Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Immunol ; 31: 137-61, 2013.
Article in English | MEDLINE | ID: mdl-23215646

ABSTRACT

Tissues such as the skin and mucosae are frequently exposed to microbial pathogens. Infectious agents must be quickly and efficiently controlled by our immune system, but the low frequency of naive T cells specific for any one pathogen means dependence on primary responses initiated in draining lymph nodes, often allowing time for serious infection to develop. These responses imprint effectors with the capacity to home to infected tissues; this process, combined with inflammatory signals, ensures the effective targeting of primary immunity. Upon vaccination or previous pathogen exposure, increased pathogen-specific T cell numbers together with altered migratory patterns of memory T cells can greatly improve immune efficacy, ensuring infections are prevented or at least remain subclinical. Until recently, memory T cell populations were considered to comprise central memory T cells (TCM), which are restricted to the secondary lymphoid tissues and blood, and effector memory T cells (TEM), which broadly migrate between peripheral tissues, the blood, and the spleen. Here we review evidence for these two memory populations, highlight a relatively new player, the tissue-resident memory T cell (TRM), and emphasize the potential differences between the migratory patterns of CD4(+) and CD8(+) T cells. This new understanding raises important considerations for vaccine design and for the measurement of immune parameters critical to the control of infectious disease, autoimmunity, and cancer.


Subject(s)
Cell Movement/immunology , Immunologic Memory , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Adaptation, Physiological/immunology , Animals , Humans , T-Lymphocyte Subsets/classification , Tissue Distribution/immunology
2.
Int J Mol Sci ; 22(6)2021 Mar 17.
Article in English | MEDLINE | ID: mdl-33802650

ABSTRACT

As an essential modulator of IgG disposition, the neonatal Fc receptor (FcRn) governs the pharmacokinetics and functions many therapeutic modalities. In this review, we thoroughly reexamine the hitherto elucidated biological and thermodynamic properties of FcRn to provide context for our assessment of more recent advances, which covers antigen-binding fragment (Fab) determinants of FcRn affinity, transgenic preclinical models, and FcRn targeting as an immune-complex (IC)-clearing strategy. We further comment on therapeutic antibodies authorized for treating SARS-CoV-2 (bamlanivimab, casirivimab, and imdevimab) and evaluate their potential to saturate FcRn-mediated recycling. Finally, we discuss modeling and simulation studies that probe the quantitative relationship between in vivo IgG persistence and in vitro FcRn binding, emphasizing the importance of endosomal transit parameters.


Subject(s)
Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/metabolism , Receptors, Fc/chemistry , Receptors, Fc/metabolism , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacokinetics , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Receptors, Fc/immunology , Tissue Distribution/immunology , COVID-19 Drug Treatment
3.
Int J Mol Sci ; 21(4)2020 Feb 15.
Article in English | MEDLINE | ID: mdl-32075272

ABSTRACT

Non-invasively monitoring allogeneic graft rejection with a specific marker is of great importance for prognosis of patients. Recently, data revealed that IL-27Rα was up-regulated in alloreactive CD4+ T cells and participated in inflammatory diseases. Here, we evaluated whether IL-27Rα could be used in monitoring allogeneic graft rejection both in vitro and in vivo. Allogeneic (C57BL/6 donor to BALB/c recipient) and syngeneic (BALB/c both as donor and recipient) skin grafted mouse models were established. The expression of IL-27Rα in grafts was detected. The radio-probe, 125I-anti-IL-27Rα mAb, was prepared. Dynamic whole-body phosphor-autoradiography, ex vivo biodistribution and immunofluorescence staining were performed. The results showed that the highest expression of IL-27Rα was detected in allogeneic grafts on day 10 post transplantation (top period of allorejection). 125I-anti-IL-27Rα mAb was successfully prepared with higher specificity and affinity. Whole-body phosphor-autoradiography showed higher radioactivity accumulation in allogeneic grafts than syngeneic grafts on day 10. The uptake of 125I-anti-IL-27Rα mAb in allogeneic grafts could be almost totally blocked by pre-injection with excess unlabeled anti-IL-27Rα mAb. Interestingly, we found that 125I-anti-IL-27Rα mAb accumulated in allogeneic grafts, along with weaker inflammation earlier on day 6. The high uptake of 125I-anti-IL-27Rα mAb was correlated with the higher infiltrated IL-27Rα positive cells (CD3+/CD68+) in allogeneic grafts. In conclusion, IL-27Rα may be a novel molecular imaging marker to predict allorejection.


Subject(s)
Biomarkers/metabolism , Graft Rejection/genetics , Molecular Imaging , Receptors, Interleukin/genetics , Allografts , Animals , Antibodies, Monoclonal/immunology , Gene Expression Regulation, Developmental/genetics , Graft Rejection/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Interleukin/isolation & purification , Receptors, Interleukin/metabolism , Skin Transplantation/adverse effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tissue Distribution/immunology , Transplantation, Homologous
4.
J Nanobiotechnology ; 16(1): 62, 2018 Aug 30.
Article in English | MEDLINE | ID: mdl-30165861

ABSTRACT

BACKGROUND: Epidermal growth factor (EGF) can promote cell proliferation as well as migration, which is feasible in tissue wound healing. Oil bodies have been exploited as an important platform to produce exogenous proteins. The exogenous proteins were expressed in oil bodies from plant seeds. The process can reduce purification steps, thereby significantly reducing the purification cost. Mostly, the diameter of oil body particle ranges between 1.0 and 1.5 µm in the safflower seeds, however, it reduces to 700-1000 nm in the transgenic safflower seeds. The significant reduction of particle size in transgenic seeds is extremely beneficial to skin absorption. RESULTS: The diameter of oil body in the transgenic safflower seeds was recorded in the range of 700-1000 nm. The smaller particle size improved their skin absorption. The expression level of oleosin-hEGF-hEGF in T3 transgenic seeds was highest at 69.32 mg/g of seeds. The oil body expressing oleosin-hEGF-hEGF had significant proliferative activity on NIH/3T3 cells and improved skin regeneration thereby accelerating wound healing in rats. The wound coverage rate exceeded 98% after treatment for 14 days with oil body expressing oleosin-hEGF-hEGF, while the saline without EGF group and wild type oil body group both showed less than 80%. The neonatal fibroblast and collagen were found to be increased in the safflower oil body expressing oleosin-hEGF-hEGF treatment group. TGF-ß1, bFGF and VEGF were noted as important growth factors in the repair of cutaneous wounds. Their expression level increased after 4 and 7 day treatment, but decreased after 14 days. Therefore, it can promote skin regeneration to accelerate wounds healing. CONCLUSIONS: The expression of oleosin-hEGF-hEGF in T3 transgenic seeds was 80.43 ng/µL oil body. It had significant proliferative activity on NIH/3T3 cells and improved skin regeneration to accelerate wound healing in rats. The expression process of TGF-ß1, bFGF and VEGF increased at first and then gradually declined.


Subject(s)
Epidermal Growth Factor/chemistry , Lipid Droplets/chemistry , Plant Proteins/chemistry , Skin/metabolism , Wound Healing/drug effects , Animals , Cell Proliferation/drug effects , Drug Carriers/chemistry , Drug Carriers/therapeutic use , Female , Humans , Male , Mice , Mice, Inbred ICR , NIH 3T3 Cells , Particle Size , Plant Oils/chemistry , Rats , Rats, Wistar , Regeneration/drug effects , Seeds/chemistry , Surface Properties , Tissue Distribution/drug effects , Tissue Distribution/immunology
5.
BMC Biotechnol ; 17(1): 8, 2017 01 18.
Article in English | MEDLINE | ID: mdl-28100205

ABSTRACT

BACKGROUND: Preclinical research implementing fluorescence-based approaches is inevitable for drug discovery and technology. For example, a variety of contrast agents developed for biomedical imaging are usually evaluated in cell systems and animal models based on their conjugation to fluorescent dyes. Biodistribution studies of excised organs are often performed by macroscopic imaging, whereas the subcellular localization though vital, is often neglected or further validated by histological procedures. Available systems used to define the subcellular biodistribution of contrast agents such as intravital microscopes or ex vivo histological analysis are expensive and not affordable by the majority of researchers, or encompass tedious and time consuming steps that may modify the contrast agents and falsify the results. Thus, affordable and more reliable approaches to study the biodistribution of contrast agents are required. We developed fluorescent immunoliposomes specific for human fibroblast activation protein and murine endoglin, and used macroscopic fluorescence imaging and confocal microscopy to determine their biodistribution and subcellular localization in freshly excised mice organs at different time points post intravenous injection. RESULTS: Near infrared fluorescence macroscopic imaging revealed key differences in the biodistribution of the respective immunoliposomes at different time points post injection, which correlated to the first-pass effect as well as the binding of the probes to molecular targets within the mice organs. Thus, a higher accumulation and longer retention of the murine endoglin immunoliposomes was seen in the lungs, liver and kidneys than the FAP specific immunoliposomes. Confocal microscopy showed that tissue autofluorescence enables detection of organ morphology and cellular components within freshly excised, non-processed organs, and that fluorescent probes with absorption and emission maxima beyond the tissue autofluorescence range can be easily distinguished. Hence, the endoglin targeting immunoliposomes retained in some organs could be detected in the vascular endothelia cells of the organs. CONCLUSIONS: The underlying work represents a quick, effective and more reliable setup to validate the macroscopic and subcellular biodistribution of contrast agents in freshly excised animal organs. The approach will be highly beneficial to many researchers involved in nanodrug design or in fluorescence-based studies on disease pathogenesis.


Subject(s)
Antibodies, Monoclonal/immunology , Liposomes/immunology , Microscopy, Fluorescence/methods , Molecular Imaging/methods , Subcellular Fractions/immunology , Viscera/immunology , Animals , Female , In Vitro Techniques , Metabolic Clearance Rate/immunology , Mice , Mice, Nude , Microscopy, Confocal/methods , Organ Specificity/immunology , Tissue Distribution/immunology
6.
Pharm Res ; 33(3): 639-52, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26530460

ABSTRACT

PURPOSE: A double-mutant E224A/E262A full-length botulinum neurotoxin (BoNT) Type A with structural similarity to native BoNT/A but lacking the endopeptidase activity provides an ideal surrogate for testing pharmacokinetics and immunochemical characteristics of BoNT. METHODS: We determined lethality (LD50) of deactivated recombinant botulinum neurotoxin (drBoNT/A) to be 24.0 µg by intraperitoneal route (i.p). The polypeptide drBoNT/A labeled with near infra-red dye 800 (NIR 800) was used to examine its distribution to different organs using whole body imaging when administered to mice via intravenous (i.v) or i.p route. Also, drBoNT/A was used to evaluate its immunogenicity in Balb/C mice model. RESULTS: drBoNT/A was found to be highly immunogenic when tested under various in vivo conditions in Balb/C mice model. For the first time we have demonstrated that a full length 150 kDa drBoNT/A, by administering via inhalation route in mice model, has evoked both circulating immunoglobulin levels of IgG and secretory IgA at the mucosal surface. The immunoglobulin levels were sufficient enough to protect against the challenge dose of native BoNT toxin in mice model. Tissue distribution of drBoNT/A seems to be similar to that of native toxin. CONCLUSIONS: Based on the characteristics described in this report this nontoxic holotoxin protein will assist us to explore the window of opportunity available for therapeutic treatment in case of unnatural poisoning, and also it can be an effective vaccine candidate.


Subject(s)
Antibody Formation/immunology , Botulinum Toxins, Type A/immunology , Recombinant Proteins/immunology , Animals , Cell Line, Tumor , Female , Immunoglobulin A, Secretory/immunology , Immunoglobulin G/immunology , Mice , Mice, Inbred BALB C , Tissue Distribution/immunology
7.
J Immunol ; 190(6): 2479-82, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23456700

ABSTRACT

The autoimmune regulator is a critical transcription factor for generating central tolerance in the thymus. Recent studies have revealed how the autoimmune regulator targets many otherwise tissue-restricted Ag genes to enable negative selection of autoreactive T cells.


Subject(s)
Antigens/genetics , Gene Expression Regulation/immunology , Transcription Factors/physiology , Amino Acid Motifs/genetics , Animals , Antigens/biosynthesis , Antigens/chemistry , Conserved Sequence/immunology , Humans , Immune Tolerance/genetics , Mice , Mice, Knockout , Polyendocrinopathies, Autoimmune/genetics , Polyendocrinopathies, Autoimmune/immunology , Polyendocrinopathies, Autoimmune/metabolism , Protein Interaction Mapping/methods , Tissue Distribution/genetics , Tissue Distribution/immunology , Transcription Factors/chemistry , AIRE Protein
8.
J Immunol ; 190(6): 2641-9, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23408837

ABSTRACT

Genome-wide association studies of complex immune-mediated diseases have indicated that many genetic factors, each with individual low risk, contribute to overall disease. It is therefore timely and important to characterize how immune responses may be subtly modified by tissue context. In this article, we explore the role of tissue-derived molecules in influencing the function of T cells, which, owing to their migratory nature, come into contact with many different microenvironments through their lifespan. Hedgehog (Hh) proteins act as secreted morphogens, providing concentration-dependent positional and temporal cell-fate specification in solid tissues. Hh signaling is required for embryogenesis and is important in postnatal tissue renewal and in malignancy. However, the function of Hh in dynamic, fluid systems, such as in mammalian immunity, is largely unknown. In this article, we show that Hh-dependent transcription in T cells promoted Th2 transcriptional programs and differentiation, exacerbating allergic disease. Of interest, expression of Sonic Hh increased in lung epithelial cells following the induction of allergic disease, and lung T cells upregulated Hh target gene expression, indicating that T cells respond to locally secreted Hh ligands in vivo. We show that Il4, the key Th2 cytokine, is a novel transcriptional target of Hh signals in T cells, providing one mechanism for the role of Hh in Th differentiation. We propose that Hh, secreted from inflamed, remodeling, or malignant tissue, can modulate local T cell function. Our data present an unexpected and novel role for tissue-derived morphogens in the regulation of fluid immune responses, with implications for allergy and tumor responses, suggesting new uses for anti-Hh therapeutics.


Subject(s)
Asthma/immunology , Asthma/metabolism , Cell Differentiation/immunology , Hedgehog Proteins/physiology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , Asthma/pathology , Cells, Cultured , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/pathology , Tissue Distribution/immunology , Transcription, Genetic/immunology
9.
J Immunol ; 190(4): 1685-94, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23296707

ABSTRACT

TLRs are essential for sensing the invading pathogens and initiating protective immune responses. However, aberrant activation of TLR-triggered inflammatory innate responses leads to the inflammatory disorders and autoimmune diseases. The molecular mechanisms that fine-tune TLR responses remain to be fully elucidated. Protein tyrosine phosphatase with proline-glutamine-serine-threonine-rich motifs (PTP-PEST) has been shown to be important in cell adhesion, migration, and also T cell and B cell activation. However, the roles of PTP-PEST in TLR-triggered immune response remain unclear. In this study, we report that PTP-PEST expression was upregulated in macrophages by TLR ligands. PTP-PEST inhibited TNF-α, IL-6, and IFN-ß production in macrophages triggered by TLR3, TLR4, and TLR9. Overexpression of catalytically inactive mutants of PTP-PEST abolished the inhibitory effects, indicating that PTP-PEST inhibits TLR response in a phosphatase-dependent manner. Accordingly, PTP-PEST knockdown increased TLR3, -4, and -9-triggered proinflammatory cytokine and type I IFN production. PTP-PEST selectively inhibited TLR-induced NF-κB activation, whereas it had no substantial effect on MAPK and IFN regulatory factor 3 activation. Moreover, PTP-PEST directly interacted with IκB kinase ß (IKKß) then inhibited IKKß phosphorylation at Ser(177/181) and Tyr(188/199), and subsequently suppressed IKKß activation and kinase activity as well as downstream NF-κB activation, resulting in suppression of the TLR-triggered innate immune response. Thus, PTP-PEST functions as a feedback-negative regulator of TLR-triggered innate immune responses by selectively impairing IKKß/NF-κB activation.


Subject(s)
Down-Regulation/immunology , I-kappa B Kinase/antagonists & inhibitors , Immunity, Innate , NF-kappa B/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 12/physiology , Toll-Like Receptors/physiology , Amino Acid Motifs/genetics , Amino Acid Motifs/immunology , Animals , Cell Line , Cells, Cultured , Down-Regulation/genetics , Glutamine/metabolism , HEK293 Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Immunity, Innate/genetics , Macrophages, Peritoneal/enzymology , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , Proline/metabolism , Protein Interaction Mapping/methods , Protein Tyrosine Phosphatase, Non-Receptor Type 12/biosynthesis , Serine/metabolism , Threonine/metabolism , Tissue Distribution/genetics , Tissue Distribution/immunology , Toll-Like Receptors/antagonists & inhibitors , Toll-Like Receptors/genetics
10.
Mol Pharm ; 11(9): 3090-6, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25077373

ABSTRACT

Radioimmunotherapy (RIT) of solid tumors is hampered by low tumor-to-nontumor (T/NT) ratios of the radiolabeled monoclonal antibodies resulting in low tumor doses in patients. Pretargeting technologies can improve the effectiveness of RIT in cancer therapy by increasing this ratio. We showed that a pretargeting strategy employing in vivo chemistry in combination with clearing agents, proceeds efficiently in tumor-bearing mice resulting in high T/NT ratios. A dosimetry study indicated that the chemical pretargeting technology, which centered on the bioorthogonal Diels-Alder click reaction between a radiolabeled tetrazine probe and a trans-cyclooctene-oxymethylbenzamide-tagged CC49 antibody (CC49-TCO(1)), can match the performance of clinically validated high-affinity biological pretargeting approaches in mice ( Rossin J Nucl Med. 2013 , 54 , 1989 - 1995 ). Nevertheless, the increased protein surface hydrophobicity of CC49-TCO(1) led to a relatively rapid blood clearance and concomitant reduced tumor uptake compared to native CC49 antibody. Here, we present the in vivo evaluation of a TCO-oxymethylacetamide-tagged CC49 antibody (CC49-TCO(2)), which is highly reactive toward tetrazines and less hydrophobic than CC49-TCO(1). CC49-TCO(2) was administered to healthy mice to determine its blood clearance and the in vivo stability of the TCO. Next, pretargeting biodistribution and SPECT studies with CC49-TCO(2), tetrazine-functionalized clearing agent, and radiolabeled tetrazine were carried out in nude mice bearing colon carcinoma xenografts (LS174T). CC49-TCO(2) had an increased circulation half-life, a 1.5-fold higher tumor uptake, and a 2.6-fold improved in vivo TCO stability compared to the more hydrophobic TCO-benzamide-CC49. As a consequence, and despite the 2-fold lower reactivity of CC49-TCO(2) toward tetrazines compared with CC49-TCO(1), administration of radiolabeled tetrazine afforded a significantly increased tumor accumulation and improved T/NT ratios in mice pretargeted with CC49-TCO(2). In conclusion, the TCO-acetamide derivative represents a large improvement in in vivo Diels-Alder pretargeting, possibly enabling application in larger animals and eventually humans.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/radiotherapy , Cyclooctanes/chemistry , Cyclooctanes/therapeutic use , Immunoconjugates/chemistry , Immunoconjugates/pharmacology , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Carcinoma/drug therapy , Carcinoma/immunology , Carcinoma/radiotherapy , Cell Line, Tumor , Colonic Neoplasms/immunology , Cycloaddition Reaction/methods , Cyclooctanes/immunology , Female , Half-Life , Humans , Immunoconjugates/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Radioimmunotherapy/methods , Radiometry/methods , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/immunology , Radiopharmaceuticals/pharmacology , Tissue Distribution/immunology
11.
Pharm Res ; 31(1): 216-27, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23949250

ABSTRACT

PURPOSE: To monitor the biodistribution of IgG1 aggregates upon subcutaneous (SC) and intravenous (IV) administration in mice and measure their propensity to stimulate an early immune response. METHODS: A human mAb (IgG1) was fluorescently labeled, aggregated by agitation stress and injected in SKH1 mice through SC and IV routes. The biodistribution of monomeric and aggregated formulations was monitored over 47 days by fluorescence imaging and the early immune response was measured by quantifying the level of relevant cytokines in serum using a Bio-plex assay. RESULTS: The aggregates remained at the SC injection site for a longer time than monomers but after entry into the systemic circulation disappeared faster than monomers. Upon IV administration, both monomers and aggregates spread rapidly throughout the circulation, and a strong accumulation in the liver was observed for both species. Subsequent removal from the circulation was faster for aggregates than monomers. No accumulation in lymph nodes was observed after SC or IV administration. Administration of monomers and aggregates induced similar cytokine levels, but SC injection resulted in higher cytokine levels than IV administration. CONCLUSION: These results show differences in biodistribution and residence time between IgG1 aggregates and monomers. The long residence time of aggregates at the SC injection site, in conjunction with elevated cytokine levels, may contribute to an enhanced immunogenicity risk of SC injected aggregates compared to that of monomers.


Subject(s)
Immunoglobulin G/immunology , Tissue Distribution/immunology , Animals , Antibodies, Monoclonal/immunology , Cytokines/immunology , Female , Fluorescence , Humans , Injections, Intravenous/methods , Injections, Subcutaneous/methods , Lymph Nodes/immunology , Mice , Optical Imaging/methods
12.
J Immunol ; 188(10): 4866-75, 2012 May 15.
Article in English | MEDLINE | ID: mdl-22504644

ABSTRACT

Differentiation and maintenance of recirculating effector memory CD8 T cells (T(EM)) depends on prolonged cognate Ag stimulation. Whether similar pathways of differentiation exist for recently identified tissue-resident effector memory T cells (T(RM)), which contribute to rapid local protection upon pathogen re-exposure, is unknown. Memory CD8αß(+) T cells within small intestine epithelium are well-characterized examples of T(RM), and they maintain a long-lived effector-like phenotype that is highly suggestive of persistent Ag stimulation. This study sought to define the sources and requirements for prolonged Ag stimulation in programming this differentiation state, including local stimulation via cognate or cross-reactive Ags derived from pathogens, microbial flora, or dietary proteins. Contrary to expectations, we found that prolonged cognate Ag stimulation was dispensable for intestinal T(RM) ontogeny. In fact, chronic antigenic stimulation skewed differentiation away from the canonical intestinal T cell phenotype. Resident memory signatures, CD69 and CD103, were expressed in many nonlymphoid tissues including intestine, stomach, kidney, reproductive tract, pancreas, brain, heart, and salivary gland and could be driven by cytokines. Moreover, TGF-ß-driven CD103 expression was required for T(RM) maintenance within intestinal epithelium in vivo. Thus, induction and maintenance of long-lived effector-like intestinal T(RM) differed from classic models of T(EM) ontogeny and were programmed through a novel location-dependent pathway that was required for the persistence of local immunological memory.


Subject(s)
Cell Differentiation/immunology , Epitopes, T-Lymphocyte/physiology , Immunologic Memory/immunology , T-Lymphocyte Subsets/immunology , Animals , Cell Line , Female , Immunophenotyping , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/pathogenicity , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Organ Specificity/immunology , Signal Transduction/immunology , T-Lymphocyte Subsets/pathology , T-Lymphocyte Subsets/virology , Time Factors , Tissue Distribution/immunology
13.
J Immunol ; 189(9): 4459-69, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23024272

ABSTRACT

The closely linked human IL-3 and GM-CSF genes are tightly regulated and are expressed in activated T cells and mast cells. In this study, we used transgenic mice to study the developmental regulation of this locus and to identify DNA elements required for its correct activity in vivo. Because these two genes are separated by a CTCF-dependent insulator, and the GM-CSF gene is regulated primarily by its own upstream enhancer, the main objective in this study was to identify regions of the locus required for correct IL-3 gene expression. We initially found that the previously identified proximal upstream IL-3 enhancers were insufficient to account for the in vivo activity of the IL-3 gene. However, an extended analysis of DNase I-hypersensitive sites (DHSs) spanning the entire upstream IL-3 intergenic region revealed the existence of a complex cluster of both constitutive and inducible DHSs spanning the -34- to -40-kb region. The tissue specificity of these DHSs mirrored the activity of the IL-3 gene, and included a highly inducible cyclosporin A-sensitive enhancer at -37 kb that increased IL-3 promoter activity 40-fold. Significantly, inclusion of this region enabled correct in vivo regulation of IL-3 gene expression in T cells, mast cells, and myeloid progenitor cells.


Subject(s)
Enhancer Elements, Genetic/immunology , Gene Expression Regulation, Developmental/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Interleukin-3/biosynthesis , Interleukin-3/genetics , Animals , Cell Line , Cell Line, Tumor , Cells, Cultured , Deoxyribonuclease I/genetics , Enhancer Elements, Genetic/genetics , Genetic Loci/immunology , Humans , Jurkat Cells , Mice , Mice, Transgenic , Tissue Distribution/genetics , Tissue Distribution/immunology
14.
J Immunol ; 188(2): 531-5, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22180615

ABSTRACT

The development of chronic allergic dermatitis in early life has been associated with increased onset and severity of allergic asthma later in life. However, the mechanisms linking these two diseases are poorly understood. In this study, we report that the development of oxazolone-induced chronic allergic dermatitis, in a mouse model, caused enhanced OVA-induced allergic asthma after the resolution of the former disease. Our findings show that oxazolone-induced dermatitis caused a marked increase in tissue mast cells, which persisted long after the resolution of this disease. Subsequent OVA sensitization and airway challenge of mice that had recovered from dermatitis resulted in increased allergic airway hyperreactivity. The findings demonstrate that the accumulation of mast cells during dermatitis has the detrimental effect of increasing allergic airway hypersensitivity. Importantly, our findings also show that exposure to a given allergen can modify the immune response to an unrelated allergen.


Subject(s)
Dermatitis, Atopic/immunology , Dermatitis, Atopic/pathology , Mast Cells/immunology , Mast Cells/pathology , Respiratory Hypersensitivity/immunology , Respiratory Hypersensitivity/pathology , Animals , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/pathology , Cell Count , Chronic Disease , Dermatitis, Atopic/chemically induced , Disease Models, Animal , Disease Progression , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Ovalbumin/administration & dosage , Oxazolone/administration & dosage , Tissue Distribution/genetics , Tissue Distribution/immunology
15.
J Immunol ; 189(6): 3054-63, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22855708

ABSTRACT

B7x (B7-H4 or B7S1), a member of the B7 family, inhibits in vitro T cell proliferation and cytokine production by binding to an unidentified receptor on activated T cells, but its in vivo function remains largely unclear. We show that B7x protein was expressed in epithelial cells of the lung, but not in lymphoid tissues. To investigate the role of B7x in the lung, we determined the susceptibility of B7x-deficient (B7x(-/-)) mice to a lethal pulmonary infection with Streptococcus pneumoniae. B7x(-/-), but not B7-H3-deficient, mice were significantly more resistant to S. pneumoniae pulmonary infection than their wild-type (Wt) counterparts. B7x(-/-) mice had significantly lower bacterial burdens and levels of inflammatory cytokines in lungs as early as 12 h postinfection. They also had milder immunopathology that was localized in alveolar spaces, whereas Wt mice had severe inflammation that was perivascular. Control of infection in B7x(-/-) mice was associated with a marked increase in activated CD4 and CD8 T cells and fewer neutrophils in lungs, whereas the susceptible Wt mice had the opposite cellular profile. In B7x(-/-)Rag1(-/-) mice that lack T cells, reduction in bacterial burden was no longer observed. Control of S. pneumoniae and the increased survival observed was specific to the lung, because systemically infected B7x(-/-) mice were not resistant to infection. These data indicate that lung-expressed B7x negatively regulates T cells, and that in its absence, in B7x(-/-) mice, an enhanced T cell response contributed to reduced lethality in a pulmonary infection model with S. pneumoniae.


Subject(s)
Pneumonia, Pneumococcal/immunology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/physiology , Animals , Disease Resistance/genetics , Disease Resistance/immunology , Lung/immunology , Lung/microbiology , Lung/pathology , Lymphoid Tissue/immunology , Lymphoid Tissue/microbiology , Lymphoid Tissue/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Outcome Assessment, Health Care , Pneumonia, Pneumococcal/mortality , Pneumonia, Pneumococcal/pathology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , T-Lymphocytes/pathology , Tissue Distribution/genetics , Tissue Distribution/immunology , V-Set Domain-Containing T-Cell Activation Inhibitor 1/deficiency , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics
16.
J Immunol ; 188(5): 2387-98, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22291183

ABSTRACT

Ectoenzymes expressed on the surface of vascular cells and leukocytes modulate the ambient nucleotide milieu. CD73 is an ecto-5' nucleotidase that catalyzes the terminal phosphohydrolysis of AMP and resides in the brain on glial cells, cells of the choroid plexus, and leukocytes. Though CD73 tightens epithelial barriers, its role in the ischemic brain remains undefined. When subjected to photothrombotic arterial occlusion, CD73(-/-) mice exhibited significantly larger (49%) cerebral infarct volumes than wild-type mice, with concordant increases in local accumulation of leukocyte subsets (neutrophils, T lymphocytes, macrophages, and microglia). CD73(-/-) mice were rescued from ischemic neurologic injury by soluble 5'-nucleotidase. In situ, CD73(-/-) macrophages upregulated expression of costimulatory molecules far more than wild-type macrophages, with a sharp increase of the CD80/CD86 ratio. To define the CD73-bearing cells responsible for ischemic cerebroprotection, mice were subjected to irradiative myeloablation, marrow reconstitution, and then stroke following engraftment. Chimeric mice lacking CD73 in tissue had larger cerebral infarct volumes and more tissue leukosequestration than did mice lacking CD73 on circulating cells. These data show a cardinal role for CD73 in suppressing ischemic tissue leukosequestration. This underscores a critical role for CD73 as a modulator of brain inflammation and immune function.


Subject(s)
5'-Nucleotidase/physiology , Brain Ischemia/immunology , Brain Ischemia/pathology , Cell Movement/genetics , Cell Movement/immunology , Leukocytes/immunology , Leukocytes/pathology , 5'-Nucleotidase/deficiency , 5'-Nucleotidase/genetics , Adenosine/biosynthesis , Adenosine/physiology , Animals , Bone Marrow Transplantation/immunology , Bone Marrow Transplantation/pathology , Brain Edema/enzymology , Brain Edema/immunology , Brain Edema/pathology , Brain Ischemia/enzymology , Extracellular Fluid/enzymology , Extracellular Fluid/immunology , Infarction, Middle Cerebral Artery/enzymology , Infarction, Middle Cerebral Artery/immunology , Infarction, Middle Cerebral Artery/pathology , Inflammation/enzymology , Inflammation/immunology , Inflammation/prevention & control , Leukocytes/enzymology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Signal Transduction/immunology , Tissue Distribution/genetics , Tissue Distribution/immunology
17.
Blood ; 118(11): 3039-50, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21791416

ABSTRACT

Memory T cells (T(M)s) have been detected in many tissues but their quantitative distribution remains largely undefined. We show that in mice there is a remarkably biased accumulation of long-term CD4 T(M)s into mucosal sites (mainly gut, especially Peyer patches), and CD8 T(M)s into lymph nodes and spleen (in particular, peripheral lymph nodes [PLNs]). This distinction correlates with their differentiated expression of PLN- and gut-homing markers. CD8 and CD4 T(M)s selectively require the expression of PLN-homing marker CCR7 or gut-homing marker α4ß7 for maintenance. PLNs and gut supply CD8 and CD4 T(M)s with their individually favored homeostatic cytokine, IL-15, or IL-7. Cytokine stimulation in turn regulates the different gut-homing marker expression on CD4 and CD8 T(M)s. IL-15 plays a major role in vivo regulating CD8 T(M)s homing to PLNs. Thus, the reservoir segregation of CD4 and CD8 T(M)s meets their individual needs for homeostatic cytokines and is under feedback control of cytokine stimulation.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Chemotaxis, Leukocyte/physiology , Cytokines/physiology , Animals , Biomarkers/analysis , Biomarkers/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chemotaxis, Leukocyte/immunology , Cytokines/metabolism , Female , Homeostasis/immunology , Humans , Lymph Nodes/cytology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Stem Cell Niche/immunology , Tissue Distribution/immunology
18.
Allergy ; 68(11): 1467-70, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24118000

ABSTRACT

Overproduction of periostin, an IL-13-inducible matricellular protein, despite corticosteroid treatment is thought to be involved in the chronicity of allergic inflammation seen in corticosteroid-refractory tissue fibrosis. Therefore, we hypothesized that some tissue cells must produce periostin in a corticosteroid-insensitive manner. Here, we show that IL-4 and IL-13 each induced comparable levels of periostin production by primary normal human fibroblasts and microvascular endothelial cells derived from lung and skin. Dexamethasone, a corticosteroid, completely inhibited IL-4/13-induced, but did not affect TGF-ß-induced, periostin production by fibroblasts. In contrast, dexamethasone synergistically enhanced IL-4/13-induced periostin production by microvascular endothelial cells. TGF-ß did not induce periostin production by microvascular endothelial cells. Our novel findings suggest that IL-4/13-induced microvascular endothelium-derived and/or TGF-ß-induced fibroblast-derived periostin might play a pivotal role in corticosteroid-refractory tissue fibrosis, leading to chronic allergic inflammation in the lung and/or skin.


Subject(s)
Cell Adhesion Molecules/biosynthesis , Dexamethasone/pharmacology , Cell Adhesion Molecules/antagonists & inhibitors , Cell-Free System , Fibroblasts/immunology , Fibroblasts/pathology , Fibrosis/immunology , Human Umbilical Vein Endothelial Cells , Humans , Inflammation Mediators/physiology , Interleukin-13/physiology , Interleukin-4/physiology , Lung/immunology , Lung/pathology , Skin/immunology , Skin/pathology , Tissue Distribution/immunology , Transforming Growth Factor beta1/physiology
19.
Mol Pharm ; 10(12): 4527-33, 2013 Dec 02.
Article in English | MEDLINE | ID: mdl-24147882

ABSTRACT

Accumulating evidence suggests that overexpression of the tyrosine kinase receptor EphB4, a mediator of vascular development, is a novel target for tumor diagnosis, prognosis and therapy. Noninvasive imaging of EphB4 expression could therefore be valuable for evaluating disease course and therapeutic efficacy at the earliest stages of anti-EphB4 treatment. In this study, we systematically investigated the use of anti-EphB4 antibody h131 (150 kDa) and its fragments (h131-F(ab')2, 110 kDa; h131-Fab, 50 kDa) for near-infrared fluorescence (NIRF) imaging of EphB4 expression in vivo. h131-F(ab')2 and h131-Fab were produced through pepsin and papain digestion of h131 respectively, whose purity was confirmed by FPLC and SDS-PAGE. After conjugation with Cy5.5, in vivo characteristics of h131, h131-F(ab')2 and h131-Fab were evaluated in EphB4-positive HT29 tumor model. Although h131-Cy5.5 demonstrated highest tumor uptake among these probes, its optimal tumor uptake level was obtained at 2 days post injection (p.i.). For h131-Fab-Cy5.5, maximum tumor uptake was achieved at 4 h p.i. However, no significant difference was observed between h131-Fab-Cy5.5 and hIgG-Fab-Cy5.5, indicating the tumor accumulation was mainly caused by passive targeting. In contrast, h131-F(ab')2-Cy5.5 demonstrated prominent tumor uptake at 6 h p.i. The target specificity was confirmed by hIgG-F(ab')2-Cy5.5 control and immunofluorescent staining. Collectively, h131-F(ab')2 exhibited prominent and specific tumor uptake at early time points, which suggests it is a promising agent for EphB4-targeted imaging.


Subject(s)
Antibodies, Monoclonal/immunology , Neoplasms/diagnosis , Neoplasms/immunology , Receptor, EphB4/immunology , Cell Line, Tumor , Diagnostic Imaging/methods , HT29 Cells , Humans , Tissue Distribution/immunology
20.
J Immunol ; 186(6): 3283-8, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21368237

ABSTRACT

IL-9 was first described in the late 1980s as a member of a growing number of cytokines that had pleiotropic functions in the immune system. Although many biological functions have been attributed to IL-9, it remains an understudied cytokine. A resurgence of interest in IL-9 has been spurred by recent work demonstrating a role for IL-9 in regulating inflammatory immunity and defining the transcription factors that activate the Il9 gene in cells that most efficiently produce IL-9. In this review, we summarize the characterization of IL-9 biological activities, highlight roles for the cytokine that are clearly defined, and outline questions regarding IL-9 functions that still require further exploration.


Subject(s)
Interleukin-9/chemistry , Interleukin-9/physiology , Animals , Gene Expression Regulation/immunology , Humans , Immunity, Cellular , Interleukin-9/biosynthesis , Interleukin-9/genetics , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Receptors, Interleukin-9/biosynthesis , Receptors, Interleukin-9/genetics , Receptors, Interleukin-9/physiology , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tissue Distribution/genetics , Tissue Distribution/immunology
SELECTION OF CITATIONS
SEARCH DETAIL