Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 375
Filtrer
1.
Front Immunol ; 15: 1357716, 2024.
Article de Anglais | MEDLINE | ID: mdl-38384461

RÉSUMÉ

Objectives: Despite their efficacy, some immunotherapies have been shown to induce immune-related adverse events, including the potentially life-threatening cytokine release syndrome (CRS), calling for reliable and translational preclinical models to predict potential safety issues and investigate their rescue. Here, we tested the reliability of humanized BRGSF mice for the assessment of therapeutics-induced CRS features in preclinical settings. Methods: BRGSF mice reconstituted with human umbilical cord blood CD34+ cells (BRGSF-CBC) were injected with anti-CD3 antibody (OKT3), anti-CD3/CD19 bispecific T-cell engager Blinatumomab, or VISTA-targeting antibody. Human myeloid and dendritic cells' contribution was investigated in hFlt3L-boosted BRGSF-CBC mice. OKT3 treatment was also tested in human PBMC-reconstituted BRGSF mice (BRGSF-PBMC). Cytokine release, immune cell distribution, and clinical signs were followed. Results: OKT3 injection in BRGSF-CBC mice induced hallmark features of CRS, specifically inflammatory cytokines release, modifications of immune cell distribution and activation, body weight loss, and temperature drop. hFlt3L-boosted BRGSF-CBC mice displayed enhanced CRS features, revealing a significant role of myeloid and dendritic cells in this process. Clinical CRS-managing treatment Infliximab efficiently attenuated OKT3-induced toxicity. Comparison of OKT3 treatment's effect on BRGSF-CBC and BRGSF-PBMC mice showed broadened CRS features in BRGSF-CBC mice. CRS-associated features were also observed in hFlt3L-boosted BRGSF-CBC mice upon treatment with other T-cell or myeloid-targeting compounds. Conclusions: These data show that BRGSF-CBC mice represent a relevant model for the preclinical assessment of CRS and CRS-managing therapies. They also confirm a significant role of myeloid and dendritic cells in CRS development and exhibit the versatility of this model for therapeutics-induced safety assessment.


Sujet(s)
Syndrome de libération de cytokines , Muromonab-CD3 , Humains , Souris , Animaux , Muromonab-CD3/pharmacologie , Agranulocytes , Reproductibilité des résultats , Cytokines , Cellules dendritiques
2.
Front Immunol ; 13: 938106, 2022.
Article de Anglais | MEDLINE | ID: mdl-36189306

RÉSUMÉ

CD3+/CD56+ Natural killer (NK) cell-like T-cells (NKT-like cells) represent <5% of blood lymphocytes, display a cytotoxic phenotype, and can kill various cancers. NKT-like cells can be expanded ex vivo into cytokine-induced killer (CIK) cells, however this therapeutic cell product has had mixed results against hematological malignancies in clinical trials. The aim of this study was to determine if NKT-like cells mobilized during acute cycling exercise could be used to generate more potent anti-tumor CIK cells from healthy donors. An acute exercise bout increased NKT-like cell numbers in blood 2-fold. Single cell RNA sequencing revealed that exercise mobilized NKT-like cells have an upregulation of genes and transcriptomic programs associated with enhanced anti-tumor activity, including cytotoxicity, cytokine responsiveness, and migration. Exercise, however, did not augment the ex vivo expansion of CIK cells or alter their surface phenotypes after 21-days of culture. CIK cells expanded at rest, during exercise (at 60% and 80% VO2max) or after (1h post) were equally capable of killing leukemia, lymphoma, and multiple myeloma target cells with and without cytokine (IL-2) and antibody (OKT3) priming in vitro. We conclude that acute exercise in healthy donors mobilizes NKT-like cells with an upregulation of transcriptomic programs involved in anti-tumor activity, but does not augment the ex vivo expansion of CIK cells.


Sujet(s)
Cellules CIK , Tumeurs , Cytotoxicité immunologique , Exercice physique , Humains , Interleukine-2/pharmacologie , Muromonab-CD3/pharmacologie , Transcriptome
3.
J Immunol Methods ; 502: 113231, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35122772

RÉSUMÉ

Cytokine release syndrome (CRS) is an undesired immune reaction that may cause dangerous side effects after the administration of novel biological therapies. In vitro cytokine release assays (CRA) are used for preclinical safety assessment prior to first-in-man dose administration of therapeutic monoclonal antibodies (mAbs). A variety of CRA platforms has been developed where the analysis of secreted cytokines is performed. Analysis of T cell activation markers is not performed routinely in CRA platforms and few studies have described intracellular cytokine levels after stimulation with therapeutic mAbs. In the present study, we performed a CRA using intracellular cytokine staining and assessment of extracellular T cell activation markers by flow cytometry. We used commercially available reference mAbs for the stimulation of peripheral blood mononuclear cells (PBMCs). We found that stimulation using solid phase (SP) dry coating with two different CD28 antibodies and muromonab-CD3 increased the percentage of IFN-É£ + CD4+ and CD8+ T cells as well as of CD3-CD56+ NK cells compared to stimulation with antibodies in aqueous phase (AP). Expression of the T cell activation markers CD25 and CD69 on CD4+ and CD8+ T cells was also increased upon SP muromonab-CD3 stimulation. Using multiplex cytokine assessment, we showed that stimulation in AP using ANC28.1, CD28.2 and muromonab-CD3 led to an increase of IFN-É£, GM-CSF, TNF-α, and IL-2 secretion. Stimulation of PBMCs preincubated at high-density culture led to an increase in IFN-É£ production but not in the expression of activation markers compared to low-density culture. Our findings demonstrated that flow cytometry analyses for assessing relevant T cell and NK cell markers may be used as a supplement to multiplex cytokine analysis in CRAs. The approach may be a valuable addition that enables a more precise description of the mechanisms leading to CRS.


Sujet(s)
Antinéoplasiques immunologiques , Produits pharmaceutiques biosimilaires , Anticorps monoclonaux , Antinéoplasiques immunologiques/pharmacologie , Antigène CD28 , Antigènes CD3 , Syndrome de libération de cytokines/induit chimiquement , Cytokines/métabolisme , Cytométrie en flux , Humains , Agranulocytes , Activation des lymphocytes , Muromonab-CD3/métabolisme , Muromonab-CD3/pharmacologie
4.
Front Immunol ; 10: 2391, 2019.
Article de Anglais | MEDLINE | ID: mdl-31681284

RÉSUMÉ

Background: Microvesicles (MVs) expressing the type 1 interferon (IFN)-inducible protein galectin-3 binding protein (G3BP) may play a pathogenic role in systemic lupus erythematosus (SLE). Co-expression of double-stranded DNA (dsDNA) on such MVs may render them immunogenic and targets for anti-dsDNA antibodies. Little is known about the mechanisms underlying generation of this MV population. In this study, we investigated how Toll-like receptors (TLRs), IFN-α, and T cells are involved in this process in healthy subjects. Methods: Peripheral blood mononuclear cells (PBMCs) isolated from 12 healthy donors were stimulated in-vitro for 24 h with a series of TLR-agonists or the T cell activating antibody OKT3 or were subjected to apoptosis by incubation with staurosporine. MVs in the supernatants were subsequently isolated by differential centrifugation and were quantified and characterized with respect to expression of G3BP and dsDNA by flow cytometry. Results: Stimulation of PBMCs with the TLR9-agonist and strong IFN-α inducer ODN2395 significantly increased the release of MVs expressing G3BP. The production of MVs with this phenotype was markedly enhanced by co-stimulation of T cells. Furthermore, dependency on IFN-α in the generation of G3BP-expressing MVs was indicated by a marked reduction following addition of the IFN-α inhibitor IFN alpha-IFNAR-IN-1 hydrochloride. Conclusion: Release of G3BP-expressing MVs from healthy donor PBMCs is induced by stimulation of TLR9 in an IFN-α-dependent manner and is enhanced by co-stimulation of T cells.


Sujet(s)
Microparticules membranaires/immunologie , ADN/immunologie , Interféron alpha/immunologie , Agranulocytes/immunologie , Récepteur-9 de type Toll-like/immunologie , Adulte , Antigènes néoplasiques , Marqueurs biologiques tumoraux , Femelle , Humains , Mâle , Muromonab-CD3/pharmacologie , Récepteur à l'interféron alpha-bêta/immunologie
5.
J Immunol ; 203(10): 2621-2629, 2019 11 15.
Article de Anglais | MEDLINE | ID: mdl-31578268

RÉSUMÉ

Oral tolerance is defined as the specific suppression of cellular and/or humoral immune responses to an Ag by prior administration of the Ag through the oral route. Although the investigation of oral tolerance has classically involved Ag feeding, we have found that oral administration of anti-CD3 mAb induced tolerance through regulatory T (Treg) cell generation. However, the mechanisms underlying this effect remain unknown. In this study, we show that conventional but not plasmacytoid dendritic cells (DCs) are required for anti-CD3-induced oral tolerance. Moreover, oral anti-CD3 promotes XCL1 secretion by small intestine lamina propria γδ T cells that, in turn, induces tolerogenic XCR1+ DC migration to the mesenteric lymph node, where Treg cells are induced and oral tolerance is established. Consistent with this, TCRδ-/- mice did not develop oral tolerance upon oral administration of anti-CD3. However, XCL1 was not required for oral tolerance induced by fed Ags, indicating that a different mechanism underlies this effect. Accordingly, oral administration of anti-CD3 enhanced oral tolerance induced by fed MOG35-55 peptide, resulting in less severe experimental autoimmune encephalomyelitis, which was associated with decreased inflammatory immune cell infiltration in the CNS and increased Treg cells in the spleen. Thus, Treg cell induction by oral anti-CD3 is a consequence of the cross-talk between γδ T cells and tolerogenic DCs in the gut. Furthermore, anti-CD3 may serve as an adjuvant to enhance oral tolerance to fed Ags.


Sujet(s)
Antigènes CD3/immunologie , Chimiokines C/métabolisme , Tolérance immunitaire/effets des médicaments et des substances chimiques , Lymphocytes intra-épithéliaux/immunologie , Muromonab-CD3/administration et posologie , Muromonab-CD3/pharmacologie , Administration par voie orale , Animaux , Mouvement cellulaire/immunologie , Cellules dendritiques/immunologie , Modèles animaux de maladie humaine , Encéphalomyélite auto-immune expérimentale/induit chimiquement , Encéphalomyélite auto-immune expérimentale/immunologie , Femelle , Techniques de knock-out de gènes , Gènes de la chaine delta du récepteur des lymphocytes T/génétique , Muqueuse intestinale/immunologie , Noeuds lymphatiques/immunologie , Mâle , Mésentère , Souris , Souris de lignée C57BL , Souris knockout , Glycoprotéine MOG/pharmacologie , Fragments peptidiques/pharmacologie , Lymphocytes T régulateurs/immunologie
6.
Immunohorizons ; 3(10): 498-510, 2019 10 21.
Article de Anglais | MEDLINE | ID: mdl-31636084

RÉSUMÉ

Immune cells express γ-aminobutyric acid receptors (GABA-R), and GABA administration can inhibit effector T cell responses in models of autoimmune disease. The pharmacokinetic properties of GABA, however, may be suboptimal for clinical applications. The amino acid homotaurine is a type A GABA-R (GABAA-R) agonist with good pharmacokinetics and appears safe for human consumption. In this study, we show that homotaurine inhibits in vitro T cell proliferation to a similar degree as GABA but at lower concentrations. In vivo, oral homotaurine treatment had a modest ability to reverse hyperglycemia in newly hyperglycemic NOD mice but was ineffective after the onset of severe hyperglycemia. In severely diabetic NOD mice, the combination of homotaurine and low-dose anti-CD3 treatment significantly increased 1) disease remission, 2) the percentages of splenic CD4+and CD8+ regulatory T cells compared with anti-CD3 alone, and 3) the frequencies of CD4+ and CD8+ regulatory T cells in the pancreatic lymph nodes compared with homotaurine monotherapy. Histological examination of their pancreata provided no evidence of the large-scale GABAA-R agonist-mediated replenishment of islet ß-cells that has been reported by others. However, we did observe a few functional islets in mice that received combined therapy. Thus, GABAA-R activation enhanced CD4+and CD8+ regulatory T cell responses following the depletion of effector T cells, which was associated with the preservation of some functional islets. Finally, we observed that homotaurine treatment enhanced ß-cell replication and survival in a human islet xenograft model. Hence, GABAA-R agonists, such as homotaurine, are attractive candidates for testing in combination with other therapeutic agents in type 1 diabetes clinical trials.


Sujet(s)
Lymphocytes T CD4+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Diabète expérimental/traitement médicamenteux , Diabète de type 1/traitement médicamenteux , Agonistes GABA/pharmacologie , Agonistes GABA/usage thérapeutique , Muromonab-CD3/usage thérapeutique , Taurine/analogues et dérivés , Animaux , Glycémie/effets des médicaments et des substances chimiques , Diabète expérimental/immunologie , Diabète de type 1/immunologie , Synergie des médicaments , Femelle , Humains , Cellules à insuline/effets des médicaments et des substances chimiques , Transplantation d'ilots de Langerhans , Souris de lignée NOD , Souris SCID , Muromonab-CD3/pharmacologie , Taurine/pharmacologie , Taurine/usage thérapeutique
7.
J Appl Physiol (1985) ; 127(6): 1539-1547, 2019 12 01.
Article de Anglais | MEDLINE | ID: mdl-31545153

RÉSUMÉ

Critically ill patients are at risk for sepsis, and immunosuppressive mechanisms may prevail. Whether functional tests are helpful to detect immune alterations is largely unknown. Therefore, we tested the hypotheses that reactivity of peripheral blood mononuclear cells (PBMCs) to secrete interferon-γ (IFNγ) following stimulation in vitro is decreased in patients with early sepsis compared with postoperative patients. IFNγ secretion [enzyme-linked immunospot (ELISpot)] in response to stimulation with cytomegalovirus (CMV), pokeweed mitogen (PWM), muromonab-anti-CD3 (OKT3), and human leukocyte antigen (HLA)-DRA-mRNA expression and serum cytokine concentrations were repeatedly [days 1, 3, 5, and 7 after intensive care unit (ICU) admission] determined in patients with sepsis (n = 7) and patients undergoing major abdominal surgery (radical prostatectomy, cystectomy, n = 10). In a second cohort, HLA-DRA expression was assessed in 80 patients with sepsis, 30 postoperative patients, and 44 healthy volunteers (German clinical trials database no. 00007694). In patients with sepsis, IFNγ secretion (ELISpot) was decreased compared with controls after stimulation with CMV (P = 0.01), OKT3 (P = 0.02), and PWM (P = 0.02 on day 5), whereas unstimulated IFNγ secretion did not differ. HLA-DRA expression was also significantly decreased in patients with sepsis at all time points (P = 0.004) compared with postoperative surgical patients, a finding confirmed in the larger cohort. Reactivity of PBMCs to stimulation with CMV, PWM, and OKT3 as well as HLA-DRA expression was already decreased upon ICU admission in patients with sepsis when compared with postoperative controls, suggesting early depression of acquired immunity. ELISpot assays may help to clinically characterize the time course of immunocompetence in patients with sepsis.NEW & NOTEWORTHY We observed suppression of reactivity to stimulation with cytomegalovirus, muromonab-anti-CD3, and pokeweed mitogen in mononuclear blood cells of patients with early sepsis when compared with postoperative controls. Thus, there is early depression of acquired immunity in sepsis. Enzyme-linked immunospot assays may help to characterize immunocompetence in patients with sepsis.


Sujet(s)
Cytomegalovirus/pathogénicité , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/virologie , Muromonab-CD3/pharmacologie , Mitogène phytolaque/pharmacologie , Sepsie/traitement médicamenteux , Sepsie/virologie , Adulte , Sujet âgé , Femelle , Humains , Interféron gamma/métabolisme , Mâle , Adulte d'âge moyen
8.
Transl Res ; 210: 43-56, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31082370

RÉSUMÉ

Cytokine release syndrome (CRS) is a serious and potentially life-threatening complication that can be associated with biological drug products. In vitro assays or in vivo tests using nonhuman primates may fail to predict CRS due to species differences and the complexity of immune system. Therefore, model species that have human-specific immune components may improve the ability to identify CRS and enhance product safety. In this study we used bone marrow-liver-thymus (BLT) humanized mice to test muromonab (OKT3), an anti-CD3 antibody with a black box warning for CRS. Initially, we completed pilot and dose escalation studies with muromonab and showed that when the dose was increased sufficiently, BLT-humanized mice experienced serious adverse outcomes including moribundity. Full studies compared muromonab treatment with adalimumab, saline, and a group pretreated with methylprednisolone prior to muromonab. We evaluated immune cell activation using flow cytometry and cytokine expression using a custom 10-plex cytokine assay to assess levels of human TNF-α, IFN-γ, IL-2, IL-6, IL-8, IL-10, IL-13, IL-17A, IL12/23p40, and GM-CSF. Muromonab treated mice had significant increases in all cytokines tested with T-cell depletion and T-cell activation noted. Adalimumab (active) and saline (inactive) control groups did not demonstrate cytokine expression changes or alterations in T-cell numbers or activation. Further, pretreatment with methylprednisolone blunted or abrogated cytokine increases. This study demonstrates that BLT-humanized mice are capable of experiencing CRS, and could be used to screen biologics for this adverse event to enhance patient safety.


Sujet(s)
Moelle osseuse/immunologie , Cytokines/métabolisme , Foie/immunologie , Thymus (glande)/immunologie , Animaux , Cellules de la moelle osseuse/cytologie , Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Humains , Foie/cytologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Numération des lymphocytes , Souris , Muromonab-CD3/pharmacologie , Rate/cytologie , Syndrome , Lymphocytes T/effets des médicaments et des substances chimiques
9.
J Crohns Colitis ; 12(12): 1410-1417, 2018 Nov 28.
Article de Anglais | MEDLINE | ID: mdl-30169593

RÉSUMÉ

BACKGROUND AND AIMS: Discontinuation of thiopurine analogues is common prior to live vaccines, during infection or when de-escalating therapy. Data regarding clearance of active metabolites and immune re-constitution is scant. We aimed to determine drug elimination and immune re-constitution following thiopurine cessation. METHODS: The elimination kinetics of 6-thioguanine nucleotides (6-TGN) were determined in nine inflammatory bowel disease [IBD] patients discontinuing thiopurines. Immune reconstitution was evaluated by toxic shock syndrome toxin 1 [TSST1] or anti-CD3 [OKT3]-induced CD4+ T-cell proliferation, following an initial exposure to TSST1 and 6-mercaptopurine [6MP], separately or combined. RESULTS: All patients discontinuing thiopurines displayed first-order elimination kinetics of 6-TGN, with a median elimination half-life of 6.8 days [interquartile range 5.9-8.4]. Resting CD4+ T-cells exposed to 6MP preserved their response to subsequent polyclonal or Vß2+-preferential stimulation. By contrast, exposure of TSST1-activated CD4+ T-cells to 6MP inhibited their subsequent Vß2+clonal response to further stimulation [p = 0.008], whereas overall response to further non-Vß2-selective stimulation with OKT3 was unaltered [p = 0.9]. CONCLUSIONS: Upon 6MP/azathioprine discontinuation, a 6-TGN elimination half-life of less than 10 days is expected in most patients. Immune reconstitution, however, may take longer for T-cell clones exposed to stimulation during thiopurine treatment. These findings may be useful when considering thiopurine cessation.


Sujet(s)
Azathioprine/pharmacocinétique , Toxines bactériennes/immunologie , Entérotoxines/immunologie , Nucléotides guanyliques/pharmacocinétique , Reconstitution immunitaire/immunologie , Maladies inflammatoires intestinales , Mercaptopurine/pharmacocinétique , Muromonab-CD3/pharmacologie , Superantigènes/immunologie , Thionucléotides/pharmacocinétique , Adulte , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Humains , Immunosuppresseurs/pharmacocinétique , Maladies inflammatoires intestinales/traitement médicamenteux , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/métabolisme , Mâle , Taux de clairance métabolique , Lymphocytes T/immunologie , Abstention thérapeutique
10.
Brain ; 139(Pt 7): 1939-57, 2016 07.
Article de Anglais | MEDLINE | ID: mdl-27246324

RÉSUMÉ

SEE WINGER AND ZAMVIL DOI101093/BRAIN/AWW121 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE: The innate immune system plays a central role in the chronic central nervous system inflammation that drives neurological disability in progressive forms of multiple sclerosis, for which there are no effective treatments. The mucosal immune system is a unique tolerogenic organ that provides a physiological approach for the induction of regulatory T cells. Here we report that nasal administration of CD3-specific antibody ameliorates disease in a progressive animal model of multiple sclerosis. This effect is IL-10-dependent and is mediated by the induction of regulatory T cells that share a similar transcriptional profile to Tr1 regulatory cells and that suppress the astrocyte inflammatory transcriptional program. Treatment results in an attenuated inflammatory milieu in the central nervous system, decreased microglia activation, reduced recruitment of peripheral monocytes, stabilization of the blood-brain barrier and less neurodegeneration. These findings suggest a new therapeutic approach for the treatment of progressive forms of multiple sclerosis and potentially other types of chronic central nervous system inflammation.


Sujet(s)
Astrocytes/immunologie , Antigènes CD3/immunologie , Encéphalomyélite auto-immune expérimentale/immunologie , Facteurs immunologiques/pharmacologie , Interleukine-10/immunologie , Muromonab-CD3/pharmacologie , Lymphocytes T régulateurs/immunologie , Administration par voie nasale , Animaux , Encéphalomyélite auto-immune expérimentale/traitement médicamenteux , Femelle , Facteurs immunologiques/administration et posologie , Souris , Souris de lignée C57BL , Souris de lignée NOD , Muromonab-CD3/administration et posologie , Pneumonie à pneumocoques/immunologie
11.
PLoS One ; 11(3): e0149093, 2016.
Article de Anglais | MEDLINE | ID: mdl-26959227

RÉSUMÉ

Therapeutic monoclonal antibodies (mAbs) such as the superagonistic, CD28-specific antibody TGN1412, or OKT3, an anti-CD3 mAb, can cause severe adverse events including cytokine release syndrome. A predictive model for mAb-mediated adverse effects, for which no previous knowledge on severe adverse events to be expected or on molecular mechanisms underlying is prerequisite, is not available yet. We used a humanized mouse model of human peripheral blood mononuclear cell-reconstituted NOD-RAG1-/-Aß-/-HLADQ(tg+ or tg-)IL-2Rγc-/- mice to evaluate its predictive value for preclinical testing of mAbs. 2-6 hours after TGN1412 treatment, mice showed a loss of human CD45+ cells from the peripheral blood and loss of only human T cells after OKT3 injection, reminiscent of effects observed in mAb-treated humans. Moreover, upon OKT3 injection we detected selective CD3 downmodulation on T cells, a typical effect of OKT3. Importantly, we detected release of human cytokines in humanized mice upon both OKT3 and TGN1412 application. Finally, humanized mice showed severe signs of illness, a rapid drop of body temperature, and succumbed to antibody application 2-6 hours after administration. Hence, the humanized mouse model used here reproduces several effects and adverse events induced in humans upon application of the therapeutic mAbs OKT3 and TGN1412.


Sujet(s)
Anticorps monoclonaux humanisés/effets indésirables , Cytokines/sang , Lymphopénie/sang , Lymphopénie/induit chimiquement , Animaux , Antigènes CD/métabolisme , Humains , Immunomodulation/effets des médicaments et des substances chimiques , Souris , Modèles animaux , Muromonab-CD3/pharmacologie , Lymphocytes T/immunologie
12.
Sci Signal ; 8(398): ra102, 2015 Oct 13.
Article de Anglais | MEDLINE | ID: mdl-26462735

RÉSUMÉ

The activation of T cells is the fundamental on switch for the adaptive immune system. Ca(2+) signaling is essential for T cell activation and starts as initial, short-lived, localized Ca(2+) signals. The second messenger nicotinic acid adenine dinucleotide phosphate (NAADP) forms rapidly upon T cell activation and stimulates early Ca(2+) signaling. We developed a high-resolution imaging technique using multiple fluorescent Ca(2+) indicator dyes to characterize these early signaling events and investigate the channels involved in NAADP-dependent Ca(2+) signals. In the first seconds of activation of either primary murine T cells or human Jurkat cells with beads coated with an antibody against CD3, we detected Ca(2+) signals with diameters close to the limit of detection and that were close to the activation site at the plasma membrane. In Jurkat cells in which the ryanodine receptor (RyR) was knocked down or in primary T cells from RyR1(-/-) mice, either these early Ca(2+) signals were not detected or the number of signals was markedly reduced. Local Ca(2+) signals observed within 20 ms upon microinjection of Jurkat cells with NAADP were also sensitive to RyR knockdown. In contrast, TRPM2 (transient receptor potential channel, subtype melastatin 2), a potential NAADP target channel, was not required for the formation of initial Ca(2+) signals in primary T cells. Thus, through our high-resolution imaging method, we characterized early Ca(2+) release events in T cells and obtained evidence for the involvement of RyR and NAADP in such signals.


Sujet(s)
Signalisation calcique , Calcium/métabolisme , NADP/analogues et dérivés , Canal de libération du calcium du récepteur à la ryanodine/métabolisme , Lymphocytes T/métabolisme , Dérivés de l'aniline/composition chimique , Animaux , Benzofuranes/composition chimique , Cellules cultivées , Fluorimétrie/méthodes , Humains , Imidazoles/composition chimique , Cellules Jurkat , Activation des lymphocytes/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Souris knockout , Microscopie de fluorescence , Muromonab-CD3/pharmacologie , NADP/métabolisme , Canal de libération du calcium du récepteur à la ryanodine/génétique , Lymphocytes T/effets des médicaments et des substances chimiques , Canaux cationiques TRPM/métabolisme , Facteurs temps , Xanthènes/composition chimique
13.
J Neural Transm (Vienna) ; 122(1): 109-22, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25377522

RÉSUMÉ

Lithium is an important psychopharmacological agent for the treatment of unipolar as well as bipolar affective disorders. Lithium has a number of side effects such as hypothyroidism and aggravation of psoriasis. On the other hand, lithium has pro-inflammatory effects, which appear beneficial in some disorders associated with immunological deficits, such as human immunodeficiency virus (HIV) infection and systemic lupus erythematosus (SLE). Therefore, immunological characteristics of lithium may be an important consideration in individualized therapeutic decisions. We measured the levels of the cytokines interleukin (IL)-1ß, IL-2, IL-4, IL-6, IL-22, IL-17 and tumour necrosis factor (TNF)-α in the stimulated blood of thirty healthy subjects supplemented with lithium alone, the antidepressants citalopram, escitalopram or mirtazapine alone, the combination of each antidepressant with lithium, and a no drug control. These drugs were tested under three blood stimulant conditions: murine anti-human CD3 monoclonal antibody OKT3 and the 5C3 monoclonal antibody (OKT3/5C3), phytohemagglutinin (PHA), and unstimulated blood. Lithium, alone and in combination with any of the tested antidepressants, led to a consistent increase of IL-1ß, IL-6 and TNF-α levels in the unstimulated as well as the stimulated blood. In the OKT3/5C3- and PHA-stimulated blood, IL-17 production was significantly enhanced by lithium. Lithium additionally increased IL-2 concentrations significantly in PHA-stimulated blood. The data support the view that lithium has pro-inflammatory properties. These immunological characteristics may contribute to side effects of lithium, but may also explain its beneficial effects in patients suffering from HIV infection or SLE.


