Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
J Immunol ; 205(12): 3358-3371, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33158954

ABSTRACT

Immune dysregulation during sepsis is mediated by an imbalance of T cell costimulatory and coinhibitory signaling. CD28 is downregulated during sepsis and is significantly altered on memory versus naive T cells. Thus, to study the role of CD28 during sepsis in a more physiologically relevant context, we developed a "memory mouse" model in which animals are subjected to pathogen infections to generate immunologic memory, followed by sepsis induction via cecal ligation and puncture. Using this system, we show that agonistic anti-CD28 treatment resulted in worsened survival in naive septic animals but conferred a significant survival advantage in immunologically experienced septic animals. Mechanistically, this differential response was driven by the ability of CD28 agonism to elicit IL-10 production from regulatory T cells uniquely in memory but not naive mice. Moreover, elevated IL-10 released by activated regulatory T cells in memory mice inhibited sepsis-induced T cell apoptosis via the antiapoptotic protein Bcl-xL. Together, these data demonstrate that immunologic experience is an important parameter that affects sepsis pathophysiology and can fundamentally change the outcome of modulating the CD28 pathway during sepsis. This study suggests that testing therapeutic strategies in immunologically experienced hosts may be one way to increase the physiologic relevance of rodent models in sepsis research.


Subject(s)
CD28 Antigens , Immunologic Memory , Interleukin-10/immunology , Sepsis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/immunology , Male , Mice , bcl-X Protein/immunology
2.
Eur J Immunol ; 50(5): 695-704, 2020 05.
Article in English | MEDLINE | ID: mdl-31943150

ABSTRACT

CD5 is expressed on T cells and a subset of B cells (B1a). It can attenuate TCR signalling and impair CTL activation and is a therapeutic targetable tumour antigen expressed on leukemic T and B cells. However, the potential therapeutic effect of functionally blocking CD5 to increase T cell anti-tumour activity against tumours (including solid tumours) has not been explored. CD5 knockout mice show increased anti-tumour immunity: reducing CD5 on CTLs may be therapeutically beneficial to enhance the anti-tumour response. Here, we show that ex vivo administration of a function-blocking anti-CD5 MAb to primary mouse CTLs of both tumour-naïve mice and mice bearing murine 4T1 breast tumour homografts enhanced their capacity to respond to activation by treatment with anti-CD3/anti-CD28 MAbs or 4T1 tumour cell lysates. Furthermore, it enhanced TCR signalling (ERK activation) and increased markers of T cell activation, including proliferation, CD69 levels, IFN-γ production, apoptosis and Fas receptor and Fas ligand levels. Finally, CD5 function-blocking MAb treatment enhanced the capacity of CD8+ T cells to kill 4T1-mouse tumour cells in an ex vivo assay. These data support the potential of blockade of CD5 function to enhance T cell-mediated anti-tumour immunity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , CD28 Antigens/immunology , CD5 Antigens/immunology , Mammary Neoplasms, Experimental/drug therapy , T-Lymphocytes, Cytotoxic/drug effects , Animals , Antineoplastic Agents, Immunological , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/genetics , CD5 Antigens/antagonists & inhibitors , CD5 Antigens/genetics , Cell Extracts/pharmacology , Cell Line, Tumor , Cell Proliferation , Fas Ligand Protein/genetics , Fas Ligand Protein/immunology , Female , Gene Expression Regulation , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymphocyte Activation/drug effects , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/immunology , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , fas Receptor/genetics , fas Receptor/immunology
3.
J Neuroinflammation ; 18(1): 104, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33931070

ABSTRACT

BACKGROUND: Cell transplantation-based treatments for neurological disease are promising, yet graft rejection remains a major barrier to successful regenerative therapies. Our group and others have shown that long-lasting tolerance of transplanted stem cells can be achieved in the brain with systemic application of monoclonal antibodies blocking co-stimulation signaling. However, it is unknown if subsequent injury and the blood-brain barrier breach could expose the transplanted cells to systemic immune system spurring fulminant rejection and fatal encephalitis. Therefore, we investigated whether delayed traumatic brain injury (TBI) could trigger graft rejection. METHODS: Glial-restricted precursor cells (GRPs) were intracerebroventricularly transplanted in immunocompetent neonatal mice and co-stimulation blockade (CoB) was applied 0, 2, 4, and 6 days post-grafting. Bioluminescence imaging (BLI) was performed to monitor the grafted cell survival. Mice were subjected to TBI 12 weeks post-transplantation. MRI and open-field test were performed to assess the brain damage and behavioral change, respectively. The animals were decapitated at week 16 post-transplantation, and the brains were harvested. The survival and distribution of grafted cells were verified from brain sections. Hematoxylin and eosin staining (HE) was performed to observe TBI-induced brain legion, and neuroinflammation was evaluated immunohistochemically. RESULTS: BLI showed that grafted GRPs were rejected within 4 weeks after transplantation without CoB, while CoB administration resulted in long-term survival of allografts. BLI signal had a steep rise following TBI and subsequently declined but remained higher than the preinjury level. Open-field test showed TBI-induced anxiety for all animals but neither CoB nor GRP transplantation intensified the symptom. HE and MRI demonstrated a reduction in TBI-induced lesion volume in GRP-transplanted mice compared with non-transplanted mice. Brain sections further validated the survival of grafted GRPs and showed more GRPs surrounding the injured tissue. Furthermore, the brains of post-TBI shiverer mice had increased activation of microglia and astrocytes compared to post-TBI wildtype mice, but infiltration of CD45+ leukocytes remained low. CONCLUSIONS: CoB induces sustained immunological tolerance towards allografted cerebral GRPs which is not disrupted following TBI, and unexpectedly TBI may enhance GRPs engraftment and contribute to post-injury brain tissue repair.


Subject(s)
Brain Injuries, Traumatic , Graft Rejection/immunology , Immune Tolerance/immunology , Neural Stem Cells/transplantation , Stem Cell Transplantation/methods , Allografts , Animals , Antibodies, Monoclonal/pharmacology , B7-1 Antigen/antagonists & inhibitors , B7-2 Antigen/antagonists & inhibitors , CD28 Antigens/antagonists & inhibitors , CD40 Antigens/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Neuroglia/transplantation
4.
Eur J Immunol ; 49(7): 1117-1126, 2019 07.
Article in English | MEDLINE | ID: mdl-31002172

ABSTRACT

The first-in-human clinical trial of the CD28-specific monoclonal antibody (mAb) TGN1412 resulted in a life-threatening cytokine release syndrome. Although TGN1412 was designed as IgG4, known for weak Fc:Fcγ receptor (FcγR) interactions, these interactions contributed to TGN1412-induced T-cell activation. Using cell lines (TFs) expressing human FcγRI, -IIa, -IIb, or -III, we show that TGN1412 and TGN1412 as IgG1 and IgG2 are bound by FcγRs as it can be deduced from literature. However, upon coculture of TGN1412-decorated T cells with TFs or human primary blood cells, we observed that binding capacities by FcγRs do not correlate with the strength of the mediated effector function. FcγRIIa and FcγRIIb, showing no or very minor binding to TGN1412, mediated strongest T cell proliferation, while high-affinity FcγRI, exhibiting strong TGN1412 binding, mediated hardly any T-cell proliferation. These findings are of biological relevance because we show that FcγRI binds TGN1412, thus prevents binding to FcγRIIa or FcγRIIb, and consequently disables T-cell proliferation. In line with this, FcγRI- FcγRII+ but not FcγRI+ FcγRII+ monocytes mediate TGN1412-induced T-cell proliferation. Collectively, by using TGN1412 as example, our results indicate that binding of monomeric IgG subclasses does not predict the FcγR-mediated effector function, which has major implications for the design of therapeutic mAbs.


Subject(s)
Antibodies, Monoclonal, Humanized/metabolism , Cytokine Release Syndrome/immunology , Immunoglobulin G/metabolism , Immunotherapy/adverse effects , Monocytes/immunology , Receptors, IgG/metabolism , T-Lymphocytes/immunology , Animals , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , CD28 Antigens/antagonists & inhibitors , Cell Line , Cell Proliferation , Cytokine Release Syndrome/etiology , Humans , Lymphocyte Activation , Mice , Protein Binding , Receptors, IgG/genetics
5.
J Nat Prod ; 83(6): 1804-1809, 2020 06 26.
Article in English | MEDLINE | ID: mdl-32539381

ABSTRACT

Hypersonins A-D (1-4), four 1,2-seco-homoadamantane type polycyclic polyprenylated acylphloroglucinols (PPAPs) possessing a new bicyclo[4.3.1]decane-3-methoxycarbonyl architecture, were obtained from Hypericum wilsonii. The structures of hypersonins A-D were identified by spectroscopic data, electronic circular dichroism comparison, and X-ray crystallographic data. Hypersonins A-D are the first seco-homoadamantane-type PPAPs with cleavage at the C-1-C-2 bond. Hypersonin A (1) showed moderate inhibitory activity to anti-CD3/anti-CD28 monoclonal antibody-induced proliferation of murine splenocytes, with an IC50 value of 8.3 ± 0.2 µM.


Subject(s)
Hypericum/chemistry , Polycyclic Compounds/pharmacology , Animals , Antibodies, Blocking , Antineoplastic Agents, Phytogenic/chemistry , CD28 Antigens/antagonists & inhibitors , CD3 Complex/antagonists & inhibitors , Cell Proliferation/drug effects , Circular Dichroism , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred BALB C , Molecular Structure , Polycyclic Compounds/chemistry , Spleen/cytology , X-Ray Diffraction
6.
J Nat Prod ; 83(9): 2672-2678, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32897070

ABSTRACT

Varioxepine B (1), an oxepine-containing diketopiperazine derivative, was isolated from a marine-derived Aspergillus terreus strain. The structure of 1 was identified by spectroscopic experiments, single-crystal X-ray diffraction analysis, and electronic circular dichroism calculations. It is noteworthy that 1 could suppressed murine splenocyte proliferation activated by concanavalin A (Con A) in vitro. More importantly, in Con A-challenged mice, pretreatment with 1 obviously decreased the generation of proinflammatory cytokines and ameliorated liver injury. Meanwhile, 1 also exhibited inhibitory activity in anti-CD3/anti-CD28 monoclonal antibodies (mAbs)-induced murine splenocytes and human T cell proliferation as well as both Th1 and Th2 cytokine production.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Oxepins/chemistry , Piperazines/chemistry , Piperazines/therapeutic use , Animals , Aspergillus/chemistry , CD28 Antigens/antagonists & inhibitors , CD3 Complex , Cell Proliferation , Chemical and Drug Induced Liver Injury/pathology , Concanavalin A/toxicity , Cytokines/biosynthesis , Diketopiperazines , Humans , Liver/pathology , Male , Mice , Mice, Inbred BALB C , Molecular Structure , Spleen/cytology , Spleen/drug effects , T-Lymphocytes/drug effects , X-Ray Diffraction
7.
Genes Immun ; 20(6): 462-472, 2019 07.
Article in English | MEDLINE | ID: mdl-29977032

ABSTRACT

The low affinity Fcγ receptor, FcγRIIA, harbors a common missense mutation, rs1801274 (G>A, Arg131His) that modifies binding affinity to human IgG2 and mouse IgG1 antibodies and is associated with increased risk of autoimmune disease. Despite the important role of the Arg131His variant, little is understood about heterozygous genotype effects on global gene expression and cytokine production during an FcγR-dependent response. To address this gap in knowledge, we treated human whole-blood samples from 130 individuals with mouse IgG1 anti-CD3 and anti-CD28 antibodies and characterized the genome-wide gene expression profiles and cytokine production among individuals stratified by rs1801274 genotype. Our analysis revealed that the levels of four cytokines (IFNγ, IL-12, IL-2, TNFα) and global gene expression patterns differed between all three genotype classes. Surprisingly, the heterozygotes showed suboptimal T cell activation compared to cells from individuals homozygous for the higher-affinity FcγRIIA allele (GG; Arg/Arg). The results of this study demonstrate that IgG response varies among all rs1801274 genotype classes and results in profound differences in both cytokine responses and gene expression patterns in blood leukocytes. Because even heterozygotes showed dampened global responses, our data may provide insight into the heterogeneity of outcomes in cytokine release assays and immunotherapy efficacy.


Subject(s)
Antibodies/pharmacology , CD28 Antigens/immunology , CD3 Complex/immunology , Leukocytes/immunology , Polymorphism, Genetic , Receptors, IgG/genetics , Adolescent , Adult , Aged , Alleles , CD28 Antigens/antagonists & inhibitors , CD3 Complex/antagonists & inhibitors , Child , Genotype , Heterozygote , Homozygote , Humans , Interferon-gamma/blood , Interferon-gamma/metabolism , Interleukin-12/blood , Interleukin-12/metabolism , Interleukin-2/blood , Interleukin-2/metabolism , Leukocytes/metabolism , Middle Aged , T-Lymphocytes/metabolism , Transcriptome/immunology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism , Young Adult
8.
Am J Transplant ; 19(8): 2199-2209, 2019 08.
Article in English | MEDLINE | ID: mdl-30801917

ABSTRACT

CTLA-4Ig (belatacept) blocks the CD80/CD86 ligands for both CD28 and CTLA-4; thus, in addition to the intended effect of blocking CD28-mediated costimulation, belatacept also has the unintended effect of blocking CTLA-4-mediated coinhibition. Recently, anti-CD28 domain antibodies (dAb) that selectively target CD28 while leaving CTLA-4 intact were shown to more effectively inhibit alloimmune responses and prolong graft survival. However, the impact of selective CD28 blockade on protective immunity has not been extensively investigated. Here, we sought to compare the impact of CTLA-4Ig vs anti-CD28dAb on CD8+ T cell immunity to a transplant-relevant pathogen, a murine homolog of Epstein-Barr virus. Mice were infected with murine gammaherpesvirus-68 (MHV) and treated with vehicle, CTLA-4Ig, or anti-CD28dAb. Although anti-CD28dAb resulted in a decrease in virus-specific CD8+ T cell numbers as compared to CTLA-4Ig, cytolytic function and the expression of markers of high-quality effectors were not different from CTLA-4Ig treated animals. Importantly, MHV-68 viral load was not different between the treatment groups. These results suggest that preserved CTLA-4 coinhibition limits MHV-specific CD8+ T cell accumulation, but the population that remains retains cytolytic function and migratory capacity and is not inferior in its ability to control viral burden relative to T cell responses in CTLA-4Ig-treated animals.


Subject(s)
Abatacept/pharmacology , Antibodies, Monoclonal/pharmacology , CD28 Antigens/antagonists & inhibitors , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/immunology , Animals , CD28 Antigens/immunology , Epstein-Barr Virus Infections/drug therapy , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/drug effects , Mice , Mice, Inbred C57BL
9.
Am J Transplant ; 19(8): 2350-2357, 2019 08.
Article in English | MEDLINE | ID: mdl-30891931

ABSTRACT

Naïve T cell activation requires antigen presentation combined with costimulation through CD28, both of which optimally occur in secondary lymphoid tissues such as lymph nodes and the spleen. Belatacept impairs CD28 costimulation by binding its ligands, CD80 and CD86, and in doing so, impairs de novo alloimmune responses. However, in most patients belatacept is ineffective in preventing allograft rejection when used as a monotherapy, and adjuvant therapy is required for control of costimulation-blockade resistant rejection (CoBRR). In rodent models, impaired access to secondary lymphoid tissues has been demonstrated to reduce alloimmune responses to vascularized allografts. Here we show that surgical maneuvers, lymphatic ligation, and splenectomy, designed to anatomically limit access to secondary lymphoid tissues, control CoBRR and facilitate belatacept monotherapy in a nonhuman primate model of kidney transplantation without adjuvant immunotherapy. We further demonstrate that animals sustained on belatacept monotherapy progressively develop an increasingly naïve T and B cell repertoire, an effect that is accelerated by splenectomy and lost at the time of belatacept withdrawal and rejection. These pilot data inform the role of secondary lymphoid tissues on the development of CoBRR and the use of costimulation molecule-focused therapies.


Subject(s)
Abatacept/therapeutic use , CD28 Antigens/antagonists & inhibitors , Graft Rejection/prevention & control , Graft Survival/immunology , Immunosuppressive Agents/therapeutic use , Kidney Transplantation/mortality , Lymphoid Tissue/immunology , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Graft Rejection/etiology , Graft Rejection/pathology , Graft Survival/drug effects , Immunologic Memory , Immunotherapy , Kidney Transplantation/adverse effects , Lymphoid Tissue/drug effects , Primates , Splenectomy , Survival Rate , Transplantation, Homologous
10.
J Autoimmun ; 101: 121-130, 2019 07.
Article in English | MEDLINE | ID: mdl-31030958

ABSTRACT

Despite the development of several targeted therapies for rheumatoid arthritis (RA), there is still no reliable drug-specific predictor to assist rheumatologists in selecting the most effective targeted therapy for each patient. Recently, a gene signature caused by impaired induction of PTPN22 in anti-CD3 stimulated peripheral blood mononuclear cells (PBMC) was observed in healthy at-risk individuals. However, the downstream target genes of PTPN22 and the molecular mechanisms regulating its expression are still poorly understood. Here we report that the PTPN22 gene signature is also present in PBMC from patients with active RA and can be reversed after effective treatment. The expression of PTPN22 correlates with that of more than 1000 genes in Th cells of anti-CD3 stimulated PBMC of healthy donors and is inhibited by TNFα or CD28 signals, but not IL-6, through distinct mechanisms. In addition, the impaired induction of PTPN22 in PBMC of patients with active RA can be normalized in vitro by several targeted therapies. More importantly, the in vitro normalization of PTPN22 expression correlates with clinical response to the targeted therapies in a longitudinal RA cohort. Thus, in vitro normalization of PTPN22 expression by targeted therapies can potentially be used to predict clinical response.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Gene Expression Regulation , Molecular Targeted Therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Adult , Aged , Arthritis, Rheumatoid/diagnosis , Biomarkers , CD28 Antigens/antagonists & inhibitors , Female , Humans , Middle Aged , Molecular Targeted Therapy/methods , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Tumor Necrosis Factor-alpha/antagonists & inhibitors
11.
Toxicol Appl Pharmacol ; 372: 57-69, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30914376

ABSTRACT

Cytokine release syndrome (CRS) is a serious and potentially life-threatening complication typically associated with biological drug products. Pre-clinical testing in vitro and in vivo studies using non-human primates had failed to reliably predict CRS. To determine if bone marrow-thymus-liver (BLT) humanized mice with a fully engrafted human immune system or a CD34-humanized mouse model could predict CRS, we tested an anti-CD28 monoclonal antibody (mAb) similar to TGN1412. This TGN1412 analogue (TGN1412A) was initially tested in vitro and found to produce significant dose-dependent increases in cytokine production. For in vivo studies, adalimumab, an anti-tumor necrosis factor-alpha antibody known not to cause CRS, served as a negative control. We evaluated immune cell activation and cytokine expression in three independent experiments. In BLT humanized mice, significant increases in levels of human cytokines were identified in animals treated with anti-CD28 mAb. As expected, CD28+ cell detection was strongly reduced in the anti-CD28 treated group. Increased T cell activation was also observed. The control group did not show reductions in CD28+ T-cells and did not experience increased cytokine levels. Responses by CD34-humanized mice showed no significant differences between adalimumab and anti-CD28 treatment at doses used to test BLT-humanized mice. These results suggest that the TGN1412A produces similar results in vitro to the original TGN1412 monoclonal antibody. The BLT immune humanized mice but not the CD34 humanized mice produce both robust and specific cytokine responses and may represent a pre-clinical model to identify CRS.


Subject(s)
Antibodies, Monoclonal, Humanized/toxicity , CD28 Antigens/antagonists & inhibitors , Cytokine Release Syndrome/etiology , Cytokines/blood , T-Lymphocytes/drug effects , Animals , Antigens, CD34/immunology , CD28 Antigens/blood , CD28 Antigens/immunology , Cells, Cultured , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/immunology , Cytokines/immunology , Female , Hematopoietic Stem Cell Transplantation , Humans , Liver Transplantation , Mice, Inbred NOD , Mice, SCID , Risk Assessment , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Thymus Gland/embryology , Thymus Gland/transplantation
12.
Diabetologia ; 61(8): 1811-1816, 2018 08.
Article in English | MEDLINE | ID: mdl-29845333

ABSTRACT

AIMS/HYPOTHESIS: The CD28/B7 interaction is critical for both effector T cell activation and forkhead box P3 (FOXP3)+ regulatory T cell (Treg) generation and homeostasis, which complicates the therapeutic use of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4)-immunoglobulin fusion protein (CTLA-4Ig) in autoimmunity. Here, we evaluated the impact of a simultaneous and selective blockade of the CD28 and mammalian target of rapamycin (mTOR) pathways in the NOD mouse model of type 1 diabetes. METHODS: NOD mice were treated with PEGylated anti-CD28 Fab' antibody fragments (PV1-polyethylene glycol [PEG], 10 mg/kg i.p., twice weekly), rapamycin (1 mg/kg i.p., twice weekly) or a combination of both drugs. Diabetes incidence, pancreatic islet infiltration and autoreactive T cell responses were analysed. RESULTS: We report that 4 week administration of PV1-PEG combined with rapamycin effectively controlled the progression of autoimmune diabetes in NOD mice at 10 weeks of age by reducing T cell activation and migration into the pancreas. Treatment with rapamycin alone was without effect, as was PV1-PEG monotherapy initiated at 4, 6 or 10 weeks of age. Prolonged PV1-PEG administration (for 10 weeks) accelerated diabetes development associated with impaired peripheral Treg homeostasis. This effect was not observed with the combined treatment. CONCLUSIONS/INTERPRETATION: CD28 antagonist and rapamycin treatment act in a complementary manner to limit T cell activation and infiltration of pancreatic islets and diabetes development. These data provide new perspectives for the treatment of autoimmune diabetes and support the therapeutic potential of protocols combining antagonists of CD28 (presently in clinical development) and the mTOR pathway.


Subject(s)
CD28 Antigens/antagonists & inhibitors , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/prevention & control , Immunoglobulin Fab Fragments/pharmacology , Sirolimus/pharmacology , Animals , Cell Movement , Disease Progression , Drug Synergism , Female , Homeostasis , Interferon-gamma/metabolism , Islets of Langerhans/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, Nude , Pancreas/metabolism , T-Lymphocytes/cytology , T-Lymphocytes, Regulatory/immunology
13.
Am J Transplant ; 18(1): 89-101, 2018 01.
Article in English | MEDLINE | ID: mdl-28637095

ABSTRACT

Donor-specific antibodies (DSAs) are a barrier to improved long-term outcomes after kidney transplantation. Costimulation blockade with CTLA4-Ig has shown promise as a potential therapeutic strategy to control DSAs. T follicular helper (Tfh) cells, a subset of CD4+ T cells required for optimal antibody production, are reliant on the CD28 costimulatory pathway. We have previously shown that selective CD28 blockade leads to superior allograft survival through improved control of CD8+ T cells relative to CTLA4-Ig, but the impact of CD28-specific blockade on CD4+ Tfh cells is unknown. Thus, we identified and characterized donor-reactive Tfh cells in a murine skin transplant model and then used this model to evaluate the impact of selective CD28 blockade with an anti-CD28 domain antibody (dAb) on the donor-specific Tfh cell-mediated immune response. We observed that the anti-CD28 dAb led to superior inhibition of donor-reactive CXCR5+ PD-1high Tfh cells, CD95+ GL7+ germinal center B cells and DSA formation compared with CTLA4-Ig. Interestingly, donor-reactive Tfh cells differentially upregulated CTLA4 expression, suggesting an important role for CTLA4 in mediating the superior inhibition observed with the anti-CD28 dAb. Therefore, selective CD28 blockade as a novel approach to control Tfh cell responses and prevent DSA after kidney transplantation warrants further study.


Subject(s)
Abatacept/immunology , Antibody Formation/immunology , CD28 Antigens/antagonists & inhibitors , Graft Rejection/immunology , Skin Transplantation/adverse effects , T-Lymphocytes, Helper-Inducer/immunology , Tissue Donors , Animals , Graft Rejection/etiology , Graft Survival/immunology , Mice , Mice, Inbred C57BL
14.
Eur J Immunol ; 47(8): 1368-1376, 2017 08.
Article in English | MEDLINE | ID: mdl-28631301

ABSTRACT

Systemic lupus erythematosus (SLE) is a chronic systemic inflammatory disease. Autoantibodies (autoAbs) against double-stranded DNA (ds DNA), the hallmark of lupus, are produced and maintained by the interaction between auto-reactive B cells and CD4+ T cells. This interplay is controlled by the CD28/CD80-86/CTLA-4 axis. Here we investigated whether selective blockade of CD28-CD80/86 co-stimulatory interactions abrogates lupus nephritis development in a murine model of SLE. To this aim, NZB/NZW F1 mice were treated for 3 months, either with an anti-CD28 Fab' fragment or a control Fab'-IgG. The effect of CD28 blockade on lupus nephritis onset, survival, production of anti-ds DNA antibodies and costimulatory molecules was evaluated. CD28 blockade prevented the development of lupus nephritis and prolonged survival during the 3-month treatment and 12 weeks after. Furthermore, the production of anti-ds DNA autoAbs was decreased. Lastly, the protective effect of CD28 blockade was associated with increased intrarenal expression of the immunoregulatory molecule, Indoleamine 2, 3-dioxygenase, of the co-inhibitory receptor programmed cell-Death - 1 (PD-1) and of its ligand programmed death ligand - 1 (PDL-1).In conclusion, CD28 blockade prevented the development of lupus nephritis in NZB/NZW F1 mice. This immunomodulatory strategy is a promising candidate for SLE therapy in humans.


Subject(s)
Antibodies, Antinuclear/blood , Autoantibodies/blood , CD28 Antigens/antagonists & inhibitors , Immunotherapy/methods , Lupus Nephritis/prevention & control , Animals , Antibodies, Antinuclear/immunology , Autoantibodies/immunology , B-Lymphocytes/immunology , B7-1 Antigen/antagonists & inhibitors , B7-1 Antigen/immunology , B7-1 Antigen/metabolism , B7-2 Antigen/antagonists & inhibitors , B7-2 Antigen/immunology , B7-2 Antigen/metabolism , CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Immunoglobulin Fab Fragments/administration & dosage , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Kidney/immunology , Kidney/pathology , Lupus Erythematosus, Systemic/immunology , Lupus Nephritis/immunology , Mice , Mice, Inbred NZB , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology
15.
J Autoimmun ; 94: 45-55, 2018 11.
Article in English | MEDLINE | ID: mdl-30054208

ABSTRACT

A missense C1858T single nucleotide polymorphism within PTPN22 is a strong genetic risk factor for the development of multiple autoimmune diseases. PTPN22 encodes a protein tyrosine phosphatase that negatively regulates immuno-receptor proximal Src and Syk family kinases. Notably, PTPN22 negatively regulates kinases downstream of T-cell receptor (TCR) and LFA-1, thereby setting thresholds for T-cell activation. Alterations to the quality of TCR and LFA-1 engagement at the immune synapse and the regulation of downstream signals can have profound effects on the type of effector T-cell response induced. Here we describe how IFNγ+ Th1 responses are potentiated in Ptpn22-/- T-cells and in T-cells from mice expressing Ptpn22R619W (the mouse orthologue of the human genetic variant) as they age, or following repeated immune challenge, and explore the mechanisms contributing to the expansion of Th1 cells. Specifically, we uncover two LFA-1-ICAM dependent mechanisms; one T-cell intrinsic, and one T-cell extrinsic. Firstly, we found that in vitro anti-CD3/LFA-1 induced Th1 responses were enhanced in Ptpn22-/- T-cells compared to WT, whereas anti-CD3/anti-CD28 induced IFNy responses were similar. These data were associated with an enhanced ability of Ptpn22-/- T-cells to engage ICAM-1 at the immune synapse when incubated on planar lipid bilayers, and to form conjugates with dendritic cells. Secondly, we observed a T-cell extrinsic mechanism whereby repeated stimulation of WT OT-II T-cells with LPS and OVA323-339 pulsed Ptpn22-/- bone marrow derived dendritic cells (BMDCs) was sufficient to enhance Th1 cell development compared to WT BMDCs. Furthermore, this response could be reversed by LFA-1 blockade. Our data point to two related but distinct mechanisms by which PTPN22 regulates LFA-1 dependent signals to enhance Th1 development, highlighting how perturbations to PTPN22 function over time to regulate the balance of the immune response.


Subject(s)
Arthritis, Experimental/immunology , Dendritic Cells/immunology , Lymphocyte Function-Associated Antigen-1/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/immunology , Th1 Cells/immunology , Animals , Antibodies/pharmacology , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/antagonists & inhibitors , CD3 Complex/genetics , CD3 Complex/immunology , Cell Proliferation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/pathology , Gene Expression Regulation , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Lipid Bilayers/chemistry , Lipid Bilayers/immunology , Lipopolysaccharides/pharmacology , Lymphocyte Function-Associated Antigen-1/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/pharmacology , Peptide Fragments/pharmacology , Polymorphism, Single Nucleotide , Protein Tyrosine Phosphatase, Non-Receptor Type 22/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Th1 Cells/drug effects , Th1 Cells/pathology
16.
J Immunol ; 197(6): 2045-50, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27591335

ABSTRACT

T cell activation is a complex process that requires multiple cell signaling pathways, including a primary recognition signal and additional costimulatory signals. TCR signaling in the absence of costimulatory signals can lead to an abortive attempt at activation and subsequent anergy. One of the best-characterized costimulatory pathways includes the Ig superfamily members CD28 and CTLA-4 and their ligands CD80 and CD86. The development of the fusion protein CTLA-4-Ig as an experimental and subsequent therapeutic tool is one of the major success stories in modern immunology. Abatacept and belatacept are clinically approved agents for the treatment of rheumatoid arthritis and renal transplantation, respectively. Future interventions may include selective CD28 blockade to block the costimulatory potential of CD28 while exploiting the coinhibitory effects of CTLA-4.


Subject(s)
Autoimmunity , CD28 Antigens/physiology , Animals , Antibodies, Monoclonal/therapeutic use , CD28 Antigens/antagonists & inhibitors , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/physiology , Clinical Trials as Topic , Humans , Immunosuppressive Agents/pharmacology , Kidney Transplantation , Signal Transduction/physiology
17.
J Immunol ; 197(2): 533-40, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27288533

ABSTRACT

Blocking of costimulatory CD28/B7 and CD40/CD40L interactions is an experimental approach to immune suppression and tolerance induction. We previously reported that administration of a combination of CTLA-4Ig and MR1 (anti-CD40L mAb) for blockade of these interactions induces tolerance in a fully mismatched allogeneic splenocyte transfer model in mice. We now used this model to study whether regulatory T cells (Tregs) contribute to immune suppression and why both pathways have to be blocked simultaneously. Mice were injected with allogeneic splenocytes, CD4(+) T cells, or CD8(+) T cells and treated with MR1 mAb and different doses of CTLA-4Ig. The graft-versus-host reaction of CD4(+) T cells, but not of CD8(+) T cells, was inhibited by MR1. CTLA-4Ig was needed to cover CD8(+) T cells but had only a weak effect on CD4(+) T cells. Consequently, only the combination provided full protection when splenocytes were transferred. Importantly, MR1 and low-dose CTLA-4Ig treatment resulted in a relative increase in Tregs, and immune suppressive efficacy was abolished in the absence of Tregs. High-dose CTLA-4Ig treatment, in contrast, prevented Treg expansion and activity, and in combination with MR1 completely inhibited CD4(+) and CD8(+) T cell activation in a Treg-independent manner. In conclusion, MR1 and CTLA-4Ig act synergistically as they target different T cell populations. The contribution of Tregs to immune suppression by costimulation blockade depends on the concentration of CTLA-4Ig and thus on the degree of available CD28 costimulation.


Subject(s)
Graft vs Host Disease/immunology , Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B7-1 Antigen/antagonists & inhibitors , B7-1 Antigen/immunology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/immunology , CD40 Antigens/antagonists & inhibitors , CD40 Antigens/immunology , CD40 Ligand/antagonists & inhibitors , CD40 Ligand/immunology , Disease Models, Animal , Flow Cytometry , Immunosuppression Therapy , Mice
18.
J Immunol ; 197(12): 4593-4602, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27849166

ABSTRACT

FR104 is a monovalent pegylated Fab' Ab, antagonist of CD28, under development for treatment of transplant rejection and autoimmune diseases. In contrast to CD80/86 antagonists (CTLA4-Ig), FR104 selectively blunts CD28 costimulation while sparing CTLA-4 and PD-L1 coinhibitory signals. In the present work, FR104 has been evaluated in a first-in-human study to evaluate the safety, pharmacokinetics, pharmacodynamics, and potency of i.v. administrations in healthy subjects. Sixty-four subjects were randomly assigned to four single ascending dose groups, two double dose groups and four single ascending dose groups challenged with keyhole limpet hemocyanin. Subjects were followed up over a maximum of 113 d. Overall, the pharmacokinetics of FR104 after a single and double infusions was approximately linear at doses ≥0.200 mg/kg. CD28 receptor occupancy by FR104 was saturated at the first sampling time point (0.5 h) at doses above 0.02 mg/kg and returned to 50% in a dose-dependent manner, by day 15 (0.020 mg/kg) to 85 (1.500 mg/kg). FR104 was well tolerated, with no evidence of cytokine-release syndrome and no impact on blood lymphocyte subsets. Inhibition of anti-keyhole limpet hemocyanin Ab response was dose-dependent in FR104 recipients and was already apparent at a dose of 0.02 mg/kg. Abs to FR104 were detected in 22/46 (48%) of FR104 recipients and only 1/46 (2.2%) was detected during drug exposure. In conclusion, selective blockade of CD28 with FR104 was safe and well tolerated at the doses tested. The observed immunosuppressive activity indicated that FR104 has potential to show clinical activity in the treatment of immune-mediated diseases.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases/therapy , Graft Rejection/prevention & control , Immunoglobulin Fab Fragments/therapeutic use , Immunotherapy/methods , Organ Transplantation , Administration, Intravenous , Adult , Antibodies, Monoclonal/pharmacology , Autoimmune Diseases/immunology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/immunology , Clinical Protocols , Cohort Studies , Female , Follow-Up Studies , Graft Rejection/immunology , Healthy Volunteers , Humans , Immunity, Humoral/drug effects , Immunosuppressive Agents , Lymphocyte Count , Male , Middle Aged
19.
J Immunol ; 196(1): 274-83, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26597009

ABSTRACT

Novel therapies that specifically target activation and expansion of pathogenic immune cell subsets responsible for autoimmune attacks are needed to confer long-term remission. Pathogenic cells in autoimmunity include memory T lymphocytes that are long-lived and present rapid recall effector functions with reduced activation requirements. Whereas the CD28 costimulation pathway predominantly controls priming of naive T cells and hence generation of adaptive memory cells, the roles of CD28 costimulation on established memory T lymphocytes and the recall of memory responses remain controversial. In contrast to CD80/86 antagonists (CTLA4-Ig), selective CD28 antagonists blunt T cell costimulation while sparing CTLA-4 and PD-L1-dependent coinhibitory signals. Using a new selective CD28 antagonist, we showed that Ag-specific reactivation of human memory T lymphocytes was prevented. Selective CD28 blockade controlled both cellular and humoral memory recall in nonhuman primates and induced long-term Ag-specific unresponsiveness in a memory T cell-mediated inflammatory skin model. No modification of memory T lymphocytes subsets or numbers was observed in the periphery, and importantly no significant reactivation of quiescent viruses was noticed. These findings indicate that pathogenic memory T cell responses are controlled by both CD28 and CTLA-4/PD-L1 cosignals in vivo and that selectively targeting CD28 would help to promote remission of autoimmune diseases and control chronic inflammation.


Subject(s)
Autoimmune Diseases/drug therapy , CD28 Antigens/antagonists & inhibitors , Immunologic Memory/immunology , Inflammation/drug therapy , Skin/immunology , T-Lymphocyte Subsets/immunology , Animals , Autoimmune Diseases/immunology , Autoimmunity/immunology , B7-H1 Antigen/immunology , CD28 Antigens/immunology , CTLA-4 Antigen/immunology , Humans , Inflammation/immunology , Lymphocyte Activation/immunology , Papio anubis , Signal Transduction/immunology , Skin/pathology , Virus Activation/immunology
20.
Article in English | MEDLINE | ID: mdl-27993846

ABSTRACT

Therapeutic strategies that target the latent HIV-1 reservoir in resting CD4+ T cells of infected individuals are always administered in the presence of combination antiretroviral therapy. Using a primary cell of HIV-1 latency, we evaluated whether different antiviral drug classes affected latency reversal (as assessed by extracellular virus production) by anti-CD3/CD28 monoclonal antibodies or bryostatin 1. We found that the nonnucleoside reverse transcriptase inhibitors efavirenz and rilpivirine significantly decreased HIV-1 production, by ≥1 log.


Subject(s)
Anti-HIV Agents/pharmacology , Benzoxazines/pharmacology , Bryostatins/pharmacology , HIV-1/drug effects , Rilpivirine/pharmacology , Virus Replication/drug effects , Adjuvants, Immunologic/pharmacology , Alkynes , Antibodies, Monoclonal/pharmacology , Antibodies, Viral/pharmacology , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/genetics , CD28 Antigens/immunology , CD3 Complex/genetics , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/virology , Cyclopropanes , Gene Expression , HIV-1/genetics , HIV-1/growth & development , Humans , Interleukin-2/pharmacology , Primary Cell Culture , Virus Latency/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL