Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters

Affiliation country
Publication year range
1.
Nat Immunol ; 20(12): 1692-1699, 2019 12.
Article in English | MEDLINE | ID: mdl-31745340

ABSTRACT

High-throughput 3' single-cell RNA-sequencing (scRNA-seq) allows cost-effective, detailed characterization of individual immune cells from tissues. Current techniques, however, are limited in their ability to elucidate essential immune cell features, including variable sequences of T cell antigen receptors (TCRs) that confer antigen specificity. Here, we present a strategy that enables simultaneous analysis of TCR sequences and corresponding full transcriptomes from 3'-barcoded scRNA-seq samples. This approach is compatible with common 3' scRNA-seq methods, and adaptable to processed samples post hoc. We applied the technique to identify transcriptional signatures associated with T cells sharing common TCRs from immunized mice and from patients with food allergy. We observed preferential phenotypes among subsets of expanded clonotypes, including type 2 helper CD4+ T cell (TH2) states associated with food allergy. These results demonstrate the utility of our method when studying diseases in which clonotype-driven responses are critical to understanding the underlying biology.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Peanut Hypersensitivity/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Th2 Cells/immunology , 2S Albumins, Plant/immunology , Animals , Antigens, Plant/immunology , Cells, Cultured , Complementarity Determining Regions/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Immunization , Immunoglobulin E/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Papillomavirus E7 Proteins/immunology , Single-Cell Analysis , T-Cell Antigen Receptor Specificity/genetics , Transcriptome
2.
Immunity ; 55(5): 827-846.e10, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35483355

ABSTRACT

Mycobacterium tuberculosis lung infection results in a complex multicellular structure: the granuloma. In some granulomas, immune activity promotes bacterial clearance, but in others, bacteria persist and grow. We identified correlates of bacterial control in cynomolgus macaque lung granulomas by co-registering longitudinal positron emission tomography and computed tomography imaging, single-cell RNA sequencing, and measures of bacterial clearance. Bacterial persistence occurred in granulomas enriched for mast, endothelial, fibroblast, and plasma cells, signaling amongst themselves via type 2 immunity and wound-healing pathways. Granulomas that drove bacterial control were characterized by cellular ecosystems enriched for type 1-type 17, stem-like, and cytotoxic T cells engaged in pro-inflammatory signaling networks involving diverse cell populations. Granulomas that arose later in infection displayed functional characteristics of restrictive granulomas and were more capable of killing Mtb. Our results define the complex multicellular ecosystems underlying (lack of) granuloma resolution and highlight host immune targets that can be leveraged to develop new vaccine and therapeutic strategies for TB.


Subject(s)
Mycobacterium tuberculosis , Pulmonary Fibrosis , Tuberculosis , Animals , Ecosystem , Granuloma , Lung , Macaca fascicularis , Pulmonary Fibrosis/pathology
3.
Immunity ; 53(4): 878-894.e7, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053333

ABSTRACT

High-throughput single-cell RNA-sequencing (scRNA-seq) methodologies enable characterization of complex biological samples by increasing the number of cells that can be profiled contemporaneously. Nevertheless, these approaches recover less information per cell than low-throughput strategies. To accurately report the expression of key phenotypic features of cells, scRNA-seq platforms are needed that are both high fidelity and high throughput. To address this need, we created Seq-Well S3 ("Second-Strand Synthesis"), a massively parallel scRNA-seq protocol that uses a randomly primed second-strand synthesis to recover complementary DNA (cDNA) molecules that were successfully reverse transcribed but to which a second oligonucleotide handle, necessary for subsequent whole transcriptome amplification, was not appended due to inefficient template switching. Seq-Well S3 increased the efficiency of transcript capture and gene detection compared with that of previous iterations by up to 10- and 5-fold, respectively. We used Seq-Well S3 to chart the transcriptional landscape of five human inflammatory skin diseases, thus providing a resource for the further study of human skin inflammation.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Inflammation/genetics , RNA, Small Cytoplasmic/genetics , Skin/pathology , Animals , Cell Line , DNA, Complementary/genetics , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcription, Genetic/genetics , Transcriptome/genetics
4.
Nat Immunol ; 15(2): 128-35, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24448570

ABSTRACT

The complex heterogeneity of cells, and their interconnectedness with each other, are major challenges to identifying clinically relevant measurements that reflect the state and capability of the immune system. Highly multiplexed, single-cell technologies may be critical for identifying correlates of disease or immunological interventions as well as for elucidating the underlying mechanisms of immunity. Here we review limitations of bulk measurements and explore advances in single-cell technologies that overcome these problems by expanding the depth and breadth of functional and phenotypic analysis in space and time. The geometric increases in complexity of data make formidable hurdles for exploring, analyzing and presenting results. We summarize recent approaches to making such computations tractable and discuss challenges for integrating heterogeneous data obtained using these single-cell technologies.


Subject(s)
Immune System/metabolism , Immunologic Techniques , Monitoring, Immunologic/methods , Single-Cell Analysis/methods , Animals , Computational Biology , Humans , Immune System/pathology , Statistics as Topic
5.
Nat Methods ; 14(4): 395-398, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28192419

ABSTRACT

Single-cell RNA-seq can precisely resolve cellular states, but applying this method to low-input samples is challenging. Here, we present Seq-Well, a portable, low-cost platform for massively parallel single-cell RNA-seq. Barcoded mRNA capture beads and single cells are sealed in an array of subnanoliter wells using a semipermeable membrane, enabling efficient cell lysis and transcript capture. We use Seq-Well to profile thousands of primary human macrophages exposed to Mycobacterium tuberculosis.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , 3T3 Cells , Animals , HEK293 Cells , High-Throughput Nucleotide Sequencing/economics , High-Throughput Nucleotide Sequencing/instrumentation , Humans , Leukocytes, Mononuclear/physiology , Macrophages/microbiology , Macrophages/physiology , Mice , Mycobacterium tuberculosis/pathogenicity , RNA, Messenger/genetics , Sequence Analysis, RNA/economics , Sequence Analysis, RNA/instrumentation , Single-Cell Analysis/economics , Single-Cell Analysis/instrumentation
6.
J Neurosci ; 36(5): 1730-46, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26843653

ABSTRACT

Secreted factors play a central role in normal and pathological processes in every tissue in the body. The brain is composed of a highly complex milieu of different cell types and few methods exist that can identify which individual cells in a complex mixture are secreting specific analytes. By identifying which cells are responsible, we can better understand neural physiology and pathophysiology, more readily identify the underlying pathways responsible for analyte production, and ultimately use this information to guide the development of novel therapeutic strategies that target the cell types of relevance. We present here a method for detecting analytes secreted from single human induced pluripotent stem cell (iPSC)-derived neural cells and have applied the method to measure amyloid ß (Aß) and soluble amyloid precursor protein-alpha (sAPPα), analytes central to Alzheimer's disease pathogenesis. Through these studies, we have uncovered the dynamic range of secretion profiles of these analytes from single iPSC-derived neuronal and glial cells and have molecularly characterized subpopulations of these cells through immunostaining and gene expression analyses. In examining Aß and sAPPα secretion from single cells, we were able to identify previously unappreciated complexities in the biology of APP cleavage that could not otherwise have been found by studying averaged responses over pools of cells. This technique can be readily adapted to the detection of other analytes secreted by neural cells, which would have the potential to open new perspectives into human CNS development and dysfunction. SIGNIFICANCE STATEMENT: We have established a technology that, for the first time, detects secreted analytes from single human neurons and astrocytes. We examine secretion of the Alzheimer's disease-relevant factors amyloid ß (Aß) and soluble amyloid precursor protein-alpha (sAPPα) and present novel findings that could not have been observed without a single-cell analytical platform. First, we identify a previously unappreciated subpopulation that secretes high levels of Aß in the absence of detectable sAPPα. Further, we show that multiple cell types secrete high levels of Aß and sAPPα, but cells expressing GABAergic neuronal markers are overrepresented. Finally, we show that astrocytes are competent to secrete high levels of Aß and therefore may be a significant contributor to Aß accumulation in the brain.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Astrocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , Single-Cell Analysis/methods , Amyloid beta-Peptides/analysis , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/analysis , Animals , Astrocytes/chemistry , CHO Cells , Cricetinae , Cricetulus , Female , Humans , Induced Pluripotent Stem Cells/chemistry , Male , Neurons/chemistry
7.
J Virol ; 90(19): 8875-90, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27466419

ABSTRACT

UNLABELLED: An effective preventive vaccine is highly sought after in order to stem the current HIV-1 pandemic. Both conservation of contiguous gp41 membrane-proximal external region (MPER) amino acid sequences across HIV-1 clades and the ability of anti-MPER broadly neutralizing antibodies (BNAbs) to block viral hemifusion/fusion establish the MPER as a prime vaccination target. In earlier studies, we described the development of an MPER vaccine formulation that takes advantage of liposomes to array the MPER on a lipid bilayer surface, paralleling its native configuration on the virus membrane while also incorporating molecular adjuvant and CD4 T cell epitope cargo. Here we demonstrate that several immunizations with MPER/liposomes induce high levels of bone marrow long-lived plasma cell (LLPC) antibody production. Single-cell immunoglobulin gene retrieval analysis shows that these plasma cells are derived from a germ line repertoire of B cells with a diverse representation of immunoglobulin genes, exhibiting antigen-driven positive selection. Characterization of LLPC recombinant monoclonal antibodies (rMAbs) indicates that antigen recognition is achieved through convergence on a common epitopic focus by utilizing various complementarity-determining region H3 (CDRH3) lengths. Importantly, the vast majority of rMAbs produced from these cells lack polyreactivity yet manifest antigen specificity in the context of lipids, shaping MPER-specific paratopes through selective pressure. Taken together, these findings demonstrate that the MPER is a vaccine target with minimal risk of generating off-target autoimmunity. IMPORTANCE: A useful vaccine must generate desired long-term, antigen-specific antibody responses devoid of polyreactivity or autoreactivity. The common polyreactive features of some HIV-1 BNAbs have raised concern about elicitation of anti-MPER antibodies. Utilizing single-LLPC repertoire analysis and biophysical characterization of anti-MPER rMAbs, we show that their fine specificities require a structural fitness of the antibody combining site involving heavy and light chain variable domains shaped by somatic hypermutation and affinity maturation of B cells in the germinal center. Perhaps more importantly, our results demonstrate that the majority of MPER-specific antibodies are not inherently polyspecific and/or autoreactive, suggesting that polyreactivity of MPER-specific antibodies is separable from their antigen specificity.


Subject(s)
HIV Antibodies/immunology , HIV Antibodies/metabolism , HIV Antigens/immunology , HIV Envelope Protein gp41/immunology , Plasma Cells/immunology , Antibodies, Monoclonal/immunology , Epitopes, B-Lymphocyte/immunology , Membrane Lipids/metabolism
8.
Trends Immunol ; 34(2): 59-66, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23200626

ABSTRACT

Activated T cells have classically been thought to progress unidirectionally through discrete phenotypic states and differentiate into static lineages. It is increasingly evident, however, that T cells exhibit much more complex and flexible dynamic behaviors than initially appreciated, and that these behaviors influence the efficacy of T cell responses to immunological challenges. In this review, we discuss how new technologies for monitoring the dynamics of T cells are enhancing the resolution of the fine phenotypic and functional heterogeneity within populations of T cells and revealing how individual T cells transition among a continuum of states. Such insights into the dynamic properties of T cells should improve immune monitoring and inform strategies for therapeutic interventions.