Sujet(s)
Antidépresseurs/pharmacologie , Cellules sanguines/effets des médicaments et des substances chimiques , Cytokines/métabolisme , Lithium/pharmacologie , Adulte , Relation dose-effet des médicaments , Femelle , Cytométrie en flux , Humains , Immunosuppresseurs/pharmacologie , Mâle , Adulte d'âge moyen , Mitogènes/pharmacologie , Muromonab-CD3/pharmacologie , Phytohémagglutinine/pharmacologie , Statistique non paramétrique , Facteurs temps , Jeune adulte
14.
Oxid Med Cell Longev ; 2014: 806162, 2014.
Article de Anglais | MEDLINE | ID: mdl-24757498

RÉSUMÉ

Increased cytokine production possibly due to oxidative stress has repeatedly been shown to play a pivotal role in the pathophysiology of epilepsy and bipolar disorder. Recent in vitro and animal studies of valproic acid (VPA) report antioxidative and anti-inflammatory properties, and suppression of interleukin (IL)-6 and tumor necrosis factor (TNF)-α. We tested the effect of drugs with antiepileptic or mood stabilizer properties, namely, primidone (PRM), carbamazepine (CBZ), levetiracetam (LEV), lamotrigine (LTG), VPA, oxcarbazepine (OXC), topiramate (TPM), phenobarbital (PB), and lithium on the production of the following cytokines in vitro: interleukin (IL)-1ß, IL-2, IL-4, IL-6, IL-17, IL-22, and TNF-α. We performed a whole blood assay with stimulated blood of 14 healthy female subjects. Anti-human CD3 monoclonal antibody OKT3, combined with 5C3 antibody against CD40, was used as stimulant. We found a significant reduction of IL-1 and IL-2 levels with all tested drugs other than lithium in the CD3/5C3-stimulated blood; VPA led to a decrease in IL-1ß, IL-2, IL-4, IL-6, IL-17, and TNF-α production, which substantiates and adds knowledge to current hypotheses on VPA's anti-inflammatory properties.


Sujet(s)
Affect/effets des médicaments et des substances chimiques , Anticonvulsivants/pharmacologie , Antigènes CD3/métabolisme , Antigènes CD40/métabolisme , Cytokines/sang , Adulte , Femelle , Humains , Adulte d'âge moyen , Muromonab-CD3/pharmacologie , Jeune adulte
15.
PLoS One ; 8(2): e56820, 2013.
Article de Anglais | MEDLINE | ID: mdl-23441216

RÉSUMÉ

BACKGROUND AND PURPOSE: Although gene-modification of T cells to express tumor-related antigen-specific T-cell receptor (TCR) or chimeric antigen receptor (CAR) has clinically proved promise, there still remains room to improve the clinical efficacy of re-directed T-cell based antitumor adoptive therapy. In order to achieve more objective clinical responses using ex vivo-expanded tumor-responsive T cells, the infused T cells need to show adequate localized infiltration into the tumor. METHODOLOGY/PRINCIPAL FINDINGS: Human lung cancer cells variously express a tumor antigen, Wilms' Tumor gene product 1 (WT1), and an inflammatory chemokine, CCL2. However, CCR2, the relevant receptor for CCL2, is rarely expressed on activated T-lymphocytes. A HLA-A2402(+) human lung cancer cell line, LK79, which expresses high amounts of both CCL2 and WT1 mRNA, was employed as a target. Normal CD8(+) T cells were retrovirally gene-modified to express both CCR2 and HLA-A*2402-restricted and WT1(235-243) nonapeptide-specific TCR as an effector. Anti-tumor functionality mediated by these effector cells against LK79 cells was assessed both in vitro and in vivo. Finally the impact of CCL2 on WT1 epitope-responsive TCR signaling mediated by the effector cells was studied. Introduced CCR2 was functionally validated using gene-modified Jurkat cells and human CD3(+) T cells both in vitro and in vivo. Double gene-modified CD3(+) T cells successfully demonstrated both CCL2-tropic tumor trafficking and cytocidal reactivity against LK79 cells in vitro and in vivo. CCL2 augmented the WT1 epitope-responsive TCR signaling shown by relevant luciferase production in double gene-modified Jurkat/MA cells to express luciferase and WT1-specific TCR, and CCL2 also dose-dependently augmented WT1 epitope-responsive IFN-γ production and CD107a expression mediated by these double gene-modified CD3(+) T cells. CONCLUSION/SIGNIFICANCE: Introduction of the CCL2/CCR2 axis successfully potentiated in vivo anti-lung cancer reactivity mediated by CD8(+) T cells double gene-modified to express WT1-specific TCR and CCR2 not only via CCL2-tropic tumor trafficking, but also CCL2-enhanced WT1-responsiveness.


Sujet(s)
Tumeurs du poumon/génétique , Tumeurs du poumon/immunologie , Récepteurs aux antigènes des cellules T/génétique , Récepteurs CCR2/génétique , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Protéines WT1/immunologie , Animaux , Lymphocytes T CD8+/immunologie , Lymphocytes T CD8+/métabolisme , Lignée cellulaire tumorale , Modèles animaux de maladie humaine , Femelle , Régulation de l'expression des gènes tumoraux , Humains , Interleukine-2/pharmacologie , Tumeurs du poumon/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/génétique , Activation des lymphocytes/immunologie , Souris , Muromonab-CD3/pharmacologie , Récepteurs CCR2/métabolisme , Reproductibilité des résultats , Transfection , Transplantation hétérologue
16.
DNA Cell Biol ; 31(11): 1628-35, 2012 Nov.
Article de Anglais | MEDLINE | ID: mdl-23057733

RÉSUMÉ

T-cell immunodeficiency is a common feature in patients with chronic myeloid leukemia (CML), and deficiency in CD3 levels was detected in T cells from these patients, which may represent a characteristic that is related to a lower T cell activation. In this study, we explored the possibility that forced TCRζ gene expression may upreg-u-late T cell receptor (TCR) signaling activation and reverse interleukin-2 (IL-2) production in T cells from patients with CML. A recombinant eukaryotic vector expressing TCRζ was transfected into T cells by nucleofection. Phosphorylated TCRζ, phosphorylated NF-κB, and the IL-2 level in TCRζ-transfected CD3+T cells that were activated with anti-CD3 and anti-CD28 antibodies were measured by Western blot and enzyme-linked immunosorbent assay (ELISA). Significantly increased TCRζ levels were found in TCRζ-transfected CD3+T cells. After CD3 and CD28 antibody stimulation, a significantly higher phosphorylated TCRζ chain level was demonstrated, and an increased IL-2 production in TCRζ-upregulated T cells was associated with the increased expression of the phosphorylated NF-κB. In conclusion, TCRζ gene transfection could restore TCRζ chain deficiency and enhance IL-2 production in T cells from patients with CML. It is possible that TCRζ chain reconstitution in leukemia-specific, clonally expanded T cells will effectively increase their activation of antileukemia cytotoxicity.


Sujet(s)
Interleukine-2/immunologie , Leucémie myéloïde chronique BCR-ABL positive/immunologie , Récepteurs aux antigènes des cellules T/immunologie , Lymphocytes T/immunologie , Technique de Western , Antigènes CD3/immunologie , Antigènes CD3/métabolisme , Cellules cultivées , Test ELISA , Cytométrie en flux , Protéines à fluorescence verte/génétique , Protéines à fluorescence verte/métabolisme , Cellules HEK293 , Humains , Interleukine-2/biosynthèse , Cellules Jurkat , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Microscopie de fluorescence , Muromonab-CD3/immunologie , Muromonab-CD3/pharmacologie , Facteur de transcription NF-kappa B/immunologie , Facteur de transcription NF-kappa B/métabolisme , Phosphorylation , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/métabolisme , Lymphocytes T/métabolisme , Transfection , Régulation positive
17.
Cancer Res ; 72(19): 4931-43, 2012 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-22875025

RÉSUMÉ

CD200 is a transmembrane molecule with an important immunoregulatory role that is overexpressed on most chronic lymphocytic leukemia (CLL) cells. In this study, we characterized a previously unknown soluble form of this molecule in human plasma termed sCD200. Levels of sCD200 were elevated in the plasma of patients with CLL as compared with healthy controls, and there was a significant correlation with CLL disease stage. Infusion of sCD200(hi) CLL plasma into severely immunocompromised NOD.SCIDγ(c)(null) (NSG) mice enhanced the engraftment of CLL splenocytes as compared with mice receiving sCD200(lo) normal plasma. CLL cells were detected in both the spleen and peritoneal cavity of animals for up to 75 days. Engraftment of CLL cells did not occur after infusion of CLL plasma depleted of sCD200 and was abolished in mice treated with anti-CD200 or OKT3 monoclonal antibody (mAb), suggesting a role for both sCD200 and T cells in CLL engraftment. Notably, anti-CD200 mAb was as effective as rituximab in eliminating engrafted CLL cells when administered 21 days after engraftment. Taken together, our findings point to sCD200 as a novel prognostic marker and therapeutic target for CLL. Furthermore, the humanized mouse model described here may prove valuable to preclinically assess new treatment regimens for CLL.


Sujet(s)
Antigènes CD/immunologie , Sujet immunodéprimé/immunologie , Leucémie chronique lymphocytaire à cellules B/immunologie , Rate/immunologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux d'origine murine/pharmacologie , Antigènes CD/sang , Antinéoplasiques/pharmacologie , Cellules cultivées , Femelle , Humains , Sous-unité gamma commune aux récepteurs des interleukines/déficit , Sous-unité gamma commune aux récepteurs des interleukines/génétique , Sous-unité gamma commune aux récepteurs des interleukines/immunologie , Leucémie chronique lymphocytaire à cellules B/traitement médicamenteux , Leucémie chronique lymphocytaire à cellules B/anatomopathologie , Mâle , Souris , Souris de lignée NOD , Souris knockout , Souris SCID , Adulte d'âge moyen , Muromonab-CD3/pharmacologie , Rituximab , Rate/effets des médicaments et des substances chimiques , Rate/anatomopathologie , Transplantation hétérologue , Résultat thérapeutique , Cellules cancéreuses en culture
18.
J Immunol Methods ; 384(1-2): 33-42, 2012 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-22796190

RÉSUMÉ

CD3-specific antibodies have shown clinical efficacy in both transplantation and autoimmunity. However, targeting CD3 in this way can lead to T-cell activation and a serious cytokine release syndrome mediated by Fcγ receptor binding. An in vivo mouse model has been developed using severe combined immunodeficient (SCID) mice to detect human T-cell depletion and cytokine release into the circulation after administration of OKT3. This system has been used to evaluate OKT3 antibody fragments lacking the entire Fc region alongside whole antibody constructs. These data clearly show that cytokine release is detected with all OKT3 antibody constructs and fragments tested and these can be ranked from highest to lowest as follows: mIgG2a>hIgG1 (Ala-Ala)>hIgG1 diFab' maleimide (DFM)>hIgG1 F(ab')2>mIgG2a F(ab')2>hIgG1 Fab'. Furthermore, the monovalent hIgG1 Fab' fragment gives the least cytokine release but it does not deplete human T-cells in this assay format. This suggests that T-cell activation may be playing a role in the mechanism of action of anti-CD3 antibodies and consequently the unwanted cytokine release is potentially unavoidable for this class of molecules. This model system provides a useful tool to aid in understanding and reducing the potential risks of cytokine release following antibody therapy.


Sujet(s)
Antigènes CD3/immunologie , Cytokines/immunologie , Muromonab-CD3/immunologie , Lymphocytes T/immunologie , Animaux , Cellules cultivées , Cytokines/sang , Cytokines/métabolisme , Cytométrie en flux , Humains , Fragments Fab d'immunoglobuline/immunologie , Fragments Fab d'immunoglobuline/pharmacologie , Fragments Fc des immunoglobulines/immunologie , Fragments Fc des immunoglobulines/pharmacologie , Immunoglobuline G/immunologie , Immunoglobuline G/pharmacologie , Techniques immunologiques/méthodes , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/immunologie , Agranulocytes/métabolisme , Activation des lymphocytes/effets des médicaments et des substances chimiques , Activation des lymphocytes/immunologie , Déplétion lymphocytaire , Souris , Souris SCID , Muromonab-CD3/pharmacologie , Reproductibilité des résultats , Lymphocytes T/effets des médicaments et des substances chimiques , Lymphocytes T/métabolisme
19.
PLoS One ; 7(1): e30229, 2012.
Article de Anglais | MEDLINE | ID: mdl-22279573

RÉSUMÉ

BACKGROUND: Using in vivo mouse models, the mechanisms of CD4+ T cell help have been intensively investigated. However, a mechanistic analysis of human CD4+ T cell help is largely lacking. Our goal was to elucidate the mechanisms of human CD4+ T cell help of CD8+ T cell proliferation using a novel in vitro model. METHODS/PRINCIPAL FINDINGS: We developed a genetically engineered novel human cell-based artificial APC, aAPC/mOKT3, which expresses a membranous form of the anti-CD3 monoclonal antibody OKT3 as well as other immune accessory molecules. Without requiring the addition of allogeneic feeder cells, aAPC/mOKT3 enabled the expansion of both peripheral and tumor-infiltrating T cells, regardless of HLA-restriction. Stimulation with aAPC/mOKT3 did not expand Foxp3+ regulatory T cells, and expanded tumor infiltrating lymphocytes predominantly secreted Th1-type cytokines, interferon-γ and IL-2. In this aAPC-based system, the presence of autologous CD4+ T cells was associated with significantly improved CD8+ T cell expansion in vitro. The CD4+ T cell derived cytokines IL-2 and IL-21 were necessary but not sufficient for this effect. However, CD4+ T cell help of CD8+ T cell proliferation was partially recapitulated by both adding IL-2/IL-21 and by upregulation of IL-21 receptor on CD8+ T cells. CONCLUSIONS: We have developed an in vitro model that advances our understanding of the immunobiology of human CD4+ T cell help of CD8+ T cells. Our data suggests that human CD4+ T cell help can be leveraged to expand CD8+ T cells in vitro.


Sujet(s)
Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Prolifération cellulaire , Antigènes CD3/immunologie , Antigènes CD3/métabolisme , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD8+/effets des médicaments et des substances chimiques , Lymphocytes T CD8+/métabolisme , Cellules cultivées , Techniques de coculture , Cytokines/immunologie , Cytokines/métabolisme , Cytométrie en flux , Humains , Interféron gamma/immunologie , Interféron gamma/métabolisme , Interleukine-2/immunologie , Interleukine-2/métabolisme , Interleukine-2/pharmacologie , Interleukines/immunologie , Interleukines/métabolisme , Interleukines/pharmacologie , Cellules K562 , Lymphocytes TIL/effets des médicaments et des substances chimiques , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Muromonab-CD3/immunologie , Muromonab-CD3/métabolisme , Muromonab-CD3/pharmacologie , Récepteurs à l'interleukine-21/immunologie , Récepteurs à l'interleukine-21/métabolisme , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Régulation positive/effets des médicaments et des substances chimiques
20.
Chin J Cancer ; 31(6): 287-94, 2012 Jun.
Article de Anglais | MEDLINE | ID: mdl-22257383

RÉSUMÉ

Establishing Epstein-Barr virus(EBV)-specific cytolytic T lymphocytes(EBV-CTLs) from peripheral blood mononuclear cells(PBMCs) for adoptive immunotherapy has been reported in EBV-associated malignancies including Hodgkin's lymphoma and nasopharyngeal carcinoma(NPC). In the current study, we performed ex vivo expansion of tumor-infiltrating lymphocytes(TILs) obtained from NPC biopsy specimens with a rapid expansion protocol using anti-CD3 monoclonal antibody(OKT3), recombinant human interleukin(IL)-2, and irradiated PBMCs from healthy donors to initiate the growth of TILs. Young TIL cultures comprised of more than 90% of CD3+ T cells, a variable percentage of CD3+CD8+ and CD3+CD4+ T cells, and less than 10% of CD3-CD16+ natural killer cells, a similar phenotype of EBV-CTL cultures from PBMCs. Interestingly, TIL cultures secreted high levels of the Th1 cytokines, interferon gamma (IFNγ) and tumor necrosis factor-alpha (TNF-α), and low levels of the Th2 cytokines, IL-4 and IL-10. Moreover, young TILs could recognize autologous EBV-transformed B lymphoblast cell lines, but not autologous EBV-negative blast cells or allogeneic EBV-negative tumor cells. Taken together, these data suggest that ex vivo expansion of TILs from NPC biopsy tissue is an appealing alternative method to establish T cell-based immunotherapy for NPC.


Sujet(s)
Immunothérapie adoptive , Lymphocytes TIL/immunologie , Muromonab-CD3/pharmacologie , Tumeurs du rhinopharynx/anatomopathologie , Tumeurs du rhinopharynx/thérapie , Lymphocytes T cytotoxiques/immunologie , Biopsie , Antigènes CD3/analyse , Antigènes CD4/analyse , Antigènes CD8/analyse , Cellules cultivées , Herpèsvirus humain de type 4/immunologie , Humains , Interféron gamma/métabolisme , Interleukine-10/métabolisme , Interleukine-2/pharmacologie , Interleukine-4/métabolisme , Lymphocytes TIL/virologie , Monocytes/anatomopathologie , Tumeurs du rhinopharynx/immunologie , Tumeurs du rhinopharynx/virologie , Récepteurs du fragment Fc des IgG/analyse , Lymphocytes T cytotoxiques/virologie , Facteur de nécrose tumorale alpha/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE