Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (521)

Search Parameters:
Keywords = Fc receptors

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
41 pages, 4351 KB  
Review
Autoantibodies as Precision Tools in Connective Tissue Diseases: From Epiphenomenon to Endophenotype
by Muhammad Soyfoo and Julie Sarrand
Antibodies 2026, 15(1), 7; https://doi.org/10.3390/antib15010007 - 13 Jan 2026
Viewed by 69
Abstract
Autoantibodies have long been regarded as passive reflections of immune dysregulation in connective tissue diseases (CTDs). Recent advances in systems immunology and molecular pathology have fundamentally redefined them as active molecular fingerprints that delineate distinct disease endophenotypes with predictive power for clinical trajectories [...] Read more.
Autoantibodies have long been regarded as passive reflections of immune dysregulation in connective tissue diseases (CTDs). Recent advances in systems immunology and molecular pathology have fundamentally redefined them as active molecular fingerprints that delineate distinct disease endophenotypes with predictive power for clinical trajectories and therapeutic responses. Rather than mere epiphenomena, autoantibodies encode precise information about dominant immune pathways, organ tropism, and pathogenic mechanisms. This review synthesizes emerging evidence that autoantibody repertoires—defined by specificity, structural properties, and functional characteristics—stratify patients beyond traditional clinical taxonomy into discrete pathobiological subsets. Specific signatures such as anti-MDA5 in rapidly progressive interstitial lung disease, anti-RNA polymerase III in scleroderma renal crisis, and anti-Ro52/TRIM21 in systemic overlap syndromes illustrate how serological profiles predict outcomes with remarkable precision. Mechanistically, autoantibody pathogenicity is modulated by immunoglobulin isotype distribution, Fc glycosylation patterns, and tissue-specific receptor expression—variables that determine whether an antibody functions as a biomarker or pathogenic effector. The structural heterogeneity of autoantibodies, shaped by cytokine microenvironments and B-cell subset imprinting, creates a dynamic continuum between pro-inflammatory and regulatory states. The integration of serological, transcriptomic, and imaging data establishes a precision medicine framework: autoantibodies function simultaneously as disease classifiers and therapeutic guides. This endophenotype-driven approach is already influencing trial design and patient stratification in systemic lupus erythematosus, systemic sclerosis, and inflammatory myopathies, and is reshaping both clinical practice and scientific taxonomy in CTDs. Recognizing autoantibodies as endophenotypic determinants aligns disease classification with pathogenic mechanism and supports the transition towards immunologically informed therapeutic strategies. Full article
(This article belongs to the Special Issue Antibody and Autoantibody Specificities in Autoimmunity)
Show Figures

Graphical abstract

25 pages, 2792 KB  
Review
B-Cells and Plasmablasts as Architects of Autoimmune Disease: From Molecular Footprints to Precision Therapeutics
by Julie Sarrand and Muhammad Soyfoo
Cells 2026, 15(2), 119; https://doi.org/10.3390/cells15020119 - 9 Jan 2026
Viewed by 342
Abstract
B-cells and plasmablasts have emerged as central organizers of autoimmune pathogenesis, extending far beyond their classical role as antibody-producing cells to orchestrate immune circuits, tissue microenvironments, and therapeutic trajectories. Advances in single-cell technologies, high-dimensional cytometry, and B-cell receptor sequencing have uncovered a dynamic [...] Read more.
B-cells and plasmablasts have emerged as central organizers of autoimmune pathogenesis, extending far beyond their classical role as antibody-producing cells to orchestrate immune circuits, tissue microenvironments, and therapeutic trajectories. Advances in single-cell technologies, high-dimensional cytometry, and B-cell receptor sequencing have uncovered a dynamic continuum of B-cell differentiation programs that drive clinical heterogeneity across systemic autoimmune diseases. Plasmablasts, in particular, have gained recognition as highly responsive sensors of immune activation: they expand during flares, encode interferon-driven and extrafollicular responses, and correlate with disease severity. Autoantibody profiles, long viewed as static diagnostic signatures, are now understood as durable molecular footprints of distinct B-cell pathways. In this review, we propose an endotype-based framework integrating B-cell circuits with clinical phenotypes, illustrate therapeutic decision-making through mechanistic case vignettes, and outline future strategies combining immunomonitoring, multi-omics, and precision therapeutics. We further address translational challenges and discuss complementary approaches, including T-cell modulation, FcRn inhibition, and antigen-specific tolerization. Full article
Show Figures

Graphical abstract

21 pages, 8112 KB  
Article
Transcriptomic Profiling of Cutibacterium acnes IA1—Infected Keratinocytes Reveal Hub Genes and CLR Pathway in Acne Pathogenesis
by Jiawen Li, Fuxin Wang, Dangsheng Liu, Weichao Yang, Hao Sun, Mingfu Gao, Dawei Chen and Hui Xu
Curr. Issues Mol. Biol. 2026, 48(1), 34; https://doi.org/10.3390/cimb48010034 - 26 Dec 2025
Viewed by 356
Abstract
Acne vulgaris is a prevalent chronic inflammatory skin disorder affecting over 85% of adolescents. Emerging evidence indicates that Cutibacterium acnes phylotype IA1 contributes to acne initiation and progression, yet its precise mechanisms in epidermal keratinocytes remain unclear. This study investigated C. acnes [...] Read more.
Acne vulgaris is a prevalent chronic inflammatory skin disorder affecting over 85% of adolescents. Emerging evidence indicates that Cutibacterium acnes phylotype IA1 contributes to acne initiation and progression, yet its precise mechanisms in epidermal keratinocytes remain unclear. This study investigated C. acnes IA1’s effects on keratinocyte behavior using an in vitro HaCaT cell model. Cells were co-cultured with live C. acnes IA1 (CICC 10864) for 24 h. Transcriptomic profiling identified 769 differentially expressed genes (DEGs; adjusted p < 0.05, |log2FC| > 1), including 392 upregulated and 377 downregulated. The protein–protein interaction network analysis via Cytoscape revealed key hub genes (HNRNPA2B1, HNRNPM, RBM39). Enrichment analyses (GO, KEGG, Reactome, DO) highlighted significant involvement of the C-type lectin receptor (CLR) signaling pathway. Validation experiments showed cellular morphological changes, altered structure, and markedly elevated interleukin-6 (IL-6; p < 0.01), underscoring its role in inflammation. These findings suggest C. acnes IA1 drives acne pathogenesis by regulating hub genes that influence sebaceous gland inflammation, immune activity, and keratinocyte proliferation, positioning them as potential biomarkers for microbiome-targeted therapies. Limitations include the in vitro model’s lack of in vivo skin microenvironment complexity and use of only one representative IA1 strain. Full article
Show Figures

Figure 1

19 pages, 1028 KB  
Review
Venezuelan Equine Encephalitis Virus: Structural Biology, Vaccines, and Advances in Functional Antibodies
by Rui Tang, Daojing Wang, Guojiang Chen, Chenghua Liu, Liang Zhang, Fenghao Peng, Jijun Yu, Xinying Li, Heng Luo, Yan Wen and Chunxia Qiao
Vaccines 2026, 14(1), 23; https://doi.org/10.3390/vaccines14010023 - 24 Dec 2025
Viewed by 342
Abstract
Background: Venezuelan equine encephalitis virus (VEEV) poses a significant public health and biodefense threat due to periodic epidemics of severe neurological disease in the Americas, yet no licensed human vaccines or specific antiviral therapies exist. Methods: We comprehensively reviewed the current literature across [...] Read more.
Background: Venezuelan equine encephalitis virus (VEEV) poses a significant public health and biodefense threat due to periodic epidemics of severe neurological disease in the Americas, yet no licensed human vaccines or specific antiviral therapies exist. Methods: We comprehensively reviewed the current literature across three core domains: structural biology, vaccine development, and therapeutic antibodies. Results: First, we detail the complex viral structural proteome (including E1/E2 glycoproteins and Capsid), focusing on the LDLRAD3 entry receptor interaction. Second, we overview vaccine strategies, covering live-attenuated, VLP, and nucleic acid platforms designed to induce robust neutralizing antibodies. Finally, we examine the evolution of therapeutic antibodies, highlighting that optimal protection often relies on both neutralization and Fc effector functions, particularly for antibodies targeting the fusion loop or receptor-binding sites. Conclusions: Integrating structural insights with advanced antibody and vaccine engineering is essential for establishing effective clinical interventions capable of preventing future outbreaks and treating infected individuals. Full article
(This article belongs to the Section Pathogens-Host Immune Boundaries)
Show Figures