Subject(s)
Cell Differentiation , Lymphocyte Activation/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Flow Cytometry/methods , Humans , Immunologic Memory/immunology , Mice , Microscopy, Fluorescence, Multiphoton , Phenotype , T-Lymphocytes/physiology
9.
J Proteome Res ; 13(2): 362-71, 2014 Feb 07.
Article in English | MEDLINE | ID: mdl-24417579

ABSTRACT

Biological assays formatted as microarrays have become a critical tool for the generation of the comprehensive data sets required for systems-level understanding of biological processes. Manual annotation of data extracted from images of microarrays, however, remains a significant bottleneck, particularly for protein microarrays due to the sensitivity of this technology to weak artifact signal. In order to automate the extraction and curation of data from protein microarrays, we describe an algorithm called Crossword that logically combines information from multiple approaches to fully automate microarray segmentation. Automated artifact removal is also accomplished by segregating structured pixels from the background noise using iterative clustering and pixel connectivity. Correlation of the location of structured pixels across image channels is used to identify and remove artifact pixels from the image prior to data extraction. This component improves the accuracy of data sets while reducing the requirement for time-consuming visual inspection of the data. Crossword enables a fully automated protocol that is robust to significant spatial and intensity aberrations. Overall, the average amount of user intervention is reduced by an order of magnitude and the data quality is increased through artifact removal and reduced user variability. The increase in throughput should aid the further implementation of microarray technologies in clinical studies.


Subject(s)
Algorithms , Automation , Protein Array Analysis
10.
Anal Chem ; 84(24): 10531-6, 2012 Dec 18.
Article in English | MEDLINE | ID: mdl-23205933

ABSTRACT

We present a method that uses fluorescent cellular barcodes to increase the number of unique samples that can be analyzed simultaneously by microengraving, a nanowell array-based technique for quantifying the secretory responses of thousands of single cells in parallel. Using n different fluorescent dyes to generate 2(n) unique cellular barcodes, we achieved a 2(n)-fold reduction in the number of arrays and quantity of reagents required per sample. The utility of this approach was demonstrated in three applications of interest in clinical and experimental immunology. Using barcoded human peripheral blood mononuclear cells and T cells, we constructed dose-response curves, profiled the secretory behavior of cells treated with mechanistically distinct stimuli, and tracked the secretory behaviors of different lineages of CD4(+) T helper cells. In addition to increasing the number of samples analyzed by generating secretory profiles of single cells from multiple populations in a time- and reagent-efficient manner, we expect that cellular barcoding in combination with microengraving will facilitate unique experimental opportunities for quantitatively analyzing interactions among heterogeneous cells isolated in small groups (~2-5 cells).


Subject(s)
Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/metabolism , Single-Cell Analysis/methods , T-Lymphocytes/chemistry , T-Lymphocytes/metabolism , Fluorescent Dyes/analysis , Fluorescent Dyes/chemistry , Humans
11.
J Clin Invest ; 132(2)2022 01 18.
Article in English | MEDLINE | ID: mdl-34813505

ABSTRACT

Food allergy affects an estimated 8% of children in the United States. Oral immunotherapy (OIT) is a recently approved treatment, with outcomes ranging from sustained tolerance to food allergens to no apparent benefit. The immunological underpinnings that influence clinical outcomes of OIT remain largely unresolved. Using single-cell RNA-Seq and paired T cell receptor α/ß (TCRα/ß) sequencing, we assessed the transcriptomes of CD154+ and CD137+ peanut-reactive T helper (Th) cells from 12 patients with peanut allergy longitudinally throughout OIT. We observed expanded populations of cells expressing Th1, Th2, and Th17 signatures that further separated into 6 clonally distinct subsets. Four of these subsets demonstrated a convergence of TCR sequences, suggesting antigen-driven T cell fates. Over the course of OIT, we observed suppression of Th2 and Th1 gene signatures in effector clonotypes but not T follicular helper-like (Tfh-like) clonotypes. Positive outcomes were associated with stronger suppression of Th2 signatures in Th2A-like cells, while treatment failure was associated with the expression of baseline inflammatory gene signatures that were present in Th1 and Th17 cell populations and unmodulated by OIT. These results demonstrate that differential clinical responses to OIT are associated with both preexisting characteristics of peanut-reactive CD4+ T cells and suppression of a subset of Th2 cells.


Subject(s)
Arachis , Desensitization, Immunologic , Peanut Hypersensitivity , RNA-Seq , Receptors, Antigen, T-Cell, alpha-beta , Single-Cell Analysis , T-Lymphocytes, Helper-Inducer/immunology , Child , Female , Humans , Male , Peanut Hypersensitivity/genetics , Peanut Hypersensitivity/immunology , Peanut Hypersensitivity/therapy , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/immunology
12.
Anal Chem ; 83(17): 6890-5, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21812465

ABSTRACT

We present here a new method to enhance the detection of secreted cytokines and chemokines from single human mononuclear cells. The technique uses a hybridization chain reaction (HCR) to amplify signals resulting from sandwich immunoassays. This immuno-HCR employs oligonucleotide-based initiators covalently linked to antibodies to propagate a chain reaction of hybridization events involving a pair of complementary hairpin oligomers bearing fluorescent labels. Integrating this strategy for signal amplification with microengraving (a soft lithographic method for printing arrays of secreted proteins from thousands of single cells) improves both the limits of detection and sensitivity for cytokines and chemokines captured from individual cells by an average of 200-fold relative to methods for direct detection by fluoresence. This approach should enhance the utility of microengraving for defining the immunological signatures of diseases and responses to interventional therapies based on multiplexed single-cell analysis.


Subject(s)
Cytokines/analysis , Immunoassay/methods , Leukocytes, Mononuclear/metabolism , Antibodies, Immobilized/immunology , Cells, Cultured , Chemokines/analysis , Chemokines/immunology , Cytokines/immunology , Dimethylpolysiloxanes/chemistry , Fluorescent Dyes/chemistry , Glass/chemistry , Humans , Leukocytes, Mononuclear/immunology , Oligonucleotides/chemistry , Protein Array Analysis/methods
13.
Nat Commun ; 12(1): 7116, 2021 12 10.
Article in English | MEDLINE | ID: mdl-34893587

ABSTRACT

Mammary morphogenesis is an orchestrated process involving differentiation, proliferation and organization of cells to form a bi-layered epithelial network of ducts and lobules embedded in stromal tissue. We have engineered a 3D biomimetic human breast that makes it possible to study how stem cell fate decisions translate to tissue-level structure and function. Using this advancement, we describe the mechanism by which breast epithelial cells build a complex three-dimensional, multi-lineage tissue by signaling through a collagen receptor. Discoidin domain receptor tyrosine kinase 1 induces stem cells to differentiate into basal cells, which in turn stimulate luminal progenitor cells via Notch signaling to differentiate and form lobules. These findings demonstrate how human breast tissue regeneration is triggered by transmission of signals from the extracellular matrix through an epithelial bilayer to coordinate structural changes that lead to formation of a complex ductal-lobular network.


Subject(s)
Breast/cytology , Breast/physiology , Cell Communication/physiology , Cell Differentiation/physiology , Discoidin Domain Receptor 1/metabolism , Biocompatible Materials , Biomedical Engineering , Cell Line , Discoidin Domain Receptor 1/genetics , Epithelial Cells/cytology , Extracellular Matrix , Humans , Regeneration , Signal Transduction , Stem Cells/cytology
14.
PLoS One ; 12(9): e0183738, 2017.
Article in English | MEDLINE | ID: mdl-28910279

ABSTRACT

BACKGROUND: The efficacy of protein-conjugated pneumococcal polysaccharide vaccines has been well characterized for children. The level of protection conferred by unconjugated polysaccharide vaccines remains less clear, particularly for elderly individuals who have had prior antigenic experience through immunization with unconjugated polysaccharide vaccines or natural exposure to Streptococcus pneumoniae. METHODS: We compared the magnitude, diversity and genetic biases of antigen-specific memory B cells in two groups of adult cynomolgus macaques that were immunized with a 7-valent conjugated vaccine and boosted after five years with either a 13-valent pneumococcal polysaccharide conjugate vaccine (13vPnC) or a 23-valent unconjugated pneumococcal polysaccharide vaccine (23vPS) using microengraving (a single-cell analysis method) and single-cell RT-PCR. RESULTS: Seven days after boosting, the mean frequency of antigen-specific memory B cells was significantly increased in macaques vaccinated with 13vPnC compared to those receiving 23vPS. The 13vPnC-vaccinated macaques also exhibited a more even distribution of antibody specificities to four polysaccharides in the vaccine (PS4, 6B, 14, 23F) that were examined. However, single-cell analysis of the antibody variable region sequences from antigen-specific B cells elicited by unconjugated and conjugated vaccines indicated that both the germline gene segments forming the heavy chains and the average lengths of the Complementary Determining Region 3 (CDR3) were similar. CONCLUSIONS: Our results confirm that distinctive differences can manifest between antigen-specific memory B cell repertoires in nonhuman primates immunized with conjugated and unconjugated pneumococcal polysaccharide vaccines. The study also supports the notion that the conjugated vaccines have a favorable profile in terms of both the frequency and breadth of the anamnestic response among antigen-specific memory B cells.


Subject(s)
B-Lymphocytes/metabolism , Heptavalent Pneumococcal Conjugate Vaccine/administration & dosage , Macaca/immunology , Pneumococcal Vaccines/administration & dosage , Animals , Antibodies, Bacterial/immunology , Heptavalent Pneumococcal Conjugate Vaccine/immunology , Immunization, Secondary , Immunologic Memory , Pneumococcal Vaccines/immunology , Single-Cell Analysis , Streptococcus pneumoniae/immunology
15.
Clin Vaccine Immunol ; 22(11): 1206-18, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26446421

ABSTRACT

Chlamydia trachomatis is the causative agent of the most frequently reported bacterial sexually transmitted infection, the total burden of which is underestimated due to the asymptomatic nature of the infection. Untreated C. trachomatis infections can cause significant morbidities, including pelvic inflammatory disease and tubal factor infertility (TFI). The human immune response against C. trachomatis, an obligate intracellular bacterium, is poorly characterized but is thought to rely on cell-mediated immunity, with CD4(+) and CD8(+) T cells implicated in protection. In this report, we present immune profiling data of subjects enrolled in a multicenter study of C. trachomatis genital infection. CD4(+) and CD8(+) T cells from subjects grouped into disease-specific cohorts were screened using a C. trachomatis proteomic library to identify the antigen specificities of recall T cell responses after natural exposure by measuring interferon gamma (IFN-γ) levels. We identified specific T cell responses associated with the resolution of infection, including unique antigens identified in subjects who spontaneously cleared infection and different antigens associated with C. trachomatis-related sequelae, such as TFI. These data suggest that novel and unique C. trachomatis T cell antigens identified in individuals with effective immune responses can be considered as targets for vaccine development, and by excluding antigens associated with deleterious sequelae, immune-mediated pathologies may be circumvented.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Chlamydia Infections/immunology , Chlamydia trachomatis/immunology , Reproductive Tract Infections/immunology , Adolescent , Adult , Antibodies, Bacterial/immunology , Antigens, Bacterial/immunology , Chlamydia Infections/microbiology , Chlamydia trachomatis/genetics , Epitopes, T-Lymphocyte/immunology , Female , Humans , Immunity, Cellular , Interferon-gamma/immunology , Male , Middle Aged , Proteomics , Reproductive Tract Infections/microbiology , Young Adult
16.
Virology ; 464-465: 296-311, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25108380

ABSTRACT

Reactivation of latent herpes simplex virus 2 (HSV-2) infections can be characterized by episodic recurrent genital lesions and/or viral shedding. We hypothesize that infected (HSV-2(pos)) asymptomatic individuals have acquired T cell responses to specific HSV-2 antigen(s) that may be an important factor in controlling their recurrent disease symptoms. Our proteomic screening technology, ATLAS, was used to characterize the antigenic repertoire of T cell responses in infected (HSV-2(pos)) and virus-exposed seronegative (HSV-2(neg)) subjects. T cell responses, determined by IFN-γ secretion, were generated to gL, UL2, UL11, UL21, ICP4, ICP0, ICP47 and UL40 with greater magnitude and/or frequency among cohorts of exposed HSV-2(neg) or asymptomatic HSV-2(pos) individuals, compared to symptomatic recurrent HSV-2(pos) subjects. T cell antigens recognized preferentially among individuals who are resistant to infection or who are infected and have mild or no clinical disease may provide new targets for the design of vaccines aimed at treating and/or preventing HSV-2 infection.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Herpes Genitalis/immunology , Herpesvirus 2, Human/immunology , Adult , Aged , Antibodies, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Cohort Studies , Epitopes, T-Lymphocyte/genetics , Female , Herpes Genitalis/genetics , Herpes Genitalis/virology , Herpesvirus 2, Human/genetics , Humans , Male , Middle Aged , Young Adult
17.
Integr Biol (Camb) ; 6(4): 388-98, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24522233

ABSTRACT

Ample evidence supports genetic and functional heterogeneity in primary tumors, but it remains unclear whether circulating tumor cells (CTCs) also exhibit the same hierarchical organization. We examined the functional diversity of viable, single CTCs using an array of subnanoliter wells (nanowells). The compartmentalization of single cells by nanowells allowed clonal comparison and mapping of heterogeneity of single cells or preformed clusters of cells. By measuring the short-term viability, invasiveness and secretory profiles of individual CTCs, it was evident that only a rare subset of CTCs possessed malignant traits indicative of metastatic potential in late-stage, progressing metastatic castration-resistant prostate cancer (mCRPC) patients. These CTCs were resistant to anoikis after being in the circulation, were invasive in their epithelial state, or secreted proteases capable of cleaving peptide substrates. Every CTC observed, however, did not exhibit such metastatic potential, suggesting that enumeration of CTCs alone may be insufficient to understand metastasis or stratify patients.


Subject(s)
Anoikis/physiology , Nanotechnology/methods , Neoplasm Proteins/physiology , Neoplasms, Hormone-Dependent/pathology , Neoplastic Cells, Circulating/pathology , Prostatic Neoplasms/pathology , Cell Survival/physiology , Fluorescence Resonance Energy Transfer , Humans , Male , Microscopy, Fluorescence , Neoplastic Cells, Circulating/metabolism
18.
Vaccine ; 30(29): 4387-93, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22682294

ABSTRACT

A comprehensive proteomic screening technology was previously used to characterize T cell responses to Chlamydia trachomatis infection. In this study, we demonstrated that T cells specific for protein antigens identified through this comprehensive technology home to the site of infection after mucosal challenge with C. trachomatis. In addition, T cell responses to these proteins were elicited in multiple genetic backgrounds. Two protein antigens, CT823 and CT144, were evaluated as vaccine candidates. When administered with AbISCO-100 adjuvant, these antigens stimulated potent CD8(+) T cell responses, polyfunctional T(H)1-polarized CD4(+) T cell responses, and high titer protein-specific T(H)1-skewed antibody responses. Vaccination with either antigen with AbISCO-100 provided long-lived protection against intravaginal challenge with C. trachomatis. Adoptive transfer of immune T cells also conferred protection in the challenge model whereas passive transfer of immune serum did not, indicating the critical role for T cell responses in control of this infection. The ability of these antigens to induce potent immune responses and provide long-lived protection in response to challenge provides a basis for the rational design of a C. trachomatis subunit vaccine.


Subject(s)
Antibody Formation , Antigens, Bacterial/immunology , Chlamydia Infections/prevention & control , Chlamydia trachomatis/immunology , T-Lymphocytes/immunology , Vagina/immunology , Adjuvants, Immunologic/administration & dosage , Adoptive Transfer , Animals , Antibodies, Bacterial/blood , Bacterial Vaccines/immunology , Cytokines/immunology , Female , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Proteomics , Recombinant Proteins/immunology , Vagina/microbiology
19.
Cell Host Microbe ; 9(2): 158-65, 2011 Feb 17.
Article in English | MEDLINE | ID: mdl-21320698

ABSTRACT

Streptococcus pneumoniae is a leading cause of mortality in young children. While successful conjugate polysaccharide vaccines exist, a less expensive serotype-independent protein-based pneumococcal vaccine offers a major advancement for preventing life-threatening pneumococcal infections, particularly in developing nations. IL-17A-secreting CD4+ T cells (T(H)17) mediate resistance to mucosal colonization by multiple pathogens including S. pneumoniae. Screening an expression library containing >96% of predicted pneumococcal proteins, we identified antigens recognized by T(H)17 cells from mice immune to pneumococcal colonization. The identified antigens also elicited IL-17A secretion from colonized mouse splenocytes and human PBMCs suggesting that similar responses are primed during natural exposure. Immunization of two mouse strains with identified antigens provided protection from pneumococcal colonization that was significantly diminished in animals treated with blocking CD4 or IL-17A antibodies. This work demonstrates the potential of proteomic screening approaches to identify specific antigens for the design of subunit vaccines against mucosal pathogens via harnessing T(H)17-mediated immunity.


Subject(s)
Pneumococcal Infections/microbiology , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/immunology , Streptococcus pneumoniae/immunology , Th17 Cells/immunology , Animals , Antigens, Bacterial/genetics , Antigens, Bacterial/immunology , Humans , Immunization , Interleukin-17/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pneumococcal Infections/immunology , Pneumococcal Vaccines/administration & dosage , Streptococcus pneumoniae/genetics , Th17 Cells/microbiology
20.
Proc Natl Acad Sci U S A ; 103(32): 12069-74, 2006 Aug 08.
Article in English | MEDLINE | ID: mdl-16880389

ABSTRACT

To date, it has not been possible to study antigen-specific T cell responses during primary infection of the genital tract. The low frequency of pathogen-specific T cells in a naïve mouse makes it difficult to monitor the initial events after antigen encounter. We developed a system to examine the response of pathogen-specific T cells in the genital mucosa after intrauterine infection. We identified the protective CD4(+) T cell antigen Cta1 from Chlamydia trachomatis and generated T cell receptor (TCR) transgenic (tg) mice with specificity for this protein. By transferring TCR tg T cells into naïve animals, we determined that Chlamydia-specific T cells were activated and proliferated in the lymph nodes draining the genital tract after primary intrauterine infection. Activated T cells migrated into the genital mucosa and secreted IFN-gamma. The development of Chlamydia-specific TCR tg mice provides an approach for dissecting how pathogen-specific T cells function in the genital tract.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Chlamydia Infections/immunology , Chlamydia Infections/microbiology , Animals , Bone Marrow Cells/metabolism , Cell Proliferation , Chlamydia Infections/blood , Chlamydia trachomatis/metabolism , Interferon-gamma/metabolism , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell/metabolism , Spleen/metabolism , Spleen/microbiology , Tumor Necrosis Factor Receptor Superfamily, Member 7/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL