Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 834
Filter
1.
Dev Comp Immunol ; 157: 105197, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38763479

ABSTRACT

Toll-like receptors (TLRs) are pivotal pattern recognition receptors (PRRs) and key mediators of innate immunity. Despite the significance of channel catfish (Ictalurus punctatus) in comparative immunology and aquaculture, its 20 TLR genes remain largely functionally uncharacterized. In this study, our aim was to determine the catfish TLR7 agonists, signaling potential, and cellular localization. Using a mammalian reporter system, we identified imiquimod and resiquimod, typical ssRNA analogs, as potent catfish TLR7 agonists. Notably, unlike grass carp TLR7, catfish TLR7 lacks the ability to respond to poly (I:C). Confocal microscopy revealed predominant catfish TLR7 expression in lysosomes, co-localizing with the endosomal chaperone protein, UNC93B1. Furthermore, imiquimod stimulation elicited robust IFNb transcription in peripheral blood leukocytes isolated from adult catfish. These findings underscore the conservation of TLR7 signaling in catfish, reminiscent of mammalian TLR7 responses. Our study sheds light on the functional aspects of catfish TLR7 and contributes to a better understanding of its role in immune defense mechanisms.


Subject(s)
Fish Proteins , Ictaluridae , Imidazoles , Imiquimod , Immunity, Innate , Lysosomes , Toll-Like Receptor 7 , Animals , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/genetics , Imidazoles/pharmacology , Ictaluridae/immunology , Lysosomes/metabolism , Fish Proteins/genetics , Fish Proteins/metabolism , Signal Transduction , Humans , Aminoquinolines/pharmacology , Poly I-C/immunology
2.
Microbes Infect ; 26(5-6): 105346, 2024.
Article in English | MEDLINE | ID: mdl-38670217

ABSTRACT

Vaccine adjuvants, such as liposome-based cationic adjuvant formulations (CAFs), are able to boost immune responses and, by incorporation of distinct immunomodulators, steer immunity towards a desired direction in mice, non-human primates and humans, while less studied in pigs. Here we used commercial pigs to investigate polarizing adjuvant effects of CAFs with immunomodulators: C-type lectin receptor ligands trehalose-6,6'-dibehenate and monomycolyl glycerol, toll-like receptor 3 ligand Poly(I:C) or retinoic acid. Vaccines were formulated with a recombinant Chlamydia model protein antigen and administered via three injection routes. All adjuvants significantly increased antigen-specific IgG in serum, compared to non-adjuvanted antigen. Administering the vaccines through intramuscular and intraperitoneal routes induced significantly higher antigen-specific IgG and IgA serum antibodies, than the perirectal route. Although immunizations triggered cell-mediated immunity, no significant differences between adjuvants or injection sites were detected. Genes depicting T cell subtypes revealed only minor differences. Our findings suggest that specific signatures of the tested adjuvant immunomodulation do not translate well from mice to pigs in standard two-dose immunizations. This study provides new insights into immune responses to CAFs in pigs, and highlights that adjuvant development should ideally be carried out in the intended species of interest or in models with high predictive validity/translational value.


Subject(s)
Adjuvants, Immunologic , Immunoglobulin G , Liposomes , Animals , Liposomes/immunology , Liposomes/administration & dosage , Swine , Adjuvants, Immunologic/administration & dosage , Immunoglobulin G/blood , Immunoglobulin A/blood , Immunoglobulin A/immunology , Antibodies, Bacterial/blood , Adjuvants, Vaccine/administration & dosage , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Poly I-C/administration & dosage , Poly I-C/immunology , Chlamydia/immunology , Tretinoin/administration & dosage , Tretinoin/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/administration & dosage , Immunomodulating Agents/administration & dosage , Immunomodulating Agents/pharmacology , Immunomodulating Agents/immunology , Immunity, Cellular , Glycolipids
3.
Dev Comp Immunol ; 157: 105182, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38636700

ABSTRACT

Galectin 8 belongs to the tandem repeat subclass of the galectin superfamily. It possesses two homologous carbohydrate recognition domains linked by a short peptide and preferentially binds to ß-galactoside-containing glycol-conjugates in a calcium-independent manner. This study identified Galectin-8-like isoform X1 (PhGal8X1) from red-lip mullet (Planiliza haematocheilus) and investigated its role in regulating fish immunity. The open reading frame of PhGal8X1 was 918bp, encoding a soluble protein of 305 amino acids. The protein had a theoretical isoelectric (pI) point of 7.7 and an estimated molecular weight of 34.078 kDa. PhGal8X1 was expressed in various tissues of the fish, with prominent levels in the brain, stomach, and intestine. PhGal8X1 expression was significantly (p < 0.05) induced in the blood and spleen upon challenge with different immune stimuli, including polyinosinic:polycytidylic acid, lipopolysaccharide, and Lactococcus garvieae. The recombinant PhGal8X1 protein demonstrated agglutination activity towards various bacterial pathogens at a minimum effective concentration of 50 µg/mL or 100 µg/mL. Subcellular localization observations revealed that PhGal8X1 was primarily localized in the cytoplasm. PhGal8X1 overexpression in fathead minnow cells significantly (p < 0.05) inhibited viral hemorrhagic septicemia virus (VHSV) replication. The expression levels of four proinflammatory cytokines and two chemokines were significantly (p < 0.05) upregulated in PhGal8X1 overexpressing cells in response to VHSV infection. Furthermore, overexpression of PhGal8X1 exhibited protective effects against oxidative stress induced by H2O2 through the upregulation of antioxidant enzymes. Taken together, these findings provide compelling evidence that PhGal8X1 plays a crucial role in enhancing innate immunity and promoting cell survival through effective regulation of antibacterial, antiviral, and antioxidant defense mechanisms in red-lip mullet.


Subject(s)
Antioxidants , Fish Proteins , Galectins , Smegmamorpha , Animals , Fish Proteins/genetics , Fish Proteins/metabolism , Fish Proteins/immunology , Smegmamorpha/immunology , Smegmamorpha/genetics , Galectins/metabolism , Galectins/genetics , Antioxidants/metabolism , Fish Diseases/immunology , Cytokines/metabolism , Immunity, Innate , Poly I-C/immunology , Lactococcus/physiology , Lipopolysaccharides/immunology , Chemokines/metabolism , Chemokines/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Novirhabdovirus/physiology , Novirhabdovirus/immunology , Antiviral Agents/metabolism
4.
Dev Comp Immunol ; 156: 105175, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38574831

ABSTRACT

Peroxiredoxin-1 (Prdx1) is a thiol-specific antioxidant enzyme that detoxifies reactive oxygen species (ROS) and regulates the redox status of cells. In this study, the Prdx1 cDNA sequence was isolated from the pre-established Amphiprion clarkii (A. clarkii) (AcPrdx1) transcriptome database and characterized structurally and functionally. The AcPrdx1 coding sequence comprises 597 bp and encodes 198 amino acids with a molecular weight of 22.1 kDa and a predicted theoretical isoelectric point of 6.3. AcPrdx1 is localized and functionally available in the cytoplasm and nucleus of cells. The TXN domain of AcPrdx1 comprises two peroxiredoxin signature VCP motifs, which contain catalytic peroxidatic (Cp-C52) and resolving cysteine (CR-C173) residues. The constructed phylogenetic tree and sequence alignment revealed that AcPrdx1 is evolutionarily conserved, and its most closely related counterpart is Amphiprion ocellaris. Under normal physiological conditions, AcPrdx1 was ubiquitously detected in all tissues examined, with the most robust expression in the spleen. Furthermore, AcPrdx1 transcripts were significantly upregulated in the spleen, head kidney, and blood after immune stimulation by polyinosinic:polycytidylic acid (poly (I:C)), lipopolysaccharide (LPS), and Vibrio harveyi injection. Recombinant AcPrdx1 (rAcPrdx1) demonstrated antioxidant and DNA protective properties in a concentration-dependent manner, as evidenced by insulin disulfide reduction, peroxidase activity, and metal-catalyzed oxidation (MCO) assays, whereas cells transfected with pcDNA3.1(+)/AcPrdx1 showed significant cytoprotective function under oxidative and nitrosative stress. Overexpression of AcPrdx1 in fathead minnow (FHM) cells led to a lower viral copy number following viral hemorrhagic septicemia virus (VHSV) infection, along with upregulation of several antiviral genes. Collectively, this study provides insights into the function of AcPrdx1 in defense against oxidative stressors and its role in the immune response against pathogenic infections in A. clarkii.


Subject(s)
Fish Proteins , Peroxiredoxins , Phylogeny , Vibrio Infections , Animals , Peroxiredoxins/metabolism , Peroxiredoxins/genetics , Peroxiredoxins/immunology , Fish Proteins/genetics , Fish Proteins/metabolism , Fish Proteins/immunology , Vibrio Infections/immunology , Poly I-C/immunology , Fish Diseases/immunology , Immunity, Innate , Vibrio/immunology , Vibrio/physiology , Cloning, Molecular , Amino Acid Sequence , Perciformes/immunology , Lipopolysaccharides/immunology , Sequence Alignment , Reactive Oxygen Species/metabolism
5.
Dev Comp Immunol ; 156: 105181, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38636698

ABSTRACT

Interferon regulatory factor 7 (IRF7) is considered the master regulator of virus-induced interferon (IFN) production. However, to avoid an autoimmune response, the expression of IRF7 must be tightly controlled. In this study, we report that zebrafish ubiquitin-specific protease 8 (USP8) promotes IRF7 degradation through an autophagy-lysosome-dependent pathway to inhibit IFN production. First, zebrafish usp8 is induced upon spring viremia of carp virus (SVCV) infection and polyinosinic/polycytidylic acid (poly I:C) stimulation. Second, overexpression of USP8 suppresses SVCV or poly I:C-mediated IFN expression. Mechanistically, USP8 interacts with IRF7 and promotes its degradation via an autophagy-lysosome-dependent pathway. Finally, USP8 significantly suppresses cellular antiviral responses and enhances SVCV proliferation. In summary, our discoveries offer a perspective on the role of zebrafish USP8 and provide additional understanding of the regulation of IRF7 in host antiviral immune response.


Subject(s)
Autophagy , Interferon Regulatory Factor-7 , Interferon Regulatory Factors , Lysosomes , Rhabdoviridae , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/immunology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Autophagy/immunology , Lysosomes/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/genetics , Rhabdoviridae/physiology , Rhabdoviridae/immunology , Interferons/metabolism , Poly I-C/immunology , Rhabdoviridae Infections/immunology , Proteolysis , Fish Diseases/immunology , Fish Diseases/virology , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Humans , Immunity, Innate
6.
Dev Comp Immunol ; 156: 105171, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38537729

ABSTRACT

Trace amine-associated receptors (TAARs) are a class of G protein-coupled receptors, playing an immunomodulatory function in the neuroinflammatory responses. In the present study, a TAAR homologue with a 7tm_classA_rhodopsin-like domain (designated as CgTAAR1L) was identified in oyster Crassostrea gigas. The abundant CgTAAR1L transcripts were detected in visceral ganglia and haemocytes compared to other tissues, which were 55.35-fold and 32.95-fold (p < 0.01) of those in adductor muscle, respectively. The mRNA expression level of CgTAAR1L in haemocytes significantly increased and reached the peak level at 3 h after LPS or Poly (I:C) stimulation, which was 4.55-fold and 12.35-fold of that in control group, respectively (p < 0.01). After the expression of CgTAAR1L was inhibited by the injection of its targeted siRNA, the mRNA expression levels of interleukin17s (CgIL17-1, CgIL17-5 and CgIL17-6), and defensin (Cgdefh1) significantly decreased at 3 h after LPS stimulation, which was 0.51-fold (p < 0.001), 0.39-fold (p < 0.01), 0.48-fold (p < 0.05) and 0.41-fold (p < 0.05) of that in the control group, respectively. The nuclear translocation of Cgp65 protein was suppressed in the CgTAAR1L-RNAi oysters. Furthermore, the number of Vibrio splendidus in the haemolymph of CgTAAR1L-RNAi oysters significantly increased (4.11-fold, p < 0.001) compared with that in the control group. In contrast, there was no significant difference in phagocytic rate of haemocytes to V. splendidus in the CgTAAR1L-RNAi oysters. These results indicated that CgTAAR1L played an important role in the immune defense against bacterial infection by inducing the expressions of interleukin and defensin.


Subject(s)
Crassostrea , Defensins , Hemocytes , Lipopolysaccharides , Receptors, G-Protein-Coupled , Vibrio , Animals , Crassostrea/immunology , Hemocytes/immunology , Hemocytes/metabolism , Vibrio/immunology , Vibrio/physiology , Lipopolysaccharides/immunology , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Defensins/genetics , Defensins/metabolism , Immunity, Innate , Interleukin-17/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , Poly I-C/immunology , RNA, Small Interfering/genetics , Vibrio Infections/immunology , Trace Amine-Associated Receptors
7.
J Biol Chem ; 299(9): 105114, 2023 09.
Article in English | MEDLINE | ID: mdl-37524131

ABSTRACT

Exosomes, extracellular vesicles (EVs) produced within cells, mediate both the disposal of intracellular waste and communication with distant cells, and they are involved in a variety of disease processes. Although disease modifications of exosome cargos have been well studied, it has been poorly investigated how disease processes, such as endoplasmic reticulum (ER) stress, affect EV production. We previously reported that adiponectin, an adipocyte-secreted salutary factor, increases systemic exosome levels through T-cadherin-mediated enhancement of exosome biogenesis. In the present study, we demonstrated that adiponectin/T-cadherin-dependent EV production was susceptible to ER stress and that low-dose tunicamycin significantly reduced EV production in the presence, but not in the absence, of adiponectin. Moreover, pharmacological or genetic activation of inositol-requiring enzyme 1α, a central regulator of ER stress, downregulated T-cadherin at the mRNA and protein levels as well as attenuated EV production. In addition, adiponectin/T-cadherin-independent EV production was attenuated under ER stress conditions. Repeated administration of tunicamycin to mice decreased circulating small EVs without decreasing tissue T-cadherin expression. Mechanistically, inositol-requiring enzyme 1α activation by silencing of the X-box binding protein 1 transcription factor upregulated the canonical interferon pathway and decreased EV production. The interferon pathway, when it was activated by polyinosinic-polycytidylic acid, also significantly attenuated EV production. Thus, we concluded that ER stress decreases exosome production through adiponectin/T-cadherin-dependent and -independent pathways.


Subject(s)
Adiponectin , Cadherins , Endoplasmic Reticulum Stress , Exosomes , Animals , Mice , Adiponectin/metabolism , Cadherins/biosynthesis , Cadherins/genetics , Cadherins/metabolism , Exosomes/drug effects , Exosomes/metabolism , Inositol/metabolism , Interferons/immunology , Poly I-C/immunology , Tunicamycin/pharmacology
8.
Proc Natl Acad Sci U S A ; 119(26): e2116738119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35749366

ABSTRACT

Tumor infiltration by T cells profoundly affects cancer progression and responses to immunotherapy. However, the tumor immunosuppressive microenvironment can impair the induction, trafficking, and local activity of antitumor T cells. Here, we investigated whether intratumoral injection of virus-derived peptide epitopes could activate preexisting antiviral T cell responses locally and promote antitumor responses or antigen spreading. We focused on a mouse model of cytomegalovirus (CMV), a highly prevalent human infection that induces vigorous and durable T cell responses. Mice persistently infected with murine CMV (MCMV) were challenged with lung (TC-1), colon (MC-38), or melanoma (B16-F10) tumor cells. Intratumoral injection of MCMV-derived T cell epitopes triggered in situ and systemic expansion of their cognate, MCMV-specific CD4+ or CD8+ T cells. The MCMV CD8+ T cell epitopes injected alone provoked arrest of tumor growth and some durable remissions. Intratumoral injection of MCMV CD4+ T cell epitopes with polyinosinic acid:polycytidylic acid (pI:C) preferentially elicited tumor antigen-specific CD8+ T cells, promoted tumor clearance, and conferred long-term protection against tumor rechallenge. Notably, secondary proliferation of MCMV-specific CD8+ T cells correlated with better tumor control. Importantly, intratumoral injection of MCMV-derived CD8+ T cell-peptide epitopes alone or CD4+ T cell-peptide epitopes with pI:C induced potent adaptive and innate immune activation of the tumor microenvironment. Thus, CMV-derived peptide epitopes, delivered intratumorally, act as cytotoxic and immunotherapeutic agents to promote immediate tumor control and long-term antitumor immunity that could be used as a stand-alone therapy. The tumor antigen-agnostic nature of this approach makes it applicable across a broad range of solid tumors regardless of their origin.


Subject(s)
CD8-Positive T-Lymphocytes , Cytomegalovirus Infections , Cytomegalovirus , Epitopes, T-Lymphocyte , Neoplasms , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/virology , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Epitopes, T-Lymphocyte/administration & dosage , Epitopes, T-Lymphocyte/immunology , Immunotherapy , Mice , Neoplasms/immunology , Neoplasms/therapy , Poly I-C/administration & dosage , Poly I-C/immunology , Tumor Microenvironment
9.
Proc Natl Acad Sci U S A ; 119(15): e2113310119, 2022 04 12.
Article in English | MEDLINE | ID: mdl-35377817

ABSTRACT

Gestational maternal immune activation (MIA) in mice induces persistent brain microglial activation and a range of neuropathologies in the adult offspring. Although long-term phenotypes are well documented, how MIA in utero leads to persistent brain inflammation is not well understood. Here, we found that offspring of mothers treated with polyriboinosinic­polyribocytidylic acid [poly(I:C)] to induce MIA at gestational day 13 exhibit blood­brain barrier (BBB) dysfunction throughout life. Live MRI in utero revealed fetal BBB hyperpermeability 2 d after MIA. Decreased pericyte­endothelium coupling in cerebral blood vessels and increased microglial activation were found in fetal and 1- and 6-mo-old offspring brains. The long-lasting disruptions result from abnormal prenatal BBB formation, driven by increased proliferation of cyclooxygenase-2 (COX2; Ptgs2)-expressing microglia in fetal brain parenchyma and perivascular spaces. Targeted deletion of the Ptgs2 gene in fetal myeloid cells or treatment with the inhibitor celecoxib 24 h after immune activation prevented microglial proliferation and disruption of BBB formation and function, showing that prenatal COX2 activation is a causal pathway of MIA effects. Thus, gestational MIA disrupts fetal BBB formation, inducing persistent BBB dysfunction, which promotes microglial overactivation and behavioral alterations across the offspring life span. Taken together, the data suggest that gestational MIA disruption of BBB formation could be an etiological contributor to neuropsychiatric disorders.


Subject(s)
Blood-Brain Barrier , Cyclooxygenase 2 , Encephalitis , Maternal-Fetal Exchange , Microglia , Prenatal Exposure Delayed Effects , Animals , Blood-Brain Barrier/abnormalities , Blood-Brain Barrier/physiopathology , Celecoxib/pharmacology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Encephalitis/immunology , Female , Gene Deletion , Maternal-Fetal Exchange/immunology , Mice , Microglia/enzymology , Poly I-C/immunology , Pregnancy , Prenatal Exposure Delayed Effects/immunology
10.
Viruses ; 14(2)2022 01 19.
Article in English | MEDLINE | ID: mdl-35215785

ABSTRACT

SARS-CoV-2, a member of the coronavirus family, is the causative agent of the COVID-19 pandemic. Currently, there is still an urgent need in developing an efficient therapeutic intervention. In this study, we aimed at evaluating the therapeutic effect of a single intranasal treatment of the TLR3/MDA5 synthetic agonist Poly(I:C) against a lethal dose of SARS-CoV-2 in K18-hACE2 transgenic mice. We demonstrate here that early Poly(I:C) treatment acts synergistically with SARS-CoV-2 to induce an intense, immediate and transient upregulation of innate immunity-related genes in lungs. This effect is accompanied by viral load reduction, lung and brain cytokine storms prevention and increased levels of macrophages and NK cells, resulting in 83% mice survival, concomitantly with long-term immunization. Thus, priming the lung innate immunity by Poly(I:C) or alike may provide an immediate, efficient and safe protective measure against SARS-CoV-2 infection.


Subject(s)
COVID-19/immunology , COVID-19/prevention & control , Immunity, Innate , Poly I-C/immunology , Poly I-C/therapeutic use , SARS-CoV-2/drug effects , Toll-Like Receptor 3/agonists , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/immunology , Animals , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/prevention & control , Disease Models, Animal , Female , Humans , Lung/immunology , Lung/virology , Mice , Mice, Transgenic , SARS-CoV-2/immunology , Toll-Like Receptor 3/immunology , Viral Load/drug effects , COVID-19 Drug Treatment
11.
Front Immunol ; 13: 807454, 2022.
Article in English | MEDLINE | ID: mdl-35145520

ABSTRACT

Background: Innate immunity, armed with pattern recognition receptors including Toll-like receptors (TLR), is critical for immune cell activation and the connection to anti-microbial adaptive immunity. However, information regarding the impact of age on the innate immunity in response to SARS-CoV2 adenovirus vector vaccines and its association with specific immune responses remains scarce. Methods: Fifteen subjects between 25-35 years (the young group) and five subjects between 60-70 years (the older adult group) were enrolled before ChAdOx1 nCoV-19 (AZD1222) vaccination. We determined activation markers and cytokine production of monocyte, natural killer (NK) cells and B cells ex vivo stimulated with TLR agonist (poly (I:C) for TLR3; LPS for TLR4; imiquimod for TLR7; CpG for TLR9) before vaccination and 3-5 days after each jab with flow cytometry. Anti-SARS-CoV2 neutralization antibody titers (surrogate virus neutralization tests, sVNTs) were measured using serum collected 2 months after the first jab and one month after full vaccination. Results: The older adult vaccinees had weaker vaccine-induced sVNTs than young vaccinees after 1st jab (47.2±19.3% vs. 21.2±22.2%, p value<0.05), but this difference became insignificant after the 2nd jab. Imiquimod, LPS and CpG strongly induced CD86 expression in IgD+CD27- naïve and IgD-CD27+ memory B cells in the young group. In contrast, only the IgD+ CD27- naïve B cells responded to these TLR agonists in the older adult group. Imiquimode strongly induced the CD86 expression in CD14+ monocytes in the young group but not in the older adult group. After vaccination, the young group had significantly higher IFN-γ expression in CD3- CD56dim NK cells after the 1st jab, whilst the older adult group had significantly higher IFN-γ and granzyme B expression in CD56bright NK cells after the 2nd jab (all p value <0.05). The IFN-γ expression in CD56dim and CD56bright NK cells after the first vaccination and CD86 expression in CD14+ monocyte and IgD-CD27-double-negative B cells after LPS and imiquimod stimulation correlated with vaccine-induced antibody responses. Conclusions: The innate immune responses after the first vaccination correlated with the neutralizing antibody production. Older people may have defective innate immune responses by TLR stimulation and weak or delayed innate immune activation profile after vaccination compared with young people.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , B-Lymphocytes/immunology , ChAdOx1 nCoV-19/immunology , Killer Cells, Natural/immunology , SARS-CoV-2/immunology , Adult , Aged , COVID-19/prevention & control , Female , Humans , Imiquimod/pharmacology , Immunity, Innate/immunology , Immunosenescence/immunology , Interferon-gamma/blood , Male , Middle Aged , Poly I-C/administration & dosage , Poly I-C/immunology , Toll-Like Receptors/immunology , Vaccination
12.
Dev Comp Immunol ; 129: 104351, 2022 04.
Article in English | MEDLINE | ID: mdl-35033573

ABSTRACT

Subcellular localization analysis implicated that CiPRMT6 was mainly located in the nucleus, with a small part of them located in the cytoplasm. PRMT6, namely protein arginine methyltransferase 6, was first identified and demonstrated to catalyze the methylation of arginine residue on the chromatin histones in mammals. Mammalian PRMT6 usually acts as an arginine methyltransferase in the nucleus, but induces antiviral innate immune response in the cytoplasm. Nowadays, there have been few reports about PRMT6 in teleost. In this study, we investigated the potential molecular mechanisms underlying the interaction of PRMT6 expression and IFN1 response in grass carp. We first cloned and identified a grass carp PRMT6 (named CiPRMT6, MN781672.1), which is 1068bp in length encoding a deduced polypeptide of 355 amino acids. In CIK cell, CiPRMT6 expression was up-regulated upon stimulation with poly (I:C); while overexpression of PRMT6 suppressed the promoter activity of grass carp IFN1 and reduced the phosphorylation of IRF3; however, the amount of PRMT6 mutant (lack of methyltransferase domain) was increased in the cytoplasm. Our results also showed that grass carp PRMT6 and IRF3 (but not TBK1) were co-located and bound to each other in the cytoplasm. The binding of CiPRMT6 to IRF3 impairs the interaction between TBK1 and IRF3, indicating that CiPRMT6 is a negative regulator for IFN1 expression through TBK1-IRF3 signaling pathway in grass carp. In conclusion, we identified that CiPRMT6 negatively regulated IFN1 expression by inhibiting the TBK1-IRF3 interaction as well as IRF3 phosphorylation.


Subject(s)
Carps/metabolism , Animals , Fish Proteins/genetics , Immunity, Innate , Interferon Regulatory Factor-3 , Interferon Type I/metabolism , Intracellular Signaling Peptides and Proteins , Phosphorylation , Poly I-C/immunology , Protein Serine-Threonine Kinases , Protein-Arginine N-Methyltransferases , Signal Transduction , Transcriptional Activation , Up-Regulation
13.
J Neuroimmunol ; 364: 577813, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35093761

ABSTRACT

Maternal immune activation (MIA) with poly(I:C) is a preclinical paradigm for schizophrenia and autism research. Methodological variations, including poly(I:C) molecular weight, contribute to inconsistencies in behavioural and molecular outcomes. We established in Wistar rats that 4 mg/kg high molecular weight (HMW)-poly(I:C) on GD19 induces maternal sickness, smaller litters and maternal elevations of serum cytokines, including increases in monocyte chemoattractants. In adult offspring, we found that males have higher serum cytokines than females, and MIA did not alter peripheral cytokines in either sex. Our study will contribute to the effective use of the MIA model to elucidate the neurobiology of neurodevelopmental disorders.


Subject(s)
Monocyte Chemoattractant Proteins/immunology , Neurodevelopmental Disorders/immunology , Poly I-C/toxicity , Pregnancy Complications, Infectious/immunology , Prenatal Exposure Delayed Effects/immunology , Animals , Cytokines/blood , Cytokines/immunology , Disease Models, Animal , Female , Male , Poly I-C/immunology , Pregnancy , Rats , Rats, Wistar
14.
J Immunol Methods ; 500: 113172, 2022 01.
Article in English | MEDLINE | ID: mdl-34673003

ABSTRACT

A promising strategy for controlling animal brucellosis is vaccination with commercial vaccine strains (Brucella melitensis Rev.1 and Brucella abortus RB51). Owing to safety concerns associated with these vaccines, developing a more effective and safe vaccine is essential. In this study, we examined the capacity of BhuA, 7α-HSDH or FliC antigens in the presence or absence of adjuvant in eliciting immune responses against brucellosis. After cloning, expression and purification, these proteins were used to examine immunologic responses. All immunized mice induced a vigorous IgG, with a predominant IgG2a response. Moreover, splenocytes of immunized mice proliferated and produced IL-2 and IFN-γ, suggesting the induction of cellular immunity. The high IgG2a/IgG1 ratio and IL-2 and IFN-γ indicated a Th1-oriented immune response in test groups. BhuA-, 7α-HSDH- or FliC- poly I:C formulations were the most effective at inducing Th1 immune response compared to groups immunized with naked proteins. Immunization with proteins protected mice against B. melitensis 16M and B. abortus 544. The proteins in adjuvant induced higher levels of protection than proteins only and exhibited similar degree of protection to live attenuated vaccines. Our results, for first time, introduced five potential candidates for subunit vaccine development against B. melitensis and B. abortus infection.


Subject(s)
Bacterial Proteins/immunology , Brucella Vaccine/immunology , Brucella abortus/physiology , Brucella melitensis/physiology , Brucellosis, Bovine/immunology , Flagellin/immunology , Hydroxysteroid Dehydrogenases/immunology , Membrane Transport Proteins/immunology , Th1 Cells/immunology , Adjuvants, Immunologic , Animals , Antibodies, Bacterial/blood , Cattle , Disease Models, Animal , Female , Immunity, Humoral , Immunoglobulin G/blood , Interferon-gamma/metabolism , Mice , Poly I-C/immunology , Vaccines, Subunit
15.
Scand J Immunol ; 95(3): e13128, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34923667

ABSTRACT

Targeting antigens to dendritic cells represent a promising method for enhancing immune responses against specific antigens. However, many studies have focused on systemic delivery (intravenous or intraperitoneally) of targeted antigen, approaches that are not easily transferable to humans. Here we evaluate the efficacy of an influenza vaccine targeting Xcr1+ cDC1 administered by intranasal immunization. Intranasal delivery of antigen fused to the chemokine Xcl1, the ligand of Xcr1, resulted in specific uptake by lung CD103+ cDC1. Interestingly, intranasal immunization with influenza A/PR/8/34 haemagglutinin (HA) fused to Xcl1, formulated with poly(I:C), resulted in enhanced induction of antigen-specific IFNγ+ CD4+ and IFNγ+ CD8+ T cell responses in lung compared non-targeted anti-NIP-HA (αNIP-HA). Induction of antibody responses was, however, similar in Xcl1-HA and αNIP-HA immunized mice, but significantly higher than in mice immunized with monomeric HA. Both Xcl1-HA and αNIP-HA vaccines induced full protection when mice were challenged with a lethal dose of influenza PR8 virus, reflecting the strong induction of HA-specific antibodies. Our results demonstrate that i.n. delivery of Xcl1-HA is a promising vaccine strategy for enhancing T cell responses in addition to inducing strong antibody responses.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chemokines, C/metabolism , Influenza Vaccines/immunology , Influenza, Human/immunology , Orthomyxoviridae Infections/immunology , Poly I-C/immunology , Animals , Antibodies, Viral/immunology , Antibody Formation/immunology , Antigens/immunology , Antigens, CD/immunology , Cell Line , Dendritic Cells/immunology , Dogs , Female , HEK293 Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Integrin alpha Chains/immunology , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C
16.
Front Immunol ; 12: 767666, 2021.
Article in English | MEDLINE | ID: mdl-34899719

ABSTRACT

Bronchial epithelial cells are front sentinels eliciting innate and adaptive immunity to respiratory viral pathogens. Recognition of viral double-stranded RNA induces antiviral interferon (IFN) responses in bronchial epithelial cells. Co-inhibitory molecules programmed cell death 1 ligand 1 (PD-L1) and ligand 2 (PD-L2) were also induced on bronchial epithelial cells, which bind programmed cell death 1 on T cell and inhibit the function of virus-specific cytotoxic T lymphocyte. A previous study showed that antiviral type I IFN increased PD-L1 and PD-L2 expression in cultured melanoma cells. However, it remains unknown whether antiviral IFNs affect PD-L1 and PD-L2 expression in bronchial epithelial cells. In addition, we previously reported that inhibition of PI3Kδ signaling enhanced antiviral IFN responses in human primary bronchial epithelial cells (PBECs). Here we assessed the effect of exogenous IFNs or a selective PI3Kδ inhibitor IC87114 on PD-L1 and PD-L2 in PBECs stimulated with a synthetic double-stranded RNA poly I:C or human metapneumovirus. Treatment with IFNß or IFNλ increased PD-L1 and PD-L2, and IFNß or IFNλ treatment plus poly I:C further increased both expressions. Treatment with IC87114 or transfection with siRNA targeting PI3K p110δ enhanced poly I:C-induced gene and protein expression of PD-L2, whereas IC87114 suppressed poly I:C-induced PD-L1. IC87114 enhanced poly I:C-induced gene expression of IFNß, IFNλ, and IFN-regulated genes via increased TBK1 and IRF3 phosphorylation. Transfection with siIRF3 counteracted the enhancement of poly I:C-induced PD-L2 by IC87114, whereas IC87114 suppressed poly I:C-induced PD-L1 regardless of transfection with siNC or siIRF3. Similar effects of IC87114 on PD-L1 and PD-L2 expression were observed in human metapneumovirus-infected PBECs. We showed for the first time that type I and type III IFNs induced the expression of PD-L1 and PD-L2 in PBECs. Our findings suggest that during viral infections, inhibition of PI3Kδ differentially regulates PD-L1 and PD-L2 expression in bronchial epithelial cells.


Subject(s)
Adenine/analogs & derivatives , B7-H1 Antigen/immunology , Epithelial Cells/immunology , Metapneumovirus/immunology , Poly I-C/immunology , Programmed Cell Death 1 Ligand 2 Protein/immunology , Quinazolines/pharmacology , Adenine/pharmacology , Asthma/genetics , Asthma/immunology , Asthma/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Bronchi/cytology , Cells, Cultured , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferons/pharmacology , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphorylation/drug effects , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism
17.
PLoS Pathog ; 17(10): e1009928, 2021 10.
Article in English | MEDLINE | ID: mdl-34695164

ABSTRACT

Non-specific protective effects of certain vaccines have been reported, and long-term boosting of innate immunity, termed trained immunity, has been proposed as one of the mechanisms mediating these effects. Several epidemiological studies suggested cross-protection between influenza vaccination and COVID-19. In a large academic Dutch hospital, we found that SARS-CoV-2 infection was less common among employees who had received a previous influenza vaccination: relative risk reductions of 37% and 49% were observed following influenza vaccination during the first and second COVID-19 waves, respectively. The quadrivalent inactivated influenza vaccine induced a trained immunity program that boosted innate immune responses against various viral stimuli and fine-tuned the anti-SARS-CoV-2 response, which may result in better protection against COVID-19. Influenza vaccination led to transcriptional reprogramming of monocytes and reduced systemic inflammation. These epidemiological and immunological data argue for potential benefits of influenza vaccination against COVID-19, and future randomized trials are warranted to test this possibility.


Subject(s)
COVID-19/immunology , Cross Protection/physiology , Immunity, Innate/physiology , Influenza Vaccines/administration & dosage , COVID-19/epidemiology , COVID-19/prevention & control , Cytokines/immunology , Cytokines/metabolism , Down-Regulation , Imidazoles/immunology , Incidence , Influenza Vaccines/immunology , Netherlands/epidemiology , Personnel, Hospital , Poly I-C/immunology , Proteomics , Risk Factors , Sequence Analysis, RNA
18.
Immunohorizons ; 5(10): 818-829, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667099

ABSTRACT

In this study, we report that the TLR4 ligand, LPS, and TLR3 ligand polyinosinic:polycytidylic acid failed to activate IRF3 or STAT1 in bone marrow-derived macrophages (BMMs) isolated from two independently generated lines of Rosa26-integrated Cas9-expressing C57BL/6J (B6) mice. RNA-sequencing analysis reveals that hundreds to thousands of genes including IFN-stimulated genes were differentially expressed in BMMs from these Cas9 strains compared with B6 upon LPS stimulation. Furthermore, the NF-κB signaling axis and TRIF-mediated necroptosis were also strongly reduced in response to LPS and polyinosinic:polycytidylic acid. In contrast, there were no defects in the responses of BMMs to ligands of the RIG-I, STING, TLR2, TLR9, and IFN receptors. Defects in TLR3 and TLR4 signaling were observed in mice with the B6 but not 129 background, and when Cas9 was integrated at the Rosa26 but not H11 locus. However, integration at the Rosa26 site, CAG promoter-driven Cas9 or eGFP were not individually sufficient to cause the defect. Taken together, the results of this study suggest a putative TRIF-mediated defect in TLR-3/4 signaling in BMMs from commercially available and widely used B6-Cas9-expressing mice.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Macrophages/immunology , Animals , CRISPR-Associated Protein 9/genetics , Cells, Cultured , Female , Lipopolysaccharides/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Necroptosis/immunology , Poly I-C/immunology , Primary Cell Culture , RNA, Untranslated/genetics , Signal Transduction/genetics , Signal Transduction/immunology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism
19.
Immunol Lett ; 239: 32-41, 2021 11.
Article in English | MEDLINE | ID: mdl-34418488

ABSTRACT

Tumor-derived exosomes (TEXs) could be harnessed as an immunotherapeutic cancer vaccine. These nanovesicles are inherently possesses rich tumor antigen reservoirs. Due to their undesirable features such as poor or limited immunogenicity as well as facilitation of cancer development via mediating communication between tumor cells TEXs could be transformed into an effective immune adjuvant delivery system that initiates a strong humoral and cell-mediated tumor-specific immune response. Engineering TEXs to harbor immunostimulatory molecules still remains a challenge. Previously, we demonstrated that nucleic acid ligand encapsulated liposomes could trigger synergistic strong humoral, and cell mediated immune responses and provokes tumor regression to that of their standalone counterparts. In this study, we evaluated to immunogenicity of 4T1/Her2 cell-derived exosomes upon loading them with two potent immuno adjuvant, a TLR9 ligand, K-type CpG ODN and a TLR3 ligand, p(I:C). Engineered TEXs co-encapsulating both ligands displayed boosted immunostimulatory properties by activating antigen-specific primary and memory T cell responses. Furthermore, our exosome-based vaccine candidate elicited robust Th1-biased immunity as evidenced by elevated secretion of IgG2a and IFNγ. In a therapeutic cancer model, administration of4T1 tumor derived exosomes loaded with CpG ODN and p(I:C) to animals regress tumor growth in 4T1 tumor-bearing mice. Taken together this work implicated that an exosome-based therapeutic vaccine promoted strong cellular and humoral anti-tumor immunity that is sufficient to reverse established tumors. This approach offers a personalized tumor therapy strategy that could be implemented in the clinic.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antigens, Neoplasm/administration & dosage , Breast Neoplasms/therapy , Cancer Vaccines/administration & dosage , Exosomes/immunology , Animals , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cancer Vaccines/immunology , Cell Line, Tumor/transplantation , Disease Models, Animal , Female , Humans , Memory T Cells/immunology , Mice , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Poly I-C/administration & dosage , Poly I-C/immunology , Th1 Cells/immunology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 9/metabolism
20.
Int Immunopharmacol ; 100: 108070, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34454292

ABSTRACT

RESEARCH QUESTION: In previous studies, we demonstrated that the human decidua and decidual stromal cells express high levels of CCL2 (chemokine (C-C motif) ligand 2, also known as monocyte chemotactic protein-1) and its receptor CCR2 (chemokine receptor 2). DSC-derived CCL2 interacts with CCR2 on DICs, causing the production and secretion of Th2-type cytokines, which promotes a Th2 bias at the maternal-foetal interface. Many pathogens may be present in the genital tract during pregnancy, but whether they affect immune regulation, especially Th2 regulation remains unknown. Toll-like receptors (TLRs) are a family of pattern-recognition receptors that recognise specific components of microbes and certain host molecules and play an important role in the host innate immune response. We examined TLR expression and evaluated whether TLRs could affect CCL2 secretion and subsequently induce Th1/Th2 responses. DESIGN: We used quantitative real-time PCR to measure TLR expression in the decidua and DSCs (decidual stromal cells). DSCs were cultured in the presence or absence of the TLR2 agonists PAM3CSK4, PGN-Sa, and zymosan, the TLR3 agonist poly (I:C) and the TLR4 agonist LPS. Then, the supernatants were assayed for CCL2 secreted by DSCs and IL-4, IFN-γ, IL-10, and TNF-α produced by DICs. RESULTS: Costimulation with TLR2, TLR3 and TLR4 agonists resulted in enhancing CCL2 production compared with that in the controls. Additionally, these TLR2, 3, and 4 agonists stimulated CD80/CD86 on DSCs and regulated IL-4 and IL-10 secretion on DICs. TLR2 and TLR3 agonists may promote Th1/Th2 immune bias. CONCLUSIONS: TLRs may induce Th1/Th2 responses by affecting the secretion of CCL2 at the maternal-foetal interface.


Subject(s)
Chemokine CCL2/metabolism , Decidua/metabolism , Maternal-Fetal Exchange/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Adult , Cells, Cultured , Decidua/cytology , Decidua/immunology , Female , Humans , Killer Cells, Natural/immunology , Lymphocyte Activation , Poly I-C/immunology , Pregnancy , Primary Cell Culture , Stromal Cells/immunology , Stromal Cells/metabolism , Th1 Cells/metabolism , Th2 Cells/metabolism , Toll-Like Receptors/agonists , Toll-Like Receptors/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...