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

Publication year range
1.
EMBO J ; 41(23): e111192, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36314682

ABSTRACT

Intracerebral hemorrhages are recognized risk factors for neurodevelopmental disorders and represent early biomarkers for cognitive dysfunction and mental disability, but the pathways leading to their occurrence are not well defined. We report that a single intrauterine exposure of the immunostimulant Poly I:C to pregnant mice at gestational day 9, which models a prenatal viral infection and the consequent maternal immune activation, induces the defective formation of brain vessels and causes intracerebral hemorrhagic events, specifically in male offspring. We demonstrate that maternal immune activation promotes the production of the TGF-ß1 active form and the consequent enhancement of pSMAD1-5 in males' brain endothelial cells. TGF-ß1, in combination with IL-1ß, reduces the endothelial expression of CD146 and claudin-5, alters the endothelium-pericyte interplay resulting in low pericyte coverage, and increases hemorrhagic events in the adult offspring. By showing that exposure to Poly I:C at the beginning of fetal cerebral angiogenesis results in sex-specific alterations of brain vessels, we provide a mechanistic framework for the association between intragravidic infections and anomalies of the neural vasculature, which may contribute to neuropsychiatric disorders.


Subject(s)
Cerebral Hemorrhage , Prenatal Exposure Delayed Effects , Animals , Female , Male , Mice , Pregnancy , Behavior, Animal , Brain/blood supply , Brain/pathology , Cerebral Hemorrhage/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Poly I-C/adverse effects , Prenatal Exposure Delayed Effects/pathology , Transforming Growth Factor beta1/metabolism
2.
Brain Behav Immun ; 108: 162-175, 2023 02.
Article in English | MEDLINE | ID: mdl-36503051

ABSTRACT

Exposure to inflammatory stressors during fetal development is a major risk factor for neurodevelopmental disorders (NDDs) in adult offspring. Maternal immune activation (MIA), induced by infection, causes an acute increase in pro-inflammatory cytokines which can increase the risk for NDDs directly by inducing placental and fetal brain inflammation, or indirectly through affecting maternal care behaviours thereby affecting postnatal brain development. Which of these two potential mechanisms dominates in increasing offspring risk for NDDs remains unclear. Here, we show that acute systemic maternal inflammation induced by the viral mimetic polyinosinic:polycytidylic acid (poly I:C) on gestational day 15 of rat pregnancy affects offspring and maternal behaviour, offspring cognition, and expression of NDD-relevant genes in the offspring brain. Dams exposed to poly I:C elicited an acute increase in the pro-inflammatory cytokine tumour necrosis factor (TNF; referred to here as TNFα), which predicted disruption of key maternal care behaviours. Offspring of poly I:C-treated dams showed early behavioural and adult cognitive deficits correlated to the maternal TNFα response, but, importantly, not with altered maternal care. We also found interacting effects of sex and treatment on GABAergic gene expression and DNA methylation in these offspring in a brain region-specific manner, including increased parvalbumin expression in the female adolescent frontal cortex. We conclude that the MIA-induced elevation of TNFα in the maternal compartment affects fetal neurodevelopment leading to altered offspring behaviour and cognition. Our results suggest that a focus on prenatal pathways affecting fetal neurodevelopment would provide greater insights into the mechanisms underpinning the TNFα-mediated genesis of altered offspring behaviour and cognition following maternal inflammation.


Subject(s)
Neurodevelopmental Disorders , Prenatal Exposure Delayed Effects , Rats , Animals , Female , Pregnancy , Humans , Tumor Necrosis Factor-alpha/pharmacology , Behavior, Animal/physiology , Placenta/metabolism , Cytokines , Poly I-C/adverse effects , Maternal Behavior , Inflammation/metabolism , Disease Models, Animal
3.
Pancreatology ; 23(7): 767-776, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37661465

ABSTRACT

BACKGROUND: IgG4-related autoimmune pancreatitis (AIP) is considered to be a T cell-mediated autoimmune disease. However, CD8+ T cells have only received brief mention, and have yet to be completely studied. The study aimed to investigate the expression of signaling lymphocytic activation molecule family 7 (SLAMF7) on CD8+ T cells and the features of SLAMF7+CD8+ T cells in MRL/Mp mice with AIP. METHODS: A murine model of AIP was established by intraperitoneal injection with polyinosinic:polycytidylic acid (poly I:C) for 8 weeks. Dexamethasone treatment was daily administrated for the last 2 weeks during a 6-week course of poly I:C. SLAMF7 expression on CD8+ T cells in the spleen and pancreas was detected by flow cytometry. Granzyme B (GZMB) and cytokines including IFN-γ, TNF-α, and IL-2, were monitored in an in vitro T cell activation assay. Dexamethasone suppression assays were performed to downregulate SLAMF7 expression on T cells upon T cell receptor stimulation. RESULTS: AIP in MRL/Mp mice was induced by repeated intraperitoneal administration of poly I:C and CD8+ T cells were increased in the inflamed pancreas. SLAMF7+CD8+ T cells were elevated in the spleen and pancreas of AIP mice. SLAMF7+CD8+ T subsets produced more GZMB, IFN-γ, TNF-α and IL-2 than SLAMF7-CD8+ T subsets. Dexamethasone treatment ameliorated pancreatic inflammatory and fibrosis of AIP. Dexamethasone could downregulate SLAMF7+CD8+ T cells and reduce GZMB, IFN-γ and TNF-α levels both in vitro and in vivo. CONCLUSIONS: Increased SLAMF7+CD8+ T cells exhibit enhanced cytotoxicity and cytokines secretion capacity, which may be involved in the pathogenesis of AIP.


Subject(s)
Autoimmune Diseases , Autoimmune Pancreatitis , Mice , Animals , CD8-Positive T-Lymphocytes , Interleukin-2/adverse effects , Tumor Necrosis Factor-alpha , Autoimmune Diseases/pathology , Poly I-C/adverse effects , Dexamethasone/adverse effects , Signaling Lymphocytic Activation Molecule Family/metabolism
4.
Contact Dermatitis ; 89(4): 230-240, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37463838

ABSTRACT

INTRODUCTION: Poly(I:C) is recognised by endosomal Toll-like receptor 3 (TLR3) and activates cytotoxic CD8(+) lymphocytes and natural killer (NK) cells. It has been shown that the viral TLR3 agonist induces robust and long-lasting T-cell-mediated responses. In addition, TLR3 modulates the contact hypersensitivity reaction. OBJECTIVE: This study aimed to determine whether poly(I:C) injection can induce NK-mediated hapten reactivity in mice. METHODS: Mice were treated with poly(I:C), and their response to dinitrofluorobenzene hapten was measured by assessing ear swelling and serum interferon gamma (IFN-γ) production. Adoptive cell transfer and cell sorting were used to investigate the mechanism of the reaction, and the phenotype of poly(I:C)-activated liver NK cells was determined by flow cytometry analysis. RESULTS: The results showed that poly(I:C) administration increased ear swelling, serum IFN-γ levels and the response to hapten in both immunocompetent and T- and B-cell-deficient mice. Only liver poly(I:C)-activated DX5(+) NK cells were able to transfer reactivity to hapten into a naive recipient. Induction of liver NK cells after poly(I:C) administration was TLR3/TRIF- and IFN-γ-dependent, interleukin 12-independent, and not modulated by MyD88. CONCLUSION: This study provides new insights into how poly(I:C) stimulates NK-mediated reactivity to hapten and suggests that liver NK cells may modulate the immune response to non-pathogenic factors during viral infection.


Subject(s)
Dermatitis, Allergic Contact , Toll-Like Receptor 3 , Mice , Animals , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/genetics , Ligands , Dermatitis, Allergic Contact/etiology , Killer Cells, Natural , Poly I-C/adverse effects , Interferon-gamma , Mice, Inbred C57BL
5.
Biochem Biophys Res Commun ; 588: 90-96, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34953211

ABSTRACT

Here we investigated the gender difference in murine cholangitis resembling human primary biliary cholangitis (PBC) caused by synthetic double-stranded RNA, and underlying hepatic innate immune responses. Female C57Bl/6 mice given repeated injections of polyinosinic-polycytidylic acid (poly I:C) for 24 weeks developed overt cholangitis with positive serum anti-mitochondria-M2 antibody, whereas male mice showed minimal pathological changes without induction in autoantibody. Poly I:C induced hepatic inflammatory cytokines and type-I interferons predominantly in females. Hepatic expression levels of toll-like receptor (TLR) 3 and melanoma differentiation-associated protein (MDA) 5 were equivalent in both genders; however, both mRNA and protein levels of retinoic acid-inducible gene (RIG)-I were nearly doubled in female livers. Following 4-week injections of poly I:C, not only hepatic RIG-I, but also TLR3 and MDA5 showed female-predominance. Moreover, hepatic RIG-I levels were 25% lower in ovariectomized mice, whereas supplementation of 17 ß-estradiol enhanced hepatic RIG-I expression, as well as cytokine induction. These results clearly indicate that hepatic RIG-I expression is potentiated by estrogen, and triggers gender-dependent hepatic innate immune response against double-stranded RNA, which most likely play a pivotal role in the pathogenesis of autoimmune cholangiopathies including PBC.


Subject(s)
Cholangitis/pathology , RNA, Double-Stranded/adverse effects , Sex Characteristics , Animals , Autoantibodies/blood , Cholangitis/blood , Cholangitis/immunology , Cytokines/metabolism , DEAD Box Protein 58/metabolism , Estrogens/pharmacology , Female , Interferon-Induced Helicase, IFIH1/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Poly I-C/adverse effects , Receptors, Pattern Recognition/metabolism , Toll-Like Receptor 3/metabolism
6.
Dev Psychobiol ; 64(5): e22278, 2022 07.
Article in English | MEDLINE | ID: mdl-35603415

ABSTRACT

The mechanisms that link maternal immune activation (MIA) with the onset of neurodevelopmental disorders remain largely unclear. Accelerated puberty is also associated with a heightened risk for psychopathology in later life, but there is a dearth of evidence on the impacts of maternal infection on pubertal timing. We examined the effects of MIA on reproductive development, mechanical allodynia, and sensorimotor gating in juvenile, adolescent, and adult male and female mice. Moreover, we investigated hypothalamic neural markers associated with the reproductive and stress axes. Finally, we tested the mitigating effects of environmental enrichment (EE), which has clinical relevancy in human rehabilitation settings. Our results show that administration of polyinosinic-polycytidylic acid (poly(I:C)) on gestational day 12.5 led to early preputial separation, vaginal openings, and age of first estrus in offspring. MIA exposure altered pain sensitivity across development and modestly altered prepulse inhibition. The downregulation of Nr3c1 and Oprk mRNA in the hypothalamus of juvenile mice suggests that MIA's effects may be mediated through disruption of hypothalamic-pituitary-adrenal axis activity. In contrast, life-long housing with EE rescued many of these MIA-induced consequences. Overall, our findings suggest that accelerated puberty may be associated with the deleterious effects of infection during pregnancy and the onset of psychopathology.


Subject(s)
Hypothalamo-Hypophyseal System , Prenatal Exposure Delayed Effects , Adolescent , Animals , Behavior, Animal/physiology , Disease Models, Animal , Female , Humans , Hyperalgesia , Male , Mice , Mice, Inbred C57BL , Pituitary-Adrenal System , Poly I-C/adverse effects , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Puberty
7.
Mol Pharm ; 18(8): 3099-3107, 2021 08 02.
Article in English | MEDLINE | ID: mdl-34228470

ABSTRACT

Celiac disease is a chronic inflammatory condition characterized by activation of the immune system in response to deamidation of gluten peptides brought about by tissue transglutaminase-2 (TG2). Overexpression of interleukin-15 (IL-15) in the intestinal epithelium and the lamina propria leads to the dysregulation of the immune system, leading to epithelial damage. The goal of this study was to develop an RNA interference therapeutic strategy for celiac disease using a combination of TG2 and IL-15 gene silencing in the inflamed intestine. TG2 and IL-15 silencing siRNA sequences, along with scrambled control, were encapsulated in a nanoparticle-in-microsphere oral system (NiMOS) and administered in a poly(I:C) mouse model of celiac disease. Single TG2 and IL-15 siRNA therapy and the combination showed effective gene silencing in vivo. Additionally, it was found that IL-15 gene silencing alone and combination in the NiMOS significantly reduced other proinflammatory cytokines. The tissue histopathology data also confirmed a reduction in immune cell infiltration and restoration of the mucosal architecture and barrier function in the intestine upon treatment. Overall, the results of this study show evidence that celiac disease can be potentially treated with an oral microsphere formulation using a combination of TG2 and IL-15 RNA interference therapeutic strategies.


Subject(s)
Celiac Disease/drug therapy , Celiac Disease/genetics , Gastroenteritis/drug therapy , Gastroenteritis/genetics , Interleukin-15/genetics , Microspheres , Nanoparticle Drug Delivery System/chemistry , Nanoparticles/chemistry , Protein Glutamine gamma Glutamyltransferase 2/genetics , RNA Interference , Administration, Oral , Animals , Celiac Disease/chemically induced , Disease Models, Animal , Drug Compounding/methods , Gastroenteritis/chemically induced , Interleukin-15/administration & dosage , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Poly I-C/adverse effects , Protein Glutamine gamma Glutamyltransferase 2/administration & dosage , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Treatment Outcome
8.
J Pharmacol Sci ; 144(4): 189-196, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33070837

ABSTRACT

Pneumonia is a common illness that continues to be the major killer of remaining to be a significant source of morbidity and mortality in the patient population. Many microorganisms cause pneumonia, and now concern is turning to the importance of the cause the new therapies for viral pneumonia. In the current study, we report the effect of andrographolide sulfonate, a water-soluble form of andrographolide (trade name: Xi-Yan-Ping Injection), on poly I: C-induced pneumonia. Andrographolide sulfonate was administrated through intraperitoneal injection to mice with poly I: C-induced pneumonia. Recruitment of airway inflammatory cells, alteration of lung histological induced by Poly I: C were significantly ameliorated by andrographolide sulfonate. The protein levels of pro-inflammatory cytokines in bronchoalveolar fluid (BALF) and serum were reduced by andrographolide sulfonate treatment. The levels of MUC5AC and MUC5B in lung tissue were also suppressed. These results reveal that andrographolide sulfate remarkably alleviated pneumonia induced by poly I:C in mice. Moreover, andrographolide sulfonate markedly inhibited the activation of nuclear factor-κB (NF-κB). Taken together, we demonstrated that andrographolide sulfonate ameliorated poly I: C-induced pneumonia in mice, suggesting the possible use of andrographolide sulfonate for virus-induced pneumonia in clinical.


Subject(s)
Diterpenes/administration & dosage , Diterpenes/pharmacology , NF-kappa B/metabolism , Phytotherapy , Pneumonia/drug therapy , Pneumonia/metabolism , Poly I-C/adverse effects , Animals , Cytokines/blood , Cytokines/metabolism , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Male , Mice, Inbred C57BL , Mucin 5AC/metabolism , Mucin-5B/metabolism , Pneumonia/chemically induced , Pneumonia/pathology , Pneumonia, Viral/drug therapy
9.
Brain Behav Immun ; 80: 525-535, 2019 08.
Article in English | MEDLINE | ID: mdl-31029796

ABSTRACT

Evidence is accumulating to suggest that viral infections and consequent viral-mediated neuroinflammation may contribute to the etiology of idiopathic Parkinson's disease. Moreover, viruses have been shown to influence α-synuclein oligomerization as well as the autophagic clearance of abnormal intra-cellular proteins aggregations, both of which are key neuropathological events in Parkinson's disease pathogenesis. To further investigate the interaction between viral-mediated neuroinflammation and α-synuclein aggregation in the context of Parkinson's disease, this study sought to determine the impact of viral neuroinflammatory priming on α-synuclein aggregate-induced neuroinflammation and neurotoxicity in the rat nigrostriatal pathway. To do so, male Sprague-Dawley rats were intra-nigrally injected with a synthetic mimetic of viral dsRNA (poly I:C) followed two weeks later by a peptidomimetic small molecule which accelerates α-synuclein fibril formation (FN075). The impact of the viral priming on α-synuclein aggregation-induced neuroinflammation, neurodegeneration and motor dysfunction was assessed. We found that prior administration of the viral mimetic poly I:C significantly exacerbated or precipitated the α-synuclein aggregate induced neuropathological and behavioral effects. Specifically, sequential exposure to the two challenges caused a significant increase in nigral microgliosis (p < 0.001) and astrocytosis (p < 0.01); precipitated a significant degeneration of the nigrostriatal cell bodies (p < 0.05); and precipitated a significant impairment in forelimb kinesis (p < 0.01) and sensorimotor integration (p < 0.01). The enhanced sensitivity of the nigrostriatal neurons to pathological α-synuclein aggregation after viral neuroinflammatory priming further suggests that viral infections may contribute to the etiology and pathogenesis of Parkinson's disease.


Subject(s)
Parkinson Disease/etiology , Poly I-C/adverse effects , alpha-Synuclein/metabolism , Animals , Biomimetic Materials , Corpus Striatum/metabolism , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Gliosis/metabolism , Male , Motor Activity/drug effects , Neurodegenerative Diseases/etiology , Neurodegenerative Diseases/physiopathology , Neuroimmunomodulation/physiology , Neurons/metabolism , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Poly I-C/administration & dosage , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/virology , Rats , Rats, Sprague-Dawley , Substantia Nigra/metabolism , Tyrosine 3-Monooxygenase/metabolism , alpha-Synuclein/physiology
10.
Behav Brain Funct ; 15(1): 3, 2019 Mar 05.
Article in English | MEDLINE | ID: mdl-30836963

ABSTRACT

BACKGROUND: Maternal immune activation (MIA) during gestation can increase the later risk of schizophrenia in adult offspring. Neuroinflammation is believed to underlie this process. Postmortem brain studies have found changes in the neuroimmune systems of patients with schizophrenia. However, little is known about the dynamic changes in cerebral inflammation and behavior during the course of the disease. METHODS: Here, the prepulse inhibition (PPI) test was conducted in adolescent and adult Sprague-Dawley rats prenatally challenged with polyriboinosinic-polyribocytidylic acid (Poly I:C) on gestational day 9 to determine the behavioral trajectory triggered by early exposure to Poly I:C. Brain immune changes were determined in the prefrontal cortex (PFC) and hippocampus (HC) at both ages. The status of the microglia and astrocytes was determined with immunohistochemical staining. The levels of IL-6, IL-1ß, and TNF-α in both brain regions were evaluated with enzyme-linked immunosorbent assays. RESULTS: Disrupted PPI, the core phenotype of schizophrenia, only emerged in adulthood. Behavioral changes during puberty and adulthood were both accompanied by the activation of microglia (PFC and HC). Astrocytes were only activated at PN60. The levels of proinflammatory cytokines (IL-1ß, IL-6, and TNF-α) in the offspring of the Poly I:C-exposed mothers differed with brain region and time, with more cytokines elevated during periadolescence than during adulthood. CONCLUSIONS: Our findings indicate that immune activation emerged before symptom manifestation in the offspring of MIA rats. We conclude that early prenatal Poly I:C challenge can lead to age-related behavioral and neuroinflammatory changes. These data provide new insight into the neuroinflammatory and neuropathological mechanisms underlying the development of schizophrenia. They also suggest that periadolescence could be more important than adulthood in the prevention and treatment of schizophrenia.


Subject(s)
Poly I-C/adverse effects , Age Factors , Animals , Behavior, Animal/drug effects , Brain/drug effects , Brain/pathology , Disease Models, Animal , Female , Hippocampus/drug effects , Male , Microglia/drug effects , Neuroimmunomodulation/physiology , Poly I-C/pharmacology , Prefrontal Cortex/drug effects , Pregnancy , Prenatal Exposure Delayed Effects , Prepulse Inhibition/physiology , Rats , Rats, Sprague-Dawley
11.
Fish Shellfish Immunol ; 95: 411-421, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31586678

ABSTRACT

Glutaredoxins are oxidoreductases present in almost all living organisms. They belong to the thioredoxin superfamily and share the thioredoxin structure and catalytic motif. Glutaredoxin 2 has been identified as a mitochondrial protein in vertebrates. In this study, the sequence of Glutaredoxin 2 from Hippocampus abdominalis (HaGrx2) was analyzed by molecular, transcriptional, and functional assays. In-silico analysis revealed that HaGrx2 shows the highest homology with Hippocampus comes, while distinctly cluster with fish Grx2 orthologs. Tissue distribution analysis showed that HaGrx2 is ubiquitously expressed in all tissues tested, and the highest expression was observed in the brain and skin. Significant HaGrx2 transcript modulation was identified in blood and liver upon injecting bacterial and Pathogen Associated Molecular Patterns. The redox activity of HaGrx2 was revealed by Dehydroascorbic reduction and insulin disulfide reduction activity assays. Further, the deglutathionylation activity of 1 nM HaGrx2 was found to be equivalent to that of 0.84 nM HaGrx1. HaGrx2 exhibited antiapoptotic activity against H2O2-induced oxidative stress in FHM cells. Altogether, the results of this study suggest that HaGrx2 plays a role in redox homeostasis and innate immune responses in fish.


Subject(s)
Fish Diseases/immunology , Gene Expression Regulation/immunology , Glutaredoxins/genetics , Glutaredoxins/immunology , Immunity, Innate/genetics , Smegmamorpha/genetics , Smegmamorpha/immunology , Amino Acid Sequence , Animals , Base Sequence , Edwardsiella tarda/physiology , Enterobacteriaceae Infections/immunology , Enterobacteriaceae Infections/veterinary , Female , Fish Proteins/chemistry , Fish Proteins/genetics , Fish Proteins/immunology , Gene Expression Profiling/veterinary , Glutaredoxins/chemistry , Homeostasis , Lipopolysaccharides/adverse effects , Male , Oxidation-Reduction , Phylogeny , Poly I-C/adverse effects , Sequence Alignment/veterinary , Streptococcal Infections/immunology , Streptococcal Infections/veterinary , Streptococcus iniae/physiology
12.
An Acad Bras Cienc ; 90(2): 1659-1663, 2018.
Article in English | MEDLINE | ID: mdl-29898114

ABSTRACT

Clozapine is an antipsychotic that produces serious withdrawal effects in schizophrenic patients. Olfactory deficits are well known as part of negative symptoms, but it is not known whether antipsychotic use and/or withdrawal are implicated. Then, we tested clozapine withdrawal in association with two widely used schizophrenia models: Neonatal immune challenge by Polycitidilic-polyinosinic acid (polyI:C) and ketamine. PolyI:C (or saline) was injected subcutaneously in neonatal period, dose of 5 mg/kg from 2 to 6 Post Natal Days, and ketamine or saline at the dose 25mg/kg intraperitoneally (i.p.), daily for 7 days from 53 to 60 post natal day. Clozapine 10mg/kg (or saline) was administered i.p. from 46 to 60 post natal day. Olfactory discrimination test (sensorial and cognitive deficit) was performed at 61 post natal day, 24h after the last injections. The association of PolyI:C, ketamine and clozapine disrupted Olfactory Discrimination, equating time in familiar and non-familiar compartments. PolyI:C plus ketamine increased crossings between compartments. It was produced, for the first time, an olfactory deficit induced by clozapine withdrawal in Wistar rats subjected to schizophrenia animal models.


Subject(s)
Antipsychotic Agents/adverse effects , Clozapine/adverse effects , Olfaction Disorders/chemically induced , Schizophrenia/chemically induced , Substance Withdrawal Syndrome , Animals , Disease Models, Animal , Drug Therapy, Combination/methods , Ketamine/adverse effects , Male , Neuropsychological Tests , Olfaction Disorders/diagnosis , Poly I-C/adverse effects , Preliminary Data , Rats, Wistar
13.
Hepatology ; 59(4): 1555-63, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24677196

ABSTRACT

UNLABELLED: Cell types and mechanisms involved in type I interferon (IFN)-mediated anti-inflammatory effects are poorly understood. Upon injection of artificial double-stranded RNA (poly(I:C)), we observed severe liver damage in type I IFN-receptor (IFNAR) chain 1-deficient mice, but not in wild-type (WT) controls. Studying mice with conditional IFNAR ablations revealed that IFNAR triggering of myeloid cells is essential to protect mice from poly(I:C)-induced liver damage. Accordingly, in poly(I:C)-treated WT, but not IFNAR-deficient mice, monocytic myeloid-derived suppressor cells (MDSCs) were recruited to the liver. Comparing WT and IFNAR-deficient mice with animals deficient for the IFNAR on myeloid cells only revealed a direct IFNAR-dependent production of the anti-inflammatory cytokine interleukin-1 receptor antagonist (IL-1RA) that could be assigned to liver-infiltrating cells. Upon poly(I:C) treatment, IFNAR-deficient mice displayed both a severe lack of IL-1RA production and an increased production of proinflammatory IL-1ß, indicating a severely imbalanced cytokine milieu in the liver in absence of a functional type I IFN system. Depletion of IL-1ß or treatment with recombinant IL-1RA both rescued IFNAR-deficient mice from poly(I:C)-induced liver damage, directly linking the deregulated IL-1ß and IL-1RA production to liver pathology. CONCLUSION: Type I IFN signaling protects from severe liver damage by recruitment of monocytic MDSCs and maintaining a balance between IL-1ß and IL-1RA production.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Interferon Type I/metabolism , Interleukin 1 Receptor Antagonist Protein/metabolism , Myeloid Cells/metabolism , RNA, Double-Stranded/adverse effects , Animals , Cells, Cultured , Disease Models, Animal , Injections , Interleukin 1 Receptor Antagonist Protein/deficiency , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1beta/metabolism , Liver/metabolism , Liver/pathology , Mice , Mice, Knockout , Myeloid Cells/pathology , Phenotype , Poly I-C/administration & dosage , Poly I-C/adverse effects , Poly I-C/pharmacology , RNA, Double-Stranded/administration & dosage , RNA, Double-Stranded/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
14.
J Immunol ; 190(7): 3639-47, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23455498

ABSTRACT

Infection and inflammation can disturb immune tolerance at the maternal-fetal interface, resulting in adverse pregnancy outcomes. However, the underlying mechanisms for detrimental immune responses remain ill defined. In this study, we provide evidence for immune programming of fetal loss in response to polyinosinic:polycytidylic acid (polyI:C), a viral mimic and an inducer of inflammatory milieu. IL-10 and uterine NK (uNK) cells expressing the activating receptor NKG2D play a critical role in poly(I:C)-induced fetal demise. In wild type (WT) mice, poly(I:C) treatment induced expansion of NKG2D(+) uNK cells and expression of Rae-1 (an NKG2D ligand) on uterine macrophages and led to fetal resorption. In IL-10(-/-) mice, NKG2D(-) T cells instead became the source of fetal resorption during the same gestation period. Interestingly, both uterine NK and T cells produced TNF-α as the key cytotoxic factor contributing to fetal loss. Treatment of WT mice with poly(I:C) resulted in excessive trophoblast migration into the decidua and increased TUNEL-positive signal. IL-10(-/-) mice supplemented with recombinant IL-10 induced fetal loss through NKG2D(+) uNK cells, similar to the response in WT mice. Blockade of NKG2D in poly(I:C)-treated WT mice led to normal pregnancy outcome. Thus, we demonstrate that pregnancy-disrupting inflammatory events mimicked by poly(I:C) are regulated by IL-10 and depend on the effector function of uterine NKG2D(+) NK cells in WT mice and NKG2D(-) T cells in IL-10 null mice.


Subject(s)
Embryo Loss/chemically induced , Embryo Loss/genetics , Interleukin-10/genetics , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , Poly I-C/adverse effects , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Embryo Loss/prevention & control , Female , Fetal Resorption/chemically induced , Fetal Resorption/immunology , Interleukin-10/deficiency , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Pregnancy , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation , Uterus/drug effects , Uterus/immunology , Uterus/metabolism
15.
J Neurosci ; 33(18): 7654-66, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23637159

ABSTRACT

Schizophrenia is thought to result from interactions between susceptible genotypes and environmental risk factors. DISC1 is an important gene for schizophrenia and mood disorders based on both human and animal studies. In the present study we sought to investigate interactions between two distinct point mutations in the mouse Disc1 gene (L100P and Q31L) and maternal immune activation (MIA) during pregnancy with polyinosinic:polycytidylic acid (polyI:C). PolyI:C given at 5 mg/kg impaired cognitive and social behavior in both wild-type (WT) and Disc1-Q31L(+/-) offspring, and reduced prepulse inhibition at 16 but not 8 weeks of age. Disc1-L100P(+/-) mutants were more sensitive to MIA than WT or Disc1-Q31L(+/-) mice. Interleukin-6 (IL-6) is a critical cytokine for mediating the behavioral and transcriptional effects of polyI:C. We found a more pronounced increase of IL-6 in response to polyI:C in fetal brain in Disc1-L100P(+/-) mice compared with WT or Disc1-Q31L(+/-) mice. Coadministration of an anti-IL-6 antibody with polyI:C reversed schizophrenia-related behavioral phenotypes in Disc1-L100P(+/-) mice. In summary, we found specific interactions between discrete genetic (Disc1-L100P(+/-)) and environmental factors (MIA) that exacerbate schizophrenia-related phenotypes. IL-6 may be important in the pathophysiology of this interaction.


Subject(s)
Nerve Tissue Proteins/genetics , Point Mutation/genetics , Prenatal Exposure Delayed Effects/immunology , Prenatal Exposure Delayed Effects/physiopathology , Schizophrenia/genetics , Schizophrenia/immunology , Acoustic Stimulation , Analysis of Variance , Animals , Cytokines/metabolism , Disease Models, Animal , Female , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C/adverse effects , Pregnancy , Recognition, Psychology/physiology , Reflex, Startle/drug effects , Reflex, Startle/genetics , Schizophrenia/metabolism , Schizophrenia/physiopathology , Social Behavior
16.
Fish Shellfish Immunol ; 39(2): 483-91, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24945573

ABSTRACT

Two sRNA libraries with or without poly(I:C) infection of large yellow croaker Pseudosciaena crocea were constructed and sequenced using the high-throughput Illumina/Solexa deep sequencing technology. The high-throughput sequencing pipeline yielded 163,79,272 and 217,07,070 raw reads corresponding to 132,27,594 and 206,86,409 clean reads for the normal and infected libraries, respectively. Bioinfromatic analysis identified 534 miRNAs, of which, 158 miRNAs were known in miRBase 20.0 and the remaining 376 were not found homology to any known metazoan miRNAs, suggesting a possible species-specificity. We analyzed the significance of differently expressed miRNAs between two libraries using pairwise comparison. There was significant differential expression of 112 miRNAs (p < 0.001) between two libraries. Thereinto, a number of known miRNAs were identified immune-related. Real-time quantitative PCR experiments (RT-qPCR) were preformed for 6 miRNAs of the two samples, and agreement was found between the sequencing and RT-qPCR data. To our knowledge, this is the first comprehensive study of miRNAs in P. crocea and of expression analysis of P. crocea miRNAs in response to poly(I:C) infection, and many miRNAs were differentially regulated under normal and infection conditions. These findings deepened our understanding of the role of miRNAs in the intricate host's immune system, and should be useful to develop new control strategies for host immune defense against various foreign infection in P. crocea.


Subject(s)
Gene Expression Regulation/drug effects , MicroRNAs/immunology , Perciformes/genetics , Perciformes/immunology , Poly I-C/adverse effects , Animals , Base Pairing , Computational Biology , DNA Primers/genetics , Gene Expression Profiling , Gene Expression Regulation/immunology , High-Throughput Nucleotide Sequencing , MicroRNAs/genetics , Molecular Sequence Data , Poly I-C/immunology , Real-Time Polymerase Chain Reaction
17.
J Pediatr Hematol Oncol ; 36(6): 451-7, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24309609

ABSTRACT

Brain tumors are the most common solid tumor diagnosed in childhood that account for significant morbidity and mortality. New therapies are urgently needed; hence, we conducted the first ever prospective open-label phase II trials of the biological response modifier, poly-ICLC, in children with brain tumors. Poly-ICLC is a synthetic double-stranded RNA that has direct antiviral, antineoplastic, and immune adjuvant effects. A total of 47 children representing a variety of brain tumor histopathologic subtypes were treated with poly-ICLC. On the basis of the results of the initial phase II trial, an expanded prospective phase II trial in low-grade glioma (LGG) has been initiated. MRI was used to acquire volume-based measures of tumor response. No dose-limiting toxicities have been observed. In the initial study 3 of 12 subjects with progressive high-grade gliomas (HGGs) responded, and 2 of 4 children with progressive LGG experienced stable disease for 18 to 24 months. In the follow-up LGG phase II study, 2 of 5 LGG patients were stable over 18 months, with 1 stable for 6 months. Overall 5 of 10 LGG patients have responded. On the basis of low toxicity and the promising LGG response, poly-ICLC may be effective for childhood LGG, and the results justify biomarker studies for personalization of poly-ICLC as a single agent or adjuvant.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/therapy , Carboxymethylcellulose Sodium/analogs & derivatives , Glioma/therapy , Poly I-C/administration & dosage , Polylysine/analogs & derivatives , Adolescent , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Brain Neoplasms/pathology , Carboxymethylcellulose Sodium/administration & dosage , Carboxymethylcellulose Sodium/adverse effects , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Glioma/pathology , Humans , Infant , Magnetic Resonance Angiography , Male , Neoplasm Grading , Poly I-C/adverse effects , Polylysine/administration & dosage , Polylysine/adverse effects , Prospective Studies , Treatment Outcome , Young Adult
18.
Eur Respir J ; 41(5): 1147-56, 2013 May.
Article in English | MEDLINE | ID: mdl-22835616

ABSTRACT

Recently, statins have been shown to have anti-inflammatory effects on lung inflammatory diseases. However, the mechanisms of action of simvastatin in viral pneumonia have yet to be elucidated, although viral infection remains a considerable health threat. In this study, we hypothesised that simvastatin inhibits polyinosinic-polycytidylic acid (poly I:C)-induced airway inflammation, such as RANTES (regulated on activation, normal T-cell expressed and secreted) expression and inflammatory cell recruitment. In bronchial cells, the effect of simvastatin on poly I:C-induced RANTES expression and signal transducer and activator of transcription (STAT)3-mediated signal transduction was determined using an ELISA and short hairpin (sh)RNA system. In a poly I:C-induced pneumonia mouse model, immunological changes in the lungs after simvastatin inhalation, such as inflammatory cell recruitment and cytokine/chemokine release, were examined. In poly I:C-stimulated bronchial cells, RANTES secretion was increased by STAT3 activation, and simvastatin suppressed poly I:C-induced STAT3 activation, resulting in inhibition of RANTES expression. In BALB/c mice stimulated with inhaled poly I:C, RANTES expression and neutrophil infiltration into the airway were elevated. However, simvastatin treatment attenuated STAT3 activation, RANTES release and subsequent neutrophilia in the lungs. These findings suggest that simvastatin inhibits airway inflammation, but there are other mechanisms that need to be fully elucidated.


Subject(s)
Chemokine CCL5/metabolism , Neutrophils/drug effects , Pneumonia/chemically induced , Poly I-C/adverse effects , Simvastatin/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Bronchi/cytology , Cell Line, Tumor , Disease Models, Animal , Doxycycline/pharmacology , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/cytology , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Inflammation , Lung/drug effects , Mice , Mice, Inbred BALB C , RNA, Small Interfering/metabolism , STAT3 Transcription Factor/metabolism
19.
Brain Behav Immun ; 33: 33-45, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23643646

ABSTRACT

Epidemiological studies have associated infection during pregnancy with increased risk of neurodevelopmental disorders in children, which is modeled in rodents by stimulating the immune system of pregnant dams with microorganisms or their mimics, such as poly(I:C) or LPS. In two prenatal mouse models, we show that in utero exposure of the fetus to cytokines/inflammatory mediators elicited by maternal immune stimulation with poly(I:C) yields offspring that exhibit a proinflammatory phenotype due to alterations in developmental programming of their immune system. Changes in the innate and adaptive immune elements of these pro-inflammatory offspring result in more robust responses following exposure to immune stimuli than those observed in control offspring from PBS-injected pregnant dams. In the first model, offspring from poly(I:C)-injected immunologically naïve dams showed heightened cellular and cytokine responses 4 h after injection of zymosan, a TLR2 agonist. In the second model, using dams with immunological memory, poly(I:C) injection during pregnancy produced offspring that showed preferential differentiation toward Th17 cell development, earlier onset of clinical symptoms of EAE, and more severe neurological deficits following immunization with MOG35-55. Such "fetal programming" in offspring from poly(I:C)-injected dams not only persists into neonatal and adult life, but also can have profound consequences on health and disease.


Subject(s)
Immunomodulation/immunology , Immunophenotyping , Mothers , Animals , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/epidemiology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Illness Behavior/physiology , Injections, Intraperitoneal , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/administration & dosage , Peptide Fragments/administration & dosage , Poly I-C/administration & dosage , Poly I-C/adverse effects , Pregnancy , Th17 Cells/immunology , Th17 Cells/metabolism , Th17 Cells/pathology , Zymosan/administration & dosage
20.
Mol Psychiatry ; 17(1): 62-70, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21079609

ABSTRACT

Approximately 40-50% of individuals affected by tuberous sclerosis (TSC) develop autism spectrum disorders (ASDs). One possible explanation for this partial penetrance is an interaction between TSC gene mutations and other risk factors such as gestational immune activation. In this study, we report the interactive effects of these two ASD risk factors in a mouse model of TSC. Combined, but not single, exposure had adverse effects on intrauterine survival. Additionally, provisional results suggest that these factors synergize to disrupt social approach behavior in adult mice. Moreover, studies in human populations are consistent with an interaction between high seasonal flu activity in late gestation and TSC mutations in ASD. Taken together, our studies raise the possibility of a gene × environment interaction between heterozygous TSC gene mutations and gestational immune activation in the pathogenesis of TSC-related ASD.


Subject(s)
Child Development Disorders, Pervasive , Haploinsufficiency/genetics , Immunity, Active/physiology , Pregnancy Complications/physiopathology , Social Behavior , Tumor Suppressor Proteins/deficiency , Age Factors , Animals , Animals, Newborn , Behavior, Animal , Child Development Disorders, Pervasive/etiology , Child Development Disorders, Pervasive/genetics , Child Development Disorders, Pervasive/immunology , Disease Models, Animal , Embryo, Mammalian , Exploratory Behavior , Female , Humans , Immunity, Active/drug effects , Infant, Newborn , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Poly I-C/adverse effects , Pregnancy , Pregnancy Complications/chemically induced , Pregnancy Complications/immunology , Pregnancy Complications/mortality , Tuberous Sclerosis Complex 2 Protein
SELECTION OF CITATIONS
SEARCH DETAIL