Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
MAbs ; 16(1): 2406539, 2024.
Article in English | MEDLINE | ID: mdl-39306747

ABSTRACT

A critical attribute of therapeutic antibodies is their ability to engage with humoral or cellular effector mechanisms, and this depends on the ability of the Fc region to bind to complement (C1q) or Fc receptors. Investigators have sought to optimize these effects by engineering the Fc region to bind to a greater or lesser extent to individual receptors. Different approaches have been used in the clinic, but they have not been systematically compared. We have now produced a matched set of anti-CD20 antibodies representing a range of variants and compared their activity in cell-based assays for complement-dependent cytotoxicity, antibody-dependent cell-mediated cytotoxicity, and antibody-dependent phagocytosis using a range of individual Fc receptors. We have also compared the thermal stability of the variants by differential scanning fluorimetry (DSF). The results reveal a spectrum of activities which may be appropriate for different applications.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Immunoglobulin Fc Fragments , Receptors, IgG , Humans , Receptors, IgG/genetics , Receptors, IgG/metabolism , Receptors, IgG/immunology , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/chemistry , Mutation , Phagocytosis , Protein Binding , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology
2.
MAbs ; 16(1): 2402701, 2024.
Article in English | MEDLINE | ID: mdl-39279104

ABSTRACT

Elimination of the binding of immunoglobulin Fc to Fc gamma receptors is highly desirable for the avoidance of unwanted inflammatory responses to therapeutic antibodies and fusion proteins. Many different approaches have been used in the clinic, but they have not been systematically compared. We have now produced a matched set of anti-CD20 antibodies with different Fc subclasses and variants and compared their activity for binding to C1q, Fc-gamma receptors and in cell-based assays. Most of the variants still have significant levels of activity in one or more of these assays and many of them have impaired temperature stability compared with the corresponding wild-type antibody.


Subject(s)
Immunoglobulin Fc Fragments , Receptors, IgG , Receptors, IgG/genetics , Receptors, IgG/metabolism , Receptors, IgG/immunology , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism , Mutation , Protein Binding , Antigens, CD20/immunology , Antigens, CD20/genetics , Antigens, CD20/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/genetics
3.
ACS Appl Bio Mater ; 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39316382

ABSTRACT

Extracellular vesicles are ideal therapeutic potentiators for various diseases. However, they commonly lack targeting capability and are rapidly cleared by phagocytes. This requires appropriate administration at high doses, which can lead to toxic and adverse reactions. To overcome these limitations, we developed bleb nanovesicles containing human Fcγ receptor I (hCD64), known for their strong affinity to monomeric IgG. In this study, we focused on prostate cancer, which has a specific membrane antigen. We have utilized the hCD64-expressing bleb nanovesicles attaching anti-prostate-specific membrane antigen (PSMA) antibodies and confirmed their targeting ability in PSMA-related cell lines and prostate cancer xenograft models. Our findings underscore the promising potential of nanovesicle Fcγ receptor-IgG as a platform for cancer diagnosis and therapy systems, inspiring further research.

4.
Front Immunol ; 15: 1400177, 2024.
Article in English | MEDLINE | ID: mdl-38953027

ABSTRACT

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.


Subject(s)
Neoplasms , Receptors, IgG , T-Lymphocytes , Tumor Microenvironment , Receptors, IgG/immunology , Receptors, IgG/metabolism , Humans , Animals , Mice , Neoplasms/immunology , Neoplasms/therapy , Neoplasms/drug therapy , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Xenograft Model Antitumor Assays , Immunotherapy, Adoptive/methods , Receptor, ErbB-2/immunology , Receptor, ErbB-2/antagonists & inhibitors , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Female , Antigens, CD20/immunology
5.
Viruses ; 16(4)2024 04 12.
Article in English | MEDLINE | ID: mdl-38675937

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal , Receptors, IgG , Receptors, IgG/immunology , Receptors, IgG/metabolism , Animals , Mice , Humans , Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Immunization , Single-Chain Antibodies/immunology , Single-Chain Antibodies/genetics , Peptide Library , Cell Surface Display Techniques , Hybridomas , Antibody Specificity , Female , Mice, Inbred BALB C
6.
Front Immunol ; 14: 1213375, 2023.
Article in English | MEDLINE | ID: mdl-37622123

ABSTRACT

Therapeutic monoclonal antibodies (mAb) targeting the immune checkpoint inhibitor programmed cell death protein 1 (PD-1) have achieved considerable clinical success in anti-cancer therapy through relieving T cell exhaustion. Blockade of PD-1 interaction with its ligands PD-L1 and PD-L2 is an important determinant in promoting the functional recovery of exhausted T cells. Here, we show that anti-PD-1 mAbs act through an alternative mechanism leading to the downregulation of PD-1 surface expression on memory CD4+ and CD8+ T cells. PD-1 receptor downregulation is a distinct process from receptor endocytosis and occurs in a CD14+ monocyte dependent manner with the CD64/Fcγ receptor I acting as the primary factor for this T cell extrinsic process. Importantly, downregulation of surface PD-1 strongly enhances antigen-specific functional recovery of exhausted PD-1+CD8+ T cells. Our study demonstrates a novel mechanism for reducing cell surface levels of PD-1 and limiting the inhibitory targeting by PD-L1/2 and thereby enhancing the efficacy of anti-PD-1 Ab in restoring T cell functionality.


Subject(s)
CD8-Positive T-Lymphocytes , Receptors, IgG , B7-H1 Antigen , Cell Membrane , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use
7.
Vaccines (Basel) ; 11(4)2023 Apr 15.
Article in English | MEDLINE | ID: mdl-37112761

ABSTRACT

The global rollout of COVID-19 vaccines has played a critical role in reducing pandemic spread, disease severity, hospitalizations, and deaths. However, the first-generation vaccines failed to block severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and transmission, partially due to the limited induction of mucosal immunity, leading to the continuous emergence of variants of concern (VOC) and breakthrough infections. To meet the challenges from VOC, limited durability, and lack of mucosal immune response of first-generation vaccines, novel approaches are being investigated. Herein, we have discussed the current knowledge pertaining to natural and vaccine-induced immunity, and the role of the mucosal immune response in controlling SARS-CoV2 infection. We have also presented the current status of the novel approaches aimed at eliciting both mucosal and systemic immunity. Finally, we have presented a novel adjuvant-free approach to elicit effective mucosal immunity against SARS-CoV-2, which lacks the safety concerns associated with live-attenuated vaccine platforms.

8.
Front Immunol ; 14: 1100499, 2023.
Article in English | MEDLINE | ID: mdl-36814926

ABSTRACT

Understanding the molecular mechanism underlying the hierarchic binding between FcγRs and IgG antibodies is critical for therapeutic antibody engineering and FcγR functions. The recent determination of crystal structures of FcγRI-Fc complexes, however, resulted in two controversial mechanisms for the high affinity receptor binding to IgG. Here, we describe high resolution structures of a bovine FG-loop variant of FcγRI in complex with the Fc fragment of IgG1 crystallized in three different conditions at neutral pH, confirming the characteristic FG loop-Fc interaction is critical to the high affinity immunoglobulin binding. We showed that the FcγRI D2-domain FG-loop functioned as a pH-sensing switch for IgG binding. Further live cell imaging of FcγRI-mediated internalization of immune complexes showed a pH sensitive temporal-spatial antibody-antigen uptake and release. Taken together, we demonstrate that the structures of FcγRI-Fc crystallized at neutral and acidic pH, respectively, represent the high and low affinity binding states of the receptor for IgG uptake and release. These results support a role for FcγRI in antigen delivery, highlight the importance of Fc glycan in antibody binding to the high affinity receptor and provide new insights to future antibody engineering.


Subject(s)
Immunoglobulin G , Receptors, IgG , Animals , Cattle , Receptors, IgG/metabolism , Protein Binding , Phagocytosis , Hydrogen-Ion Concentration
9.
Front Immunol ; 13: 889286, 2022.
Article in English | MEDLINE | ID: mdl-35833115

ABSTRACT

Persistent arthritis pain after resolution of joint inflammation represents a huge health burden in patients with rheumatoid arthritis (RA). However, the underling mechanisms are poorly understood. We and other groups recently revealed that FcγRI, a key immune receptor, is functionally expressed in joint nociceptors. Thus, we investigated a potential role of sensory neuron expressed FcγRI in postinflammatory arthritis pain in a mouse model of collagen antibody-induced arthritis (CAIA). Here, we show that global deletion of Fcgr1 significantly attenuated mechanical hyperalgesia in the ankle and hind paw of female mice in both inflammatory and postinflammatory phases of CAIA. No obvious differences in cartilage destruction were observed after resolution of joint inflammation between genotypes. In situ hybridization (ISH) revealed that a larger proportion of dorsal root ganglion (DRG) neurons expressed Fcgr1 mRNA signal in the late phase of CAIA. Conditional deletion of Fcgr1 in primary sensory neurons produced similar analgesic effects without affecting joint swelling. Knockdown of Fcgr1 expression within DRG in the postinflammatory phase of CAIA alleviated persistent pain. Inflammation within DRG after resolution of joint inflammation in the CAIA model was evidenced by T cell and neutrophil infiltration and upregulated mRNA expression of numerous inflammatory mediators. Yet, such changes were not altered by genetic deletion of Fcgr1. We suggest that neuroinflammation within the DRG after resolution of joint inflammation might upregulate FcγRI signaling in DRG neurons. Sensory neuron expressed FcγRI thus merits exploration as a potential target for the treatment of arthritis pain that persists in RA patients in remission.


Subject(s)
Arthritis, Experimental , Arthritis, Rheumatoid , Receptors, IgG , Animals , Antibodies , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Female , Mice , Pain , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, IgG/genetics , Receptors, IgG/metabolism , Sensory Receptor Cells/metabolism
10.
Microorganisms ; 11(1)2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36677333

ABSTRACT

FcγRI is an important cell surface receptor reported to be involved in multiple immune responses, although it has not yet been extensively studied in intracellular bacterial infections. Here, using a mouse model of C. muridarum respiratory infection, we were able to determine how FcγRI regulates the host resistance against chlamydial invasion. According to our findings, the chlamydial loads and pulmonary pathology were both reduced in FcγRI deficient (Fcgr1-/-) animals. Being infected, monocytes, macrophages, neutrophils, DCs, CD4+/CD8+ T cells, and effector Th1 subsets displayed increased FcγRI expression patterns. Altered infiltration of these cells in the lungs of Fcgr1-/- mice further demonstrated the regulation of FcγRI in the immune system and identified Th1 cells and macrophages as its target cell populations. As expected, we observed that the Th1 response was augmented in Fcgr1-/- mice, while the pro-inflammatory M1 macrophage polarization was constrained. These findings might indicate FcγRI as a potential regulator for host immunity and inflammatory response during chlamydial infection.

11.
Eur J Immunol ; 51(2): 459-470, 2021 02.
Article in English | MEDLINE | ID: mdl-33078845

ABSTRACT

Ulcerative colitis (UC) is a disease that frequently relapses and affects more than 0.1% general population; the underlying mechanism is poorly understood. Published data show that polymorphonuclear neutrophils (PMN) contribute to the pathogenesis of UC. This study aims to identify antigen (Ag)-specific PMNs and investigate their role in UC relapse. In this study, the correlation between PMN activities and UC relapse was assessed in a group of UC patients. A UC mouse model was developed to expand the findings of UC patient study. The results showed that a positive correlation was detected between the high PMN activities and the food Ag-specific IgG amounts in colon biopsies of UC patients. UC patient-derived Ag-specific PMNs could be activated upon exposure to food specific Ag. The Ag/FcγRI complexes were detected on the surface of PMNs in UC patients. Re-exposure of sensitized PMNs to specific Ag triggered PMN activation and induced UC-like inflammation in the mouse colon. We conclude that FcγRI plays a critical role in UC relapse. Inhibition of FcγRI can efficiently inhibits experimental UC.


Subject(s)
Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Receptors, IgG/metabolism , Adult , Animals , Cells, Cultured , Colon/metabolism , Colon/pathology , Female , Humans , Immunoglobulin G/metabolism , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Neutrophil Activation/physiology , Neutrophils/metabolism , Neutrophils/pathology , Reactive Oxygen Species/metabolism , Recurrence
12.
Antibodies (Basel) ; 9(4)2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32977708

ABSTRACT

A variety of Fc domain engineering approaches for abrogating the effector functions of mAbs exists. To address some of the limitations of the current Fc domain silencing approaches, we are exploring a less commonly considered option which relies on the deletion of the hinge. Removal of the hinge domain in humanized IgG1 and IgG4 mAbs obliterates their ability to bind to activating human Fc gamma receptors I and IIIA, while leaving their ability to engage their target antigen intact. Deletion of the hinge also reduces binding to the Fc neonatal receptor, although Fc engineering allows partial recovery of affinity. Engineering of the CH3 domain, stabilizes hinge deleted IgG4s and prevents Fab arm exchange. The faster clearing properties together with the pacified Fc make modality of the hinge deleted mAb an appealing solution for therapeutic and diagnostic applications.

13.
Viruses ; 12(2)2020 02 08.
Article in English | MEDLINE | ID: mdl-32046249

ABSTRACT

Antibody-dependent enhancement (ADE) contributes to the pathogenesis of porcine reproductive and respiratory syndrome virus (PRRSV)-persistent infection. However, the mechanisms of PRRSV-ADE infection are still confusing. A clear understanding of the event upon virus infection by the ADE pathway has become crucial for developing efficient intervention of the PRRSV infection. In this study, an ADE assay showed that PRRSV-ADE infection in porcine alveolar macrophages (AMs) significantly decreased the production of interferon-α (IFN-α) and tumor necrosis factor-α (TNF-α), and significantly increased the production of interleukine-10 (IL-10). A gene knockdown assay based on small interfering RNA (siRNA) showed that both Fc gamma receptor I (FcγRI) and FcγRIII in porcine AMs were involved in PRRSV-ADE infection. An activation assay showed that specific activation of FcγRI or FcγRIII in porcine AMs during PRRSV infection not only significantly decreased the production of IFN-α and TNF-α, but also significantly increased the production of IL-10 and significantly facilitated PRRSV replication. In conclusion, our studies suggested that ADE downregulated the production of IFN-α and TNF-α in porcine AMs maybe via FcγRI and FcγRIII, thereby leading to enhanced PRRSV infection.


Subject(s)
Antibodies, Viral/immunology , Interferon-alpha/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , Porcine Reproductive and Respiratory Syndrome/immunology , Receptors, IgG/genetics , Tumor Necrosis Factor-alpha/immunology , Animals , Antibody-Dependent Enhancement , Cell Line , Cells, Cultured , Down-Regulation , Porcine respiratory and reproductive syndrome virus , Swine , Virus Replication
14.
Int Immunopharmacol ; 81: 106268, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32062072

ABSTRACT

Porcine activating Fc gamma receptors (FcγRI and FcγRIII) have been cloned and characterized for many years. However, their roles in interferon (IFN) antiviral immune response to porcine reproductive and respiratory syndrome virus (PRRSV) infection have not yet been investigated extensively. In this study, PRRSV infection assay showed that PRRSV increased significantly the transcription of IFN-ß, IFN-γ and IFN-λ1 in porcine alveolar macrophages (PAMs) in early infection and decreased significantly the transcription of IFN-ß, IFN-γ and IFN-λ1 in PAMs in late infection. Activation assay showed that specific activation of FcγRI or FcγRIII in PAMs decreased significantly the transcription of IFN-ß, IFN-γ and IFN-λ1 and increased significantly the transcription of transforming growth factor ß1 (TGF-ß1). PRRSV infection assay mediated by FcγRI and FcγRIII showed that specific activation of FcγRI or FcγRIII in PAMs during PRRSV infection decreased significantly the transcription of IFN-ß, IFN-γ and IFN-λ1, but increased significantly the transcription of TGF-ß1 and enhanced significantly viral replication. In conclusion, our studies suggested that activating FcγR signaling inhibited the transcriptional levels of IFN-ß, IFN-γ and IFN-λ1 in PAMs in response to PRRSV infection.


Subject(s)
Interferon-beta/metabolism , Interferon-gamma/metabolism , Interferons/metabolism , Macrophages, Alveolar/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/physiology , Receptors, IgG/metabolism , Animals , Cells, Cultured , Down-Regulation , Immune Evasion , Macrophages, Alveolar/virology , Signal Transduction , Swine , Transforming Growth Factor beta1/metabolism , Virus Replication
15.
Mol Brain ; 12(1): 83, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31640731

ABSTRACT

The Fc gamma receptor I (FcγRI; CD64) is the high-affinity receptor of the immunoglobulin G protein (IgG). It is usually expressed in immune cells and has recently been identified to distribute in the nervous system and play critical roles in various neurological disorders. Presently, the impacts of FcγRI in neuropathic pain was largely unknown. Here, we aimed to investigate the impacts of FcγRI in neuropathic pain through pain-related neurobehavioral studies and underlying mechanisms by biochemical methods in animal and cell models. Specifically, we first utilized the chronic constriction injury (CCI) rat model that displayed neuropathic pain related symptoms and signs, including thermal hyperalgesia and mechanical allodynia. These neurobehavioral defects were significantly attenuated by the anti-FcγRI antibody, which was associated with reduced levels of neuropeptide substance P, C3, and TNF-α. Furthermore, we validated our animal findings using the embryonically neural crest-originated PC12 cell model. We found that stimulation of the IgG immune complex led to increased levels of FcγRI and inflammatory mediators, which were attenuated by the anti-FcγRI antibody in these cells. Collectively, our results from animal and cell-based studies suggest that FcγRI is a critical player for peripheral nerve injury-induced neuropathic pain by mediating pain-related immunological events, which therefore may provide a new therapeutic target for protection against chronic pain.


Subject(s)
Neuralgia/etiology , Neuralgia/metabolism , Peripheral Nerve Injuries/complications , Receptors, IgG/metabolism , Animals , PC12 Cells , Rats , Rats, Sprague-Dawley
16.
J Nucl Med ; 60(8): 1174-1182, 2019 08.
Article in English | MEDLINE | ID: mdl-30733320

ABSTRACT

Antibodies are promising vectors for PET imaging. However, the high uptake of radioimmunoconjugates in nontarget tissues such as the liver and spleen hampers their performance as radiotracers. This off-target uptake can lead to suboptimal tumor-to-background activity concentration ratios, decreasing the contrast of images and reducing their diagnostic utility. A possible cause of this uptake is the sequestration of radioimmunoconjugates by immune cells bearing Fc-γ-receptors (FcγR) that bind to the Fc regions of antibodies. Methods: Since the heavy chain glycans influence the affinity of FcγR for the Fc domain, we set out to investigate whether radioimmunoconjugates with truncated glycans would exhibit altered binding to FcγRI and, in turn, improved in vivo performance. Using the HER2-targeting antibody trastuzumab, we synthesized a series of desferrioxamine-bearing immunoconjugates with differing glycosylation states and interrogated their FcγRI binding via surface plasmon resonance, enzyme-linked immunosorbent assay, and flow cytometry. Furthermore, we labeled these immunoconjugates with 89Zr and explored their biodistribution in athymic nude, NSG, and humanized NSG mice bearing human epidermal growth factor receptor 2-expressing human breast cancer xenografts. Results: We observed a strong correlation between the impaired in vitro FcγRI binding of deglycosylated immunoconjugates and significant decreases in the in vivo off-target uptake of the corresponding 89Zr-labeled radioimmunoconjugates (i.e., liver activity concentrations are reduced by ∼3.5-fold in humanized NSG mice). These reductions in off-target uptake were accompanied by concomitant increases in the tumoral activity concentrations of the glycoengineered radioimmunoconjugates, ultimately yielding improved tumor-to-healthy organ contrast and higher quality PET images. Conclusion: Our findings suggest that the deglycosylation of antibodies represents a facile strategy for improving the quality of immuno-PET in animal models as well as in certain patient populations.


Subject(s)
Immunoconjugates/chemistry , Positron-Emission Tomography , Receptors, IgG/chemistry , Animals , Breast Neoplasms/diagnostic imaging , Cell Line, Tumor , Deferoxamine/chemistry , Female , Glycosylation , Humans , Immunoglobulin G/chemistry , In Vitro Techniques , Kinetics , Mice , Mice, Nude , Neoplasm Transplantation , Radiopharmaceuticals , Receptor, ErbB-2/metabolism , Recombinant Proteins/chemistry , Surface Plasmon Resonance , Tissue Distribution , Trastuzumab/chemistry , Zirconium/chemistry
17.
Dev Comp Immunol ; 90: 186-198, 2019 01.
Article in English | MEDLINE | ID: mdl-30273630

ABSTRACT

Receptors for the Fc region of IgG (FcγRs) play a key role in protecting the immune system and host from infection. In this study, we described the cloning, sequencing and characterization of porcine FcγRI, and reported six different FcγRI isoforms, four of which have never been reported before. Further analysis revealed that FcγR isoforms are generated by alternative splicing mechanisms, including two membrane isoforms and four soluble isoforms. Importantly, we found FcγRI splice variants differentially influence PRRSV antibody-dependent enhancement (ADE) effects. Membrane pCD64-T1 promotes endocytosis of the PRRSV-antibody complex to enhance PRRSV replication, and soluble pCD64-T3 has no ADE effect on PRRSV proliferation, but shows an inflammation enhancement effect. The differential expression of selective splicing in primary PAM cells and 3D4/21 cell lines are altered and regulated by PRRSV infection and inflammatory environment. Our results indicated that porcine FcγRI plays dual regulatory roles in PRRSV multiplication and PRRSV inflammation process by the alternatively spliced mechanism, which will be a new target in PRRSV prevention and control.


Subject(s)
Antibodies, Viral/metabolism , Inflammation/immunology , Macrophages, Alveolar/immunology , Porcine Reproductive and Respiratory Syndrome/immunology , Porcine respiratory and reproductive syndrome virus/physiology , Receptors, IgG/genetics , Receptors, IgG/metabolism , Swine/immunology , Alternative Splicing , Animals , Antibody-Dependent Enhancement , Cell Line , Cloning, Molecular , Cytokines/metabolism , Inflammation Mediators/metabolism , Macrophages, Alveolar/virology , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Splicing , Virus Replication
18.
J Pharmacol Sci ; 137(4): 342-349, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30190171

ABSTRACT

Lupus nephritis, one of the most serious complications of systemic lupus erythematosus (SLE), has been confirmed in a large number of clinical surveys. Current studies have suggested that inflammatory situation is generally considered to facilitate the occurrence and development of lupus nephritis. Previous research found that Fcγ receptor I (FcγRI) was compulsory for several autoimmune and inflammatory diseases, and it might be involved in the treatment of lupus nephritis. Furthermore, the possible molecular mechanism of the role of FcγRI in lupus nephritis still needs a further study. In the present study, in order to evaluate the effect of FcγRI on kidney function in lupus-prone MLR/lpr mice, FcγRI knockdown was implemented utilizing FcγRI-RNAi lentivirus. We reported that the administration of FcγRI-RNAi lentivirus (1) mainly inhibited FcγRI expression on macrophage of the kidneys, lowered the levels of urinary protein and serum anti-dsDNA antibody and prevented the impairment of renal function; (2) reduced the renal inflammatory cytokines (IL-1ß and IL-18); (3) decreased NF-κB p65 nuclear migration, suppressed NOD-like receptor protein 3 (NLRP3) inflammasome activation, and finally inhibited renal inflammation. Together, these results showed the role of FcγRI on macrophages to involve in renal inflammatory response, potentially via regulating the NLRP3 inflammasome-associated signaling.


Subject(s)
Gene Knockdown Techniques/methods , Genetic Therapy/methods , Genetic Vectors , Inflammasomes/genetics , Lentivirus/genetics , Lupus Nephritis/genetics , Lupus Nephritis/therapy , NF-kappa B/genetics , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , RNA Interference , Receptors, IgG/genetics , Signal Transduction/genetics , Animals , Female , Inflammasomes/metabolism , Macrophages/metabolism , Mice, Transgenic , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
19.
Life Sci ; 207: 296-303, 2018 Aug 15.
Article in English | MEDLINE | ID: mdl-29920250

ABSTRACT

AIMS: Fcγ receptor I (FcγRI/CD64) that is restrictedly expressed on monocytes and macrophages, acts as the single high-affinity receptor of immunoglobulin G (IgG) in human. The expression of FcγRI is positively correlated with immune inflammation. The primary aim of this study was to explore the effects of FcγRI expression on immune-related inflammatory response and investigate the potential mechanisms. MAIN METHODS: FcγRI-expressing Ba/F3 cells are the ideal models for evaluating the functions of FcγRI. Nuclear factor kappa B (NF-κB) and NOD-like receptor protein 3 (NLRP3) inflammasome-associated protein expressions and inflammatory cytokine (IL-1ß and IL-18) release were detected in the presence or absence of NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC). Besides, the effects of FcγRI on the activation of the NLRP3 inflammasomes were also investigated in THP-1 macrophages deficient for FcγRI. KEY FINDINGS: FcγRI-expressing Ba/F3 cells appeared increased NLRP3 inflammasome formation and IL-1ß and IL-18 release via activating NF-κB signaling. Interestingly, this alteration could be reversed in THP-1 macrophages after FcγRI was silenced. SIGNIFICANCE: These results indicated that FcγRI functioned as a regulator for immune inflammation via acceleration of NF-κB regulating NLRP3 inflammasome signaling.


Subject(s)
Inflammasomes/metabolism , NF-kappa B p50 Subunit/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, IgG/metabolism , Animals , Biomarkers/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation , Gene Silencing , Humans , Inflammation , Interleukin-18/metabolism , Interleukin-1beta/metabolism , Interleukin-3/metabolism , Macrophages/metabolism , Mice , Risk Factors , Signal Transduction , THP-1 Cells
20.
Cancer Immunol Immunother ; 67(7): 1079-1090, 2018 07.
Article in English | MEDLINE | ID: mdl-29687231

ABSTRACT

Antibodies targeting PD-1 have been demonstrated durable anti-cancer activity in certain cancer types. However, the anti-PD-1 antibodies are less or not efficacious in many situations, which might be attributed to co-expression of multiple inhibitory receptors or presence of immunosuppressive cells in the tumor microenvironment. Most of the anti-PD-1 antibodies used in clinical studies are of IgG4 isotype with the S228P mutation (IgG4S228P). The functional impact by the interaction of anti-PD-1 IgG4S228P antibody with Fc gamma receptors (FcγRs) is poorly understood. To assess the effects, we generated a pair of anti-PD-1 antibodies: BGB-A317/IgG4S228P and BGB-A317/IgG4-variant (abbreviated as BGB-A317), with the same variable regions but two different IgG4 Fc-hinge sequences. There was no significant difference between these two antibodies in binding to PD-1. However, BGB-A317/IgG4S228P binds to human FcγRI with high affinity and mediates crosslinking between PD-1 and FcγRI. In contrast, BGB-A317 does neither. Further cell-based assays showed that such crosslinking could reverse the function of an anti-PD-1 antibody from blocking to activating. More importantly, the crosslinking induces FcγRI+ macrophages to phagocytose PD-1+ T cells. In a mouse model transplanted with allogeneic human cancer cells and PBMCs, BGB-A317 showed significant tumor growth inhibition, whereas BGB-A317/IgG4S228P had no such inhibition. Immunohistochemistry study revealed an inverse correlation between FcγRI+ murine macrophage infiltration and the density of CD8+PD-1+ human T cells within tumors in the BGB-A317/IgG4S228P-treated group. These evidences suggested that FcγRI+ binding and crosslinking had negative impact on the anti-PD-1 antibody-mediated anti-cancer activity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Carcinoma, Squamous Cell/immunology , Immunoglobulin G/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, IgG/metabolism , Skin Neoplasms/immunology , Animals , Apoptosis , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Cell Proliferation , Humans , Immunoglobulin G/drug effects , Immunoglobulin G/metabolism , Lymphocyte Activation , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Programmed Cell Death 1 Receptor/immunology , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Cells, Cultured , Tumor Microenvironment , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL