Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters

Publication year range
1.
J Neuroinflammation ; 19(1): 319, 2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36587195

ABSTRACT

BACKGROUND: Regulatory CD4+CD25+FoxP3+ T cells (Treg) are a subgroup of T lymphocytes involved in maintaining immune balance. Disturbance of Treg number and impaired suppressive function of Treg correlate with Parkinson's disease severity. Superagonistic anti-CD28 monoclonal antibodies (CD28SA) activate Treg and cause their expansion to create an anti-inflammatory environment. METHODS: Using the AAV1/2-A53T-α-synuclein Parkinson's disease mouse model that overexpresses the pathogenic human A53T-α-synuclein (hαSyn) variant in dopaminergic neurons of the substantia nigra, we assessed the neuroprotective and disease-modifying efficacy of a single intraperitoneal dose of CD28SA given at an early disease stage. RESULTS: CD28SA led to Treg expansion 3 days after delivery in hαSyn Parkinson's disease mice. At this timepoint, an early pro-inflammation was observed in vehicle-treated hαSyn Parkinson's disease mice with elevated percentages of CD8+CD69+ T cells in brain and increased levels of interleukin-2 (IL-2) in the cervical lymph nodes and spleen. These immune responses were suppressed in CD28SA-treated hαSyn Parkinson's disease mice. Early treatment with CD28SA attenuated dopaminergic neurodegeneration in the SN of hαSyn Parkinson's disease mice accompanied with reduced brain numbers of activated CD4+, CD8+ T cells and CD11b+ microglia observed at the late disease-stage 10 weeks after AAV injection. In contrast, a later treatment 4 weeks after AAV delivery failed to reduce dopaminergic neurodegeneration. CONCLUSIONS: Our data indicate that immune modulation by Treg expansion at a timepoint of overt inflammation is effective for treatment of hαSyn Parkinson's disease mice and suggest that the concept of early immune therapy could pose a disease-modifying option for Parkinson's disease patients.


Subject(s)
Parkinson Disease , Mice , Humans , Animals , Parkinson Disease/pathology , T-Lymphocytes, Regulatory , alpha-Synuclein/metabolism , CD8-Positive T-Lymphocytes/metabolism , CD28 Antigens , Antibodies/pharmacology , Substantia Nigra/metabolism , Dopaminergic Neurons/metabolism , Dopamine , Disease Models, Animal , Mice, Inbred C57BL
2.
Haematologica ; 106(2): 363-374, 2021 02 01.
Article in English | MEDLINE | ID: mdl-31879324

ABSTRACT

Recurrence of cytomegalovirus reactivation remains a major cause of morbidity and mortality following allogeneic hematopoietic stem cell transplantation. Monitoring cytomegalovirus-specific cellular immunity using a standardized assay might improve the risk stratification of patients. A prospective multicenter study was conducted in 175 intermediate- and high-risk allogeneic hematopoietic stem cell transplant recipients under preemptive antiviral therapy. Cytomegalovirus-specific cellular immunity was measured using a standardized IFN-γ ELISpot assay (T-Track® CMV). Primary aim was to evaluate the suitability of measuring cytomegalovirus-specific immunity after end of treatment for a first cytomegalovirus reactivation to predict recurrent reactivation. 40/101 (39.6%) patients with a first cytomegalovirus reactivation experienced recurrent reactivations, mainly in the high-risk group (cytomegalovirus-seronegative donor/cytomegalovirus-seropositive recipient). The positive predictive value of T-Track® CMV (patients with a negative test after the first reactivation experienced at least one recurrent reactivation) was 84.2% in high-risk patients. Kaplan-Meier analysis revealed a higher probability of recurrent cytomegalovirus reactivation in high-risk patients with a negative test after the first reactivation (hazard ratio 2.73; p=0.007). Interestingly, a post-hoc analysis considering T-Track® CMV measurements at day 100 post-transplantation, a time point highly relevant for outpatient care, showed a positive predictive value of 90.0% in high-risk patients. Our results indicate that standardized cytomegalovirus-specific cellular immunity monitoring may allow improved risk stratification and management of recurrent cytomegalovirus reactivation after hematopoietic stem cell transplantation. This study was registered at www.clinicaltrials.gov as #NCT02156479.


Subject(s)
Cytomegalovirus Infections , Hematopoietic Stem Cell Transplantation , Cytomegalovirus , Cytomegalovirus Infections/diagnosis , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Prospective Studies , Risk Assessment , Virus Activation
3.
Immunity ; 37(1): 134-46, 2012 Jul 27.
Article in English | MEDLINE | ID: mdl-22683122

ABSTRACT

Myelin-specific CD8(+) T cells are thought to contribute to the pathogenesis of multiple sclerosis. Here we modeled this contribution in mice with CD8(+) T cells recognizing ovalbumin (OVA) expressed in oligodendrocytes (ODCs). Surprisingly, even very high numbers of activated OVA-reactive CD8(+) T cells failed to induce disease and were cleared from the immune system after antigen encounter in the central nervous system (CNS). Peripheral infection with OVA-expressing Listeria (Lm-OVA) enabled CD8(+) T cells to enter the CNS, leading to the deletion of OVA-specific clones after OVA recognition on ODCs. In contrast, intracerebral infection allowed OVA-reactive CD8(+) T cells to cause demyelinating disease. Thus, in response to infection, CD8(+) T cells also patrol the CNS. If the CNS itself is infected, they destroy ODCs upon cognate antigen recognition in pursuit of pathogen eradication. In the sterile brain, however, antigen recognition on ODCs results in their deletion, thereby maintaining tolerance.


Subject(s)
Autoimmunity , Brain/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance , Oligodendroglia/immunology , Animals , Antibodies, Monoclonal/immunology , Apoptosis/immunology , Autoantigens/immunology , Brain/microbiology , Hematopoiesis/immunology , Integrin alpha4beta1/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oligodendroglia/metabolism , Oligodendroglia/transplantation , Ovalbumin/immunology , Stromal Cells/immunology , Stromal Cells/metabolism , Thymus Gland/cytology , Thymus Gland/immunology , fas Receptor/immunology
4.
Eur J Immunol ; 49(3): 413-427, 2019 03.
Article in English | MEDLINE | ID: mdl-30666625

ABSTRACT

Increasing evidence suggests a role of CD8 T cells in autoimmune demyelinating CNS disease, which, however, is still controversially discussed. Mice, which express ovalbumin (OVA) as cytosolic self-antigen in oligodendrocytes (ODC-OVA mice), respond to CNS infection induced by OVA-expressing attenuated Listeria with CD8 T cell-mediated inflammatory demyelination. This model is suitable to decipher the contribution of CD8 T cells and the pathogen in autoimmune CNS disease. Here, we show that both antigen and pathogen are required in the CNS for disease induction, though not in a physically linked fashion. Intracerebral challenge with combined toll like receptor (TLR) TLR2 and TLR9 as well as TLR7 and TLR9 agonists substituted for the bacterial stimulus, but not with individual TLR agonists (TLR2, TLR3,TLR5,TLR7, TLR9). Furthermore, MyD88 inactivation rendered ODC-OVA mice resistant to disease induction. Collectively, CD8 T cell-mediated destruction of oligodendrocytes is activated if (i) an antigen shared with an infectious agent is provided in the CNS microenvironment and (ii) innate immune signals inform the CNS microenvironment that pathogen removal warrants an immune attack by CD8 T cells, even at the expense of locally restricted demyelination.


Subject(s)
Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Oligodendroglia/immunology , Ovalbumin/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , Animals , Antigens/genetics , Antigens/metabolism , Autoimmune Diseases/immunology , Autoimmune Diseases/microbiology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Central Nervous System/immunology , Central Nervous System/microbiology , Central Nervous System/pathology , Demyelinating Diseases/immunology , Demyelinating Diseases/microbiology , Listeria monocytogenes/immunology , Listeria monocytogenes/physiology , Listeriosis/immunology , Listeriosis/microbiology , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Oligodendroglia/metabolism , Oligodendroglia/pathology , Ovalbumin/genetics , Ovalbumin/metabolism , Toll-Like Receptors/metabolism
5.
Eur J Immunol ; 48(2): 366-379, 2018 02.
Article in English | MEDLINE | ID: mdl-29023682

ABSTRACT

The identification of regulatory T cells (Treg cells) in human peripheral blood is an important tool in diagnosis, research, and therapeutic intervention. As compared to lymphoid tissues, the frequencies of circulating Treg cells identified as CD4+ CD25+ Foxp3+ are, however, low. We here show that many of these cells remain undetected due to transient down regulation of Foxp3, which rapidly decays in the absence of cytokine-mediated STAT5 signals. Short-term incubation of PBMCs or isolated CD4+ T cells, but not of lymph node cells, with IL-2, -7, or -15 more than doubles the frequency of Foxp3+ CD25+ among CD4+ T cells detectable by flow cytometry. This increase is not due to cell division but to upregulation of both proteins. At the same time, the uncovered Treg cells up-regulate CD25 and down-regulate CD127, making them accessible to viable cell sorting. "Latent" Treg cells have a demethylated FOXP3 TSDR sequence, are enriched in naïve, non-cycling cells, and are functional. The confirmation of our findings in RA and SLE patients shows the feasibility of uncovering latent Treg cells for immune monitoring in clinical settings. Finally, our results suggest that unmasking of latent Treg cells contributes to the increase in circulating CD4+ CD25+ Foxp3+ cells reported in IL-2 treated patients.


Subject(s)
Arthritis, Rheumatoid/immunology , CD4-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Lupus Erythematosus, Systemic/immunology , T-Lymphocytes, Regulatory/immunology , Arthritis, Rheumatoid/therapy , Blood Circulation , Cells, Cultured , Down-Regulation , Flow Cytometry , Forkhead Transcription Factors/metabolism , Humans , Interleukin-15/metabolism , Interleukin-2/metabolism , Interleukin-2/therapeutic use , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-7/metabolism , Interleukin-7 Receptor alpha Subunit/metabolism , Lupus Erythematosus, Systemic/therapy , Lymphocyte Activation , Male , Methylation , Monitoring, Physiologic
6.
Transpl Int ; 31(4): 436-450, 2018 04.
Article in English | MEDLINE | ID: mdl-29284181

ABSTRACT

Impaired cytomegalovirus (CMV)-specific cell-mediated immunity (CMV-CMI) is a major cause of CMV reactivation and associated complications in solid-organ transplantation. Reliably assessing CMV-CMI is desirable to individually adjust antiviral and immunosuppressive therapy. This study aimed to evaluate the suitability of T-Track® CMV, a novel IFN-γ ELISpot assay based on the stimulation of peripheral blood mononuclear cells with pp65 and IE-I CMV proteins, to monitor CMV-CMI following kidney transplantation. A prospective longitudinal multicenter study was conducted in 86 intermediate-risk renal transplant recipients. CMV-CMI, CMV viral load, and clinical complications were monitored over 6 months post-transplantation. Ninety-five percent and 88-92% ELISpot assays were positive pre- and post-transplantation, respectively. CMV-specific response was reduced following immunosuppressive treatment and increased in patients with graft rejection, indicating the ability of the ELISpot assay to monitor patients' immunosuppressive state. Interestingly, median pp65-specific response was ninefold higher in patients with self-clearing viral load compared to antivirally treated patients prior to first viral load detection (P < 0.001), suggesting that reactivity to pp65 represents a potential immunocompetence marker. Altogether, T-Track® CMV is a highly sensitive IFN-γ ELISpot assay, suitable for the immunomonitoring of CMV-seropositive renal transplant recipients, and with a potential use for the risk assessment of CMV-related clinical complications (ClinicalTrials.gov Identifier: NCT02083042).


Subject(s)
Cytomegalovirus Infections/diagnosis , Enzyme-Linked Immunosorbent Assay/methods , Immunity, Cellular , Phosphoproteins/immunology , Postoperative Complications/diagnosis , Viral Matrix Proteins/immunology , Adult , Aged , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Female , Graft Rejection/immunology , Graft Rejection/virology , Humans , Immunosuppressive Agents , Kidney Transplantation , Longitudinal Studies , Male , Middle Aged , Opportunistic Infections , Postoperative Complications/immunology , Prospective Studies , Young Adult
7.
Eur J Immunol ; 46(7): 1644-55, 2016 07.
Article in English | MEDLINE | ID: mdl-27122236

ABSTRACT

The role of CD28-mediated costimulation in secondary CD8(+) T-cell responses remains controversial. Here, we have used two tools - blocking mouse anti-mouse CD28-specific antibodies and inducible CD28-deleting mice - to obtain definitive answers in mice infected with ovalbumin-secreting Listeria monocytogenes. We report that both blockade and global deletion of CD28 reveal its requirement for full clonal expansion and effector functions such as degranulation and IFN-γ production during the secondary immune response. In contrast, cell-intrinsic deletion of CD28 in transferred TCR-transgenic CD8(+) T cells before primary infection leads to impaired clonal expansion but an increase in cells able to express effector functions in both primary and secondary responses. We suggest that the proliferation-impaired CD8(+) T cells respond to CD28-dependent help from their environment by enhanced functional differentiation. Finally, we report that cell-intrinsic deletion of CD28 after the peak of the primary response does not affect the establishment, maintenance, or recall of long-term memory. Thus, if given sufficient time, the progeny of primed CD8(+) T cells adapt to the absence of this costimulator.


Subject(s)
Antigens/immunology , CD28 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunologic Memory , Lymphocyte Activation/immunology , Animals , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/metabolism , CD28 Antigens/antagonists & inhibitors , CD28 Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/immunology , Lymphocyte Activation/genetics , Mice , Mice, Knockout , Mice, Transgenic
8.
Eur J Immunol ; 46(5): 1193-202, 2016 05.
Article in English | MEDLINE | ID: mdl-26711629

ABSTRACT

Although regulatory T (Treg) cells are necessary to prevent autoimmune diseases, including arthritis, whether Treg cells can ameliorate established inflammatory disease is controversial. Using the glucose-6-phosphate isomerase (G6PI)-induced arthritis model in mice, we aimed to determine the therapeutic efficacy of increasing Treg cell number and function during chronic destructive arthritis. Chronic destructive arthritis was induced by transient depletion of Treg cells prior to immunization with G6PI. At different time points after disease induction, mice were treated with a CD28 superagonistic antibody (CD28SA). CD28SA treatment during the induction phase of arthritis ameliorated the acute signs of arthritis and completely prevented the development of chronic destructive arthritis. CD28SA treatment of mice with fully developed arthritis induced a significant reduction in clinical and histological signs of arthritis. When given during the chronic destructive phase of arthritis, 56 days after disease induction, CD28SA treatment resulted in a modest reduction of clinical signs of arthritis and a reduction in histopathological signs of joint inflammation. Our data show that increasing the number and activation of Treg cells by a CD28SA is therapeutically effective in experimental arthritis.


Subject(s)
Antibodies, Monoclonal/immunology , Arthritis, Experimental/prevention & control , Autoimmune Diseases/prevention & control , CD28 Antigens/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Autoimmune Diseases/immunology , CD28 Antigens/agonists , Chronic Disease/prevention & control , Disease Models, Animal , Glucose-6-Phosphate Isomerase/administration & dosage , Inflammation/prevention & control , Inflammation/therapy , Joints/immunology , Joints/physiopathology , Mice , Th1 Cells/immunology
9.
Cancer Immunol Immunother ; 66(3): 319-332, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27896368

ABSTRACT

Due to its immunogenicity and overexpression concomitant with leukemia progression, Wilms tumor protein 1 (WT1) is of particular interest for immunotherapy of AML relapse after allogeneic hematopoietic stem cell transplantation (allo-HSCT). So far, WT1-specific T-cell responses have mainly been induced by vaccination with peptides presented by certain HLA alleles. However, this approach is still not widely applicable in clinical practice due to common limitations of HLA restriction. Dendritic cell (DC) vaccines electroporated with mRNA encoding full-length protein have also been tested for generating WT1-derived peptides for presentation to T-cells. Alternatively, an efficient and broad WT1 peptide presentation could be elicited by triggering receptor-mediated protein endocytosis of DCs. Therefore, we developed antibody fusion proteins consisting of an antibody specific for the DEC205 endocytic receptor on human DCs and various fragments of WT1 as DC-targeting recombinant WT1 vaccines (anti-hDEC205-WT1). Of all anti-hDEC205-WT1 fusion proteins designed for overcoming insufficient expression, anti-hDEC205-WT110-35, anti-hDEC205-WT191-138, anti-hDEC205-WT1223-273, and anti-hDEC205-WT1324-371 were identified in good yields. The anti-hDEC205-WT191-138 was capable of directly inducing ex vivo T-cell responses by co-incubation of the fusion protein-loaded monocyte-derived mature DCs and autologous T-cells of either healthy or HSCT individuals. Furthermore, the DC-targeted WT191-138-induced specific T-cells showed a strong cytotoxic activity by lysing WT1-overexpressing THP-1 leukemia cells in vitro while sparing WT1-negative hematopoietic cells. In conclusion, our approach identifies four WT1 peptide-antibody fusion proteins with sufficient production and introduces an alternative vaccine that could be easily translated into clinical practice to improve WT1-directed antileukemia immune responses after allo-HSCT.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , T-Lymphocytes, Cytotoxic/immunology , WT1 Proteins/immunology , Amino Acid Sequence , Animals , Antigen Presentation , CHO Cells , Cancer Vaccines/genetics , Cricetulus , Electroporation , HEK293 Cells , Humans , Lymphocyte Activation , RNA, Messenger/genetics , RNA, Messenger/immunology , WT1 Proteins/genetics
10.
Blood ; 126(2): 185-94, 2015 Jul 09.
Article in English | MEDLINE | ID: mdl-26024876

ABSTRACT

Peripheral blood mononuclear cells (PBMCs) are the only source of human lymphoid cells routinely available for immunomonitoring of T-cell responses to microbial and tumor-associated antigens. However, previous work in mice and humans had indicated that CD4 T cells transiently lose antigen sensitivity when cellular contacts are lost (eg, by entering the circulation). Using the simple and robust protocol for resetting T cells to original reactivity (RESTORE; ie, preculturing PBMCs for 2 days at a high cell density before initiation of antigenic stimulation), we show that CD8 T-cell responses to viral and tumor-associated antigens are greatly underestimated in blood, and sometimes even remain undetected, if conventional, unprocessed PBMC cultures are used. The latter finding is particularly striking with regard to the appearance of Wilms tumor 1 protein-specific CD8 T-cell responses in leukemia patients after allogeneic bone marrow transplantation. The dramatic increase in antigen sensitivity of "restored" CD8 T cells is associated with phosphorylation of proximal T-cell receptor signaling components, and with the upregulation of genes involved in aerobic glycolysis, thereby increasing T-cell functionality. The RESTORE protocol permits a more meaningful monitoring of CD8 memory T-cell responses to viral infections and tumors and vaccination success. Furthermore, when generating T-cell lines for adoptive T-cell therapy, it avoids the loss of those clones, which strictly depend on the primed status conferred by cellular interactions in the tissue context for their initial reactivation by antigen. The data reported in this article have been deposited in the Gene Expression Omnibus database (accession number GSE63430).


Subject(s)
Antigens, Neoplasm/immunology , Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Culture Techniques/methods , Leukocytes, Mononuclear/cytology , T-Cell Antigen Receptor Specificity , Cell Count , Cells, Cultured , Child , Humans , Leukocytes, Mononuclear/physiology
11.
Eur J Immunol ; 45(7): 1997-2007, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25907100

ABSTRACT

Upon transplantation of T cells from a CD28 superagonist (CD28-SA) treated donor into an irradiated allogeneic host, the CD28-SA-induced activation and expansion of Treg cells inhibits acute graft versus host disease (aGvHD), while not abrogating the desired graft versus tumor effect. Human peripheral blood CD4(+) T cells, however, harbor only very few Treg cells. Therefore, we studied whether polyclonal in vitro prestimulation of conventional, that is Treg -cell-depleted, CD4(+) T cells of C57BL/6 mice with CD28-SA-coated paramagnetic beads is sufficient to protect recipient BALB/c mice from aGvHD. CD28-SA prestimulation of conventional CD4(+) T cells efficiently protected BALB/c recipient mice from aGvHD and CD28-SA-stimulated CD4(+) and CD8(+) T cells were capable of mediating long-term protection from the BCL1 lymphoma. The recently completed successful phase I testing of the human CD28-SA TGN1412/TAB08 should greatly facilitate further development of this straightforward method into a novel immunotherapy for patients.


Subject(s)
Bone Marrow Transplantation/methods , CD28 Antigens/agonists , CD4-Positive T-Lymphocytes/immunology , Graft vs Host Disease/prevention & control , Lymphocyte Activation/immunology , Allografts , Animals , Bone Marrow Transplantation/adverse effects , CD8-Positive T-Lymphocytes/immunology , Cell Culture Techniques/methods , Cell Separation , Disease Models, Animal , Flow Cytometry , Graft vs Host Disease/immunology , In Vitro Techniques , Lymphoma/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
12.
Eur J Immunol ; 45(2): 480-91, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25382658

ABSTRACT

Anergic T cells can survive for long time periods passively in a hyporesponsive state without obvious active functions. Thus, the immunological reason for their maintenance is unclear. Here, we induced peptide-specific anergy in T cells from mice by coculturing these cells with immature murine dendritic cells (DCs). We found that these anergic, nonsuppressive IL-10(-) Foxp3(-) CTLA-4(+) CD25(low) Egr2(+) T cells could be converted into suppressive IL-10(+) Foxp3(-) CTLA-4(+) CD25(high) Egr2(+) cells resembling type-1 Treg cells (Tr1) when stimulated a second time by immature DCs in vitro. Addition of TGF-ß during anergy induction favored Foxp3(+) Treg-cell induction, while TGF-ß had little effect when added to the second stimulation. Expression of both CD28 and CTLA-4 molecules on anergic T cells was required to allow their conversion into Tr1-like cells. Suppressor activity was enabled via CD28-mediated CD25 upregulation, acting as an IL-2 sink, together with a CTLA-4-mediated inhibition of NFATc1/α activation to shut down IL-2-mediated proliferation. Together, these data provide evidence and mechanistical insights into how persistent anergic T cells may serve as a resting memory pool for Tr1-like cells.


Subject(s)
CD28 Antigens/genetics , CTLA-4 Antigen/genetics , Clonal Anergy , Dendritic Cells/immunology , Forkhead Transcription Factors/genetics , Interleukin-10/genetics , T-Lymphocytes, Regulatory/immunology , Animals , CD28 Antigens/immunology , CTLA-4 Antigen/immunology , Cell Communication , Cell Differentiation , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/immunology , Forkhead Transcription Factors/immunology , Gene Expression Regulation/immunology , Interleukin-10/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Signal Transduction , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/pharmacology
13.
PLoS Pathog ; 10(2): e1003906, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24516382

ABSTRACT

IL-13 driven Th2 immunity is indispensable for host protection against infection with the gastrointestinal nematode Nippostronglus brasiliensis. Disruption of CD28 mediated costimulation impairs development of adequate Th2 immunity, showing an importance for CD28 during the initiation of an immune response against this pathogen. In this study, we used global CD28⁻/⁻ mice and a recently established mouse model that allows for inducible deletion of the cd28 gene by oral administration of tamoxifen (CD28(-/lox)Cre⁺/⁻+TM) to resolve the controversy surrounding the requirement of CD28 costimulation for recall of protective memory responses against pathogenic infections. Following primary infection with N. brasiliensis, CD28⁻/⁻ mice had delayed expulsion of adult worms in the small intestine compared to wild-type C57BL/6 mice that cleared the infection by day 9 post-infection. Delayed expulsion was associated with reduced production of IL-13 and reduced serum levels of antigen specific IgG1 and total IgE. Interestingly, abrogation of CD28 costimulation in CD28(-/lox)Cre⁺/⁻ mice by oral administration of tamoxifen prior to secondary infection with N. brasiliensis resulted in impaired worm expulsion, similarly to infected CD28⁻/⁻ mice. This was associated with reduced production of the Th2 cytokines IL-13 and IL-4, diminished serum titres of antigen specific IgG1 and total IgE and a reduced CXCR5⁺ T(FH) cell population. Furthermore, total number of CD4⁺ T cells and B220⁺ B cells secreting Th1 and Th2 cytokines were significantly reduced in CD28⁻/⁻ mice and tamoxifen treated CD28(-/lox)Cre⁺/⁻ mice compared to C57BL/6 mice. Importantly, interfering with CD28 costimulatory signalling before re-infection impaired the recruitment and/or expansion of central and effector memory CD4⁺ T cells and follicular B cells to the draining lymph node of tamoxifen treated CD28(-/lox)Cre⁺/⁻ mice. Therefore, it can be concluded that CD28 costimulation is essential for conferring host protection during secondary N. brasiliensis infection.


Subject(s)
CD28 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Nippostrongylus , Strongylida Infections/immunology , Animals , CD28 Antigens/metabolism , Flow Cytometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Strongylida Infections/metabolism
14.
Exp Dermatol ; 25(4): 293-8, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26661498

ABSTRACT

Pemphigus vulgaris (PV) is a potentially life-threatening autoimmune disease of the skin and mucous membranes. Its pathogenesis is based on IgG autoantibodies that target the desmosomal cadherins, desmoglein 3 (Dsg3) and desmoglein 1 (Dsg1) and induce intra-epidermal loss of adhesion. Although the PV pathogenesis is well-understood, therapeutic options are still limited to immunosuppressive drugs, particularly corticosteroids, which are associated with significant side effects. Dsg3-reactive T regulatory cells (Treg) have been previously identified in PV and healthy carriers of PV-associated HLA class II alleles. Ex vivo, Dsg3-specific Treg cells down-regulated the activation of pathogenic Dsg3-specific T-helper (Th) 2 cells. In this study, in a HLA-DRB1*04:02 transgenic mouse model of PV, peripheral Treg cells were modulated by the use of Treg-depleting or expanding monoclonal antibodies, respectively. Our findings show that, in vivo, although not statistically significant, Treg cells exert a clear down-regulatory effect on the Dsg3-driven T-cell response and, accordingly, the formation of Dsg3-specific IgG antibodies. These observations confirm the powerful immune regulatory functions of Treg cells and identify Treg cells as potential therapeutic modulators in PV.


Subject(s)
Autoantibodies/chemistry , CD28 Antigens/immunology , Desmoglein 3/genetics , HLA-DRB1 Chains/immunology , Pemphigus/immunology , T-Lymphocytes, Regulatory/metabolism , Alleles , Animals , Antibodies, Monoclonal/chemistry , CD28 Antigens/genetics , Cell Proliferation , Desmoglein 1/genetics , Down-Regulation , HLA-DRB1 Chains/genetics , Humans , Immunoglobulin G/chemistry , Inflammation , Mice , Mice, Transgenic , Pemphigus/genetics , Recombinant Proteins/chemistry , T-Lymphocytes, Regulatory/cytology , Th2 Cells/cytology , Th2 Cells/metabolism
15.
Clin Exp Rheumatol ; 34(4 Suppl 98): 45-8, 2016.
Article in English | MEDLINE | ID: mdl-27586803

ABSTRACT

CD28 superagonists (CD28SA) are CD28-specific monoclonal antibodies which are able to activate T-cells without overt TCR engagement. In rodents, CD28SA efficiently activate regulatory T-cells and are therapeutically effective in multiple models of autoimmunity, inflammation and transplantation. However, a phase I study of the human CD28SA TGN1412 in 2006 resulted in a life-threatening cytokine storm. This brief review summarises preclinical work before and since the failed phase I trial with an emphasis on understanding the reasons why there had been no warning of toxicity, and how a novel assay paved the way for a new phase I, phase Ib (both completed), and an ongoing phase II study.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/drug therapy , CD28 Antigens/antagonists & inhibitors , Animals , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Humans , Lymphocyte Activation/drug effects , Molecular Targeted Therapy , Risk Assessment , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
16.
J Immunol ; 192(5): 2091-8, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24470499

ABSTRACT

Following inconspicuous preclinical testing, the superagonistic anti-CD28 mAb TGN1412 was applied to six study participants who all developed a devastating cytokine storm. We verified that TGN1412 treatment of fresh PBMCs induced only moderate responses, whereas restoration of tissue-like conditions by high-density preculture (HDC) allowed vigorous cytokine production. TGN1412 treatment of T cells isolated from HDC-PBMCs induced moderate cytokine responses, which upon additional anti-IgG crosslinking were significantly boosted. Moreover, coincubation of TGN1412-treated T cells with B cells expressing the intermediate affinity Fcγ receptor IIB (CD32B), or coincubation with CD32B(+) transfectants, resulted in robust T cell activation. This was surprising because TGN1412 was expressed as an Ig of the subclass 4 (IgG4), which was shown before to exhibit only minor affinity to FcγRs. Transcriptome analysis of TGN1412-treated T cells revealed that similar gene signatures were induced irrespective of whether T cells derived from fresh or HDC-PBMCs were studied. Collectively, these data indicate that HDC-PBMCs and HDC-PBMC-derived T cells mount rapid TGN1412 responses, which are massively boosted by FcγR crosslinking, in particular by CD32-expressing B cells. These results qualify HDC-PBMCs as a valuable in vitro test system for the analysis of complex mAb functions.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Cytokines/immunology , Gene Expression Regulation/drug effects , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/pharmacology , Receptors, IgG/immunology , Antibodies, Monoclonal, Humanized/immunology , Female , Gene Expression Profiling , Gene Expression Regulation/immunology , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin G/immunology , Male , Transcriptome/drug effects , Transcriptome/immunology
17.
Stroke ; 46(1): 212-20, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25378432

ABSTRACT

BACKGROUND AND PURPOSE: Neuroinflammation plays an important role in ischemic brain injury. Regulatory T cells (Treg) are important endogenous immune modulators. We tested the hypothesis that Treg amplification with a CD28 superagonistic monoclonal antibody (CD28SA) reduces brain damage in murine cerebral ischemia. METHODS: Cerebral ischemia was induced by coagulation of the distal middle cerebral artery or by 60 minutes filament occlusion of the proximal middle cerebral artery in C57BL6 mice. 150 µg CD28SA was injected intraperitoneally 3 or 6 hours after ischemia onset. Outcome was determined by infarct volumetry and behavioral testing. Brain-infiltrating leukocyte subpopulations were analyzed by flow cytometry and immunohistochemistry 3 and 7 days after middle cerebral artery occlusion. RESULTS: CD28SA reduced infarct size in both models and attenuated functional deficit 7 days after stroke induction. Mice treated with CD28SA increased numbers of Treg in spleen and brain. Tregs were functionally active and migrated into the brain where they accumulated and proliferated in the peri-infarct area. More than 60% of brain infiltrating Treg produced interleukin-10 in CD28SA compared with 30% in control. CONCLUSIONS: In vivo expansion and amplification of Treg by CD28SA attenuates the inflammatory response and improves outcome after experimental stroke.


Subject(s)
Brain/immunology , CD28 Antigens/agonists , Infarction, Middle Cerebral Artery/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Monoclonal/pharmacology , Brain/drug effects , Brain Ischemia/complications , Brain Ischemia/immunology , Disease Models, Animal , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/pathology , Mice , Mice, Inbred C57BL , Stroke/etiology , Stroke/immunology , T-Lymphocytes, Regulatory/drug effects
18.
Eur J Immunol ; 44(4): 1225-36, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24374661

ABSTRACT

CD28 superagonists (CD28SAs) are potent T-cell-activating monoclonal antibodies (mAbs). In contrast to their benign behavior and marked therapeutic efficacy as activators of regulatory T (Treg) cells in preclinical rodent models, a phase I trial of the human CD28SA TGN1412 (now called TAB08) in 2006 resulted in a life-threatening cytokine release syndrome (CRS). We studied TAB08-mediated Treg-cell activation in a recently developed in vitro system of human PBMCs, which also reproduces the CRS experienced by the healthy volunteers. We show that just as in rodents, CD28SAs are potent activators and expanders of Treg cells from healthy donors and rheumatoid arthritis patients, even under effective blockade of pro-inflammatory cytokine release by a corticosteroid. Moreover, CD28SA titration identifies a dose range where pro-inflammatory cytokine secretion from conventional T cells is absent while appreciable Treg-cell activation is maintained. Finally, we report that low-dose application of TAB08 to healthy volunteers results in dose-dependent systemic release of the Treg-cell signature cytokine IL-10 in the absence of the pro-inflammatory factors associated with the CRS of the 2006 TGN1412 study. These results demonstrate the potential of appropriately dosed CD28SA and corticosteroid comedication to mobilize human Treg cells for the treatment of autoimmune and inflammatory conditions.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , CD28 Antigens/immunology , Lymphocyte Activation/drug effects , T-Lymphocytes, Regulatory/immunology , Adrenal Cortex Hormones/pharmacology , Antibodies, Monoclonal, Humanized/administration & dosage , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , CD28 Antigens/agonists , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dose-Response Relationship, Drug , Flow Cytometry , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Interleukin-10/immunology , Interleukin-10/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/metabolism
19.
Histochem Cell Biol ; 144(3): 195-208, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25976155

ABSTRACT

Immune responses are generally accompanied by antigen presentation and proliferation and differentiation of antigen-specific lymphocytes (immunoproliferation), but analysis of these events in situ on tissue sections is very difficult. We have developed a new method of simultaneous multicolor immunofluorescence staining for immunohistology and flow cytometry using a thymidine analogue, 5-ethynyl-2'-deoxyuridine (EdU). Because of the small size of azide dye using click chemistry and elimination of DNA denaturation steps, EdU staining allowed for immunofluorescence staining of at least four colors including two different markers on a single-cell surface, which is impossible with the standard 5-bromo-2'-deoxyuridine method. By using two rat models, successfully detected parameters were the cluster of differentiation antigens including phenotypic and functional markers of various immune cells, histocompatibility complex antigens, and even some nuclear transcription factors. Proliferating cells could be further sorted and used for RT-PCR analysis. This method thus enables functional in situ time-kinetic analysis of immunoproliferative responses in a distinct domain of the lymphoid organs, which are quantitatively confirmed by flow cytometry.


Subject(s)
Antimetabolites/pharmacology , Cell Proliferation/drug effects , Deoxyuridine/analogs & derivatives , Immunity, Cellular/physiology , Animals , Bromodeoxyuridine/pharmacology , CD28 Antigens/biosynthesis , Dendritic Cells/drug effects , Dendritic Cells/immunology , Deoxyuridine/pharmacology , Flow Cytometry , Immunohistochemistry , Lymphocytes/immunology , Major Histocompatibility Complex/immunology , Male , Rats , Rats, Inbred Lew , S Phase/drug effects , Spleen/cytology , Spleen/immunology , Transcription Factors/biosynthesis
20.
Eur J Immunol ; 43(1): 188-93, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23065717

ABSTRACT

While the requirement for CD28 and its ligands for the generation and function of "natural" (n)Treg cells is well established, it has not been possible yet to investigate cell-intrinsic effects after interrupted CD28 expression. Here, we demonstrate a selective loss of Treg cells after disruption of the CD28 gene. The decline in Treg-cell number was accompanied by reduced homeostatic proliferation, probably due to lack of costimulation during self-antigen recognition, and by impaired Treg-cell function including downregulation of CTLA-4. The decline in Treg-cell number was unaffected by thymectomy or by the presence of CD28 expressing T cells within the same animal, indicating that impairment of peripheral homeostasis and function of nTreg cells by CD28 deletion is cell-intrinsic. In contrast, downregulation of CD25, the α chain of the IL-2R, did not occur in the presence of WT T cells, indicating that its expression does not depend on CD28 signals in cis.


Subject(s)
CD28 Antigens/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , CD28 Antigens/genetics , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Cell Proliferation , Cells, Cultured , Down-Regulation , Homeostasis , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphocyte Count , Mice , Mice, Inbred C57BL , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL