Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Mol Biol Rep ; 50(5): 4261-4272, 2023 May.
Article in English | MEDLINE | ID: mdl-36918433

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) is the leading cause of mortality and morbidity in children under the age of five. Despite this, there is still a lack of safe and effective vaccines and antiviral agents for clinical use. Andrographolide exerts antiviral functions against a variety of viruses, but whether (and how) it exerts antiviral effects on RSV remains unclear. METHODS AND RESULTS: In vitro RSV infection models using A549 and 16HBE cell lines were established, and the effects of andrographolide on RSV were analyzed via RSV N gene load and proinflammatory cytokine levels. The RNA transcriptome was sequenced, and data were analyzed by R software. Andrographolide-related target genes were extracted via network pharmacology using online databases. Lentiviral transfection was applied to knockdown the heme oxygenase-1 gene (Hmox1, HO-1). Results showed that andrographolide suppressed RSV replication and attenuated subsequent inflammation. Network pharmacology and RNA sequencing analysis indicated that the hub gene HO-1 may play a pivotal role in the anti-RSV effects of andrographolide. Furthermore, andrographolide exerted antiviral effects against RSV partially by inducing HO-1 but did not activate the antiviral interferon response. CONCLUSION: Our findings demonstrated that andrographolide exerted anti-RSV activity by up-regulating HO-1 expression in human airway epithelial cells, providing novel insights into potential therapeutic targets and drug repurposing in RSV infection.


Subject(s)
Diterpenes , Heme Oxygenase-1 , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Child , Humans , Antiviral Agents/pharmacology , Epithelial Cells/metabolism , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/genetics , Interferons/drug effects , Interferons/metabolism , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/pathogenicity , Diterpenes/pharmacology , Diterpenes/therapeutic use
2.
Elife ; 102021 12 23.
Article in English | MEDLINE | ID: mdl-34939923

ABSTRACT

Hematopoietic stem cells (HSCs) must ensure adequate blood cell production following distinct external stressors. A comprehensive understanding of in vivo heterogeneity and specificity of HSC responses to external stimuli is currently lacking. We performed single-cell RNA sequencing (scRNA-Seq) on functionally validated mouse HSCs and LSK (Lin-, c-Kit+, Sca1+) progenitors after in vivo pharmacological perturbation of niche signals interferon, granulocyte colony-stimulating factor (G-CSF), and prostaglandin. We identified six HSC states that are characterized by enrichment but not exclusive expression of marker genes. External signals induced rapid transitions between HSC states but transcriptional response varied both between external stimulants and within the HSC population for a given perturbation. In contrast to LSK progenitors, HSCs were characterized by a greater link between molecular signatures at baseline and in response to external stressors. Chromatin analysis of unperturbed HSCs and LSKs by scATAC-Seq suggested some HSC-specific, cell intrinsic predispositions to niche signals. We compiled a comprehensive resource of HSC- and LSK progenitor-specific chromatin and transcriptional features that represent determinants of signal receptiveness and regenerative potential during stress hematopoiesis.


Most organs in the human body are maintained by a type of immature cells known as adult stem cells, which ensure a constant supply of new, mature cells. Adult stem cells monitor their environment through external signalling molecules and replace damaged cells as needed. Stem cell therapy takes advantage of the regenerative ability of immature stem cells and can be helpful for conditions such as blood diseases, autoimmune diseases, neurodegeneration and cancer. For example, hematopoietic stem-cell transplantation is a treatment for some types of cancer and blood disorders, in which stem cells are harvested from the blood or bone marrow and reintroduced into the body, where they can develop into all types of blood cells, including white blood cells, red blood cells and platelets. Hematopoietic stem-cell transplants have been in use for over 30 years, but they remain a highly risky procedure. One of the challenges is that outcomes can vary between patients and many of the factors that can influence the 'regenerative' potential of hematopoietic stem cells, such as external signalling molecules, are not well understood. To fill this gap, Fast et al. analysed which genes are turned on and off in hematopoietic stem cells in response to several external signalling molecules. To do so, three signalling pathways in mice were altered by injecting them with different chemicals. After two hours, the hematopoietic stem cells were purified and the gene expression for each cell was analysed. This revealed that the types of genes and the strength at which they were affected by each chemical was unique. Moreover, hematopoietic stem cells responded rapidly to external signals, with substantial differences in gene expression between individual groups of cells. Contrary to more specialised cells, the external signalling genes in some hematopoietic stem cells were already activated without being injected with external signalling molecules. This suggest that low levels of external signalling molecules released from their microenvironment may prepare stem cells to better respond to future stress or injuries. These results help to better understand stem cells and to evaluate how the signalling state of hematopoietic stem cells affects regeneration, and ultimately improve hematopoietic stem cell transplantation for patients.


Subject(s)
Gene Expression Regulation/physiology , Hematopoietic Stem Cells/metabolism , Transcriptome , Animals , Cell Lineage , Female , Granulocyte Colony-Stimulating Factor/drug effects , Hematopoietic Stem Cells/drug effects , Interferons/drug effects , Male , Mice , Multipotent Stem Cells/drug effects , Multipotent Stem Cells/metabolism , Prostaglandins/metabolism , Sequence Analysis, RNA , Signal Transduction
3.
Cells ; 10(10)2021 09 30.
Article in English | MEDLINE | ID: mdl-34685580

ABSTRACT

Host cell-intrinsic antiviral responses are largely mediated by pattern-recognition receptor (PRR) signaling and the interferon (IFN) system. The IFN regulatory factor (IRF) family of transcription factors takes up a central role in transcriptional regulation of antiviral innate immunity. IRF3 and IRF7 are known to be key players downstream of PRRs mediating the induction of type I and III IFNs. IFN signaling then requires IRF9 for the expression of the full array of interferon stimulated genes (ISGs) ultimately defining the antiviral state of the cell. Other members of the IRF family clearly play a role in mediating or modulating IFN responses, such as IRF1, IRF2 or IRF5, however their relative contribution to mounting a functional antiviral response is much less understood. In this study, we systematically and comparatively assessed the impact of six members of the IRF family on antiviral signaling in alveolar epithelial cells. We generated functional knockouts of IRF1, -2, -3, -5, -7, and -9 in A549 cells, and measured their impact on the expression of IFNs and further cytokines, ISGs and other IRFs, as well as on viral replication. Our results confirmed the vital importance of IRF3 and IRF9 in establishing an antiviral state, whereas IRF1, 5 and 7 were largely dispensable. The previously described inhibitory activity of IRF2 could not be observed in our experimental system.


Subject(s)
Alveolar Epithelial Cells/metabolism , Antiviral Agents/pharmacology , Interferon Regulatory Factor-7/drug effects , Interferon-Stimulated Gene Factor 3, gamma Subunit/drug effects , Humans , Immunity, Innate/drug effects , Immunity, Innate/physiology , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factors/metabolism , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Interferons/drug effects , Interferons/metabolism , Signal Transduction/drug effects , Virus Replication/drug effects , Virus Replication/physiology
4.
Clin Cancer Res ; 27(20): 5528-5535, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34433652

ABSTRACT

PURPOSE: Activation of STING (stimulator of interferon genes) can trigger a robust, innate antitumor immune response in immunologically "cold" tumors such as glioblastoma. PATIENTS AND METHODS: A small-molecule STING agonist, IACS-8779, was stereotactically administered using intraoperative navigation intratumorally in dogs with spontaneously arising glioblastoma. The phase I trial used an escalating dose design, ascending through four dose levels (5-20 µg). Treatment was repeated every 4-6 weeks for a minimum of two cycles. Radiographic response to treatment was determined by response assessment in neuro-oncology (RANO) criteria applied to isovoxel postcontrast T1-weighted MR images obtained on a single 3T magnet. RESULTS: Six dogs were enrolled and completed ≥1 cycle of treatment. One dog was determined to have an abscess and was removed from further analysis. One procedure-related fatality was observed. Radiographic responses were dose dependent after the first cycle. The first subject had progressive disease, whereas there was 25% volumetric reduction in one subject and greater than 50% in the remaining surviving subjects. The median progression-free survival time was 14 weeks (range: 0-22 weeks), and the median overall survival time was 32 weeks (range: 11-39 weeks). CONCLUSIONS: Intratumoral STING agonist (IACS-8779) administration was well tolerated in dogs with glioblastoma to a dose of 15 µg. Higher doses of IACS-8779 were associated with radiographic responses.


Subject(s)
Brain Neoplasms , Glioblastoma , Animals , Dogs , Female , Brain Neoplasms/drug therapy , Brain Neoplasms/veterinary , Glioblastoma/drug therapy , Glioblastoma/veterinary , Injections, Intralesional , Interferons/drug effects , Interferons/genetics , Treatment Outcome
5.
Anticancer Drugs ; 32(10): 1019-1028, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34261921

ABSTRACT

Ribosomal protein S14 (RPS14) is a component of the 40S ribosomal subunit and is considered to be indispensable for ribosomal biogenesis. Previously, we found that RPS14 was significantly downregulated in estrogen receptor-positive (ER+) breast cancer cells following treatment with 4-hydroxytamoxifen (4-OH-TAM). However, its role in breast cancer remains poorly understood. In the present study, we sought to demonstrate, for the first time, that RPS14 is highly expressed in ER+ breast cancer tissues and its downregulation can significantly inhibit the proliferation, cycle, and metastasis of ER+ breast cancer cells, as well as induce cell apoptosis. Quantitative RT-PCR and western blotting were used to determine the expression of target genes. Herein, lentivirus-mediated small hairpin RNA (shRNA) targeting RPS14 was designed to determine the impact of RPS14 knockdown on ER+ breast cancer cells. Further, bioinformatics analysis was used to reveal the significance of differentially expressed genes in RPS14 knockdown breast cancer cells. RPS14 was highly expressed in ER+ breast cancer tissues compared to ER- tissues. The downregulation of RPS14 in two ER+ breast cancer cell lines suppressed cell proliferation, cell cycle and metastasis, and induced apoptosis. Based on bioinformatics analysis, the expression level of several significant genes, such as ASNS, Ret, and S100A4, was altered in breast cancer cells after RPS14 downregulation. Furthermore, the BAG2 and interferon signaling pathways were identified to be significantly activated. The downregulation of RPS14 in ER+ breast cancer cells can inhibit their proliferation and metastasis.


Subject(s)
Breast Neoplasms/pathology , Down-Regulation/drug effects , RNA, Small Interfering/pharmacology , Receptors, Estrogen/biosynthesis , Ribosomal Proteins/drug effects , Apoptosis , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Interferons/drug effects , Neoplasm Metastasis , Signal Transduction/drug effects
6.
Rheumatol Int ; 41(9): 1593-1600, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34165604

ABSTRACT

To re-analyse the clinical outcomes and interferon (IFN) activity data from the JOQUER trial, a phase III trial investigating hydroxychloroquine (HCQ) in patients with primary Sjögren's syndrome (pSS), after stratifying patients into putative pathobiological subgroups utilizing the Newcastle Sjögren's Stratification Tool (NSST) based on patient-reported symptoms of dryness, pain, fatigue, anxiety and depression. 107 patients were assigned to one of four subgroups using NSST at baseline-the high symptom burden (HSB), pain dominant with fatigue (PDF), dryness dominant with fatigue (DDF) and low symptom burden (LSB). Endpoints were re-analysed after stratification, testing for treatment differences within subgroups and adjusting for baseline differences using a repeated measures covariate model. The HSB subgroup (n = 32) showed a relative improvement in ESSPRI of 1.49 points (95% CI 0.54-2.43; p = 0.002) within 12 weeks in patients taking HCQ compared to placebo, with no further changes after 24 weeks. For the LSB subgroup (n = 14), the ESSPRI worsened in the placebo but not the HCQ arm after 12 weeks (mean difference 1.44, 95% CI 0.05-2.83, p = 0.042). Neither the HSB nor the LSB patients showed significant changes in IFN activity at 24 weeks. There were no significant differences in ESSPRI in the PDF (n = 39) and DDF (n = 22) patients taking HCQ. However, significant reductions in overall IFN score at 24 weeks were seen in both PDF (difference at 24 weeks; 6.41, 95% CI, 2.48-10.34, p = 0.002) and DDF (difference at 24 weeks; 7.23, 95% CI, 1.85-12.6, p = 0.009) without improvement in ESSPRI. Although the JOQUER trial reported no overall benefit from HCQ in pSS patients, stratification suggests that both HSB and LSB subgroups may respond to HCQ. However, these patients may benefit through mechanisms other than the reduction of IFN activities.


Subject(s)
Antirheumatic Agents/therapeutic use , Hydroxychloroquine/therapeutic use , Sjogren's Syndrome/drug therapy , Antirheumatic Agents/pharmacology , Female , Follow-Up Studies , Humans , Hydroxychloroquine/pharmacology , Interferons/drug effects , Male
7.
BMC Complement Med Ther ; 21(1): 141, 2021 May 12.
Article in English | MEDLINE | ID: mdl-33980308

ABSTRACT

BACKGROUND: Herbal remedies of Echinacea purpurea tinctures are widely used today to reduce common cold respiratory tract infections. METHODS: Transcriptome, epigenome and kinome profiling allowed a systems biology level characterisation of genomewide immunomodulatory effects of a standardized Echinacea purpurea (L.) Moench extract in THP1 monocytes. RESULTS: Gene expression and DNA methylation analysis revealed that Echinaforce® treatment triggers antiviral innate immunity pathways, involving tonic IFN signaling, activation of pattern recognition receptors, chemotaxis and immunometabolism. Furthermore, phosphopeptide based kinome activity profiling and pharmacological inhibitor experiments with filgotinib confirm a key role for Janus Kinase (JAK)-1 dependent gene expression changes in innate immune signaling. Finally, Echinaforce® treatment induces DNA hypermethylation at intergenic CpG, long/short interspersed nuclear DNA repeat elements (LINE, SINE) or long termininal DNA repeats (LTR). This changes transcription of flanking endogenous retroviral sequences (HERVs), involved in an evolutionary conserved (epi) genomic protective response against viral infections. CONCLUSIONS: Altogether, our results suggest that Echinaforce® phytochemicals strengthen antiviral innate immunity through tonic IFN regulation of pattern recognition and chemokine gene expression and DNA repeat hypermethylated silencing of HERVs in monocytes. These results suggest that immunomodulation by Echinaforce® treatment holds promise to reduce symptoms and duration of infection episodes of common cold corona viruses (CoV), Severe Acute Respiratory Syndrome (SARS)-CoV, and new occurring strains such as SARS-CoV-2, with strongly impaired interferon (IFN) response and weak innate antiviral defense.


Subject(s)
COVID-19 Drug Treatment , Echinacea , Immunologic Factors/pharmacology , Monocytes/drug effects , Plant Extracts/pharmacology , SARS-CoV-2/drug effects , Gene Expression , Humans , Immunity, Innate/drug effects , Immunologic Factors/therapeutic use , Interferons/drug effects , Phytotherapy , Plant Extracts/therapeutic use
8.
Viruses ; 12(10)2020 09 27.
Article in English | MEDLINE | ID: mdl-32992478

ABSTRACT

Influenza A virus is a major global pathogen of humans, and there is an unmet need for effective antivirals. Current antivirals against influenza A virus directly target the virus and are vulnerable to mutational resistance. Harnessing an effective host antiviral response is an attractive alternative. We show that brief exposure to low, non-toxic doses of thapsigargin (TG), an inhibitor of the sarcoplasmic/endoplasmic reticulum (ER) Ca2+ ATPase pump, promptly elicits an extended antiviral state that dramatically blocks influenza A virus production. Crucially, oral administration of TG protected mice against lethal virus infection and reduced virus titres in the lungs of treated mice. TG-induced ER stress unfolded protein response appears as a key driver responsible for activating a spectrum of host antiviral defences that include an enhanced type I/III interferon response. Our findings suggest that TG is potentially a viable host-centric antiviral for the treatment of influenza A virus infection without the inherent problem of drug resistance.


Subject(s)
Antiviral Agents/pharmacology , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H3N8 Subtype/growth & development , Thapsigargin/pharmacology , Virus Replication/drug effects , Animals , Cell Line , Chick Embryo , Chlorocebus aethiops , Dogs , Endoplasmic Reticulum Stress/drug effects , Female , Host-Pathogen Interactions/drug effects , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Influenza, Human/drug therapy , Interferon Type I/drug effects , Interferon Type I/immunology , Interferons/drug effects , Interferons/immunology , Mice , Mice, Inbred BALB C , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Swine , Unfolded Protein Response/drug effects , Vero Cells , Interferon Lambda
9.
Eur Respir J ; 56(5)2020 Nov.
Article in English | MEDLINE | ID: mdl-32616594

ABSTRACT

While severe coronavirus infections, including Middle East respiratory syndrome coronavirus (MERS-CoV), cause lung injury with high mortality rates, protective treatment strategies are not approved for clinical use.We elucidated the molecular mechanisms by which the cyclophilin inhibitors cyclosporin A (CsA) and alisporivir (ALV) restrict MERS-CoV to validate their suitability as readily available therapy in MERS-CoV infection.Calu-3 cells and primary human alveolar epithelial cells (hAECs) were infected with MERS-CoV and treated with CsA or ALV or inhibitors targeting cyclophilin inhibitor-regulated molecules including calcineurin, nuclear factor of activated T-cells (NFATs) or mitogen-activated protein kinases. Novel CsA-induced pathways were identified by RNA sequencing and manipulated by gene knockdown or neutralising antibodies. Viral replication was quantified by quantitative real-time PCR and 50% tissue culture infective dose. Data were validated in a murine MERS-CoV infection model.Both CsA and ALV reduced MERS-CoV titres and viral RNA replication in Calu-3 cells and hAECs, improving epithelial integrity. While neither calcineurin nor NFAT inhibition reduced MERS-CoV propagation, blockade of c-Jun N-terminal kinase diminished infectious viral particle release but not RNA accumulation. Importantly, CsA induced interferon regulatory factor 1 (IRF1), a pronounced type III interferon (IFNλ) response and expression of antiviral genes. Downregulation of IRF1 or IFNλ increased MERS-CoV propagation in the presence of CsA. Importantly, oral application of CsA reduced MERS-CoV replication in vivo, correlating with elevated lung IFNλ levels and improved outcome.We provide evidence that cyclophilin inhibitors efficiently decrease MERS-CoV replication in vitro and in vivo via upregulation of inflammatory antiviral cell responses, in particular IFNλ. CsA might therefore represent a promising candidate for treating MERS-CoV infection.


Subject(s)
Coronavirus Infections/prevention & control , Cyclophilins/antagonists & inhibitors , Cyclosporine/pharmacology , Interferons/metabolism , Middle East Respiratory Syndrome Coronavirus/drug effects , Alveolar Epithelial Cells/drug effects , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/virology , Animals , Calcineurin Inhibitors/pharmacology , Cell Culture Techniques , Coronavirus Infections/metabolism , Disease Models, Animal , Humans , Interferon Regulatory Factor-1/drug effects , Interferon Regulatory Factor-1/metabolism , Interferons/drug effects , Mice , Middle East Respiratory Syndrome Coronavirus/physiology , Virus Replication/drug effects , Interferon Lambda
10.
Viruses ; 11(6)2019 06 01.
Article in English | MEDLINE | ID: mdl-31159430

ABSTRACT

Influenza virus is among the most common causes of respiratory illness worldwide and can be complicated by secondary bacterial pneumonia, a frequent cause of mortality. When influenza virus infects the lung, the innate immune response is activated, and interferons and inflammatory mediators are released. This "cytokine storm" is thought to play a role in influenza-induced lung pathogenesis. Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of the nuclear hormone receptor super-family. PPARγ has numerous functions including enhancing lipid and glucose metabolism and cellular differentiation and suppressing inflammation. Synthetic PPARγagonists (thiazolidinediones or glitazones) have been used clinically in the treatment of type II diabetes. Using data from the National Health and Nutrition Examination Survey (NHANES), diabetic participants taking rosiglitazone had an increased risk of mortality from influenza/pneumonia compared to those not taking the drug. We examined the effect of rosiglitazone treatment during influenza and secondary bacterial (Methicillin resistant Staphylococcus aureus) pneumonia in mice. We found decreased influenza viral burden, decreased numbers of neutrophils and macrophages in bronchoalveolar lavage, and decreased production of cytokines and chemokines in influenza infected, rosiglitazone-treated mice when compared to controls. However, rosiglitazone treatment compromised bacterial clearance during influenza-bacterial super-infection. Both human and mouse data suggest that rosiglitazone treatment worsens the outcome of influenza-associated pneumonia.


Subject(s)
Bacterial Infections , Coinfection/drug therapy , Inflammation/pathology , Influenza, Human , Rosiglitazone/adverse effects , Animals , Bacterial Infections/complications , Bacterial Infections/drug therapy , Bacterial Infections/immunology , Coinfection/immunology , Coinfection/microbiology , Coinfection/virology , Cytokines/drug effects , Cytokines/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/virology , Female , Gene Expression Regulation/drug effects , Humans , Inflammation/genetics , Inflammation/microbiology , Inflammation/virology , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/complications , Influenza, Human/drug therapy , Influenza, Human/immunology , Interferons/drug effects , Interferons/metabolism , Lung/immunology , Lung/microbiology , Lung/pathology , Lung/virology , Male , Methicillin-Resistant Staphylococcus aureus/drug effects , Methicillin-Resistant Staphylococcus aureus/growth & development , Methicillin-Resistant Staphylococcus aureus/immunology , Mice , PPAR gamma/agonists , Rosiglitazone/pharmacology , Signal Transduction/drug effects , Viral Load/drug effects
11.
Antiviral Res ; 166: 42-55, 2019 06.
Article in English | MEDLINE | ID: mdl-30928439

ABSTRACT

Liver injury is one of the hallmark features of severe dengue virus (DENV) infection since DENV can replicate in the liver and induce hepatocytes to undergo apoptosis. N-acetyl cysteine (NAC), which is a clinically-used drug for treating acetaminophen toxicity, was found to benefit patients with DENV-induced liver injury; however, its mechanism of action remains unclear. Accordingly, our aim was to repurpose NAC in the preclinical studies to investigate its mechanism of action. Time of addition experiments in HepG2 cells elucidated effectiveness of NAC to reduce infectious virion at pre-, during- and post infection. In DENV-infected mice, NAC improved DENV-associated clinical manifestations, including leucopenia and thrombocytopenia, and reduced liver injury and hepatocyte apoptosis. Interestingly, we discovered that NAC significantly reduced DENV production in HepG2 cells and in liver of DENV-infected mice by induction of antiviral responses via interferon signaling. NAC treatment in DENV-infected mice helped to maintain antioxidant enzymes and redox balance in the liver. Therefore, NAC reduces DENV production and oxidative damage to ameliorate DENV-induced liver injury. Taken together, these findings suggest the novel therapeutic potential of NAC in DENV-induced liver injury and recommend evaluating its efficacy and safety in humans with DENV-induced liver injury.


Subject(s)
Acetylcysteine/pharmacology , Dengue Virus/drug effects , Dengue/drug therapy , Animals , Antiviral Agents/pharmacology , Disease Models, Animal , Drug Repositioning , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/virology , Humans , Interferons/drug effects , Interferons/metabolism , Liver/drug effects , Liver/pathology , Liver/virology , Mice , Oxidative Stress/drug effects , Virus Replication/drug effects
12.
BMC Res Notes ; 11(1): 759, 2018 Oct 25.
Article in English | MEDLINE | ID: mdl-30359300

ABSTRACT

OBJECTIVE: The use of beneficial bacteria may stimulate wound healing. We performed a randomized, placebo-controlled double-blind cross-over study comprising ten healthy volunteers. The aim was to investigate the impact of topical and systemic applications of probiotic lactobacilli (Lactobacillus reuteri) on the healing of standardized wounds (punch biopsies) in the oral mucosa. The expression of selected matrix metalloproteinases (MMP'S) and interferons (IFN's) was analyzed with multiplex immunoassays in the wound exudate during the first healing week (day 2, 5 and 8). RESULTS: All participants completed the study and in all cases, the healing after the punch biopsies was uneventful. The concentrations of MMP-1, MMP-2, MMP-3 decreased with time in both the test- and control group. The MMP levels were consistently lower during the probiotic intervention when compared with placebo but the differences were not statistically significant. Likewise, the concentrations if IFN-alpha2, IFN-beta and IFN-gamma decreased with time with no significant differences between the test and placebo interventions. Within the limitations of this pilot study, we were unable to demonstrate an influence of probiotic supplements containing L. reuteri on the concentrations of selected matrix metalloproteinases and interferons from mucosal wounds within 1 week after a standardized punch biopsy. Trial registration ClinicalTrials.gov Identifier NCT03210779. Date of registration: July 7, 2017.


Subject(s)
Dietary Supplements , Interferons/drug effects , Limosilactobacillus reuteri , Matrix Metalloproteinases/drug effects , Mouth Mucosa/drug effects , Mouth Mucosa/injuries , Probiotics/pharmacology , Wound Healing/drug effects , Adult , Aged , Cross-Over Studies , Double-Blind Method , Female , Humans , Male , Middle Aged , Pilot Projects , Probiotics/administration & dosage , Treatment Failure , Young Adult
13.
Cancer Cell ; 31(6): 848-848.e1, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28609660

ABSTRACT

Immunotherapy has changed the landscape of cancer treatment. Checkpoint blockade therapies unleash breaks in the immune system and induce long-lasting responses. However, a significant number of patients do not respond (innate resistance), and a subset progress after responding (acquired resistance). A better understanding of the molecular mechanisms underlying checkpoint blockade therapies will facilitate the design of novel strategies to treat and prevent resistance. To view this SnapShot, open or download the PDF.


Subject(s)
Immunotherapy/methods , Neoplasms/drug therapy , Adaptive Immunity/drug effects , Cell Cycle Checkpoints/drug effects , Drug Resistance, Neoplasm/immunology , Humans , Interferons/drug effects , Interferons/physiology , Neoplasms/immunology
14.
Viruses ; 9(5)2017 05 12.
Article in English | MEDLINE | ID: mdl-28498306

ABSTRACT

The non-structural protein, NS1, is a virulence factor encoded by influenza A viruses (IAVs). In this report, we provide evidence that the conserved residue, tyrosine (Y) 84, in a conserved putative SH2-binding domain in A/Duck/Hubei/2004/L-1 [H5N1] NS1 is critical for limiting an interferon (IFN) response to infection. A phenylalanine (F) substitution of this Y84 residue abolishes NS1-mediated downregulation of IFN-inducible STAT phosphorylation, and surface IFNAR1 expression. Recombinant IAV (rIAV) [H1N1] expressing A/Grey Heron/Hong Kong/837/2004 [H5N1] NS1-Y84F (rWSN-GH-NS1-Y84F) replicates to lower titers in human lung epithelial cells and is more susceptible to the antiviral effects of IFN-ß treatment compared with rIAV expressing the intact H5N1 NS1 (rWSN-GH-NS1-wt). Cells infected with rWSN-GH-NS1-Y84F express higher levels of IFN stimulated genes (ISGs) associated with an antiviral response compared with cells infected with rWSN-GH-NS1-wt. In mice, intranasal infection with rWSN-GH-NS1-Y84F resulted in a delay in onset of weight loss, reduced lung pathology, lower lung viral titers and higher ISG expression, compared with mice infected with rWSN-GH-NS1-wt. IFN-ß treatment of mice infected with rWSN-GH-NS1-Y84F reduced lung viral titers and increased lung ISG expression, but did not alter viral titers and ISG expression in mice infected with rWSN-GH-NS1-wt. Viewed altogether, these data suggest that the virulence associated with this conserved Y84 residue in NS1 is, in part, due to its role in regulating the host IFN response.


Subject(s)
Influenza A Virus, H5N1 Subtype/metabolism , Influenza, Human/virology , Interferons/drug effects , Signal Transduction/drug effects , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Diseases/metabolism , A549 Cells , Animals , Antiviral Agents/pharmacology , Disease Models, Animal , Dogs , Epithelial Cells/virology , Fibroblasts , HEK293 Cells , HeLa Cells , Host-Pathogen Interactions/drug effects , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N1 Subtype/physiology , Influenza A virus/genetics , Influenza A virus/metabolism , Influenza A virus/physiology , Interferon-beta , Lung/pathology , Lung/virology , Madin Darby Canine Kidney Cells , Male , Mice , Mice, Inbred C57BL , Mutagenesis, Site-Directed , Neutrophils/pathology , Neutrophils/virology , Proto-Oncogene Proteins c-akt/metabolism , Reverse Genetics , Transfection , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/physiology , Virulence , Virulence Factors
15.
Antiviral Res ; 142: 185-192, 2017 06.
Article in English | MEDLINE | ID: mdl-28356234

ABSTRACT

Rhinoviruses (RVs) cause the common cold and are associated with exacerbations of chronic inflammatory respiratory diseases, especially asthma and chronic obstructive pulmonary disease (COPD). We have assessed the antiviral drugs Anaferon for Children (AC) and Ergoferon (containing AC as one of the active pharmaceutical ingredients) in in vitro and in vivo experimental models, in order to evaluate their anti-rhinoviral and immunomodulatory potential. HeLa cells were pretreated with AC, and levels of the interferon-stimulated gene (ISG), 2'-5'-oligoadenylate synthetase 1 (OAS1-A) and viral replication were analyzed. In a mouse model of RV-induced exacerbation of allergic airway inflammation we administered Ergoferon and analyzed its effect on type I (IFN-ß), type II (IFN-γ) and type III (IFN-λ) IFNs induction, cell counts in bronchoalveolar lavage (BAL), cytokine (interleukin (IL)-4; IL-6) and chemokine (CXCL10/IP-10; CXCL1/KC) levels. It was shown that AC increased OAS1-А production and significantly decreased viral replication in vitro. Increased IFNs expression together with reduced neutrophils/lymphocytes recruitment and correlated IL-4/IL-6 declination was demonstrated for Ergoferon in vivo. However, there was no effect on examined chemokines. We conclude that AC and Ergoferon possess effects against RV infection and may have potential as novel therapies against RV-induced exacerbations of asthma.


Subject(s)
Antibodies/pharmacology , Antiviral Agents/immunology , Antiviral Agents/pharmacology , Picornaviridae Infections/drug therapy , Rhinovirus/drug effects , 2',5'-Oligoadenylate Synthetase/analysis , Animals , Asthma/immunology , Asthma/virology , Cell Line , Chemokines/metabolism , Child , Cytokines/drug effects , Cytokines/metabolism , Disease Models, Animal , Gene Expression/drug effects , HeLa Cells , Humans , Inflammation , Interferons/drug effects , Mice , Mice, Inbred BALB C , Respiratory System/immunology , Respiratory System/virology , Rhinovirus/pathogenicity
16.
Poult Sci ; 94(10): 2382-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26240389

ABSTRACT

Real time quantitative polymerase chain reaction (RT-qPCR) based on SYBR-Green I binding is a quick, reliable, and easy method for analyzing small amounts of mRNA. Viral pathogens are recognized at the time of infection by pattern recognition receptors; thus, the inflammatory cytokines (IL1ß, IL6, and IL18) and antiviral cytokines (IFNα, IFNγ) are secreted by innate immune cells and induced to respond to the pathogens. The objective of this study was to develop an effective and sensitive RT-qPCR assay for the rapid and accurate quantification of goose cytokines: IFNα, IFNγ, IL1ß, IL6, and IL18. Subsequently, the established methods were employed to detect the immune response in agonist-stimulated goose spleen cells in vitro. These data indicated that the established RT-qPCR is a reliable method for determining relative gene expression. The results revealed that Imiquimod led to the significant upregulation of goose IFNα (P < 0.01), IFNγ (P < 0.01), IL1ß (P < 0.01), IL6 (P < 0.01), and IL18 (P < 0.05). The established methods are important for scientific research and clinical applications, which require rapid and accurate results in a short period of time. The technique can potentially be used in the further research of goose molecular immunology, which will help us understand the interactions between hosts and pathogens.


Subject(s)
Avian Proteins/genetics , Geese/genetics , Gene Expression , Interferons/genetics , Interleukins/genetics , Organic Chemicals , Real-Time Polymerase Chain Reaction/veterinary , Aminoquinolines/pharmacology , Animals , Avian Proteins/drug effects , Benzothiazoles , Diamines , Imiquimod , Interferon Inducers/pharmacology , Interferons/drug effects , Leukocytes, Mononuclear/drug effects , Quinolines , Spleen/drug effects
17.
J Psychosom Res ; 76(3): 227-32, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24529042

ABSTRACT

OBJECTIVE: The aim of the study was to research the relationship between interferon (IFN) induced depression and sociodemographic characteristics, neurotrophic factors and oxidative stress. METHODS: Sixty four cases, 34 with Chronic Hepatitis B (CHB) and 30 with Chronic Hepatitis C (CHC), were included in the study. The patients were assessed with Structured Clinical Interview for DSM-IV (SCID-I), Hamilton Anxiety Rating Scale (HARS) and Hamilton Depression Rating Scale (HDRS) at baseline on the 2nd and 6th weeks of treatment. S100 calcium binding protein B (S100B), brain-derived neurotrophic factor (BDNF), total antioxidant status (TAS) and total oxidative stress (TOS) levels were measured at the same visits. RESULTS: In total, 20 patients were diagnosed with major depression (MD) on the sixth week. A significant relationship was found between depression developed after IFN therapy and baseline HARS scores and the type of IFN-α. When the pretreatment levels of HDRS, HARS, S100B, BDNF, TAS, and TOS were compared to those after treatment on the 2nd week, there was a significant increase in HDRS and HARS levels and a significant decrease in the levels of S100B and BDNF. No significant change was determined for TAS and TOS levels. CONCLUSIONS: Our study suggests that the pathogenesis of IFN induced depression may involve neurotrophic factors.


Subject(s)
Antidepressive Agents/therapeutic use , Brain-Derived Neurotrophic Factor/physiology , Depression/drug therapy , Hepatitis B, Chronic/drug therapy , Hepatitis C, Chronic/drug therapy , Interferons/adverse effects , Oxidative Stress , S100 Calcium Binding Protein beta Subunit/physiology , Adolescent , Adult , Aged , Antidepressive Agents/adverse effects , Brain-Derived Neurotrophic Factor/blood , Depression/blood , Depression/chemically induced , Depressive Disorder, Major/blood , Depressive Disorder, Major/chemically induced , Depressive Disorder, Major/drug therapy , Female , Hepatitis B, Chronic/psychology , Hepatitis C, Chronic/psychology , Humans , Interferons/drug effects , Male , Middle Aged , Prospective Studies , S100 Calcium Binding Protein beta Subunit/blood , Young Adult
18.
Magy Onkol ; 57(2): 79-83, 2013 Jun.
Article in Hungarian | MEDLINE | ID: mdl-23795352

ABSTRACT

Malignant melanoma biologically can be divided into non-metastatic and metastatic forms which cannot be predicted precisely using classical clinicopathological parameters, therefore studies on novel genetic or protein markers are abundant in the literature. These studies did not result in clinically useful markers because mostly ignored the results of studies on the genetic basis of metastatic potential of malignant melanoma. Accordingly, the list of promising novel markers is short (BCL2, CDK2, MART-1, OPN). Similar to other solid malignancies, introduction of targeted therapy into clinical practice of melanoma turned the attention toward the genetic basis of resistance to chemo- and targeted therapies. These novel data could lead to the development of molecular diagnostics which can help in designing more effective therapeutic strategies of malignant melanoma.


Subject(s)
Biomarkers, Tumor/genetics , Drug Resistance, Neoplasm , Melanoma/diagnosis , Molecular Targeted Therapy , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-kit/genetics , Skin Neoplasms/diagnosis , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 2/genetics , Diagnosis, Differential , Drug Resistance, Neoplasm/genetics , Humans , Interferons/drug effects , Interferons/metabolism , Interleukin-2/metabolism , MART-1 Antigen/genetics , Melanoma/drug therapy , Melanoma/genetics , Molecular Targeted Therapy/trends , Osteopontin/genetics , Patient Selection , Peptide Fragments/genetics , Predictive Value of Tests , Prognosis , Proto-Oncogene Proteins c-bcl-2/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics
19.
Biomed Pharmacother ; 67(7): 665-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23582788

ABSTRACT

Native human interferon-α (nHuIFN-α) has a stronger reductive effect on procollagen type I mRNA expression than recombinant human interferon-α (rHuIFN-α). It is partially due to the additive activity of interleukin-1ß (IL-1ß), which is present in small concentrations in nHuIFN-α. Here, we show that the reductive effect is also the result of the endogenous cytokines induced by the activity of nHuIFN-α. In the culture of MRC5 fibroblasts, we have further found that nHuIFN-α induces endogenous interferons in higher amounts than rHuIFN-α, measured with PCR. That is more pronounced when interferon-γ (IFN-γ) is measured. This result puts a new light on IFN-γ activity in the nHuIFN-α treatment because its role was neglected due to the loss of its activity during the nHuIFN-α preparation process. The findings lead to the conclusion that endogenous cytokines play a significant role in the nHuIFN-α -mediated reduction of procollagen type I mRNA and are therefore an important factor in potential therapeutic usage.


Subject(s)
Collagen Type I/biosynthesis , Cytokines/metabolism , Interferon-alpha/pharmacology , Interferons/biosynthesis , Procollagen/biosynthesis , Cycloheximide/pharmacology , Cytokines/biosynthesis , Gene Expression/drug effects , Humans , Interferon-alpha/antagonists & inhibitors , Interferons/drug effects , Interleukin-1beta/metabolism , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...