Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.675
Filtrar
1.
Nat Commun ; 15(1): 5842, 2024 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-38992037

RESUMO

Activating interferon responses with STING agonists (STINGa) is a current cancer immunotherapy strategy, and therapeutic modalities that enable tumor-targeted delivery via systemic administration could be beneficial. Here we demonstrate that tumor cell-directed STING agonist antibody-drug-conjugates (STINGa ADCs) activate STING in tumor cells and myeloid cells and induce anti-tumor innate immune responses in in vitro, in vivo (in female mice), and ex vivo tumor models. We show that the tumor cell-directed STINGa ADCs are internalized into myeloid cells by Fcγ-receptor-I in a tumor antigen-dependent manner. Systemic administration of STINGa ADCs in mice leads to STING activation in tumors, with increased anti-tumor activity and reduced serum cytokine elevations compared to a free STING agonist. Furthermore, STINGa ADCs induce type III interferons, which contribute to the anti-tumor activity by upregulating type I interferon and other key chemokines/cytokines. These findings reveal an important role for type III interferons in the anti-tumor activity elicited by STING agonism and provide rationale for the clinical development of tumor cell-directed STINGa ADCs.


Assuntos
Imunidade Inata , Imunoconjugados , Interferons , Proteínas de Membrana , Animais , Proteínas de Membrana/agonistas , Proteínas de Membrana/imunologia , Imunidade Inata/efeitos dos fármacos , Feminino , Humanos , Camundongos , Linhagem Celular Tumoral , Imunoconjugados/farmacologia , Imunoconjugados/administração & dosagem , Interferons/metabolismo , Interferon lambda , Neoplasias/imunologia , Neoplasias/tratamento farmacológico , Interferon Tipo I/imunologia , Citocinas/metabolismo , Células Mieloides/imunologia , Células Mieloides/efeitos dos fármacos , Imunoterapia/métodos , Camundongos Endogâmicos C57BL , Receptores de IgG/agonistas , Receptores de IgG/metabolismo , Receptores de IgG/imunologia
2.
Front Immunol ; 15: 1400177, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38953027

RESUMO

Background: Chimeric antigen receptor T (CAR-T) cell therapies have achieved remarkable success in the treatment of hematological tumors. However, given the distinct features of solid tumors, particularly heterogeneity, metabolic aggressiveness, and fewer immune cells in tumor microenvironment (TME), the practical utility of CAR-T cells for solid tumors remains as a challenging issue. Meanwhile, although anti-PD-1 monoclonal antibody (mAb) has shown clinical efficacy, most mAbs also show limited clinical benefits for solid tumors due mainly to the issues associated with the lack of immune cells in TME. Thus, the infiltration of targeted immunological active cells into TME could generate synergistic efficacy for mAbs. Methods: We present a combinational strategy for solid tumor treatment, which combines armored-T cells to express Fc-gamma receptor I (FcγRI) fragment on the surfaces for targeting various tumors with therapeutically useful mAbs. Choosing CD20 and HER-2 as the targets, we characterized the in vitro and in vivo efficacy and latent mechanism of the combination drug by using flow cytometry, ELISA and other methods. Results: The combination and preprocessing of armored T-cells with corresponding antibody of Rituximab and Pertuzumab exerted profound anti-tumor effects, which is demonstrated to be mediated by synergistically produced antibody-dependent cellular cytotoxicity (ADCC) effects. Meanwhile, mAb was able to carry armored-T cell by preprocessing for the infiltration to TME in cell derived xenograft (CDX) model. Conclusions: This combination strategy showed a significant increase of safety profiles from the reduction of antibody doses. More importantly, the present strategy could be a versatile tool for a broad spectrum of cancer treatment, with a simple pairing of engineered T cells and a conventional antibody.


Assuntos
Neoplasias , Receptores de IgG , Linfócitos T , Microambiente Tumoral , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Humanos , Animais , Camundongos , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/tratamento farmacológico , Linfócitos T/imunologia , Microambiente Tumoral/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais/imunologia , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Imunoterapia Adotiva/métodos , Receptor ErbB-2/imunologia , Receptor ErbB-2/antagonistas & inibidores , Antineoplásicos Imunológicos/farmacologia , Antineoplásicos Imunológicos/uso terapêutico , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/genética , Feminino , Antígenos CD20/imunologia
3.
Elife ; 122024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38885133

RESUMO

Despite the central role of T cells in tumor immunity, attempts to harness their cytotoxic capacity as a therapy have met limited efficacy, partially as a result of the suppressive microenvironment which limits their migration and activation. In contrast, myeloid cells massively infiltrate tumors and are well adapted to survive these harsh conditions. While they are equipped with cell-killing abilities, they often adopt an immunosuppressive phenotype upon migration to tumors. Therefore, the questions of how to modify their activation programming against cancer, and what signaling cascades should be activated in myeloid cells to elicit their cytotoxicity have remained unclear. Here, we found that activation of IgM-induced signaling in murine myeloid cells results in secretion of lytic granules and massive tumor cell death. These findings open venues for designing novel immunotherapy by equipping monocytes with chimeric receptors that target tumor antigens and consequently, signal through IgM receptor. Nonetheless, we found that myeloid cells do not express the antibody-derived portion used to recognize the tumor antigen due to the induction of an ER stress response. To overcome this limitation, we designed chimeric receptors that are based on the high-affinity FcγRI for IgG. Incubation of macrophages expressing these receptors along with tumor-binding IgG induced massive tumor cell killing and secretion of reactive oxygen species and Granzyme B. Overall, this work highlights the challenges involved in genetically reprogramming the signaling in myeloid cells and provides a framework for endowing myeloid cells with antigen-specific cytotoxicity.


Assuntos
Células Mieloides , Receptores de IgG , Animais , Receptores de IgG/metabolismo , Receptores de IgG/imunologia , Camundongos , Células Mieloides/imunologia , Células Mieloides/metabolismo , Camundongos Endogâmicos C57BL , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Imunoglobulina M/metabolismo , Imunoglobulina M/imunologia , Transdução de Sinais , Macrófagos/imunologia , Macrófagos/metabolismo , Neoplasias/imunologia
4.
Front Immunol ; 15: 1409333, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38919608

RESUMO

Introduction: Therapeutic antibodies have become a major strategy to treat oncologic diseases. For chronic lymphocytic leukemia, antibodies against CD20 are used to target and elicit cytotoxic responses against malignant B cells. However, efficacy is often compromised due to a suppressive microenvironment that interferes with cellular immune responses. To overcome this suppression, agonists of pattern recognition receptors have been studied which promote direct cytotoxicity or elicit anti-tumoral immune responses. NOD2 is an intracellular pattern recognition receptor that participates in the detection of peptidoglycan, a key component of bacterial cell walls. This detection then mediates the activation of multiple signaling pathways in myeloid cells. Although several NOD2 agonists are being used worldwide, the potential benefit of these agents in the context of antibody therapy has not been explored. Methods: Primary cells from healthy-donor volunteers (PBMCs, monocytes) or CLL patients (monocytes) were treated with versus without the NOD2 agonist L18-MDP, then antibody-mediated responses were assessed. In vivo, the Eµ-TCL1 mouse model of CLL was used to test the effects of L18-MDP treatment alone and in combination with anti-CD20 antibody. Results: Treatment of peripheral blood mononuclear cells with L18-MDP led to activation of monocytes from both healthy donors and CLL patients. In addition, there was an upregulation of activating FcγR in monocytes and a subsequent increase in antibody-mediated phagocytosis. This effect required the NF-κB and p38 signaling pathways. Treatment with L18-MDP plus anti-CD20 antibody in the Eµ-TCL model of CLL led to a significant reduction of CLL load, as well as to phenotypic changes in splenic monocytes and macrophages. Conclusions: Taken together, these results suggest that NOD2 agonists help overturn the suppression of myeloid cells, and may improve the efficacy of antibody therapy for CLL.


Assuntos
Leucemia Linfocítica Crônica de Células B , Macrófagos , Proteína Adaptadora de Sinalização NOD2 , Receptores de IgG , Proteína Adaptadora de Sinalização NOD2/agonistas , Proteína Adaptadora de Sinalização NOD2/metabolismo , Proteína Adaptadora de Sinalização NOD2/imunologia , Animais , Humanos , Receptores de IgG/metabolismo , Receptores de IgG/imunologia , Camundongos , Macrófagos/imunologia , Macrófagos/metabolismo , Leucemia Linfocítica Crônica de Células B/imunologia , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Linfocítica Crônica de Células B/metabolismo , Acetilmuramil-Alanil-Isoglutamina/farmacologia , Feminino , Camundongos Endogâmicos C57BL , Transdução de Sinais , Fagocitose , Rituximab/farmacologia , Rituximab/uso terapêutico
5.
Sci Rep ; 14(1): 14595, 2024 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-38918496

RESUMO

There are two known mechanisms by which natural killer (NK) cells recognize and kill diseased targets: (i) direct killing and (ii) antibody-dependent cell-mediated cytotoxicity (ADCC). We investigated an indirect NK cell activation strategy for the enhancement of human NK cell killing function. We did this by leveraging the fact that toll-like receptor 9 (TLR9) agonism within pools of human peripheral blood mononuclear cells (PBMCs) results in a robust interferon signaling cascade that leads to NK cell activation. After TLR9 agonist stimulation, NK cells were enriched and incorporated into assays to assess their ability to kill tumor cell line targets. Notably, differential impacts of TLR9 agonism were observed-direct killing was enhanced while ADCC was not increased. To ensure that the observed differential effects were not attributable to differences between human donors, we recapitulated the observation using our Natural Killer-Simultaneous ADCC and Direct Killing Assay (NK-SADKA) that controls for human-to-human differences. Next, we observed a treatment-induced decrease in NK cell surface CD16-known to be shed by NK cells post-activation. Given the essential role of CD16 in ADCC, such shedding could account for the observed differential impact of TLR9 agonism on NK cell-mediated killing capacity.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Células Matadoras Naturais , Receptor Toll-Like 9 , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Receptores de IgG/metabolismo , Receptores de IgG/imunologia , Linhagem Celular Tumoral , Citotoxicidade Imunológica/efeitos dos fármacos
6.
Nat Commun ; 15(1): 5280, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38902261

RESUMO

The regulatory circuits dictating CD8+ T cell responsiveness versus exhaustion during anti-tumor immunity are incompletely understood. Here we report that tumor-infiltrating antigen-specific PD-1+ TCF-1- CD8+ T cells express the immunosuppressive cytokine Fgl2. Conditional deletion of Fgl2 specifically in mouse antigen-specific CD8+ T cells prolongs CD8+ T cell persistence, suppresses phenotypic and transcriptomic signatures of T cell exhaustion, and improves control of the tumor. In a mouse model of chronic viral infection, PD-1+ CD8+ T cell-derived Fgl2 also negatively regulates virus-specific T cell responses. In humans, CD8+ T cell-derived Fgl2 is associated with poorer survival in patients with melanoma. Mechanistically, the dampened responsiveness of WT Fgl2-expressing CD8+ T cells, when compared to Fgl2-deficient CD8+ T cells, is underpinned by the cell-intrinsic interaction of Fgl2 with CD8+ T cell-expressed FcγRIIB and concomitant caspase 3/7-mediated apoptosis. Our results thus illuminate a cell-autonomous regulatory axis by which PD-1+ CD8+ T cells both express the receptor and secrete its ligand in order to mediate suppression of anti-tumor and anti-viral immunity.


Assuntos
Linfócitos T CD8-Positivos , Melanoma , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1 , Receptores de IgG , Animais , Feminino , Humanos , Camundongos , Apoptose , Linfócitos T CD8-Positivos/imunologia , Melanoma/imunologia , Melanoma/genética , Camundongos Knockout , Receptor de Morte Celular Programada 1/metabolismo , Receptor de Morte Celular Programada 1/genética , Receptores de IgG/metabolismo , Receptores de IgG/genética , Receptores de IgG/imunologia
7.
Front Immunol ; 15: 1374818, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38827738

RESUMO

Activated lung ILC2s produce large quantities of IL-5 and IL-13 that contribute to eosinophilic inflammation and mucus production following respiratory syncytial virus infection (RSV). The current understanding of ILC2 activation during RSV infection, is that ILC2s are activated by alarmins, including IL-33, released from airway epithelial cells in response to viral-mediated damage. Thus, high levels of RSV neutralizing maternal antibody generated from maternal immunization would be expected to reduce IL-33 production and mitigate ILC2 activation. Here we report that lung ILC2s from mice born to RSV-immunized dams become activated despite undetectable RSV replication. We also report, for the first time, expression of activating and inhibitory Fcgamma receptors on ILC2s that are differentially expressed in offspring born to immunized versus unimmunized dams. Alternatively, ex vivo IL-33-mediated activation of ILC2s was mitigated following the addition of antibody: antigen immune complexes. Further studies are needed to confirm the role of Fcgamma receptor ligation by immune complexes as an alternative mechanism of ILC2 regulation in RSV-associated eosinophilic lung inflammation.


Assuntos
Interleucina-33 , Pulmão , Camundongos Endogâmicos BALB C , Infecções por Vírus Respiratório Sincicial , Vírus Sinciciais Respiratórios , Animais , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções por Vírus Respiratório Sincicial/prevenção & controle , Camundongos , Feminino , Pulmão/imunologia , Pulmão/virologia , Interleucina-33/imunologia , Vírus Sinciciais Respiratórios/imunologia , Linfócitos/imunologia , Imunização , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Anticorpos Antivirais/imunologia , Gravidez , Vacinas contra Vírus Sincicial Respiratório/imunologia
8.
J Vis Exp ; (206)2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38738886

RESUMO

Monoclonal antibody-based immunotherapy targeting tumor antigens is now a mainstay of cancer treatment. One of the clinically relevant mechanisms of action of the antibodies is antibody-dependent cellular cytotoxicity (ADCC), where the antibody binds to the cancer cells and engages the cellular component of the immune system, e.g., natural killer (NK) cells, to kill the tumor cells. The effectiveness of these therapies could be improved by identifying adjuvant compounds that increase the sensitivity of the cancer cells or the potency of the immune cells. In addition, undiscovered drug interactions in cancer patients co-medicated for previous conditions or cancer-associated symptoms may determine the success of the antibody therapy; therefore, such unwanted drug interactions need to be eliminated. With these goals in mind, we created a cancer ADCC model and describe here a simple protocol to find ADCC-modulating drugs. Since 3D models such as cancer cell spheroids are superior to 2D cultures in predicting in vivo responses of tumors to anticancer therapies, spheroid co-cultures of EGFP-expressing HER2+ JIMT-1 breast cancer cells and the NK92.CD16 cell lines were set up and induced with Trastuzumab, a monoclonal antibody clinically approved against HER2-positive breast cancer. JIMT-1 spheroids were allowed to form in cell-repellent U-bottom 96-well plates. On day 3, NK cells and Trastuzumab were added. The spheroids were then stained with Annexin V-Alexa 647 to measure apoptotic cell death, which was quantitated in the peripheral zone of the spheroids with an automated microscope. The applicability of our assay to identify ADCC-modulating molecules is demonstrated by showing that Sunitinib, a receptor tyrosine kinase inhibitor approved by the FDA against metastatic cancer, almost completely abolishes ADCC. The generation of the spheroids and image acquisition and analysis pipelines are compatible with high-throughput screening for ADCC-modulating compounds in cancer cell spheroids.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Esferoides Celulares , Humanos , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/imunologia , Descoberta de Drogas/métodos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/efeitos dos fármacos , Linhagem Celular Tumoral , Receptores de IgG/imunologia , Antineoplásicos Imunológicos/farmacologia , Trastuzumab/farmacologia
9.
Bioconjug Chem ; 35(6): 855-866, 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38789102

RESUMO

Antibody effector functions including antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) are mediated through the interaction of the antibody Fc region with Fcγ receptors present on immune cells. Several approaches have been used to modulate antibody Fc-Fcγ interactions with the goal of driving an effective antitumor immune response, including Fc point mutations and glycan modifications. However, robust antibody-Fcγ engagement and immune cell binding of Fc-enhanced antibodies in the periphery can lead to the unwanted induction of systemic cytokine release and other dose-limiting infusion-related reactions. Creating a balance between effective engagement of Fcγ receptors that can induce antitumor activity without incurring systemic immune activation is an ongoing challenge in the field of antibody and immuno-oncology therapeutics. Herein, we describe a method for the reversible chemical modulation of antibody-Fcγ interactions using simple poly(ethylene glycol) (PEG) linkers conjugated to antibody interchain disulfides with maleimide attachments. This method enables dosing of a therapeutic with muted Fcγ engagement that is restored in vivo in a time-dependent manner. The technology was applied to an effector function enhanced agonist CD40 antibody, SEA-CD40, and experiments demonstrate significant reductions in Fc-induced immune activation in vitro and in mice and nonhuman primates despite showing retained efficacy and improved pharmacokinetics compared to the parent antibody. We foresee that this simple, modular system can be rapidly applied to antibodies that suffer from systemic immune activation due to peripheral FcγR binding immediately upon infusion.


Assuntos
Receptores de IgG , Animais , Camundongos , Receptores de IgG/imunologia , Humanos , Polietilenoglicóis/química , Citotoxicidade Celular Dependente de Anticorpos , Fagocitose/efeitos dos fármacos
10.
Clin Immunol ; 263: 110231, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38692449

RESUMO

Moderate-to-severe systemic lupus erythematosus (SLE) is characterized by extensive autoantibody deposition and persistent autoinflammation. As the existing animal models are limited in accurately reproducing the pathological characteristics of human SLE, we introduced a novel animal model simulating multi-organ autoinflammation through intra-organ injections. The model closely mimicked key features of SLE, including IgG deposition, inflammation, and tissue damage. The model could be used to assess the roles of IgG, immune cells, cytokines, and Fc gamma receptor (FcγR) in the pathogenesis of autoinflammation. The results obtained from this model could be confirmed by lupus MRL/lpr mice. The review suggested that the diagnostic criteria should be reconsidered to incorporate IgG deposition in tissues and highlighted the limitations of current T-cell and B-cell-focused treatments. To summarize, the IgG deposition model can be used to investigate the pathogenesis and treatment of multi-organ tissue damage associated with SLE.


Assuntos
Modelos Animais de Doenças , Imunoglobulina G , Lúpus Eritematoso Sistêmico , Animais , Lúpus Eritematoso Sistêmico/imunologia , Lúpus Eritematoso Sistêmico/patologia , Camundongos , Imunoglobulina G/imunologia , Humanos , Camundongos Endogâmicos MRL lpr , Inflamação/imunologia , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Linfócitos B/imunologia
11.
Front Immunol ; 15: 1341013, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38655263

RESUMO

Recombinant Factor VIII-Fc fusion protein (rFVIIIFc) is an enhanced half-life therapeutic protein product used for the management of hemophilia A. Recent studies have demonstrated that rFVIIIFc interacts with Fc gamma receptors (FcγR) resulting in the activation or inhibition of various FcγR-expressing immune cells. We previously demonstrated that rFVIIIFc, unlike recombinant Factor IX-Fc (rFIXFc), activates natural killer (NK) cells via Fc-mediated interactions with FcγRIIIA (CD16). Additionally, we showed that rFVIIIFc activated CD16+ NK cells to lyse a FVIII-specific B cell clone. Here, we used human NK cell lines and primary NK cells enriched from peripheral blood leukocytes to study the role of the FVIII moiety in rFVIIIFc-mediated NK cell activation. Following overnight incubation of NK cells with rFVIIIFc, cellular activation was assessed by measuring secretion of the inflammatory cytokine IFNγ by ELISA or by cellular degranulation. We show that anti-FVIII, anti-Fc, and anti-CD16 all inhibited indicating that these molecules were involved in rFVIIIFc-mediated NK cell activation. To define which domains of FVIII were involved, we used antibodies that are FVIII domain-specific and demonstrated that blocking FVIII C1 or C2 domain-mediated membrane binding potently inhibited rFVIIIFc-mediated CD16+ NK cell activation, while targeting the FVIII heavy chain domains did not. We also show that rFVIIIFc binds CD16 with about five-fold higher affinity than rFIXFc. Based on our results we propose that FVIII light chain-mediated membrane binding results in tethering of the fusion protein to the cell surface, and this, together with increased binding affinity for CD16, allows for Fc-CD16 interactions to proceed, resulting in NK cellular activation. Our working model may explain our previous results where we observed that rFVIIIFc activated NK cells via CD16, whereas rFIXFc did not despite having identical IgG1 Fc domains.


Assuntos
Fator VIII , Proteínas Ligadas por GPI , Fragmentos Fc das Imunoglobulinas , Células Matadoras Naturais , Ativação Linfocitária , Receptores de IgG , Proteínas Recombinantes de Fusão , Humanos , Degranulação Celular/imunologia , Fator VIII/química , Fator VIII/imunologia , Proteínas Ligadas por GPI/imunologia , Proteínas Ligadas por GPI/metabolismo , Hemofilia A/imunologia , Hemofilia A/tratamento farmacológico , Fragmentos Fc das Imunoglobulinas/imunologia , Interferon gama/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/imunologia , Ativação Linfocitária/efeitos dos fármacos , Ligação Proteica , Receptores de IgG/metabolismo , Receptores de IgG/imunologia
12.
Vox Sang ; 119(7): 712-719, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38597364

RESUMO

BACKGROUND AND OBJECTIVES: The isolation of neutrophils and subsequent detection of anti-human neutrophil antigens (HNA) antibodies are crucial in clinical medicine for the diagnosis of autoimmune neutropenia, neonatal alloimmune neutropenia (NAIN) and transfusion-related acute lung injury (TRALI). This study reports two cases of maternal anti-Fc-gamma-receptor-IIIb (FcγRIIIb) isoimmunization without NAIN symptoms and compares the efficiency of immunomagnetic negative selection (IMNS) with traditional dextran/Ficoll for neutrophil isolation in HNA serological assays. MATERIALS AND METHODS: Investigating two cases of maternal anti-FcγRIIIb isoimmunization, neutrophils from three donors were isolated from 8 mL of whole blood using IMNS and dextran/Ficoll. Serological assays included the granulocyte agglutination and immunofluorescence test, monoclonal antibody immobilization of granulocyte antigens and the LABScreen Multi (One Lambda). IMNS and dextran/Ficoll were compared in terms of cell yield, viability, time, cost and purity. RESULTS: Maternal anti-FcγRIIIb isoantibodies with FCGR3B gene deletion were detected in both cases. Newborns and fathers exhibited specific gene combinations: FCGR3B*02/FCGR3B*02 (Case 1) and FCGR3B*02/FCGR3B*03 (Case 2). IMNS outperformed dextran/Ficoll, yielding four times more neutrophils (average neutrophil counts: 18.5 × 103/µL vs. 4.5 × 103/µL), efficiently removing non-neutrophil cells and reducing processing time (30-40 min vs. 70-90 min), although it incurred a higher cost (2.7 times). CONCLUSION: Two cases of maternal anti-FcγRIIIb isoantibodies, unrelated to NAIN, were identified. Although neutropenia has not been described in these cases, we emphasize the importance of identifying asymptomatic cases with the potential for severe neutropenia. Additionally, IMNS is introduced as a rapid, high-yield, high-purity neutrophil isolation technique, beneficial for serological assays detecting anti-HNA antibodies.


Assuntos
Isoanticorpos , Neutrófilos , Receptores de IgG , Humanos , Neutrófilos/imunologia , Feminino , Receptores de IgG/imunologia , Isoanticorpos/imunologia , Isoanticorpos/sangue , Recém-Nascido , Proteínas Ligadas por GPI/imunologia , Masculino , Separação Imunomagnética/métodos , Adulto , Gravidez , Neutropenia/imunologia , Neutropenia/sangue
13.
Clin Immunol ; 263: 110206, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38599263

RESUMO

Patients suffering from sepsis-induced acute lung injury (ALI) exhibit a high mortality rate, and their prognosis is closely associated with infiltration of neutrophils into the lungs. In this study, we found a significant elevation of CD64+ neutrophils, which highly expressed p75 neurotrophin receptor (p75NTR) in peripheral blood of mice and patients with sepsis-induced ALI. p75NTR+CD64+ neutrophils were also abundantly expressed in the lung of ALI mice induced by lipopolysaccharide. Conditional knock-out of the myeloid lineage's p75NTR gene improved the survival rates, attenuated lung tissue inflammation, reduced neutrophil infiltration and enhanced the phagocytic functions of CD64+ neutrophils. In vitro, p75NTR+CD64+ neutrophils exhibited an upregulation and compromised phagocytic activity in blood samples of ALI patients. Blocking p75NTR activity by soluble p75NTR extracellular domain peptide (p75ECD-Fc) boosted CD64+ neutrophils phagocytic activity and reduced inflammatory cytokine production via regulation of the NF-κB activity. The findings strongly indicate that p75NTR+CD64+ neutrophils are a novel pathogenic neutrophil subpopulation promoting sepsis-induced ALI.


Assuntos
Lesão Pulmonar Aguda , Camundongos Endogâmicos C57BL , Neutrófilos , Fagocitose , Receptores de IgG , Receptores de Fator de Crescimento Neural , Sepse , Animais , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/etiologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Sepse/imunologia , Sepse/complicações , Humanos , Receptores de IgG/metabolismo , Receptores de IgG/genética , Receptores de IgG/imunologia , Camundongos , Masculino , Fagocitose/imunologia , Receptores de Fator de Crescimento Neural/metabolismo , Receptores de Fator de Crescimento Neural/genética , Receptores de Fator de Crescimento Neural/imunologia , Camundongos Knockout , Lipopolissacarídeos , Citocinas/metabolismo , Citocinas/imunologia , Pulmão/imunologia , Pulmão/patologia , Feminino , NF-kappa B/metabolismo , NF-kappa B/imunologia , Proteínas do Tecido Nervoso
14.
Clin Immunol ; 263: 110223, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38636890

RESUMO

Idiopathic severe aplastic anemia (SAA) is a disease of bone marrow failure caused by T-cell-induced destruction of hematopoietic stem and progenitor cells (HSPCs), however the mechanism remains unclear. We performed single-cell RNA sequencing of PBMCs and BMMCs from SAA patients and healthy donors and identified a CD8+ T cell subset with a tissue residency phenotype (Trm) in bone marrow that exhibit high IFN-γ and FasL expression and have a higher ability to induce apoptosis in HSPCs in vitro through FasL expression. CD8+ Trm cells were induced by IL-15 presented by IL-15Rα on monocytes, especially CD16+ monocytes, which were increased in SAA patients. CD16+ monocytes contributed to IL-15-induced CD38+CXCR6+ pre-Trm differentiation into CD8+ Trm cells, which can be inhibited by the CD38 inhibitor 78c. Our results demonstrate that IL-15-induced CD8+ Trm cells are pathogenic cells that mediate HSPC destruction in SAA patients and are therapeutic targets for future treatments.


Assuntos
Anemia Aplástica , Linfócitos T CD8-Positivos , Proteínas Ligadas por GPI , Células-Tronco Hematopoéticas , Interleucina-15 , Monócitos , Receptores de IgG , Humanos , Anemia Aplástica/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Interleucina-15/farmacologia , Interleucina-15/imunologia , Receptores de IgG/metabolismo , Receptores de IgG/imunologia , Monócitos/imunologia , Monócitos/efeitos dos fármacos , Feminino , Masculino , Adulto , Células-Tronco Hematopoéticas/imunologia , Proteínas Ligadas por GPI/metabolismo , Proteínas Ligadas por GPI/imunologia , Pessoa de Meia-Idade , Proteína Ligante Fas/metabolismo , Proteína Ligante Fas/imunologia , Adulto Jovem , Adolescente , Interferon gama/imunologia , Interferon gama/metabolismo , Receptores de Interleucina-15/metabolismo , Receptores de Interleucina-15/imunologia , Apoptose/efeitos dos fármacos , Diferenciação Celular/imunologia
15.
Viruses ; 16(4)2024 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-38675937

RESUMO

Antibodies that specifically bind to individual human fragment crystallizable γ receptors (FcγRs) are of interest as research tools in studying immune cell functions, as well as components in bispecific antibodies for immune cell engagement in cancer therapy. Monoclonal antibodies for human low-affinity FcγRs have been successfully generated by hybridoma technology and are widely used in pre-clinical research. However, the generation of monoclonal antibodies by hybridoma technology that specifically bind to the high-affinity receptor FcγRI is challenging. Monomeric mouse IgG2a, IgG2b, and IgG3 bind human FcγRI with high affinity via the Fc part, leading to an Fc-mediated rather than a fragment for antigen binding (Fab)-mediated selection of monoclonal antibodies. Blocking the Fc-binding site of FcγRI with an excess of human IgG or Fc during screening decreases the risk of Fc-mediated interactions but can also block the potential epitopes of new antibody candidates. Therefore, we replaced hybridoma technology with phage display of a single-chain fragment variable (scFv) antibody library that was generated from mice immunized with FcγRI-positive cells and screened it with a cellular panning approach assisted by next-generation sequencing (NGS). Seven new FcγRI-specific antibody sequences were selected with this methodology, which were produced as Fc-silent antibodies showing FcγRI-restricted specificity.


Assuntos
Anticorpos Monoclonais , Receptores de IgG , Receptores de IgG/imunologia , Receptores de IgG/metabolismo , Animais , Camundongos , Humanos , Anticorpos Monoclonais/imunologia , Imunoglobulina G/imunologia , Imunização , Anticorpos de Cadeia Única/imunologia , Anticorpos de Cadeia Única/genética , Biblioteca de Peptídeos , Técnicas de Visualização da Superfície Celular , Hibridomas , Especificidade de Anticorpos , Feminino , Camundongos Endogâmicos BALB C
16.
Front Immunol ; 15: 1360615, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646521

RESUMO

Introduction: Malignant ascites indicates ovarian cancer progression and predicts poor clinical outcome. Various ascites components induce an immunosuppressive crosstalk between tumor and immune cells, which is poorly understood. In our previous study, imbalanced electrolytes, particularly high sodium content in malignant ascites, have been identified as a main immunosuppressive mechanism that impaired NK and T-cell activity. Methods: In the present study, we explored the role of high concentrations of ascites proteins and immunoglobulins on antitumoral NK effector functions. To this end, a coculture system consisting of healthy donor NK cells and ovarian cancer cells was used. The anti-EGFR antibody Cetuximab was added to induce antibody-dependent cellular cytotoxicity (ADCC). NK activity was assessed in the presence of different patient ascites samples and immunoglobulins that were isolated from ascites. Results: Overall high protein concentration in ascites impaired NK cell degranulation, conjugation to tumor cells, and intracellular calcium signaling. Immunoglobulins isolated from ascites samples competitively interfered with NK ADCC and inhibited the conjugation to target cells. Furthermore, downregulation of regulatory surface markers CD16 and DNAM-1 on NK cells was prevented by ascites-derived immunoglobulins during NK cell activation. Conclusion: Our data show that high protein concentrations in biological fluids are able to suppress antitumoral activity of NK cells independent from the mechanism mediated by imbalanced electrolytes. The competitive interference between immunoglobulins of ascites and specific therapeutic antibodies could diminish the efficacy of antibody-based therapies and should be considered in antibody-based immunotherapies.


Assuntos
Citotoxicidade Celular Dependente de Anticorpos , Ascite , Células Matadoras Naturais , Neoplasias Ovarianas , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ascite/imunologia , Feminino , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Neoplasias Ovarianas/imunologia , Neoplasias Ovarianas/metabolismo , Linhagem Celular Tumoral , Imunoglobulinas/metabolismo , Receptores de IgG/metabolismo , Receptores de IgG/imunologia , Degranulação Celular/imunologia , Degranulação Celular/efeitos dos fármacos , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígenos de Diferenciação de Linfócitos T/imunologia , Cetuximab/farmacologia
17.
J Immunol ; 212(10): 1564-1578, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38551350

RESUMO

HIV-1 infection greatly alters the NK cell phenotypic and functional repertoire. This is highlighted by the expansion of a rare population of FcRγ- NK cells exhibiting characteristics of traditional immunologic memory in people with HIV (PWH). Although current antiretroviral therapy (ART) effectively controls HIV-1 viremia and disease progression, its impact on HIV-1-associated NK cell abnormalities remains unclear. To address this, we performed a longitudinal analysis detailing conventional and memory-like NK cell characteristics in n = 60 PWH during the first 4 y of ART. Throughout this regimen, a skewed repertoire of cytokine unresponsive FcRγ- memory-like NK cells persisted and accompanied an overall increase in NK surface expression of CD57 and KLRG1, suggestive of progression toward immune senescence. These traits were linked to elevated serum inflammatory biomarkers and increasing Ab titers to human CMV, with human CMV viremia detected in approximately one-third of PWH at years 1-4 of ART. Interestingly, 40% of PWH displayed atypical NK cell subsets, representing intermediate stages of NK-poiesis based on single-cell multiomic trajectory analysis. Our findings indicate that NK cell irregularities persist in PWH despite long-term ART, underscoring the need to better understand the causative mechanisms that prevent full restoration of immune health in PWH.


Assuntos
Antígenos CD57 , Infecções por HIV , HIV-1 , Células Matadoras Naturais , Humanos , Células Matadoras Naturais/imunologia , Infecções por HIV/imunologia , Infecções por HIV/tratamento farmacológico , Infecções por HIV/virologia , HIV-1/imunologia , Masculino , Feminino , Antígenos CD57/imunologia , Adulto , Pessoa de Meia-Idade , Memória Imunológica/imunologia , Lectinas Tipo C/imunologia , Receptores Imunológicos , Viremia/imunologia , Viremia/tratamento farmacológico , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/tratamento farmacológico , Receptores de IgG/imunologia , Estudos Longitudinais , Antirretrovirais/uso terapêutico
18.
Expert Opin Pharmacother ; 25(3): 281-294, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38465524

RESUMO

INTRODUCTION: Fcγ-receptors (FcγR) are membrane receptors expressed on a variety of immune cells, specialized in recognition of the Fc part of immunoglobulin G (IgG) antibodies. FcγRIIA-dependent platelet activation in platelet factor 4 (PF4) antibody-related disorders have gained major attention, when these antibodies were identified as the cause of the adverse vaccination event termed vaccine-induced immune thrombocytopenia and thrombosis (VITT) during the COVID-19 vaccination campaign. With the recognition of anti-PF4 antibodies as cause for severe spontaneous and sometimes recurrent thromboses independent of vaccination, their clinical relevance extended far beyond heparin-induced thrombocytopenia (HIT) and VITT. AREAS COVERED: Patients developing these disorders show life-threatening thromboses, and the outcome is highly dependent on effective treatment. This narrative literature review summarizes treatment options for HIT and VITT that are currently available for clinical application and provides the perspective toward new developments. EXPERT OPINION: Nearly all these novel approaches are based on in vitro, preclinical observations, or case reports with only limited implementation in clinical practice. The therapeutic potential of these approaches still needs to be proven in larger cohort studies to ensure treatment efficacy and long-term patient safety.


Assuntos
Vacinas contra COVID-19 , Heparina , Receptores de IgG , Trombocitopenia , Trombose , Humanos , Anticoagulantes/efeitos adversos , COVID-19/complicações , COVID-19/prevenção & controle , COVID-19/imunologia , Vacinas contra COVID-19/efeitos adversos , Vacinas contra COVID-19/imunologia , Heparina/efeitos adversos , Ativação Plaquetária/efeitos dos fármacos , Fator Plaquetário 4/imunologia , Púrpura Trombocitopênica Idiopática/induzido quimicamente , Púrpura Trombocitopênica Idiopática/imunologia , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Receptores de IgG/metabolismo , Receptores de IgG/imunologia , Trombocitopenia/induzido quimicamente , Trombocitopenia/imunologia , Tromboinflamação/tratamento farmacológico , Trombose/tratamento farmacológico , Trombose/imunologia
19.
J Clin Invest ; 134(11)2024 Mar 26.
Artigo em Inglês | MEDLINE | ID: mdl-38530369

RESUMO

Antibodies can initiate lung injury in a variety of disease states such as autoimmunity, in reactions to transfusions, or after organ transplantation, but the key factors determining in vivo pathogenicity of injury-inducing antibodies are unclear. Harmful antibodies often activate the complement cascade. A model for how IgG antibodies trigger complement activation involves interactions between IgG Fc domains driving the assembly of IgG hexamer structures that activate C1 complexes. The importance of IgG hexamers in initiating injury responses was not clear, so we tested their relevance in a mouse model of alloantibody- and complement-mediated acute lung injury. We used 3 approaches to block alloantibody hexamerization (antibody carbamylation, the K439E Fc mutation, or treatment with domain B from staphylococcal protein A), all of which reduced acute lung injury. Conversely, Fc mutations promoting spontaneous hexamerization made a harmful alloantibody into a more potent inducer of acute lung injury and rendered an innocuous alloantibody pathogenic. Treatment with a recombinant Fc hexamer "decoy" therapeutic protected mice from lung injury, including in a model with transgenic human FCGR2A expression that exacerbated pathology. These results indicate an in vivo role of IgG hexamerization in initiating acute lung injury and the potential for therapeutics that inhibit or mimic hexamerization to treat antibody-mediated diseases.


Assuntos
Lesão Pulmonar Aguda , Imunoglobulina G , Receptores de IgG , Animais , Camundongos , Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/patologia , Imunoglobulina G/imunologia , Humanos , Receptores de IgG/imunologia , Receptores de IgG/genética , Receptores de IgG/metabolismo , Ativação do Complemento/imunologia , Camundongos Transgênicos , Isoanticorpos/imunologia , Mutação de Sentido Incorreto , Modelos Animais de Doenças , Substituição de Aminoácidos , Fragmentos Fc das Imunoglobulinas/imunologia , Fragmentos Fc das Imunoglobulinas/genética , Fragmentos Fc das Imunoglobulinas/metabolismo
20.
Cell Immunol ; 399-400: 104823, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38520831

RESUMO

AAV-mediated gene transfer is a promising platform still plagued by potential host-derived, antagonistic immune responses to therapeutic components. CpG-mediated TLR9 stimulation activates innate immune cells and leads to cognate T cell activation and suppression of transgene expression. Here, we demonstrate that CpG depletion increased expression of an antibody transgene product by 2-3-fold as early as 24 h post-vector administration in mice. No significant differences were noted in anti-transgene product/ anti-AAV capsid antibody production or cytotoxic gene induction. Instead, CpG depletion significantly reduced the presence of a pDC-like myeloid cell population, which was able to directly bind the antibody transgene product via Fc-FcγR interactions. Thus, we extend the mechanisms of TLR9-mediated antagonism of transgene expression in AAV gene therapy to include the actions of a previously unreported pDC-like cell population.


Assuntos
Células Dendríticas , Dependovirus , Terapia Genética , Vetores Genéticos , Camundongos Endogâmicos C57BL , Receptor Toll-Like 9 , Transgenes , Animais , Células Dendríticas/imunologia , Dependovirus/genética , Camundongos , Terapia Genética/métodos , Receptor Toll-Like 9/imunologia , Ilhas de CpG/genética , Ilhas de CpG/imunologia , Receptores de IgG/imunologia , Receptores de IgG/genética , Receptores de IgG/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...