Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.266
Filter
1.
Dev Comp Immunol ; 161: 105261, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39241936

ABSTRACT

Low molecular weight proteins, known as chemokines, facilitate the migration and localization of immune cells to the site of infection and injury. One of the first chemokines identified, CXCL8 functions as a key neutrophil activator, recruiting neutrophils to sites of inflammation. Several viral infections, including zoonotic coronaviruses and poxviruses, have been reported to induce the expression of CXCL8. Dromedary camels are known to harbor several potentially zoonotic pathogens, but critical immune molecules such as chemokines remain unidentified. We report here the identification of CXCL8 from the dromedary camel - the first chemokine identified from camelids. The complete dromedary CXCL8 cDNA sequence as well as the corresponding gene sequence from dromedary and two New World camelids - alpaca and llama were cloned. CXCL8 mRNA expression was relatively higher in PBMC, spleen, lung, intestine, and liver. Poly(I:C) and lipopolysaccharide stimulated CXCL8 expression in vitro, while interferon treatment inhibited it. In vitro infection with potentially zoonotic camelpox virus induced the expression of CXCL8 in camel kidney cells. Toxicological studies on camelids have been limited, and no biomarkers have been identified. Hence, we also evaluated CXCL8 mRNA expression as a potential biomarker to assess heavy metal toxicity in camel kidney cells in vitro. CXCL8 expression was increased after in vitro exposure to heavy metal compounds of cobalt and cadmium, suggesting potential utility as a biomarker for renal toxicity in camels. The results of our study demonstrate that camel CXCL8 plays a significant role in immunomodulatory and induced toxicity responses in dromedary camels.


Subject(s)
Camelus , Interleukin-8 , Animals , Interleukin-8/metabolism , Interleukin-8/genetics , Camelus/immunology , Poly I-C/immunology , Metals, Heavy/toxicity , Camelids, New World/immunology , Poxviridae Infections/immunology , Poxviridae Infections/veterinary , Cloning, Molecular , Poxviridae/immunology , Poxviridae/genetics , Lipopolysaccharides/immunology , Cells, Cultured
2.
Exp Clin Transplant ; 22(7): 551-558, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39223813

ABSTRACT

OBJECTIVES: Mesenchymal stem cells are gaining attention in medicine because of their anti-inflammatory and immunosuppressive properties. Inflammatory conditions can modulate immune responses in mesenchymal stem cells.We investigated the expression of long noncoding RNAs (RMRP, MALT1, NKILA,THRIL, and Linc-MAF-4) in humanWharton jelly mesenchymal stem cells primed with polyinosinicpolycytidylic acid. MATERIALS AND METHODS: Mesenchymal stem cells were isolated from human Wharton jelly by the explant method. To determine the stem nature of the cells, we performed a differentiation test on bone and fat cells. We used flow cytometry analysis to determine surface markers. Umbilical cord mesenchymal stem cells (1 × 105) were cultured in T75 culture flasks in Dulbecco's modified Eagle medium containing 10% fetal bovine serum. After cells reached approximately 80% confluency, cells were exposed to 50 µg/mL of polyinosinic-polycytidylic acid, a Toll-like receptor 3 ligand, for 24, 48, and 72 hours. The control group were cells not exposed to polyinosinic-polycytidylic acid. Real-time polymerase chain reaction evaluated RMRP, MALAT1, NKILA, THRIL, and Linc-MAF-4 long noncoding RNAs. RESULTS: We observed significantly increased expression of NKILA inWharton jelly mesenchymal stem cells stimulated with polyinosinic-polycytidylic acid at 72 hours compared with expression level in the control group (P < .001). CONCLUSIONS: Results indicated that a potential mechanism by which the Toll-like receptor 3 ligand improves immunosuppression of mesenchymal stem cells can be attributed to the regulatory role of long noncoding RNAs, possibly through increased expression of anti-inflammatory long noncoding RNAs such as NKILA.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cells , Poly I-C , RNA, Long Noncoding , Toll-Like Receptor 3 , Wharton Jelly , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/drug effects , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/genetics , Wharton Jelly/cytology , Cells, Cultured , Poly I-C/pharmacology , Cell Differentiation/drug effects , Time Factors , Gene Expression Regulation , Osteogenesis/drug effects
3.
Mol Biol Rep ; 51(1): 974, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39259342

ABSTRACT

BACKGROUND: One of the causes of tubulointerstitial nephritis is viral infection, with innate immune responses affecting its pathogenesis. Toll-like receptor 3 (TLR3) recognizes viral infections and acts antivirally by activating signaling to produce inflammatory cytokines/chemokines, including C-C motif chemokine ligand 5 (CCL5) and interferon-ß (IFN-ß). Although cylindromatosis lysine 63 deubiquitinase (CYLD) is known to be associated with tubulointerstitial nephritis and renal function, its role in the antiviral innate immune response in tubular epithelial cells remains unknown. In this study, we investigated the association between CYLD and TLR3-mediated CCL5 production in cultured human renal proximal tubular epithelial cells (hRPTECs). METHODS AND RESULTS: Polyinosinic-polycytidylic acid (poly IC), a synthetic TLR3 ligand, was used to stimulate hRPTECs. mRNA expression was measured using reverse transcription-quantitative polymerase chain reaction. Protein expression was assayed using western blotting or an enzyme-linked immunosorbent assay. Knockdown of IFN-ß, nuclear factor-kappa B (NF-κB) p65, and CYLD was performed by transfecting cells with specific small interfering RNAs. The intracellular localization of CYLD in hRPTECs was analyzed using immunofluorescence. Poly IC induced CCL5 expression in a time- and concentration-dependent manner, and knockdown of either IFN-ß or p65 reduced poly IC-induced CCL5 expression. CYLD knockdown increased the poly IC-induced CCL5, phosphorylated IκB kinase α/ß (IKK complex), and phosphorylated p65 expression. The CYLD protein was localized in the cytoplasm, and poly IC did not alter its expression. CONCLUSION: CYLD may prevent excessive inflammation due to an antiviral innate immune response by suppressing IKK complex and NF-κB activation downstream of TLR3 in hRPTECs.


Subject(s)
Chemokine CCL5 , Deubiquitinating Enzyme CYLD , Epithelial Cells , Kidney Tubules, Proximal , Poly I-C , Toll-Like Receptor 3 , Humans , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 3/genetics , Deubiquitinating Enzyme CYLD/metabolism , Deubiquitinating Enzyme CYLD/genetics , Chemokine CCL5/metabolism , Chemokine CCL5/genetics , Kidney Tubules, Proximal/metabolism , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Poly I-C/pharmacology , Interferon-beta/metabolism , Interferon-beta/genetics , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Immunity, Innate , NF-kappa B/metabolism , Cell Line
4.
Viruses ; 16(9)2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39339860

ABSTRACT

Allergic asthma is characterized by increased type 2 inflammation, including eosinophils. Subjects with allergic asthma have recurrent symptoms due to their constant exposure to environmental allergens, such as house dust mite (HDM), which can be further exacerbated by respiratory infections like rhinovirus. The immunoproteasome (IP) is a proteolytic machinery that is induced by inflammatory mediators during virus infection, but the role of the IP in airway allergic inflammation during rhinovirus infection remains unknown. Wild-type (WT) and IP knockout (KO) mice were challenged with HDM. At 48 h after the last HDM challenge, mice were infected with rhinovirus 1B (RV-A1B) for 24 h. After HDM and RV-A1B treatment, IP KO (vs. WT) mice had significantly more lung eosinophils and neutrophils, as well as a significantly higher viral load, but less IFN-beta expression, compared to WT mice. A TLR3 agonist polyinosinic-polycytidylic acid (Poly I:C) treatment after RV-A1B infection in HDM-challenged IP KO mice significantly increased IFN-beta expression and reduced viral load, with a minimal effect on the number of inflammatory cells. Our data suggest that immunoproteasome is an important mechanism functioning to prevent excessive inflammation and viral infection in allergen-exposed mice, and that Poly I:C could be therapeutically effective in enhancing the antiviral response and lessening the viral burden in lungs with IP deficiency.


Subject(s)
Mice, Knockout , Poly I-C , Proteasome Endopeptidase Complex , Rhinovirus , Toll-Like Receptor 3 , Animals , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/genetics , Mice , Proteasome Endopeptidase Complex/metabolism , Poly I-C/pharmacology , Picornaviridae Infections/immunology , Viral Load , Pyroglyphidae/immunology , Lung/pathology , Lung/immunology , Lung/virology , Asthma/immunology , Asthma/drug therapy , Eosinophils/immunology , Inflammation/immunology , Hypersensitivity/immunology , Hypersensitivity/drug therapy , Mice, Inbred C57BL , Disease Models, Animal , Interferon-beta/genetics , Interferon-beta/immunology
5.
Viruses ; 16(9)2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39339867

ABSTRACT

The establishment of effective antiviral responses within host cells is intricately related to their metabolic status, shedding light on immunometabolism. In this study, we investigated the hypothesis that cellular reliance on glutamine metabolism contributes to the development of a potent antiviral response. We evaluated the antiviral response in the presence or absence of L-glutamine in the culture medium, revealing a bivalent response hinging on cellular metabolism. While certain interferon-stimulated genes (ISGs) exhibited higher expression in an oxidative phosphorylation (OXPHOS)-dependent manner, others were surprisingly upregulated in a glycolytic-dependent manner. This metabolic dichotomy was influenced in part by variations in interferon-ß (IFN-ß) expression. We initially demonstrated that the presence of L-glutamine induced an enhancement of OXPHOS in A549 cells. Furthermore, in cells either stimulated by poly:IC or infected with dengue virus and Zika virus, a marked increase in ISGs expression was observed in a dose-dependent manner with L-glutamine supplementation. Interestingly, our findings unveiled a metabolic dependency in the expression of specific ISGs. In particular, genes such as ISG54, ISG12 and ISG15 exhibited heightened expression in cells cultured with L-glutamine, corresponding to higher OXPHOS rates and IFN-ß signaling. Conversely, the expression of viperin and 2'-5'-oligoadenylate synthetase 1 was inversely related to L-glutamine concentration, suggesting a glycolysis-dependent regulation, confirmed by inhibition experiments. This study highlights the intricate interplay between cellular metabolism, especially glutaminergic and glycolytic, and the establishment of the canonical antiviral response characterized by the expression of antiviral effectors, potentially paving the way for novel strategies to modulate antiviral responses through metabolic interventions.


Subject(s)
Glutamine , Interferon-beta , Oxidative Phosphorylation , Poly I-C , Zika Virus , Humans , Glutamine/metabolism , A549 Cells , Poly I-C/pharmacology , Interferon-beta/metabolism , Oxidative Phosphorylation/drug effects , Zika Virus/drug effects , Zika Virus/physiology , Antiviral Agents/pharmacology , Glycolysis/drug effects , Dengue Virus/drug effects , Dengue Virus/physiology , Virus Replication/drug effects , Host-Pathogen Interactions , Viperin Protein
6.
Cytokine ; 183: 156731, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39168064

ABSTRACT

Subunit vaccines drive immune cell-cell interactions in the lymph node (LN), yet it remains unclear how distinct adjuvants influence the chemokines responsible for this interaction in the tissue. Here, we tested the hypothesis that classic Th1-polarizing vaccines elicit a unique chemokine signature in the LN compared to other adjuvants. Polyinosinic:polycytidylic acid (Poly I:C) vaccination resulted in dynamic upregulation of CXCL9 that was localized in the interfollicular region, a response not observed after vaccination with alum or a combination of alum and poly I:C. Experiments using in vivo mouse models and live ex vivo LN slices revealed that poly I:C vaccination resulted in a type-I IFN response in the LN that led to the secretion of IFNγ, and type-I IFN and IFNγ were required for CXCL9 expression in this context. CXCL9 expression in the LN was correlated with an IgG2c antibody polarization after vaccination; however, genetic depletion of the receptor for CXCL9 did not prevent the development of this polarization. Additionally, we measured secretion of CXCL9 from ex vivo LN slices after stimulation with a variety of adjuvants and confirmed that adjuvants that induced IFNγ responses also promoted CXCL9 expression. Taken together, these results identify a CXCL9 signature in a suite of Th1-polarizing adjuvants and determined the pathway involved in driving CXCL9 in the LN, opening avenues to target this chemokine pathway in future vaccines.


Subject(s)
B-Lymphocytes , Chemokine CXCL9 , Interferon Type I , Interferon-gamma , Lymph Nodes , Mice, Inbred C57BL , Poly I-C , Signal Transduction , Vaccination , Animals , Chemokine CXCL9/metabolism , Poly I-C/pharmacology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Interferon Type I/metabolism , Interferon-gamma/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Adjuvants, Immunologic/pharmacology , Female
7.
Curr Microbiol ; 81(9): 296, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105989

ABSTRACT

Duck enteritis virus (DEV) may lead to vascular injury, gastrointestinal mucosal erosion, lymphoid organ injury, and Polyinosinic-polycytidylic acid (Poly I:C) has an antiviral effect by inducing low levels of interferon. The purpose of this study was to explore the pathogenesis of DEV-induced intestinal injury in ducks and to verify the therapeutic effects of different concentrations of Poly I:C. In this study, duck enteritis model was established by infecting healthy Pekin ducks with DEV. Duck intestinal tissues were extracted from normal control group, model group, and treatment group with different doses of Poly I:C. In vivo, HE and TUNEL staining were used to observe the morphological changes and apoptosis. In vitro, the proliferation and apoptosis of duck intestinal epithelial cells were evaluated by MTT assay, TUNEL staining, and flow cytometry. The results showed that Poly I:C protected ducks from DEV toxicity by improving intestinal morphology and inhibiting apoptosis. In addition, the antiviral effect of Poly I:C on DEV was found in a dose-dependent manner, with a more relatively obvious effect at a high dose of Poly I:C. All in all, these results demonstrated that Poly I:C played a vital role in the apoptosis induced by DEV in ducks and modest dose of Poly I:C treatment worked well and may provide important reference for the development of new antiviral drugs in the future.


Subject(s)
Apoptosis , Ducks , Enteritis , Poly I-C , Animals , Ducks/virology , Poly I-C/pharmacology , Poly I-C/administration & dosage , Apoptosis/drug effects , Enteritis/virology , Enteritis/drug therapy , Enteritis/veterinary , Poultry Diseases/virology , Poultry Diseases/drug therapy , Intestines/virology , Intestines/pathology , Antiviral Agents/pharmacology , Mardivirus/drug effects , Intestinal Mucosa/virology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology
8.
Oncoimmunology ; 13(1): 2395067, 2024.
Article in English | MEDLINE | ID: mdl-39188754

ABSTRACT

Toll-like receptor (TLR) agonists are being developed as anti-cancer therapeutics due to their potent immunostimulatory properties. However, clinical trials testing TLR agonists as monotherapy have often failed to demonstrate significant improvement over standard of care. We hypothesized that the anti-cancer efficacy of TLR agonist immunotherapy could be improved by combinatorial approaches. To prevent increased toxicity, often seen with systemic combination therapies, we developed a hydrogel to deliver TLR agonist combinations at low doses, locally, during cancer debulking surgery. Using tumor models of WEHI 164 and bilateral M3-9-M sarcoma and CT26 colon carcinoma, we assessed the efficacy of pairwise combinations of poly(I:C), R848, and CpG in controlling local and distant tumor growth. We show that combination of the TLR3 agonist poly(I:C) and TLR7/8 agonist R848 drives anti-tumor immunity against local and distant tumors. In addition, combination of local poly(I:C) and R848 sensitized tumors to systemic immune checkpoint blockade, improving tumor control. Mechanistically, we demonstrate that local therapy with poly(I:C) and R848 recruits inflammatory monocytes to the tumor draining lymph nodes early in the anti-tumor response. Finally, we provide proof of concept for intraoperative delivery of poly(I:C) and R848 together via a surgically applicable biodegradable hydrogel.


Subject(s)
Imidazoles , Immune Checkpoint Inhibitors , Poly I-C , Animals , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/therapeutic use , Mice , Poly I-C/administration & dosage , Poly I-C/pharmacology , Poly I-C/therapeutic use , Imidazoles/pharmacology , Imidazoles/administration & dosage , Imidazoles/therapeutic use , Immunotherapy/methods , Humans , Toll-Like Receptors/agonists , Cell Line, Tumor , Female , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/therapy , Mice, Inbred C57BL , Hydrogels/administration & dosage , Hydrogels/chemistry , Toll-Like Receptor Agonists
9.
Fish Shellfish Immunol ; 153: 109806, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39102971

ABSTRACT

Blood transcriptomics has emerged as a vital tool for tracking the immune system and supporting disease diagnosis, prognosis, treatment, and research. The present study was conducted to analyze the gene expression profile and potential biomarker candidates using the whole blood of mandarin fish (Siniperca chuatsi) infected with LPS or poly (I:C) at 0 h, 3 h, 6 h, and 12 h. Our data suggest that 310 shared differentially expressed genes (DEGs) were identified among each comparison group after LPS infection, and 137 shared DEGs were identified after poly (I:C) infection. A total of 62 shared DEGs were differentially expressed in all compared groups after LPS or poly (I:C) infection. Pathways analysis for DEGs in all different compared groups showed that cytokine-cytokine receptor interaction was the most enrichment pathway. The expression levels of genes C-X-C chemokine receptor type 2-like (cxcr2), chemokine (C-C motif) receptor 9a (ccr9a), chemokine (C-C motif) receptor 9b (ccr9b), chemokine (C-X-C motif) receptor 4b (cxcr4b), and interleukin 10 receptor alpha (il10ra) were significantly different in all compared groups and most enriched in cytokine-cytokine receptor interaction pathway. The protein-protein interactions analysis among all shared DEGs showed that cxcr4 was the hub gene with the highest degree. The biomarker candidates discovered in this study may, following validation, prove effective as diagnostic tools in monitoring mandarin fish diseases.


Subject(s)
Biomarkers , Fish Diseases , Fish Proteins , Lipopolysaccharides , Perciformes , Poly I-C , Transcriptome , Animals , Fish Diseases/immunology , Poly I-C/pharmacology , Biomarkers/blood , Lipopolysaccharides/pharmacology , Perciformes/genetics , Perciformes/immunology , Perciformes/blood , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/blood , Gene Expression Profiling/veterinary , Immunity, Innate/genetics , Gene Expression Regulation/immunology , Gene Expression Regulation/drug effects
10.
Neurosci Biobehav Rev ; 164: 105844, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39106940

ABSTRACT

This systematic review explored the impact of maternal immune activation (MIA) on learning and memory behavior in offspring, with a particular focus on sexual dimorphism. We analyzed 20 experimental studies involving rodent models (rats and mice) exposed to either lipopolysaccharide (LPS) or POLY I:C during gestation following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Our findings reveal that most studies report a detrimental impact of MIA on the learning and memory performance of offspring, highlighting the significant role of prenatal environmental factors in neurodevelopment. Furthermore, this review underscores the complex effects of sex, with males often exhibiting more pronounced cognitive impairment compared to females. Notably, a small subset of studies report enhanced cognitive function following MIA, suggesting complex, context-dependent outcomes of prenatal immune challenges. This review also highlights sex differences caused by the effects of MIA in terms of cytokine responses, alterations in gene expression, and differences in microglial responses as factors that contribute to the cognitive outcomes observed.


Subject(s)
Learning , Memory , Prenatal Exposure Delayed Effects , Animals , Female , Pregnancy , Prenatal Exposure Delayed Effects/immunology , Learning/physiology , Memory/physiology , Sex Characteristics , Mice , Lipopolysaccharides/pharmacology , Poly I-C/pharmacology , Rats , Male
11.
Respir Res ; 25(1): 314, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160577

ABSTRACT

BACKGROUND: While asthma exacerbations remain a major challenge in patient management, few animal models exist to explore the underlying mechanisms. Here, we established an animal model of asthma that can be used to study pathophysiological mechanisms and therapeutic strategies on asthma exacerbation. METHODS: Female BALB/c mice were sensitized and exposed to PBS or Dermatophagoides pteronyssinus (DerP) extract for 11 weeks. Asthmatic phenotype was assessed through lung inflammation, bronchial hyperresponsiveness and bronchial smooth muscle remodeling. Asthmatic and control mice were exposed once or three times to poly(I:C) to simulate virus-induced inflammation. RESULTS: Fourteen days after exposure to DerP, asthmatic mice showed resolution of inflammation with sustained bronchial hyperresponsiveness and bronchial smooth muscle remodeling compared to control. At this stage, when mice were subjected to a single exposure to poly(I:C), control and asthmatic mice were characterized by a significant increase in neutrophilic inflammation and bronchial hyperresponsiveness. When mice were repeatedly exposed to poly(I:C), control mice showed a significant decrease in neutrophilic inflammation and bronchial hyperresponsiveness, while asthmatic mice experienced worsening of these outcomes. CONCLUSIONS: This observational study report an asthmatic mouse model that can undergo exacerbation after repeated exposure to poly(I:C). Our findings on pulmonary adaptation in control mice may also pave the way for further research into the mechanism of adaptation that may be impaired in asthma and raise the question of whether asthma exacerbation may be a loss of adaptation.


Subject(s)
Asthma , Lung , Mice, Inbred BALB C , Poly I-C , Animals , Asthma/physiopathology , Female , Poly I-C/toxicity , Mice , Lung/physiopathology , Lung/drug effects , Adaptation, Physiological/physiology , Disease Models, Animal , Bronchial Hyperreactivity/physiopathology , Bronchial Hyperreactivity/chemically induced , Airway Remodeling/drug effects , Airway Remodeling/physiology
12.
Proc Natl Acad Sci U S A ; 121(35): e2406928121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39178233

ABSTRACT

Autism spectrum disorders (ASD) can be caused by environmental factors. These factors act early in the development of the nervous system and induce stereotyped repetitive behaviors and diminished social interactions, among other outcomes. Little is known about how these behaviors are produced. In pregnant women, delivery of valproic acid (VPA) (to control seizure activity or stabilize mood) or immune activation by a virus increases the incidence of ASD in offspring. We found that either VPA or Poly Inosine:Cytosine (which mimics a viral infection), administered at mouse embryonic day 12.5, induced a neurotransmitter switch from GABA to glutamate in PV- and CCK-expressing interneurons in the medial prefrontal cortex by postnatal day 10. The switch was present for only a brief period during early postnatal development, observed in male and female mice at postnatal day 21 and reversed in both males and females by postnatal day 30. At postnatal day 90, male mice exhibited stereotyped repetitive behaviors and diminished social interaction while female mice exhibited only stereotyped repetitive behavior. Transfecting GAD1 in PV- and CCK-expressing interneurons at postnatal day 10, to reintroduce GABA expression, overrode the switch and prevented expression of autistic-like behavior. These findings point to an important role of neurotransmitter switching in mediating the environmental causes of autism.


Subject(s)
Valproic Acid , gamma-Aminobutyric Acid , Animals , Female , Mice , Male , Pregnancy , Valproic Acid/toxicity , gamma-Aminobutyric Acid/metabolism , Interneurons/metabolism , Animals, Newborn , Behavior, Animal , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/pathology , Glutamate Decarboxylase/metabolism , Glutamate Decarboxylase/genetics , Autistic Disorder/etiology , Autistic Disorder/metabolism , Glutamic Acid/metabolism , Neurotransmitter Agents/metabolism , Poly I-C , Prefrontal Cortex/metabolism , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/etiology , Autism Spectrum Disorder/pathology , Cholecystokinin/metabolism , Parvalbumins/metabolism , Mice, Inbred C57BL , Stereotyped Behavior/drug effects
13.
Front Immunol ; 15: 1439465, 2024.
Article in English | MEDLINE | ID: mdl-39211041

ABSTRACT

The economic importance of lumpfish (Cyclopterus lumpus) is increasing, but several aspects of its immune responses are not well understood. To discover genes and mechanisms involved in the lumpfish antiviral response, fish were intraperitoneally injected with either the viral mimic polyinosinic:polycytidylic acid [poly(I:C)] or phosphate-buffered saline (PBS; vehicle control), and head kidneys were sampled 24 hours post-injection (hpi) for transcriptomic analyses. RNA sequencing (RNA-Seq) (adjusted p-value <0.05) identified 4,499 upregulated and 3,952 downregulated transcripts in the poly(I:C)-injected fish compared to the PBS-injected fish. Eighteen genes identified as differentially expressed by RNA-Seq were included in a qPCR study that confirmed the upregulation of genes encoding proteins with antiviral immune response functions (e.g., rsad2) and the downregulation of genes (e.g., jarid2b) with potential cellular process functions. In addition, transcript expression levels of 12 members of the interferon regulatory factor (IRF) family [seven of which were identified as poly(I:C)-responsive in this RNA-Seq study] were analyzed using qPCR. Levels of irf1a, irf1b, irf2, irf3, irf4b, irf7, irf8, irf9, and irf10 were significantly higher and levels of irf4a and irf5 were significantly lower in the poly(I:C)-injected fish compared to the PBS-injected fish. This research and associated new genomic resources enhance our understanding of the genes and molecular mechanisms underlying the lumpfish response to viral mimic stimulation and help identify possible therapeutic targets and biomarkers for viral infections in this species.


Subject(s)
Head Kidney , Interferon Regulatory Factors , Poly I-C , Transcriptome , Animals , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Head Kidney/immunology , Head Kidney/metabolism , Poly I-C/immunology , Perciformes/immunology , Perciformes/genetics , Gene Expression Profiling , Fish Proteins/genetics , Fish Proteins/immunology , Fish Diseases/immunology , Fish Diseases/virology , Fishes/immunology , Fishes/genetics
14.
Brain Behav Immun ; 121: 351-364, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39089536

ABSTRACT

BACKGROUND: Maternal immune activation (MIA) triggers neurobiological changes in offspring, potentially reshaping the molecular synaptic landscape, with the hippocampus being particularly vulnerable. However, critical details regarding developmental timing of these changes and whether they differ between males and females remain unclear. METHODS: We induced MIA in C57BL/6J mice on gestational day nine using the viral mimetic poly(I:C) and performed mass spectrometry-based proteomic analyses on hippocampal synaptoneurosomes of embryonic (E18) and adult (20 ± 1 weeks) MIA offspring. RESULTS: In the embryonic synaptoneurosomes, MIA led to lipid, polysaccharide, and glycoprotein metabolism pathway disruptions. In the adult synaptic proteome, we observed a dynamic shift toward transmembrane trafficking, intracellular signalling cascades, including cell death and growth, and cytoskeletal organisation. In adults, many associated pathways overlapped between males and females. However, we found distinct sex-specific enrichment of dopaminergic and glutamatergic pathways. We identified 50 proteins altered by MIA in both embryonic and adult samples (28 with the same directionality), mainly involved in presynaptic structure and synaptic vesicle function. We probed human phenome-wide association study data in the cognitive and psychiatric domains, and 49 of the 50 genes encoding these proteins were significantly associated with the investigated phenotypes. CONCLUSIONS: Our data emphasise the dynamic effects of viral-like MIA on developing and mature hippocampi and provide novel targets for study following prenatal immune challenges. The 22 proteins that changed directionality from the embryonic to adult hippocampus, suggestive of compensatory over-adaptions, are particularly attractive for future investigations.


Subject(s)
Hippocampus , Mice, Inbred C57BL , Prenatal Exposure Delayed Effects , Proteome , Synapses , Animals , Hippocampus/metabolism , Female , Proteome/metabolism , Pregnancy , Male , Mice , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/metabolism , Synapses/metabolism , Poly I-C/pharmacology , Proteomics/methods , Humans
15.
Int J Mol Sci ; 25(16)2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39201612

ABSTRACT

Although therapies based on mesenchymal stromal cells (MSCs) are being implemented in clinical settings, many aspects regarding these procedures require further optimization. Domestic dogs suffer from numerous immune-mediated diseases similar to those found in humans. This study aimed to assess the immunomodulatory activity of canine (c) Wharton jelly (WJ)-derived MSCs and refer them to human (h) MSCs isolated from the same tissue. Canine MSC(WJ)s appeared to be more prone to in vitro aging than their human counterparts. Both canine and human MSC(WJ)s significantly inhibited the activation as well as proliferation of CD4+ and CD8+ T cells. The treatment with IFNγ significantly upregulated indoleamine-2,3-dioxygenase 1 (IDO1) synthesis in human and canine MSC(WJ)s, and the addition of poly(I:C), TLR3 ligand, synergized this effect in cells from both species. Unstimulated human and canine MSC(WJ)s released TGFß at the same level (p > 0.05). IFNγ significantly increased the secretion of TGFß in cells from both species (p < 0.05); however, this response was significantly stronger in human cells than in canine cells. Although the properties of canine and human MSC(WJ)s differ in detail, cells from both species inhibit the proliferation of activated T cells to a very similar degree and respond to pro-inflammatory stimulation by enhancing their anti-inflammatory activity. These results suggest that testing MSC transplantation in naturally occurring immune-mediated diseases in dogs may have high translational value for human clinical trials.


Subject(s)
Cell Proliferation , Mesenchymal Stem Cells , Wharton Jelly , Dogs , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/immunology , Mesenchymal Stem Cells/cytology , Animals , Humans , Wharton Jelly/cytology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Immunomodulation , Interferon-gamma/metabolism , Cells, Cultured , Transforming Growth Factor beta/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lymphocyte Activation/immunology , Poly I-C/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism
16.
Fish Shellfish Immunol ; 153: 109835, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39147180

ABSTRACT

Galectin-4 belongs to the galactoside-binding protein family and is a type of tandem repeat galectin. Despite previous studies indicating its importance in fish immunology, a comprehensive investigation is necessary to fully understand its role in immunomodulatory functions and cellular dynamics. Therefore, this study aimed to explore the immunomodulatory functions of galectin-4 with a particular focus on its antimicrobial and cellular proliferative properties. The open reading frame of PhGal4 spans 1092 base pairs and encodes a soluble protein of 363 amino acids with a theoretical isoelectric point (IEP) of 6.39 and a molecular weight of 39.411 kDa. Spatial expression analysis under normal physiological conditions revealed ubiquitous expression of PhGal4 across all examined tissues, with the highest level observed in intestinal tissue. Upon stimulation with poly I:C, LPS, and L. garvieae, a significant increase (p < 0.05) in PhGal4 expression was observed in both blood and spleen tissues. Subsequent subcellular localization assay demonstrated that PhGal4 was predominantly localized in the cytoplasm. The recombinant PhGal4 (rPhGal4) exhibited specific binding capabilities to pathogen-associated molecular patterns (PAMPs), including LPS and peptidoglycan, but not poly I:C. The rPhGal4 negatively affected the bacterial growth kinetics. Additionally, rPhGal4 demonstrated complete hemagglutination of fish erythrocytes, which could be inhibited by the presence of D-galactose and α-lactose. The overexpression of PhGal4 in FHM epithelial cells demonstrated a significant suppression of viral replication during VHSV infection. Furthermore, the in vitro scratch assay and WST-1 assay demonstrated a wound healing effect of PhGal4 overexpression in FHM cells, potentially achieved through the promotion of cell proliferation by activating genes involved in cell cycle regulation. In conclusion, the responsive expression to immune stimuli, antimicrobial properties, and cell proliferation promotion of PhGal4 suggest that it plays a crucial role in immunomodulation and cellular dynamics of red-lip mullet. The findings in this study shed light on the multifunctional nature of galectin-4 in teleost fish.


Subject(s)
Cell Proliferation , Fish Proteins , Galectin 4 , Smegmamorpha , Animals , Fish Proteins/genetics , Fish Proteins/immunology , Fish Proteins/chemistry , Cell Proliferation/drug effects , Galectin 4/genetics , Galectin 4/immunology , Galectin 4/chemistry , Smegmamorpha/immunology , Smegmamorpha/genetics , Immunity, Innate/genetics , Phylogeny , Amino Acid Sequence , Gene Expression Profiling/veterinary , Fish Diseases/immunology , Gene Expression Regulation/immunology , Poly I-C/pharmacology , Sequence Alignment/veterinary , Lipopolysaccharides/pharmacology
17.
Fish Shellfish Immunol ; 153: 109829, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39142373

ABSTRACT

As a vital pathway for cellular energy production, mitochondrial fatty acid ß-oxidation (FAO) is essential in regulating immune responses to bacterial pathogens and maintaining intracellular homeostasis in vertebrates. However, the specific role of FAO in antiviral innate immune response in macrophages remains insufficiently understood. In this study, virus infection simulated by poly(I:C) inhibited FAO, as indicated by the reduced expression of FAO-related genes and proteins in the head kidney of large yellow croaker, with similar results observed in poly(I:C)-stimulated macrophages. Then, inhibition of FAO by supplementary mildronate in vivo and etomoxir treatment in vitro revealed varying increases in the mRNA expression of antiviral innate immune response genes after stimulated by poly(I:C) in the head kidney and macrophages. Notably, etomoxir significantly facilitated the transcriptional up-regulation of the IFNh promoter by IRF3. Moreover, inhibiting FAO by knockdown of cpt1b promoted antiviral innate immune response triggered by poly(I:C) in macrophages. Conversely, activating FAO through overexpression of cpt1b or cpt2 significantly reduced the mRNA levels of antiviral response genes in macrophages stimulated by poly(I:C). Unlike etomoxir, cpt1b overexpression inhibited the transcriptional up-regulation of the IFNh promoter by IRF3. Furthermore, in vivo dietary palm oil feeding and in vitro exposure to palmitic acid inhibited the antiviral innate immune response triggered by poly(I:C) in the head kidney and macrophages, respectively. These effects were partly associated with FAO activation, as evidenced by etomoxir. In summary, this study elucidates FAO's critical role in regulating antiviral innate immune response in head kidney macrophages. These findings not only deepen insights into the interaction between metabolic remodeling and host immune responses, but also offer valuable guidance for developing nutritional strategies to improve antiviral immunity in aquaculture.


Subject(s)
Fatty Acids , Fish Diseases , Head Kidney , Immunity, Innate , Macrophages , Perciformes , Poly I-C , Animals , Immunity, Innate/drug effects , Immunity, Innate/genetics , Perciformes/immunology , Head Kidney/immunology , Macrophages/immunology , Macrophages/drug effects , Fish Diseases/immunology , Poly I-C/pharmacology , Mitochondria , Oxidation-Reduction , Fish Proteins/genetics , Fish Proteins/immunology
18.
Fish Shellfish Immunol ; 153: 109840, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39153579

ABSTRACT

Infectious diseases have significantly impacted Atlantic salmon aquaculture worldwide. Modulating fish immunity with immunostimulant-containing functional feeds could be an effective strategy in mitigating disease problems. Previously, we characterized the impact of polyriboinosinic polyribocytidylic acid (pIC) and formalin-killed typical Aeromonas salmonicida bacterin on miRNA expression in Atlantic salmon fed a commercial diet with and without immunostimulant CpG. A set of miRNA biomarkers of Atlantic salmon head kidney responding to pIC and/or bacterin immune stimulations was identified (Xue et al., 2019) [1]. Herein, we report a complementary qPCR study that investigated the impact of the pIC, bacterin and dietary CpG on the expression of immune-relevant mRNAs (n = 31) using the same samples as in the previous study (Xue et al., 2019) [1]. Twenty-six of these genes were predicted target transcripts of the pIC- and/or bacterin-responsive miRNAs identified in the earlier study. The current data showed that pIC and/or bacterin stimulations significantly modulated the majority of the qPCR-analyzed genes involved in various immune pathways. Some genes responded to both stimulations (e.g. tnfa, il10rb, ifng, irf9, cxcr3, campb) while others appeared to be stimulation specific [e.g. irf3, irf7a, il1r1, mxa, mapk3 (pIC only); clra (bacterin only)]. A. salmonicida bacterin stimulation produced a strong inflammatory response (e.g. higher expression of il1b, il8a and tnfa), while salmon stimulated with pIC showed robust interferon responses (both type I and II). Furthermore, the current data indicated significant down-regulation of immune-relevant transcripts (e.g. tlr9, irf5, il1r1, hsp90ab1, itgb2) by dietary immunostimulant CpG, especially among pre-injection and PBS-injected fish. Together with our prior miRNA study, the present research provided complementary information on Atlantic salmon anti-viral and anti-bacterial immune responses and on how dietary CpG may modulate these responses.


Subject(s)
Adjuvants, Immunologic , Aeromonas salmonicida , Animal Feed , Diet , RNA, Messenger , Salmo salar , Animals , Salmo salar/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Animal Feed/analysis , Diet/veterinary , RNA, Messenger/genetics , RNA, Messenger/metabolism , Aeromonas salmonicida/physiology , Immunity, Innate/drug effects , Biomarkers , Fish Diseases/immunology , Dietary Supplements/analysis , Oligodeoxyribonucleotides/pharmacology , Oligodeoxyribonucleotides/administration & dosage , MicroRNAs/genetics , Head Kidney/immunology , Poly I-C/pharmacology , Poly I-C/administration & dosage
19.
Fish Shellfish Immunol ; 153: 109867, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39214263

ABSTRACT

The study of mussels (Mytilus galloprovincialis) has grown in importance in recent years due to their high economic value and resistance to pathogens. Because of the biological characteristics revealed by mussel genome sequencing, this species is a valuable research model. The high genomic variability and diversity, particularly in immune genes, may be responsible for their resistance to pathogens found in seawater and continuously filtered and internalized by them. These facts, combined with the lack of proven mussel susceptibility to viruses in comparison to other bivalves such as oysters, result in a lack of studies on mussel antiviral response. We used RNA-seq to examine the genomic response of mussel hemocytes after they were exposed to poly I:C, simulating immune cell contact with viral dsRNA. Apoptosis and the molecular axis IRFs/STING-IFI44/IRGC1 were identified as the two main pathways in charge of the response but we also found a modulation of lncRNAs. Finally, in order to obtain new information about the response of mussels to putative natural challenges, we used VHSV virus (Viral Hemorrhagic Septicemia Virus) to run some functional analysis and confirm poly I:C's activity as an immunomodulator in a VHSV waterborne stimulation. Both, poly I:C as well as an injury stimulus (filtered sea water injection) accelerated the viral clearance by hemocytes and altered the expression of several immune genes, including IL-17, IRF1 and viperin.


Subject(s)
Immunity, Innate , Mytilus , Poly I-C , Transcriptome , Animals , Poly I-C/pharmacology , Mytilus/immunology , Mytilus/genetics , Mytilus/virology , Immunity, Innate/genetics , Novirhabdovirus/physiology , Hemocytes/immunology , Gene Expression Profiling/veterinary
20.
Fish Shellfish Immunol ; 153: 109853, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39173983

ABSTRACT

Inhibitors of NF-κB (IκBs) have been implicated as major components of the Rel/NF-κB signaling pathway, playing an important negative regulatory role in host antiviral immunity such as in the activation of interferon (IFN) in vertebrates. In the present study, the immunomodulatory effect of IκB (CgIκB2) on the expression of interferon-like protein (CgIFNLP) was evaluated in Pacific oyster (Crassostrea gigas). After poly (I:C) stimulation, the mRNA expression level of CgIκB2 in haemocytes was significantly down-regulated at 3-12 h while up-regulated at 48-72 h. The mRNA expression of CgIκB2 in haemocytes was significantly up-regulated at 3 h after rCgIFNLP stimulation. In the CgIκB2-RNAi oysters, the mRNA expression of CgIFNLP, interferon regulatory factor-8 (CgIRF8) and NF-κB subunit (CgRel), the abundance of CgIFNLP and CgIRF8 protein in haemocytes, as well as the abundance of CgRel protein in nucleus were significantly increased after poly (I:C) stimulation. Immunofluorescence assay showed that nuclear translocation of CgIRF8 and CgRel protein was promoted in CgIκB2-RNAi oysters compared with that in EGFP-RNAi group. In the CgRel-RNAi oysters, the mRNA and protein expression level of CgIFNLP significantly down-regulated after poly (I:C) stimulation. The collective results indicated that CgIκB2 plays an important role in regulating CgIFNLP expression through its effects on Rel/NF-κB and IRF signaling pathways.


Subject(s)
Crassostrea , Gene Expression Regulation , Interferons , NF-kappa B , Poly I-C , Signal Transduction , Animals , Crassostrea/genetics , Crassostrea/immunology , Poly I-C/pharmacology , NF-kappa B/genetics , NF-kappa B/metabolism , Gene Expression Regulation/immunology , Interferons/genetics , Interferons/immunology , Interferons/metabolism , Immunity, Innate/genetics , I-kappa B Proteins/genetics , I-kappa B Proteins/metabolism , Hemocytes/immunology , Hemocytes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL