resistant sooty mangabey monkeys in the presence and absence of SIV infection. The phenotype of DC-SIGN+ cells in susceptible and resistant species was identical and most consistent with macrophage identity. Significantly lower levels of DC-SIGN expression were identified in spleen, mesenteric lymph node, and bone marrow of macaques with AIDS (P<0.05). Reduced levels of splenic DC-SIGN correlated significantly with CD4T cell depletion in long-term pathogenic infection of macaques (P<0.01), whereas SIV-infected mangabeys retained high levels of DC-SIGN expression in spleen despite persistent infection. Reduced expression of DC-SIGN in spleen specifically characterizes pathogenic forms of SIV infection, correlates with disease progression, and may contribute to SIV pathogenesis.
Collapse
Affiliation(s)
- Jennifer H Yearley
- Division of Comparative Pathology, New England Primate Research Center, Harvard Medical School, One Pine Hill Dr. P.O. Box 9102, Southborough, MA 01772, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Irache JM, Salman HH, Gamazo C, Espuelas S. Mannose-targeted systems for the delivery of therapeutics. Expert Opin Drug Deliv 2008; 5:703-24. [DOI: 10.1517/17425247.5.6.703] [Citation(s) in RCA: 234] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Troth SP, Dean AD, Hoover EA. In vivo CXCR4 expression, lymphoid cell phenotype, and feline immunodeficiency virus infection. Vet Immunol Immunopathol 2008; 123:97-105. [PMID: 18295345 PMCID: PMC2423945 DOI: 10.1016/j.vetimm.2008.01.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Primary isolates of feline immunodeficiency virus (FIV) appear to require binding to CD134 in conjunction with CXCR4(X4) to infect IL-2-dependent T-cell-derived cells in culture. However, much less is known about the role of X4 for the infection of cells in vivo. To investigate the correlation between X4 expression and FIV infection in cats acutely infected with FIV-C-Pgmr we used high-speed fluorescence-activated cell sorting and realtime PCR to co-analyze cell phenotypes from lymph node, thymus, bone marrow and blood for FIV infection and X4 expression. X4 expression was greatest in lymph node, both in frequency and in mean fluorescence intensity. The thymus demonstrated a higher proviral burden in X4+ thymic T cells ( approximately 14% in X4+ thymic T cells and 7% in X4- cells) whereas, proviral loads were similar between X4+ and X4- cell populations in all other tissues examined. Assuming a minimum of one proviral copy per cell, a maximum of approximately 50% of FIV-positive cells were X4+. The highest fraction of FIV-infected X4- cells was present in bone marrow. Regardless of X4 status, proviral loads were higher in lymph node and blood T cells than in B cells. These studies provide both a positive association between X4 expression and FIV infection and introduce the probability that X4-independent infection occurs in other target cells in vivo.
Collapse
Affiliation(s)
- Sean P. Troth
- Department of Microbiology Immunology and Pathology, Colorado State University
| | | | - Edward A. Hoover
- Department of Microbiology Immunology and Pathology, Colorado State University
| |
Collapse
|
50
|
Gramberg T, Soilleux E, Fisch T, Lalor PF, Hofmann H, Wheeldon S, Cotterill A, Wegele A, Winkler T, Adams DH, Pöhlmann S. Interactions of LSECtin and DC-SIGN/DC-SIGNR with viral ligands: Differential pH dependence, internalization and virion binding. Virology 2008; 373:189-201. [PMID: 18083206 PMCID: PMC7103327 DOI: 10.1016/j.virol.2007.11.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Revised: 10/15/2007] [Accepted: 11/02/2007] [Indexed: 01/20/2023]
Abstract
The calcium-dependent lectins DC-SIGN and DC-SIGNR (collectively termed DC-SIGN/R) bind to high-mannose carbohydrates on a variety of viruses. In contrast, the related lectin LSECtin does not recognize mannose-rich glycans and interacts with a more restricted spectrum of viruses. Here, we analyzed whether these lectins differ in their mode of ligand engagement. LSECtin and DC-SIGNR, which we found to be co-expressed by liver, lymph node and bone marrow sinusoidal endothelial cells, bound to soluble Ebola virus glycoprotein (EBOV-GP) with comparable affinities. Similarly, LSECtin, DC-SIGN and the Langerhans cell-specific lectin Langerin readily bound to soluble human immunodeficiency virus type-1 (HIV-1) GP. However, only DC-SIGN captured HIV-1 particles, indicating that binding to soluble GP is not necessarily predictive of binding to virion-associated GP. Capture of EBOV-GP by LSECtin triggered ligand internalization, suggesting that LSECtin like DC-SIGN might function as an antigen uptake receptor. However, the intracellular fate of lectin-ligand complexes might differ. Thus, exposure to low-pH medium, which mimics the acidic luminal environment in endosomes/lysosomes, released ligand bound to DC-SIGN/R but had no effect on LSECtin interactions with ligand. Our results reveal important differences between pathogen capture by DC-SIGN/R and LSECtin and hint towards different biological functions of these lectins.
Collapse
Affiliation(s)
- Thomas Gramberg
- Institute of Virology, University Hospital Erlangen, 91054 Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|