1
|
Abstract
CpG Oligonucleotides (ODN) are immunomodulatory synthetic oligonucleotides specifically designed to stimulate Toll-like receptor 9. TLR9 is expressed on human plasmacytoid dendritic cells and B cells and triggers an innate immune response characterized by the production of Th1 and pro-inflammatory cytokines. This chapter reviews recent progress in understanding the mechanism of action of CpG ODN and provides an overview of human clinical trial results using CpG ODN to improve vaccines for the prevention/treatment of cancer, allergy, and infectious disease.
Collapse
Affiliation(s)
| | | | - Dennis M Klinman
- National Cancer Institute, NIH, Frederick, MD, USA.
- Leitman Klinman Consulting, Potomac, MD, USA.
| |
Collapse
|
2
|
Montamat G, Leonard C, Poli A, Klimek L, Ollert M. CpG Adjuvant in Allergen-Specific Immunotherapy: Finding the Sweet Spot for the Induction of Immune Tolerance. Front Immunol 2021; 12:590054. [PMID: 33708195 PMCID: PMC7940844 DOI: 10.3389/fimmu.2021.590054] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 01/04/2021] [Indexed: 01/16/2023] Open
Abstract
Prevalence and incidence of IgE-mediated allergic diseases have increased over the past years in developed and developing countries. Allergen-specific immunotherapy (AIT) is currently the only curative treatment available for allergic diseases that has long-term efficacy. Although AIT has been proven successful as an immunomodulatory therapy since its beginnings, it still faces several unmet needs and challenges today. For instance, some patients can experience severe side effects, others are non-responders, and prolonged treatment schedules can lead to lack of patient adherence and therapy discontinuation. A common strategy to improve AIT relies on the use of adjuvants and immune modulators to boost its effects and improve its safety. Among the adjuvants tested for their clinical efficacy, CpG oligodeoxynucleotide (CpG-ODN) was investigated with limited success and without reaching phase III trials for clinical allergy treatment. However, recently discovered immune tolerance-promoting properties of CpG-ODN place this adjuvant again in a prominent position as an immune modulator for the treatment of allergic diseases. Indeed, it has been shown that the CpG-ODN dose and concentration are crucial in promoting immune regulation through the recruitment of pDCs. While low doses induce an inflammatory response, high doses of CpG-ODN trigger a tolerogenic response that can reverse a pre-established allergic milieu. Consistently, CpG-ODN has also been found to stimulate IL-10 producing B cells, so-called B regulatory cells (Bregs). Accordingly, CpG-ODN has shown its capacity to prevent and revert allergic reactions in several animal models showing its potential as both preventive and active treatment for IgE-mediated allergy. In this review, we describe how CpG-ODN-based therapies for allergic diseases, despite having shown limited success in the past, can still be exploited further as an adjuvant or immune modulator in the context of AIT and deserves additional attention. Here, we discuss the past and current knowledge, which highlights CpG-ODN as a potential adjuvant to be reevaluated for the enhancement of AIT when used in appropriate conditions and formulations.
Collapse
Affiliation(s)
- Guillem Montamat
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Cathy Leonard
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Aurélie Poli
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Ludger Klimek
- Centre for Rhinology and Allergology, Wiesbaden, Germany
| | - Markus Ollert
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg.,Department of Dermatology and Allergy Centre, Odense University Hospital, Odense, Denmark
| |
Collapse
|
3
|
Seyfizadeh N, Muthuswamy R, Mitchell DA, Nierkens S, Seyfizadeh N. Migration of dendritic cells to the lymph nodes and its enhancement to drive anti-tumor responses. Crit Rev Oncol Hematol 2016; 107:100-110. [PMID: 27823637 DOI: 10.1016/j.critrevonc.2016.09.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 12/29/2022] Open
Abstract
Better prognoses associated with increased T cell infiltration of tumors, as seen with chimeric antigen receptor (CAR) T cell therapies and immune checkpoint inhibitors, portray the importance and potential of the immune system in controlling tumors. This has rejuvenated the field of cancer immunotherapy leading to an increasing number of immunotherapies developed for cancer patients. Dendritic Cells (DCs) vaccines represent an appealing option for cancer immunotherapy since DCs have the ability to circumvent tolerance to tumors by its adjuvant properties and to induce memory T cells that can become persistent after initial tumor clearance to engage potential metastatic tumors. In the past, DC-based cancer vaccines have elicited only poor clinical response in cancer patients, which can be attributed to complex and a multitude of issues associated with generation, implementing, delivery of DC vaccine and their potential interaction with effector cells. The current review mainly focuses on migration/trafficking of DCs, as one of the key issues that affect the success of DC-based cancer vaccines, and discusses strategies to enhance it for cancer immunotherapy. Additionally, impact of maturation, route of DC delivery and negative effects of tumor microenvironment (TME) on DC homing to LN are reviewed. Moreover, strategies to increase the expression of genes involved in Lymph node homing, preconditioning of the vaccination site, enhancing lymph node ability to attract and receive DCs, while limiting negative impact of TME on DC migration are discussed.
Collapse
Affiliation(s)
- Narges Seyfizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Duane A Mitchell
- Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Stefan Nierkens
- Laboratory of Translational Immunology, U-DAIR, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nayer Seyfizadeh
- Umbilical Cord Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Clinical Biochemistry and Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
4
|
The effects of subcutaneous and intraocular administration of class B ODN CpG in chicken on the expression of TLR21, IFN-γ and IL-1β. Pol J Vet Sci 2015; 17:593-9. [PMID: 25638972 DOI: 10.2478/pjvs-2014-0089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The synthetic unmethylated oligodeoxynucleotides with CpG motifs (CpG ODN) were shown to activate Toll-like receptor 21 (TLR21) and stimulate the innate and adaptive immune system. In this study we tested the expression of TLR21, interferon (IFN)-γ and interleukin (IL)-1β mRNA in the blood after subcutaneous and intraocular application of the class B CpG ODN in chicken. The relative expression of mRNA of TLR21, IFN-γ and IL-1β were quantified at 3, 6, 12, 24 and 72 h post-stimulation. The study revealed that IFN-γ mRNA expression was significantly upregulated 12 h after subcutaneous stimulation with a high and low dose of ODN CpG, whereas the IL-1β mRNA expression levels were significantly upregulated 3 and 72 h after subcutaneous administration. After intraocular administration, the IL-1β mRNA levels were the highest at 24 h post-application, albeit not specifically. This data indicates that class B CpG ODN has the ability to induce TLR21 response in blood when administered parenterally in chicken. In contrast, intraocular administration of CpG ODN was not able to produce a significant increase in cytokine mRNA expression in blood. The data suggest that additional stimulus, e.g. the antigen, may be needed on the site of mucosal administration to activate systemic immune response.
Collapse
|
5
|
Parlet CP, Schlueter AJ. Mechanisms by which chronic ethanol feeding impairs the migratory capacity of cutaneous dendritic cells. Alcohol Clin Exp Res 2013; 37:2098-107. [PMID: 23895590 DOI: 10.1111/acer.12201] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 04/22/2013] [Indexed: 01/18/2023]
Abstract
BACKGROUND Chronic alcoholism is associated with increased incidence and severity of skin infection. Cutaneous dendritic cells (CDCs) play a pivotal role in skin immunity, and chronic ethanol (EtOH) feeding in mice has been shown to inhibit CDC migration to skin-draining lymph nodes (dLNs) following epicutaneous sensitization. Because CDC subsets differentially initiate T-cell responses, it is important to determine how EtOH feeding affects migration of each subset and identify mechanisms responsible for observed defects. METHODS Mice received EtOH in the drinking water for ≥ 16 weeks. Baseline numbers of CDC subsets and their migration to the dLNs following fluorescein 5-isothiocyanate (FITC) sensitization were assessed by flow cytometry. Epidermal cell suspension and skin explant cultures were used to measure the impact of EtOH upon molecules that influence CDC migration. Cytokine arrays performed on explant culture supernatants assessed local production of inflammatory cytokines. RESULTS Chronic EtOH feeding reduced migration of all CDC subsets to the dLNs following FITC sensitization. Reduced migration of dermal-resident CDCs did not correspond with reduced baseline numbers of these cells. For Langerhans cells (LCs), EtOH-induced migratory dysfunction corresponded with delayed down-regulation of E-cadherin, chemokine receptor 1 (CCR1), and CCR6 and impaired up-regulation of matrix metalloproteinases (MMPs) 2 and 9. In skin explant assays, EtOH blunted CDC mobilization following stimulation with CCL21/CPG 1826. No alteration in CD54 or CCR7 expression was observed, but production of skin-derived tumor necrosis factor alpha (TNF-α) was reduced. Poor migratory responses in vitro could be improved by supplementing explant cultures from EtOH-fed mice with TNF-α. CONCLUSIONS Chronic EtOH consumption does not alter baseline dermal-resident CDC numbers. However, like LCs, migratory responsiveness of dermal CDCs was decreased following FITC sensitization. Inefficient down-regulation of both CCRs and adhesion molecules and the inability to up-regulate MMPs indicate that EtOH impedes LC acquisition of a promigratory phenotype. These defects, combined with improvement of the migratory defect with in vitro TNF-α replacement, demonstrate intrinsic as well as environmental contributions to defective CDC migration. These findings provide novel mechanisms to explain the observed increased incidence and severity of skin infections in chronic alcoholics.
Collapse
Affiliation(s)
- Corey P Parlet
- Department of Pathology and Interdisciplinary Graduate Program in Immunology , University of Iowa Carver College of Medicine, Iowa City, Iowa
| | | |
Collapse
|
6
|
Pearton M, Saller V, Coulman SA, Gateley C, Anstey AV, Zarnitsyn V, Birchall JC. Microneedle delivery of plasmid DNA to living human skin: Formulation coating, skin insertion and gene expression. J Control Release 2012; 160:561-9. [PMID: 22516089 DOI: 10.1016/j.jconrel.2012.04.005] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Revised: 03/21/2012] [Accepted: 04/02/2012] [Indexed: 10/28/2022]
Abstract
Microneedle delivery of nucleic acids, in particular plasmid DNA (pDNA), to the skin represents a potential new approach for the clinical management of genetic skin diseases and cutaneous cancers, and for intracutaneous genetic immunisation. In this study excised human skin explants were used to investigate and optimise key parameters that will determine stable and effective microneedle-facilitated pDNA delivery. These include (i) high dose-loading of pDNA onto microneedle surfaces, (ii) stability and functionality of the coated pDNA, (iii) skin penetration capability of pDNA-coated microneedles, and (iv) efficient gene expression in human skin. Optimisation of a dip-coating method enabled significant increases in the loading capacity, up to 100μg of pDNA per 5-microneedle array. Coated microneedles were able to reproducibly perforate human skin at low (<1N) insertion forces. The physical stability of the coated pDNA was partially compromised on storage, although this was improved through the addition of saccharide excipients without detriment to the biological functionality of pDNA. The pDNA-coated microneedles facilitated reporter gene expression in viable human skin. The efficiency of gene expression from coated microneedles will depend upon suitable DNA loading, efficient and reproducible skin puncture and rapid in situ dissolution of the plasmid at the site of delivery.
Collapse
Affiliation(s)
- Marc Pearton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | | | | | | | | | | | | |
Collapse
|
7
|
Bal SM, Slütter B, Verheul R, Bouwstra JA, Jiskoot W. Adjuvanted, antigen loaded N-trimethyl chitosan nanoparticles for nasal and intradermal vaccination: adjuvant- and site-dependent immunogenicity in mice. Eur J Pharm Sci 2011; 45:475-81. [PMID: 22009113 DOI: 10.1016/j.ejps.2011.10.003] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 09/30/2011] [Accepted: 10/01/2011] [Indexed: 10/17/2022]
Abstract
N-trimethyl chitosan (TMC) nanoparticles have been shown to increase the immunogenicity of subunit antigens after nasal and intradermal administration. This work describes a second generation of TMC nanoparticles containing ovalbumin as a model antigen (TMC/OVA nanoparticles) and an immunopotentiator (TMC/OVA/immunopotentiator nanoparticles). The selection of immunopotentiators included Toll-like receptor (TLR) ligands lipopolysaccharide (LPS), PAM(3)CSK(4) (PAM), CpG DNA, the NOD-like receptor 2 ligand muramyl dipeptide (MDP) and the GM1 ganglioside receptor ligand, cholera toxin B subunit (CTB). The TMC/OVA/immunopotentiator nanoparticles were characterised physico-chemically and their immunogenicity was assessed by determining the serum IgG, IgG1, IgG2a titres and secretory IgA levels in nasal washes after intradermal and nasal vaccination in mice. After nasal vaccination, TMC/OVA nanoparticles containing LPS or MDP elicited higher IgG, IgG1 and sIgA levels than non-adjuvanted TMC/OVA particles, whereas nanoparticles containing CTB, PAM or CpG did not. After intradermal vaccination, the TMC/OVA/CpG and TMC/OVA/LPS nanoparticles provoked higher IgG titres than plain TMC/OVA particles. Altogether, our results show that co-encapsulation of an additional immunopotentiator with the antigen into TMC nanoparticles can further improve the immunogenicity of the vaccine. However, the strength and quality of the response depends on the immunopotentiator as well as the route of administration.
Collapse
Affiliation(s)
- Suzanne M Bal
- Division of Drug Delivery Technology, Leiden/Amsterdam Center for Drug Research, Leiden University, Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
8
|
Ruutu MP, Chen X, Joshi O, Kendall MA, Frazer IH. Increasing mechanical stimulus induces migration of Langerhans cells and impairs the immune response to intracutaneously delivered antigen. Exp Dermatol 2011; 20:534-6. [DOI: 10.1111/j.1600-0625.2010.01234.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
9
|
Kigasawa K, Kajimoto K, Nakamura T, Hama S, Kanamura K, Harashima H, Kogure K. Noninvasive and efficient transdermal delivery of CpG-oligodeoxynucleotide for cancer immunotherapy. J Control Release 2011; 150:256-65. [PMID: 21256903 DOI: 10.1016/j.jconrel.2011.01.018] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2010] [Revised: 11/30/2010] [Accepted: 01/17/2011] [Indexed: 12/13/2022]
Abstract
Oligodeoxynucleotides containing unmethylated cytosine-phosphate-guanosine motifs (CpG-ODN) possess immunostimulatory effects and potential antitumor activity. Since the skin is an easily available site of administration of CpG-ODN due to its accessibility and the presence of abundant antigen presenting cells, it is expected that the application of CpG-ODN to the skin would induce systemic immune response and antitumor activity. However, it is difficult to deliver hydrophilic macromolecules including CpG-ODN through the skin. We have previously demonstrated that small interfering RNA (siRNA) was efficiently delivered into rat epidermis by iontophoresis. In this report, we investigate the effect of transdermal iontophoretic delivery of CpG-ODN on the induction of immune responses and antitumor activity against B16F1 melanoma in mice. Iontophoresis promoted CpG-ODN delivery into the epidermis and dermis. Furthermore, iontophoretic delivery of CpG-ODN to the skin induced the expression of proinflammatory and Th1-type cytokines in the skin and draining lymph node. Finally, transdermal iontophoretic delivery of CpG-ODN led to antitumor activity against B16F1 melanoma. Interestingly, the CpG-ODN administration site is not restricted to the tumor area. In conclusion, CpG-ODN delivered transdermally induced potent antitumor activity, and our system is expected to serve as a simple and noninvasive approach for cancer immunotherapy.
Collapse
Affiliation(s)
- Kaoru Kigasawa
- Graduate School of Life Science, Hokkaido University, Kita 12 Nishi 6, Kita-ku, Sapporo-City, Hokkaido 060-0812 Japan
| | | | | | | | | | | | | |
Collapse
|
10
|
Bal SM, Ding Z, van Riet E, Jiskoot W, Bouwstra JA. Advances in transcutaneous vaccine delivery: Do all ways lead to Rome? J Control Release 2010; 148:266-82. [DOI: 10.1016/j.jconrel.2010.09.018] [Citation(s) in RCA: 141] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 09/13/2010] [Indexed: 01/09/2023]
|
11
|
Lai MD, Chen CS, Yang CR, Yuan SY, Tsai JJ, Tu CF, Wang CC, Yen MC, Lin CC. An HDAC inhibitor enhances the antitumor activity of a CMV promoter-driven DNA vaccine. Cancer Gene Ther 2009; 17:203-11. [PMID: 19851354 DOI: 10.1038/cgt.2009.65] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The cytomegalovirus (CMV) promoter is considered to be one of the strongest promoters for driving the in vivo expression of genes encoded by DNA vaccines. However, the efficacy of DNA vaccines has so far been disappointing (particularly in humans), and this might be explained in part by histone deacetylase (HDAC)-mediated chromatin condensation. Hence, we sought to investigate whether increasing the expression of DNA vaccine antigens with the HDAC inhibitor OSU-HDAC42 would enhance the efficacy of DNA vaccines in vivo. A luciferase assay was used to determine the effects of OSU-HDAC42 on CMV promoter-driven DNA plasmids in vitro and in vivo. Three HDAC inhibitors were able to activate expression from the CMV promoter in NIH3T3 cells and MBT-2 bladder cancer cells. The expression of luciferase was significantly enhanced by co-administration of pCMV-luciferase and OSU-HDAC42 in mice. To explore whether OSU-HDAC42 could enhance the specific antitumor activity of a neu DNA vaccine driven by the CMV promoter, we evaluated therapeutic effects and immune responses in a mouse tumor natively overexpressing HER2/neu. Mice receiving OSU-HDAC42 in combination with the CMV-promoter neu DNA vaccine exhibited stronger antitumor effects than mice given the DNA vaccine only. In addition, a correlation between the antitumor effects and the specific cellular immune responses was observed in the mice receiving the DNA vaccine and OSU-HDAC42.
Collapse
Affiliation(s)
- M-D Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Ng KW, Pearton M, Coulman S, Anstey A, Gateley C, Morrissey A, Allender C, Birchall J. Development of an ex vivo human skin model for intradermal vaccination: tissue viability and Langerhans cell behaviour. Vaccine 2009; 27:5948-55. [PMID: 19679220 PMCID: PMC2753709 DOI: 10.1016/j.vaccine.2009.07.088] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Revised: 06/29/2009] [Accepted: 07/23/2009] [Indexed: 01/16/2023]
Abstract
The presence of resident Langerhans cells (LCs) in the epidermis makes the skin an attractive target for DNA vaccination. However, reliable animal models for cutaneous vaccination studies are limited. We demonstrate an ex vivo human skin model for cutaneous DNA vaccination which can potentially bridge the gap between pre-clinical in vivo animal models and clinical studies. Cutaneous transgene expression was utilised to demonstrate epidermal tissue viability in culture. LC response to the culture environment was monitored by immunohistochemistry. Full-thickness and split-thickness skin remained genetically viable in culture for at least 72 h in both phosphate-buffered saline (PBS) and full organ culture medium (OCM). The epidermis of explants cultured in OCM remained morphologically intact throughout the culture duration. LCs in full-thickness skin exhibited a delayed response (reduction in cell number and increase in cell size) to the culture conditions compared with split-thickness skin, whose response was immediate. In conclusion, excised human skin can be cultured for a minimum of 72 h for analysis of gene expression and immune cell activation. However, the use of split-thickness skin for vaccine formulation studies may not be appropriate because of the nature of the activation. Full-thickness skin explants are a more suitable model to assess cutaneous vaccination ex vivo.
Collapse
Affiliation(s)
- Keng Wooi Ng
- Gene Delivery Research Group, Welsh School of Pharmacy, Cardiff University, Cardiff CF10 3NB, UK
| | - Marc Pearton
- Gene Delivery Research Group, Welsh School of Pharmacy, Cardiff University, Cardiff CF10 3NB, UK
| | - Sion Coulman
- Gene Delivery Research Group, Welsh School of Pharmacy, Cardiff University, Cardiff CF10 3NB, UK
| | - Alexander Anstey
- Gwent Healthcare NHS Trust, Royal Gwent Hospital, Cardiff Road, Newport NP20 2UB, UK
| | - Christopher Gateley
- Gwent Healthcare NHS Trust, Royal Gwent Hospital, Cardiff Road, Newport NP20 2UB, UK
| | - Anthony Morrissey
- Biomedical Microsystems Team, Tyndall National Institute, Prospect Row, Cork, Ireland
| | - Christopher Allender
- Gene Delivery Research Group, Welsh School of Pharmacy, Cardiff University, Cardiff CF10 3NB, UK
| | - James Birchall
- Gene Delivery Research Group, Welsh School of Pharmacy, Cardiff University, Cardiff CF10 3NB, UK
| |
Collapse
|
13
|
Cognasse F, Hamzeh-Cognasse H, Garraud O. [Platelets "Toll-like receptor" engagement stimulates the release of immunomodulating molecules]. Transfus Clin Biol 2008; 15:139-47. [PMID: 18842435 DOI: 10.1016/j.tracli.2008.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Accepted: 07/30/2008] [Indexed: 01/14/2023]
Abstract
Platelets are nonnucleated cellular elements that play a role in the process of haemostasis, and also in various ways in innate immunity and in inflammation. Platelets also contain numerous secretory products and can exert critical roles in several aspects of haemostasis. In addition, they house and secrete a variety of cytokines, chemokines and associated molecules which behave as ligands for receptors/counterparts displayed by endothelial cells lining tissue vessels and most leukocyte subsets. These latter studies show that platelets have an important role in innate as well as adaptive immunity; thus platelets can take part in an immune directive response. Moreover, platelets display receptors for several types of cytokines/chemokines along with FcgammaRII receptors. Finally, platelets not only express a variety of Toll-like receptors, with recently identified functions or not as-yet fully identified, but have also been demonstrated to express the key tandem pair of inflammatory and antigen presentation molecules (CD40 and CD40-ligand/CD154), this latter function making them the major purveyors of soluble CD40L in the plasma. It appears that platelets may be regarded as one of the neglected components of immune cell regulators, and platelets contribute to some interesting aspects in bridging innate and adaptive immunity. We propose that platelets discriminate danger signals and adapt the subsequent responses, with polarized cytokine secretion. Platelets may recognize several types of infectious pathogens and limit microbial colonization by sequestering these pathogens and releasing immunomodulatory factors. This review allows us to re-explore indications that platelets exert direct anti-infection immunity and we will present experimentally-driven arguments in favour of a role of platelet TLR in regulating certain immune activities.
Collapse
Affiliation(s)
- F Cognasse
- EFS Auvergne-Loire, French Regional Blood Bank, Saint-Etienne, France.
| | | | | |
Collapse
|
14
|
Sudowe S, Dominitzki S, Montermann E, Bros M, Grabbe S, Reske-Kunz AB. Uptake and presentation of exogenous antigen and presentation of endogenously produced antigen by skin dendritic cells represent equivalent pathways for the priming of cellular immune responses following biolistic DNA immunization. Immunology 2008; 128:e193-205. [PMID: 18800984 DOI: 10.1111/j.1365-2567.2008.02947.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Gene gun-mediated biolistic DNA vaccination with beta-galactosidase (betaGal)-encoding plasmid vectors efficiently modulated antigen-induced immune responses in an animal model of type I allergy, including the inhibition of immunoglobulin E (IgE) production. Here we show that CD4(+) as well as CD8(+) T cells from mice biolistically transfected with a plasmid encoding betaGal under the control of the fascin promoter (pFascin-betaGal) are capable of inhibiting betaGal-specific IgE production after adoptive transfer into naïve recipients. Moreover, suppression of IgE production was dependent on interferon (IFN)-gamma. To analyse the modalities of activation of CD4(+) and CD8(+) T cells regarding the localization of antigen synthesis following gene gun-mediated DNA immunization, we used the fascin promoter and the keratin 5 promoter (pK5-betaGal) to direct betaGal production mainly to dendritic cells (DCs) and to keratinocytes, respectively. Gene gun-mediated DNA immunization with each vector induced considerable activation of betaGal-specific CD8(+) cytotoxic T cells. Cytokine production by re-stimulated CD4(+) T cells in draining lymph nodes and immunoglobulin isotype profiles in sera of immunized mice indicated that immunization with pFascin-betaGal induced a T helper type 1 (Th1)-biased immune response, whereas immunization with pK5-betaGal generated a mixed Th1/Th2 immune response. Nevertheless, DNA vaccination with pFascin-betaGal and pK5-betaGal, respectively, efficiently inhibited specific IgE production in the mouse model of type I allergy. In conclusion, our data show that uptake of exogenous antigen produced by keratinocytes and its presentation by untransfected DCs as well as the presentation of antigen synthesized endogenously in DCs represent equivalent pathways for efficient priming of cellular immune responses.
Collapse
Affiliation(s)
- Stephan Sudowe
- Department of Dermatology, Johannes Gutenberg-University Mainz, Mainz, Germany.
| | | | | | | | | | | |
Collapse
|
15
|
Verdijk P, Aarntzen EHJG, Punt CJA, de Vries IJM, Figdor CG. Maximizing dendritic cell migration in cancer immunotherapy. Expert Opin Biol Ther 2008; 8:865-74. [PMID: 18549318 DOI: 10.1517/14712598.8.7.865] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND The success of dendritic cell (DC)-based immunotherapy in inducing cellular immunity against tumors is highly dependent on accurate delivery and trafficking of the DC to T-cell-rich areas of secondary lymphoid tissues. OBJECTIVE To provide an overview of DC migration in vivo and how migration to peripheral lymph nodes might be improved to optimize DC therapy. METHODS We focused on DC migration in preclinical models and human skin explants and on clinical vaccination trials studying migration of in vitro-generated DC. RESULTS/CONCLUSIONS DC migration requires an intricate interplay between the cell and its environment. To maximize migration for cellular therapy, it is important to optimize the generation of migratory DC as well as treatment strategies.
Collapse
Affiliation(s)
- Pauline Verdijk
- Radboud University Nijmegen Medical Centre, Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Geert Grooteplein 28, Nijmegen, The Netherlands
| | | | | | | | | |
Collapse
|
16
|
Ness KJ, Fan J, Wilke WW, Coleman RA, Cook RT, Schlueter AJ. Chronic ethanol consumption decreases murine Langerhans cell numbers and delays migration of Langerhans cells as well as dermal dendritic cells. Alcohol Clin Exp Res 2008; 32:657-68. [PMID: 18241312 PMCID: PMC2276693 DOI: 10.1111/j.1530-0277.2007.00614.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Chronic alcoholics experience increased incidence and severity of infections, the mechanism of which is incompletely understood. Dendritic cells (DC) migrate from peripheral locations to lymph nodes (LN) to initiate adaptive immunity against infection. Little is known about how chronic alcohol exposure affects skin DC numbers or migration. METHODS Mice received 20% EtOH in the drinking water for up to 35 weeks. Baseline Langerhans cell (LC) and dermal DC (dDC) numbers were enumerated by immunofluorescence (IF). LC repopulation after inflammation was determined following congenic bone marrow (BM) transplant and ultraviolet (UV) irradiation. Net LC loss from epidermis was determined by IF following TNF-alpha or CpG stimulation. LC and dDC migration into LN was assessed by flow cytometry following epicutaneous FITC administration. RESULTS Chronic EtOH consumption caused a baseline reduction in LC but not dDC numbers. The deficit was not corrected following transplantation with non-EtOH-exposed BM and UV irradiation, supporting the hypothesis that the defect is intrinsic to the skin environment rather than LC precursors. Net loss of LC from epidermis following inflammation was greatly reduced in EtOH-fed mice versus controls. Ethanol consumption for at least 4 weeks led to delayed LC migration into LN, and consumption for at least 8 weeks led to delayed dDC migration into LN following epicutaneous FITC application. CONCLUSIONS Chronic EtOH consumption causes decreased density of epidermal LC, which likely results in decreased epidermal immunosurveillance. It also results in altered migratory responsiveness and delayed LC and dDC migration into LN, which likely delays activation of adaptive immunity. Decreased LC density at baseline appears to be the result of an alteration in the skin environment rather than an intrinsic LC defect. These findings provide novel mechanisms to at least partially explain why chronic alcoholics are more susceptible to infections, especially those following skin penetration.
Collapse
Affiliation(s)
- Kristin J Ness
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | |
Collapse
|
17
|
Abstract
Aptamers constitute a new class of oligonucleotides that have gained therapeutic importance. With the approval of the first aptamer drug, pegaptanib, interest in this class of oligonucleotides, often referred to as 'chemical antibodies', has increased. This article discusses aptamers in relation to other oligonucleotide molecules such as antisense nucleotides, short inhibitory sequences, ribozymes and so on. The development of pegaptanib is looked at from the point of view of the challenges faced in converting aptamers into therapeutic molecules. Cases of other aptamers, which show promise as drugs, are discussed in slightly greater detail. Comparison with antibodies and small molecules, which have hitherto held monopoly in this area, is also made.
Collapse
Affiliation(s)
- Gurjot Kaur
- National Institute of Pharmaceutical Education and Research (NIPER), Department of Biotechnology, Sector 67, SAS. Nagar, Punjab 160 062, India.
| | | |
Collapse
|
18
|
Ness KJ, Fan J, Wilke WW, Coleman RA, Cook RT, Schlueter AJ. Chronic ethanol consumption decreases murine Langerhans cell numbers and delays migration of Langerhans cells as well as dermal dendritic cells. Alcohol Clin Exp Res 2008. [PMID: 18241312 DOI: 10.1111/j.1530-0277.2007.00614.x.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Chronic alcoholics experience increased incidence and severity of infections, the mechanism of which is incompletely understood. Dendritic cells (DC) migrate from peripheral locations to lymph nodes (LN) to initiate adaptive immunity against infection. Little is known about how chronic alcohol exposure affects skin DC numbers or migration. METHODS Mice received 20% EtOH in the drinking water for up to 35 weeks. Baseline Langerhans cell (LC) and dermal DC (dDC) numbers were enumerated by immunofluorescence (IF). LC repopulation after inflammation was determined following congenic bone marrow (BM) transplant and ultraviolet (UV) irradiation. Net LC loss from epidermis was determined by IF following TNF-alpha or CpG stimulation. LC and dDC migration into LN was assessed by flow cytometry following epicutaneous FITC administration. RESULTS Chronic EtOH consumption caused a baseline reduction in LC but not dDC numbers. The deficit was not corrected following transplantation with non-EtOH-exposed BM and UV irradiation, supporting the hypothesis that the defect is intrinsic to the skin environment rather than LC precursors. Net loss of LC from epidermis following inflammation was greatly reduced in EtOH-fed mice versus controls. Ethanol consumption for at least 4 weeks led to delayed LC migration into LN, and consumption for at least 8 weeks led to delayed dDC migration into LN following epicutaneous FITC application. CONCLUSIONS Chronic EtOH consumption causes decreased density of epidermal LC, which likely results in decreased epidermal immunosurveillance. It also results in altered migratory responsiveness and delayed LC and dDC migration into LN, which likely delays activation of adaptive immunity. Decreased LC density at baseline appears to be the result of an alteration in the skin environment rather than an intrinsic LC defect. These findings provide novel mechanisms to at least partially explain why chronic alcoholics are more susceptible to infections, especially those following skin penetration.
Collapse
Affiliation(s)
- Kristin J Ness
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | |
Collapse
|
19
|
Hovav AH, Panas MW, Rahman S, Sircar P, Gillard G, Cayabyab MJ, Letvin NL. Duration of antigen expression in vivo following DNA immunization modifies the magnitude, contraction, and secondary responses of CD8+ T lymphocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 179:6725-33. [PMID: 17982062 PMCID: PMC2190620 DOI: 10.4049/jimmunol.179.10.6725] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The duration of Ag expression in vivo has been reported to have a minimal impact on both the magnitude and kinetics of contraction of a pathogen-induced CD8(+) T cell response. In this study, we controlled the duration of Ag expression by excising the ear pinnae following intradermal ear pinnae DNA immunization. This resulted in decreased magnitude, accelerated contraction and differentiation, and surprisingly greater secondary CD8(+) T cell responses. Furthermore, we found delayed and prolonged Ag presentation in the immunized mice; however, this presentation was considerably decreased when the depot Ag was eliminated. These findings suggest that the magnitude and the contraction phase of the CD8(+) T cell response following intradermal DNA immunization is regulated by the duration rather than the initial exposure to Ag.
Collapse
Affiliation(s)
- Avi-Hai Hovav
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Michael W. Panas
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Shaila Rahman
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Piya Sircar
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Geoffrey Gillard
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Mark J. Cayabyab
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| | - Norman L. Letvin
- Division of Viral Pathogenesis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
20
|
Abstract
Cutaneous dendritic cells (DC) include epidermal Langerhans cells (LC), interstitial/dermal dendritic cells (DDC), as well as plasmacytoid DC (pDC) that occur under pathological conditions. These immune cells have a spectrum of different functions with implications that extend far beyond the skin. They have the potential to internalize particulate agents and macromolecules, and display migratory properties that endow them with the unique capacity to journey between skin and draining lymph nodes where they encounter antigen-specific T lymphocytes. Herein, we will review the features of human and mouse cutaneous DC, emphasizing characteristics representative of their life-cycle stages that occur within the skin.
Collapse
Affiliation(s)
- Jenny Valladeau
- Université Claude Bernard Lyon I, Dermatologie-Pavillon R, EA 3732, Hopital Ed Herriot, Pav R, 69437 Lyon cedex, France.
| | | |
Collapse
|
21
|
Flacher V, Bouschbacher M, Verronèse E, Massacrier C, Sisirak V, Berthier-Vergnes O, de Saint-Vis B, Caux C, Dezutter-Dambuyant C, Lebecque S, Valladeau J. Human Langerhans cells express a specific TLR profile and differentially respond to viruses and Gram-positive bacteria. THE JOURNAL OF IMMUNOLOGY 2007; 177:7959-67. [PMID: 17114468 DOI: 10.4049/jimmunol.177.11.7959] [Citation(s) in RCA: 192] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Dendritic cells (DC) are APCs essential for the development of primary immune responses. In pluristratified epithelia, Langerhans cells (LC) are a critical subset of DC which take up Ags and migrate toward lymph nodes upon inflammatory stimuli. TLR allow detection of pathogen-associated molecular patterns (PAMP) by different DC subsets. The repertoire of TLR expressed by human LC is uncharacterized and their ability to directly respond to PAMP has not been systematically investigated. In this study, we show for the first time that freshly purified LC from human skin express mRNA encoding TLR1, TLR2, TLR3, TLR5, TLR6 and TLR10. In addition, keratinocytes ex vivo display TLR1-5, TLR7, and TLR10. Accordingly, highly enriched immature LC efficiently respond to TLR2 agonists peptidoglycan and lipoteichoic acid from Gram-positive bacteria, and to dsRNA which engages TLR3. In contrast, LC do not directly sense TLR7/8 ligands and LPS from Gram-negative bacteria, which signals through TLR4. TLR engagement also results in cytokine production, with marked differences depending on the PAMP detected. TLR2 and TLR3 ligands increase IL-6 and IL-8 production, while dsRNA alone stimulates TNF-alpha release. Strikingly, only peptidoglycan triggers IL-10 secretion, thereby suggesting a specific function in tolerance to commensal Gram-positive bacteria. However, LC do not produce IL-12p70 or type I IFNs. In conclusion, human LC are equipped with TLR that enable direct detection of PAMP from viruses and Gram-positive bacteria, subsequent phenotypic maturation, and differential cytokine production. This implies a significant role for LC in the control of skin immune responses.
Collapse
Affiliation(s)
- Vincent Flacher
- Laboratory for Immunological Research, Schering-Plough, Dardilly, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Adel-Patient K, Ah-Leung S, Bernard H, Durieux-Alexandrenne C, Créminon C, Wal JM. Oral sensitization to peanut is highly enhanced by application of peanut extracts to intact skin, but is prevented when CpG and cholera toxin are added. Int Arch Allergy Immunol 2006; 143:10-20. [PMID: 17191005 DOI: 10.1159/000098221] [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: 06/07/2006] [Accepted: 09/12/2006] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND CpG oligonucleotides might offer an alternative to conventional immunotherapy in preventing and potentially reversing Th2-biased immune deregulation which leads to allergy. However, non-invasive ways of administration, especially in peanut-allergic patients, should be explored. METHODS One hundred micrograms of whole peanut protein extract (PE) alone, or mixed with cholera toxin (CT, 50 microg) plus CpG (100 microg) as adjuvant, was applied on intact skin of mice (40 min, twice). Initiation of an immune response was monitored by detection of specific antibodies in sera. The effect of this pretreatment on a further oral sensitization by PE was then evaluated by assaying antibodies and cytokines specific for PE and purified allergens. Cytokine production in liver 40 min after skin application was also assayed. RESULTS Two brief skin applications of PE alone highly potentiated further oral sensitization, as demonstrated by very intense specific IgE, IL-4 and IL-5 productions. Conversely, skin pretreatment with PE and CT + CpG efficiently prevented further sensitization via gastro-intestinal exposure. In both cases, the specificity of the antibodies and cytokines was the same as in control mice. CT + CpG treatment allowed the rapid production of IL-12 and TGFbeta in liver and of specific IgG2a in sera, suggesting the activation of Th1 and/or regulatory T cells. CONCLUSIONS Oral sensitization to peanut is highly enhanced by a previous short exposure of allergens to intact skin. Conversely, the use of CT + CpG adjuvant for skin application efficiently prevents further oral sensitization. The potential of such treatment in specific immunotherapy needs to be evaluated.
Collapse
Affiliation(s)
- Karine Adel-Patient
- Laboratoire Inra d'Immuno-Allergie Alimentaire, CEA de Saclay, Gif-sur-Yvette, France.
| | | | | | | | | | | |
Collapse
|
23
|
Mohan J, Hopkins J, Mabbott NA. Skin-derived dendritic cells acquire and degrade the scrapie agent following in vitro exposure. Immunology 2005; 116:122-33. [PMID: 16108824 PMCID: PMC1802412 DOI: 10.1111/j.1365-2567.2005.02207.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The accumulation of the scrapie agent in lymphoid tissues following inoculation via the skin is critical for efficient neuroinvasion, but how the agent is initially transported from the skin to the draining lymph node is not known. Langerhans cells (LCs) are specialized antigen-presenting cells that continually sample their microenvironment within the epidermis and transport captured antigens to draining lymph nodes. We considered LCs probable candidates to acquire and transport the scrapie agent after inoculation via the skin. XS106 cells are dendritic cells (DCs) isolated from mouse epidermis with characteristics of mature LC cells. To investigate the potential interaction of LCs with the scrapie agent XS106 cells were exposed to the scrapie agent in vitro. We show that XS106 cells rapidly acquire the scrapie agent following in vitro exposure. In addition, XS106 cells partially degrade the scrapie agent following extended cultivation. These data suggest that LCs might acquire and degrade the scrapie agent after inoculation via the skin, but data from additional experiments demonstrate that this ability could be lost in the presence of lipopolysaccharide or other immunostimulatory molecules. Our studies also imply that LCs would not undergo maturation following uptake of the scrapie agent in the skin, as the expression of surface antigens associated with LC maturation were unaltered following exposure. In conclusion, although LCs or DCs have the potential to acquire the scrapie agent within the epidermis our data suggest it is unlikely that they become activated and stimulated to transport the agent to the draining lymph node.
Collapse
|
24
|
Cognasse F, Acquart S, Beniguel L, Sabido O, Chavarin P, Genin C, Garraud O. Differential production of immunoglobulin classes and subclasses by mucosal-type human B-lymphocytes exposed in vitro to CpG oligodeoxynucleotides. Clin Chem Lab Med 2005; 43:22-31. [PMID: 15653438 DOI: 10.1515/cclm.2005.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
As B-lymphocytes play an important role in innate and adaptive immunity, we aimed to examine the effects of CpG oligodeoxynucleotides (ODNs) on purified tonsil-originating CD19+ B-cells, representing mucosal B-cells. We screened various K-type ODNs, reactive with human B-cells, and tested for the production of immunoglobulins in vitro. Using one CpG-ODN, DSP30, we observed that it could upregulate not only Toll-like receptor 9 (TLR9) mRNA expression in activated B-cells, but also the early expression of CD69 followed by the sequential expression of CD80, CD86 and the nuclear factor (NF)-kappaB pathway. Furthermore, mRNA expression of certain B-cell-derived cytokines was influenced by exposure to DSP30, with a strong upregulation of interleukin 6 (IL-6) and downregulation of IL1-beta. Stimulation of B-cells, co-stimulated with IL-2, IL-10 and soluble CD40 ligand (sCD40L) with different CpG-ODNs, had differing effects on the terminal differentiation in vitro of B-cells into immunoglobulin-secreting cells. TLR9 is involved in innate immunity and the recognition of bound CpG DNA from invading bacterial pathogens. As tonsillar B-cells are mucosal-type B-lymphocytes, this study suggests that CpG-ODNs show promise as mucosal adjuvants in modulating the local production of immunoglobulins of certain classes and subclasses, a crucial issue in vaccine perspectives.
Collapse
Affiliation(s)
- Fabrice Cognasse
- GIMAP-EA 3064, Université de Saint-Etienne, Faculté de Médecine, Saint-Etienne, France and EFS Auvergne-Loire, Saint-Etienne, France
| | | | | | | | | | | | | |
Collapse
|
25
|
Yang GX, Lian ZX, Kikuchi K, Liu YJ, Ansari AA, Ikehara S, Gershwin ME. CD4- plasmacytoid dendritic cells (pDCs) migrate in lymph nodes by CpG inoculation and represent a potent functional subset of pDCs. THE JOURNAL OF IMMUNOLOGY 2005; 174:3197-203. [PMID: 15749849 DOI: 10.4049/jimmunol.174.6.3197] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We have recently identified two groups of plasmacytoid dendritic cells (pDCs) isolated from murine liver based on the expression of CD4 and other cell surface markers uniquely expressed by pDCs. Herein, we describe the identification of both CD4+ and CD4- pDCs that clearly exist in lymph nodes (LNs), spleen, liver, thymus, bone marrow, and lung. Normally, CD4+ pDCs are enriched in LNs. However, after in vivo systemic injection with bacterial CpG, a larger number of CD4- pDCs are recruited to the LNs and local inoculation by CpG drives CD4- pDCs migrating into local sentinel LNs, suggesting that CD4- pDCs are the main subpopulation migrating to the peripheral LNs. Furthermore, although both freshly isolated CD4+ pDCs and CD4- pDCs appear as an immature plasmacytoid cell and develop into a DC morphology following activation, the two subsets have strikingly different immune features, including differences in the production pattern of cytokines stimulated with CpG and in T cell activation.
Collapse
Affiliation(s)
- Guo-Xiang Yang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA 95616, USA
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
Bacterial DNA and synthetic oligodeoxynucleotides containing CpG motifs (CpG-ODN) are the ligands for the Toll-like receptor 9 (TLR9), which is expressed by B-lymphocytes and a subset of dendritic cells. CpG-ODN are strong activators of both innate and specific immunity, and drive the immune response towards the Th1 phenotype. Given the promising results obtained in several experimental models of allergies or infections, CpG-ODN are now entering clinical trials for these diseases. In cancer, promising approaches combined CpG-ODN with tumor antigens, monoclonal antibodies or dendritic cells. When no relevant tumor antigen is known, CpG-ODN can be used alone to activate locally the innate immunity and trigger a tumor-specific immune response, overcoming the need for the identification of a tumoral antigen. Preclinical models have shown impressive results and several clinical trials are on-going worldwide in melanoma, lymphoma, renal carcinoma, breast cancer and glioblastoma.
Collapse
Affiliation(s)
- Antoine F Carpentier
- Fédération de Neurologie Mazarin et Inserm U.495, Hôpital de la Salpêtrière, 47, boulevard de l'Hôpital, 75651 Paris Cedex 13, France.
| |
Collapse
|
27
|
Switaj T, Jalili A, Jakubowska AB, Drela N, Stoksik M, Nowis D, Basak G, Golab J, Wysocki PJ, Mackiewicz A, Sasor A, Socha K, Jakóbisiak M, Lasek W. CpG immunostimulatory oligodeoxynucleotide 1826 enhances antitumor effect of interleukin 12 gene-modified tumor vaccine in a melanoma model in mice. Clin Cancer Res 2005; 10:4165-75. [PMID: 15217954 DOI: 10.1158/1078-0432.ccr-04-0022] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The effectiveness of interleukin (IL)-12-secreting tumor vaccines in the treatment of mouse tumors could be enhanced by concurrent application of cytokines and costimulatory molecules. We investigated the therapeutic potential of IL-12 gene-transduced melanoma vaccine in combination with CpG immunostimulatory oligodeoxynucleotide (ODN) 1826, an adjuvant known to favor development of Th1-biased immune response, in a B78-H1 (B78) melanoma model in mice. EXPERIMENTAL DESIGN Mice injected with B78 melanoma cells were treated with irradiated IL-12 gene-transduced B78 cells [B78/IL-12(X)] and/or ODN 1826. Mechanisms responsible for the antitumor effects of the treatment were investigated using fluorescence-activated cell sorter analysis, a standard (51)Cr releasing assay, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and ELISA. RESULTS Single injection of B78/IL-12(X) cells had no effect on tumor growth, whereas seven consecutive daily injections of ODN 1826 markedly inhibited tumor progression with occasional curative effects. When used in combination, B78/IL-12(X) cells and ODN 1826 caused additional tumor growth reduction and eradication of tumors in 62% of treated mice. The combined treatment activated local inflammatory response against tumor but also induced systemic antitumor immunity. In vitro studies have shown that when used together, B78/IL-12(X) cells and ODN 1826 induced a potent Th1 response and suggested the role of IFN-gamma in activation of the host immune response. The antitumor effects in double-treated mice were accompanied by the development of cytotoxic effectors in the spleen and activation of macrophages. CONCLUSIONS The results provided the evidence that the combination of IL-12 gene-modified melanoma vaccine and ODN 1826 induces synergistically systemic and local antitumor immunity.
Collapse
Affiliation(s)
- Tomasz Switaj
- Department of Immunology, Center of Biostructure, Medical University of Warsaw, Warsaw, Poland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Akiba H, Satoh M, Iwatsuki K, Kaiserlian D, Nicolas JF, Kaneko F. CpG immunostimulatory sequences enhance contact hypersensitivity responses in mice. J Invest Dermatol 2004; 123:488-93. [PMID: 15304088 DOI: 10.1111/j.0022-202x.2004.23318.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Bacterial DNA and synthetic cytidine-phosphate-guanosine-oligodeoxynucleotides (CpG ODN) potently activate dendritic cells (DC) and therefore have been proposed as adjuvants for vaccination strategies. Although CpG ODN are considered as safe adjuvants this study shows that CpG ODN are responsible for enhanced antigen-specific skin inflammatory reactions. We used the murine model of contact hypersensitivity (CHS) to 2,4-dinitrofluorobenzene (DNFB) in which hapten-specific CD8+T cytotoxic 1 cells are effector cells. Subcutaneous injection of CpG ODN, 1 d before sensitization enhanced the CHS response to DNFB and resulted in increased recruitment of CD8+ T cells at the challenge sites, whereas control ODN injection did not have any effect. This effect was local and not systemic as it was only observed when DNFB was applied at the same site as the CpG motifs. CpG ODN-induced enhancement of CHS was due to increased antigen-presenting cell functions of DC since: (i) CpG ODN-injected skin revealed upregulated expression of major histocompatibility complex class II, CD80, and CD86 molecules and (ii) CpG ODN treatment of DNFB-derivatized DC enhanced the intensity of CHS responses after in vivo transfer. Taken together, the results show that CpG ODN may be responsible for immune side-effects such as worsening of T cell-mediated skin diseases.
Collapse
Affiliation(s)
- Hitoshi Akiba
- Department of Dermatology, Fukushima Medical University School of Medicine, Hikarigaoka-1, Fukushima 960-1295, Japan.
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
The prevalence and severity of atopic diseases (atopic dermatitis, asthma, and allergic rhinitis) have increased over recent decades, particularly in industrialized nations. Atopic dermatitis, like asthma, is more common in older siblings and in less crowded houses and with late entry to day care, increased maternal education, and higher socio-economic status. The inverse relationship between the incidence of atopy and childhood infections has led to the "hygiene hypothesis," which suggests that diminished exposure to childhood infections in modern society has led to decreased TH1-type responses. Reduced TH1 may lead to enhanced TH2-type inflammation, which is important in promoting asthma and allergic disease. Corticosteroids, commonly used to treat these conditions, inhibit the function of inflammatory cells, but they are ineffective in altering the initial TH2-type response to allergens in a sensitized individual. Treatment with TH1 cytokines not only has failed to make any significant impact on the outcome of these diseases, but it also has caused significant adverse reactions. A novel therapeutic approach, recently reported in the preclinical setting, is the use of oligodeoxynucleotides, which contain unmethylated motifs centered on CG dinucleotides. These CpG oligodeoxynucleotides potently induce TH1 cytokines and suppress TH2 cytokines, and can prevent manifestations of asthma and other allergic diseases in animal models. They have the potential to reverse TH2-type responses to allergens and thus restore balance to the immune system without the adverse effects of TH1 cytokines.
Collapse
Affiliation(s)
- Iftikhar Hussain
- Department of Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | |
Collapse
|
30
|
Kunikata N, Sano K, Honda M, Ishii K, Matsunaga J, Okuyama R, Takahashi K, Watanabe H, Tamura G, Tagami H, Terui T. Peritumoral CpG oligodeoxynucleotide treatment inhibits tumor growth and metastasis of B16F10 melanoma cells. J Invest Dermatol 2004; 123:395-402. [PMID: 15245441 DOI: 10.1111/j.0022-202x.2004.23233.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although melanoma mostly affects the skin, it is notorious for its propensity to easily develop metastasis. Metastatic melanoma is highly resistant to a variety of therapies. We examined the anti-metastatic potential of peritumoral monotherapy against murine cutaneous B16F10 melanoma with synthetic oligodeoxynucleotides (ODN) containing unmethylated CpG motifs. We demonstrated that repeated peritumoral injections of CpG ODN significantly reduced skin tumor size. Peritumoral CpG ODN-treatment of skin tumors prevented the development of pulmonary B16F10 colonies. Adoptive transfer of splenocytes obtained from CpG ODN-treated mice markedly reduced the number of previously established pulmonary colonies in recipient naïve mice. T-lymphocyte depletion studies indicated that the anti-metastatic effect was dependent on both CD4+ and CD8+ T cells. These results suggest that CpG ODN are promising as a preventive and therapeutic anti-metastatic measure against melanoma.
Collapse
Affiliation(s)
- Nagisa Kunikata
- Department of Dermatology, tOHOKU University, Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Klimuk SK, Najar HM, Semple SC, Aslanian S, Dutz JP. Epicutaneous application of CpG oligodeoxynucleotides with peptide or protein antigen promotes the generation of CTL. J Invest Dermatol 2004; 122:1042-9. [PMID: 15102096 DOI: 10.1111/j.0022-202x.2004.22411.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Immunostimulatory oligodeoxynucleotides (ODN) are effective adjuvants in the induction of humoral and cellular immune responses when administered parenterally with antigen. The skin has recently become a target organ for the design of non-invasive vaccine technologies. Using ovalbumin (OVA) as a model antigen, we demonstrate that the application of ODN sequences to tape-stripped skin promotes the induction of potent cytotoxic T lymphocyte (CTL) responses to co-administered peptide. Induction of peptide-specific CTL required the presence of CpG motifs within the ODN. CTL afforded tumor protection against a tumor expressing an immunodominant OVA CTL epitope. CTL could also be induced to whole protein administered onto the skin. Differential CpG sequence activity was noted with respect to the induction of CTL to epicutaneous protein with an ODN sequence containing a poly-G motif having an optimal effect. Peptide-specific CTL could be detected in the peripheral blood as early as 6 d after a single immunization. These results highlight the potential of the bare skin as a route for vaccine development and indicate an important role for immunostimulatory ODN as adjuvants to generate functional CTL with the help of the skin immune system.
Collapse
Affiliation(s)
- Sandra K Klimuk
- Inex Pharmaceuticals Corporation, Burnaby, British Columbia, Canada
| | | | | | | | | |
Collapse
|
32
|
Nichani AK, Kaushik RS, Mena A, Popowych Y, Dent D, Townsend HGG, Mutwiri G, Hecker R, Babiuk LA, Griebel PJ. CpG oligodeoxynucleotide induction of antiviral effector molecules in sheep. Cell Immunol 2004; 227:24-37. [PMID: 15051512 DOI: 10.1016/j.cellimm.2004.01.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2003] [Accepted: 01/15/2004] [Indexed: 12/12/2022]
Abstract
Immunostimulatory CpG oligodeoxynucleotide (ODN) can protect mice against infection by many pathogens but the mechanisms mediating disease protection are not well defined. Furthermore, the mechanisms of CpG ODN induced disease protection in vivo have not been investigated in other species. We investigated the induction of antiviral effector molecules in sheep treated with a class B CpG ODN (2007). Subcutaneous injection of ODN 2007 induced a dose-dependent increase in serum levels of the antiviral effector molecule, 2'5'-A synthetase. Peak levels of enzyme were observed 4 days following ODN injection and enzyme levels remained elevated for the following 3-5 days. Repeated ODN injections induced a more sustained elevation of serum 2'5'-A synthetase activity. Finally, formulation of ODN 2007 in emulsigen increased the level of serum 2'5'-A synthetase activity and this response was CpG-specific. Elevated serum 2'5'-A synthetase activity suggested that CpG ODN acted through the induction of either interferon (IFN)-alpha or IFN-gamma. ODN 2007 did not induce detectable levels of IFN-alpha or IFN-gamma when incubated with peripheral blood mononuclear cells, but both IFN-alpha and IFN-gamma were detected following stimulation of lymph node cells with ODN 2007. CpG ODN induction of 2'5'-A synthetase in vitro correlated with the secretion of both IFN-alpha and IFN-gamma. Furthermore, immunohistochemical staining of skin revealed a marked cellular infiltration at the site of ODN 2007 injection. This cellular infiltration was CpG-specific and consisted of primarily CD172(+) myeloid cells. Many of the cells recruited to the site of ODN 2007 injection expressed IFN-alpha and some IFN-gamma. These observations support the conclusion that localized cell recruitment and activation contribute to CpG ODN induction of antiviral effector molecules, such as interferon and 2'5'-A synthetase.
Collapse
Affiliation(s)
- Anil K Nichani
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada S7N 5E3
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Krepler C, Wacheck V, Strommer S, Hartmann G, Polterauer P, Wolff K, Pehamberger H, Jansen B. CpG oligonucleotides elicit antitumor responses in a human melanoma NOD/SCID xenotransplantation model. J Invest Dermatol 2004; 122:387-91. [PMID: 15009720 DOI: 10.1046/j.0022-202x.2004.22202.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
For patients with advanced malignant melanoma, the 5 y survival rate with current treatment modalities is low. There is an urgent need for more effective therapeutic concepts. One approach with great potential is to stimulate the body's own immune defense to reject cancer cells using CpG oligonucleotides. Distinct oligonucleotides containing nonmethylated cytidine residues in cytidine-guanosine dinucleotides with particular flanking bases (CpG motifs) are capable of eliciting powerful immune stimulation by mimicking infectious disease. We evaluated the in vivo antitumoral effects of CpG oligonucleotides against human malignant melanoma xenografts in NOD/SCID mice. CpG oligonucleotides administered in single peritumoral subcutaneous injections three times per week resulted in elevated plasma levels of interleukin-12 and significant inhibition of the growth of established tumor xenografts by 60% (p<0.016) compared to the saline control. In addition to this a significant invasion of macrophages into tumor xenografts and increased numbers of Langerhans-cell-derived dendritic cells in draining lymph nodes could be observed. Our findings demonstrate the antitumor activity of oligonucleotides containing immune-stimulatory CpG motifs in a xenotransplantation model with absent B, T cells and a lack of natural killer cell function.
Collapse
Affiliation(s)
- Clemens Krepler
- Department of Dermatology, Division of General Dermatology, University of Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Cornélie S, Poulain-Godefroy O, Lund C, Vendeville C, Ban E, Capron M, Riveau G. Methylated CpG-Containing Plasmid Activates the Immune System. Scand J Immunol 2004; 59:143-51. [PMID: 14871290 DOI: 10.1111/j.0300-9475.2004.01373.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bacterial DNA differs from mammalian DNA by the presence of unmethylated cytosine-phosphate-guanosine (CpG) motifs. The immunostimulatory properties of a DNA vaccine have been suspected to be associated with these motifs. The aim of this study was to assess the inactivation of the immunostimulatory potential of a plasmid after methylation of its CpG motifs. We constructed two identical non-coding plasmids, and one of these was de novo methylated on its CG sequences. A single administration of recombinant antigen with methylated or unmethylated CpG-containing plasmid was performed in mice. As expected, only unmethylated CpG-containing plasmid enhanced the specific immune response. However, a study of in vivo activation of Langerhans' cells and analysis of mRNA synthesis indicated that both the plasmids promoted cell emigration and cytokine induction. These data highlight that a methylated CpG-containing plasmid is not inert and carries immunomodulatory properties. The results further emphasize the necessity to definitively identify the mode of action of plasmids used for DNA vaccination.
Collapse
Affiliation(s)
- S Cornélie
- INSERM U547, Pasteur Institut de Lille, 59019 Lille Cedex, France
| | | | | | | | | | | | | |
Collapse
|
35
|
Mempel M, Ronet C, Lemaitre F, Lim A, Kourilsky P, Gachelin G, Musette P. The intratumoral application of poly-G-oligodeoxynucleotides does not augment the naturally induced antitumoral CD8-T-cell response in P815 mastocytomas. Immunol Lett 2003; 89:47-57. [PMID: 12946864 DOI: 10.1016/s0165-2478(03)00111-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
DNA sequences containing CpG have been described to induce a strong immune reaction by acting on a variety of immune cells including a strong and pronounced antitumoral response. Poly-G-oligodeoxynucleotides (ODNs) on the other hand have been attributed the preferential induction of CD8-T-cell proliferation when used in vitro. This activity led us to the investigation of the possible antitumoral properties of poly-G-ODNs in an established CD8-dependent tumor eradication model. We used the well described poly-G-ODN 1628 in its capacity to enhance antitumoral CD8 response in the cutaneous mastocytoma P815. When injecting 30 microg of the purified phosphothioate-modified oligo into the tumor bearing area of P815 challenged mice for up to 12 consecutive days we did not observe increased tumor rejection as compared to the group of mice injected with a control oligo. The 1628-injected mice did not produce higher numbers of P815-specific CD8 cells as measured by P1A-, and P1E-tetramer staining and Immunoscope analysis. Furthermore, tumor-specific CD8 cells in 1628 did not show enhanced antitumoral cytotoxicity when analyzing lymphocyte-tumor cell co-cultures or transcription of the cytotoxic CD8-cell associated molecules interferon gamma, FAS ligand, perforin, or granzyme B by quantitative real-time RT-PCR. These experiments show that there is no enhanced induction of an antitumoral CD8 response after in situ administration of poly-G-ODNs in the P815 mastocytoma model.
Collapse
Affiliation(s)
- Martin Mempel
- Department of Immunology, Institut Pasteur, Unité de Biologie Moléculaire du Gène, INSERM U277, 25 rue du Dr. Roux, 75015 Paris, France.
| | | | | | | | | | | | | |
Collapse
|
36
|
Mölne L, Collins LV, Tarkowski A. Inflammatogenic properties of bacterial DNA following cutaneous exposure. J Invest Dermatol 2003; 121:294-9. [PMID: 12880421 DOI: 10.1046/j.1523-1747.2003.12352.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Bacterial DNA and oligodeoxynucleotides containing cytosine-phosphate-guanosine sequences and thereby mimicking prokaryotic DNA, have recently been shown to exert potent immunostimulatory properties. As skin normally harbors bacteria, and as the bacterial content and the levels of bacterial degradation products increase during skin infection, we analyzed the potential inflammatogenic role of bacterial DNA and oligodeoxynucleotides in a mouse model of cutaneous inflammation. Bacterial DNA from Staphylococcus aureus was injected intradermally into mice and its inflammatogenic properties were compared with synthetic phosphodiester and phosphorothioate cytosine-phosphate-guanosine- or GpC-containing oligodeoxynucleotides. A peak inflammatory infiltrate in the skin was seen already 2 d after injection with either bacterial DNA or the phosphodiester cytosine-phosphate-guanosine-oligodeoxynucleotides. In contrast, nuclease-resistant phosphorothioate cytosine-phosphate-guanosine-induced dermatitis peaked 7 d after intradermal injection. The inflammatory infiltrates consisted mainly of macrophages, and depletion of this cell population resulted in a significant (p=0.0001) decrease in the severity of inflammation, which suggests that macrophages play a central part in inflammatory responses in the skin following exposure to cytosine-phosphate-guanosine-containing oligodeoxynucleotides. A significant decrease in local inflammatory infiltrate was also seen in mice with deficiencies in neutrophil or lymphocyte populations, which indicates that these cell populations may also be involved in mediating inflammatory signals after the injection of immunostimulatory DNA sequences. In summary, our results suggest that bacterial DNA is an important virulence determinant and inflammatory stimulus during skin infections.
Collapse
Affiliation(s)
- Lena Mölne
- Department of Dermatology, Sahlgrenska University Hospital, University of Göteborg, Sweden.
| | | | | |
Collapse
|
37
|
Godefroy S, Corvaia N, Schmitt D, Aubry JP, Bonnefoy JY, Jeannin P, Staquet MJ. Outer membrane protein A (OmpA) activates human epidermal Langerhans cells. Eur J Cell Biol 2003; 82:193-200. [PMID: 12751905 DOI: 10.1078/0171-9335-00303] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Outer membrane protein (Omp)A is highly represented and conserved in the Enterobacteriaceae family. Using a recombinant OmpA from Klebsiella pneumoniae (kpOmpA), we have analysed the interaction between this bacterial cell wall protein and human Langerhans cells (LC), the antigen-presenting cells of the epidermis and mucosa. We showed that biotinylated kpOmpA binds to human LC freshly isolated from epidermis. kpOmpA up-regulated MHC class II, CD86 and CCR7 expression, enhanced migration in response to macrophage inflammatory protein-3beta (MIP-3beta) through a reconstituted basement membrane mimicking the prerequisite passage through the dermal-epidermal basement membrane on the way to lymph nodes. The allostimulatory function of kpOmpA-treated LC was more potent than that of untreated cells. Even though the proportion of LC which binds kpOmpA was shown to vary between individuals, our data indicate that kpOmpA binds to and activates LC, and suggest that recognition of OmpA by LC may be an initiating event in the antibacterial host response.
Collapse
|
38
|
Abstract
Invading pathogens are controlled by the innate and adaptive arms of the immune system. Adaptive immunity, which is mediated by B and T lymphocytes, recognises pathogens by rearranged high affinity receptors. However, the establishment of adaptive immunity is often not rapid enough to eradicate microorganisms as it involves cell proliferation, gene activation and protein synthesis. More rapid defense mechanisms are provided by innate immunity, which recognises invading pathogens by germ-line-encoded pattern recognition receptors (PRR). Recent evidence shows that this recognition can mainly be attributed to the family of TOLL-like receptors (TLR). Binding of pathogen-associated molecular patterns (PAMP) to TLR induces the production of reactive oxygen and nitrogen intermediates (ROI and RNI), pro-inflammatory cytokines, and up-regulates expression of co-stimulatory molecules, subsequently initiating the adaptive immunity. In this review, we will summarize the discovery and the critical roles of the TLR family in host defense, briefly allude to signaling mechanisms mediating the response to TLR ligands, and will provide an update on current knowledge regarding the ligand specificity of these receptors and their role in immunity of domestic animals, particularly cattle.
Collapse
Affiliation(s)
- Dirk Werling
- Institute of Veterinary Virology, University of Berne, Länggass-Str. 122, CH-3012 Bern, Switzerland
| | | |
Collapse
|
39
|
Harandi AM, Eriksson K, Holmgren J. A protective role of locally administered immunostimulatory CpG oligodeoxynucleotide in a mouse model of genital herpes infection. J Virol 2003; 77:953-62. [PMID: 12502811 PMCID: PMC140825 DOI: 10.1128/jvi.77.2.953-962.2003] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Unmethylated CpG dinucleotides in bacterial DNA or synthetic oligodeoxynucleotides (ODNs) are known as potent activators of the immune system and inducers of several Th1-associated immunomodulatory cytokines. We therefore investigated whether such a CpG-containing ODN (CpG ODN) given mucosally in the female genital tract could enhance innate immunity and protect against genital herpes infection. Groups of C57BL/6 mice were treated intravaginally with either CpG ODN or a non-CpG ODN control in the absence of any antigen either 2 days before or 4 h after an intravaginal challenge with a normally lethal dose of herpes simplex virus type 2 (HSV-2). Mice treated with CpG ODN exhibited significantly decreased titers of HSV-2 in their vaginal fluids compared with non-CpG ODN-treated mice. Furthermore, CpG ODN pretreatment significantly protected against development of disease and death compared to non-CpG ODN pretreatment. Most strikingly, CpG ODN conferred protection against disease and death even when given after the viral challenge. The CpG ODN-induced protection was associated with a rapid production of gamma interferon (IFN-gamma), interleukin-12 (IL-12), IL-18, and RANTES in the genital tract mucosa following CpG ODN treatment. The observed protection appeared to be dependent on IFN-gamma, IL-12, IL-18, and T cells, as CpG ODN pretreatment did not confer any significant protection in mice deficient in IFN-gamma, IL-12, IL-18, or T cells. Further, a complete protective immunity to reinfection was elicited in CpG ODN-treated, HSV-2-challenged mice, suggesting a role for mucosally administered CpG ODN in inducing the development of an acquired immune response in addition to its potent stimulation of innate immunity.
Collapse
Affiliation(s)
- Ali M Harandi
- Department of Medical Microbiology & Immunology, Göteborg University Vaccine Research Institute, Göteborg University, Sweden.
| | | | | |
Collapse
|
40
|
Fausch SC, Da Silva DM, Rudolf MP, Kast WM. Human papillomavirus virus-like particles do not activate Langerhans cells: a possible immune escape mechanism used by human papillomaviruses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:3242-9. [PMID: 12218143 DOI: 10.4049/jimmunol.169.6.3242] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
High-risk human papillomaviruses are linked to several malignancies including cervical cancer. Because human papillomavirus-infected women do not always mount protective antiviral immunity, we explored the interaction of human papillomavirus with Langerhans cells, which would be the first APCs the virus comes into contact with during infection. We determined that dendritic cells, normally targeted by vaccination procedures and Langerhans cells, normally targeted by the natural virus equally internalize human papillomavirus virus-like particles. However, in contrast to dendritic cells, Langerhans cells are not activated by human papillomavirus virus-like particles, illustrated by the lack of: up-regulating activation markers, secreting IL-12, stimulating T cells in an MLR, inducing human papillomavirus-specific immunity, and migrating from epidermal tissue. Langerhans cells, like dendritic cells, can display all of these characteristics when stimulated by proinflammatory agents. These data may define an intriguing immune escape mechanism used by human papillomavirus and form the basis for designing optimal vaccination strategies.
Collapse
Affiliation(s)
- Steven C Fausch
- Cancer Immunology Program, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL 60153, USA
| | | | | | | |
Collapse
|
41
|
Baca-Estrada ME, Ewen C, Mahony D, Babiuk LA, Wilkie D, Foldvari M. The haemopoietic growth factor, Flt3L, alters the immune response induced by transcutaneous immunization. Immunology 2002; 107:69-76. [PMID: 12225364 PMCID: PMC1782769 DOI: 10.1046/j.1365-2567.2002.01488.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Topical application of antigen induces antigen-specific humoral and cellular immune responses. In this study we examined whether expansion of dendritic cells (DC) by Flt3 ligand (Flt3L) treatment influences the induction of immune responses following transcutaneous immunization. Mice were treated intraperitoneally with Flt3L or phosphate-buffered saline (PBS) and immunized transcutaneously with hen egg lysozyme (HEL). Flt3L-treated mice developed lower HEL-specific cellular and humoral immune responses than PBS-treated mice. However, in the presence of cholera toxin (CT), a potent adjuvant for mucosal and transcutaneous immunization, Flt3L-treated mice developed significantly higher cellular and humoral immune responses to HEL when compared to PBS-treated mice. We assessed whether the immunomodulatory effects of CT were a result of activation of epidermal dendritic cells (Langerhans' cells; LC). Our results indicate that within 8-12 hr of topical application of CT, epidermal LC cells lose their dendritic morphology and become rounder in appearance. In addition, we observed enhanced expression of major histocompatibility complex (MHC) class II, and of adhesion molecules CD11c and intracellular adhesion molecule-1 (ICAM-1). Our observations support the concept that the state of activation of DC in the skin is central to the regulation of immune responses. This information is relevant to the design of effective transcutaneous vaccination strategies.
Collapse
Affiliation(s)
- Maria E Baca-Estrada
- Veterinary Infectious Disease Organization, Department of Animal and Poultry Science and College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada.
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
Unmethylated CpG motifs are prevalent in bacterial but not vertebrate genomic DNAs. Oligodeoxynucleotides (ODN) containing CpG motifs activate host defense mechanisms leading to innate and acquired immune responses. The recognition of CpG motifs requires Toll-like receptor (TLR) 9, which triggers alterations in cellular redox balance and the induction of cell signaling pathways including the mitogen activated protein kinases (MAPKs) and NF kappa B. Cells that express TLR-9, which include plasmacytoid dendritic cells (PDCs) and B cells, produce Th1-like proinflammatory cytokines, interferons, and chemokines. Certain CpG motifs (CpG-A) are especially potent at activating NK cells and inducing IFN-alpha production by PDCs, while other motifs (CpG-B) are especially potent B cell activators. CpG-induced activation of innate immunity protects against lethal challenge with a wide variety of pathogens, and has therapeutic activity in murine models of cancer and allergy. CpG ODN also enhance the development of acquired immune responses for prophylactic and therapeutic vaccination.
Collapse
Affiliation(s)
- Arthur M Krieg
- Department of Veterans Affairs Medical Center, Iowa City, Iowa 52246, USA.
| |
Collapse
|
43
|
Merad M, Sugie T, Engleman EG, Fong L. In vivo manipulation of dendritic cells to induce therapeutic immunity. Blood 2002; 99:1676-82. [PMID: 11861283 DOI: 10.1182/blood.v99.5.1676] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Efficient antigen presentation and T-cell priming are essential components of effective antitumor immunity. Dendritic cells are critical to both of these functions but to date no method has been devised that both targets antigen to these cells and activates them, in situ, in a manner that induces systemic immunity. In this study we combined a dendritic cell growth factor, Flt3 ligand, with a dendritic cell activator, immunostimulatory DNA, and a tumor antigen to activate and load dendritic cells in vivo. Initial studies showed that immunostimulatory DNA not only activates dendritic cells but also prolongs their survival in vivo and in vitro. Following treatment of mice with Flt3 ligand, coadministration of immunostimulatory DNA and antigen induced potent antitumor immunity, resulting in both tumor prevention and regression of existing tumors. CD8 cytotoxic T lymphocytes but not CD4 T cells were required for tumor protection. Natural killer cells also contributed to tumor protection. These results show that dendritic cells can be loaded with antigen and activated, in situ, and provide the basis for dendritic cell- targeted clinical strategies.
Collapse
Affiliation(s)
- Miriam Merad
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | | | | | |
Collapse
|
44
|
Kawarada Y, Ganss R, Garbi N, Sacher T, Arnold B, Hämmerling GJ. NK- and CD8(+) T cell-mediated eradication of established tumors by peritumoral injection of CpG-containing oligodeoxynucleotides. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5247-53. [PMID: 11673539 DOI: 10.4049/jimmunol.167.9.5247] [Citation(s) in RCA: 163] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Unmethylated cytosine-phosphorothioate-guanine (CpG) containing oligodeoxynucleotides (CpG-ODN) are known to act as adjuvants and powerful activators of the innate immune system. We investigated the therapeutic effect of CpG-ODN on a variety of established mouse tumors including AG104A, IE7 fibrosarcoma, B16 melanoma, and 3LL lung carcinoma. These tumors are only weakly immunogenic and notoriously difficult to treat. Repeated peritumoral injection of CpG-ODN resulted in complete rejection or strong inhibition of tumor growth, whereas systemic application had only partial effects. The CpG-ODN-induced tumor rejection was found to be mediated by both NK and tumor-specific CD8(+) T cells. Comparison of parental tumors and variants rendered more antigenic by transfection with tumor Ags suggested that the efficiency of the CpG-ODN therapy correlated with the antigenicity of the tumors. Peritumoral CpG-ODN treatment was even effective in a situation where the immune system was tolerant for the tumor Ag, as shown by breakage of tolerance and tumor elimination. These results suggest that peritumoral application of CpG-ODN acts locally by inducing NK cells, and also leads to efficient presentation of tumor Ags and stimulation of CD8(+) effector and memory T cells, thus providing a powerful antitumor therapy that can be also applied without knowledge of the tumor Ag.
Collapse
Affiliation(s)
- Y Kawarada
- Division of Molecular Immunology, Tumor Immunology Program, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
Mobilization of dendritic cells into lymphatic vessels requires cytokine stimulation and induction of the chemokine receptor CCR7. The respective roles of the CCR7 ligands CCL19 and CCL21 in mediating migration are not fully defined, but chemotaxis to CCL19 mediates Langerhans cell exit from the epidermis. Optimal chemotaxis to CCL19 occurs when DCs are triggered with exogenous leukotriene C(4), an eicosanoid transported out of the cell via the ATP binding cassette (ABC) transporter multidrug resistance related protein 1 (MRP1, ABCC1). Indeed, MRP1 and the related multidrug resistance protein 1 (MDR1, p-glycoprotein, ABCB1) may control the intracellular and extracellular accumulation of key signaling lipids that regulate dendritic cell migration.
Collapse
Affiliation(s)
- G J Randolph
- Institute for Gene Therapy and Molecular Medicine, Mt. Sinai School of Medicine, 1425 Madison Avenue, Box 1496, New York, NY 10029, USA.
| |
Collapse
|
46
|
Coombes BK, Mahony JB. Dendritic cell discoveries provide new insight into the cellular immunobiology of DNA vaccines. Immunol Lett 2001; 78:103-11. [PMID: 11672594 DOI: 10.1016/s0165-2478(01)00242-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The evolution of increasingly virulent human pathogens, together with the rapid onset of antimicrobial resistance has created a need for new vaccination strategies. Nucleic acid vaccines, based on recombinant DNA technology are a promising new vaccine formulation capable of eliciting both humoral and cellular immune responses. This technology has been experimentally validated in animal models of pathogen challenge and tumor protection following administration of a DNA vaccine and has led to extensive research into the mechanisms of protective immunity. We focus here on the cellular and molecular mechanisms leading to cell-mediated immune responses to DNA vaccines and discuss these mechanisms in light of recent advances in the field of dendritic cell immunobiology. In particular, the potential involvement of: (i) the CpG pattern-recognition receptor, toll-like receptor-9; (ii) the dendritic cell-specific surface adhesion molecule, DC-SIGN; and (iii) the molecular interactions between CD40 and CD154 in the evolution of protective cell-mediated immunity to DNA vaccines are discussed. An improved understanding of the precise mechanisms leading to protective cellular immunity following DNA vaccination may help in the design of novel DNA constructs containing immunostimulatory features that target one or more of these mechanisms, with the aim of increasing the immunogenic potential and protective efficacy of DNA vaccines.
Collapse
Affiliation(s)
- B K Coombes
- Department of Medical Sciences, Father Sean O'Sullivan Research Centre, St. Joseph's Hospital, McMaster University, 50 Charlton Avenue East, Hamilton, Ontario, Canada L8N 4A6.
| | | |
Collapse
|
47
|
Parsa AT, Chi JH, Hurley PT, Jeyapalan SA, Bruce JN. Immunomodulation of glioma cells after gene therapy: induction of major histocompatibility complex class I but not class II antigen in vitro. Neurosurgery 2001; 49:681-8; discussion 688-9. [PMID: 11523680 DOI: 10.1097/00006123-200109000-00028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE Acquired immunity has been demonstrated in Fischer rats bearing syngeneic 9L tumors after herpes simplex virus (HSV) thymidine kinase (TK) gene transfection and ganciclovir treatment. The nature of this immunity in rats and its relevance to the HSV TK/ganciclovir protocol for human subjects remain to be determined. In this study, levels of major histocompatibility complex (MHC) Class I and II antigen expression were measured before and after HSV TK transfection, in an effort to document immunomodulatory changes caused by gene therapy. METHODS Tumor cells from the 9L gliosarcoma cell line, three primary human glioma cultures, and the human glioma cell line U87 MG were transduced with HSV TK vector-containing supernatant from fibroblast-producing cells (titer of 5 x 10(6) colony-forming units/ml) and selected in G418 medium for neomycin resistance. Clones were pooled or individually selected for cell-killing assays with ganciclovir, to confirm TK expression (10(3) cells/well in a 96-well dish). Northern analyses using MHC Class I and Class II complementary deoxyribonucleic acid probes were performed on blots containing total ribonucleic acid from wild-type tumor cells and HSV TK transfectants. A beta-actin complementary deoxyribonucleic acid probe served as an internal control. Cell surface expression was confirmed with flow cytometry. The induction of MHC Class I was tested for cycloheximide and genistein sensitivity. RESULTS All cell cultures exhibited increases in MHC Class I but not MHC Class II expression, as determined by Northern analysis densitometry and flow cytometry. Cycloheximide treatment did not diminish the up-regulation of MHC Class I after retroviral transfection, implicating a signal transduction pathway that does not require ongoing protein synthesis. Genistein pretreatment of cell cultures did diminish the up-regulation of MHC Class I, implicating a tyrosine kinase in the signaling cascade. CONCLUSION Induction of MHC Class I in rat and human glioma cells after HSV TK retroviral gene therapy is a primary effect that is dependent on tyrosine kinase activity. Specific immune responses generated after transfection may represent an important general side effect of gene therapy protocols. Elucidation of the mechanism of immunomodulation after gene therapy will likely yield safer and more effective clinical protocols.
Collapse
Affiliation(s)
- A T Parsa
- Bartoli Brain Tumor Research Laboratory, Department of Neurological Surgery, Neurological Institute of New York, College of Physicians and Surgeons of Columbia University, New York, USA
| | | | | | | | | |
Collapse
|
48
|
Parsa AT, Chi JH, Hurley PT, Jeyapalan SA, Bruce JN. Immunomodulation of Glioma Cells after Gene Therapy: Induction of Major Histocompatibility Complex Class I but Not Class II Antigen in Vitro. Neurosurgery 2001. [DOI: 10.1227/00006123-200109000-00028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
49
|
Hervé M, Dupré L, Ban E, Schacht AM, Capron A, Riveau G. Features of the antibody response attributable to plasmid backbone adjuvanticity after DNA immunization. Vaccine 2001; 19:4549-56. [PMID: 11483283 DOI: 10.1016/s0264-410x(01)00174-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
DNA vaccination induces antigen-specific immune responses with characteristics distinct from other vaccination modes. In the present study, the contribution of the plasmid backbone adjuvant effect to the quality of the DNA-raised antibody response was investigated. For this purpose, three intradermal primings were compared in mice using: (1) the recombinant Schistosoma haematobium glutathione S-transferase antigen (rSh28GST): (2) rSh28GST supplemented with a non-coding plasmid; and (3) a Sh28GST-encoding plasmid. In contrast to immunization with the protein, DNA immunization elicited a very stable antibody (Ab) response over a prolonged period of time. This feature was attributed to the plasmid backbone, because co-administration of the non-coding plasmid with rSh28GST allowed the maintenance of the specific Ab response. A strong anamnestic Ab response was induced after intradermal boost with rSh28GST only in the mice primed with pMSh. This indicated that the selective ability of DNA vaccination to induce memory humoral response was independent of the plasmid backbone. In contrast the plasmid backbone was found to strongly participate in the preferential IgG2a Ab production observed. These results suggest that, following DNA immunization, the Th1-biased profile and the maintenance of the long-lived Ab response could be attributed to an adjuvant effect of the plasmid backbone during priming, whereas the strength of B-cell memory was independent of this effect.
Collapse
Affiliation(s)
- M Hervé
- Relations Hôte-Parasite et Stratégies Vaccinales, INSERM U 167, Institut Pasteur de Lille, 1 Rue du professeur Calmette, BP-245, F-59019, Cedex, Lille, France.
| | | | | | | | | | | |
Collapse
|
50
|
Angeli V, Faveeuw C, Roye O, Fontaine J, Teissier E, Capron A, Wolowczuk I, Capron M, Trottein F. Role of the parasite-derived prostaglandin D2 in the inhibition of epidermal Langerhans cell migration during schistosomiasis infection. J Exp Med 2001; 193:1135-47. [PMID: 11369785 PMCID: PMC2193325 DOI: 10.1084/jem.193.10.1135] [Citation(s) in RCA: 219] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Epidermal Langerhans cells (LCs) play a key role in immune defense mechanisms and in numerous immunological disorders. In this report, we show that percutaneous infection of C57BL/6 mice with the helminth parasite Schistosoma mansoni leads to the activation of LCs but, surprisingly, to their retention in the epidermis. Moreover, using an experimental model of LC migration induced by tumor necrosis factor (TNF)-α, we show that parasites transiently impair the departure of LCs from the epidermis and their subsequent accumulation as dendritic cells in the draining lymph nodes. The inhibitory effect is mediated by soluble lipophilic factors released by the parasites and not by host-derived antiinflammatory cytokines, such as interleukin-10. We find that prostaglandin (PG)D2, but not the other major eicosanoids produced by the parasites, specifically impedes the TNF-α–triggered migration of LCs through the adenylate cyclase–coupled PGD2 receptor (DP receptor). Moreover, the potent DP receptor antagonist BW A868C restores LC migration in infected mice. Finally, in a model of contact allergen-induced LC migration, we show that activation of the DP receptor not only inhibits LC emigration but also dramatically reduces the contact hypersensitivity responses after challenge. Taken together, we propose that the inhibition of LC migration could represent an additional stratagem for the schistosomes to escape the host immune system and that PGD2 may play a key role in the control of cutaneous immune responses.
Collapse
Affiliation(s)
- Véronique Angeli
- Centre d'Immunologie et de Biologie Parasitaire, Institut National de la Santé et de la Recherche Médicale (INSERM) U547
| | - Christelle Faveeuw
- Centre d'Immunologie et de Biologie Parasitaire, Institut National de la Santé et de la Recherche Médicale (INSERM) U547
| | - Olivier Roye
- Centre National de la Recherche Scientifique 8527, Institut de Biologie de Lille
| | - Josette Fontaine
- Centre d'Immunologie et de Biologie Parasitaire, Institut National de la Santé et de la Recherche Médicale (INSERM) U547
| | | | - André Capron
- Centre d'Immunologie et de Biologie Parasitaire, Institut National de la Santé et de la Recherche Médicale (INSERM) U547
| | - Isabelle Wolowczuk
- Centre National de la Recherche Scientifique 8527, Institut de Biologie de Lille
| | - Monique Capron
- Centre d'Immunologie et de Biologie Parasitaire, Institut National de la Santé et de la Recherche Médicale (INSERM) U547
| | - François Trottein
- Centre d'Immunologie et de Biologie Parasitaire, Institut National de la Santé et de la Recherche Médicale (INSERM) U547
| |
Collapse
|