1
|
Chitta S, Nehete BP, Delise AB, Simmons JH, Nehete PN. Reactivity of HLADR antibody manifests expression of surface MHC II molecules on peripheral blood T lymphocytes in new world monkeys. Immun Inflamm Dis 2024; 12:e1318. [PMID: 38923761 PMCID: PMC11194976 DOI: 10.1002/iid3.1318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/20/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Major histocompatibility complex (MHC) class II molecules expressed on B cells, monocytes and dendritic cells present processed peptides to CD4+ T cells as one of the mechanisms to combat infection and inflammation. AIM To study MHC II expression in a variety of nonhuman primate species, including New World (NWM) squirrel monkeys (Saimiri boliviensis boliviensis), owl monkeys (Aotus nancymae), common marmosets (Callithrix spp.), and Old World (OWM) rhesus (Macaca mulatta), baboons (Papio anubis). METHODS Two clones of cross-reactive mouse anti-human HLADR monoclonal antibodies (mAb) binding were analyzed by flow cytometry to evaluate MHC II expression on NHP immune cells, including T lymphocytes in whole blood (WB) and peripheral blood mononuclear cells (PBMC). RESULTS MHC class II antibody reactivity is seen with CD20+ B cells, CD14+ monocytes and CD3+ T lymphocytes. Specific reactivity with both clones was demonstrated in T lymphocytes: this reactivity was not inhibited by purified CD16 antibody but was completely inhibited when pre-blocked with purified unconjugated MHC II antibody. Freshly prepared PBMC also showed reactivity with T lymphocytes without any stimulation. Interestingly, peripheral blood from rhesus macaques and olive baboons (OWM) showed no such T lymphocyte associated MHCII antibody reactivity. DISCUSSION & CONCLUSION Our results from antibody (MHC II) reactivity clearly show the potential existence of constitutively expressed (with no stimulation) MHC II molecules on T lymphocytes in new world monkeys. These results suggest that additional study is warranted to evaluate the functional and evolutionary significance of these finding and to better understand MHC II expression on T lymphocytes in new world monkeys.
Collapse
Affiliation(s)
- Sriram Chitta
- Department of Comparative Medicine, Keeling Center for Comparative Medicine and ResearchUT‐MD Anderson Cancer CenterBastropTexasUSA
| | - Bharti P. Nehete
- Department of Comparative Medicine, Keeling Center for Comparative Medicine and ResearchUT‐MD Anderson Cancer CenterBastropTexasUSA
| | - Ashley B. Delise
- Department of Comparative Medicine, Keeling Center for Comparative Medicine and ResearchUT‐MD Anderson Cancer CenterBastropTexasUSA
| | - Joe H. Simmons
- Department of Comparative Medicine, Keeling Center for Comparative Medicine and ResearchUT‐MD Anderson Cancer CenterBastropTexasUSA
| | - Pramod N. Nehete
- Department of Comparative Medicine, Keeling Center for Comparative Medicine and ResearchUT‐MD Anderson Cancer CenterBastropTexasUSA
- Department of Comparative MedicineThe University of Texas Graduate School of Biomedical SciencesHoustonTexasUSA
| |
Collapse
|
2
|
Layton ED, Barman S, Wilburn DB, Yu KKQ, Smith MT, Altman JD, Scriba TJ, Tahiri N, Minnaard AJ, Roederer M, Seder RA, Darrah PA, Seshadri C. T Cells Specific for a Mycobacterial Glycolipid Expand after Intravenous Bacillus Calmette-Guérin Vaccination. THE JOURNAL OF IMMUNOLOGY 2021; 206:1240-1250. [PMID: 33536255 DOI: 10.4049/jimmunol.2001065] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/01/2021] [Indexed: 12/12/2022]
Abstract
Intradermal vaccination with Mycobacterium bovis bacillus Calmette-Guérin (BCG) protects infants from disseminated tuberculosis, and i.v. BCG protects nonhuman primates (NHP) against pulmonary and extrapulmonary tuberculosis. In humans and NHP, protection is thought to be mediated by T cells, which typically recognize bacterial peptide Ags bound to MHC proteins. However, during vertebrate evolution, T cells acquired the capacity to recognize lipid Ags bound to CD1a, CD1b, and CD1c proteins expressed on APCs. It is unknown whether BCG induces T cell immunity to mycobacterial lipids and whether CD1-restricted T cells are resident in the lung. In this study, we developed and validated Macaca mulatta (Mamu) CD1b and CD1c tetramers to probe ex vivo phenotypes and functions of T cells specific for glucose monomycolate (GMM), an immunodominant mycobacterial lipid Ag. We discovered that CD1b and CD1c present GMM to T cells in both humans and NHP. We show that GMM-specific T cells are expanded in rhesus macaque blood 4 wk after i.v. BCG, which has been shown to protect NHP with near-sterilizing efficacy upon M. tuberculosis challenge. After vaccination, these T cells are detected at high frequency within bronchoalveolar fluid and express CD69 and CD103, markers associated with resident memory T cells. Thus, our data expand the repertoire of T cells known to be induced by whole cell mycobacterial vaccines, such as BCG, and show that lipid Ag-specific T cells are resident in the lungs, where they may contribute to protective immunity.
Collapse
Affiliation(s)
- Erik D Layton
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109
| | - Soumik Barman
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109
| | - Damien B Wilburn
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195
| | - Krystle K Q Yu
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109
| | - Malisa T Smith
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109
| | - John D Altman
- National Institutes of Health Tetramer Core Facility, Emory University, Atlanta, GA 30329
| | - Thomas J Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 9747, South Africa
| | - Nabil Tahiri
- Stratingh Institute for Chemistry, University of Groningen 7925, Groningen, the Netherlands
| | - Adriaan J Minnaard
- Stratingh Institute for Chemistry, University of Groningen 7925, Groningen, the Netherlands
| | - Mario Roederer
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892; and
| | - Robert A Seder
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892; and
| | - Patricia A Darrah
- Vaccine Research Center, National Institutes of Health, Bethesda, MD 20892; and
| | - Chetan Seshadri
- Department of Medicine, University of Washington School of Medicine, Seattle, WA 98109; .,Tuberculosis Research and Training Center, University of Washington, Seattle, WA 98109
| |
Collapse
|
3
|
Crowley AR, Ackerman ME. Mind the Gap: How Interspecies Variability in IgG and Its Receptors May Complicate Comparisons of Human and Non-human Primate Effector Function. Front Immunol 2019; 10:697. [PMID: 31024542 PMCID: PMC6463756 DOI: 10.3389/fimmu.2019.00697] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 03/13/2019] [Indexed: 01/08/2023] Open
Abstract
The field of HIV research relies heavily on non-human primates, particularly the members of the macaque genus, as models for the evaluation of candidate vaccines and monoclonal antibodies. A growing body of research suggests that successful protection of humans will not solely rely on the neutralization activity of an antibody's antigen binding fragment. Rather, immunological effector functions prompted by the interaction of the immunoglobulin G constant region and its cognate Fc receptors help contribute to favorable outcomes. Inherent differences in the sequences, expression, and activities of human and non-human primate antibody receptors and immunoglobulins have the potential to produce disparate results in the observations made in studies conducted in differing species. Having a more complete understanding of these differences, however, should permit the more fluent translation of observations between model organisms and the clinic. Here we present a guide to such translations that encompasses not only what is presently known regarding the affinity of the receptor-ligand interactions but also the influence of expression patterns and allelic variation, with a focus on insights gained from use of this model in HIV vaccines and passive antibody therapy and treatment.
Collapse
Affiliation(s)
- Andrew R. Crowley
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH, United States
| | - Margaret E. Ackerman
- Molecular and Cellular Biology Program, Dartmouth College, Hanover, NH, United States
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| |
Collapse
|
4
|
Boesch AW, Miles AR, Chan YN, Osei-Owusu NY, Ackerman ME. IgG Fc variant cross-reactivity between human and rhesus macaque FcγRs. MAbs 2017; 9:455-465. [PMID: 28055295 DOI: 10.1080/19420862.2016.1274845] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Non-human primate (NHP) studies are often an essential component of antibody development efforts before human trials. Because the efficacy or toxicity of candidate antibodies may depend on their interactions with Fcγ receptors (FcγR) and their resulting ability to induce FcγR-mediated effector functions such as antibody-dependent cell-meditated cytotoxicity and phagocytosis (ADCP), the evaluation of human IgG variants with modulated affinity toward human FcγR is becoming more prevalent in both infectious disease and oncology studies in NHP. Reliable translation of these results necessitates analysis of the cross-reactivity of these human Fc variants with NHP FcγR. We report evaluation of the binding affinities of a panel of human IgG subclasses, Fc amino acid point mutants and Fc glycosylation variants against the common allotypes of human and rhesus macaque FcγR by applying a high-throughput array-based surface plasmon resonance platform. The resulting data indicate that amino acid variation present in rhesus FcγRs can result in disrupted, matched, or even increased affinity of IgG Fc variants compared with human FcγR orthologs. These observations emphasize the importance of evaluating species cross-reactivity and developing an understanding of the potential limitations or suitability of representative in vitro and in vivo models before human clinical studies when either efficacy or toxicity may be associated with FcγR engagement.
Collapse
Affiliation(s)
- Austin W Boesch
- a Thayer School of Engineering, Dartmouth College , Hanover , NH , USA
| | - Adam R Miles
- b Wasatch Microfluidics , Salt Lake City , UT , USA
| | - Ying N Chan
- a Thayer School of Engineering, Dartmouth College , Hanover , NH , USA
| | - Nana Y Osei-Owusu
- c Department of Microbiology and Immunology , Geisel School of Medicine , Lebanon , NH , USA
| | - Margaret E Ackerman
- a Thayer School of Engineering, Dartmouth College , Hanover , NH , USA.,c Department of Microbiology and Immunology , Geisel School of Medicine , Lebanon , NH , USA
| |
Collapse
|
5
|
Abstract
ABSTRACT
The aim of this review is to provide a coherent framework for understanding dendritic cells (DCs). It has seven sections. The introduction provides an overview of the immune system and essential concepts, particularly for the nonspecialist reader. Next, the “History” section outlines the early evolution of ideas about DCs and highlights some sources of confusion that still exist today. The “Lineages” section then focuses on five different populations of DCs: two subsets of “classical” DCs, plasmacytoid DCs, monocyte-derived DCs, and Langerhans cells. It highlights some cellular and molecular specializations of each, and also notes other DC subsets that have been proposed. The following “Tissues” section discusses the distribution and behavior of different DC subsets within nonlymphoid and secondary lymphoid tissues that are connected by DC migration pathways between them. In the “Tolerance” section, the role of DCs in central and peripheral tolerance is considered, including their ability to drive the differentiation of different populations of regulatory T cells. In contrast, the “Immunity” section considers the roles of DCs in sensing of infection and tissue damage, the initiation of primary responses, the T-cell effector phase, and the induction of immunological memory. The concluding section provides some speculative ideas about the evolution of DCs. It also revisits earlier concepts of generation of diversity and clonal selection in terms of DCs driving the evolution of T-cell responses. Throughout, this review highlights certain areas of uncertainty and suggests some avenues for future investigation.
Collapse
|
6
|
Lahaye X, Satoh T, Gentili M, Cerboni S, Silvin A, Conrad C, Ahmed-Belkacem A, Rodriguez EC, Guichou JF, Bosquet N, Piel M, Le Grand R, King MC, Pawlotsky JM, Manel N. Nuclear Envelope Protein SUN2 Promotes Cyclophilin-A-Dependent Steps of HIV Replication. Cell Rep 2016; 15:879-892. [PMID: 27149839 PMCID: PMC4850421 DOI: 10.1016/j.celrep.2016.03.074] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 02/12/2016] [Accepted: 03/20/2016] [Indexed: 12/01/2022] Open
Abstract
During the early phase of replication, HIV reverse transcribes its RNA and crosses the nuclear envelope while escaping host antiviral defenses. The host factor Cyclophilin A (CypA) is essential for these steps and binds the HIV capsid; however, the mechanism underlying this effect remains elusive. Here, we identify related capsid mutants in HIV-1, HIV-2, and SIVmac that are restricted by CypA. This antiviral restriction of mutated viruses is conserved across species and prevents nuclear import of the viral cDNA. Importantly, the inner nuclear envelope protein SUN2 is required for the antiviral activity of CypA. We show that wild-type HIV exploits SUN2 in primary CD4+ T cells as an essential host factor that is required for the positive effects of CypA on reverse transcription and infection. Altogether, these results establish essential CypA-dependent functions of SUN2 in HIV infection at the nuclear envelope. HIV capsid mutants reveal that Cyclophilin A can restrict viral nuclear import Nuclear envelope protein SUN2 is implicated in the restriction of HIV mutants SUN2 is essential for wild-type HIV infection in CD4+ T cells and dendritic cells In CD4+ T cells, the activities of CypA on HIV-1 infection require SUN2
Collapse
Affiliation(s)
- Xavier Lahaye
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France
| | - Takeshi Satoh
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France
| | - Matteo Gentili
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France
| | - Silvia Cerboni
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France
| | - Aymeric Silvin
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France
| | - Cécile Conrad
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France
| | | | - Elisa C Rodriguez
- Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520-8002, USA
| | - Jean-François Guichou
- CNRS UMR5048, INSERM U1054 - Université de Montpellier, Centre de Biochimie Structurale, 34090 Montpellier, France
| | - Nathalie Bosquet
- Université Paris Sud, INSERM, CEA, DRF-Immunology of Viral Infections and Autoimmune Diseases department (IMVA), UMR1184, IDMIT Infrastructure, iMETI, 92260 Fontenay-aux-Roses, France
| | - Matthieu Piel
- Subcellular Structure and Cellular Dynamics Department, Institut Curie, PSL Research University, CNRS UMR144, 75005 Paris, France; Institut Pierre-Gilles de Gennes, PSL Research University, 75005 Paris, France
| | - Roger Le Grand
- Université Paris Sud, INSERM, CEA, DRF-Immunology of Viral Infections and Autoimmune Diseases department (IMVA), UMR1184, IDMIT Infrastructure, iMETI, 92260 Fontenay-aux-Roses, France
| | - Megan C King
- Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520-8002, USA
| | - Jean-Michel Pawlotsky
- INSERM U955, 94010 Créteil, France; Department of Virology, National Reference Center for Viral Hepatitis B, C and D, Hôpital Henri Mondor, Université Paris-Est, 94010 Créteil, France
| | - Nicolas Manel
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France.
| |
Collapse
|
7
|
Moreau A, Varey E, Bériou G, Hill M, Bouchet-Delbos L, Segovia M, Cuturi MC. Tolerogenic dendritic cells and negative vaccination in transplantation: from rodents to clinical trials. Front Immunol 2012; 3:218. [PMID: 22908013 PMCID: PMC3414843 DOI: 10.3389/fimmu.2012.00218] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2012] [Accepted: 07/06/2012] [Indexed: 12/12/2022] Open
Abstract
The use of immunosuppressive (IS) drugs to treat transplant recipients has markedly reduced the incidence of acute rejection and early graft loss. However, such treatments have numerous adverse side effects and fail to prevent chronic allograft dysfunction. In this context, therapies based on the adoptive transfer of regulatory cells are promising strategies to induce indefinite transplant survival. The use of tolerogenic dendritic cells (DC) has shown great potential, as preliminary experiments in rodents have demonstrated that administration of tolerogenic DC prolongs graft survival. Recipient DC, Donor DC, or Donor Ag-pulsed recipient DC have been used in preclinical studies and administration of these cells with suboptimal immunosuppression increases their tolerogenic potential. We have demonstrated that autologous unpulsed tolerogenic DC injected in the presence of suboptimal immunosuppression are able to induce Ag-specific allograft tolerance. We derived similar tolerogenic DC in different animal models (mice and non-human primates) and confirmed their protective abilities in vitro and in vivo. The mechanisms involved in the tolerance induced by autologous tolerogenic DC were also investigated. With the aim of using autologous DC in kidney transplant patients, we have developed and characterized tolerogenic monocyte-derived DC in humans. In this review, we will discuss the preclinical studies and describe our recent results from the generation and characterization of tolerogenic monocyte-derived DC in humans for a clinical application. We will also discuss the limits and difficulties in translating preclinical experiments to theclinic.
Collapse
|
8
|
Romain G, van Gulck E, Epaulard O, Oh S, Li D, Zurawski G, Zurawski S, Cosma A, Adam L, Chapon C, Todorova B, Banchereau J, Dereuddre-Bosquet N, Vanham G, Le Grand R, Martinon F. CD34-derived dendritic cells transfected ex vivo with HIV-Gag mRNA induce polyfunctional T-cell responses in nonhuman primates. Eur J Immunol 2012; 42:2019-30. [PMID: 22585548 DOI: 10.1002/eji.201242478] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 04/07/2012] [Accepted: 04/30/2012] [Indexed: 12/23/2022]
Abstract
The pivotal role of DCs in initiating immune responses led to their use as vaccine vectors. However, the relationship between DC subsets involved in antigen presentation and the type of elicited immune responses underlined the need for the characterization of the DCs generated in vitro. The phenotypes of tissue-derived APCs from a cynomolgus macaque model for human vaccine development were compared with ex vivo-derived DCs. Monocyte/macrophages predominated in bone marrow (BM) and blood. Myeloid DCs (mDCs) were present in all tested tissues and were more highly represented than plasmacytoid DCs (pDCs). As in human skin, Langerhans cells (LCs) resided exclusively in the macaque epidermis, expressing CD11c, high levels of CD1a and langerin (CD207). Most DC subsets were endowed with tissue-specific combinations of PRRs. DCs generated from CD34(+) BM cells (CD34-DCs) were heterogeneous in phenotype. CD34-DCs shared properties (differentiation and PRR) of dermal and epidermal DCs. After injection into macaques, CD34-DCs expressing HIV-Gag induced Gag-specific CD4(+) and CD8(+) T cells producing IFN-γ, TNF-α, MIP-1β, or IL-2. In high responding animals, the numbers of polyfunctional CD8(+) T cells increased with the number of booster injections. This DC-based vaccine strategy elicited immune responses relevant to the DC subsets generated in vitro.
Collapse
Affiliation(s)
- Gabrielle Romain
- CEA, Division of Immuno-Virology, Institute for Emerging Diseases and Innovative Therapies, DSV, Fontenay-aux-Roses, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
In vitro assessment of the feline cell-mediated immune response against feline panleukopeniavirus, calicivirus and felid herpesvirus 1 using 5-bromo-2'-deoxyuridine labeling. Vet Immunol Immunopathol 2012; 146:177-84. [PMID: 22460172 PMCID: PMC7112514 DOI: 10.1016/j.vetimm.2012.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 02/16/2012] [Accepted: 03/05/2012] [Indexed: 02/06/2023]
Abstract
In this study an in vitro assay was optimized to detect feline proliferating lymphocytes as an assessment for the cell-mediated immune response. For this purpose, 5-bromo-2′-deoxyuridine (BrdU) labeling was chosen because of its sensitivity and the possibility of further characterization of proliferating cells. The assay was optimized by selecting the best batch and concentration of fetal bovine serum, β-mercaptoethanol concentration, cell density, BrdU incubation time and antigen presenting cell type. Cats were vaccinated with the attenuated Nobivac vaccine Tricat and the peripheral blood lymphocyte proliferation responses were quantified upon in vitro restimulation with inactivated and infectious feline panleukopenia virus (FPV), feline calicivirus (FCV) and felid herpesvirus 1 (FeHV-1). Proliferation signals were detected with inactivated FeHV-1 in the CD8+ but not in the CD8− T lymphocyte population, with inactivated FCV and FPV in both CD8− and CD8+ T lymphocyte populations. Restimulation with infectious FCV caused significant proliferation in the CD8− T lymphocyte population only while infectious FPV and FeHV-1 seemed to suppress lymphocyte proliferation in both T cell populations. Additional IFN-γ quantification in the culture supernatant revealed a large correlation between the proliferation signals and IFN-γ production, indicating that BrdU labeling is a very reliable technique to assess and characterize feline lymphoproliferative responses to viral antigens in vitro.
Collapse
|
10
|
Teleshova N, Derby N, Martinelli E, Pugach P, Calenda G, Robbiani M. Simian immunodeficiency virus interactions with macaque dendritic cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 762:155-81. [PMID: 22975875 DOI: 10.1007/978-1-4614-4433-6_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This chapter summarizes advances in the following areas: (1) dendritic cell (DC)-mediated simian immunodeficiency virus (SIV) transmission, (2) role of DCs in innate and adaptive immunity against SIV, and (3) approaches to harness DC function to induce anti-SIV responses. The nonhuman primate (NHP) model of human immunodeficiency virus (HIV) infection in rhesus macaques and other Asian NHP species is highly relevant to advance the understanding of virus-host interactions critical for transmission and disease pathogenesis. HIV infection is associated with changes in frequency, phenotype, and function of the two principal subsets of DCs, myeloid DCs and plasmacytoid DCs. DC biology during pathogenic SIV infection is strikingly similar to that observed in HIV-infected patients. The NHP models provide an opportunity to dissect the requirements for DC-driven SIV infection and to understand how SIV distorts the DC system to its advantage. Furthermore, the SIV model of mucosal transmission enables the study of the earliest events of infection at the portal of entry that cannot be studied in humans, and, importantly, the involvement of DCs. Nonpathogenic infection in African NHP hosts allows investigations into the role of DCs in disease control. Understanding how DCs are altered during SIV infection is critical to the design of therapeutic and preventative strategies against HIV.
Collapse
Affiliation(s)
- Natalia Teleshova
- HIV and AIDS Program, Center for Biomedical Research, Population Council, New York, NY 10065, USA.
| | | | | | | | | | | |
Collapse
|
11
|
Jesudason S, Collins MG, Rogers NM, Kireta S, Coates PTH. Non-human primate dendritic cells. J Leukoc Biol 2011; 91:217-28. [PMID: 22124138 DOI: 10.1189/jlb.0711355] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Non-human primates (NHP) are essential translational models for biomedical research. Dendritic cells (DC) are a group of antigen presenting cells (APC) that play pivotal roles in the immunobiology of health and disease and are attractive cells for adoptive immunotherapy to stimulate and suppress immunity. DC have been studied extensively in humans and mice but until recently, have not been well characterized in NHP. This review considers the available data about DC across a range of NHP species and summarizes the understanding of in vitro-propagated DC and in vivo-isolated DC, which is now established. It is clear that although NHP DC exist within the paradigm of human DC, there are important functional and phenotypic differences when compared with human DC subsets. These differences need to be taken into account when designing preclinical, translational studies of DC therapy using NHP models.
Collapse
Affiliation(s)
- Shilpanjali Jesudason
- Transplantation Immunology Laboratory and Department of Medicine, University of Adelaide, The Queen Elizabeth Hospital Campus, Adelaide, South Australia, Australia
| | | | | | | | | |
Collapse
|
12
|
Prasad S, Kireta S, Leedham E, Russ GR, Coates PTH. Propagation and characterisation of dendritic cells from G-CSF mobilised peripheral blood monocytes and stem cells in common marmoset monkeys. J Immunol Methods 2009; 352:59-70. [PMID: 19931270 DOI: 10.1016/j.jim.2009.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2009] [Revised: 11/09/2009] [Accepted: 11/10/2009] [Indexed: 01/06/2023]
Abstract
The common marmoset is a small New World Primate that has been used as an immunological model for a number of human diseases. Dendritic cells (DC) have not been extensively characterised in this species and in particular protocols to derive DC from living donors without the need for animal sacrifice are presently lacking. This study establishes new protocols to generate substantial numbers of marmoset DC for use in cell therapy studies. Recombinant human G-CSF was used to mobilise peripheral blood monocytes and CD34(+) stem cells in sufficient numbers for large scale in-vitro DC propagation using cytokine conditioning including IL-4, GM-CSF, FLT3-L, stem cell factor and thrombopoietin. Marmoset DC exhibited morphology similar to human DC, were capable of antigen uptake and presentation and had moderate allo-stimulatory ability. Monocyte-derived DC had a maturation-resistant immature phenotype, whereas haematopoietic precursor-derived DC were semi-mature in phenotype and function. This study confirms the feasibility of the marmoset as a unique small primate model in which to pursue DC-based immunotherapy strategies.
Collapse
Affiliation(s)
- Shilpanjali Prasad
- Transplantation Immunology Laboratory and Department of Medicine, University of Adelaide, The Queen Elizabeth Hospital Campus, 28 Woodville Road, Woodville, SA 5011, Australia
| | | | | | | | | |
Collapse
|
13
|
Grandi G, Genchi C, Bazzocchi C, Mortarino M, Borghetti P, De Angelis E, Kramer LH. Generation and infection of bovine PBMC-derived dendritic cells with Neospora caninum. Vet Res Commun 2008; 32 Suppl 1:S207-9. [PMID: 18683066 DOI: 10.1007/s11259-008-9159-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- G Grandi
- Dipartimento di Produzioni Animali, Biotecnologie Veterinarie, Qualità e Sicurezza degli Alimenti, Via del Taglio 8, Parma 43100, Italy.
| | | | | | | | | | | | | |
Collapse
|
14
|
Jasny E, Eisenblätter M, Mätz-Rensing K, Tenner-Racz K, Tenbusch M, Schrod A, Stahl-Hennig C, Moos V, Schneider T, Racz P, Uberla K, Kaup FJ, Ignatius R. IL-12-impaired and IL-12-secreting dendritic cells produce IL-23 upon CD154 restimulation. THE JOURNAL OF IMMUNOLOGY 2008; 180:6629-39. [PMID: 18453582 DOI: 10.4049/jimmunol.180.10.6629] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Experimental studies in monkeys on the basis of ex vivo-generated, reinjected dendritic cells (DCs) allow investigations of primate DC biology in vivo. To study in vitro and in vivo properties of DCs with a reduced capacity to produce IL-12, we adapted findings obtained in vitro with human cells to the rhesus macaque model. Following exposure of immature monocyte-derived monkey DCs to the immunomodulating synthetic polypeptide glatiramer acetate (GA) and to dibutyryl-cAMP (d-cAMP; i.e., a cAMP enhancer that activates DCs but inhibits the induction of Th1 immune responses), the resulting DCs displayed a mature phenotype with enhanced Ag-specific T cell stimulatory function, notably also for memory Th1 cells. Phosphorylation of p38 MAPK was not induced in GA/d-cAMP-activated DCs. Accordingly, these cells secreted significantly less IL-12p40 (p < or = 0.001) than did cytokine-activated cells. However, upon restimulation with rhesus macaque CD154, GA/d-cAMP-activated DCs produced IL-12p40/IL-23. Additionally, DCs activated by proinflammatory cytokines following protocols for the generation of cells used in clinical studies secreted significantly more IL-23 upon CD154 restimulation than following prior activation. Two days after intradermal injection, GA/d-cAMP-activated fluorescence-labeled DCs were detected in the T cell areas of draining lymph nodes. When similarly injected, GA/d-cAMP as well as cytokine-activated protein-loaded DCs induced comparable Th immune responses characterized by secretion of IFN-gamma, TNF, and IL-17, and transiently expanded FOXP3(+) regulatory T cells. Reactivation of primate DCs through CD154 considerably influences their immmunostimulatory properties. This may have a substantial impact on the development of innovative vaccine approaches.
Collapse
Affiliation(s)
- Edith Jasny
- Institute of Microbiology and Hygiene, Department of Infection Immunology, Charité-University Medicine Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Moreau A, Chiffoleau E, Beriou G, Deschamps JY, Heslan M, Ashton-Chess J, Rolling F, Josien R, Moullier P, Cuturi MC, Alliot-Licht B. Superiority of Bone Marrow-Derived Dendritic Cells Over Monocyte-Derived Ones for the Expansion of Regulatory T Cells in the Macaque. Transplantation 2008; 85:1351-6. [DOI: 10.1097/tp.0b013e31816f22d6] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
16
|
Toda M. Analysis of dendritic cells from common marmosets for the treatment of CNS injury. Inflamm Regen 2008. [DOI: 10.2492/inflammregen.28.174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
17
|
Ohta S, Ueda Y, Yaguchi M, Matsuzaki Y, Nakamura M, Toyama Y, Tanioka Y, Tamaoki N, Nomura T, Okano H, Kawakami Y, Toda M. Isolation and characterization of dendritic cells from common marmosets for preclinical cell therapy studies. Immunology 2007; 123:566-74. [PMID: 18005037 DOI: 10.1111/j.1365-2567.2007.02727.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Dendritic cells (DCs) have important functions as modulators of immune responses, and their ability to activate T cells is of great value in cancer immunotherapy. The isolation of DCs from the peripheral blood of rhesus and African green monkeys has been reported, but the immune system in the common marmoset remains poorly characterized, although it offers many potential advantages for preclinical studies. In the present study, we devised methods, based on techniques developed for mouse and human DC preparation, for isolating DCs from three major tissue sources in the common marmoset: bone marrow (BM), spleen and peripheral blood. Each set of separated cells was analysed using the cell surface DC-associated markers CD11c, CD80, CD83, CD86 and human leucocyte antigen (HLA)-DR, all of which are antibodies against human antigens, and the cells were further characterized both functionally and morphologically as antigen-presenting cells. BM proved to be an excellent cell source for the isolation of DCs intended for preclinical studies on cell therapy, for which large quantities of cells are required. In the BM-derived CD11c(+) cell population, cells exhibiting the characteristic features of DCs were enriched, with the typical DC morphology and the abilities to undergo endocytosis, to secrete interleukin (IL)-12, and to stimulate Xenogenic T cells. Moreover, BM-derived DCs produced the neurotrophic factor NT-3, which is also found in murine splenic DCs. These results suggest that BM-derived DCs from the common marmoset may be useful for biological analysis and for preclinical studies on cell therapy for central nervous system diseases and cancer.
Collapse
Affiliation(s)
- Shigeki Ohta
- Neuroimmunology Research Group, Keio University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Nehete PN, Nehete BP, Hill L, Manuri PR, Baladandayuthapani V, Feng L, Simmons J, Sastry KJ. Selective induction of cell-mediated immunity and protection of rhesus macaques from chronic SHIV(KU2) infection by prophylactic vaccination with a conserved HIV-1 envelope peptide-cocktail. Virology 2007; 370:130-41. [PMID: 17920095 DOI: 10.1016/j.virol.2007.08.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Revised: 07/25/2007] [Accepted: 08/21/2007] [Indexed: 10/22/2022]
Abstract
Infection of Indian-origin rhesus macaques by the simian human immunodeficiency virus (SHIV) is considered to be a suitable preclinical model for directly testing efficacy of vaccine candidates based on the HIV-1 envelope. We used this model for prophylactic vaccination with a peptide-cocktail comprised of highly conserved HIV-1 envelope sequences immunogenic/antigenic in macaques and humans. Separate groups of macaques were immunized with the peptide-cocktail by intravenous and subcutaneous routes using autologous dendritic cells (DC) and Freund's adjuvant, respectively. The vaccine elicited antigen specific IFN-gamma-producing cells and T-cell proliferation, but not HIV-neutralizing antibodies. The vaccinated animals also exhibited efficient cross-clade cytolytic activity against target cells expressing envelope proteins corresponding to HIV-1 strains representative of multiple clades that increased after intravenous challenge with pathogenic SHIV(KU2). Virus-neutralizing antibodies were either undetectable or present only transiently at low levels in the control as well as vaccinated monkeys after infection. Significant control of plasma viremia leading to undetectable levels was achieved in majority of vaccinated monkeys compared to mock-vaccinated controls. Monkeys vaccinated with the peptide-cocktail using autologous DC, compared to Freund's adjuvant, and the mock-vaccinated animals, showed significantly higher IFN-gamma production, higher levels of vaccine-specific IFN-gamma producing CD4(+) cells and significant control of plasma viremia. These results support DC-based vaccine delivery and the utility of the conserved HIV-1 envelope peptide-cocktail, capable of priming strong cell-mediated immunity, for potential inclusion in HIV vaccination strategies.
Collapse
Affiliation(s)
- Pramod N Nehete
- Department of Veterinary Sciences, The University of Texas M. D. Anderson Cancer Center, Bastrop, TX 78602, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Zahorchak AF, Kean LS, Tokita D, Turnquist HR, Abe M, Finke J, Hamby K, Rigby MR, Larsen CP, Thomson AW. Infusion of Stably Immature Monocyte-Derived Dendritic Cells Plus CTLA4Ig Modulates Alloimmune Reactivity in Rhesus Macaques. Transplantation 2007; 84:196-206. [PMID: 17667811 DOI: 10.1097/01.tp.0000268582.21168.f6] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Immature dendritic cells (DC) can promote long-term transplant survival in rodents. We assessed the impact of stably immature, donor-derived DC on alloimmune reactivity in rhesus macaques. METHODS CD14 monocytes isolated from leukapheresis products of Macacca mulatta were cultured in granulocyte-macrophage colony stimulating factor plus interleukin (IL)-4+/-vitamin (vit) D3, and IL-10. Major histocompatibility complex class II and cosignaling molecule expression was determined on CD11c cells by flow cytometry. T-cell allostimulatory capacity of the DC, including DC exposed to proinflammatory cytokines, was determined in mixed leukocyte reaction. To test their influence in vivo, purified DC were infused intravenously into allogeneic recipients, either alone or followed by CTLA4Ig, 24 hr later. Proliferative responses of recipient CFSE-labeled T cells to donor or third party DC, cytokine production by stimulated T cells, and circulating alloantibody levels were determined by flow cytometry, up to 100 days postinfusion. RESULTS VitD3/IL-10-conditioned, monocyte-derived DC were resistant to maturation and failed to induce allogeneic T cell proliferation in vitro. After their infusion, an increase in anti-donor and anti-third party T-cell reactivity was observed, that subsequently subsided to fall significantly below pretreatment levels (by day 56) only in animals also given CTLA4Ig. No increase in circulating immunoglobulin (Ig) M or IgG anti-donor alloantibody titers compared with pretreatment values was detected. With DC+CTLA4Ig infusion, alloreactive IL-10-producing T cells were prevalent in the circulation after day 28. CONCLUSIONS Maturation-resistant rhesus DC infusion is well-tolerated. DC+CTLA4Ig infusion modulates allogeneic T-cell responses and results in hyporesponsiveness to donor and third party alloantigens.
Collapse
Affiliation(s)
- Alan F Zahorchak
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Kim N, Dabrowska A, Jenner RG, Aldovini A. Human and simian immunodeficiency virus-mediated upregulation of the apoptotic factor TRAIL occurs in antigen-presenting cells from AIDS-susceptible but not from AIDS-resistant species. J Virol 2007; 81:7584-97. [PMID: 17494085 PMCID: PMC1933355 DOI: 10.1128/jvi.02616-06] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Human immunodeficiency virus (HIV) and simian immunodeficiency virus (SIV) infections lead to AIDS in humans and rhesus macaques (RM), while they are asymptomatic in species naturally infected with SIV, such as chimpanzees, sooty mangabeys (SM), and African green monkeys (AGM). Differential CD4(+) T-cell apoptosis may be responsible for these species-specific differences in susceptibility to disease. To identify factors that influence the different apoptotic responses of these species, we analyzed virus-infected human and nonhuman primate peripheral blood mononuclear cells (PBMC). We found that the apoptotic factor TRAIL was present at higher levels in human and RM PBMC cultures and was mediating, at least in part, CD4(+) T-cell apoptosis in these cultures. The species-specific increase in TRAIL and death receptor expression observed with cultures also occurred in vivo in SIV-infected RM but not in SIV-infected SM. In human and RM myeloid immature dendritic cells and macrophages, the virus-induced expression of TRAIL and other interferon-inducible genes, which did not occur in the same cells from chimpanzee, SM, and AGM, was Tat dependent. Our results link the differential induction of TRAIL in human and nonhuman primate cells to species-specific differences in disease susceptibility.
Collapse
Affiliation(s)
- Nayoung Kim
- Children's Hospital Boston, Department of Medicine, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
21
|
Morelli AE, Coates PTH, Shufesky WJ, Barratt-Boyes SM, Fung JJ, Demetris AJ, Thomson AW. Growth factor-induced mobilization of dendritic cells in kidney and liver of rhesus macaques: implications for transplantation. Transplantation 2007; 83:656-62. [PMID: 17353790 DOI: 10.1097/01.tp.0000255320.00061.e9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Hematopoietic growth factors (HGF) mobilize potential tolerogenic cells in transplant donors. Fms-like tyrosine kinase 3 ligand (Flt3L) mobilizes stem cells and dendritic cells (DCs) in human and nonhuman primate blood. Blood and renal and liver biopsies were obtained from untreated and Flt3L-mobilized rhesus macaques. Flt3L increased the number of myeloid CD11c(hi) and plasmacytoid CD123(hi) precursors in blood and both myeloid CD11c(+) HLA-DR(+) fascin(+) (CD45RA(-)) DCs and putative plasmacytoid CD11c(lo) CD45RA(hi) DC precursors in liver and kidneys, without affecting organ function. DC in Flt3L-treated monkeys were concentrated in the glomeruli and interstitium of kidneys, and in the portal triads and parenchyma of liver. These DCs exhibited the phenotype of immature antigen-presenting cells (APCs; CD83(-) CD86(lo) CCR5(+) CCR7(-)). HGF-induced changes reversed significantly within 7 days of Flt3L withdrawal. Therapeutic protocols that mobilize donor hematopoietic cells should consider the influence of HGF on the APC constituency of prospective organ allografts.
Collapse
Affiliation(s)
- Adrian E Morelli
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Bautista EM, Nfon C, Ferman GS, Golde WT. IL-13 replaces IL-4 in development of monocyte derived dendritic cells (MoDC) of swine. Vet Immunol Immunopathol 2007; 115:56-67. [PMID: 17070934 DOI: 10.1016/j.vetimm.2006.09.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2006] [Revised: 07/25/2006] [Accepted: 09/07/2006] [Indexed: 11/21/2022]
Abstract
Dendritic cells (DCs) are a critical aspect of innate immune responses in addition to initiating adaptive immunity. In vitro generation of monocyte derived dendritic cells (MoDC) by culturing cells in IL-4 and granulocyte/macrophage colony stimulating factor (GM-CSF) has been reported for multiple species including swine. However, IL-4 is not a prominent cytokine detected in the periphery of common breeds of swine such as Yorkshire pigs. In this study, we report the generation and characterization of porcine MoDC in vitro using porcine IL-13 and porcine GM-CSF. These cells have the predicted expression of Class II MHC and T cell costimulatory molecules, phagocytic capacity and the ability to process and present antigen. Critically, porcine IL-13/GM-CSF MoDC have the unique ability to stimulate a primary mixed lymphocyte response in vitro. The type I interferon response of these MoDC to poly I:C (TLR3 ligand), LPS (TLR4 ligand) and CpG (TLR9 ligand) was tested. Of these TLR agonists, LPS or CpG did not stimulate induction of type I interferons, but a strong response was observed to poly I:C. This analysis shows that the generation of MoDCs in IL-13 yields cells of equivalent phenotype and function as IL-4 generated DC. However, for swine, in vitro generation of MoDC in IL-13 is likely to induce a more physiological cell population to study given expression of IL-4 is lacking in the periphery of these animals.
Collapse
Affiliation(s)
- Elida M Bautista
- Foot-and-Mouth Disease Research Unit, Plum Island Animal Disease Center, ARS, USDA, Greenport, NY 11944, USA
| | | | | | | |
Collapse
|
23
|
van der Meer FJUM, Schuurman NMP, Egberink HF. Feline immunodeficiency virus infection is enhanced by feline bone marrow-derived dendritic cells. J Gen Virol 2007; 88:251-258. [PMID: 17170458 DOI: 10.1099/vir.0.82450-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In the pathogenesis of feline immunodeficiency virus (FIV) infection, feline dendritic cells (feDCs) are thought to play an important role. As with DCs in other species, feDCs are believed to transport virus particles to lymph nodes and transfer them to lymphocytes. Our investigation has focused on the ability of feDCs to influence the infection of syngeneic peripheral blood mononuclear cells (PBMCs) and allogeneic thymocytes. feDCs were derived from bone marrow mononuclear cells that were cultured under the influence of feline interleukin-4 and feline granulocyte–macrophage colony-stimulating factor. By using these feDCs in co-culture with resting PBMCs, an upregulation of FIV replication was shown. An enhancement of FIV infection was also detected when co-cultures of feDCs/feline thymocytes were infected. To obtain this enhancement, direct contact of the cells in the co-culture was necessary; transwell cultures showed that the involvement of only soluble factors produced by feDCs in this process is not likely. These feDCs were also able to induce the proliferation of resting thymocytes, which might explain the enhanced FIV replication observed. Together, these data suggest that feDCs have abilities similar to those shown for simian and human DCs in the interaction with leukocytes. This system is suitable for further investigations of the interplay of DC and T cells during FIV infection in vitro.
Collapse
Affiliation(s)
- F J U M van der Meer
- Department of Immunology and Infectious Diseases, Division of Virology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - N M P Schuurman
- Department of Immunology and Infectious Diseases, Division of Virology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| | - H F Egberink
- Department of Immunology and Infectious Diseases, Division of Virology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL Utrecht, The Netherlands
| |
Collapse
|
24
|
Cassiani-Ingoni R, Cabral ES, Lünemann JD, Garza Z, Magnus T, Gelderblom H, Munson PJ, Marques A, Martin R. Borrelia burgdorferi Induces TLR1 and TLR2 in human microglia and peripheral blood monocytes but differentially regulates HLA-class II expression. J Neuropathol Exp Neurol 2006; 65:540-8. [PMID: 16783164 DOI: 10.1097/00005072-200606000-00002] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The spirochete Borrelia burgdorferi is the agent of Lyme disease, which causes central nervous system manifestations in up to 20% of patients. We investigated the response of human brain microglial cells, glial progenitors, neurons, astrocytes, as well as peripheral blood monocytes to stimulation with B. burgdorferi. We used oligoarrays to detect changes in the expression of genes important for shaping adaptive and innate immune responses. We found that stimulation with B. burgdorferi lysate increased the expression of Toll-like receptors (TLRs) 1 and 2 in all cell types except neurons. However, despite similarities in global gene profiles of monocytes and microglia, only microglial cells responded to the stimulation with a robust increase in HLA-DR, HLA-DQ, and also coexpressed CD11-c, a dendritic cell marker. In contrast, a large number of HLA-related molecules were repressed at both the RNA and the protein levels in stimulated monocytes, whereas secretion of IL-10 and TNF-alpha was strongly induced. These results show that signaling through TLR1/2 in response to B. burgdorferi can elicit opposite immunoregulatory effects in blood and in brain immune cells, which could play a role in the different susceptibility of these compartments to infection.
Collapse
Affiliation(s)
- Riccardo Cassiani-Ingoni
- Cellular Immunology Section, Neuroimmunology Branch, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Teleshova N, Kenney J, Robbiani M. Dendritic cells and HIV infection: activating dendritic cells to boost immunity. Adv Dent Res 2006; 19:36-41. [PMID: 16672547 DOI: 10.1177/154407370601900108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dendritic cells (DCs) are white blood cells that coordinate innate and adaptive immunity. They are distributed within epithelia and mucosal-associated lymphoid tissues, positioned to entrap incoming pathogens or vaccines. Human immunodeficiency virus (HIV) and the non-human primate equivalent (SIV) exploit DCs to amplify infection, underscoring the need to harness strategies that promote presentation of virus by DCs to stimulate potent anti-viral immunity instead of virus transmission. Two main subsets of DCs need to be considered: myeloid (MDC) and plasmacytoid (PDC) subsets. Using the SIV-macaque system to advance oral vaccine research, we examined macaque PDC and MDC biology, identifying ways to activate DCs and boost antiviral immunity. Immunostimulatory oligodeoxyribonucleotides (ISS-ODNs) stimulated PDC/MDC mixtures to up-regulate co-stimulatory molecule expression and to secrete both IFN-alpha and IL-12. Additionally, ISS-ODNs augmented SIV-specific IFN-gamma responses induced by virus-bearing DCs. ISS-ODN-driven DC activation is being pursued to improve oral/nasopharyngeal mucosal vaccines and therapies against HIV.
Collapse
Affiliation(s)
- N Teleshova
- Center for Biomedical Research, Population Council, 1230 York Avenue, New York, NY 10021, USA
| | | | | |
Collapse
|
26
|
Awasthi S, Cropper J. Immunophenotype and functions of fetal baboon bone-marrow derived dendritic cells. Cell Immunol 2006; 240:31-40. [PMID: 16842767 DOI: 10.1016/j.cellimm.2006.06.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2006] [Revised: 05/31/2006] [Accepted: 06/02/2006] [Indexed: 01/22/2023]
Abstract
Dendritic cells (DCs) are unique antigen-presenting cells that can take up pathogens, pathogens-derived and stress-antigens and stimulate antigen-specific immune response. Here we investigated the immunobiology of fetal DCs and compared their phenotype and activation status against infectious stimuli with those of young and adult baboons. The DCs were obtained from femoral bone-marrow (BMDCs) of fetus (140 and 175 days of gestation), young (4-5 years old) and mature adult (10-35 years old) baboons. The cells were cultured in the presence of GM-CSF and IL-4. To study phagocytic ability of BMDCs, the cells were harvested on 6th day and incubated with fluorescent-labeled Escherichia coli bioparticles. The BMDCs were also treated with E. coli O111:B4 lipopolysaccharide (LPS) for 24h and changes in expression of cell-surface markers and IL-12 were studied using distinct immunoassays. We found that the phenotype and morphology of BMDCs from fetal, young and adult baboons were similar and showed increased expression of HLA-DP, DQ, DR and T cell co-stimulatory molecules upon LPS treatment. However, significant differences were observed in phagocytic activity and IL-12 secretion among BMDCs from these sources. The ability of fetal baboon BMDCs to phagocytose E. coli bioparticles was significantly lower and they secreted lower level of LPS-stimulated IL-12 as compared to the BMDCs from adult baboon. These results suggest that compared to adult BMDCs, fetal baboon BMDCs are less efficient in mounting immune response against Gram-negative bacterial stimuli.
Collapse
Affiliation(s)
- Shanjana Awasthi
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| | | |
Collapse
|
27
|
Mortara L, Ploquin MJY, Faye A, Scott-Algara D, Vaslin B, Butor C, Hosmalin A, Barré-Sinoussi F, Diop OM, Müller-Trutwin MC. Phenotype and function of myeloid dendritic cells derived from African green monkey blood monocytes. J Immunol Methods 2005; 308:138-55. [PMID: 16325847 DOI: 10.1016/j.jim.2005.10.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2005] [Revised: 07/08/2005] [Accepted: 10/21/2005] [Indexed: 01/19/2023]
Abstract
Myeloid dendritic cells probably play an important role in the immune response against HIV and SIV, and in the enhancement of CD4+ T cell infection. Here, we have investigated phenotypic and functional features of myeloid monocyte-derived DC (MDDC) from African green monkeys (AGMs). AGMs are natural hosts of SIV and exhibit no signs of abnormal T cell activation despite high SIV plasma viremia. We identified mAbs that cross-react specifically with homologous molecules expressed on AGM DC. We adapted a protocol to derive AGM MDDC by culture in the presence of GM-CSF and IL-4. The differentiated cells possessed a typical dendritic morphology and the majority were CD11c+ DC-SIGN+. AGM MDDC displayed a high expression of typical maturation markers, such as CD83, CD86 and DC-LAMP, and moderate immunostimulatory capacity, suggesting that the cells were in a semi-mature state. Stimulation resulted in further maturation, as shown by up-regulation of CD80 and decrease of endocytosis ability. However, neither increase of HLA-DR or CD40 expression nor enhanced immunostimulatory capacity was observed. The latter was associated with a low pro-inflammatory cytokine production during mixed lymphocyte reactions and a cytokine balance in favour of IL-10 in contrast to human MDDC. This is the first characterization of AGM MDDC. The tools described here are a crucial step for future studies in vivo or in vitro on the function of myeloid DC using the AGM animal model.
Collapse
Affiliation(s)
- Lorenzo Mortara
- Unité de Régulation des Infections Rétrovirales, Institut Pasteur, 25, rue du Docteur Roux, 75015 Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sprague WS, Pope M, Hoover EA. Culture and comparison of feline myeloid dendritic cells vs macrophages. J Comp Pathol 2005; 133:136-45. [PMID: 16038926 DOI: 10.1016/j.jcpa.2005.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2004] [Accepted: 03/07/2005] [Indexed: 11/26/2022]
Abstract
To gain insight into the role of dendritic cells (DCs) in feline immunodeficiency virus (FIV) infection and immunity, methods were developed to culture feline myeloid DCs from CD14(+) monocytes with a combination of human recombinant granulocyte-macrophage colony-stimulating factor (hrGM-CSF) and interleukin-4 (hrIL-4). These cells were compared with feline macrophages cultured in the presence of hrGM-CSF. As with DCs in other species, feline DCs showed uniformly high MHC class II expression, moderate B7.1 expression, potent induction of the allogeneic mixed leucocyte reaction (MLR), and moderate uptake of fluorescein isothiocyanate-dextran (FITC-DX) in the endocytic assay. In comparison with feline macrophages, DCs showed higher expression of MHC class II, similar expression of B7.1, CD14, CXCR4 and CD1a, and lower expression of CD11b. When placed on alcian blue-coated glass slides, DCs differed from macrophages in showing a greater tendency to spread out; they also had characteristic fine cytoplasmic processes instead of the broader pseudopodia of macrophages. Basal IL-12 mRNA expression and FITC-DX uptake were greater in DCs than in macrophages. Unlike feline DCs, feline macrophages exhibited a dose-dependent suppressive effect in the MLR. Feline DCs propagated in vitro should prove useful in the development of DC-mediated vaccination and therapy for infectious and neoplastic feline diseases. Additionally, macrophages cultured with GM-CSF provide a potential means of studying the mechanism of immunosuppression in cats.
Collapse
Affiliation(s)
- W S Sprague
- Department of Microbiology, Immunology and Pathology, Colorado State University, 1619 Campus Delivery, Fort Collins, CO 80523-1619, USA
| | | | | |
Collapse
|
29
|
Miranda de Carvalho C, Bonnefont-Rebeix C, Rigal D, Chabanne L. "Dendritic cells in different animal species: an overview". ACTA ACUST UNITED AC 2005; 54:85-93. [PMID: 16019158 DOI: 10.1016/j.patbio.2005.04.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2005] [Accepted: 04/13/2005] [Indexed: 12/30/2022]
Abstract
The comprehension of the immune system and the role of DC in the pathological diseases may contribute to their use in veterinary medicine in the prevention and treatment of many diseases. Currently, most dendritic cell (DC) research occurs in the human and murine model systems on the generation of cells from the bone marrow or peripheral blood mononuclear cells (PBMC) cultured in vitro. Despite the lack of available immunological reagents such as antibodies and cytokines, analogous cells have been generated and identified in many different species and reviewed in this study.
Collapse
Affiliation(s)
- C Miranda de Carvalho
- Etablissement français du sang, 1-3, rue du Vercors, 69007 Lyon, France; Ecole nationale vétérinaire de Lyon, 1, avenue Bourgelat, 69380 Marcy l'Etoile, France.
| | | | | | | |
Collapse
|
30
|
Ibisch C, Pradal G, Bach JM, Lieubeau B. Functional canine dendritic cells can be generated in vitro from peripheral blood mononuclear cells and contain a cytoplasmic ultrastructural marker. J Immunol Methods 2005; 298:175-82. [PMID: 15847807 DOI: 10.1016/j.jim.2005.02.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2004] [Revised: 12/10/2004] [Accepted: 02/16/2005] [Indexed: 01/06/2023]
Abstract
For physiological and practical reasons the dog is a large animal model used increasingly to study the pathogenesis of human diseases and new therapeutic approaches, in particular for immune disorders. However, some immunological resources are lacking in this model, especially concerning dendritic cells. The aim of our study was to develop an efficient method to generate dendritic cells (DC) in vitro from dog peripheral blood mononuclear cells (PBMC) and to characterize their functional, structural and ultrastructural properties. PBMC were cultured in vitro with IL-4 and GM-CSF. After 1 week of culture, a great proportion of non-adherent cells displayed typical cytoplasmic processes, as evidenced both by optical and electron microscopy. Cytometric analysis revealed the presence of 41.7+/-24.6% CD14+ cells expressing both CD11c and MHC class II molecules. Allogeneic mixed lymphocyte reactions confirmed the ability of these cultures to stimulate the proliferation of allogeneic lymphocytes as already reported as a characteristic of DC in other species. In addition, we describe for the first time the presence in canine DC of cytoplasmic periodic microstructures (PMS) that could represent ultrastructural markers of canine DC. In conclusion, our study provides an easy method to generate DC from PBMC in sufficient numbers for immunological in vitro investigations in dogs, a pre-clinical model for many human diseases.
Collapse
Affiliation(s)
- C Ibisch
- Immuno-Endocrinology Unit, ENVN/INRA/University, ENVN, Atlanpole, la Chantrerie, BP 40706, F-44307 Nantes Cedex 03, France
| | | | | | | |
Collapse
|
31
|
Gabriela D, Carlos PL, Clara S, Elkin PM. Phenotypical and functional characterization of non-human primate Aotus spp. dendritic cells and their use as a tool for characterizing immune response to protein antigens. Vaccine 2005; 23:3386-95. [PMID: 15837362 DOI: 10.1016/j.vaccine.2005.01.155] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Revised: 01/14/2005] [Accepted: 01/17/2005] [Indexed: 10/25/2022]
Abstract
A population of cells exhibiting bona fide dendritic cell (DC) morphological and functional characteristics was obtained by treating Aotus spp. monocytes with human IL-4 and GM-CSF. Although the purity of mature DCs was relatively low IL-4/GM-CSF-treated monocytes (hereafter called Aotus spp. DCs) down-regulated CD14 and up-regulated discrete levels of CD80, MHC-Class II and CD1b molecules in response to different maturation stimuli. Aotus spp. DCs generated a potent allogeneic in vitro response evidenced in mixed lymphocyte reaction (MLR) where DCs were 2- to 10-fold more efficient than peripheral blood mononuclear cells (PBMCs). Aotus spp. DC ability to boost T-cells or priming naive T-cells in vivo was proved by vaccinating Aotus spp. with autologous DCs pulsed with tetanus toxoid (TT). A single dose of TT-pulsed DCs was sufficient to increase cellular response to TT in these experiments as assessed by lymphoproliferation and cytokine production. Since Aotus spp. represents a suitable animal model for evaluating anti-Plasmodium falciparum malaria vaccine, the results shown here suggest that using antigen-pulsed Aotus spp. DCs as vaccines might lead to identifying new prospects for malarial vaccines unidentified to date because they are being formulated in less efficient adjuvants.
Collapse
Affiliation(s)
- Delgado Gabriela
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50, No. 26-00, Bogotá, Colombia
| | | | | | | |
Collapse
|
32
|
Ashton-Chess J, Blancho G. An in vitro evaluation of the potential suitability of peripheral blood CD14+ and bone marrow CD34+-derived dendritic cells for a tolerance inducing regimen in the primate. J Immunol Methods 2005; 297:237-52. [PMID: 15777946 DOI: 10.1016/j.jim.2004.12.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2004] [Revised: 11/03/2004] [Accepted: 12/13/2004] [Indexed: 10/25/2022]
Abstract
Dendritic cells (DC) represent a tool not only for immune activation, but also potentially for tolerance induction in transplantation. This latter approach is yet to be explored in a pre-clinical primate model. Since no information concerning baboon DC has been available, we characterised the DC of this species derived in vitro from bone marrow (CD34(+)) and peripheral blood (CD14(+)) precursors to determine which would be the most suitable for a tolerance inducing strategy. Baboon DC were differentiated in vitro using protocols similar to those used in humans and their maturation status was assessed and compared according to their phenotype and function. Based on both phenotypic and functional criteria, the CD14-derived baboon DC appeared to be less mature DC, necessitating an additional stimulus in order to become fully mature. The CD34-derived DC on the other hand appeared more mature in nature, without necessarily requiring exposure to overt maturation signals. We suggest therefore that, in the baboon, peripheral blood CD14-derived DC may be more suitable for protocols where tolerance induction is the goal. We now aim to perform further in vitro investigations into the potential tolerance inducing effects of CD14-derived DC alone or in association with other strategies that would be applicable in vivo.
Collapse
Affiliation(s)
- Joanna Ashton-Chess
- INSERM U437 " Immunointervention en Allo et Xénotransplantation " and Institut de Transplantation et de Recherche en Transplantation, C.H.U. Hôtel-Dieu, 30 Bd Jean Monnet, 44093 Nantes Cedex 01, France
| | | |
Collapse
|
33
|
Teleshova N, Kenney J, Jones J, Marshall J, Van Nest G, Dufour J, Bohm R, Lifson JD, Gettie A, Pope M. CpG-C immunostimulatory oligodeoxyribonucleotide activation of plasmacytoid dendritic cells in rhesus macaques to augment the activation of IFN-gamma-secreting simian immunodeficiency virus-specific T cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:1647-57. [PMID: 15265893 DOI: 10.4049/jimmunol.173.3.1647] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
There are two principle subsets of dendritic cells (DCs); CD11c(+)CD123(-) myeloid DCs (MDCs) and CD11c(-)CD123(+) plasmacytoid DCs (PDCs). DC activation via TNF-TNFRs (e.g., CD40L) and TLRs (e.g., immunostimulatory oligodeoxyribonucleotides (ISS-ODNs)) is crucial for maximal stimulation of innate and adaptive immunity. Macaque DC biology is being studied to improve HIV vaccines using the SIV macaque model. Using lineage (Lin) markers to exclude non-DCs, Lin(-)HLA-DR(+)CD11c(+)CD123(-) MDCs and Lin(-)HLA-DR(+)CD11c(-)CD123(+) PDCs were identified in the blood of uninfected macaques and healthy macaques infected with SIV or simian-human immunodeficiency virus. Overnight culture of DC-enriched Lin-depleted cells increased CD80 and CD86 expression. IL-12 production and CD80/CD86 expression by MDC/PDC mixtures was further enhanced by CD40L and ISS-ODN treatment. A CpG-B ISS-ODN increased CD80/CD86 expression by PDCs, but resulted in little IFN-alpha secretion unless IL-3 was added. In contrast, a CpG-C ISS-ODN and aldrithiol-2-inactivated (AT-2) SIV induced considerable PDC activation and IFN-alpha release without needing exogenous IL-3. The CpG-C ISS-ODN also stimulated IL-12 release (unlike AT-2 SIV) and augmented DC immunostimulatory activity, increasing SIV-specific T cell IFN-gamma production induced by AT-2 SIV-presenting MDC/PDC-enriched mixtures. These data highlight the functional capacities of MDCs and PDCs in naive as well as healthy, infected macaques, revealing a promising CpG-C ISS-ODN-driven DC activation strategy that boosts immune function to augment preventative and therapeutic vaccine efficacy.
Collapse
Affiliation(s)
- Natalia Teleshova
- Center for Biomedical Research, Population Council, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Eisenblätter M, Stahl-Hennig C, Kuate S, Stolte N, Jasny E, Hahn H, Pope M, Tenner-Racz K, Racz P, Steinman RM, Uberla K, Ignatius R. Induction of neutralising antibodies restricts the use of human granulocyte/macrophage colony stimulating factor for vaccine studies in rhesus macaques. Vaccine 2004; 22:3295-302. [PMID: 15308352 DOI: 10.1016/j.vaccine.2004.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2003] [Revised: 02/05/2004] [Accepted: 03/02/2004] [Indexed: 11/29/2022]
Abstract
Granulocyte/macrophage-colony stimulating factor (GM-CSF) is a valuable adjuvant to enhance induction of cellular immune responses in rodents. Less information is available regarding its use as an adjuvant in primates or humans. We explored recombinant human GM-CSF for potential vaccine studies in rhesus macaques and focused on its effect on peripheral monocytes as progenitors of dendritic cells and its potential immunogenicity. Application of human GM-CSF to nine animals led to an average 32-fold increase in monocyte numbers. This was not observed upon re-treatment, which coincided with GM-CSF-specific neutralising antibodies. These also neutralised the activity of rhesus macaque GM-CSF. The data underscore the need to use species-specific GM-CSF for immunomodulation in primates.
Collapse
Affiliation(s)
- Martin Eisenblätter
- Department of Medical Microbiology and Immunology of Infection, Charité-University Medicine Berlin, Campus Benjamin Franklin, Hindenburgdamm 27, 12203 Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Villamide-Herrera L, Ignatius R, Eller MA, Wilkinson K, Griffin C, Mehlhop E, Jones J, Han SY, Lewis MG, Parrish S, Vancott TC, Lifson JD, Schlesinger S, Mascola JR, Pope M. Macaque dendritic cells infected with SIV-recombinant canarypox ex vivo induce SIV-specific immune responses in vivo. AIDS Res Hum Retroviruses 2004; 20:871-84. [PMID: 15320991 DOI: 10.1089/0889222041725136] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Dendritic cells (DCs) infected with recombinant avipox vectors express the introduced genes and activate antigen-specific T cells. DCs exhibit distinct differentiation-dependent immune functions. Moreover, immature DCs are readily infected by canarypox vectors, but undergo tumor necrosis factor (TNF)-alpha-dependent death, while fewer mature DCs get infected and resist dying. A pilot study was performed using the rhesus macaque system to explore whether immature and mature DCs infected with SIV-recombinant canarypox (vCP180) ex vivo could induce primary virus-specific immune responses in vivo. After subcutaneous (sc) reinjection, functional monocyte-derived DCs migrated to lymph nodes (LNs) within 1-2 days and primed T cells in vivo. This was observed by monitoring dye-labeled DCs in the draining LNs and tetanus toxoid (TT)-specific T cell responses after injection of TT-loaded DCs. DCs from simian immunodeficiency virus (SIV)-naïve rhesus macaques were infected with vCP180 (SIVmac142 gag, pol, and env genes), and sc reinjected into donor animals. Low-level SIV-specific T cell proliferation, but little if any interferon (IFN)-gamma production was detected. DCs pulsed with vCP180 in combination with TT and keyhole limpet hemocyanin (KLH) (to activate additional T cells and provide "helper" cytokines) induced SIV-, TT-, and KLH-specific T cell responses, including IFN-gamma responses not seen when vCP180-carrying DCs were used alone. Interleukin (IL)-10 and low-level antibody responses were also observed. This pilot study provides the proof of principle that sc injected ex vivo SIV-recombinant canarypox-infected DCs safely induce low-level SIV-specific immune responses in vivo.
Collapse
Affiliation(s)
- L Villamide-Herrera
- Center for Biomedical Research, Population Council, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Teleshova N, Jones J, Kenney J, Purcell J, Bohm R, Gettie A, Pope M. Short-term Flt3L treatment effectively mobilizes functional macaque dendritic cells. J Leukoc Biol 2004; 75:1102-10. [PMID: 15075365 DOI: 10.1189/jlb.1103588] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In vivo administration of soluble Flt3L increases dendritic cell (DC) numbers to favor improved DC targeting of vaccine antigens, augmenting vaccine efficiency. In addition to confirming the effectiveness of human Flt3L in macaques, we strove to determine the optimal regimen to elevate numbers of functional DCs. Circulating DCs were identified within lineage(-)human leukocyte antigen-DR(+) cells, which comprised CD11c(-)CD123(+) plasmacytoid DCs (PDCs) and CD123(-) cells including CD11c(+)CD123(-) myeloid DCs as well as CD11c(-)CD123(-) cells. Traditionally, DCs have been monitored 1-2 days after 10- to 14-day treatments with Flt3L (100 microg/kg/day). We demonstrate that although standard treatment increased macaque DC percentages, as little as 5-7 days of treatment was sufficient, if not more effective at mobilizing DCs. Moreover, DC frequency continued to escalate over the ensuing days, peaking at approximately 4 days post 7 days of treatment and ultimately decreasing thereafter. As expected, there was a more pronounced increase in the percentages and actual numbers of CD123(-) cells (CD11c(+) and CD11c(-) subsets) compared with PDCs. Flt3L-mobilized DCs exhibited slightly increased CD80/CD86 expression but typically still that of immature DCs and were resilient to freeze-thawing. Overnight culture activated the cells, up-regulating CD80/CD86 expression as well as interleukin-12 release, typically being boosted by CD40L. This was even more apparent for enriched DC cultures. These data verify that peak mobilization of large numbers of functional macaque DCs occurs a few days, not immediately, after short-term Flt3L dosing. This has important implications for improved DC-targeting vaccine strategies to prevent infection with human immunodeficiency virus and other pathogens.
Collapse
|
37
|
Teleshova N, Frank I, Pope M. Immunodeficiency virus exploitation of dendritic cells in the early steps of infection. J Leukoc Biol 2003; 74:683-90. [PMID: 12960236 DOI: 10.1189/jlb.0403178] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The unique capacity of dendritic cells (DCs) to capture and process pathogens for presentation to the immune system, combined with their capacity to express costimulatory and adhesion molecules as well as cytokines and chemokines, renders them powerful antigen-presenting cells. However, immunodeficiency viruses hijack DCs to facilitate virus dissemination while subverting effective immune activation. Depending on the activation level of the DC subset, human immunodeficiency virus can use different receptors (CD4, chemokine, and C-type lectin receptors) to bind to DCs. These aspects likely impact whether a DC is productively infected by or simply carries virus for transmission to more permissive targets. DCs efficiently transmit virus to CD4+ T cells, driving virus growth as well as providing signals to trigger virus expansion in virus-bearing CD4+ T cells. There is accumulating evidence that viral determinants (nef, tat) selectively modulate immature DC biology, fostering DC-T cell interactions and virus replication without up-regulating costimulatory molecules for effective immune function. In addition, virus-loaded, immature DCs activate CD4+ virus-specific T cells, and mature DCs stimulate CD4+ and CD8+ T cells. Thus, even if immature DCs entrap virus as it crosses the mucosae and initiate a CD4+ T cell response, this is likely insufficient to control infection. Appreciating how virus modulates DC function and what determines whether virus is processed for immune stimulation or transmitted between cells will unveil the exact role of these cells in the onset of infection and advance preventative microbicide and vaccine/therapeutic approaches.
Collapse
Affiliation(s)
- Natalia Teleshova
- Center for Biomedical Research, Population Council, New York, NY 10021, USA
| | | | | |
Collapse
|
38
|
Frank I, Santos JJ, Mehlhop E, Villamide-Herrera L, Santisteban C, Gettie A, Ignatius R, Lifson JD, Pope M. Presentation of exogenous whole inactivated simian immunodeficiency virus by mature dendritic cells induces CD4+ and CD8+ T-cell responses. J Acquir Immune Defic Syndr 2003; 34:7-19. [PMID: 14501788 DOI: 10.1097/00126334-200309010-00002] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Interactions between HIV-1 and dendritic cells (DCs) play an important role in the initial establishment and spread of infection and development of antiviral immunity. We used chemically inactivated aldrithiol-2 (AT-2) simian immunodeficiency virus (SIV) with functional envelope glycoproteins to study virus interactions with DCs and developed an in vitro system to evaluate the quality of SIV antigen (Ag) presentation by DCs to T cells. AT-2 SIV interacts authentically with T cells and DCs and thus allows assessment of natural SIV-specific responses. CD4+ and CD8+ T cells from blood or lymph nodes of SIV-infected macaques released interferon-gamma (IFN gamma) and proliferated in response to a variety of AT-2 SIV isolates. Responses did not vary significantly as a function of the quantitative envelope glycoprotein content of the virions. Presentation of Ags derived from AT-2 SIV by DCs was more potent than presentation by comparably Ag-loaded monocytes. Interestingly, SIV-pulsed mature DCs stimulated both CD4+ and CD8+ T-cell responses, whereas immature DCs primarily stimulated CD4+ T cells. Further studies using AT-2 inactivated virus may help to define better the details of the virus-DC interactions critical for infection versus induction of antiviral immune responses.
Collapse
Affiliation(s)
- Ines Frank
- Center for Biomedical Research, Population Council, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Coates PTH, Barratt-Boyes SM, Zhang L, Donnenberg VS, O'Connell PJ, Logar AJ, Duncan FJ, Murphey-Corb M, Donnenberg AD, Morelli AE, Maliszewski CR, Thomson AW. Dendritic cell subsets in blood and lymphoid tissue of rhesus monkeys and their mobilization with Flt3 ligand. Blood 2003; 102:2513-21. [PMID: 12829599 DOI: 10.1182/blood-2002-09-2929] [Citation(s) in RCA: 101] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We provide phenotypic and functional evidence of premonocytoid dendritic cells (DCs) and preplasmacytoid DCs in blood and of corresponding DC subsets in secondary lymphoid tissue of rhesus monkeys. Subsets were identified and sorted by 4-color flow cytometry using antihuman monoclonal antibodies cross-reactive with rhesus monkey. To mobilize pre-DC subsets, fms-like tyrosine 3 kinase ligand (Flt3L; 100 microg/kg subcutaneously) was administered for 10 days. Presumptive pre-DC subsets were identified within the lineage- (Lin-) major histocompatibility complex (MHC) class II+ fraction of blood mononuclear cells. Premonocytoid DCs were CD11c+CD123- (interleukin-3Ralpha- [IL-3Ralpha-]). Preplasmacytoid DCs were characterized as CD11c-CD123++ Flt3L increased the CD11c+ pre-DC (7-fold) and CD123++ pre-DC subsets (3-fold) in blood. The freshly isolated CD11c+ pre-DC subset induced modest proliferation of naive allogeneic T cells. After overnight culture with granulocyte macro-phage-colony-stimulating factor (GMCSF) and CD40L, both subsets up-regulated surface costimulatory molecules, and CD11c+ pre-DCs became potent allostimulators. Freshly isolated CD123++ pre-DCs showed typical plasmacytoid morphology and, when cultured with IL-3 and CD40L for 72 hours, developed mature DC morphology. Following stimulation with CD40L, CD11c+ pre-DCs secreted increased levels of IL-12p40. Importantly, herpes simplex virus-stimulated CD123++ pre-DCs, but not CD11c+ pre-DCs, secreted interferon-alpha (IFN-alpha). Corresponding DC subsets were identified by flow analysis and immunohistochemistry in lymph nodes wherein both populations were increased 2- to 3-fold by Flt3L administration. CD123+ pre-DCs produced IFN-alpha in response to in vivo viral infection. Thus, rhesus monkeys exhibit 2 distinct DC precursor populations that closely resemble those of humans. Both are mobilized into blood and lymphoid tissue by Flt3L, offering potential for their further characterization and possible therapeutic application.
Collapse
Affiliation(s)
- P Toby H Coates
- Thomas E. Starzl Transplantation Institute, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Angénieux C, Salamero J, Fricker D, Wurtz JM, Maître B, Cazenave JP, Hanau D, de la Salle H. Common characteristics of the human and rhesus macaque CD1e molecules: conservation of biochemical and biological properties during primate evolution. Immunogenetics 2003; 54:842-9. [PMID: 12671734 DOI: 10.1007/s00251-003-0538-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2002] [Revised: 01/09/2003] [Indexed: 11/27/2022]
Abstract
In humans, a family of five genes encodes the CD1 molecules. Four of these proteins, CD1a, b, c, and d, are expressed on the plasma membrane and traffic between the cell surface and endocytic compartments, where they are loaded with antigenic glycolipids. The existence of human CD1e was demonstrated recently. This molecule surprisingly remains inside the cell, accumulating mainly in the Golgi compartments of immature dendritic cells and in the late endosomes of mature dendritic cells. In the latter compartments, CD1e is cleaved and becomes soluble. To determine whether these properties were specific to human CD1e, we investigated the presence and characteristics of CD1e in the rhesus macaque, an evolutionarily distant species of the primate lineage. Our results show that the cellular and biochemical properties of the human and simian CD1e molecules are similar, suggesting that the particular intracellular distribution of CD1e is important for its physiological and/or immunological function.
Collapse
|
41
|
Messmer D, Jacqué JM, Santisteban C, Bristow C, Han SY, Villamide-Herrera L, Mehlhop E, Marx PA, Steinman RM, Gettie A, Pope M. Endogenously expressed nef uncouples cytokine and chemokine production from membrane phenotypic maturation in dendritic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:4172-82. [PMID: 12370346 DOI: 10.4049/jimmunol.169.8.4172] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Immature dendritic cells (DCs), unlike mature DCs, require the viral determinant nef to drive immunodeficiency virus (SIV and HIV) replication in coculture with CD4(+) T cells. Since immature DCs may capture and get infected by virus during mucosal transmission, we hypothesized that Nef associated with the virus or produced during early replication might modulate DCs to augment virus dissemination. Adenovirus vectors expressing nef were used to introduce nef into DCs in the absence of other immunodeficiency virus determinants to examine Nef-induced changes that might activate immature DCs to acquire properties of mature DCs and drive virus replication. Nef expression by immature human and macaque DCs triggered IL-6, IL-12, TNF-alpha, CXCL8, CCL3, and CCL4 release, but without up-regulating costimulatory and other molecules characteristic of mature DCs. Coincident with this, nef-expressing immature DCs stimulated stronger autologous CD4(+) T cell responses. Both SIV and HIV nef-expressing DCs complemented defective SIVmac239 delta nef, driving replication in autologous immature DC-T cell cultures. In contrast, if DCs were activated after capturing delta nef, virus growth was not exacerbated. This highlights one way in which nef-defective virus-bearing immature DCs that mature while migrating to draining lymph nodes could induce stronger immune responses in the absence of overwhelming productive infection (unlike nef-containing wild-type virus). Therefore, Nef expressed in immature DCs signals a distinct activation program that promotes virus replication and T cell recruitment but without complete DC maturation, thereby lessening the likelihood that wild-type virus-infected immature DCs would activate virus-specific immunity, but facilitating virus dissemination.
Collapse
Affiliation(s)
- Davorka Messmer
- Laboratory of Cellular Physiology and Immunology, Rockefeller University, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ignatius R, Tenner-Racz K, Messmer D, Gettie A, Blanchard J, Luckay A, Russo C, Smith S, Marx PA, Steinman RM, Racz P, Pope M. Increased macrophage infection upon subcutaneous inoculation of rhesus macaques with simian immunodeficiency virus-loaded dendritic cells or T cells but not with cell-free virus. J Virol 2002; 76:9787-97. [PMID: 12208957 PMCID: PMC136510 DOI: 10.1128/jvi.76.19.9787-9797.2002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Information on the establishment of immunodeficiency virus infection through transmission of infected cells is sparse. Dendritic cells (DCs) and T cells may be central to the onset and subsequent spread of infection following mucosal exposure. To directly investigate the consequences of virus being introduced by DCs or T cells, we reinjected ex vivo simian immunodeficiency virus (SIV)-loaded autologous immature DCs and T cells subcutaneously (s.c.) into healthy macaques. s.c. injection of cell-bound virus was used to mirror what may happen if virus-loaded cells pass through an epithelium or perhaps DCs and T cells that immediately entrap cell-free virus, having just crossed an epithelial barrier. Virus load in the plasma was monitored along with combined in situ hybridization and immunohistochemistry to identify the cells replicating virus in the lymphoid tissues. Both DCs and T cells transmitted infection after being pulsed with either wild-type or nef-defective (delta nef) SIVmac239. As seen in animals infected intravenously, replication of delta nef was attenuated compared to that of wild-type virus when introduced in either cell-bound form. Upon examination of the draining lymph nodes (LNs) during the first days of infection, virus-producing CD4(+) T cells predominated in control animals that received s.c. cell-free virus. In dramatic contrast, both SIV-positive macrophages and T cells were detected in the LNs of monkeys infected with cell-associated SIV. Therefore, although both cell-free and cell-associated viruses are infectious, the initial cells amplifying the virus differ. This may have important implications for the subsequent dissemination of infection and/or induction of antiretroviral immunity.
Collapse
Affiliation(s)
- Ralf Ignatius
- Laboratory of Cellular Physiology and Immunology, The Rockefeller University, New York, New York 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Coates PTH, Barratt-Boyes SM, Donnenberg AD, Morelli AE, Murphey-Corb M, Thomson AW. Strategies for preclinical evaluation of dendritic cell subsets for promotion of transplant tolerance in the nonhuman primate. Hum Immunol 2002; 63:955-65. [PMID: 12368048 DOI: 10.1016/s0198-8859(02)00457-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
A role for dendritic cells (DC) as critical regulators of immune reactivity has become increasingly recognized. There is evidence in rodent models that donor-derived DC, particularly in the immature state, can prolong organ allograft survival and even induce donor-specific tolerance. To allow the potential tolerogenic properties of these cells to be evaluated more fully with a view to clinical testing, it is necessary to identify DC subsets in nonhuman primates. We have identified the putative rhesus monkey equivalents of circulating human DC subset precursors as lineage(-), HLA-DR(+), CD123(lo),CD11c(hi)(pDC1) and lineage(-), HLA-DR(+), CD123(hi),CD11c(lo)(pDC2). Testing of these DC populations both in vitro and in vivo, as well as in transplant models in combination with conventional or experimental immunosuppressive reagents, will aid the development of novel strategies for the promotion of allo-antigen specific tolerance in transplantation.
Collapse
Affiliation(s)
- P Toby H Coates
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh, 200 Lathrop Street, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
44
|
Messmer D, Bromberg J, Devgan G, Jacqué JM, Granelli-Piperno A, Pope M. Human immunodeficiency virus type 1 Nef mediates activation of STAT3 in immature dendritic cells. AIDS Res Hum Retroviruses 2002; 18:1043-50. [PMID: 12396456 DOI: 10.1089/08892220260235407] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Replication of immunodeficiency viruses (HIV-1 and SIV) in immature dendritic cell (DC)-T cell cocultures is dependent on Nef. In contrast, mature DCs promote the replication of wild-type and nef-defective SIV in concert with CD4(+) T cells. Transcription factor activation occurs on DC maturation and this study aimed to investigate whether Nef triggers similar events in immature DCs, rendering them more like mature DCs. Recombinant HIV nef-expressing adenovirus was used to selectively introduce nef into immature human or macaque DCs. These data provide the first evidence that the expression of HIV nef in immature DCs induced selective activation of STAT3 and, to a lesser extent, NF-kappaB. This highlights how Nef can signal primary immature DCs, suggesting one way in which Nef may modulate immature DCs to drive virus replication in the DC-T cell milieu.
Collapse
Affiliation(s)
- D Messmer
- North Shore LIJ Research Institute, Manhasset, New York 11030, USA
| | | | | | | | | | | |
Collapse
|
45
|
Guo J, Wang B, Zhang M, Chen T, Yu Y, Regulier E, Homann HE, Qin Z, Ju DW, Cao X. Macrophage-derived chemokine gene transfer results in tumor regression in murine lung carcinoma model through efficient induction of antitumor immunity. Gene Ther 2002; 9:793-803. [PMID: 12040461 DOI: 10.1038/sj.gt.3301688] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2001] [Accepted: 01/28/2002] [Indexed: 11/09/2022]
Abstract
Chemokine gene transfer represents a promising approach in the treatment of malignancies. Macrophage-derived chemokine (MDC) (CCL22) belongs to the CC chemokine family and is a strong chemoattractant for dendritic cells (DC), NK cells and T cells. Using adenoviral vectors, human MDC gene was transferred in vivo to investigate its efficacy to induce an antitumor response and to determine the immunologic mechanisms involved. We observed that intratumoral injection of recombinant adenovirus encoding human MDC (AdMDC) resulted in marked tumor regression in a murine model with pre-established subcutaneous 3LL lung carcinoma and induced significant CTL activity. The antitumor response was demonstrated to be CD4+ T cell- and CD8+ T cell-dependent. Administration of AdMDC induced chemoattraction of DC to the tumor site, facilitated DC migration to draining lymph nodes or spleen, and finally activated DC to produce high levels of IL-12. Furthermore, a significant increase of IL-4 production within the tumors was observed early after the AdMDC administration and was followed by the increase of IL-12 and IL-2 production. The levels of IL-2, IL-12 and IFN-gamma in serum, lymph nodes and spleen were also found to be higher in mice treated with AdMDC as compared with that in AdLacZ- or PBS-treated mice. The antitumor response induced by AdMDC was markedly impaired in IL-4 knockout mice, suggesting an important role of IL-4 in the induction of antitumor immunity by MDC. These results suggest that MDC gene transfer might elicit significant antitumor effects through efficient induction of antitumor immunity and might be of therapeutic potentials for cancer.
Collapse
Affiliation(s)
- J Guo
- Institute of Immunology, Second Military Medical University, Shanghai, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Frank I, Piatak M, Stoessel H, Romani N, Bonnyay D, Lifson JD, Pope M. Infectious and whole inactivated simian immunodeficiency viruses interact similarly with primate dendritic cells (DCs): differential intracellular fate of virions in mature and immature DCs. J Virol 2002; 76:2936-51. [PMID: 11861860 PMCID: PMC135959 DOI: 10.1128/jvi.76.6.2936-2951.2002] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
As potential targets for human immunodeficiency virus type 1 and simian immunodeficiency virus (HIV-1 and SIV), dendritic cells (DCs) likely play a significant role in the onset and spread of infection as well as in the induction of antiviral immunity. Using the SIV-macaque system to study the very early events in DC-virus interactions, we compared chemically inactivated SIV having conformationally and functionally intact envelope glycoproteins (2,2'-dithiodipyridine [AT-2] SIV) to infectious and heat-treated SIV. Both human and macaque DCs interact similarly with SIV without detectable effects on DC viability, phenotype, or endocytic function. As assessed by measuring cell-associated viral RNA, considerable amounts of virus are captured by the DCs and this is reduced when the virus is heat treated or derived from a strain that expresses low levels of envelope glycoprotein. Immunostaining for SIV proteins and electron microscopy indicated that few intact virus particles are retained at the periphery of the endocytically active, immature DCs. This contrasts with a perinuclear localization of numerous virions in large vesicular compartments deeper within mature DCs (in which macropinocytosis is down-regulated). Both immature and mature DCs are capable of clathrin-coated pit-mediated uptake of SIV, supporting the notion that the receptor-mediated uptake of virus can occur readily in mature DCs. While large numbers of whole viruses were preferentially found in mature DCs, both immature and mature DCs contained similar amounts of viral RNA, suggesting that different uptake/virus entry mechanisms are active in immature and mature DCs. These findings have significant implications for cell-to-cell transmission of HIV-1 and SIV and support the use of AT-2 SIV, an authentic but noninfectious form of virus, as a useful tool for studies of processing and presentation of AT-2 SIV antigens by DCs.
Collapse
Affiliation(s)
- I Frank
- Laboratory of Cellular Physiology and Immunology, The Rockefeller University, New York, New York, 10021, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Wu L, Bashirova AA, Martin TD, Villamide L, Mehlhop E, Chertov AO, Unutmaz D, Pope M, Carrington M, KewalRamani VN. Rhesus macaque dendritic cells efficiently transmit primate lentiviruses independently of DC-SIGN. Proc Natl Acad Sci U S A 2002; 99:1568-73. [PMID: 11818554 PMCID: PMC122231 DOI: 10.1073/pnas.032654399] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2001] [Accepted: 12/07/2001] [Indexed: 11/18/2022] Open
Abstract
Here, we describe the isolation and characterization of the rhesus macaque homolog for human DC-SIGN, a dendritic cell-specific C-type lectin. mac-DC-SIGN is 92% identical to hu-DC-SIGN. mac-DC-SIGN preserves the virus transmission function of hu-DC-SIGN, capturing and efficiently transducing simian and human immunodeficiency virus to target CD4(+) T cells. Surprisingly, however, mac-DC-SIGN plays no discernable role in the ability of rhesus macaque dendritic cells to capture and transmit primate lentiviruses. Expression and neutralization analyses suggest that this process is DC-SIGN independent in macaque, although the participation of other lectin molecules cannot be ruled out. The ability of primate lentiviruses to effectively use human and rhesus dendritic cells in virus transmission without the cells becoming directly infected suggests that these viruses have taken advantage of a conserved dendritic cell mechanism in which DC-SIGN family molecules are significant contributors but not the only participants.
Collapse
Affiliation(s)
- Li Wu
- HIV Drug Resistance Program, Laboratory of Genomic Diversity, and Basic Research Program, Science Applications International Corporation Frederick, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Mehlhop E, Villamide LA, Frank I, Gettie A, Santisteban C, Messmer D, Ignatius R, Lifson JD, Pope M. Enhanced in vitro stimulation of rhesus macaque dendritic cells for activation of SIV-specific T cell responses. J Immunol Methods 2002; 260:219-34. [PMID: 11792391 DOI: 10.1016/s0022-1759(01)00544-0] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The macaque-simian immunodeficiency virus (SIV) system is one of the best animal models available to study the role of dendritic cells (DCs) in transmission and pathogenesis of HIV, as well as to test DC-based vaccine and therapeutic strategies. To better define and optimize this system, the responsiveness of macaque monocyte-derived DCs to a variety of maturation stimuli was examined. Characteristic immunophenotypic and functional DC maturation induced by standard monocyte conditioned medium (MCM) was compared to the activation induced by a panel of stimuli including soluble CD40L, LPS, Poly I:C, PGE(2)/TNFalpha, and a cocktail mixture of PGE(2)/TNFalpha/IL-1beta/IL-6. Immunophenotypic analysis confirmed that all stimuli induced stable up-regulation of CD25, CD40, CD80, CD83, CD86, HLA-DR, DC-LAMP (CD208), and DEC-205 (CD205). In general, macaque DCs exhibited weaker responses to LPS and Poly I:C than human DCs, and soluble CD40L stimulation induced variable expression of CD25. Interestingly, while the endocytic capacity of CD40L-matured cells was down-modulated comparably to DCs matured with MCM or the cocktail, the T cell stimulatory activity was not enhanced to the same extent. The particularly reproducible and potent T cell stimulatory capacity of cocktail-treated DCs correlated with a more homogenous mature DC phenotype, consistently high levels of IL-12 production, and better viability upon reculture compared to DCs activated by other stimuli. Furthermore, cocktail-matured DCs efficiently captured and presented inactivated SIV to SIV-primed T cells in vitro. Thus, the cocktail represents a particularly potent and useful stimulus for the generation of efficacious immunostimulatory macaque DCs.
Collapse
Affiliation(s)
- Erin Mehlhop
- Laboratory of Cellular Physiology and Immunology, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Krueger JG. The immunologic basis for the treatment of psoriasis with new biologic agents. J Am Acad Dermatol 2002; 46:1-23; quiz 23-6. [PMID: 11756941 DOI: 10.1067/mjd.2002.120568] [Citation(s) in RCA: 387] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Psoriasis vulgaris is the most prevalent T-cell-mediated inflammatory disease in humans. The pathogenesis of psoriasis is linked to activation of several types of leukocytes that control cellular immunity and to a T-cell-dependent inflammatory process in skin that accelerates the growth of epidermal and vascular cells in psoriasis lesions. Critical steps in immunologic activation include Langerhans cell maturation (activation), T-cell activation, differentiation and expansion of type 1 T cells, selective trafficking of activated T cells to skin, and induction of an inflammatory cytokine and chemokine cascade in skin lesions. In turn, each of these steps offers an opportunity for intervention with engineered biologic therapeutics.
Collapse
Affiliation(s)
- James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY 10021-6399, USA.
| |
Collapse
|
50
|
Pichyangkul S, Saengkrai P, Yongvanitchit K, Limsomwong C, Gettayacamin M, Walsh DS, Stewart VA, Ballou WR, Heppner DG. Isolation and characterization of rhesus blood dendritic cells using flow cytometry. J Immunol Methods 2001; 252:15-23. [PMID: 11334961 DOI: 10.1016/s0022-1759(01)00327-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Recognition of dendritic cells (DCs) as initiators and modulators of immune responses and growing use of rhesus monkeys for the preclinical optimization of vaccine formulations prompted characterization of the phenotype and function of isolated rhesus peripheral blood DCs. We developed a flow cytometric method to directly identify and isolate DCs from rhesus peripheral blood whereby a T cell depleted population negative for CD3, CD14, CD16 and CD20 but positive for CD83 yielded a cell population with surface markers, morphology, and a cytokine profile similar to human myeloid DCs. Rhesus blood DCs were more effective than monocytes and B cells in mixed lymphocyte reactions and in the presentation of recombinant malaria blood stage antigen MSP-1((42)) to autologous T cells. The ability to isolate rhesus blood DC from peripheral blood should be a useful tool for immunological investigations.
Collapse
Affiliation(s)
- S Pichyangkul
- Department of Immunology and Medicine, US Army Medical Component, US Armed Forces Research Institute of Medical Science (AFRIMS), 315/6 Rajvithi Road, 10400, Bangkok, Thailand.
| | | | | | | | | | | | | | | | | |
Collapse
|