Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.616
Filter
1.
Sci Signal ; 17(844): eado5279, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38980923

ABSTRACT

In this issue of Science Signaling, Jackson et al. present a new antibody strategy to-quite literally-strap transforming growth factor-ß1 (TGF-ß1) to latent complexes in the extracellular matrix. The antibody has no effect on latent TGF-ß1 presented on the surface of immune cells and thus allows targeting of the detrimental effects of TGF-ß1 in fibrosis without affecting its beneficial immune-suppressing activities.


Subject(s)
Extracellular Matrix , Transforming Growth Factor beta1 , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/immunology , Humans , Extracellular Matrix/metabolism , Extracellular Matrix/immunology , Animals , Fibrosis , Antibodies/immunology , Antibodies/metabolism , Mice
2.
Sci Rep ; 14(1): 15992, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987432

ABSTRACT

Aquaporins (AQPs) are a family of water permeable channels expressed on the plasma membrane with AQP5 being the major channel expressed in several human tissues including salivary and lacrimal glands. Anti-AQP5 autoantibodies have been observed in patients with Sjögren's syndrome who are characterised by dryness of both salivary and lacrimal glands, and they have been implicated in the underlying mechanisms of glandular dysfunction. AQP5 is formed by six transmembrane helices linked with three extracellular and two intracellular loops. Develop antibodies against membrane protein extracellular loops can be a challenge due to the difficulty in maintaining these proteins as recombinant in their native form. Therefore, in this work we aimed to generate an efficient stable-transfected cell line overexpressing human AQP5 (CHO-K1/AQP5) to perform primarily cell-based phage display biopanning experiments to develop new potential recombinant antibodies targeting AQP5. We also showed that the new CHO-K1/AQP5 cell line can be used to study molecular mechanisms of AQP5 sub-cellular trafficking making these cells a useful tool for functional studies.


Subject(s)
Aquaporin 5 , Cricetulus , Aquaporin 5/metabolism , Aquaporin 5/genetics , CHO Cells , Humans , Animals , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Antibodies/metabolism , Peptide Library
3.
Brief Bioinform ; 25(4)2024 May 23.
Article in English | MEDLINE | ID: mdl-38960407

ABSTRACT

The optimization of therapeutic antibodies through traditional techniques, such as candidate screening via hybridoma or phage display, is resource-intensive and time-consuming. In recent years, computational and artificial intelligence-based methods have been actively developed to accelerate and improve the development of therapeutic antibodies. In this study, we developed an end-to-end sequence-based deep learning model, termed AttABseq, for the predictions of the antigen-antibody binding affinity changes connected with antibody mutations. AttABseq is a highly efficient and generic attention-based model by utilizing diverse antigen-antibody complex sequences as the input to predict the binding affinity changes of residue mutations. The assessment on the three benchmark datasets illustrates that AttABseq is 120% more accurate than other sequence-based models in terms of the Pearson correlation coefficient between the predicted and experimental binding affinity changes. Moreover, AttABseq also either outperforms or competes favorably with the structure-based approaches. Furthermore, AttABseq consistently demonstrates robust predictive capabilities across a diverse array of conditions, underscoring its remarkable capacity for generalization across a wide spectrum of antigen-antibody complexes. It imposes no constraints on the quantity of altered residues, rendering it particularly applicable in scenarios where crystallographic structures remain unavailable. The attention-based interpretability analysis indicates that the causal effects of point mutations on antibody-antigen binding affinity changes can be visualized at the residue level, which might assist automated antibody sequence optimization. We believe that AttABseq provides a fiercely competitive answer to therapeutic antibody optimization.


