Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 391
Filter
1.
Eur J Immunol ; 54(10): e2451207, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38980268

ABSTRACT

Tertiary lymphoid structures (TLS) resemble follicles of secondary lymphoid organs and develop in nonlymphoid tissues during inflammation and cancer. Which cell types and signals drive the development of TLS is largely unknown. To investigate early events of TLS development in the lungs, we repeatedly instilled p(I:C) plus ovalbumin (Ova) intranasally. This induced TLS ranging from lymphocytic aggregates to organized and functional structures containing germinal centers. We found that TLS development is independent of FAP+ fibroblasts, alveolar macrophages, or CCL19 but crucially depends on type I interferon (IFN-I). Mechanistically, IFN-I initiates two synergistic pathways that culminate in the development of TLS. On the one hand, IFN-I induces lymphotoxin (LT)α in lymphoid cells, which stimulate stromal cells to produce the B-cell-attracting chemokine CXCL13 through LTßR-signaling. On the other hand, IFN-I is sensed by stromal cells that produce the T-cell-attracting chemokines CXCL9, CXCL10 as well as CCL19 and CCL21 independently of LTßR. Consequently, B-cell aggregates develop within a week, whereas follicular dendritic cells and germinal centers appear after 3 weeks. Thus, sustained production of IFN-I together with an antigen is essential for the induction of functional TLS in the lungs.


Subject(s)
Immunity, Innate , Interferon Type I , Tertiary Lymphoid Structures , Animals , Tertiary Lymphoid Structures/immunology , Mice , Interferon Type I/metabolism , Interferon Type I/immunology , Immunity, Innate/drug effects , Chemokine CCL19/metabolism , Lung/immunology , Chemokine CCL21/metabolism , Chemokine CXCL13/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/drug effects , Lymphotoxin beta Receptor/metabolism , Lymphotoxin beta Receptor/immunology , Mice, Inbred C57BL , Stromal Cells/immunology , Stromal Cells/drug effects , Stromal Cells/metabolism , Lymphotoxin-alpha/metabolism , Lymphotoxin-alpha/immunology , Germinal Center/immunology , Ovalbumin/immunology , Ovalbumin/administration & dosage , Signal Transduction/immunology , Signal Transduction/drug effects , Fibroblasts/immunology , Fibroblasts/drug effects , Macrophages, Alveolar/immunology , Macrophages, Alveolar/drug effects , Chemokine CXCL10/metabolism , Chemokine CXCL10/immunology , Mice, Knockout , Chemokine CXCL9/metabolism
2.
J Exp Med ; 218(5)2021 05 03.
Article in English | MEDLINE | ID: mdl-33724364

ABSTRACT

The spleen contains a myriad of conventional dendritic cell (cDC) subsets that protect against systemic pathogen dissemination by bridging antigen detection to the induction of adaptive immunity. How cDC subsets differentiate in the splenic environment is poorly understood. Here, we report that LTα1ß2-expressing Rorgt+ ILC3s, together with B cells, control the splenic cDC niche size and the terminal differentiation of Sirpα+CD4+Esam+ cDC2s, independently of the microbiota and of bone marrow pre-cDC output. Whereas the size of the splenic cDC niche depended on lymphotoxin signaling only during a restricted time frame, the homeostasis of Sirpα+CD4+Esam+ cDC2s required continuous lymphotoxin input. This latter property made Sirpα+CD4+Esam+ cDC2s uniquely susceptible to pharmacological interventions with LTßR agonists and antagonists and to ILC reconstitution strategies. Together, our findings demonstrate that LTα1ß2-expressing Rorgt+ ILC3s drive splenic cDC differentiation and highlight the critical role of ILC3s as perpetual regulators of lymphoid tissue homeostasis.


Subject(s)
Dendritic Cells/immunology , Immunity, Innate , Lymphoid Tissue/immunology , Lymphotoxin-alpha/immunology , Signal Transduction/immunology , Spleen/immunology , Animals , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Dendritic Cells/metabolism , Female , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/immunology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Signal Transduction/genetics , Spleen/cytology , Spleen/metabolism
3.
J BUON ; 25(3): 1643-1649, 2020.
Article in English | MEDLINE | ID: mdl-32862617

ABSTRACT

PURPOSE: To explore the efficacy and safety of irreversible electroporation (IRE) ablation combined with natural killer (NK) cells in the treatment of locally advanced pancreatic cancer (LAPC). METHODS: A total of 92 LAPC patients treated in our hospital from January 2016 to January 2017 were enrolled, and there were 46 cases of percutaneous IRE as IRE group, and 46 cases of IRE combined NK cell therapy as IRE-NK group. The clinical information of all the patients was collected, and the short-term efficacy, changes in the serum immunological indicators after treatment, carbohydrate antigen 19-9 (CA19-9) level, and incidence of adverse reactions were compared between the two groups of patients. Besides, the overall survival (OS) and disease-free survival (DFS) of patients were followed up and recorded. RESULTS: On 1 month after treatment, all the patients underwent efficacy assessment, which showed the overall response rate of patients in IRE-NK group was significantly superior to that in IRE group. One and 7 days after operation, the level of CA19-9 was obviously raised in the two groups, with a statistically significant difference, and it declined 30 days postoperatively. Seven and 30 days after operation IRE-NK group had a notably lower level of CA19-9 than IRE group. After treatment, all the patients exhibited considerably higher lymphocyte count and notably enhanced lymphocyte function, and all the indicators in IRE-NK group were higher than those in IRE group. Besides, the levels of serum interleukin (IL)-2, TNF-ß and IFN-γ in IRE-NK group were remarkably higher than those in IRE group, whereas there were no statistically significant differences in the levels of IL-4, IL-6 and IL-10 between the two groups. All the patients were followed up for 6-29 months, and there were no statistically significant differences in the DFS and OS between IRE group and IRE-NK group. CONCLUSION: IRE ablation combined with NK cells has excellent efficacy in treating LAPC, and they can exert a synergistic treatment effect to enhance the immune function of patients and reduce CA19-9 expression, with tolerable adverse reactions.


Subject(s)
Killer Cells, Natural/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , CA-19-9 Antigen/immunology , Combined Modality Therapy/methods , Disease-Free Survival , Electroporation/methods , Female , Humans , Immunotherapy, Adoptive/methods , Interferon-gamma/immunology , Interleukins/immunology , Lymphotoxin-alpha/immunology , Male , Middle Aged , Pancreas/immunology , Pancreas/metabolism , Prospective Studies
4.
Ginekol Pol ; 91(6): 301-307, 2020.
Article in English | MEDLINE | ID: mdl-32627150

ABSTRACT

OBJECTIVES: Human papillomavirus (HPV) ranks the first cause of cervical cancer. Cervical cancer has high prevalence rates in women around the world. The HPV-E7 oncoprotein is expressed in cervical cancer and is a target of developing immunotherapies against HPV-associated tumors. However, the antigenicity of this protein is low. Due to this reason, potent adjuvants are required to enhance its therapeutic efficacy. This preliminary study aims to evaluate whether lymphotoxin (LT) could act as an effective immune adjuvant for HPV infection in mice models. MATERIAL AND METHODS: Intranasal immunization was used to explore the effect of HPV-E7 and/or LT immune response. After the third intranasal immunization, the titer for the HPV-E7 antibody was detected in serum and vaginal washing fluid. Also, we assessed the expression of chemokine ligand 13 (CXCL13) and Peripheral Node Addressin (PNAd) in the lymph nodes after intranasal immunization with immunohistochemical analysis. RESULTS: compared to HPV-E7 immunization, intranasal immunization with HPV-E7 plus LT significantly increased HPV-E7-specific serum IgG and vaginal IgA titers. Furthermore, the combined use of HPV-E7 and LT strongly induced E7-specific CTL responses. CONCLUSIONS: LT can be effective for intranasal immunized HPV-E7 to improve E7-specific immune responses to HPV infection. It is new approach to eradicate chronic HPV infection capable of inducing an effective anti-infection method.


Subject(s)
Antigens, Viral/immunology , Lymphotoxin-alpha/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Infections/immunology , Viral Vaccines/immunology , Animals , Antibodies, Viral/blood , Disease Models, Animal , Female , Immunity , Immunotherapy , Mice , Papillomavirus Infections/prevention & control , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/prevention & control
5.
J Immunol ; 205(3): 853-863, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32591396

ABSTRACT

As a specialized subset of intestinal epithelial cells (IECs), goblet cells (GCs) play an important role during the antibacterial response via mucin production. However, the regulatory mechanisms involved in GC differentiation and function during infection, particularly the role of immune cell-IEC cross-talk, remain largely unknown. In this study, using Villin∆Ltbr conditional knockout mice, we demonstrate that LTßR, expressed on IECs, is required for GC hyperplasia and mucin 2 (MUC2) expression during Listeria infection for host defense but not homeostatic maintenance in the naive state. Analysis of single gene-deficient mice revealed that the ligand lymphotoxin (LT), but not LIGHT, and type 3 innate lymphoid cells (ILC3s), but not conventional T cells, are required for MUC2-dependent Listeria control. Conditional deficiency of LT in ILC3s further confirmed the importance of LT signals derived from ILC3s. Lack of ILC3-derived LT or IEC-derived LTßR resulted in the defective expression of genes related to GC differentiation but was not correlated with IEC proliferation and cell death, which were found to be normal by Ki-67 and Annexin V staining. In addition, the alternative NF-κB signaling pathway (involving RelB) in IECs was found to be required for the expression of GC differentiation-related genes and Muc2 and required for the anti-Listeria response. Therefore, our data together suggest a previously unrecognized ILC3-IEC interaction and LT-LTßR-RelB signaling axis governing GC differentiation and function during Listeria infection for host defense.


Subject(s)
Cell Differentiation/immunology , Goblet Cells/immunology , Listeria/immunology , Listeriosis/immunology , Lymphocytes/immunology , Lymphotoxin-alpha/immunology , Signal Transduction/immunology , Animals , Cell Differentiation/genetics , Goblet Cells/pathology , Listeriosis/genetics , Listeriosis/pathology , Lymphocytes/pathology , Lymphotoxin beta Receptor , Lymphotoxin-alpha/genetics , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , Signal Transduction/genetics
6.
BMC Evol Biol ; 19(1): 218, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31791241

ABSTRACT

BACKGROUND: Lymphotoxin-α (LTα), located in the Major Histocompatibility Complex (MHC) class III region on chromosome 6, encodes a cytotoxic protein that mediates a variety of antiviral responses among other biological functions. Furthermore, several genotypes at this gene have been implicated in the onset of a number of complex diseases, including myocardial infarction, autoimmunity, and various types of cancer. However, little is known about levels of nucleotide variation and linkage disequilibrium (LD) in and near LTα, which could also influence phenotypic variance. To address this gap in knowledge, we examined sequence variation across ~ 10 kilobases (kbs), encompassing LTα and the upstream region, in 2039 individuals from the 1000 Genomes Project originating from 21 global populations. RESULTS: Here, we observed striking patterns of diversity, including an excess of intermediate-frequency alleles, the maintenance of multiple common haplotypes and a deep coalescence time for variation (dating > 1.0 million years ago), in global populations. While these results are generally consistent with a model of balancing selection, we also uncovered a signature of positive selection in the form of long-range LD on chromosomes with derived alleles primarily in Eurasian populations. To reconcile these findings, which appear to support different models of selection, we argue that selective sweeps (particularly, soft sweeps) of multiple derived alleles in and/or near LTα occurred in non-Africans after their ancestors left Africa. Furthermore, these targets of selection were predicted to alter transcription factor binding site affinity and protein stability, suggesting they play a role in gene function. Additionally, our data also showed that a subset of these functional adaptive variants are present in archaic hominin genomes. CONCLUSIONS: Overall, this study identified candidate functional alleles in a biologically-relevant genomic region, and offers new insights into the evolutionary origins of these loci in modern human populations.


Subject(s)
Lymphotoxin-alpha/genetics , Major Histocompatibility Complex , Africa , Animals , Biological Evolution , Chromosomes, Human, Pair 6 , Evolution, Molecular , Gene Frequency , Genetics, Population , Haplotypes , Hominidae/genetics , Human Genome Project , Humans , Linkage Disequilibrium , Lymphotoxin-alpha/immunology , Polymorphism, Single Nucleotide
7.
Sci Immunol ; 4(42)2019 12 20.
Article in English | MEDLINE | ID: mdl-31862865

ABSTRACT

Redundant mechanisms support immunoglobulin A (IgA) responses to intestinal antigens. These include multiple priming sites [mesenteric lymph nodes (MLNs), Peyer's patches, and isolated lymphoid follicles] and various cytokines that promote class switch to IgA, even in the absence of T cells. Despite these backup mechanisms, vaccination against enteric pathogens such as rotavirus has limited success in some populations. Genetic and environmental signals experienced during early life are known to influence mucosal immunity, yet the mechanisms for how these exposures operate remain unclear. Here, we used rotavirus infection to follow antigen-specific IgA responses through time and in different gut compartments. Using genetic and pharmacological approaches, we tested the role of the lymphotoxin (LT) pathway-known to support IgA responses-at different developmental stages. We found that LT-ß receptor (LTßR) signaling in early life programs intestinal IgA responses in adulthood by affecting antibody class switch recombination to IgA and subsequent generation of IgA antibody-secreting cells within an intact MLN. In addition, early-life LTßR signaling dictates the phenotype and function of MLN stromal cells to support IgA responses in the adult. Collectively, our studies uncover new mechanistic insights into how early-life LTßR signaling affects mucosal immune responses during adulthood.


Subject(s)
Immunoglobulin A/immunology , Lymph Nodes/immunology , Lymphotoxin beta Receptor/immunology , Lymphotoxin-alpha/immunology , Mesentery/immunology , Stromal Cells/immunology , Animals , Feces/microbiology , Female , Immunity, Mucosal , Lymph Nodes/cytology , Lymphotoxin beta Receptor/genetics , Lymphotoxin-alpha/genetics , Male , Mesentery/cytology , Mice, Inbred C57BL , Mice, Knockout
8.
BMC Genomics ; 20(1): 878, 2019 Nov 20.
Article in English | MEDLINE | ID: mdl-31747871

ABSTRACT

BACKGROUND: The function of Toll-like receptor 2 (TLR2) in host defense against pathogens, especially Mycobacterium tuberculosis (Mtb) is poorly understood. To investigate the role of TLR2 during mycobacterial infection, we analyzed the response of tlr2 zebrafish mutant larvae to infection with Mycobacterium marinum (Mm), a close relative to Mtb, as a model for tuberculosis. We measured infection phenotypes and transcriptome responses using RNA deep sequencing in mutant and control larvae. RESULTS: tlr2 mutant embryos at 2 dpf do not show differences in numbers of macrophages and neutrophils compared to control embryos. However, we found substantial changes in gene expression in these mutants, particularly in metabolic pathways, when compared with the heterozygote tlr2+/- control. At 4 days after Mm infection, the total bacterial burden and the presence of extracellular bacteria were higher in tlr2-/- larvae than in tlr2+/-, or tlr2+/+ larvae, whereas granuloma numbers were reduced, showing a function of Tlr2 in zebrafish host defense. RNAseq analysis of infected tlr2-/- versus tlr2+/- shows that the number of up-regulated and down-regulated genes in response to infection was greatly diminished in tlr2 mutants by at least 2 fold and 10 fold, respectively. Analysis of the transcriptome data and qPCR validation shows that Mm infection of tlr2 mutants leads to decreased mRNA levels of genes involved in inflammation and immune responses, including il1b, tnfb, cxcl11aa/ac, fosl1a, and cebpb. Furthermore, RNAseq analyses revealed that the expression of genes for Maf family transcription factors, vitamin D receptors, and Dicps proteins is altered in tlr2 mutants with or without infection. In addition, the data indicate a function of Tlr2 in the control of induction of cytokines and chemokines, such as the CXCR3-CXCL11 signaling axis. CONCLUSION: The transcriptome and infection burden analyses show a function of Tlr2 as a protective factor against mycobacteria. Transcriptome analysis revealed tlr2-specific pathways involved in Mm infection, which are related to responses to Mtb infection in human macrophages. Considering its dominant function in control of transcriptional processes that govern defense responses and metabolism, the TLR2 protein can be expected to be also of importance for other infectious diseases and interactions with the microbiome.


Subject(s)
Fish Diseases/genetics , Gene Expression Regulation, Developmental , Mycobacterium Infections, Nontuberculous/genetics , Mycobacterium Infections, Nontuberculous/veterinary , Toll-Like Receptor 2/genetics , Zebrafish/genetics , Animals , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/immunology , Chemokine CXCL11/genetics , Chemokine CXCL11/immunology , Disease Resistance/genetics , Embryo, Nonmammalian , Fish Diseases/immunology , Fish Diseases/microbiology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immunity, Innate , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Larva/genetics , Larva/growth & development , Larva/immunology , Larva/microbiology , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/immunology , Macrophages/immunology , Macrophages/microbiology , Maf Transcription Factors/genetics , Maf Transcription Factors/immunology , Metabolic Networks and Pathways/genetics , Metabolic Networks and Pathways/immunology , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/microbiology , Mycobacterium marinum/immunology , Mycobacterium marinum/pathogenicity , Neutrophils/immunology , Neutrophils/microbiology , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/immunology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/immunology , Transcriptome/immunology , Zebrafish/growth & development , Zebrafish/immunology , Zebrafish/microbiology , Zebrafish Proteins/genetics , Zebrafish Proteins/immunology
9.
Cell Rep ; 27(8): 2442-2458.e5, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31116987

ABSTRACT

Adaptive type 2 immune responses against the intestinal helminth Heligmosomoides polygyrus (Hp) require the interaction of follicle-associated CXCR5+ dendritic cells with naive T cells in the draining mesenteric lymph nodes (mLNs). However, the source of CXCL13 responsible for attracting CXCR5+ dendritic cells has remained unclear. Using multiplex imaging combined with deep tissue analysis, we observed new CXCL13+ fibroblastic reticular cells surrounding paracortical and cortical B cell follicles in the mLNs of infected mice. CXCL13+ fibroblasts expressed markers of marginal reticular cells (MRCs), and their expansion required lymphotoxin (LT)-dependent interactions between IL-4Rα-expressing B cells and CCL19+ fibroblasts. Infection-induced follicles did not necessarily contain follicular dendritic cells (FDCs), indicating that CXCL13+ fibroblasts may instead drive their formation. These data reveal a role for lymphotoxin signaling to CCL19+ fibroblasts in the development of CXCL13+ MRC-like cells and adaptive type 2 immunity in response to helminth infection.


Subject(s)
B-Lymphocytes/metabolism , Chemokine CXCL13/metabolism , Receptors, Cell Surface/metabolism , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Chemokine CCL19/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Lymph Nodes/immunology , Lymph Nodes/parasitology , Lymph Nodes/pathology , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-alpha/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nematospiroides dubius/immunology , Nematospiroides dubius/pathogenicity , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/genetics , Signal Transduction , Stromal Cells/cytology , Stromal Cells/metabolism
10.
Bull Exp Biol Med ; 166(6): 770-773, 2019 Apr.
Article in English | MEDLINE | ID: mdl-31028583

ABSTRACT

Plant lipid transfer proteins and homologues of the main birch pollen allergen Bet v 1 are involved in the development of allergic reactions of varying severity to plant foods and pollen. In this study, the sera from patients with tree and weed pollen allergies in the Moscow region were examined. The levels of IL-4, IL-5, IL-9, IL-10, IL-13, IL-17A, IFNγ, TNFα, and TNFß cytokines were determined in the sera of patients with specific IgE antibodies to Bet v 1 and Pru p 3 allergens. It was confirmed that patients with pollen allergy are often characterized by Th2 response of the immune system, though other mechanisms of allergy development occurred in some cases. The data obtained demonstrate the necessity of detailed analysis of the individual mechanism of allergic reactions and patient-centered approach to the personalized allergy treatment.


Subject(s)
Antigens, Plant/immunology , Carrier Proteins/immunology , Immunoglobulin E/blood , Plant Proteins/immunology , Rhinitis, Allergic, Seasonal/blood , Adult , Antigens, Plant/chemistry , Carrier Proteins/chemistry , Case-Control Studies , Female , Gene Expression , Humans , Immunoglobulin E/genetics , Interferon-gamma/blood , Interferon-gamma/immunology , Interleukin-10/blood , Interleukin-10/immunology , Interleukin-13/blood , Interleukin-13/immunology , Interleukin-17/blood , Interleukin-17/immunology , Interleukin-4/blood , Interleukin-4/immunology , Interleukin-5/blood , Interleukin-5/immunology , Interleukin-9/blood , Interleukin-9/immunology , Lymphotoxin-alpha/blood , Lymphotoxin-alpha/immunology , Male , Middle Aged , Moscow , Plant Proteins/chemistry , Precision Medicine , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Rhinitis, Allergic, Seasonal/genetics , Rhinitis, Allergic, Seasonal/immunology , Rhinitis, Allergic, Seasonal/physiopathology , Th1 Cells/immunology , Th17 Cells/immunology , Th2 Cells/immunology , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/immunology
11.
J Biol Chem ; 294(13): 5214-5227, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30723161

ABSTRACT

Etanercept is a soluble form of the tumor necrosis factor receptor 2 (TNFR2) that inhibits pathological tumor necrosis factor (TNF) responses in rheumatoid arthritis and other inflammatory diseases. However, besides TNF, etanercept also blocks lymphotoxin-α (LTα), which has no clear therapeutic value and might aggravate some of the adverse effects associated with etanercept. Poxviruses encode soluble TNFR2 homologs, termed viral TNF decoy receptors (vTNFRs), that display unique specificity properties. For instance, cytokine response modifier D (CrmD) inhibits mouse and human TNF and mouse LTα, but it is inactive against human LTα. Here, we analyzed the molecular basis of these immunomodulatory activities in the ectromelia virus-encoded CrmD. We found that the overall molecular mechanism to bind TNF and LTα from mouse and human origin is fairly conserved in CrmD and dominated by a groove under its 50s loop. However, other ligand-specific binding determinants optimize CrmD for the inhibition of mouse ligands, especially mouse TNF. Moreover, we show that the inability of CrmD to inhibit human LTα is caused by a Glu-Phe-Glu motif in its 90s loop. Importantly, transfer of this motif to etanercept diminished its anti-LTα activity in >60-fold while weakening its TNF-inhibitory capacity in 3-fold. This new etanercept variant could potentially be used in the clinic as a safer alternative to conventional etanercept. This work is the most detailed study of the vTNFR-ligand interactions to date and illustrates that a better knowledge of vTNFRs can provide valuable information to improve current anti-TNF therapies.


Subject(s)
Ectromelia virus/immunology , Immunologic Factors/immunology , Lymphotoxin-alpha/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Tumor Necrosis Factor Decoy Receptors/immunology , Viral Proteins/immunology , Amino Acid Sequence , Animals , Ectromelia virus/chemistry , Ectromelia, Infectious/virology , Humans , Immunologic Factors/chemistry , Mice , Models, Molecular , Protein Domains , Tumor Necrosis Factor-alpha/immunology , Viral Proteins/chemistry
12.
Blood ; 133(13): 1489-1494, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30696620

ABSTRACT

Persistent NF-κB activation is a hallmark of the malignant Hodgkin/Reed-Sternberg (HRS) cells in classical Hodgkin lymphoma (cHL). Genomic lesions, Epstein-Barr virus infection, soluble factors, and tumor-microenvironment interactions contribute to this activation. Here, in an unbiased approach to identify the cHL cell-secreted key factors for NF-κB activation, we have dissected the secretome of cultured cHL cells by chromatography and subsequent mass spectrometry. We identified lymphotoxin-α (LTA) as the causative factor for autocrine and paracrine activation of canonical and noncanonical NF-κB in cHL cell lines. In addition to inducing NF-κB, LTA promotes JAK2/STAT6 signaling. LTA and its receptor TNFRSF14 are transcriptionally activated by noncanonical NF-κB, creating a continuous feedback loop. Furthermore, LTA shapes the expression of cytokines, receptors, immune checkpoint ligands and adhesion molecules, including CSF2, CD40, PD-L1/PD-L2, and VCAM1. Comparison with single-cell gene-activity profiles of human hematopoietic cells showed that LTA induces genes restricted to the lymphoid lineage, as well as those largely restricted to the myeloid lineage. Thus, LTA sustains autocrine NF-κB activation, impacts activation of several signaling pathways, and drives expression of genes essential for microenvironmental interactions and lineage ambiguity. These data provide a robust rationale for targeting LTA as a treatment strategy for cHL patients.


Subject(s)
Hodgkin Disease/immunology , Janus Kinase 2/immunology , Lymphotoxin-alpha/immunology , NF-kappa B/immunology , STAT6 Transcription Factor/immunology , Cell Line , Gene Expression Regulation, Neoplastic , Hodgkin Disease/genetics , Humans , Lymphotoxin-alpha/genetics , Reed-Sternberg Cells/immunology , Reed-Sternberg Cells/metabolism , Signal Transduction , Transcriptional Activation
13.
Eur J Immunol ; 49(3): 428-442, 2019 03.
Article in English | MEDLINE | ID: mdl-30575951

ABSTRACT

IL-4 is critical for differentiation of Th2 cells and antibody isotype switching, but our work demonstrated that it is produced in the peripheral LN under both Type 2, and Type 1 conditions, raising the possibility of other functions. We found that IL-4 is vital for proper positioning of hematopoietic and stromal cells in steady state, and the lack of IL-4 or IL-4Rα correlates with disarrangement of both follicular dendritic cells and CD31+ endothelial cells. We observed a marked disorganization of B cells in these mice, suggesting that the lymphocyte-stromal cell axis is maintained by the IL-4 signaling pathway. This study showed that absence of IL-4 correlates with significant downregulation of Lymphotoxin alpha (LTα) and Lymphotoxin beta (LTß), critical lymphokines for the development and maintenance of lymphoid organs. Moreover, immunization of IL-4 deficient mice with Type 2 antigens failed to induce lymphotoxin production, LN reorganization, or germinal center formation, while this process is IL-4 independent following Type 1 immunization. Additionally, we found that Type 1 antigen mediated LN reorganization is dependent on IFN-γ in the absence of IL-4. Our findings reveal a role of IL-4 in the maintenance of peripheral lymphoid organ microenvironments during homeostasis and antigenic challenge.


Subject(s)
Cell Proliferation , Interleukin-4/immunology , Receptors, Cell Surface/immunology , Stromal Cells/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Dendritic Cells, Follicular/immunology , Dendritic Cells, Follicular/metabolism , Endothelial Cells/immunology , Endothelial Cells/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymphotoxin-alpha/immunology , Lymphotoxin-alpha/metabolism , Lymphotoxin-beta/immunology , Lymphotoxin-beta/metabolism , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism
14.
Sci Rep ; 8(1): 17727, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30531962

ABSTRACT

Though lymphotoxin (LT) is highly expressed by type I helper T (Th1) cells, its contribution to CD4+ T cell differentiation during infections and diseases remains a mystery. In HSV-1 infection, we observed that LTßR signaling is required to limit the Th1 response. Using bone marrow chimeric mice, mixed-T-cell chimeric mice, and LTßR in vivo blockades, we unexpectedly observed that LT, especially T cell-derived LT, played an indispensable role in limiting the Th1 response. The LTßR-Ig blockade promoted the Th1 response by increasing infiltration of monocytes and monocyte-derived DCs and up-regulating IL-12 secretion in the lymphoid environment. Our findings identified a novel role for T cell-derived LT in manipulating Th1 differentiation.


Subject(s)
Herpes Simplex/immunology , Herpesvirus 1, Human/immunology , Lymphotoxin-alpha/immunology , Th1 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Interleukin-12/immunology , Lymphotoxin beta Receptor/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Monocytes/immunology , Signal Transduction/immunology , Up-Regulation/immunology
15.
J Autoimmun ; 95: 1-14, 2018 12.
Article in English | MEDLINE | ID: mdl-30446251

ABSTRACT

Susceptibility to multiple autoimmune diseases is associated with common gene polymorphisms influencing IL-2 signaling and Treg function, making Treg-specific expansion by IL-2 a compelling therapeutic approach to treatment. As an in vivo IL-2 half-life enhancer we used a non-targeted, effector-function-silent human IgG1 as a fusion protein. An IL-2 mutein (N88D) with reduced binding to the intermediate affinity IL-2Rßγ receptor was engineered with a stoichiometry of two IL-2N88D molecules per IgG, i.e. IgG-(IL-2N88D)2. The reduced affinity of IgG-(IL-2N88D)2 for the IL-2Rßγ receptor resulted in a Treg-selective molecule in human whole blood pSTAT5 assays. Treatment of cynomolgus monkeys with single low doses of IgG-(IL-2N88D)2 induced sustained preferential activation of Tregs accompanied by a corresponding 10-14-fold increase in CD4+ and CD8+ CD25+FOXP3+ Tregs; conditions that had no effect on CD4+ or CD8+ memory effector T cells. The expanded cynomolgus Tregs had demethylated FOXP3 and CTLA4 epigenetic signatures characteristic of functionally suppressive cells. Humanized mice had similar selective in vivo responses; IgG-(IL-2N88D)2 increased Tregs while wild-type IgG-IL-2 increased NK cells in addition to Tregs. The expanded human Tregs had demethylated FOXP3 and CTLA4 signatures and were immunosuppressive. These results describe a next-generation immunotherapy using a long-lived and Treg-selective IL-2 that activates and expands functional Tregsin vivo. Patients should benefit from restored immune homeostasis in a personalized fashion to the extent that their autoimmune disease condition dictates opening up the possibility for remissions and cures.


Subject(s)
Autoimmune Diseases/therapy , Immunoglobulin G/immunology , Immunotherapy/methods , Interleukin-2/immunology , Lymphotoxin-alpha/immunology , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Regulatory/drug effects , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Binding Sites , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Cell Proliferation , DNA Methylation/drug effects , Disease Models, Animal , Epigenesis, Genetic , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Immunoglobulin G/administration & dosage , Immunoglobulin G/chemistry , Immunoglobulin G/genetics , Interleukin-2/administration & dosage , Interleukin-2/chemistry , Interleukin-2/genetics , Interleukin-2 Receptor beta Subunit/genetics , Interleukin-2 Receptor beta Subunit/immunology , Lymphocyte Activation/drug effects , Lymphotoxin-alpha/administration & dosage , Lymphotoxin-alpha/chemistry , Lymphotoxin-alpha/genetics , Macaca fascicularis , Male , Mice , Mice, Transgenic , Models, Molecular , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/immunology , Protein Structure, Secondary , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
16.
Cell Host Microbe ; 24(2): 187-188, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30092193

ABSTRACT

The early response to bacterial infection requires cytokine responses by immune cells. In this issue of Cell Host & Microbe, Seo et al. (2018) demonstrate that TNF-TNFR superfamily molecules LIGHT and HVEM stimulate early IFN-γ production by type 3 innate lymphoid cells, which are critical for defense against Yersinia enterocolitica.


Subject(s)
Allogeneic Cells/immunology , Immunity, Innate , Lymphocytes/immunology , Lymphotoxin-alpha/immunology , Receptors, Tumor Necrosis Factor, Member 14/immunology , Animals , Humans , Interferon-gamma/immunology , Intestinal Diseases/immunology , Intestinal Diseases/microbiology , Yersinia Infections/immunology , Yersinia enterocolitica/immunology
17.
Inflamm Res ; 67(6): 495-501, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29541795

ABSTRACT

BACKGROUND: The role of tumor necrosis factor (TNF) in the inflammatory response in rheumatoid arthritis (RA) is well established, whereas less is known about the role of TNF's close homolog, lymphotoxin alpha (LTα). FINDINGS: Increased levels of LTα are found in the serum and synovial tissue of patients with RA, and in vitro studies found that LTα-induced proliferation of RA fibroblast-like synoviocytes was at a similar level to TNF. These findings support the idea that anti-LTα treatment could be beneficial in patients with RA, but pateclizumab, an anti-LTα antibody, was not as efficacious as the anti-TNF agent adalimumab in reducing symptoms of RA in a head-to-head study, suggesting that anti-LTα therapies might not represent a valid alternative treatment option in patients with RA. However, suppression of LTα activity might be relevant in the context of RA-related comorbidities, as patients with RA have an increased risk of myocardial infarction (MI) compared with the general population, and specific polymorphisms of the LTα gene have been linked to increased MI risk. CONCLUSIONS: In this review, we summarize the key characteristics of LTα and the most recent findings on the role of LTα in RA.


Subject(s)
Arthritis, Rheumatoid/immunology , Lymphotoxin-alpha/immunology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Etanercept/therapeutic use , Humans
18.
Expert Opin Biol Ther ; 18(4): 477-481, 2018 04.
Article in English | MEDLINE | ID: mdl-29558865

ABSTRACT

INTRODUCTION: Rheumatoid arthritis is a common inflammatory joint disease with a myriad of systemic manifestations. Over the last 20 years its treatment has been revolutionised by the introduction of a number of different biologic drugs, including the TNF-receptor Fc fusion protein, Etanercept. However, these drugs are expensive and their widespread use puts a financial burden on healthcare systems. As many biologic treatments begin to come off patent new 'biosimilar' versions are being developed which can lead to significant cost savings. GP2015 (Erelzi®) is the second biosimilar version of Etanercept which is licensed for the treatment of rheumatoid arthritis. Areas covered: We discuss the Chemistry, pharmacokinetics and pharmacodynamics of GP2015 in relation to reference Etanercept. Preclinical trials have shown pharmacokinetic equivalence between GP2015 and the reference drug. The recently completed Phase III, randomised, double blind EQUIRA study has shown equivalent efficacy and safety between GP2015 and Etanercept in patients with rheumatoid arthritis. Expert opinion: GP2015 has shown equivalent efficacy and safety to reference Etanercept. With a growing number of biosimilar medications becoming available and another biosimilar Etanercept already being widely prescribed it is likely to be the cost of the drug that will determine if it is used widely.


Subject(s)
Arthritis, Rheumatoid/drug therapy , Biosimilar Pharmaceuticals/therapeutic use , Dermatologic Agents/therapeutic use , Etanercept/therapeutic use , Antibodies, Anti-Idiotypic/blood , Arthritis, Rheumatoid/pathology , Biosimilar Pharmaceuticals/adverse effects , Biosimilar Pharmaceuticals/pharmacokinetics , Clinical Trials as Topic , Dermatologic Agents/adverse effects , Dermatologic Agents/immunology , Dermatologic Agents/pharmacokinetics , Etanercept/adverse effects , Etanercept/immunology , Etanercept/pharmacokinetics , Half-Life , Humans , Lymphotoxin-alpha/immunology , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Tumor Necrosis Factor-alpha/immunology
19.
Cytokine ; 101: 39-47, 2018 01.
Article in English | MEDLINE | ID: mdl-27623349

ABSTRACT

Lymphotoxin (LT) is a member of the tumor necrosis factor (TNF) superfamily of cytokines which serves multiple functions, including the control of lymphoid organ development and maintenance, as well as regulation of inflammation and autoimmunity. Although the role of LT in organogenesis and maintenance of lymphoid organs is well established, the contribution of LT pathway to homeostasis of lymphoid organs during the immune response to pathogens is less understood. In this review, we highlight recent advances on the role of LT pathway in antiviral immune responses. We discuss the role of LT signaling in lymphoid organ integrity, type I IFN production and regulation of protection and immunopathology during viral infections. We further discuss the potential of therapeutic targeting LT pathway for controlling immunopathology and antiviral protection.


Subject(s)
Antiviral Agents/immunology , Lymphoid Tissue/physiology , Lymphotoxin-alpha/immunology , Virus Diseases/immunology , Animals , Autoimmunity , Homeostasis/immunology , Humans , Inflammation , Interferon Type I/biosynthesis , Interferon Type I/immunology , Lymphoid Tissue/immunology , Lymphotoxin beta Receptor/immunology , Lymphotoxin-alpha/drug effects , Lymphotoxin-alpha/genetics , Mice , Signal Transduction/genetics , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/physiology , Virus Diseases/drug therapy , Virus Diseases/physiopathology
20.
Bull Exp Biol Med ; 164(2): 233-240, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29181671

ABSTRACT

We studied in vivo modifying effect of autotransfusion of human bone marrow mesenchymal stromal cells on ROS generation and production of cytokines (TNFα,TNFß, IL-1α, IL-10, IFNγ, and GM-CSF) and PGE2 by mononuclear cells of patients (N=21) with chronic heart failure. These parameters were evaluated prior to (control) and after (immediately and on day 14) intravenous administration of stromal cells in doses of 100-200×106. Immediately after autotransfusion, significant increase of in vitro zymosan-induced chemiluminescence of blood mononuclear cells from 10 patients was observed. At later terms after autotransfusion (day 14), inhibition of chemiluminescent activity of blood mononuclear cells was revealed in 50% patients. We discuss possible mechanisms of involvement of transplanted autologous bone marrow mesenchymal stromal cells in reprogramming of blood mononuclear phagocytes from the pro- to anti-inflammatory phenotype under conditions of their in vivo interaction manifesting in transition from activation to inhibition of ROS-producing activity of macrophages and significant suppression of in vitro LPS-induced production of TNFα and GM-CSF by blood mononuclears against the background of significantly elevated TNFß, IL-10, and IL-1α concentrations.


Subject(s)
Heart Failure/therapy , Leukocytes, Mononuclear/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Reactive Oxygen Species/immunology , Dinoprostone/immunology , Dinoprostone/metabolism , Gene Expression Regulation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Heart Failure/genetics , Heart Failure/immunology , Heart Failure/pathology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/pathology , Lipopolysaccharides/pharmacology , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/immunology , Mesenchymal Stem Cells/cytology , Primary Cell Culture , Reactive Oxygen Species/metabolism , Signal Transduction , Transplantation, Autologous , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
SELECTION OF CITATIONS
SEARCH DETAIL