Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
1.
Immunol Cell Biol ; 101(1): 36-48, 2023 01.
Article in English | MEDLINE | ID: mdl-36214093

ABSTRACT

Type 1 diabetes (T1D) is caused by aberrant activation of autoreactive T cells specific for the islet beta cells. How islet-specific T cells evade tolerance to become effector T cells is unknown, but it is believed that an altered gut microbiota plays a role. Possible mechanisms include bystander activation of autoreactive T cells in the gut or "molecular mimicry" from cross-reactivity between gut microbiota-derived peptides and islet-derived epitopes. To investigate these mechanisms, we use two islet-specific CD8+ T cell clones and the non-obese diabetic mouse model of type 1 diabetes. Both insulin-specific G9C8 cells and IGRP-specific 8.3 cells underwent early activation and proliferation in the pancreatic draining lymph nodes but not in the Peyer's patches or mesenteric lymph nodes. Mutation of the endogenous epitope for G9C8 cells abolished their CD69 upregulation and proliferation, ruling out G9C8 cell activation by a gut microbiota derived peptide and molecular mimicry. However, previously activated islet-specific effector memory cells but not naïve cells migrated into the Peyer's patches where they increased their cytotoxic function. Oral delivery of butyrate, a microbiota derived anti-inflammatory metabolite, reduced IGRP-specific cytotoxic function. Thus, while initial activation of islet-specific CD8+ T cells occurred in the pancreatic lymph nodes, activated cells trafficked through the gut lymphoid tissues where they gained additional effector function via non-specific bystander activation influenced by the gut microbiota.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans , Mice , Animals , CD8-Positive T-Lymphocytes , Diabetes Mellitus, Type 1/genetics , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Peptides/metabolism , Lymph Nodes , Epitopes/metabolism
2.
Immunol Cell Biol ; 100(1): 33-48, 2022 01.
Article in English | MEDLINE | ID: mdl-34668580

ABSTRACT

The autoimmune disease type 1 diabetes is predominantly mediated by CD8+ cytotoxic T-cell destruction of islet beta cells, of which islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP)206-214 is a dominant target antigen specificity. Previously, we found that a liposome-based antigen-specific immunotherapy encapsulating the CD4+ T-cell islet epitope 2.5mim together with the nuclear factor-κB inhibitor calcitriol induced regulatory T cells and protected from diabetes in NOD mice. Here we investigated whether the same system delivering IGRP206-214 could induce antigen-specific CD8+ T-cell-targeted immune regulation and delay diabetes. Subcutaneous administration of IGRP206-214 /calcitriol liposomes transiently activated and expanded IGRP-specific T-cell receptor transgenic 8.3 CD8+ T cells. Liposomal co-delivery of calcitriol was required to optimally suppress endogenous IGRP-specific CD8+ T-cell interferon-γ production and cytotoxicity. Concordantly, a short course of IGRP206-214 /calcitriol liposomes delayed diabetes progression and reduced insulitis. However, when IGRP206-214 /calcitriol liposomes were delivered together with 2.5mim /calcitriol liposomes, disease protection was not observed and the regulatory effect of 2.5mim /calcitriol liposomes was abrogated. Thus, tolerogenic liposomes that target either a dominant CD8+ or a CD4+ T-cell islet epitope can delay diabetes progression but combining multiple epitopes does not enhance protection.


Subject(s)
Diabetes Mellitus, Type 1 , Animals , CD8-Positive T-Lymphocytes , Epitopes, T-Lymphocyte , Glucose-6-Phosphatase/metabolism , Immune Tolerance , Liposomes/metabolism , Mice , Mice, Inbred NOD , T-Lymphocytes, Regulatory
3.
PLoS Pathog ; 16(7): e1008651, 2020 07.
Article in English | MEDLINE | ID: mdl-32658914

ABSTRACT

Type-2 immunity elicits tissue repair and homeostasis, however dysregulated type-2 responses cause aberrant tissue remodelling, as observed in asthma. Severe respiratory viral infections in infancy predispose to later asthma, however, the processes that mediate tissue damage-induced type-2 inflammation and the origins of airway remodelling remain ill-defined. Here, using a preclinical mouse model of viral bronchiolitis, we find that increased epithelial and mesenchymal high-mobility group box 1 (HMGB1) expression is associated with increased numbers of IL-13-producing type-2 innate lymphoid cell (ILC2s) and the expansion of the airway smooth muscle (ASM) layer. Anti-HMGB1 ablated lung ILC2 numbers and ASM growth in vivo, and inhibited ILC2-mediated ASM cell proliferation in a co-culture model. Furthermore, we identified that HMGB1/RAGE (receptor for advanced glycation endproducts) signalling mediates an ILC2-intrinsic IL-13 auto-amplification loop. In summary, therapeutic targeting of the HMGB1/RAGE signalling axis may act as a novel asthma preventative by dampening ILC2-mediated type-2 inflammation and associated ASM remodelling.


Subject(s)
Airway Remodeling/immunology , HMGB1 Protein/immunology , Inflammation/immunology , Lymphocytes/immunology , Muscle, Smooth/immunology , Animals , Mice , Muscle, Smooth/pathology , Receptor for Advanced Glycation End Products/immunology
4.
J Immunol ; 205(5): 1239-1247, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32709661

ABSTRACT

A series of layered peripheral checkpoints maintain self-reactive B cells in an unresponsive state. Autoantibody production occurs when these checkpoints are breached; however, when and how this occurs is largely unknown. In particular, how self-reactive B cells are restrained during bystander inflammation in otherwise healthy individuals is poorly understood. A weakness has been the unavailability of methods capable of dissecting physiologically relevant B cell responses without the use of an engineered BCR. Resolving this will provide insights that decipher how this process goes awry during autoimmunity or could be exploited for therapy. In this study, we use a strong adjuvant to provide bystander innate and adaptive signals that promote B cell responsiveness in conjunction with newly developed B cell detection tools to study in detail the ways that peripheral tolerance mechanisms limit the expansion and function of self-reactive B cells activated under these conditions. We show that although self-reactive B cells are recruited into the germinal center, their development does not proceed, possibly because of rapid counterselection. Consequently, differentiation of plasma cells is blunted, and Ab responses are transient and devoid of affinity maturation. We propose this approach, and these tools can be more widely applied to track Ag-specific B cell responses to more disease-relevant Ags, without the need for BCR transgenic mice, in settings where tolerance pathways are compromised or have been genetically manipulated to drive stronger insights into the biology underlying B cell-mediated autoimmunity.


Subject(s)
Antibody Formation/immunology , B-Lymphocytes/immunology , Immune Tolerance/immunology , Peripheral Tolerance/immunology , Receptors, Antigen, B-Cell/immunology , Animals , Autoantibodies/immunology , Autoantigens/immunology , Autoimmunity/immunology , Cell Differentiation/immunology , Female , Germinal Center/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Plasma Cells/immunology
5.
Cytotherapy ; 22(8): 436-444, 2020 08.
Article in English | MEDLINE | ID: mdl-32546362

ABSTRACT

BACKGROUND AIMS: Pathological activation and collaboration of T and B cells underlies pathogenic autoantibody responses. Existing treatments for autoimmune disease cause non-specific immunosuppression, and induction of antigen-specific tolerance remains an elusive goal. Many immunotherapies aim to manipulate the T-cell component of T-B interplay, but few directly target B cells. One possible means to specifically target B cells is the transfer of gene-engineered BM that, once engrafted, gives rise to widespread specific and tolerogenic antigen expression within the hematopoietic system. METHODS: Gene-engineered bone marrow encoding ubiquitous ovalbumin expression was transferred after low-dose (300-cGy) immune-preserving irradiation. B-cell responsiveness was monitored by analyzing ovalbumin-specific antibody production after immunization with ovalbumin/complete Freund's adjuvant. Ovalbumin-specific B cells and their response to immunization were analyzed using multi-tetramer staining. When antigen-encoding bone marrow was transferred under immune-preserving conditions, cognate antigen-specific B cells were purged from the recipient's preexisting B-cell repertoire and the repertoire that arose after bone marrow transfer. RESULTS: OVA-specific B-cell deletion was apparent within the established host B-cell repertoire as well as that developing after gene-engineered bone marrow transfer. OVA-specific antibody production was substantially inhibited by transfer of OVA-encoding BM and activation of OVA-specific B cells, germinal center formation and subsequent OVA-specific plasmablast differentiation were all inhibited. Low levels of gene-engineered bone marrow chimerism were sufficient to limit antigen-specific antibody production. RESULTS: These data show that antigen-specific B cells within an established B-cell repertoire are susceptible to de novo tolerance induction, and this can be achieved by transfer of gene-engineered bone marrow. This adds further dimensions to the utility of antigen-encoding bone marrow transfer as an immunotherapeutic tool.


Subject(s)
Antibody Formation , Antigens/metabolism , B-Cell Maturation Antigen/metabolism , B-Lymphocytes/immunology , Bone Marrow Transplantation , Bone Marrow/immunology , Lymphocyte Depletion , Animals , Cell Differentiation , Germinal Center/cytology , Germinal Center/metabolism , Immune Tolerance/immunology , Mice, Inbred C57BL , Ovalbumin/biosynthesis , Ovalbumin/immunology , T-Lymphocytes/immunology
6.
Eur J Immunol ; 48(7): 1251-1254, 2018 07.
Article in English | MEDLINE | ID: mdl-29572817

ABSTRACT

Detecting naïve antigen-specific B cells can be challenging. Use of multiple, complementary tetramers with different fluorochromes enhances sensitivity and specificity allowing naïve antigen-specific B cells to be readily distinguished within a polyclonal repertoire. Activated, affinity-matured B cells, however, can be detected effectively using a single tetramer.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes/immunology , Flow Cytometry/methods , Receptors, Antigen, B-Cell/immunology , Antibody Affinity , Antigens/metabolism , Cells, Cultured , Epitopes , Fluorescent Dyes/metabolism , HLA Antigens/metabolism , Humans , Lymphocyte Activation , Protein Binding , Receptors, Antigen, B-Cell/genetics , Sensitivity and Specificity
7.
Eur J Immunol ; 47(9): 1550-1561, 2017 09.
Article in English | MEDLINE | ID: mdl-28665492

ABSTRACT

Type 1 diabetes (T1D) results from autoimmune destruction of insulin-producing pancreatic ß cells. Therapies need to incorporate strategies to overcome the genetic defects that impair induction or maintenance of peripheral T-cell tolerance and contribute to disease development. We tested whether the enforced expression of an islet autoantigen in antigen-presenting cells (APC) counteracted peripheral T-cell tolerance defects in autoimmune-prone NOD mice. We observed that insulin-specific CD8+ T cells transferred to mice in which proinsulin was transgenically expressed in APCs underwent several rounds of division and the majority were deleted. Residual insulin-specific CD8+ T cells were rendered unresponsive and this was associated with TCR downregulation, loss of tetramer binding and expression of a range of co-inhibitory molecules. Notably, accumulation and effector differentiation of insulin-specific CD8+ T cells in pancreatic lymph nodes was prominent in non-transgenic recipients but blocked by transgenic proinsulin expression. This shift from T-cell priming to T-cell tolerance exemplifies the tolerogenic capacity of autoantigen expression by APC and the capacity to overcome genetic tolerance defects.


Subject(s)
Antigen-Presenting Cells/immunology , Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Islets of Langerhans/immunology , Proinsulin/immunology , Animals , Autoimmunity , Cells, Cultured , Humans , Immune Tolerance , Lymphocyte Activation , Male , Mice , Mice, Inbred NOD
8.
Diabetologia ; 60(11): 2256-2261, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28779211

ABSTRACT

AIMS/HYPOTHESIS: The aim of this study was to determine whether therapy with the cytokine IL-22 could be used to prevent the development of, or treat, autoimmune diabetes in the NOD mouse. METHODS: Six-week-old NOD mice were administered bi-weekly either recombinant mouse IL-22 (200 ng/g) or PBS (vehicle control) intraperitoneally until overt diabetes was diagnosed as two consecutive measurements of non-fasting blood glucose ≥ 11 mmol/l. At this time, NOD mice in the control arm were treated with LinBit insulin pellets and randomised to bi-weekly therapeutic injections of either PBS or IL-22 (200 ng/g) and followed until overt diabetes was diagnosed, as defined above. RESULTS: IL-22 therapy did not delay the onset of diabetes in comparison with the vehicle-treated mice. We did not observe an improvement in islet area, glycaemic control, beta cell residual function, endoplasmic reticulum stress, insulitis or macrophage and neutrophil infiltration as determined by non-fasting blood glucose, C-peptide and histological scoring. Therapeutic administration of IL-22 did not reduce circulating lipopolysaccharide, a marker of impaired gut mucosal integrity. CONCLUSIONS/INTERPRETATION: Our study suggests that, at this dosing regimen introduced either prior to overt diabetes or at diagnosis of diabetes, recombinant mouse IL-22 therapy cannot prevent autoimmune diabetes, or prolong the honeymoon period in the NOD mouse.


Subject(s)
Diabetes Mellitus, Type 1/drug therapy , Interleukins/therapeutic use , Animals , Biological Assay , Diabetes Mellitus, Type 1/immunology , Female , In Vitro Techniques , Mice , Mice, Inbred NOD , Interleukin-22
9.
Immunol Cell Biol ; 95(9): 765-774, 2017 10.
Article in English | MEDLINE | ID: mdl-28611473

ABSTRACT

Type 1 diabetes (T1D) results from T-cell-mediated autoimmune destruction of pancreatic ß cells. Effector T-cell responses emerge early in disease development and expand as disease progresses. Following ß-cell destruction, a long-lived T-cell memory is generated that represents a barrier to islet transplantation and other cellular insulin-replacement therapies. Development of effective immunotherapies that control or ablate ß-cell destructive effector and memory T-cell responses has the potential to prevent disease progression and recurrence. Targeting antigen expression to antigen-presenting cells inactivates cognate CD8+ effector and memory T-cell responses and has therapeutic potential. Here we investigated this in the context of insulin-specific responses in the non-obese diabetic mouse where genetic immune tolerance defects could impact on therapeutic tolerance induction. Insulin-specific CD8+ memory T cells transferred to mice expressing proinsulin in antigen-presenting cells proliferated in response to transgenically expressed proinsulin and the majority were rapidly deleted. A small proportion of transferred insulin-specific Tmem remained undeleted and these were antigen-unresponsive, exhibited reduced T cell receptor (TCR) expression and H-2Kd/insB15-23 tetramer binding and expressed co-inhibitory molecules. Expression of proinsulin in antigen-presenting cells also abolished the diabetogenic capacity of CD8+ effector T cells. Therefore, destructive insulin-specific CD8+ T cells are effectively inactivated by enforced proinsulin expression despite tolerance defects that exist in diabetes-prone NOD mice. These findings have important implications in developing immunotherapeutic approaches to T1D and other T-cell-mediated autoimmune diseases.


Subject(s)
Antigen-Presenting Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/physiology , Proinsulin/metabolism , Adoptive Transfer , Animals , Autoantigens/immunology , Cells, Cultured , Humans , Immune Tolerance , Immunologic Memory , Insulin/immunology , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Proinsulin/genetics , Proinsulin/immunology , Receptors, Antigen, T-Cell/metabolism
10.
J Immunol ; 194(9): 4567-76, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25810394

ABSTRACT

Inducible BALT (iBALT) can amplify pulmonary or systemic inflammatory responses to the benefit or detriment of the host. We took advantage of the age-dependent formation of iBALT to interrogate the underlying mechanisms that give rise to this ectopic, tertiary lymphoid organ. In this study, we show that the reduced propensity for weanling as compared with neonatal mice to form iBALT in response to acute LPS exposure is associated with greater regulatory T cell expansion in the mediastinal lymph nodes. Ab- or transgene-mediated depletion of regulatory T cells in weanling mice upregulated the expression of IL-17A and CXCL9 in the lungs, induced a tissue neutrophilia, and increased the frequency of iBALT to that observed in neonatal mice. Remarkably, neutrophil depletion in neonatal mice decreased the expression of the B cell active cytokines, a proliferation-inducing ligand and IL-21, and attenuated LPS-induced iBALT formation. Taken together, our data implicate a role for neutrophils in lymphoid neogenesis. Neutrophilic inflammation is a common feature of many autoimmune diseases in which iBALT are present and pathogenic, and hence the targeting of neutrophils or their byproducts may serve to ameliorate detrimental lymphoid neogenesis in a variety of disease contexts.


Subject(s)
Inflammation/immunology , Lymphoid Tissue/immunology , Neutrophils/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Animals, Newborn , Cellular Microenvironment/immunology , Cytokines/biosynthesis , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/immunology , Lymphocyte Depletion , Lymphoid Tissue/metabolism , Male , Mice , Neutrophils/metabolism , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism
11.
Immunol Cell Biol ; 94(5): 509-19, 2016 05.
Article in English | MEDLINE | ID: mdl-26763864

ABSTRACT

Enhancement of regulatory T cell (Treg cell) frequency and function is the goal of many therapeutic strategies aimed at treating type 1 diabetes (T1D). The interleukin-2 (IL-2) pathway, which has been strongly implicated in T1D susceptibility in both humans and mice, is a master regulator of Treg cell homeostasis and function. We investigated how IL-2 pathway defects impact Treg cells in T1D-susceptible nonobese diabetic (NOD) mice in comparison with protected C57BL/6 and NOD congenic mice. NOD Treg cells were reduced in frequency specifically in the lymph nodes and expressed lower levels of CD25 and CD39/CD73 immunosuppressive molecules. In the spleen and blood, Treg cell frequency was preserved through expansion of CD25(low), effector phenotype Treg cells. Reduced CD25 expression led to decreased IL-2 signaling in NOD Treg cells. In vivo, treatment with IL-2-anti-IL-2 antibody complexes led to effective upregulation of suppressive molecules on NOD Treg cells in the spleen and blood, but had reduced efficacy on lymph node Treg cells. In contrast, NOD CD8(+) and CD4(+) effector T cells were not impaired in their response to IL-2 therapy. We conclude that NOD Treg cells have an impaired responsiveness to IL-2 that reduces their ability to compete for a limited supply of IL-2.


Subject(s)
Interleukin-2/metabolism , T-Lymphocytes, Regulatory/immunology , Alleles , Animals , Antigens, CD/metabolism , Cell Movement , Cell Proliferation , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Haplotypes/genetics , Immunosuppression Therapy , Lymph Nodes/metabolism , Lymphocyte Count , Mice, Inbred C57BL , Mice, Inbred NOD , Pancreas/pathology , Phenotype , STAT5 Transcription Factor/metabolism , Signal Transduction , Spleen/metabolism , Up-Regulation
12.
J Autoimmun ; 72: 118-25, 2016 08.
Article in English | MEDLINE | ID: mdl-27255733

ABSTRACT

Reestablishment of immune tolerance to the insulin-producing beta cells is the desired goal for type 1 diabetes (T1D) treatment and prevention. Immune tolerance to multiple islet antigens is defective in individuals with T1D, but the mechanisms involved are multifaceted and may involve loss of thymic and peripheral tolerance. In this review we discuss our current understanding of the varied mechanisms by which peripheral tolerance to islet antigens is maintained in healthy individuals where genetic protection from T1D is present and how this fails in those with genetic susceptibility to disease. Novel findings in regards to expression of neo-islet antigens, non-classical regulatory cell subsets and the impact of specific genetic variants on tolerance induction are discussed.


Subject(s)
Autoantigens/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Peripheral Tolerance/immunology , Animals , Central Tolerance/immunology , Humans , Models, Immunological , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology
13.
Immunol Cell Biol ; 93(6): 540-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25601274

ABSTRACT

Atopic dermatitis is a common pruritic and inflammatory skin disorder with unknown etiology. Most commonly occurring during early childhood, atopic dermatitis is associated with eczematous lesions and lichenification, in which the epidermis becomes hypertrophied resulting in thickening of the skin. In this study, we report an atopic dermatitis-like pathophysiology results in a murine model following the expression of the high-risk human papillomavirus (HPV) 16 oncoprotein E7 in keratinocytes under the keratin 14 promoter. We show that HPV16 E7 expression in the skin is associated with skin thickening, acanthosis and light spongiosis. Locally, HPV16 E7-expressing skin secreted high levels of thymic stromal lymphopoietin (TSLP) and contained increased numbers of innate lymphoid cells (ILCs). High levels of circulating immunoglobulin E were associated with increased susceptibility to skin allergy in a model of cutaneous challenge, and to airway bronchiolar inflammation, enhanced airway goblet cell metaplasia and mucus production in a model of atopic march. Surprisingly, skin pathology occurred independently of T cells and mast cells. Thus, our findings suggest that the expression of a single HPV oncogene in the skin can drive the onset of atopic dermatitis-like pathology through the induction of TSLP and type 2 ILC infiltration.


Subject(s)
Cytokines/biosynthesis , Dermatitis, Atopic/immunology , Dermatitis, Atopic/metabolism , Gene Expression , Papillomavirus E7 Proteins/genetics , Skin/immunology , Skin/metabolism , T-Lymphocyte Subsets/immunology , Animals , Dermatitis, Atopic/pathology , Dermatitis, Atopic/virology , Disease Models, Animal , Immunity, Innate , Interleukin-33/metabolism , Interleukins/metabolism , Mast Cells/immunology , Mast Cells/pathology , Mice , Mice, Transgenic , Phenotype , Skin/pathology , Skin/virology , T-Lymphocyte Subsets/pathology , Thymic Stromal Lymphopoietin
14.
Blood ; 121(6): 1049-58, 2013 Feb 07.
Article in English | MEDLINE | ID: mdl-23233664

ABSTRACT

Bone marrow (BM) or hematopoietic stem cell (HSC) transplantation is used as curative therapy for hematologic malignancies. Incorporation of gene therapy to drive tolerogenic expression of antigens is a promising strategy to overcome the limited long-term efficacy of autologous HSC transplantation for autoimmune diseases. HSC engraftment and tolerance induction is readily achieved after myeloablative or immune-depleting conditioning regardless of the cellular compartment in which antigen is expressed. It is unclear whether the efficiency of engraftment and tolerance induction is influenced by targeting antigen to specific cellular compartments. This is particularly important when using clinically feasible low-intensity conditioning aimed at preserving infectious immunity in individuals where immunologic memory exists to the autoantigen to be expressed. Here we demonstrate that, under immune-preserving conditions, confining expression of a transgenically expressed antigen to dendritic cells permits stable, long-term engraftment of genetically modified BM even when recipients are immune to the expressed antigen. In contrast, broader expression within the hematopoietic compartment leads to graft rejection and therapeutic failure because of antigen expression in HSCs. These findings are relevant to the clinical application of genetically engineered HSCs and provide evidence that careful selection of promoters for HSC-mediated gene therapy is important, particularly where tolerance is sought under immune-preserving conditions.


Subject(s)
Antigen-Presenting Cells/immunology , Immune Tolerance/immunology , Stem Cells/immunology , Transplantation Conditioning/methods , Analysis of Variance , Animals , Antigen-Presenting Cells/metabolism , Bone Marrow Transplantation/methods , CD11c Antigen/genetics , CD11c Antigen/immunology , CD11c Antigen/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Flow Cytometry , Hematopoietic Stem Cell Transplantation/methods , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Immune Tolerance/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/metabolism , Stem Cells/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
15.
Immunol Cell Biol ; 92(2): 156-63, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24217808

ABSTRACT

Small interfering RNAs (siRNAs) to inhibit oncogene expression and also to activate innate immune responses via Toll-like receptor (TLR) recognition have been shown to be beneficial as anti-cancer therapy in certain cancer models. In this study, we investigated the effects of local versus systemic delivery of such immune-stimulating Dicer-substrate siRNAs (IS-DsiRNAs) on a human papillomavirus (HPV)-driven tumour model. Localized siRNA delivery using intratumour injection of siRNA was able to increase siRNA delivery to the tumour compared with intravenous (IV) delivery and potently activated innate immune responses. However, IV injection remained the more effective delivery route for reducing tumour growth. Although IS-DsiRNAs activated innate immune cells and required interferon-α (IFNα) for full effect on tumour growth, we found that potent silencing siRNA acting independently of IFNα were overall more effective at inhibiting TC-1 tumour growth. Other published work utilising IS-siRNAs have been carried out on tumour models with low levels of major histocompatibility complex (MHC)-class 1, a target of natural killer cells that are potently activated by IS-siRNA. As TC-1 cells used in our study express high levels of MHC-class I, the addition of the immunostimulatory motifs may not be as beneficial in this particular tumour model. Our data suggest that selection of siRNA profile and delivery method based on tumour environment is crucial to developing siRNA-based therapies.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alphapapillomavirus/immunology , Neoplasms, Experimental/drug therapy , Papillomavirus Infections/drug therapy , RNA, Small Interfering/pharmacology , Tumor Virus Infections/drug therapy , Animals , Cell Line, Tumor , Histocompatibility Antigens Class I/immunology , Humans , Interferon-alpha/immunology , Mice , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Papillomavirus Infections/immunology , Papillomavirus Infections/pathology , Tumor Virus Infections/immunology , Tumor Virus Infections/pathology
16.
Blood ; 119(24): 5918-30, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22415754

ABSTRACT

Alloreactivity after transplantation is associated with profound immune suppression, and consequent opportunistic infection results in high morbidity and mortality. This immune suppression is most profound during GVHD after bone marrow transplantation where an inflammatory cytokine storm dominates. Contrary to current dogma, which avers that this is a T-cell defect, we demonstrate that the impairment lies within conventional dendritic cells (cDCs). Significantly, exogenous antigens can only be presented by the CD8(-) cDC subset after bone marrow transplantation, and inflammation during GVHD specifically renders the MHC class II presentation pathway in this population incompetent. In contrast, both classic and cross-presentation within MHC class I remain largely intact. Importantly, this defect in antigen processing can be partially reversed by TNF inhibition or the adoptive transfer of donor cDCs generated in the absence of inflammation.


Subject(s)
Antigen Presentation/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Graft vs Host Disease/immunology , Immunosuppression Therapy , Adoptive Transfer , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Marrow Transplantation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cross-Priming/immunology , Graft vs Host Disease/pathology , Histocompatibility Antigens Class II/immunology , Inflammation/immunology , Inflammation/pathology , Interferon-gamma/metabolism , Isoantigens/immunology , Mice , Mice, Transgenic , Peptides/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Necrosis Factor-alpha/metabolism
17.
Pharm Res ; 31(11): 3150-60, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24848340

ABSTRACT

PURPOSE: Safe, targeted delivery of therapeutics remains a focus of drug/gene delivery, the aim being to achieve optimal efficacy while minimising off-target delivery. Dendrimers have a vast array of potential applications and have great potential as gene and drug delivery tools. We previously reported the development of peptide dendrimers that effectively complexed DNA and that have distinct advantages over conventional spherical dendrimers. Here, to expand the application of peptide-based low generation dendrimers we tested their capacity to be transformed into linkers for antibody-based targeting of diverse payloads. METHODS: Peptide-based low-generation asymmetric dendrimers were generated and conjugated to partially-reduced antibodies specific for B cell surface antigens or an irrelevant antigen. Preservation of antigen binding by the antibodies and targeting of the conjugated dendrimers carrying a small molecule (biotin) or plasmid DNA payloads was tested. RESULTS: Peptide-based low generation dendrimers were efficiently and site-specifically conjugated to antibodies with retention of antigen-binding capacity. Altering the branching termini of dendrimers facilitated delivery of diverse payloads in vitro and in vivo. CONCLUSIONS: We propose that safe, non-toxic peptide dendrimers, which are readily synthesised and modifiable for a variety of applications, form the basis of a new family of biocompatible "linkers" with substantial potential for targeted delivery applications.


Subject(s)
Antibodies/administration & dosage , Antibodies/chemistry , B-Lymphocytes/drug effects , Dendrimers/administration & dosage , Dendrimers/chemistry , Peptides/administration & dosage , Peptides/chemistry , DNA/chemistry , Drug Delivery Systems/methods , Plasmids/chemistry
18.
J Immunol ; 189(12): 5622-31, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23144496

ABSTRACT

Adoptive T cell therapy uses the specificity of the adaptive immune system to target cancer and virally infected cells. Yet the mechanism and means by which to enhance T cell function are incompletely described, especially in the skin. In this study, we use a murine model of immunotherapy to optimize cell-mediated immunity in the skin. We show that in vitro-derived central but not effector memory-like T cells bring about rapid regression of skin-expressing cognate Ag as a transgene in keratinocytes. Local inflammation induced by the TLR7 receptor agonist imiquimod subtly yet reproducibly decreases time to skin graft rejection elicited by central but not effector memory T cells in an immunodeficient mouse model. Local CCL4, a chemokine liberated by TLR7 agonism, similarly enhances central memory T cell function. In this model, IL-2 facilitates the development in vivo of effector function from central memory but not effector memory T cells. In a model of T cell tolerogenesis, we further show that adoptively transferred central but not effector memory T cells can give rise to successful cutaneous immunity, which is dependent on a local inflammatory cue in the target tissue at the time of adoptive T cell transfer. Thus, adoptive T cell therapy efficacy can be enhanced if CD8(+) T cells with a central memory T cell phenotype are transferred, and IL-2 is present with contemporaneous local inflammation.


Subject(s)
Immunologic Memory/immunology , Skin/immunology , Skin/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Adoptive Transfer/methods , Animals , Cell Communication/immunology , Cells, Cultured , Epidermal Cells , Epidermis/immunology , Epidermis/transplantation , Immune Tolerance , Immunity, Cellular , Inflammation/immunology , Inflammation/surgery , Inflammation/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Skin/cytology , Skin Transplantation/immunology , Skin Transplantation/methods , Skin Transplantation/pathology , T-Lymphocyte Subsets/transplantation
19.
Proc Natl Acad Sci U S A ; 108(18): 7529-34, 2011 May 03.
Article in English | MEDLINE | ID: mdl-21502514

ABSTRACT

CD4(+)CD25(+) regulatory T cells (Treg) play a crucial role in the regulation of immune responses. Although many mechanisms of Treg suppression in vitro have been described, the mechanisms by which Treg modulate CD8(+) T cell differentiation and effector function in vivo are more poorly defined. It has been proposed, in many instances, that modulation of cytokine homeostasis could be an important mechanism by which Treg regulate adaptive immunity; however, direct experimental evidence is sparse. Here we demonstrate that CD4(+)CD25(+) Treg, by critically regulating IL-2 homeostasis, modulate CD8(+) T-cell effector differentiation. Expansion and effector differentiation of CD8(+) T cells is promoted by autocrine IL-2 but, by competing for IL-2, Treg limit CD8(+) effector differentiation. Furthermore, a regulatory loop exists between Treg and CD8(+) effector T cells, where IL-2 produced during CD8(+) T-cell effector differentiation promotes Treg expansion.


Subject(s)
Adaptive Immunity/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Homeostasis/immunology , Interleukin-2/metabolism , T-Lymphocytes, Regulatory/immunology , Analysis of Variance , Animals , Antibodies, Monoclonal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukin-2/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
20.
J Immunol ; 187(8): 4018-30, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21900177

ABSTRACT

Costimulation-deficient dendritic cells (DCs) prevent autoimmune disease in mouse models. However, autoimmune-prone mice and humans fail to control expansion of peripheral autoreactive effector memory T cells (T(EMs)), which resist immunoregulation by costimulation-deficient DCs. In contrast, activation of DC costimulation may be coupled with regulatory capacity. To test whether costimulatory DCs control T(EMs) and attenuate established autoimmune disease, we used RelB-deficient mice, which have multiorgan inflammation, expanded peripheral autoreactive T(EMs), and dysfunctional Foxp3(+) regulatory T cells (Tregs) cells and conventional DCs. T(EMs) were regulated by Foxp3(+) Tregs when costimulated by CD3/CD28-coated beads or wild-type DCs but not DCs deficient in RelB or CD80/CD86. After transfer, RelB and CD80/CD86-sufficient DCs restored tolerance and achieved a long-term cure of autoimmune disease through costimulation of T(EM) and Foxp3(+) Treg IFN-γ production, as well as induction of IDO by host APCs. IDO was required for regulation of T(EMs) and suppression of organ inflammation. Our data challenge the paradigm that costimulation-deficient DCs are required to regulate established autoimmune disease to avoid T(EM) activation and demonstrate cooperative cross-talk between costimulatory DCs, IFN-γ, and IDO-dependent immune regulation. IFN-γ and IDO activity may be good surrogate biomarkers measured against clinical efficacy in trials of autoimmune disease immunoregulation.


Subject(s)
Autoimmune Diseases/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Animals , Autoimmune Diseases/therapy , Dendritic Cells/transplantation , Flow Cytometry , Immune Tolerance/immunology , Immunomagnetic Separation , Inflammation/immunology , Inflammation/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Signal Transduction/immunology , Transcription Factor RelB/deficiency , Transcription Factor RelB/genetics , Transcription Factor RelB/immunology
SELECTION OF CITATIONS
SEARCH DETAIL