Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Int J Mol Sci ; 24(6)2023 Mar 10.
Article in English | MEDLINE | ID: mdl-36982375

ABSTRACT

Pregnancy is a fascinating immunological paradox: the semi-allogeneic fetus generally grows without any complications. In the placenta, fetal trophoblast cells come into contact with maternal immune cells. Inaccurate or inadequate adaptations of the maternal immune system could lead to problems with the functioning of the placenta. Macrophages are important for tissue homeostasis, cleanup, and the repair of damaged tissues. This is crucial for a rapidly developing organ such as the placenta. The consensus on macrophages at the maternal-fetal interface in pregnancy is that a major proportion have an anti-inflammatory, M2-like phenotype, that expresses scavenger receptors and is involved in tissue remodeling and the dampening of the immune reactions. Recent multidimensional analyses have contributed to a more detailed outlook on macrophages. The new view is that this lineage represents a highly diverse phenotype and is more prevalent than previously thought. Spatial-temporal in situ analyses during gestation have identified unique interactions of macrophages both with trophoblasts and with T cells at different trimesters of pregnancy. Here, we elaborate on the role of macrophages during early human pregnancy and at later gestation. Their possible effect is reviewed in the context of HLA incompatibility between mother and fetus, first in naturally conceived pregnancies, but foremost in pregnancies after oocyte donation. The potential functional consequences of macrophages for pregnancy-related immune reactions and the outcome in patients with recurrent pregnancy loss are also discussed.


Subject(s)
Decidua , Pregnancy Complications , Female , Pregnancy , Humans , Placenta , Trophoblasts , Macrophages , Fetus
2.
J Theor Biol ; 538: 111028, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35085535

ABSTRACT

Intra-tumour heterogeneity (ITH) has a strong impact on the efficacy of the immune response against solid tumours. The number of sub-populations of cancer cells expressing different antigens and the percentage of immunogenic cells (i.e. tumour cells that are effectively targeted by immune cells) in a tumour are both expressions of ITH. Here, we present a spatially explicit stochastic individual-based model of the interaction dynamics between tumour cells and CD8+ T cells, which makes it possible to dissect out the specific impact of these two expressions of ITH on anti-tumour immune response. The set-up of numerical simulations of the model is defined so as to mimic scenarios considered in previous experimental studies. Moreover, the ability of the model to qualitatively reproduce experimental observations of successful and unsuccessful immune surveillance is demonstrated. First, the results of numerical simulations of this model indicate that the presence of a larger number of sub-populations of tumour cells that express different antigens is associated with a reduced ability of CD8+ T cells to mount an effective anti-tumour immune response. Secondly, the presence of a larger percentage of tumour cells that are not effectively targeted by CD8+ T cells may reduce the effectiveness of anti-tumour immunity. Ultimately, the mathematical model presented in this paper may provide a framework to help biologists and clinicians to better understand the mechanisms that are responsible for the emergence of different outcomes of immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Neoplasms , Humans , Immunity, Cellular , Immunotherapy , Models, Theoretical , Neoplasms/therapy
3.
Bull Math Biol ; 84(12): 141, 2022 10 31.
Article in English | MEDLINE | ID: mdl-36315362

ABSTRACT

We present a spatial hybrid discrete-continuum modelling framework for the interaction dynamics between tumour cells and cytotoxic T cells, which play a pivotal role in the immune response against tumours. In this framework, tumour cells and T cells are modelled as individual agents while chemokines that drive the chemotactic movement of T cells towards the tumour are modelled as a continuum. We formally derive the continuum counterpart of this model, which is given by a coupled system that comprises an integro-differential equation for the density of tumour cells, a partial differential equation for the density of T cells and a partial differential equation for the concentration of chemokines. We report on computational results of the hybrid model and show that there is an excellent quantitative agreement between them and numerical solutions of the corresponding continuum model. These results shed light on the mechanisms that underlie the emergence of different levels of infiltration of T cells into the tumour and elucidate how T-cell infiltration shapes anti-tumour immune response. Moreover, to present a proof of concept for the idea that, exploiting the computational efficiency of the continuum model, extensive numerical simulations could be carried out, we investigate the impact of T-cell infiltration on the response of tumour cells to different types of anti-cancer immunotherapy.


Subject(s)
Models, Biological , Neoplasms , Humans , Computer Simulation , Mathematical Concepts , Neoplasms/pathology , Immunity , Chemokines
4.
Int J Mol Sci ; 23(23)2022 Nov 29.
Article in English | MEDLINE | ID: mdl-36499301

ABSTRACT

Nanoparticles (NPs) are commonly modified with tumor-targeting moieties that recognize proteins overexpressed on the extracellular membrane to increase their specific interaction with target cells. Nanobodies (Nbs), the variable domain of heavy chain-only antibodies, are a robust targeting ligand due to their small size, superior stability, and strong binding affinity. For the clinical translation of targeted Nb-NPs, it is essential to understand how the number of Nbs per NP impacts the receptor recognition on cells. To study this, Nbs targeting the hepatocyte growth factor receptor (MET-Nbs) were conjugated to PEGylated liposomes at a density from 20 to 800 per liposome and their targeting efficiency was evaluated in vitro. MET-targeted liposomes (MET-TLs) associated more profoundly with MET-expressing cells than non-targeted liposomes (NTLs). MET-TLs with approximately 150-300 Nbs per liposome exhibited the highest association and specificity towards MET-expressing cells and retained their targeting capacity when pre-incubated with proteins from different sources. Furthermore, a MET-Nb density above 300 Nbs per liposome increased the interaction of MET-TLs with phagocytic cells by 2-fold in ex vivo human blood compared to NTLs. Overall, this study demonstrates that adjusting the MET-Nb density can increase the specificity of NPs towards their intended cellular target and reduce NP interaction with phagocytic cells.


Subject(s)
Nanoparticles , Neoplasms , Single-Domain Antibodies , Humans , Liposomes/chemistry , Ligands
5.
Int J Mol Sci ; 23(24)2022 Dec 10.
Article in English | MEDLINE | ID: mdl-36555306

ABSTRACT

A promise of cancer nanomedicine is the "targeted" delivery of therapeutic agents to tumors by the rational design of nanostructured materials. During the past several decades, a realization that in vitro and in vivo preclinical data are unreliable predictors of successful clinical translation has motivated a reexamination of this approach. Mathematical models of drug pharmacokinetics (PK) and biodistribution (BD) are essential tools for small-molecule drugs development. A key assumption underlying these models is that drug-target binding kinetics dominate blood clearance, hence recognition by host innate immune cells is not explicitly included. Nanoparticles circulating in the blood are conspicuous to phagocytes, and inevitable interactions typically trigger active biological responses to sequester and remove them from circulation. Our recent findings suggest that, instead of referring to nanoparticles as designed for active or passive "tumor targeting", we ought rather to refer to immune cells residing in the tumor microenvironment (TME) as active or passive actors in an essentially "cell-mediated tumor retention" process that competes with active removal by other phagocytes. Indeed, following intravenous injection, nanoparticles induce changes in the immune compartment of the TME because of nanoparticle uptake, irrespective of the nature of tumor targeting moieties. In this study, we propose a 6-compartment PK model as an initial mathematical framework for modeling this tumor-associated immune cell-mediated retention. Published in vivo PK and BD results obtained with bionized nanoferrite® (BNF®) nanoparticles were combined with results from in vitro internalization experiments with murine macrophages to guide simulations. As a preliminary approximation, we assumed that tumor-associated macrophages (TAMs) are solely responsible for active retention in the TME. We model the TAM approximation by relating in vitro macrophage uptake to an effective macrophage avidity term for the BNF® nanoparticles under consideration.


Subject(s)
Nanoparticles , Nanostructures , Neoplasms , Mice , Animals , Tissue Distribution , Macrophages/metabolism , Neoplasms/therapy , Nanoparticles/chemistry , Tumor Microenvironment
6.
Immunology ; 163(3): 239-249, 2021 07.
Article in English | MEDLINE | ID: mdl-33556186

ABSTRACT

Communication between stromal and immune cells is essential to maintain tissue homeostasis, mount an effective immune response and promote tissue repair. This 'crosstalk' occurs in both the steady state and following a variety of insults, for example, in response to local injury, at sites of infection or cancer. What do we mean by crosstalk between cells? Reciprocal activation and/or regulation occurs between immune and stromal cells, by direct cell contact and indirect mechanisms, including the release of soluble cytokines. Moving beyond cell-to-cell contact, this review investigates the complexity of 'cross-space' cellular communication. We highlight different examples of cellular communication by a variety of lung stromal and immune cells following tissue insults. This review examines how the 'geography of the lung microenvironment' is altered in various disease states; more specifically, we investigate how this influences lung epithelial cells and fibroblasts via their communication with immune cells and each other.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epithelial Cells/immunology , Fibroblasts/immunology , Lung/pathology , Stromal Cells/immunology , Animals , Cell Communication , Cellular Microenvironment , Humans , Immunity, Cellular
7.
Exp Cell Res ; 393(2): 112014, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32439494

ABSTRACT

Understanding interactions between immune cells and their targets is an important step on the path to fully characterizing the immune system, and in doing so, learning how it combats disease. Many studies of these interactions have a narrow focus, often looking only at a binary result of whether or not a specific treatment was successful or only focusing on the interactions between two individual cells. Therefore, in an effort to more comprehensively study multicellular interactions among immune cells and their targets, we used in vitro longitudinal time-lapse imaging and developed an automated cell cluster analysis tool, or macro, to investigate the formation of cell clusters. In particular, we investigated the behavior of cancer-specific CD8+ and CD4+ T cells on how they interact around their targets: cancer cells and antigen-presenting cells. The macro that we established allowed us to examine these large-scale clustering behaviors taking place between those four cell types. Thus, we were able to distinguish directed immune cell clustering from random cell movement. Furthermore, this macro can be generalized to be applicable to systems consisting of any number of differently labeled species and can be used to track clustering behaviors and compare them to randomized simulations.


Subject(s)
Cell Communication/physiology , Cell Culture Techniques , Cell Movement/physiology , T-Lymphocytes/cytology , Animals , Cluster Analysis , Mice, Inbred C57BL
8.
Proc Natl Acad Sci U S A ; 113(51): 14805-14810, 2016 12 20.
Article in English | MEDLINE | ID: mdl-27911843

ABSTRACT

The gut microbiota impacts many aspects of host biology including immune function. One hypothesis is that microbial communities induce epigenetic changes with accompanying alterations in chromatin accessibility, providing a mechanism that allows a community to have sustained host effects even in the face of its structural or functional variation. We used Assay for Transposase-Accessible Chromatin with high-throughput sequencing (ATAC-seq) to define chromatin accessibility in predicted enhancer regions of intestinal αß+ and γδ+ intraepithelial lymphocytes purified from germ-free mice, their conventionally raised (CONV-R) counterparts, and mice reared germ free and then colonized with CONV-R gut microbiota at the end of the suckling-weaning transition. Characterizing genes adjacent to traditional enhancers and super-enhancers revealed signaling networks, metabolic pathways, and enhancer-associated transcription factors affected by the microbiota. Our results support the notion that epigenetic modifications help define microbial community-affiliated functional features of host immune cell lineages.


Subject(s)
Enhancer Elements, Genetic , Gastrointestinal Microbiome , Intestines/microbiology , Intraepithelial Lymphocytes/microbiology , Animals , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Lineage , Chromatin/chemistry , Chromatin/metabolism , Germ-Free Life , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction
9.
ACS Nano ; 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39380440

ABSTRACT

Antibody-directed targeting of chemotherapeutic nanoparticles to primary human cancers holds promise for improving efficacy and reducing off-target toxicity. However, clinical responses to targeted nanomedicines are highly variable. Herein, we prepared and examined a matrix of 9 particles (organic and inorganic particles of three surface chemistries with and without antibody functionalization) and developed an ex vivo model to study the person-specific targeting of nanoparticles in whole blood of 15 patients with chronic lymphocytic leukemia (CLL). Generally, anti-CD20-functionalized poly(ethylene glycol) (PEG) nanoparticles efficiently targeted CLL cells, leading to low off-target phagocytosis by granulocytes and monocytes in the blood. However, there was up to 164-fold patient-to-patient variability in the CLL targeting. This was further exemplified through using clinically relevant PEGylated doxorubicin-encapsulated liposomes, which showed high interpersonal differences in CLL targeting (up to 234-fold differences) and off-target phagocytosis (up to 65- and 112-fold differences in granulocytes and monocytes, respectively). Off-target phagocytosis led to almost all monocytes being killed within 24 h of treatment. Variance of the off-target association of PEGylated liposomes with granulocytes and monocytes significantly correlated to anti-PEG immunoglobulin G levels in the blood of CLL patients. A negative correlation between CLL targeting of PEG particles and anti-PEG immunoglobulin M levels was found in the blood. Taken together, our study identifies anti-PEG antibodies as key proteins in modulating patient-specific targeting of PEGylated nanoparticles in human leukemia blood. Other factors, such as the antigen expression of targeted cells and fouling properties of nanoparticles, also play an important role in patient-specific targeting. The human leukemia blood assay we developed provides an ex vivo model to evaluate interpersonal variances in response to targeted nanomedicines.

10.
ACS Nano ; 18(39): 27077-27089, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39298422

ABSTRACT

Lipid nanoparticle mRNA vaccines are an exciting but emerging technology used in humans. There is limited understanding of the factors that influence their biodistribution and immunogenicity. Antibodies to poly(ethylene glycol) (PEG), which is on the surface of the lipid nanoparticle, are detectable in humans and boosted by human mRNA vaccination. We hypothesized that PEG-specific antibodies could increase the clearance of mRNA vaccines. To test this, we developed methods to quantify both the vaccine mRNA and ionizable lipid in frequent serial blood samples from 19 subjects receiving Moderna SPIKEVAX mRNA booster immunization. Both the vaccine mRNA and ionizable lipid peaked in blood 1-2 days post vaccination (median peak level 0.19 and 3.22 ng mL-1, respectively). The vaccine mRNA was detectable and quantifiable up to 14-15 days postvaccination in 37% of subjects. We measured the proportion of vaccine mRNA that was relatively intact in blood over time and found that the decay kinetics of the intact mRNA and ionizable lipid were identical, suggesting the intact lipid nanoparticle recirculates in blood. However, the decay rates of mRNA and ionizable lipids did not correlate with baseline levels of PEG-specific antibodies. Interestingly, the magnitude of mRNA and ionizable lipid detected in blood did correlate with the boost in the level of PEG antibodies. Furthermore, the ability of a subject's monocytes to phagocytose lipid nanoparticles was inversely related to the rise in PEG antibodies. This suggests that the circulation of mRNA lipid nanoparticles into the blood and their clearance by phagocytes influence the PEG immunogenicity of the mRNA vaccines. Overall, this work defines the pharmacokinetics of lipid nanoparticle mRNA vaccine components in human blood after intramuscular injection and the factors that influence these processes. These insights should be valuable in improving the future safety and efficacy of lipid nanoparticle mRNA vaccines and therapeutics.


Subject(s)
COVID-19 Vaccines , Nanoparticles , Humans , Nanoparticles/chemistry , COVID-19 Vaccines/immunology , COVID-19 Vaccines/chemistry , COVID-19 Vaccines/administration & dosage , SARS-CoV-2/immunology , mRNA Vaccines/immunology , Lipids/chemistry , Female , Adult , RNA, Messenger/immunology , RNA, Messenger/genetics , Male , Polyethylene Glycols/chemistry , COVID-19/prevention & control , COVID-19/immunology , Middle Aged , Tissue Distribution , Liposomes
11.
Biomolecules ; 14(8)2024 Aug 11.
Article in English | MEDLINE | ID: mdl-39199375

ABSTRACT

(1) Background: SMARCA4-deficient undifferentiated uterine sarcoma (SDUS) is a rare and aggressive cancer that urgently requires novel therapeutic strategies. Despite the proven efficacy of immunotherapy in various cancer types, its application in SDUS remains largely unexplored. This study aims to investigate the immune microenvironment of SDUS to evaluate the feasibility of utilizing immunotherapy. (2) Methods: Multiplex immunofluorescence (mIF) was employed to examine the immune microenvironment in two cases of SDUS in comparison to other subtypes of endometrial stromal sarcomas (ESSs). This research involved a comprehensive evaluation of immune cell infiltration, cellular interactions, and spatial organization within the tumor immune microenvironment (TiME). Statistical analysis was performed to assess differences in immune cell densities and interactions between SDUS and other ESSs. (3) Results: SDUS exhibited a significantly higher density of cytotoxic T lymphocytes (CTLs), T helper (Th) cells, B cells, and macrophages compared to other ESSs. Notable cellular interactions included Th-CTL and Th-B cell interactions, which were more prominent in SDUS. The spatial analysis revealed distinct immune niches characterized by lymphocyte aggregation and a vascular-rich environment, suggesting an active and engaged immune microenvironment in SDUS. (4) Conclusions: The results suggest that SDUS exhibits a highly immunogenic TiME, characterized by substantial lymphocyte infiltration and dynamic cellular interactions. These findings highlight the potential of immunotherapy as an effective treatment approach for SDUS. However, given the small number of samples evaluated, these conclusions should be drawn with caution. This study underscores the importance of additional investigation into immune-targeted therapies for this challenging cancer subtype, with a larger sample size to validate and expand upon these preliminary findings.


Subject(s)
DNA Helicases , Immunotherapy , Sarcoma , Transcription Factors , Tumor Microenvironment , Uterine Neoplasms , Humans , Female , Tumor Microenvironment/immunology , Immunotherapy/methods , Uterine Neoplasms/therapy , Uterine Neoplasms/immunology , Uterine Neoplasms/pathology , Uterine Neoplasms/genetics , Sarcoma/therapy , Sarcoma/immunology , Sarcoma/genetics , Sarcoma/pathology , Transcription Factors/genetics , DNA Helicases/genetics , DNA Helicases/deficiency , DNA Helicases/immunology , Nuclear Proteins/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/immunology , Middle Aged , Sarcoma, Endometrial Stromal/therapy , Sarcoma, Endometrial Stromal/genetics , Sarcoma, Endometrial Stromal/immunology , Sarcoma, Endometrial Stromal/pathology
12.
Adv Biol (Weinh) ; 7(5): e2200041, 2023 05.
Article in English | MEDLINE | ID: mdl-35751460

ABSTRACT

Blood and lymphatic vessels are regulators of physiological processes, including oxygenation and fluid transport. Both vessels are ubiquitous throughout the body and are critical for sustaining tissue homeostasis. The complexity of each vessel's processes has limited the understanding of exactly how the vessels maintain their functions. Both vessels have been shown to be involved in the pathogenesis of many diseases, including cancer metastasis, and it is crucial to probe further specific mechanisms involved. In vitro models are developed to better understand blood and lymphatic physiological functions and their mechanisms. In this review, blood and lymphatic in vitro model systems, including 2D and 3D designs made using Transwells, microfluidic devices, organoid cultures, and various other methods, are described. Models studying endothelial cell-extracellular matrix interactions, endothelial barrier properties, transendothelial transport and cell migration, lymph/angiogenesis, vascular inflammation, and endothelial-cancer cell interactions are particularly focused. While the field has made significant progress in modeling and understanding lymphatic and blood vasculature, more models that include coculture of multiple cell types, complex extracellular matrix, and 3D morphologies, particularly for models mimicking disease states, will help further the understanding of the role of blood and lymphatic vasculature in health and disease.


Subject(s)
Lymphatic Vessels , Lymphatic Vessels/physiology , Lymphoid Tissue , Cell Movement , Cell Communication
13.
J Leukoc Biol ; 112(3): 499-512, 2022 09.
Article in English | MEDLINE | ID: mdl-35435271

ABSTRACT

Clearance of airway intruders by immune cells is required to resolve infectious pneumonia. However, the molecular mechanisms underlying this process remain elusive. Here, we demonstrated that alveolar macrophage (AM)-derived neuropilin 2 (NRP2) plays an essential role in controlling severe pneumonia by enhancing microbial clearance. Mice with conditional deletion of the NRP2 gene in AM had persistent bacteria, uncontrolled neutrophil influx, and decreased survival during Escherichia coli-induced pneumonia. In vitro assays demonstrated that NRP2 could bind to CD11b+ Ly6Glo/+ neutrophils and promote their capacities in phagocytosis and killing of bacteria, which is partially contributed to the increased expression of TLR4 and TNF-a. These findings collectively revealed that AM-derived NRP2 protects the lungs from unwanted injury by promoting the clearance of invading pathogens. This study might provide a promising diagnostic biomarker and therapeutic target for severe pneumonia.


Subject(s)
Escherichia coli Infections , Macrophages, Alveolar , Neuropilin-2 , Pneumonia, Bacterial , Animals , Escherichia coli Infections/immunology , Lung , Lung Injury , Macrophages, Alveolar/immunology , Mice , Mice, Inbred C57BL , Neuropilin-2/metabolism , Neutrophils , Phagocytosis , Pneumonia, Bacterial/immunology
14.
Pathologie (Heidelb) ; 43(Suppl 1): 21-24, 2022 Aug.
Article in German | MEDLINE | ID: mdl-36222923

ABSTRACT

Malignant neoplasms are highly complex ecosystems consisting of tumor cells and the tumor microenvironment (TME), which is composed of structural elements (vessels, fibroblasts, extracellular matrix) and a wide variety of infiltrating immune cell types of the innate and adaptive immune systems. The TME is the main site of tumor cell-immune cell interactions and plays a critical role in antitumoral immunity. Immunotherapies can affect the interactions between immune cell types and tumor cells in the TME, boost immune responses, and lead to tumor elimination. Novel highly multiplexed microscopy techniques, which enable the detection of more than 50 simultaneous markers in tissues, facilitate the in-depth characterization of the TME at single-cell resolution in clinically relevant samples. Detailed knowledge about the cellular and spatial composition of the TME, the specific cell types and their functional properties, and cell-cell interactions-as revealed by highly multiplexed microscopy-will improve our understanding of immunotherapies' mechanisms of action and reveal new potential therapeutic targets and predictive biomarkers.


Subject(s)
Neoplasms , Tumor Microenvironment , Humans , Microscopy , Ecosystem , Immunotherapy , Neoplasms/therapy
15.
ACS Nano ; 16(2): 2088-2100, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35040637

ABSTRACT

For several decades, surface grafted polyethylene glycol (PEG) has been a go-to strategy for preserving the synthetic identity of liposomes in physiological milieu and preventing clearance by immune cells. However, the limited clinical translation of PEGylated liposomes is mainly due to the protein corona formation and the subsequent modification of liposomes' synthetic identity, which affects their interactions with immune cells and blood residency. Here we exploit the electric charge of DNA to generate unPEGylated liposome/DNA complexes that, upon exposure to human plasma, gets covered with an opsonin-deficient protein corona. The final product of the synthetic process is a biomimetic nanoparticle type covered by a proteonucleotidic corona, or "proteoDNAsome", which maintains its synthetic identity in vivo and is able to slip past the immune system more efficiently than PEGylated liposomes. Accumulation of proteoDNAsomes in the spleen and the liver was lower than that of PEGylated systems. Our work highlights the importance of generating stable biomolecular coronas in the development of stealth unPEGylated particles, thus providing a connection between the biological behavior of particles in vivo and their synthetic identity.


Subject(s)
Liposomes , Protein Corona , Humans , Opsonin Proteins , Polyethylene Glycols
16.
ACS Nano ; 16(8): 11769-11780, 2022 08 23.
Article in English | MEDLINE | ID: mdl-35758934

ABSTRACT

Humans commonly have low level antibodies to poly(ethylene) glycol (PEG) due to environmental exposure. Lipid nanoparticle (LNP) mRNA vaccines for SARS-CoV-2 contain small amounts of PEG, but it is not known whether PEG antibodies are enhanced by vaccination and what their impact is on particle-immune cell interactions in human blood. We studied plasma from 130 adults receiving either the BNT162b2 (Pfizer-BioNTech) or mRNA-1273 (Moderna) mRNA vaccines or no SARS-CoV-2 vaccine for PEG-specific antibodies. Anti-PEG IgG was commonly detected prior to vaccination and was significantly boosted a mean of 13.1-fold (range 1.0-70.9) following mRNA-1273 vaccination and a mean of 1.78-fold (range 0.68-16.6) following BNT162b2 vaccination. Anti-PEG IgM increased 68.5-fold (range 0.9-377.1) and 2.64-fold (0.76-12.84) following mRNA-1273 and BNT162b2 vaccination, respectively. The rise in PEG-specific antibodies following mRNA-1273 vaccination was associated with a significant increase in the association of clinically relevant PEGylated LNPs with blood phagocytes ex vivo. PEG antibodies did not impact the SARS-CoV-2 specific neutralizing antibody response to vaccination. However, the elevated levels of vaccine-induced anti-PEG antibodies correlated with increased systemic reactogenicity following two doses of vaccination. We conclude that PEG-specific antibodies can be boosted by LNP mRNA vaccination and that the rise in PEG-specific antibodies is associated with systemic reactogenicity and an increase of PEG particle-leukocyte association in human blood. The longer-term clinical impact of the increase in PEG-specific antibodies induced by lipid nanoparticle mRNA vaccines should be monitored. It may be useful to identify suitable alternatives to PEG for developing next-generation LNP vaccines to overcome PEG immunogenicity in the future.


Subject(s)
COVID-19 Vaccines , COVID-19 , Adult , Humans , BNT162 Vaccine , SARS-CoV-2 , COVID-19/prevention & control , Polyethylene Glycols , Antibodies , Vaccination , Antibodies, Viral , Antibodies, Neutralizing , mRNA Vaccines
17.
World J Gastrointest Oncol ; 14(9): 1808-1822, 2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36187404

ABSTRACT

BACKGROUND: Gastric cancer (GC) is one of the most common malignancies in China with a high morbidity and mortality. AIM: To determine whether interleukin (IL)-31, IL-32, and IL-33 can be used as biomarkers for the detection of GC, via evaluating the correlations between their expression and clinicopathological parameters of GC patients. METHODS: Tissue array (n = 180) gastric specimens were utilised. IL-31, IL-32, and IL-33 expression in GC and non-GC tissues was detected immunohistochemically. The correlations between IL-31, IL-32, and IL-33 expression in GC and severity of clinicopathological parameters were evaluated. Survival curves were plotted using the Kaplan-Meier method/Cox regression. Circulating IL-31, IL-32, and IL-33 were detected by ELISA. RESULTS: We found that the expression levels of IL-31, IL-32, and IL-33 were all lower in GC than in adjacent non-GC gastric tissues (P < 0.05). IL-33 in peripheral blood of GC patients was significantly lower than that of healthy individuals (1.50 ± 1.11 vs 9.61 ± 8.00 ng/mL, P <0.05). Decreased IL-31, IL-32, and IL-33 in GC were observed in younger patients (< 60 years), and IL-32 and IL-33 were lower in female patients (P < 0.05). Higher IL-32 correlated with a longer survival in two GC subgroups: T4 invasion depth and TNM I-II stage. Univariate/multivariate analysis revealed that IL-32 was an independent prognostic factor for GC in the T4 stage subgroup. Circulating IL-33 was significantly lower in GC patients at TNM stage IV than in healthy people (P < 0.05). CONCLUSION: Our findings may provide new insights into the roles of IL-31, IL-32, and IL-33 in the carcinogenesis of GC and demonstrate their relative usefulness as prognostic markers for GC. The underlying mechanism of IL-31, IL-32, and IL-33 actions in GC should be further explored.

18.
Mil Med Res ; 8(1): 45, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34389057

ABSTRACT

Single-cell RNA sequencing (scRNA-seq) is a comprehensive technical tool to analyze intracellular and intercellular interaction data by whole transcriptional profile analysis. Here, we describe the application in biomedical research, focusing on the immune system during organ transplantation and rejection. Unlike conventional transcriptome analysis, this method provides a full map of multiple cell populations in one specific tissue and presents a dynamic and transient unbiased method to explore the progression of allograft dysfunction, starting from the stress response to final graft failure. This promising sequencing technology remarkably improves individualized organ rejection treatment by identifying decisive cellular subgroups and cell-specific interactions.


Subject(s)
Organ Transplantation/instrumentation , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Gene Expression Profiling/methods , Humans , Organ Transplantation/methods , Sequence Analysis, RNA/instrumentation , Single-Cell Analysis/instrumentation
19.
Methods Mol Biol ; 2055: 595-638, 2020.
Article in English | MEDLINE | ID: mdl-31502171

ABSTRACT

Humans are living ecosystems composed of human cells and microbes. The microbiome is the collection of microbes (microbiota) and their genes. Recent breakthroughs in the high-throughput sequencing technologies have made it possible for us to understand the composition of the human microbiome. Launched by the National Institutes of Health in USA, the human microbiome project indicated that our bodies harbor a wide array of microbes, specific to each body site with interpersonal and intrapersonal variabilities. Numerous studies have indicated that several factors influence the development of the microbiome including genetics, diet, use of antibiotics, and lifestyle, among others. The microbiome and its mediators are in a continuous cross talk with the host immune system; hence, any imbalance on one side is reflected on the other. Dysbiosis (microbiota imbalance) was shown in many diseases and pathological conditions such as inflammatory bowel disease, celiac disease, multiple sclerosis, rheumatoid arthritis, asthma, diabetes, and cancer. The microbial composition mirrors inflammation variations in certain disease conditions, within various stages of the same disease; hence, it has the potential to be used as a biomarker.


Subject(s)
Bacteria/classification , Bacterial Proteins/genetics , Metagenomics/methods , Age Factors , Aged , Bacteria/genetics , Bacteria/isolation & purification , Gene Expression Profiling , Gene Expression Regulation, Bacterial , High-Throughput Nucleotide Sequencing , Humans , Microbiota , Middle Aged , Sequence Analysis, DNA
20.
ACS Nano ; 14(11): 15723-15737, 2020 11 24.
Article in English | MEDLINE | ID: mdl-33112593

ABSTRACT

When nanoparticles interact with human blood, a multitude of plasma components adsorb onto the surface of the nanoparticles, forming a biomolecular corona. Corona composition is known to be influenced by the chemical composition of nanoparticles. In contrast, the possible effects of variations in the human blood proteome between healthy individuals on the formation of the corona and its subsequent interactions with immune cells in blood are unknown. Herein, we prepared and examined a matrix of 11 particles (including organic and inorganic particles of three sizes and five surface chemistries) and plasma samples from 23 healthy donors to form donor-specific biomolecular coronas (personalized coronas) and investigated the impact of the personalized coronas on particle interactions with immune cells in human blood. Among the particles examined, poly(ethylene glycol) (PEG)-coated mesoporous silica (MS) particles, irrespective of particle size (800, 450, or 100 nm in diameter), displayed the widest range (up to 60-fold difference) of donor-dependent variance in immune cell association. In contrast, PEG particles (after MS core removal) of 860, 518, or 133 nm in diameter displayed consistent stealth behavior (negligible cell association), irrespective of plasma donor. For comparison, clinically relevant PEGylated doxorubicin-encapsulated liposomes (Doxil) (74 nm in diameter) showed significant variance in association with monocytes and B cells across all plasma donors studied. An in-depth proteomic analysis of each biomolecular corona studied was performed, and the results were compared against the nanoparticle-blood cell association results, with individual variance in the proteome driving differential association with specific immune cell types. We identified key immunoglobulin and complement proteins that explicitly enriched or depleted within the corona and which strongly correlated with the cell association pattern observed across the 23 donors. This study demonstrates how plasma variance in healthy individuals significantly influences the blood immune cell interactions of nanoparticles.


Subject(s)
Nanoparticles , Protein Corona , Blood Proteins , Humans , Particle Size , Proteomics , Silicon Dioxide
SELECTION OF CITATIONS
SEARCH DETAIL