Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
PLoS One ; 16(8): e0254125, 2021.
Article in English | MEDLINE | ID: mdl-34411144

ABSTRACT

Imaging techniques based on fluorescence and bioluminescence have been important tools in visualizing tumor progression and studying the effect of drugs and immunotherapies on tumor immune microenvironment in animal models of cancer. However, transgenic expression of foreign proteins may induce immune responses in immunocompetent syngeneic tumor transplant models and augment the efficacy of experimental drugs. In this study, we show that the growth rate of Lewis lung carcinoma (LL/2) tumors was reduced after transduction of tdTomato and luciferase (tdTomato/Luc) compared to the parental cell line. tdTomato/Luc expression by LL/2 cells altered the tumor microenvironment by increasing tumor-infiltrating lymphocytes (TILs) while inhibiting tumor-induced myeloid-derived suppressor cells (MDSCs). Interestingly, tdTomato/Luc expression did not alter the response of LL/2 tumors to anti-PD-1 and anti-CTLA-4 antibodies. These results suggest that the use of tdTomato/Luc-transduced cancer cells to conduct studies in immune competent mice may lead to cell-extrinsic tdTomato/Luc-induced alterations in tumor growth and tumor immune microenvironment that need to be taken into consideration when evaluating the efficacy of anti-cancer drugs and vaccines in immunocompetent animal models.


Subject(s)
Carcinoma, Lewis Lung , Gene Expression , Genes, Reporter/immunology , Luciferases , Luminescent Proteins , Lung Neoplasms , Tumor Microenvironment , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/immunology , Cell Line, Tumor , Luciferases/genetics , Luciferases/immunology , Luminescent Proteins/genetics , Luminescent Proteins/immunology , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mice , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Red Fluorescent Protein
2.
Cell Rep ; 35(2): 109000, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33852860

ABSTRACT

Chemotaxis and lysosomal function are closely intertwined processes essential for the inflammatory response and clearance of intracellular bacteria. We used the zebrafish model to examine the link between chemotactic signaling and lysosome physiology in macrophages during mycobacterial infection and wound-induced inflammation in vivo. Macrophages from zebrafish larvae carrying a mutation in a chemokine receptor of the Cxcr3 family display upregulated expression of vesicle trafficking and lysosomal genes and possess enlarged lysosomes that enhance intracellular bacterial clearance. This increased microbicidal capacity is phenocopied by inhibiting the lysosomal transcription factor EC, while its overexpression counteracts the protective effect of chemokine receptor mutation. Tracking macrophage migration in zebrafish revealed that lysosomes of chemokine receptor mutants accumulate in the front half of cells, preventing macrophage polarization during chemotaxis and reaching sites of inflammation. Our work shows that chemotactic signaling affects the bactericidal properties and localization during chemotaxis, key aspects of the inflammatory response.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Lysosomes/immunology , Macrophages/immunology , Mycobacterium Infections/genetics , Receptors, CXCR3/genetics , Signal Transduction/immunology , Zebrafish Proteins/genetics , Zebrafish/genetics , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/immunology , Cell Tracking , Chemotaxis/genetics , Chemotaxis/immunology , Embryo, Nonmammalian , Gene Expression Profiling , Gene Expression Regulation , Genes, Reporter , Larva/immunology , Larva/microbiology , Luminescent Proteins/genetics , Luminescent Proteins/immunology , Lysosomes/metabolism , Lysosomes/microbiology , Lysosomes/ultrastructure , Macrophage Activation , Macrophages/microbiology , Macrophages/ultrastructure , Mutation , Mycobacterium Infections/immunology , Mycobacterium Infections/microbiology , Mycobacterium marinum/immunology , Mycobacterium marinum/pathogenicity , Receptors, CXCR3/immunology , Sequence Analysis, RNA , Signal Transduction/genetics , Zebrafish/immunology , Zebrafish/microbiology , Zebrafish Proteins/immunology , Red Fluorescent Protein
3.
Anal Biochem ; 608: 113875, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32739350

ABSTRACT

mWasabi is a bright monomeric green fluorescent protein. It can be used as a fusion tag to monitor various biological events, e.g. protein localization. Here we report the selection of camelid-derived single-domain antibody fragments (nanobodies) against mWasabi. In this work, phage-display approach was employed to select the high affinity mWasabi-specific Nb (nanobodies). These nanobodies were able to recognize mWasabi or in a fused fashion with PD1. The interesting binding characteristics of these two mWasabi-specific nanobodies could be valuable for design new tools for cellular tracing or targeting based on the mWasabi-fusing protein in many different biological research fields.


Subject(s)
Cell Surface Display Techniques/methods , Luminescent Proteins/chemistry , Single-Domain Antibodies/immunology , Single-Domain Antibodies/isolation & purification , Amino Acid Sequence , Animals , Camelidae/blood , Camelidae/immunology , HEK293 Cells , Humans , Immunoglobulin G/blood , Luminescent Proteins/immunology , Luminescent Proteins/isolation & purification , Programmed Cell Death 1 Receptor/analysis , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Sequence Alignment
4.
Biotechniques ; 69(4): 302-306, 2020 10.
Article in English | MEDLINE | ID: mdl-32639163

ABSTRACT

We established a quantitative detection method for immunohistochemistry based on a reference standard light-emitting diode, protein microarray and antibody-fused bioluminescent protein. In this procedure, we calibrated the bioluminescence imaging system and prepared the calibration curve between antigen and antibody-fused bioluminescent protein using a protein microarray. Then we converted the detecting light signal to antigen count via absolute photon number in the bioluminescent images; there was a resulting threefold difference in the target antigen number between normal and cancerous tissues. Our technique can easily compare immunohistological images and evaluate tumor progression in quantitative pathological diagnosis.


Subject(s)
Antibodies/chemistry , Immunohistochemistry , Luminescent Proteins/isolation & purification , Protein Array Analysis , Antibodies/genetics , Antibodies/immunology , Humans , Luminescent Proteins/chemistry , Luminescent Proteins/immunology
5.
Cells ; 8(1)2019 01 14.
Article in English | MEDLINE | ID: mdl-30646582

ABSTRACT

DNA point accumulation for imaging in nanoscale topography (PAINT) is a rapidly developing fluorescence super-resolution technique, which allows for reaching spatial resolutions below 10 nm. It also enables the imaging of multiple targets in the same sample. However, using DNA-PAINT to observe cellular structures at such resolution remains challenging. Antibodies, which are commonly used for this purpose, lead to a displacement between the target protein and the reporting fluorophore of 20⁻25 nm, thus limiting the resolving power. Here, we used nanobodies to minimize this linkage error to ~4 nm. We demonstrate multiplexed imaging by using three nanobodies, each able to bind to a different family of fluorescent proteins. We couple the nanobodies with single DNA strands via a straight forward and stoichiometric chemical conjugation. Additionally, we built a versatile computer-controlled microfluidic setup to enable multiplexed DNA-PAINT in an efficient manner. As a proof of principle, we labeled and imaged proteins on mitochondria, the Golgi apparatus, and chromatin. We obtained super-resolved images of the three targets with 20 nm resolution, and within only 35 minutes acquisition time.


Subject(s)
Luminescent Proteins/analysis , Microscopy, Fluorescence/methods , Single Molecule Imaging/methods , Single-Domain Antibodies/immunology , Animals , COS Cells , Chlorocebus aethiops , Chromatin/chemistry , Chromatin/ultrastructure , DNA/chemistry , Golgi Apparatus/chemistry , Golgi Apparatus/ultrastructure , Luminescent Proteins/immunology , Mitochondria/chemistry , Mitochondria/ultrastructure
6.
ACS Sens ; 4(1): 20-25, 2019 01 25.
Article in English | MEDLINE | ID: mdl-30525479

ABSTRACT

Bioluminescent sensor proteins provide attractive tools for applications ranging from in vivo imaging to point-of-care testing. Here we introduce a new class of ratiometric bioluminescent sensor proteins that do not rely on direct modulation of BRET efficiency, but are based on competitive intramolecular complementation of split NanoLuc luciferase. Proof of concept for the feasibility of this sensor principle was provided by developing a blue-red light emitting sensor protein for the detection of anti-HIV1-p17 antibodies with a 500% change in emission ratio and a Kd of 10 pM. The new sensor design also improved the dynamic response of a sensor for the therapeutic antibody cetuximab 4-fold, allowing the direct quantification of this anti-EGFR antibody in undiluted blood plasma. The modular sensor architecture allows easy and systematic tuning of a sensor's dynamic range and should be generally applicable to allow rational engineering of bioluminescent sensor proteins.


Subject(s)
HIV Antibodies/blood , Luciferases/chemistry , Luminescent Proteins/chemistry , Bioluminescence Resonance Energy Transfer Techniques/methods , Cetuximab/immunology , Fluorescent Dyes/chemistry , HIV Antibodies/immunology , Luminescent Proteins/immunology , Proof of Concept Study
7.
Parasite Immunol ; 41(2): e12608, 2019 02.
Article in English | MEDLINE | ID: mdl-30500992

ABSTRACT

Parasites have been engineered to express fluorescent reporter proteins, yet the impact of red fluorescent proteins on Leishmania infections remains largely unknown. We analysed the infection outcome of Leishmania mexicana parasites engineered for the constitutive expression of mKate protein and evaluated their immunogenicity in BALB/c mice. Infection of BALB/c mice with mKate transfected L. mexicana (LmexmKate ) parasites caused enlarged lesion sizes, leading to ulceration, and containing more parasites, as compared to LmexWT . The mKate protein showed immunogenic properties inducing antibody production against the mKate protein, as well as enhancing antibody production against the parasite. The augmented lesion sizes and ulcers, together with the more elevated antibody production, were related to an enhanced number of TNF-α and IL-1ß producing cells in the infected tissues. We conclude that mKate red fluorescent protein is an immunogenic protein, capable of modifying disease evolution of L. mexicana.


Subject(s)
Leishmania mexicana/immunology , Luminescent Proteins/immunology , Animals , Female , Leishmania mexicana/genetics , Luminescent Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transfection , Red Fluorescent Protein
8.
Nat Commun ; 9(1): 4845, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451869

ABSTRACT

Circulating monocytes can compete for virtually any tissue macrophage niche and become long-lived replacements that are phenotypically indistinguishable from their embryonic counterparts. As the factors regulating this process are incompletely understood, we studied niche competition in the brain by depleting microglia with >95% efficiency using Cx3cr1CreER/+R26DTA/+ mice and monitored long-term repopulation. Here we show that the microglial niche is repopulated within weeks by a combination of local proliferation of CX3CR1+F4/80lowClec12a- microglia and infiltration of CX3CR1+F4/80hiClec12a+ macrophages that arise directly from Ly6Chi monocytes. This colonization is independent of blood brain barrier breakdown, paralleled by vascular activation, and regulated by type I interferon. Ly6Chi monocytes upregulate microglia gene expression and adopt microglia DNA methylation signatures, but retain a distinct gene signature from proliferating microglia, displaying altered surface marker expression, phagocytic capacity and cytokine production. Our results demonstrate that monocytes are imprinted by the CNS microenvironment but remain transcriptionally, epigenetically and functionally distinct.


Subject(s)
Brain/immunology , Cell Lineage/immunology , Gene Expression Regulation/immunology , Microglia/immunology , Monocytes/immunology , Adoptive Transfer , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Antigens, Ly/genetics , Antigens, Ly/immunology , Bacterial Proteins/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Brain/cytology , Brain/radiation effects , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/immunology , Cell Lineage/radiation effects , Cell Proliferation , DNA Methylation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Interferon Type I/genetics , Interferon Type I/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Luminescent Proteins/immunology , Macrophages/cytology , Macrophages/immunology , Macrophages/radiation effects , Mice , Mice, Transgenic , Microglia/cytology , Microglia/radiation effects , Monocytes/cytology , Monocytes/radiation effects , Monocytes/transplantation , Phagocytosis , Receptors, Mitogen/genetics , Receptors, Mitogen/immunology , Signal Transduction , Transplantation Chimera , Whole-Body Irradiation
9.
PLoS One ; 13(10): e0205303, 2018.
Article in English | MEDLINE | ID: mdl-30332431

ABSTRACT

Higher vertebrates have evolved innate and adaptive immune systems to defend against foreign substances and pathogens. Sophisticated regulatory circuits are needed to avoid inappropriate immune responses and inflammation. GPR108 is a seven-transmembrane family protein that activates NF-κB strongly when overexpressed. Surprisingly, its action in a physiological context is that of an antagonist of Toll-like receptor (TLR)-mediated signaling. Cells from Gpr108-null mice exhibit enhanced cytokine secretion and NF-κB and IRF3 signaling, whereas Gpr108-null macrophages reconstituted with GPR108 exhibit blunted signaling. Co-expression of TLRs and GPR108 reduces NF-κB and IFNß promoter activation compared to expression of either TLRs or GPR108 alone. Upon TLR stimulation GPR108 abundance increases and the protein engages TLRs and their partners to reduce MyD88 expression and interfere with its binding to TLR4 through blocking MyD88 ubiquitination. In turn GPR108 is antagonized by TIRAP, an adaptor protein for TLR and MyD88. The interrelationships between GPR108 and innate immune signaling components are multifactorial and point to a membrane-associated signaling structure of significant complexity.


Subject(s)
Immunity, Innate , Membrane Glycoproteins/genetics , NF-kappa B/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Interleukin-1/genetics , Toll-Like Receptor 3/genetics , Animals , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , HEK293 Cells , Humans , Lipopolysaccharides/pharmacology , Luminescent Proteins/genetics , Luminescent Proteins/immunology , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NF-kappa B/immunology , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/immunology , Receptors, Interleukin-1/immunology , Signal Transduction , THP-1 Cells , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/immunology , Toll-Like Receptor 7/genetics , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/immunology , Red Fluorescent Protein
10.
Angew Chem Int Ed Engl ; 56(25): 7112-7116, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28510347

ABSTRACT

We introduce a general method to transform antibodies into ratiometric, bioluminescent sensor proteins for the no-wash quantification of analytes. Our approach is based on the genetic fusion of antibody fragments to NanoLuc luciferase and SNAP-tag, the latter being labeled with a synthetic fluorescent competitor of the antigen. Binding of the antigen, here synthetic drugs, by the sensor displaces the tethered fluorescent competitor from the antibody and disrupts bioluminescent resonance energy transfer (BRET) between the luciferase and fluorophore. The semisynthetic sensors display a tunable response range (submicromolar to submillimolar) and large dynamic range (ΔRmax >500 %), and they permit the quantification of analytes through spotting of the samples onto paper followed by analysis with a digital camera.


Subject(s)
Antibodies/chemistry , Immunoassay/methods , Luminescent Proteins/chemistry , Point-of-Care Systems , Antibodies/immunology , Biosensing Techniques , Complementarity Determining Regions , Energy Transfer , Humans , Luminescent Measurements , Luminescent Proteins/immunology , Methotrexate/chemistry , Methotrexate/immunology , Quinine/chemistry , Quinine/immunology , Reproducibility of Results , Theophylline/chemistry , Theophylline/immunology
11.
Theranostics ; 6(9): 1453-66, 2016.
Article in English | MEDLINE | ID: mdl-27375792

ABSTRACT

In vivo optical spatio-temporal imaging of the tumor microenvironment is useful to explain how tumor immunotherapies work. However, the lack of fluorescent antigens with strong immunogenicity makes it difficult to study the dynamics of how tumors are eliminated by any given immune response. Here, we develop an effective fluorescent model antigen based on the tetrameric far-red fluorescent protein KatushkaS158A (tfRFP), which elicits both humoral and cellular immunity. We use this fluorescent antigen to visualize the dynamic behavior of immunocytes as they attack and selectively eliminate tfRFP-expressing tumors in vivo; swarms of immunocytes rush toward tumors with high motility, clusters of immunocytes form quickly, and numerous antigen-antibody complexes in the form of tfRFP(+) microparticles are generated in the tumor areas and ingested by macrophages in the tumor microenvironment. Therefore, tfRFP, as both a model antigen and fluorescent reporter, is a useful tool to visualize specific immune responses in vivo.


Subject(s)
Antigens, Neoplasm/analysis , Luminescent Proteins/analysis , Luminescent Proteins/immunology , Neoplasms/chemistry , Neoplasms/immunology , Animals , Disease Models, Animal , Intravital Microscopy , Mice, Inbred C57BL , Red Fluorescent Protein
12.
Stem Cell Rev Rep ; 12(5): 553-559, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27435468

ABSTRACT

Green Fluorescent protein (GFP), used as a cellular tag, provides researchers with a valuable method of measuring gene expression and cell tracking. However, there is evidence to suggest that the immunogenicity and cytotoxicity of GFP potentially confounds the interpretation of in vivo experimental data. Studies have shown that GFP expression can deteriorate over time as GFP tagged cells are prone to death. Therefore, the cells that were originally marked with GFP do not survive and cannot be accurately traced over time. This review will present current evidence for the immunogenicity and cytotoxicity of GFP in in vivo studies by characterizing these responses.


Subject(s)
Cell Tracking/adverse effects , Green Fluorescent Proteins/adverse effects , Green Fluorescent Proteins/immunology , Animals , Gene Expression/genetics , Luminescent Proteins/adverse effects , Luminescent Proteins/immunology
13.
Nat Commun ; 7: 12057, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27381163

ABSTRACT

Cells respond dynamically to pulsatile cytokine stimulation. Here we report that single, or well-spaced pulses of TNFα (>100 min apart) give a high probability of NF-κB activation. However, fewer cells respond to shorter pulse intervals (<100 min) suggesting a heterogeneous refractory state. This refractory state is established in the signal transduction network downstream of TNFR and upstream of IKK, and depends on the level of the NF-κB system negative feedback protein A20. If a second pulse within the refractory phase is IL-1ß instead of TNFα, all of the cells respond. This suggests a mechanism by which two cytokines can synergistically activate an inflammatory response. Gene expression analyses show strong correlation between the cellular dynamic response and NF-κB-dependent target gene activation. These data suggest that refractory states in the NF-κB system constitute an inherent design motif of the inflammatory response and we suggest that this may avoid harmful homogenous cellular activation.


Subject(s)
Interleukin-1beta/pharmacology , NF-KappaB Inhibitor alpha/genetics , NF-kappa B/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/pharmacology , Cell Line, Tumor , Feedback, Physiological , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/immunology , Luminescent Proteins/genetics , Luminescent Proteins/immunology , NF-KappaB Inhibitor alpha/immunology , NF-kappa B/immunology , Neurons , RNA, Small Interfering/genetics , RNA, Small Interfering/immunology , Receptors, Tumor Necrosis Factor, Type I/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor alpha-Induced Protein 3/antagonists & inhibitors , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/immunology , Red Fluorescent Protein
14.
J Immunol Methods ; 430: 56-60, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26775851

ABSTRACT

Antibodies made in large animals are integral to many biomedical research endeavors. Domesticated herd animals like goats, sheep, donkeys, horses and camelids all offer distinct advantages in antibody production. However, their cost of use is often prohibitive, especially where poor antigen response is commonplace; choosing a non-responsive animal can set a research program back or even prevent experiments from moving forward entirely. Over the course of production of antibodies from llamas, we found that some animals consistently produced a higher humoral antibody response than others, even to highly divergent antigens, as well as to their standard vaccines. Based on our initial data, we propose that these "high level responders" could be pre-selected by checking antibody titers against common vaccines given to domestic farm animals. Thus, time and money can be saved by reducing the chances of getting poor responding animals and minimizing the use of superfluous animals.


Subject(s)
Antibodies, Viral/blood , Antibody Formation , Camelids, New World/immunology , Rabies Vaccines/immunology , Animals , Female , Green Fluorescent Proteins/immunology , Luminescent Proteins/immunology , Male , Rabies virus/immunology , Sheep , Vaccination/veterinary , Red Fluorescent Protein
15.
Infect Immun ; 83(12): 4719-30, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26416905

ABSTRACT

Microsporidia, which belong to the kingdom Fungi, are important opportunistic pathogens in HIV-infected populations and organ transplant recipients that are often associated with a broad range of symptoms, such as diarrhea, nephritis, and encephalitis. Natural infection occurs via the oral route, and as a consequence, gut immunity plays an important role in restricting the dissemination of these pathogens. Studies from our laboratory have reported that the pathogens induce a rapid intraepithelial lymphocyte (IEL) response important for host protection. Although mucosal dendritic cells (DC) are likely involved in triggering an antigen-specific IEL response, the specific subset(s) responsible has yet to be identified. Toward this goal, we demonstrate a very important role for mucosal CD11b(-) CD8(+) DC in the initiation of an antigen-specific IEL in vivo. Effectively, after Encephalitozoon cuniculi infection, CD11b(-) CD8(+) DC were activated in the lamina propria (LP) and acquired the ability to process retinoic acid (RA). However, this subset did not produce interleukin 12 (IL-12) but upregulated CD103, which is essential for migration to the mesenteric lymph nodes (MLN). Interestingly, CD103(+) CD11b(-) CD8(+) DC in the MLN, in addition to processing RA, also secreted IL-12 and were responsible for gut imprinting specificity on mucosal CD8 T cells. To the best of our knowledge, this is the first report describing the importance of MLN CD103(+) CD11b(-) CD8(+) DC isolated from infected animals in the generation of an IEL response against a live pathogen.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Encephalitozoon cuniculi/immunology , Encephalitozoonosis/immunology , Interleukin-12/immunology , Intestinal Mucosa/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Bacterial Proteins/genetics , Bacterial Proteins/immunology , CD11b Antigen/genetics , CD11b Antigen/immunology , CD8-Positive T-Lymphocytes/microbiology , CD8-Positive T-Lymphocytes/pathology , Dendritic Cells/microbiology , Dendritic Cells/pathology , Encephalitozoon cuniculi/pathogenicity , Encephalitozoonosis/genetics , Encephalitozoonosis/microbiology , Encephalitozoonosis/pathology , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , Host-Pathogen Interactions , Immunity, Mucosal , Immunophenotyping , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Interleukin-12/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Intestines/immunology , Intestines/microbiology , Intestines/pathology , Luminescent Proteins/genetics , Luminescent Proteins/immunology , Lymph Nodes/immunology , Lymph Nodes/microbiology , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Signal Transduction
16.
PLoS One ; 10(8): e0135994, 2015.
Article in English | MEDLINE | ID: mdl-26296096

ABSTRACT

Killing of virally infected cells or tumor cells by cytotoxic T lymphocytes requires targeting of lytic granules to the junction between the CTL and its target. We used whole-cell patch clamp to measure the cell capacitance at fixed intracellular [Ca2+] to study fusion of lytic granules in human CTLs. Expression of a fluorescently labeled human granzyme B construct allowed identification of lytic granule fusion using total internal reflection fluorescence microscopy. In this way capacitance steps due to lytic granule fusion were identified. Our goal was to determine the size of fusing lytic granules and to describe their behavior at the plasma membrane. On average, 5.02 ± 3.09 (mean ± s.d.) lytic granules were released per CTL. The amplitude of lytic granule fusion events was ~ 3.3 fF consistent with a diameter of about 325 nm. Fusion latency was biphasic with time constants of 15.9 and 106 seconds. The dwell time of fusing lytic granules was exponentially distributed with a mean dwell time of 28.5 seconds. Fusion ended in spite of the continued presence of granules at the immune synapse. The mobility of fusing granules at the membrane was indistinguishable from that of lytic granules which failed to fuse. While dwelling at the plasma membrane lytic granules exhibit mobility consistent with docking interspersed with short periods of greater mobility. The failure of lytic granules to fuse when visible in TIRF at the membrane may indicate that a membrane-confined reaction is rate limiting.


Subject(s)
Cell Membrane/metabolism , Cytoplasmic Granules/metabolism , Immunological Synapses/metabolism , Membrane Fusion/immunology , T-Lymphocytes, Cytotoxic/metabolism , Calcium/immunology , Calcium/metabolism , Cell Membrane/immunology , Cell Membrane/ultrastructure , Cytoplasmic Granules/immunology , Cytoplasmic Granules/ultrastructure , Cytotoxicity, Immunologic , Electric Capacitance , Electroporation , Exocytosis , Gene Expression , Granzymes/genetics , Granzymes/immunology , Granzymes/metabolism , Humans , Immunological Synapses/immunology , Immunological Synapses/ultrastructure , Kinetics , Luminescent Proteins/genetics , Luminescent Proteins/immunology , Luminescent Proteins/metabolism , Microscopy, Fluorescence , Primary Cell Culture , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Recombinant Fusion Proteins/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/ultrastructure , Time Factors , Red Fluorescent Protein
17.
Methods ; 88: 89-97, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26123185

ABSTRACT

With the recent development of single-molecule localization-based superresolution microscopy, the imaging of cellular structures at a resolution below the diffraction-limit of light has become a widespread technique. While single fluorescent molecules can be resolved in the nanometer range, the delivery of these molecules to the authentic structure in the cell via traditional antibody-mediated techniques can add substantial error due to the size of the antibodies. Accurate and quantitative labeling of cellular molecules has thus become one of the bottlenecks in the race for highest resolution of target structures. Here we illustrate in detail how to use small, high affinity nanobody binders against GFP and RFP family proteins for highly generic labeling of fusion constructs with bright organic dyes. We provide detailed protocols and examples for their application in superresolution imaging and single particle tracking and demonstrate advantages over conventional labeling approaches.


Subject(s)
Fluorescent Antibody Technique/methods , Fluorescent Dyes , Green Fluorescent Proteins/immunology , Luminescent Proteins/immunology , Microscopy, Fluorescence/methods , Single-Domain Antibodies , Staining and Labeling/methods , Animals , Cells, Cultured , Humans , Image Processing, Computer-Assisted , Molecular Imaging/methods , Rats , Red Fluorescent Protein
18.
J Biol Chem ; 290(26): 16431-9, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25971973

ABSTRACT

Green fluorescent protein (GFP) and other fluorescent proteins are essential tools for biological research. When fused to peptides or proteins as a reporter, GFP enables localization and quantitation of gene products in otherwise unmanipulated live cells or organisms. We previously reported that a sizable fraction of nascent GFP is post-translationally converted into a 20-kDa Triton X-100-insoluble proteasome substrate (Qian, S. B., Princiotta, M. F., Bennink, J. R., and Yewdell, J. W. (2006) J. Biol. Chem. 281, 392-400; Dolan, B. P., Li, L., Veltri, C. A., Ireland, C. M., Bennink, J. R., and Yewdell, J. W. (2011) J. Immunol. 186, 2065-2072). Here, we show that a similarly sized fragment is generated by all GFP and red fluorescent protein family members we examined. We demonstrate that fragmentation is a by-product of GFP chromophore rearrangement. A non-rearranging GFP mutant fails to fragment and generates diminished levels of K(b)-SIINFEKL complexes when SIINFEKL is genetically fused to either the C- or N-terminal domains of GFP fusion proteins. Instructively, another fragmenting GFP mutant that cannot create the functional chromophore but still generates fragments also demonstrates diminished K(b)-SIINFEKL generation. However, the mutant and wild-type fragments differ fundamentally in that wild-type fragments are rapidly liberated from the intact molecule and degraded quickly, accounting for increased K(b)-SIINFEKL generation. In the fragmenting mutant, the fragments are generated slowly and remain associated, likely in a native conformation based on their original structural description (Barondeau, D. P., Kassmann, C. J., Tainer, J. A., and Getzoff, E. D. (2006) J. Am. Chem. Soc. 128, 4685-4693). The wild-type GFP fragments represent the first biochemically defined natural defective ribosomal products to contribute peptides for immunosurveillance, enabling quantitation of peptide generation efficiency from this source of defective ribosomal products. More broadly, given the wide use of fluorescent proteins, their ubiquitous and abundant fragmentation must be considered when interpreting experiments using these extremely useful probes.


Subject(s)
Green Fluorescent Proteins/chemistry , Luminescent Proteins/chemistry , Peptide Fragments/immunology , Antigen Presentation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , HeLa Cells , Humans , Luminescent Proteins/genetics , Luminescent Proteins/immunology , Monitoring, Immunologic , Peptide Fragments/chemistry , Peptide Fragments/genetics , Red Fluorescent Protein
19.
Nat Methods ; 11(12): 1253-60, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25362362

ABSTRACT

Nanobodies are single-domain antibodies derived from the variable regions of Camelidae atypical immunoglobulins. They show promise as high-affinity reagents for research, diagnostics and therapeutics owing to their high specificity, small size (∼15 kDa) and straightforward bacterial expression. However, identification of repertoires with sufficiently high affinity has proven time consuming and difficult, hampering nanobody implementation. Our approach generates large repertoires of readily expressible recombinant nanobodies with high affinities and specificities against a given antigen. We demonstrate the efficacy of this approach through the production of large repertoires of nanobodies against two antigens, GFP and mCherry, with Kd values into the subnanomolar range. After mapping diverse epitopes on GFP, we were also able to design ultrahigh-affinity dimeric nanobodies with Kd values as low as ∼30 pM. The approach presented here is well suited for the routine production of high-affinity capture reagents for various biomedical applications.


Subject(s)
Epitopes/immunology , Green Fluorescent Proteins/immunology , Luminescent Proteins/immunology , Recombinant Proteins/isolation & purification , Single-Domain Antibodies/immunology , Single-Domain Antibodies/isolation & purification , Animals , Camelids, New World , Epitopes/genetics , Epitopes/metabolism , Gene Library , Green Fluorescent Proteins/metabolism , Immunization , Luminescent Proteins/metabolism , Lymphocytes/immunology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Single-Domain Antibodies/genetics , Single-Domain Antibodies/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Red Fluorescent Protein
20.
Eur J Immunol ; 44(12): 3632-45, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25159127

ABSTRACT

Under physiological conditions, studies on the biology of naturally induced Foxp3(+) Treg cells of intra- and extrathymic origin have been hampered by the lack of unambiguous markers to discriminate the mature progeny of such developmental Treg-cell sublineages. Here, we report on experiments in double-transgenic mice, in which red fluorescent protein (RFP) is expressed in all Foxp3(+) Treg cells, whereas Foxp3-dependent GFP expression is exclusively confined to intrathymically induced Foxp3(+) Treg cells. This novel molecular genetic tool enabled us to faithfully track and characterize naturally induced Treg cells of intrathymic (RFP(+) GFP(+) ) and extrathymic (RFP(+) GFP(-) ) origin in otherwise unmanipulated mice. These experiments directly demonstrate that extrathymically induced Treg cells substantially contribute to the overall pool of mature Foxp3(+) Treg cells residing in peripheral lymphoid tissues of steady-state mice. Furthermore, we provide evidence that intra- and extrathymically induced Foxp3(+) Treg cells represent distinct phenotypic and functional sublineages.


Subject(s)
Forkhead Transcription Factors/immunology , Green Fluorescent Proteins/immunology , Luminescent Proteins/immunology , T-Lymphocytes, Regulatory , Thymus Gland , Animals , Forkhead Transcription Factors/genetics , Green Fluorescent Proteins/genetics , Luminescent Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Knockout , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Thymus Gland/cytology , Thymus Gland/immunology , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL