Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Immunity ; 56(8): 1910-1926.e7, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37478854

ABSTRACT

Highly effective vaccines elicit specific, robust, and durable adaptive immune responses. To advance informed vaccine design, it is critical that we understand the cellular dynamics underlying responses to different antigen formats. Here, we sought to understand how antigen-specific B and T cells were activated and participated in adaptive immune responses within the mucosal site. Using a human tonsil organoid model, we tracked the differentiation and kinetics of the adaptive immune response to influenza vaccine and virus modalities. Each antigen format elicited distinct B and T cell responses, including differences in their magnitude, diversity, phenotype, function, and breadth. These differences culminated in substantial changes in the corresponding antibody response. A major source of antigen format-related variability was the ability to recruit naive vs. memory B and T cells to the response. These findings have important implications for vaccine design and the generation of protective immune responses in the upper respiratory tract.


Subject(s)
Influenza Vaccines , Influenza, Human , Humans , Antibody Formation , Antibodies, Viral , T-Lymphocytes , Antigens , Organoids
2.
Trends Immunol ; 44(12): 938-944, 2023 12.
Article in English | MEDLINE | ID: mdl-37940395

ABSTRACT

Current influenza A and B virus (IABV) vaccines provide suboptimal protection and efforts are underway to develop a universal IABV vaccine. Blood neutralizing antibodies are the current gold standard for protection, but many processes that regulate human IABV-specific immunity occur in mucosal and lymphoid tissues. We need an improved mechanistic understanding of how immune cells respond within these tissues to advance our current (slow and expensive) vaccine testing model. We posit that advanced in vitro models of human adaptive immunity can bridge some of the gaps between vaccine design, animal models, and human clinical trials. Here, we highlight how they can be integrated into current practices and play a role in reverse translating the defined features of protective vaccines to rationally design new candidates.


Subject(s)
Influenza Vaccines , Influenza, Human , Animals , Humans , Antibodies, Viral , Antibodies, Neutralizing , Adaptive Immunity , Organoids , Influenza, Human/prevention & control
3.
Mol Ther ; 28(1): 189-201, 2020 01 08.
Article in English | MEDLINE | ID: mdl-31628052

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked muscle wasting disease that is caused by the loss of functional dystrophin protein in cardiac and skeletal muscles. DMD patient muscles become weakened, leading to eventual myofiber breakdown and replacement with fibrotic and adipose tissues. Inflammation drives the pathogenic processes through releasing inflammatory cytokines and other factors that promote skeletal muscle degeneration and contributing to the loss of motor function. Selective inhibitors of nuclear export (SINEs) are a class of compounds that function by inhibiting the nuclear export protein exportin 1 (XPO1). The XPO1 protein is an important regulator of key inflammatory and neurological factors that drive inflammation and neurotoxicity in various neurological and neuromuscular diseases. Here, we demonstrate that SINE compound KPT-350 can ameliorate dystrophic-associated pathologies in the muscles of DMD models of zebrafish and mice. Thus, SINE compounds are a promising novel strategy for blocking dystrophic symptoms and could be used in combinatorial treatments for DMD.


Subject(s)
Active Transport, Cell Nucleus/drug effects , Karyopherins/antagonists & inhibitors , Muscular Dystrophy, Duchenne/drug therapy , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Zebrafish/genetics , Administration, Oral , Animals , Biomarkers/blood , Cytokines/antagonists & inhibitors , Cytokines/blood , Disease Models, Animal , Locomotion/drug effects , Macrophages/drug effects , Membrane Proteins/genetics , Mice , Mice, Inbred DBA , Mice, Inbred mdx , Muscle Proteins/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Mutation , Zebrafish Proteins/genetics , Exportin 1 Protein
4.
J Immunol ; 199(9): 3326-3335, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28978694

ABSTRACT

We describe a novel B cell-associated cytokine, encoded by an uncharacterized gene (C17orf99; chromosome 17 open reading frame 99), that is expressed in bone marrow and fetal liver and whose expression is also induced in peripheral B cells upon activation. C17orf99 is only present in mammalian genomes, and it encodes a small (∼27-kDa) secreted protein unrelated to other cytokine families, suggesting a function in mammalian immune responses. Accordingly, C17orf99 expression is induced in the mammary gland upon the onset of lactation, and a C17orf99-/- mouse exhibits reduced levels of IgA in the serum, gut, feces, and lactating mammary gland. C17orf99-/- mice have smaller and fewer Peyer's patches and lower numbers of IgA-secreting cells. The microbiome of C17orf99-/- mice exhibits altered composition, likely a consequence of the reduced levels of IgA in the gut. Although naive B cells can express C17orf99 upon activation, their production increases following culture with various cytokines, including IL-4 and TGF-ß1, suggesting that differentiation can result in the expansion of C17orf99-producing B cells during some immune responses. Taken together, these observations indicate that C17orf99 encodes a novel B cell-associated cytokine, which we have called IL-40, that plays an important role in humoral immune responses and may also play a role in B cell development. Importantly, IL-40 is also expressed by human activated B cells and by several human B cell lymphomas. The latter observations suggest that it may play a role in the pathogenesis of certain human diseases.


Subject(s)
B-Lymphocytes/immunology , Gene Expression Regulation/immunology , Interleukins/immunology , Peyer's Patches/immunology , Animals , Humans , Immunoglobulin A/immunology , Interleukins/genetics , Jurkat Cells , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/immunology , Mice , Mice, Knockout
5.
Cell Stem Cell ; 31(3): 410-420.e4, 2024 03 07.
Article in English | MEDLINE | ID: mdl-38402619

ABSTRACT

Heterogeneity in the tumor microenvironment (TME) of follicular lymphomas (FLs) can affect clinical outcomes. Current immunotherapeutic strategies, including antibody- and cell-based therapies, variably overcome pro-tumorigenic mechanisms for sustained disease control. Modeling the intact FL TME, with its native, syngeneic tumor-infiltrating leukocytes, is a major challenge. Here, we describe an organoid culture method for cultivating patient-derived lymphoma organoids (PDLOs), which include cells from the native FL TME. We define the robustness of this method by successfully culturing cryopreserved FL specimens from diverse patients and demonstrate the stability of TME cellular composition, tumor somatic mutations, gene expression profiles, and B/T cell receptor dynamics over 3 weeks. PDLOs treated with CD3:CD19 and CD3:CD20 therapeutic bispecific antibodies showed B cell killing and T cell activation. This stable system offers a robust platform for advancing precision medicine efforts in FL through patient-specific modeling, high-throughput screening, TME signature identification, and treatment response evaluation.


Subject(s)
Lymphoma, Follicular , Humans , Lymphoma, Follicular/therapy , Lymphoma, Follicular/diagnosis , Lymphoma, Follicular/genetics , Tumor Microenvironment , B-Lymphocytes , Receptors, Antigen, T-Cell , Organoids
6.
Front Immunol ; 15: 1373537, 2024.
Article in English | MEDLINE | ID: mdl-38812520

ABSTRACT

Sex-based differences in immune cell composition and function can contribute to distinct adaptive immune responses. Prior work has quantified these differences in peripheral blood, but little is known about sex differences within human lymphoid tissues. Here, we characterized the composition and phenotypes of adaptive immune cells from male and female ex vivo tonsils and evaluated their responses to influenza antigens using an immune organoid approach. In a pediatric cohort, female tonsils had more memory B cells compared to male tonsils direct ex vivo and after stimulation with live-attenuated but not inactivated vaccine, produced higher influenza-specific antibody responses. Sex biases were also observed in adult tonsils but were different from those measured in children. Analysis of peripheral blood immune cells from in vivo vaccinated adults also showed higher frequencies of tissue homing CD4 T cells in female participants. Together, our data demonstrate that distinct memory B and T cell profiles are present in male vs. female lymphoid tissues and peripheral blood respectively and suggest that these differences may in part explain sex biases in response to vaccines and viruses.


Subject(s)
Palatine Tonsil , Humans , Female , Male , Child , Palatine Tonsil/immunology , Adult , Influenza Vaccines/immunology , Influenza, Human/immunology , Sex Characteristics , Child, Preschool , Adolescent , Antibodies, Viral/blood , Antibodies, Viral/immunology , Memory B Cells/immunology , Organ Specificity/immunology , Young Adult , Sex Factors , CD4-Positive T-Lymphocytes/immunology , B-Lymphocytes/immunology , Immunologic Memory
7.
Sci Adv ; 9(27): eadd9984, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37418531

ABSTRACT

Macrophages are essential for skeletal muscle homeostasis, but how their dysregulation contributes to the development of fibrosis in muscle disease remains unclear. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six clusters and unexpectedly found that none corresponded to traditional definitions of M1 or M2 macrophages. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 (gal-3) and osteopontin (Spp1). Spatial transcriptomics, computational inferences of intercellular communication, and in vitro assays indicated that macrophage-derived Spp1 regulates stromal progenitor differentiation. Gal-3+ macrophages were chronically activated in dystrophic muscle, and adoptive transfer assays showed that the gal-3+ phenotype was the dominant molecular program induced within the dystrophic milieu. Gal-3+ macrophages were also elevated in multiple human myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining their transcriptional programs and reveal Spp1 as a major regulator of macrophage and stromal progenitor interactions.


Subject(s)
Macrophages , Transcriptome , Mice , Animals , Humans , Mice, Inbred C57BL , Macrophages/metabolism , Muscle, Skeletal/metabolism , Galectin 3/genetics , Galectin 3/metabolism , Fibrosis
8.
bioRxiv ; 2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37131694

ABSTRACT

The monocytic/macrophage system is essential for skeletal muscle homeostasis, but its dysregulation contributes to the pathogenesis of muscle degenerative disorders. Despite our increasing knowledge of the role of macrophages in degenerative disease, it still remains unclear how macrophages contribute to muscle fibrosis. Here, we used single-cell transcriptomics to determine the molecular attributes of dystrophic and healthy muscle macrophages. We identified six novel clusters. Unexpectedly, none corresponded to traditional definitions of M1 or M2 macrophage activation. Rather, the predominant macrophage signature in dystrophic muscle was characterized by high expression of fibrotic factors, galectin-3 and spp1. Spatial transcriptomics and computational inferences of intercellular communication indicated that spp1 regulates stromal progenitor and macrophage interactions during muscular dystrophy. Galectin-3 + macrophages were chronically activated in dystrophic muscle and adoptive transfer assays showed that the galectin-3 + phenotype was the dominant molecular program induced within the dystrophic milieu. Histological examination of human muscle biopsies revealed that galectin-3 + macrophages were also elevated in multiple myopathies. These studies advance our understanding of macrophages in muscular dystrophy by defining the transcriptional programs induced in muscle macrophages, and reveal spp1 as a major regulator of macrophage and stromal progenitor interactions.

9.
Sci Transl Med ; 14(628): eabi9196, 2022 01 19.
Article in English | MEDLINE | ID: mdl-35044790

ABSTRACT

Sporadic inclusion body myositis (IBM) is the most common acquired muscle disease in adults over age 50, yet it remains unclear whether the disease is primarily driven by T cell­mediated autoimmunity. IBM muscle biopsies display nuclear clearance and cytoplasmic aggregation of TDP-43 in muscle cells, a pathologic finding observed initially in neurodegenerative diseases, where nuclear loss of TDP-43 in neurons causes aberrant RNA splicing. Here, we show that loss of TDP-43­mediated splicing repression, as determined by inclusion of cryptic exons, occurs in skeletal muscle of subjects with IBM. Of 119 muscle biopsies tested, RT-PCR­mediated detection of cryptic exon inclusion was able to diagnose IBM with 84% sensitivity and 99% specificity. To determine the role of T cells in pathogenesis, we generated a xenograft model by transplanting human IBM muscle into the hindlimb of immunodeficient mice. Xenografts from subjects with IBM displayed robust regeneration of human myofibers and recapitulated both inflammatory and degenerative features of the disease. Myofibers in IBM xenografts showed invasion by human, oligoclonal CD8+ T cells and exhibited MHC-I up-regulation, rimmed vacuoles, mitochondrial pathology, p62-positive inclusions, and nuclear clearance and cytoplasmic aggregation of TDP-43, associated with cryptic exon inclusion. Reduction of human T cells within IBM xenografts by treating mice intraperitoneally with anti-CD3 (OKT3) suppressed MHC-I up-regulation. However, rimmed vacuoles and loss of TDP-43 function persisted. These data suggest that T cell depletion does not alter muscle degenerative pathology in IBM.


Subject(s)
DNA-Binding Proteins/metabolism , Myositis, Inclusion Body , Myositis , Animals , CD8-Positive T-Lymphocytes , DNA-Binding Proteins/genetics , Heterografts , Humans , Mice , Muscle, Skeletal/pathology , Myositis/diagnosis , Myositis/pathology , Myositis, Inclusion Body/diagnosis , Myositis, Inclusion Body/pathology , Vacuoles/pathology
10.
Cell Rep ; 35(2): 108997, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852849

ABSTRACT

Despite the well-accepted view that chronic inflammation contributes to the pathogenesis of Duchenne muscular dystrophy (DMD), the function and regulation of eosinophils remain an unclear facet of type II innate immunity in dystrophic muscle. We report the observation that group 2 innate lymphoid cells (ILC2s) are present in skeletal muscle and are the principal regulators of muscle eosinophils during muscular dystrophy. Eosinophils were elevated in DMD patients and dystrophic mice along with interleukin (IL)-5, a major eosinophil survival factor that was predominantly expressed by muscle ILC2s. We also find that IL-33 was upregulated in dystrophic muscle and was predominantly produced by fibrogenic/adipogenic progenitors (FAPs). Exogenous IL-33 and IL-2 complex (IL-2c) expanded muscle ILC2s and eosinophils, decreased the cross-sectional area (CSA) of regenerating myofibers, and increased the expression of genes associated with muscle fibrosis. The deletion of ILC2s in dystrophic mice mitigated muscle eosinophilia and impaired the induction of IL-5 and fibrosis-associated genes. Our findings highlight a FAP/ILC2/eosinophil axis that promotes type II innate immunity, which influences the balance between regenerative and fibrotic responses during muscular dystrophy.


Subject(s)
Eosinophils/immunology , Fibroblasts/immunology , Interleukin-5/immunology , Lymphocytes/immunology , Mesenchymal Stem Cells/immunology , Muscular Dystrophy, Duchenne/immunology , Animals , Cell Proliferation , Chemokines, CC/genetics , Chemokines, CC/immunology , Eosinophils/drug effects , Eosinophils/pathology , Fibroblasts/drug effects , Fibroblasts/pathology , Fibrosis , Gene Expression , Gene Expression Profiling , Humans , Immunity, Innate , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-33/immunology , Interleukin-33/pharmacology , Interleukin-5/genetics , Intestines/drug effects , Intestines/immunology , Intestines/pathology , Lung/drug effects , Lung/immunology , Lung/pathology , Lymphocytes/drug effects , Lymphocytes/pathology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred mdx , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology
11.
Cell Host Microbe ; 28(6): 769-770, 2020 12 09.
Article in English | MEDLINE | ID: mdl-33301713

ABSTRACT

Antibodies are typically thought to mediate antiviral immunity by blocking host-cell infection. However, immunoglobulin G (IgG) can also stimulate antiviral responses through its constant region. A recent study in Nature (Bournazos et al., 2020) shows that modifying Fc binding affinities to better target FcÉ£RIIa enhances the influenza CD8 T cell response.


Subject(s)
Influenza, Human , Orthomyxoviridae , Antibodies, Viral , CD8-Positive T-Lymphocytes , Humans , Immunoglobulin Fc Fragments
12.
Front Physiol ; 10: 1416, 2019.
Article in English | MEDLINE | ID: mdl-31849692

ABSTRACT

Skeletal muscle injury provokes a regenerative response, characterized by the de novo generation of myofibers that are distinguished by central nucleation and re-expression of developmentally restricted genes. In addition to these characteristics, myofiber cross-sectional area (CSA) is widely used to evaluate muscle hypertrophic and regenerative responses. Here, we introduce QuantiMus, a free software program that uses machine learning algorithms to quantify muscle morphology and molecular features with high precision and quick processing-time. The ability of QuantiMus to define and measure myofibers was compared to manual measurement or other automated software programs. QuantiMus rapidly and accurately defined total myofibers and measured CSA with comparable performance but quantified the CSA of centrally-nucleated fibers (CNFs) with greater precision compared to other software. It additionally quantified the fluorescence intensity of individual myofibers of human and mouse muscle, which was used to assess the distribution of myofiber type, based on the myosin heavy chain isoform that was expressed. Furthermore, analysis of entire quadriceps cross-sections of healthy and mdx mice showed that dystrophic muscle had an increased frequency of Evans blue dye+ injured myofibers. QuantiMus also revealed that the proportion of centrally nucleated, regenerating myofibers that express embryonic myosin heavy chain (eMyHC) or neural cell adhesion molecule (NCAM) were increased in dystrophic mice. Our findings reveal that QuantiMus has several advantages over existing software. The unique self-learning capacity of the machine learning algorithms provides superior accuracy and the ability to rapidly interrogate the complete muscle section. These qualities increase rigor and reproducibility by avoiding methods that rely on the sampling of representative areas of a section. This is of particular importance for the analysis of dystrophic muscle given the "patchy" distribution of muscle pathology. QuantiMus is an open source tool, allowing customization to meet investigator-specific needs and provides novel analytical approaches for quantifying muscle morphology.

13.
Methods Mol Biol ; 1687: 43-56, 2018.
Article in English | MEDLINE | ID: mdl-29067655

ABSTRACT

Although mutations of the dystrophin gene are the causative defect in Duchenne muscular dystrophy (DMD) patients, secondary disease processes such as inflammation contribute greatly to the pathogenesis of DMD. Genetic and histological studies have shown that distinct facets of the immune system promote muscle degeneration or regeneration during muscular dystrophy through mechanisms that are only beginning to be defined. Although histological methods have allowed the enumeration and localization of immune cells within dystrophic muscle, they are limited in their ability to assess the full spectrum of phenotypic states of an immune cell population and its functional characteristics. This chapter highlights flow cytometry methods for the isolation and functional study of immune cell populations from muscle of the mdx mouse model of DMD. We include a detailed description of preparing single-cell suspensions of dystrophic muscle that maintain the integrity of cell-surface markers used to identify macrophages, eosinophils, group 2 innate lymphoid cells, and regulatory T cells. This method complements the battery of histological assays that are currently used to study the role of inflammation in muscular dystrophy, and provides a platform capable of being integrated with multiple downstream methodologies for the mechanistic study of immunity in muscle degenerative diseases.


Subject(s)
Flow Cytometry/methods , Inflammation/diagnosis , Muscle, Skeletal/diagnostic imaging , Muscular Dystrophy, Duchenne/diagnosis , Animals , Disease Models, Animal , Dystrophin/genetics , Humans , Inflammation/diagnostic imaging , Inflammation/genetics , Inflammation/pathology , Macrophages/pathology , Mice , Mice, Inbred mdx , Muscular Dystrophy, Duchenne/diagnostic imaging , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology
SELECTION OF CITATIONS
SEARCH DETAIL