Subject(s)
Antigen-Antibody Complex , Deep Learning , Antigen-Antibody Complex/chemistry , Antigens/chemistry , Antigens/genetics , Antigens/metabolism , Antigens/immunology , Antibody Affinity , Amino Acid Sequence , Computational Biology/methods , Humans , Mutation , Antibodies/chemistry , Antibodies/immunology , Antibodies/genetics , Antibodies/metabolism
4.
Elife ; 132024 May 08.
Article in English | MEDLINE | ID: mdl-38717135

ABSTRACT

Eukaryotic chromatin is organized into functional domains, that are characterized by distinct proteomic compositions and specific nuclear positions. In contrast to cellular organelles surrounded by lipid membranes, the composition of distinct chromatin domains is rather ill described and highly dynamic. To gain molecular insight into these domains and explore their composition, we developed an antibody-based proximity biotinylation method targeting the RNA and proteins constituents. The method that we termed antibody-mediated proximity labelling coupled to mass spectrometry (AMPL-MS) does not require the expression of fusion proteins and therefore constitutes a versatile and very sensitive method to characterize the composition of chromatin domains based on specific signature proteins or histone modifications. To demonstrate the utility of our approach we used AMPL-MS to characterize the molecular features of the chromocenter as well as the chromosome territory containing the hyperactive X chromosome in Drosophila. This analysis identified a number of known RNA-binding proteins in proximity of the hyperactive X and the centromere, supporting the accuracy of our method. In addition, it enabled us to characterize the role of RNA in the formation of these nuclear bodies. Furthermore, our method identified a new set of RNA molecules associated with the Drosophila centromere. Characterization of these novel molecules suggested the formation of R-loops in centromeres, which we validated using a novel probe for R-loops in Drosophila. Taken together, AMPL-MS improves the selectivity and specificity of proximity ligation allowing for novel discoveries of weak protein-RNA interactions in biologically diverse domains.


Subject(s)
Chromatin , Mass Spectrometry , RNA , Animals , Chromatin/metabolism , Chromatin/chemistry , RNA/metabolism , RNA/chemistry , Mass Spectrometry/methods , Drosophila/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/genetics , Biotinylation , Centromere/metabolism , Antibodies/metabolism , Antibodies/chemistry , X Chromosome/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/chemistry , Staining and Labeling/methods
5.
Protein J ; 43(3): 405-424, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38724751

ABSTRACT

As the demand for immunotherapy to treat and manage cancers, infectious diseases and other disorders grows, a comprehensive understanding of amino acids and their intricate role in antibody engineering has become a prime requirement. Naturally produced antibodies may not have the most suitable amino acids at the complementarity determining regions (CDR) and framework regions, for therapeutic purposes. Therefore, to enhance the binding affinity and therapeutic properties of an antibody, the specific impact of certain amino acids on the antibody's architecture must be thoroughly studied. In antibody engineering, it is crucial to identify the key amino acid residues that significantly contribute to improving antibody properties. Therapeutic antibodies with higher binding affinity and improved functionality can be achieved through modifications or substitutions with highly suitable amino acid residues. Here, we have indicated the frequency of amino acids and their association with the binding free energy in CDRs. The review also analyzes the experimental outcome of two studies that reveal the frequency of amino acids in CDRs and provides their significant correlation between the outcomes. Additionally, it discusses the various bond interactions within the antibody structure and antigen binding. A detailed understanding of these amino acid properties should assist in the analysis of antibody sequences and structures needed for designing and enhancing the overall performance of therapeutic antibodies.


Subject(s)
Amino Acids , Complementarity Determining Regions , Protein Engineering , Amino Acids/chemistry , Complementarity Determining Regions/chemistry , Complementarity Determining Regions/genetics , Humans , Protein Engineering/methods , Antibodies/chemistry , Antibodies/immunology , Antibodies/metabolism , Antibody Affinity , Animals
6.
Nature ; 630(8016): 493-500, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718835

ABSTRACT

The introduction of AlphaFold 21 has spurred a revolution in modelling the structure of proteins and their interactions, enabling a huge range of applications in protein modelling and design2-6. Here we describe our AlphaFold 3 model with a substantially updated diffusion-based architecture that is capable of predicting the joint structure of complexes including proteins, nucleic acids, small molecules, ions and modified residues. The new AlphaFold model demonstrates substantially improved accuracy over many previous specialized tools: far greater accuracy for protein-ligand interactions compared with state-of-the-art docking tools, much higher accuracy for protein-nucleic acid interactions compared with nucleic-acid-specific predictors and substantially higher antibody-antigen prediction accuracy compared with AlphaFold-Multimer v.2.37,8. Together, these results show that high-accuracy modelling across biomolecular space is possible within a single unified deep-learning framework.


Subject(s)
Deep Learning , Ligands , Models, Molecular , Proteins , Software , Humans , Antibodies/chemistry , Antibodies/metabolism , Antigens/metabolism , Antigens/chemistry , Deep Learning/standards , Ions/chemistry , Ions/metabolism , Molecular Docking Simulation , Nucleic Acids/chemistry , Nucleic Acids/metabolism , Protein Binding , Protein Conformation , Proteins/chemistry , Proteins/metabolism , Reproducibility of Results , Software/standards
7.
Histochem Cell Biol ; 162(1-2): 41-52, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38762823

ABSTRACT

During development and differentiation, histone modifications dynamically change locally and globally, associated with transcriptional regulation, DNA replication and repair, and chromosome condensation. The level of histone H4 Lys20 monomethylation (H4K20me1) increases during the G2 to M phases of the cell cycle and is enriched in facultative heterochromatin, such as inactive X chromosomes in cycling cells. To track the dynamic changes of H4K20me1 in living cells, we have developed a genetically encoded modification-specific intracellular antibody (mintbody) probe that specifically binds to the modification. Here, we report the generation of knock-in mice in which the coding sequence of the mCherry-tagged version of the H4K20me1-mintbody is inserted into the Rosa26 locus. The knock-in mice, which ubiquitously expressed the H4K20me1-mintbody, developed normally and were fertile, indicating that the expression of the probe does not disturb the cell growth, development, or differentiation. Various tissues isolated from the knock-in mice exhibited nuclear fluorescence without the need for fixation. The H4K20me1-mintbody was enriched in inactive X chromosomes in developing embryos and in XY bodies during spermatogenesis. The knock-in mice will be useful for the histochemical analysis of H4K20me1 in any cell types.


Subject(s)
Gene Knock-In Techniques , Histones , Luminescent Proteins , Animals , Mice , Histones/metabolism , Luminescent Proteins/metabolism , Luminescent Proteins/genetics , Antibodies/metabolism , Red Fluorescent Protein , Male , Mice, Inbred C57BL , Mice, Transgenic
8.
J Microbiol Biotechnol ; 34(6): 1222-1228, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38783697

ABSTRACT

Protein-specific antibodies are essential for various aspects of protein research, including detection, purification, and characterization. When specific antibodies are unavailable, protein tagging is a useful alternative. Small epitope tags, typically less than 10 amino acids, are widely used in protein research due to the simple modification through PCR and reduced impact on the target protein's function compared to larger tags. The 2B8 epitope tag (RDPLPFFPP), reported by us in a previous study, has high specificity and sensitivity to the corresponding antibody. However, when attached to the C-terminus of the target protein in immunoprecipitation experiments, we observed a decrease in detection signal with reduced immunity and low protein recovery. This phenomenon was not unique to 2B8 and was also observed with the commercially available Myc tag. Our study revealed that C-terminal tagging of small epitope tags requires the addition of more than one extra amino acid to enhance (restore) antibody immunities. Moreover, among the amino acids we tested, serine was the best for the 2B8 tag. Our findings demonstrated that the interaction between a small epitope and a corresponding paratope of an antibody requires an extra amino acid at the C-terminus of the epitope. This result is important for researchers planning studies on target proteins using small epitope tags.


Subject(s)
Amino Acids , Epitopes , Epitopes/immunology , Animals , Antibodies/immunology , Antibodies/metabolism , Mice , Immunoprecipitation , Antibody Formation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/chemistry
9.
Sci Rep ; 14(1): 9003, 2024 04 18.
Article in English | MEDLINE | ID: mdl-38637614

ABSTRACT

The invasive Asian longhorned tick Haemaphysalis longicornis that vectors and transmits several animal pathogens is significantly expanding in the United States. Recent studies report that these ticks also harbor human pathogens including Borrelia burgdorferi sensu lato, Babesia microti, and Anaplasma phagocytophilum. Therefore, studies that address the interactions of these ticks with human pathogens are important. In this study, we report the characterization of H. longicornis organic anion-transporting polypeptides (OATPs) in interactions of these ticks with A. phagocytophilum. Using OATP-signature sequence, we identified six OATPs in the H. longicornis genome. Bioinformatic analysis revealed that H. longicornis OATPs are closer to other tick orthologs rather than to mammalian counterparts. Quantitative real-time PCR analysis revealed that OATPs are highly expressed in immature stages when compared to mature stages of these ticks. In addition, we noted that the presence of A. phagocytophilum upregulates a specific OATP in these ticks. We also noted that exogenous treatment of H. longicornis with xanthurenic acid, a tryptophan metabolite, influenced OATP expression in these ticks. Immunoblotting analysis revealed that antibody generated against Ixodes scapularis OATP cross-reacted with H. longicornis OATP. Furthermore, treatment of H. longicornis with OATP antibody impaired colonization of A. phagocytophilum in these ticks. These results not only provide evidence that the OATP-tryptophan pathway is important for A. phagocytophilum survival in H. longicornis ticks but also indicate OATP as a promising candidate for the development of a universal anti-tick vaccine to target this bacterium and perhaps other rickettsial pathogens of medical importance.


Subject(s)
Anaplasma phagocytophilum , Borrelia burgdorferi , Borrelia , Ixodes , Organic Anion Transporters , Animals , Humans , Haemaphysalis longicornis , Anaplasma phagocytophilum/genetics , Tryptophan , Ixodes/microbiology , Antibodies/metabolism , Organic Anion Transporters/genetics , Borrelia burgdorferi/metabolism , Mammals/metabolism
10.
Scand J Immunol ; 99(5): e13356, 2024 May.
Article in English | MEDLINE | ID: mdl-38605549

ABSTRACT

In light of increasing resistance to PD1 antibody therapy among certain patient populations, there is a critical need for in-depth research. Our study assesses the synergistic effects of a MUC1 DNA vaccine and PD1 antibody for surmounting PD1 resistance, employing a murine CT26/MUC1 colon carcinoma model for this purpose. When given as a standalone treatment, PD1 antibodies showed no impact on tumour growth. Additionally, there was no change observed in the intra-tumoural T-cell ratios or in the functionality of T-cells. In contrast, the sole administration of a MUC1 DNA vaccine markedly boosted the cytotoxicity of CD8+ T cells by elevating IFN-γ and granzyme B production. Our compelling evidence highlights that combination therapy more effectively inhibited tumour growth and prolonged survival compared to either monotherapy, thus mitigating the limitations intrinsic to single-agent therapies. This enhanced efficacy was driven by a significant alteration in the tumour microenvironment, skewing it towards pro-immunogenic conditions. This assertion is backed by a raised CD8+/CD4+ T-cell ratio and a decrease in immunosuppressive MDSC and Treg cell populations. On the mechanistic front, the synergistic therapy amplified expression levels of CXCL13 in tumours, subsequently facilitating T-cell ingress into the tumour setting. In summary, our findings advocate for integrated therapy as a potent mechanism for surmounting PD1 antibody resistance, capitalizing on improved T-cell functionality and infiltration. This investigation affords critical perspectives on enhancing anti-tumour immunity through the application of innovative therapeutic strategies.


Subject(s)
Antibodies , Mucin-1 , Neoplasms , Programmed Cell Death 1 Receptor , Vaccines, DNA , Animals , Mice , Antibodies/metabolism , CD8-Positive T-Lymphocytes , Cell Line, Tumor , Mucin-1/genetics , Neoplasms/metabolism , Programmed Cell Death 1 Receptor/metabolism , Tumor Microenvironment
11.
Methods Mol Biol ; 2790: 405-416, 2024.
Article in English | MEDLINE | ID: mdl-38649583

ABSTRACT

Antibodies are a valuable research tool, with uses including detection and quantification of specific proteins. By using peptide fragments to raise antibodies, they can be designed to differentiate between structurally similar proteins, or to bind conserved motifs in divergent proteins. Peptide sequence selection and antibody validation are crucial to ensure reliable results from antibody-based experiments. This chapter describes the steps for the identification of peptide sequences to produce protein- or isoform-specific antibodies using recombinant technologies as well as the subsequent validation of such antibodies. The photosynthetic protein Rubisco activase is used as a case study to explain the various steps involved and key aspects to take into consideration.


Subject(s)
Antibodies , Protein Isoforms , Antibodies/chemistry , Antibodies/immunology , Antibodies/metabolism , Photosynthesis , Amino Acid Sequence , Plant Proteins/metabolism
12.
Biochem J ; 481(10): 643-651, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38683688

ABSTRACT

GCN5L1, also known as BLOC1S1 and BLOS1, is a small intracellular protein involved in many key biological processes. Over the last decade, GCN5L1 has been implicated in the regulation of protein lysine acetylation, energy metabolism, endo-lysosomal function, and cellular immune pathways. An increasing number of published papers have used commercially-available reagents to interrogate GCN5L1 function. However, in many cases these reagents have not been rigorously validated, leading to potentially misleading results. In this report we tested several commercially-available antibodies for GCN5L1, and found that two-thirds of those available did not unambiguously detect the protein by western blot in cultured mouse cells or ex vivo liver tissue. These data suggest that previously published studies which used these unverified antibodies to measure GCN5L1 protein abundance, in the absence of other independent methods of corroboration, should be interpreted with appropriate caution.


Subject(s)
Antibodies , Animals , Mice , Antibodies/immunology , Antibodies/metabolism , Liver/metabolism , Liver/immunology , Mice, Knockout , Mitochondrial Proteins/immunology , Nerve Tissue Proteins/immunology
13.
Sci Transl Med ; 16(741): eadj9052, 2024 Apr 03.
Article in English | MEDLINE | ID: mdl-38569016

ABSTRACT

Microglia help limit the progression of Alzheimer's disease (AD) by constraining amyloid-ß (Aß) pathology, effected through a balance of activating and inhibitory intracellular signals delivered by distinct cell surface receptors. Human leukocyte Ig-like receptor B4 (LILRB4) is an inhibitory receptor of the immunoglobulin (Ig) superfamily that is expressed on myeloid cells and recognizes apolipoprotein E (ApoE) among other ligands. Here, we find that LILRB4 is highly expressed in the microglia of patients with AD. Using mice that accumulate Aß and carry a transgene encompassing a portion of the LILR region that includes LILRB4, we corroborated abundant LILRB4 expression in microglia wrapping around Aß plaques. Systemic treatment of these mice with an anti-human LILRB4 monoclonal antibody (mAb) reduced Aß load, mitigated some Aß-related behavioral abnormalities, enhanced microglia activity, and attenuated expression of interferon-induced genes. In vitro binding experiments established that human LILRB4 binds both human and mouse ApoE and that anti-human LILRB4 mAb blocks such interaction. In silico modeling, biochemical, and mutagenesis analyses identified a loop between the two extracellular Ig domains of LILRB4 required for interaction with mouse ApoE and further indicated that anti-LILRB4 mAb may block LILRB4-mApoE by directly binding this loop. Thus, targeting LILRB4 may be a potential therapeutic avenue for AD.


Subject(s)
Alzheimer Disease , Microglia , Humans , Mice , Animals , Microglia/metabolism , Antibodies/metabolism , Receptors, Cell Surface/metabolism , Amyloid/metabolism , Disease Models, Animal , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Apolipoproteins E , Leukocytes/metabolism , Mice, Transgenic , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism
14.
Methods Mol Biol ; 2791: 23-33, 2024.
Article in English | MEDLINE | ID: mdl-38532089

ABSTRACT

Epigenetic programming plays a vital role in regulating pluripotency genes, which become activated or inactivated during the processes of dedifferentiation and differentiation during an organism's development. The analysis of epigenetic modifications has become possible through the technique of immunostaining, where specific antibodies allow the identification of a single target protein. This chapter describes a detailed protocol for the analysis of the epigenetic modifications with the use of confocal microscopy, subsequent image, and statistical analysis on the example of Fagopyrum calli with the use of nine antibodies raised against histone H3 and H4 methylation and acetylation on several lysines as well as DNA methylation.


Subject(s)
Fagopyrum , Fagopyrum/metabolism , Histones/metabolism , Cell Nucleus/metabolism , DNA Methylation , Antibodies/metabolism , Epigenesis, Genetic , Acetylation
15.
Methods Mol Biol ; 2791: 15-22, 2024.
Article in English | MEDLINE | ID: mdl-38532088

ABSTRACT

Immunostaining is a well-established technique for identifying specific proteins in tissue samples with specific antibodies to identify a single target protein. It is commonly used in research and provides information about cellular localization and protein expression levels. This chapter describes a detailed protocol for immunostaining fixed Fagopyrum calli embedded in Steedman's wax using nine antibodies raised against histone H3 and H4 methylation and acetylation on several lysines and DNA methylation.


Subject(s)
Fagopyrum , Fagopyrum/metabolism , Histones/metabolism , Epigenesis, Genetic , DNA Methylation , Lysine/metabolism , Antibodies/metabolism , Acetylation
16.
ACS Nano ; 18(12): 8733-8744, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38469811

ABSTRACT

Covalent conjugation of poly(ethylene glycol) (PEG) is frequently employed to enhance the pharmacokinetics and biodistribution of various protein and nanoparticle therapeutics. Unfortunately, some PEGylated drugs can induce elevated levels of antibodies that can bind PEG, i.e., anti-PEG antibodies (APA), in some patients. APA in turn can reduce the efficacy and increase the risks of allergic reactions, including anaphylaxis. There is currently no intervention available in the clinic that specifically mitigates allergic reactions to PEGylated drugs without the use of broad immunosuppression. We previously showed that infusion of high molecular weight free PEG could safely and effectively suppress the induction of APA in mice and restore prolonged circulation of various PEGylated therapeutics. Here, we explored the effectiveness of free PEG as a prophylaxis against anaphylaxis induced by PEG-specific allergic reactions in swine. Injection of PEG-liposomes (PL) resulted in anaphylactoid shock (pseudoanaphylaxis) within 1-3 min in both naïve and PL-sensitized swine. In contrast, repeated injection of free PEG alone did not result in allergic reactions, and injection of free PEG effectively suppressed allergic reactions to PL, including in previously PL-sensitized swine. These results strongly support the further investigation of free PEG for reducing APA and allergic responses to PEGylated therapeutics.


Subject(s)
Anaphylaxis , Humans , Animals , Swine , Mice , Anaphylaxis/chemically induced , Anaphylaxis/drug therapy , Anaphylaxis/prevention & control , Tissue Distribution , Nanomedicine , Polyethylene Glycols/pharmacology , Antibodies/metabolism , Liposomes/pharmacology
17.
Methods Mol Biol ; 2754: 437-444, 2024.
Article in English | MEDLINE | ID: mdl-38512680

ABSTRACT

Microglia are brain-resident phagocytic cells, considered to be the innate immune cells of the central nervous system. Microglia respond to both infectious pathogens in the brain and sterile cellular debris, including the proteinaceous aggregates that accumulate in the brains of patients with Alzheimer's disease (AD). Microtubule-associated protein Tau is an intracellular protein that self-aggregates into neurofibrillary tangles in Alzheimer's disease and many other neurodegenerative diseases. Ongoing clinical trials are testing whether therapeutic antibodies specific to Tau protein aggregates can reduce pathological protein deposition and improve the course of disease. Data suggest that Tau-specific antibodies act on extracellular Tau aggregates by promoting uptake into microglia cells and thus preventing its prion-like spread to unaffected neurons. Here we describe a protocol to test the effect of Tau-specific antibodies on Tau uptake into microglia by flow cytometry. Recombinant Tau protein is fibrillized in vitro and tagged with a fluorescent label. Then, fibrillized Tau is incubated with the antibody of interest and applied to microglial cells in culture. Uptake of Tau into microglia is then assessed by measuring fluorescence intensity by flow cytometry. With slight modifications, this assay can be used to test effects of many antibodies, various Tau protein compositions, and different microglial sources in a high-throughput format.


Subject(s)
Alzheimer Disease , tau Proteins , Humans , tau Proteins/metabolism , Alzheimer Disease/metabolism , Microglia/metabolism , Flow Cytometry , Antibodies/metabolism , Brain/metabolism
18.
Methods Mol Biol ; 2754: 387-410, 2024.
Article in English | MEDLINE | ID: mdl-38512678

ABSTRACT

A region-specific catheter-based intranasal administration method was successfully developed, established, and validated as reported previously. By using this method, drugs can be applicated specifically to the olfactory region. Thereby, intranasally administered drugs could be delivered via neuronal connections to the central nervous system. Here, we present a detailed protocol with a step-by-step procedure for nose-to-brain delivery via the olfactory mucosa.Fc receptors such as the neonatal Fc receptor (FcRn) and potentially Fcγ receptor IIb (FcγRIIb) are involved in the uptake and transport of antibodies via the olfactory nasal mucosa. To better characterize their expression levels and their role in CNS drug delivery via the nose, an in situ hybridization (ISH) protocol was adapted for nasal mucosa samples and described in abundant details.


Subject(s)
Brain , Nasal Mucosa , Mice , Animals , Administration, Intranasal , Brain/metabolism , Pharmaceutical Preparations/metabolism , Antibodies/metabolism , Receptors, Fc/genetics , Receptors, Fc/metabolism , In Situ Hybridization , Drug Delivery Systems/methods
19.
Handb Clin Neurol ; 200: 347-364, 2024.
Article in English | MEDLINE | ID: mdl-38494289

ABSTRACT

Among patients with paraneoplastic neurologic syndromes (PNS), emphasis has historically been placed on neural antibodies against intracellular proteins that have a strong association with malignancy. Because of the intracellular location of their antigenic targets, these antibodies are typically considered to be non-pathogenic surrogate markers of immune cell-mediated neural injury. Unfortunately, patients with these antibodies often have suboptimal response to immunotherapy and poor prognosis. Over the last two decades, however, dramatic advancements have been made in the discovery and clinical characterization of neural antibodies against extracellular targets. These antibodies are generally considered to be pathogenic, given their potential to directly alter antigen structure or function, and patients with these antibodies often respond favorably to prompt immunotherapy. These antibodies also associate with tumors and may thus occur as PNS, albeit more variably than neural antibodies against intracellular targets. The updated 2021 PNS diagnostic criteria, which classifies antibodies as high-risk, intermediate-risk, or lower-risk for an associated cancer, better clarifies how neural antibodies against extracellular targets relate to PNS. Using this recently created framework, the clinical presentations, ancillary test findings, oncologic associations, and treatment responses of syndromes associated with these antibodies are discussed.


Subject(s)
Neoplasms , Paraneoplastic Syndromes, Nervous System , Humans , Antibodies/metabolism , Neoplasms/complications , Biomarkers , Autoantibodies
20.
ACS Nano ; 18(14): 10142-10155, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38526307

ABSTRACT

Fully mobilizing the activities of multiple immune cells is crucial to achieve the desired tumor immunotherapeutic efficacy yet still remains challenging. Herein, we report a nanomedicine formulation based on phosphorus dendrimer (termed AK128)/programmed cell death protein 1 antibody (aPD1) nanocomplexes (NCs) that are camouflaged with M1-type macrophage cell membranes (M1m) for enhanced immunotherapy of orthotopic glioma. The constructed AK128-aPD1@M1m NCs with a mean particle size of 160.3 nm possess good stability and cytocompatibility. By virtue of the decorated M1m having α4 and ß1 integrins, the NCs are able to penetrate the blood-brain barrier to codeliver both AK128 with intrinsic immunomodulatory activity and aPD1 to the orthotopic glioma with prolonged blood circulation time. We show that the phosphorus dendrimer AK128 can boost natural killer (NK) cell proliferation in peripheral blood mononuclear cells, while the delivered aPD1 enables immune checkpoint blockade (ICB) to restore the cytotoxic T cells and NK cells, thus promoting tumor cell apoptosis and simultaneously decreasing the tumor distribution of regulatory T cells vastly for improved glioma immunotherapy. The developed nanomedicine formulation with a simple composition achieves multiple modulations of immune cells by utilizing the immunomodulatory activity of nanocarrier and antibody-mediated ICB therapy, providing an effective strategy for cancer immunotherapy.


Subject(s)
Dendrimers , Glioma , Humans , Phosphorus , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Biomimetics , Glioma/therapy , Glioma/pathology , Immunotherapy , Killer Cells, Natural , Antibodies/metabolism , T-Lymphocytes, Cytotoxic , Blood-Brain Barrier/metabolism , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...