Figure 1

20 pages, 2114 KB  
Article
Does the Chimerization Process Affect the Immunochemical Properties of WNV-Neutralizing Antibody 900?
by Anastasiya A. Isaeva, Valentina S. Nesmeyanova, Daniil V. Shanshin, Nikita D. Ushkalenko, Ekaterina A. Volosnikova, Tatiana I. Esina, Elena V. Protopopova, Victor A. Svyatchenko, Valery B. Loktev, Sergey E. Olkin, Elena D. Danilenko, Elena I. Kazachinskaia and Dmitriy N. Shcherbakov
Int. J. Mol. Sci. 2025, 26(24), 12181; https://doi.org/10.3390/ijms262412181 - 18 Dec 2025
Viewed by 393
Abstract
West Nile fever is an infectious disease caused by the West Nile virus (WNV), which is transmitted by mosquitoes. Epidemiological surveillance confirms the potential risk of WNV infection in human populations. The lack of specific antiviral therapeutics and vaccines against WNV underscores the [...] Read more.
West Nile fever is an infectious disease caused by the West Nile virus (WNV), which is transmitted by mosquitoes. Epidemiological surveillance confirms the potential risk of WNV infection in human populations. The lack of specific antiviral therapeutics and vaccines against WNV underscores the urgent need to develop effective therapeutic approaches. In this study, a recombinant chimeric monoclonal antibody (mAb) 900 was generated based on the broadly neutralizing and protective murine mAb 9E2. The antigen-binding regions of the murine mAb were fused with the constant domains (CH2–CH3) of human IgG1. Two key amino acid clusters, M252/S254/T256 and H433/N434, were introduced into the CH2–CH3 domains to enhance the affinity of mAb 900 for the neonatal Fc receptor (FcRn). The engineered mAb 900 was produced in CHO cells and purified to high homogeneity. Biophysical characterization confirmed its stability and correct dimeric assembly. Comparative analysis demonstrated that mAb 900 retained the high antigen-binding affinity and potent virus-neutralizing activity of its murine predecessor. Most importantly, mAb 900 demonstrated significant protective efficacy in a lethal mouse model of WNV infection. These results establish the proof of concept for mAb 900 as a promising candidate for further preclinical development against WNV infection. Full article
Show Figures

Graphical abstract

21 pages, 7960 KB  
Article
Production of a Dulaglutide Analogue by Apoptosis-Resistant Chinese Hamster Ovary Cells in a 3-Week Fed-Batch Process
by Rolan R. Shaifutdinov, Maria V. Sinegubova, Ivan I. Vorobiev, Polina E. Prokhorova, Alexey B. Podkorytov and Nadezhda A. Orlova
Pharmaceuticals 2025, 18(12), 1896; https://doi.org/10.3390/ph18121896 - 16 Dec 2025
Viewed by 603
Abstract
Background: Dulaglutide, a GLP-1-IgG4 Fc fusion, is a long-acting GLP-1 receptor agonist used for type 2 diabetes therapy and other emerging indications. It is produced commercially in Chinese hamster ovary (CHO) cells. The supply of the original drug is now limited in some [...] Read more.
Background: Dulaglutide, a GLP-1-IgG4 Fc fusion, is a long-acting GLP-1 receptor agonist used for type 2 diabetes therapy and other emerging indications. It is produced commercially in Chinese hamster ovary (CHO) cells. The supply of the original drug is now limited in some regions, so creation of highly productive biosimilar manufacturing platforms is important. Methods: Two expression plasmids (p1.1-Tr2-Dul, p1.2-GS-Dul) encoding dulaglutide were sequentially transfected into apoptosis-resistant CHO 4BGD cells. Two-step transgene amplifications with methotrexate (MTX), followed by methionine sulfoximine (MSX) selection and subsequent cell cloning pipeline, were employed. Candidate clonal cell lines were selected using fed-batch culturing and long-term productivity testing. Results: Transfection with a second plasmid encoding glutamine synthetase (p1.2-GS-Dul) and selection with MSX resulted in a further ~30% increase titer in polyclonal population even after MTX-driven amplification. Top clone 4BGD/Dul #73 reached 1.05 g/L product titer in fed-batch culture (qP up to 22 pg·cell−1·day−1) and remained stable for 69 days in medium without MTX/MSX. Size exclusion-high-performance liquid chromatography showed ≥95% monomer; EC50 of the purified GLP-1-Fc in a GLP-1R/CRE-Luc assay was 52 pM for the obtained product versus 76 pM for the original reference drug. Conclusions: The sequential transfection and dual-marker selection approach enables the efficient generation of a robust, high-yield, and glutamine-independent CHO producer, representing a productive strategy suitable for industrial biosimilar development. Full article
(This article belongs to the Section Pharmaceutical Technology)
Show Figures

Figure 1

18 pages, 1528 KB  
Article
Detection of Microbial Contamination in Nanomaterials Using LAL, rFC and Cell-Based Assays: Implications for Nanotoxicological Hazard Assessment
by Peng Lei, Fikirte Debebe Zegeye, Mayes Alswady-Hoff, Chiara Marcolungo, Pernille Høgh Danielsen, Anne Mette Madsen, Håkan Wallin, Ulla Vogel, Shan Zienolddiny-Narui and Johanna Samulin Erdem
Nanomaterials 2025, 15(24), 1871; https://doi.org/10.3390/nano15241871 - 13 Dec 2025
Viewed by 355
Abstract
Accurate detection of microbial contamination is essential in the assessment of toxicological and immunological responses to various materials, as low-level contaminants can lead to confounding results. Traditional endotoxin testing relies on the Limulus Amebocyte Lysate (LAL) assay, which depends on horseshoe crab blood [...] Read more.
Accurate detection of microbial contamination is essential in the assessment of toxicological and immunological responses to various materials, as low-level contaminants can lead to confounding results. Traditional endotoxin testing relies on the Limulus Amebocyte Lysate (LAL) assay, which depends on horseshoe crab blood and raises both ecological and ethical concerns. Sustainable alternatives such as recombinant Factor C (rFC) provide a promising solution, yet validation for the detection of endotoxin in nanomaterials remains incomplete. In this study, we have used rFC alongside Toll-like receptor (TLR) reporter assays to detect both endotoxin and broader microbial contaminants in 31 nanomaterials from diverse classes. Special attention was given to assay interference by nanomaterials to ensure reliable detection. The rFC assay demonstrated a sensitive detection limit of 0.005 EU/mL, equivalent to the LAL assay, and showed that more than 50% of tested nanomaterials contained low-level endotoxin contamination. Additionally, several nanomaterials activated the TLR2 reporter, indicative of microbial contaminants beyond endotoxin. These results suggest that rFC can serve as a sustainable and reliable replacement for LAL in nanomaterial endotoxin testing but also emphasize the limitations of relying solely on endotoxin-specific assays. We recommend that future nanotoxicological evaluations integrate rFC with complementary methods, such as TLR-based approaches, and include thorough interference controls to ensure robust and comprehensive microbial contamination assessment. Full article
(This article belongs to the Special Issue Nanosafety Assessment, Implications and Mitigations)
Show Figures

Graphical abstract

13 pages, 851 KB  
Article
Cost-Effectiveness Analysis of Recombinant Tumor Necrosis Factor Receptor: Fc Fusion Protein as First-Line Treatment for Active Rheumatoid Arthritis in China
by Rui Zhang and Aixia Ma
Healthcare 2025, 13(24), 3267; https://doi.org/10.3390/healthcare13243267 - 12 Dec 2025
Viewed by 436
Abstract
Background/Objectives: To evaluate the cost-effectiveness of recombinant tumor necrosis factor receptor Fc fusion protein compared with methotrexate as first-line therapy for active rheumatoid arthritis in China using evidence from a Chinese head-to-head randomized trial. Methods: A Markov model with 6 months [...] Read more.
Background/Objectives: To evaluate the cost-effectiveness of recombinant tumor necrosis factor receptor Fc fusion protein compared with methotrexate as first-line therapy for active rheumatoid arthritis in China using evidence from a Chinese head-to-head randomized trial. Methods: A Markov model with 6 months per cycle was developed to estimate costs and health utilization in the lifetime of patients with RA from the Chinese healthcare system. The analysis data were derived from the randomized clinical trial in China. The primary cost includes drug and other medical costs. The health utilities quality-adjusted life years (QALYs) were derived using EQ-5D-5L mapping from disease-specific health assessment questionnaire (HAQ) scores obtained in clinical trials. The cost-effectiveness analysis was conducted by calculating the incremental cost-effectiveness ratio (ICER) values for rhTNFR:Fc and MTX. One-way and probabilistic sensitivity analyses were conducted to test the robustness of the base-case result. Results: In the base case, rhTNFR:Fc yielded 8.20 QALYs versus 7.46 with methotrexate, resulting in an ICER of CNY 12,783.56 per QALY. Scenario ICERs for bDMARD group combination treatment were 11,776.31 per QALY. Scenario ICERs were CNY 8079.04 per QALY for the patient perspective and CNY 7630.34 per QALY for the medical insurance perspective. One-way analysis highlighted utility inputs as the main drivers, and probabilistic analysis indicated a high probability of cost-effectiveness across common willingness-to-pay thresholds. Conclusions: The fusion protein strategy achieved an incremental cost-effectiveness ratio far below the 2024 China per capita gross domestic product threshold of CNY 95,749 per quality-adjusted life year. As first-line therapy for active rheumatoid arthritis, it is cost-effective relative to methotrexate in the Chinese setting. Full article
Show Figures

Figure 1

21 pages, 1208 KB  
Review
Efgartigimod for Generalized Myasthenia Gravis and Beyond: A Narrative Review of Its Pharmacological Profile, Clinical Utility, and Expanding Applications
by Ghaith K. Mansour, Leen Alangari, Leen Khosyfan, Reem Alhammad and Ahmad W. Hajjar
Biomedicines 2025, 13(12), 2975; https://doi.org/10.3390/biomedicines13122975 - 4 Dec 2025
Viewed by 1643
Abstract
Efgartigimod is a novel neonatal Fc receptor (FcRn) antagonist that reduces pathogenic immunoglobulin G (IgG) autoantibodies, offering a targeted therapeutic approach for generalized myasthenia gravis (gMG) and other antibody-mediated autoimmune diseases. This narrative review synthesizes clinical trial data, pharmacological insights, and real-world evidence [...] Read more.
Efgartigimod is a novel neonatal Fc receptor (FcRn) antagonist that reduces pathogenic immunoglobulin G (IgG) autoantibodies, offering a targeted therapeutic approach for generalized myasthenia gravis (gMG) and other antibody-mediated autoimmune diseases. This narrative review synthesizes clinical trial data, pharmacological insights, and real-world evidence to evaluate efgartigimod’s efficacy, safety, and emerging applications. Phase 3 randomized controlled trials and extension studies demonstrate rapid and sustained improvements in muscle strength and patient-reported outcomes with a favorable safety profile, including reduced reliance on corticosteroids and intravenous immunoglobulin (IVIg). Additionally, observational studies highlight its expanding utility in diverse IgG-mediated disorders such as immune thrombocytopenia (ITP) and autoimmune encephalitis. Efgartigimod thus represents a paradigm shift in autoimmune disease management, enabling precision immunomodulation with the potential for broad clinical impact and improved patient quality of life (QOL). Full article
(This article belongs to the Section Drug Discovery, Development and Delivery)
Show Figures

Graphical abstract

20 pages, 1223 KB  
Review
Mechanistic Insights and Advances of Bispecific T Cell Engaging Antibodies Therapy in Multiple Myeloma
by Ting Fang Tang, Chin Sum Cheong, Chung Yeng Looi, Won Fen Wong and Gin Gin Gan
Medicina 2025, 61(12), 2113; https://doi.org/10.3390/medicina61122113 - 27 Nov 2025
Viewed by 1470
Abstract
Multiple myeloma (MM) is a clonal malignancy of terminally differentiated plasma cells characterized by bone marrow infiltration and excessive production of monoclonal immunoglobulins, leading to end-organ damage such as osteolytic bone lesions. Despite substantial therapeutic progress achieved with proteasome inhibitors, immunomodulatory drugs, and [...] Read more.
Multiple myeloma (MM) is a clonal malignancy of terminally differentiated plasma cells characterized by bone marrow infiltration and excessive production of monoclonal immunoglobulins, leading to end-organ damage such as osteolytic bone lesions. Despite substantial therapeutic progress achieved with proteasome inhibitors, immunomodulatory drugs, and anti-CD38 monoclonal antibodies, multiple myeloma remains incurable, and outcomes for triple-class-refractory patients remain dismal, with median survival below one year. Bispecific T cell engaging antibodies (TCEs) have recently emerged as a promising immunotherapeutic approach capable of redirecting cytotoxic T cells to eliminate malignant plasma cells. These engineered antibodies simultaneously engage CD3 on T cells and a tumor-associated antigen such as B cell maturation antigen (BCMA), G protein-coupled receptor family C group 5 member D (GPRC5D), or Fc receptor homolog 5 (FcRH5), thereby forming an immune synapse that triggers T cell activation, cytokine secretion, and perforin–granzyme-mediated apoptosis of the targeted B cell. This review summarizes the molecular design, mechanism of action, and clinical development of TCEs in MM, encompassing early bi-specific T cell engagers (BiTE) constructs such as AMG 420 and next-generation IgG-like molecules including teclistamab. Pivotal clinical trials have demonstrated overall response rates between 43% and 73%, accompanied by durable remissions and manageable safety profiles. Future directions include earlier-line integration, synergistic combinations with immunomodulatory or costimulatory agents, and the development of trispecific formats to overcome antigen escape and T cell exhaustion. Collectively, TCEs represent a paradigm shift toward durable, immune-mediated disease control in multiple myeloma. Full article
Show Figures

Figure 1

25 pages, 2715 KB  
Article
Characterization of Novel Variants in P2YRY12, GP6 and TBXAS1 in Patients with Lifelong History of Bleeding
by Ana Zamora-Cánovas, Ana Marín-Quílez, Lorena Díaz-Ajenjo, Ana Sánchez-Fuentes, Pedro Luis Gómez-González, Marilena Crescente, Nuria Fernández-Mosteirín, José Padilla, José Ramón González-Porras, Rocío Benito, María Luisa Lozano, José María Bastida and José Rivera Pozo
Biomolecules 2025, 15(12), 1639; https://doi.org/10.3390/biom15121639 - 21 Nov 2025
Viewed by 562
Abstract
Inherited platelet function disorders (IPFDs) are rare diseases caused by defects in platelet surface receptors, enzymes, granules, or signaling proteins. In humans, GPVI and P2Y12 deficiency cause autosomal recessive bleeding disorders, while TBXAS1 deficiency is related to Ghosal hematodiaphyseal dysplasa, a rare autosomal [...] Read more.
Inherited platelet function disorders (IPFDs) are rare diseases caused by defects in platelet surface receptors, enzymes, granules, or signaling proteins. In humans, GPVI and P2Y12 deficiency cause autosomal recessive bleeding disorders, while TBXAS1 deficiency is related to Ghosal hematodiaphyseal dysplasa, a rare autosomal recessive disorder characterized by increased long bone density and platelet dysfunction without bleeding. To date, at least 20 patients have been identified with molecular defects in P2RY12, 12 cases with molecular defects in GP6, and 34 cases with molecular defects in TBXAS1. Here, we report a novel nonsense and missense variants in P2RY12, a novel nonsense variant in GP6, and a novel missense variant in TBXAS1. These variants selectively affect the platelet reactivity to ADP and collagen/CRP, predisposing to bleeding. P2RY12 c.835 G>A [p.Val279Met] variant did not affect receptor expression whereas P2RY12 c.44delG [p.Ser15Ilefs*33] lead to decreased levels of the receptor in one of the patients. This was confirmed both by RT-qPCR and immunoblotting analysis. Decreased expression of both GPVI and FcRγ-chain was detected in patients carrying GPVI nonsense variant in heterozygosis. The deleterious effect of these variants was also confirmed in a transfected cell line model. TBXAS1 variant triggered decreased TxA2 production using a cell line model. These variants expand the genetic landscape of P2RY12, GPVI and TBXAS1 inherited deficiency. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

21 pages, 17128 KB  
Article
Changes in the Interaction Properties of Antibodies with Fc Receptors upon Binding to Target Antigens
by Artem S. Grevtsev, Anton A. Kommer, Irina S. Zelmanchuk, Andrei S. Avdiushkin, Elizaveta O. Ermolaeva, Aleksandr A. Tiulin, Darya O. Chernyshova, Alexandra D. Azarian, Alexandr A. Gordeev and Aleksey K. Misorin
Biosensors 2025, 15(11), 759; https://doi.org/10.3390/bios15110759 - 14 Nov 2025
Viewed by 1036
Abstract
The interaction of therapeutic antibodies with Fc receptors is an important property that is actively modified to improve pharmacokinetic profiles and optimize antibody-dependent mechanisms of action. Various modifications of the Fc and hinge regions of antibodies, leading to a change in affinity with [...] Read more.
The interaction of therapeutic antibodies with Fc receptors is an important property that is actively modified to improve pharmacokinetic profiles and optimize antibody-dependent mechanisms of action. Various modifications of the Fc and hinge regions of antibodies, leading to a change in affinity with various Fc receptors, are widely covered in the literature. However, data on changes in antibody and Fc receptor interactions after antibody binding to the target antigen are poorly covered in the literature. In this work, we demonstrated a change in the affinity of the interaction of antibodies with Fc receptors after binding to the target antigen via the method of biolayer interferometry. An interesting result was a significant weakening of the interaction of FcRn and FcγRIIIa with some of the antibodies when the latter bound to the target antigen, which suggests the importance of this effect for the pharmacokinetic properties and effector mechanisms of action necessary in the treatment of oncological diseases. The sensor-based biolayer interferometry methods presented in this paper allow antibody screening to be performed to detect the effects of the reduced affinity of interactions with Fc receptors, and can be a useful tool in the early development of therapeutic antibodies. Full article
(This article belongs to the Section Biosensors and Healthcare)
Show Figures

Figure 1

18 pages, 1936 KB  
Article
Does a Polycistronic 2A Design Enable Functional FcRn Production for Antibody Pharmacokinetic Studies?
by Valentina S. Nesmeyanova, Nikita D. Ushkalenko, Sergei E. Olkin, Maksim N. Kosenko, Elena A. Rukhlova, Ivan M. Susloparov and Dmitry N. Shcherbakov
Pharmaceutics 2025, 17(11), 1463; https://doi.org/10.3390/pharmaceutics17111463 - 13 Nov 2025
Viewed by 821
Abstract
Background/Objectives: The neonatal Fc receptor (FcRn) is a heterodimeric protein composed of a heavy α-chain with an MHC class I-like fold and β2-microglobulin. It plays a crucial role in maintaining the homeostasis and pharmacokinetics of immunoglobulin G (IgG) and albumin through [...] Read more.
Background/Objectives: The neonatal Fc receptor (FcRn) is a heterodimeric protein composed of a heavy α-chain with an MHC class I-like fold and β2-microglobulin. It plays a crucial role in maintaining the homeostasis and pharmacokinetics of immunoglobulin G (IgG) and albumin through pH-dependent recycling. The production of soluble recombinant FcRn is technically challenging due to its heterodimeric structure and the presence of a transmembrane domain. This study aimed to develop a polycistronic construct enabling the co-expression of FcRn subunits from a single transcript and to evaluate the functional activity of the resulting protein in CHO-K1 cells. Methods: Integration vectors (pComV-FcRn-B2M) were designed to encode FcRn and β2-microglobulin linked via self-cleaving 2A peptides (P2A, E2A, F2A, T2A). Stable producer cell lines were generated using the Sleeping Beauty transposon system. The purified proteins were characterized by SDS-PAGE, Western blotting, and size-exclusion chromatography (SEC). Functional activity was assessed by ELISA and bio-layer interferometry (BLI). Results: Electrophoretic and chromatographic analyses confirmed the expected subunit composition and demonstrated that over 95% of the recombinant protein was monomeric. Functional assays revealed pH-dependent IgG binding, with strong interaction at pH 6.0 and negligible binding at pH 7.5. BLI measurements showed high affinity consistent with native FcRn function (KD = 3.15 nM at pH 6.0). Conclusions: The developed polycistronic construct containing a P2A peptide with a GSG linker enabled efficient production of functional FcRn in CHO-K1 cells (yield up to 2.23 mg/mL). The P2A variant demonstrated the highest efficiency and can serve as a reference system for screening Fc-engineered antibodies with optimized pharmacokinetic properties. Full article
Show Figures

Figure 1

21 pages, 2723 KB  
Article
miRNA-Mediated Regulation of Meloidogyne arenaria Responses in Wild Arachis
by Patricia Messenberg Guimaraes, Andressa da Cunha Quintana Martins, Roberto Coiti Togawa, Mario Alfredo de Passos Saraiva, Ana Luiza Machado Lacerda, Ana Cristina Miranda Brasileiro and Priscila Grynberg
Int. J. Mol. Sci. 2025, 26(22), 10824; https://doi.org/10.3390/ijms262210824 - 7 Nov 2025
Viewed by 509
Abstract
MicroRNAs (miRNAs) are key post-transcriptional regulators of plant development and stress responses, with many being conserved across diverse plant lineages. In this study, we investigated the expression profiles of miRNAs and their corresponding target genes in Arachis stenosperma, a wild peanut relative [...] Read more.
MicroRNAs (miRNAs) are key post-transcriptional regulators of plant development and stress responses, with many being conserved across diverse plant lineages. In this study, we investigated the expression profiles of miRNAs and their corresponding target genes in Arachis stenosperma, a wild peanut relative that exhibits robust resistance to root-knot nematodes (RKN). Small RNA sequencing of nematode-infected roots identified 107 miRNA loci, of which 93 corresponded to conserved miRNA families and 14 represented novel candidates, designated as miRNOVO. Among these, 18 miRNAs belonging to 11 conserved families were identified as differentially expressed (DEMs). Notably, miR399 and miR319 showed the highest upregulation (logFC = 4.25 and 4.20), while miR393 and miR477 were the most downregulated (logFC = −0.83 and −0.79). Integrated analysis of miRNA and transcriptome data revealed several regulatory interactions involving key defense-related genes. These included NLR genes targeted by miR393 and miR477, hormone signaling components such as the auxin response factor ARF8 targeted by miR167, and the growth regulator GRF2 targeted by miR396. Additionally, miR408 was predicted to target laccase3, a gene involved in the oxidation of phenolic compounds, lignin biosynthesis, copper homeostasis and defense responses. Remarkably, four immune receptor genes belonging to the nucleotide-binding site leucine-rich repeat (NLR) family displayed inverse expression patterns relative to their regulatory miRNAs, suggesting miRNA-mediated post-transcriptional control during the early stages of nematode infection. These findings reveal both conserved and species-specific miRNA–mRNA modules associated with nematode resistance in A. stenosperma, highlighting promising targets for developing RKN-tolerant peanut cultivars through miRNA-based strategies. Full article
(This article belongs to the Special Issue Interactions between Plants and Nematodes)
Show Figures

Figure 1

22 pages, 1228 KB  
Review
The Role of Fc-like Receptor 3 in the Pathophysiology of Rheumatoid Arthritis
by Paweł Dec, Paulina Plewa, Adam Kubisa and Andrzej Pawlik
Genes 2025, 16(11), 1318; https://doi.org/10.3390/genes16111318 - 2 Nov 2025
Viewed by 875
Abstract
The pathogenesis of rheumatoid arthritis involves a complex interplay of genetic predisposition, environmental factors, and autoimmune mechanisms that lead to chronic inflammation of the synovial membrane. Fc-like receptor 3 (FcRL3) is a receptor encoded by the FCRL3 gene, located on the long arm [...] Read more.
The pathogenesis of rheumatoid arthritis involves a complex interplay of genetic predisposition, environmental factors, and autoimmune mechanisms that lead to chronic inflammation of the synovial membrane. Fc-like receptor 3 (FcRL3) is a receptor encoded by the FCRL3 gene, located on the long arm of chromosome 1 at 1q23.1. Polymorphisms in the promoter region of FCRL3, rather than elsewhere in the gene, primarily affect the level of protein expression, which is of clinical significance. Understanding the structure of FcRL3, particularly in the context of genetic variants, is therefore important for elucidating the pathogenesis of autoimmune diseases. Detailed knowledge of the molecular architecture of immune receptors such as FcRL3 is also essential for advancing our understanding of immune function and for guiding the development of targeted therapeutic strategies in autoimmune disease. In this article, we discuss the role of FcRL3 in the pathophysiology and potential therapy of rheumatoid arthritis. Full article
(This article belongs to the Section Human Genomics and Genetic Diseases)
Show Figures

Figure 1

Back to TopTop