ABSTRACT
Chronic intake of a high-fat diet increases saturated fatty acids in the brain causing the progression of neurodegenerative diseases. Palmitic acid is a free fatty acid abundant in the diet that at high concentrations may penetrate the blood-brain barrier and stimulate the production of pro-inflammatory cytokines, leading to inflammation in astrocytes. The use of the synthetic neurosteroid tibolone in protection against fatty acid toxicity is emerging, but its transcriptional effects on palmitic acid-induced lipotoxicity remain unclear. Herein, we performed a transcriptome profiling of normal human astrocytes to investigate the molecular mechanisms by which palmitic acid causes cellular damage to astrocytes, and whether tibolone could reverse its detrimental effects. Astrocytes undergo a profound transcriptional change at 2 mM palmitic acid, affecting the expression of 739 genes, 366 upregulated and 373 downregulated. However, tibolone at 10 nM does not entirely reverse palmitic acid effects. Additionally, the protein-protein interaction reveals two novel gene clustering modules. The first module involves astrocyte defense responses by upregulation of pathways associated with antiviral innate immunity, and the second is linked to lipid metabolism. Our data suggest that activation of viral response signaling pathways might be so far, the initial molecular mechanism of astrocytes in response to a lipotoxic insult by palmitic acid, triggered particularly upon increased expression levels of IFIT2, IRF1, and XAF1. Therefore, this novel approach using a global gene expression analysis may shed light on the pleiotropic effects of palmitic acid on astrocytes, and provide a basis for future studies addressed to elucidate these responses in neurodegenerative conditions, which is highly valuable for the design of therapeutic strategies.
Subject(s)
Interferon Type I , Palmitic Acid , Humans , Palmitic Acid/toxicity , Antiviral Agents/pharmacology , Astrocytes/metabolism , Interferon Type I/metabolism , Interferon Type I/pharmacology , Fatty Acids/metabolism , Cholesterol/metabolismABSTRACT
The Chikungunya virus (CHIKV) causes Chikungunya fever, a disease characterized by symptoms such as arthralgia/polyarthralgia. Currently, there are no antivirals approved against CHIKV, emphasizing the need to develop novel therapies. The imidazonaphthyridine compound (RO8191), an interferon-α (IFN-α) agonist, was reported as a potent inhibitor of HCV. Here RO8191 was investigated for its potential to inhibit CHIKV replication in vitro. RO8191 inhibited CHIKV infection in BHK-21 and Vero-E6 cells with a selectivity index (SI) of 12.3 and 37.3, respectively. Additionally, RO8191 was capable to protect cells against CHIKV infection, inhibit entry by virucidal activity, and strongly impair post-entry steps of viral replication. An effect of RO8191 on CHIKV replication was demonstrated in BHK-21 through type-1 IFN production mechanism and in Vero-E6 cells which has a defective type-1 IFN production, also suggesting a type-1 IFN independent mode of action. Molecular docking calculations demonstrated interactions of RO8191 with the CHIKV E proteins, corroborated by the ATR-FTIR assay, and with non-structural proteins, supported by the CHIKV-subgenomic replicon cells assay.
Subject(s)
Chikungunya Fever , Chikungunya virus , Interferon Type I , Animals , Chlorocebus aethiops , Humans , Chikungunya Fever/drug therapy , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Molecular Docking Simulation , Virus Replication , Vero Cells , Interferon Type I/pharmacologyABSTRACT
Previously, we reported that the presence of multiple day 7 (D7) bovine embryos in the uterus induces systemic immune responses in circulating polymorphonuclear neutrophils (PMNs), but with unknown mechanism. Thus, this study aimed to investigate the direct impact of D7 bovine embryo on PMNs' immune responses in vitro and whether these PMNs can amplify and transfer embryo signals further to another PMN population. PMNs were directly stimulated by embryo culture media (ECM) or interferon tau (IFNT) (10 ng/ml) followed by evaluating mRNA expression by real-time PCR and phenotypic analysis by flow cytometry. To test whether PMNs can transfer embryo signals to a new PMN population, PMNs triggered by ECM or IFNT, were thoroughly washed and diluted to remove any media components, and again were incubated in fresh culture media for 3 h, from which culture supernatants were collected and used as PMN conditioned media (CM) to stimulate a new PMN population. Similar to ECM, IFNT directly stimulated expressions of IFNs (IFNA, IFNG), interferon-stimulated genes (ISGs; OAS1, ISG15, MX1), STAT1, TGFB and IL8, and downregulated TNFA in PMNs. Flow cytometrical analyses demonstrated that IFNT stimulated expressions of pregnancy-related phenotypic markers, CD16 and arginase-1 (ARG1), in PMNs. Most importantly, PMN CM induced ISGs and STAT1 mRNA in fresh PMNs. Since IFNT directly upregulated IFNA expression in PMNs, the impact of IFNA on PMNs' immune responses was further tested. Stimulation of PMNs with IFNA, especially at a low level (1 pg/ml), induced IFNT-like immune responses comparable to those induced by PMN CM. Together, these findings indicated that D7 bovine embryos induce direct anti-inflammatory responses with upregulation of ISGs expressions in PMNs mainly via IFNT. Additionally, PMNs can amplify and transfer embryo signals to a new PMN population in a cell-to-cell communication mechanism possibly mediated in part by IFNA. Such a novel immunological crosstalk might contribute to embryo tolerance and pregnancy establishment in cattle.
Subject(s)
Embryo, Mammalian/immunology , Embryo, Mammalian/metabolism , Gene Expression Regulation , Interferon Type I/immunology , Neutrophils/immunology , Pregnancy Proteins/immunology , Pregnancy/genetics , Pregnancy/immunology , Animals , Arginase/genetics , Cattle , Culture Media, Conditioned/pharmacology , Female , Gene Expression Regulation/drug effects , Immunity, Innate , In Vitro Techniques , Interferon Type I/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Phenotype , Pregnancy Proteins/pharmacology , Receptors, IgG/geneticsABSTRACT
Temozolomide (TMZ) is a chemotherapeutic used for the treatment of glioblastoma. The MGMT repair enzyme (O'-(6)-methyl guanine-DNA-methyltransferase) promoter methylation is a predictive biomarker to TMZ response; interferons (IFNs) type I can downregulate MGMT expression improving survival in patients with unmethylated MGMT promoter. HeberFERON is a co-formulation of IFNs type I and II with higher antiproliferative effect over glioblastoma cell lines than individual IFNs. We investigated the proliferative response of patient-derived glioblastoma cultures to HeberFERON and its combination with TMZ in relation to MGMT promoter methylation and the regulation of MGMT transcript after HeberFERON treatment. Eleven glioblastoma-derived cultures, molecularly classified according to TCGA and MGMT promoter methylation, were assayed for proliferation inhibition with HeberFERON at low doses (1-25 IU/mL) [alone or combined with TMZ] or at higher doses (50-200 IU/mL) using CellTiter-Glo Luminescent Cell Viability Assay (Promega). Eight cultures were further treated with 100 IU/mL of HeberFERON for 72 h, total RNA purified (Qiagen) and converted to cDNA (Superscript III kit, Invitrogen) as quantitative PCR templates. Changes of MGMT&P53 transcripts level were monitored. Response of cultures to HeberFERON is variable, dose-dependent and apparently independent from TCGA classification and MGMT methylation status, based on the eight Classical cultures data. When combining HeberFERON with TMZ there was an increase in cell death for cultures, 2/4 with methylated and 5/5 with unmethylated MGMT promoter. In two out five cultures with unmethylated MGMT status, we observed a decrease of MGMT gene levels and an increase in P53 encoding gene levels. HeberFERON and TMZ combination should be further assayed in glioblastoma, mainly for those with unmethylated MGMT promoter.
Subject(s)
Brain Neoplasms/genetics , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioblastoma/genetics , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Temozolomide/pharmacology , Tumor Suppressor Proteins/genetics , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Drug Synergism , Glioblastoma/metabolism , Humans , Promoter Regions, Genetic , Tumor Cells, CulturedABSTRACT
We aimed to evaluate the accuracy of Interferon-tau stimulated genes (ISG) abundance in peripheral blood polymorphonuclear cells (PMNs) on D20 after fixed-time artificial insemination (FTAI; D0) as a pregnancy diagnosis method against CL evaluation by Doppler ultrasonography and progesterone (P4) concentrations on D20, as well as Pregnancy Associated Glycoproteins (PAG) concentrations on D25. Additionally, we evaluated the potential of ISG abundance in PMNs as pregnancy loss predictors. Nelore heifers (n = 103) and cows (n = 144) underwent estrous synchronization and were artificially inseminated on D0. Pregnancy was diagnosed by B-mode ultrasonography on D30 and D70, and after the final diagnosis, females were classified in four groups: Pregnant; Non-pregnant; Functional CL on D20 but non-pregnant (CL-NP) and Pregnancy loss between D30 and D70 (PL). After determining cutoff values, the Sensitivity (SE), Specificity (SP), Positive Predictive Value (PPV), Negative Predictive Value (NPV) and Accuracy (ACC) were determined for each method. All methods were classified as significant (P < 0.05) predictors of pregnancy. Both ISG expression and PAG concentrations were greater (P < 0.05) in pregnant females than in non-pregnant and CL-NP females but did not differ (P > 0.05) from the PL group. ISG15 expression was greater (P < 0.05) in heifers than in cows, but this difference was not found in OAS1 expression and PAG concentrations. All the methods evaluated were proven to be adequate predictors of pregnancy, but greater accuracies were obtained through PAG concentrations and Doppler-US, due to the decreased number of false positive and false negative results.
Subject(s)
Cattle , Interferon Type I/pharmacology , Neutrophils/drug effects , Pregnancy Proteins/pharmacology , Pregnancy Tests/veterinary , Animals , Female , Gene Expression Regulation/drug effects , Neutrophils/metabolism , Parity , Pregnancy , Pregnancy Proteins/blood , Pregnancy Tests/methods , Progesterone/blood , Sensitivity and Specificity , Ultrasonography, DopplerABSTRACT
Interferons (IFNs) are cytokines with important immunomodulatory activity in vertebrates. Although type I IFNs and interleukins (IL) 29 and 28a (type III IFNs) bind to different cellular receptors and have distinct structures, most of their biological activities are redundant. Apeu virus (APEUV) is a member of the Bunyaviridae family isolated from the Brazilian rain forest. In this paper we evaluated the antiviral activity of type I and type III IFNs against APEUV. All tested IFNs were able to induce an antiviral state against the virus in a dose-dependent way. The activity of type III IFNs did not need the presence of type I IFNs. Mixing both types of IFNs did not improve the biological activity of each type alone. The tested IFNs were also able to protect human peripheral blood mononuclear cells from infection. IFN alpha2, IFN beta, IL-29 and IL-28a induced the expression of 2',5'-oligoadenylate synthetase (2'5'OAS) and 6-16 genes. Although MxA gene was related to antiviral activity against Bunyaviruses, there was no induction of MxA in our model. We were able to show activity of type I and type III IFNs against a RNA virus, and that this activity is not dependent on MxA gene.
Subject(s)
Antiviral Agents/immunology , Bunyaviridae Infections/drug therapy , Interferon Type I/immunology , Interleukins/immunology , Animals , Antiviral Agents/pharmacology , Bunyaviridae Infections/virology , Cells, Cultured , Chlorocebus aethiops , Dose-Response Relationship, Drug , Humans , Interferon Type I/pharmacology , Interferons , Interleukins/pharmacology , Leukocytes, Mononuclear/drug effects , Orthobunyavirus/drug effects , Orthobunyavirus/immunology , Vero CellsABSTRACT
Type I Interferon (IFN-alpha/beta) therapy has altered the natural course of multiple sclerosis. In this paper we evaluate the possible molecular mechanisms involved in the in vitro effects of IFN-alpha/beta on peripheral blood mononuclear cells from patients with clinically definite Relapsing-Remitting Multiple Sclerosis. The total RNA from IFN-alpha, IFN-beta treated cells and untreated cells was extracted and amplified for CD86, CD28, CTLA-4, TNF-alpha, IFN-gamma, CCL2, CCR5, IL-13, MMP-9, TIMP-1, CD25, TGF-beta, IL-10 and the transcriptional factor Foxp3 by Reverse Transcription-Polymerase Chain Reaction and the CD4+CD25high subset was evaluated using flow cytometry. In general, there were no significant differences concerning the modulation of the genes studied in the response to IFN-alpha and IFN-beta treatments, which suggest a similar mechanism of action for both interferons. However, we found a significant increment in IFN-gamma expression after IFN-alpha but not after IFN-beta treatments. The in vitro treatment of mononuclear cells from multiple sclerosis patients with both interferons significantly increased the CD25 mRNA. Furthermore, we observed a CD25/Foxp3 correlation and an increment of the CD4+CD25high subset, indicating that the induction of regulatory T cells could be a crucial mechanism involved in the type I interferon effects.
Subject(s)
Cytokines/metabolism , Interferon Type I/immunology , Leukocytes, Mononuclear/immunology , Multiple Sclerosis/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Antigen Presentation , Blood-Brain Barrier , Cells, Cultured , Cytokines/immunology , Gene Expression , Humans , Interferon Type I/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Multiple Sclerosis/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolismABSTRACT
Oropouche, Caraparu, Guama, Guaroa and Tacaiuma viruses (Orthobunyavirus genus) cause human febrile illnesses and/or encephalitis. To achieve a therapeutical agent to prevent and/or treat these diseases we evaluated the antiviral action of Interferon-alpha (IFN-alpha) on these orthobunyaviruses. In vitro results showed that all the studied orthobunyaviruses are susceptible to antiviral action of IFN-alpha, but this susceptibility is limited and dependent on both concentration of drug and treatment period. In vivo results demonstrated that IFN-alpha present antiviral action on Oropouche and Guaroa viruses when used as a prophylactic treatment. Moreover, a treatment initiated 3h after infection prevented the death of Guaroa virus infected-mice. Additionally, mortality of mice was related to the migration and replication of viruses in their brains. Our results suggest that IFN-alpha could be potentially useful in the prevention of diseases caused by Oropouche virus and in the prevention and/or treatment of diseases caused by Guaroa virus.
Subject(s)
Bunyaviridae Infections/drug therapy , Interferon-alpha/therapeutic use , Orthobunyavirus/drug effects , Animals , Animals, Suckling , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Body Weight/drug effects , Brain/drug effects , Brain/virology , Bunyaviridae Infections/mortality , Bunyaviridae Infections/prevention & control , Chlorocebus aethiops , Dose-Response Relationship, Drug , Interferon Type I/pharmacology , Interferon Type I/therapeutic use , Interferon alpha-2 , Interferon-alpha/pharmacology , Mice , Orthobunyavirus/growth & development , Recombinant Proteins , Survival Rate , Time Factors , Vero Cells , Virus Replication/drug effectsABSTRACT
Interferons (IFNs) are a family of cytokines that have many biological functions in the cell, including regulation of cellular growth, differentiation, immunomodulation, and viral replication by inducing a set of interferon stimulated genes (ISGs). Based on their structure and biological activities IFNs are subdivided into two groups: type I IFNs, which includes IFN-alpha and IFN-beta and type II IFNs, represented by IFN-gamma. The aim of this work was to investigate whether integrin alpha 11 (ITGA-11), a novel collagen-binding integrin, is responsive to type I IFN treatment. Our findings indicated that type I IFNs were able to induce the ITGA-11 mRNA levels in T98G cells. Increased levels of ITGA-11 protein were also observed in IFN-treated cells. The in vivo induction of ITGA-11 was detected in spleen and lungs of IFN-treated BALB/c mice. T98G cells infected with Murine encephalomyocarditis virus showed increased levels of ITGA-11 mRNA and protein. We observed that the ITGA-11 promoter has binding sites for transcriptional factors regulated by IFNs and the double-stranded RNA dependent protein kinase (PKR). Therefore we investigated the role of PKR in the induction of ITGA-11 by using a PKR deficient mouse embryo fibroblast cell line (MEFs). PKR(-/-) MEFs treated with IFN did not show increased levels of ITGA-11 protein nor mRNA although that could be promptly detected in wild type MEFs. Taken together our data suggest that ITGA-11 is a new interferon stimulated gene.
Subject(s)
Gene Expression Regulation/physiology , Integrin alpha Chains/genetics , Interferon Type I/pharmacology , Interferon-alpha/physiology , Interferon-beta/physiology , Animals , Cell Line, Transformed , Cell Line, Tumor , Humans , Integrin alpha Chains/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Knockout , Recombinant Proteins , eIF-2 Kinase/deficiency , eIF-2 Kinase/geneticsABSTRACT
We assessed the effect of recombinant IFN-alpha-2a (rIFN-alpha-2a) on the induction of CAs and sister-chromatid exchanges (SCEs) by the methylating compound streptozotocin (STZ), in Chinese Hamster Ovary (CHO) cells. The cytokine was added to cell cultures 30 min before STZ and left in the culture medium until the end of the treatment. A statistically significant increase in the frequency of CAs and SCEs was observed following treatment with STZ alone (p < 0.05) compared to control, whereas treatments with rIFN-alpha-2a alone did not produce any significant increase of CAs or SCEs over the control values (p < 0.05). Moreover, rIFN-alpha-2a had a marked inhibitory effect on the frequency of STZ-induced CAs--both chromosome- and chromatid-type--(p < 0.05) but was unable to prevent SCEs induced by the antibiotic (p > 0.05). A decrease in the replication index (RI) was observed in the combined treatments compared with STZ alone-treated cultures, indicating inhibition of DNA synthesis. It is suggested that rIFN-alpha-2a exerts its protective action against the induction of CAs by STZ by stimulating DNA repair.
Subject(s)
Antibiotics, Antineoplastic/toxicity , Antineoplastic Agents/pharmacology , Chromosome Aberrations/drug effects , Interferon Type I/pharmacology , Sister Chromatid Exchange/drug effects , Streptozocin/toxicity , Animals , Antineoplastic Combined Chemotherapy Protocols , Cricetinae , Cricetulus , Cyclophosphamide , DNA/biosynthesis , DNA Repair , Doxorubicin , Recombinant Proteins , VincristineABSTRACT
Both IFN-beta and TGF-beta have demonstrated their ability to antagonize several of the stimulatory activities of IFN-gamma on human macrophages, thereby classifying them as Th2-like. Aiming at a further characterization of their role in Th1/Th2 development, we studied their possible interaction with IL-12, the key Th1 cytokine. We found that IFN-beta by itself induced modest amounts of IFN-gamma, but was able to synergize with IL-12 for IFN-gamma induction. TGF-beta, on the other hand, had no effect by itself and inhibited significantly the IL-12-induced IFN-gamma secretion. The differential effect of IFN-beta and TGF-b on IL-12 bioactivity was most pronounced upon IFN-gamma synthesis, since IFN-beta induced only marginal amounts of IL-10 and IL-12 and TGF-beta diminished constitutive IL-10 production, while neither had a significant effect on TNF-alpha production. Although monocytes did not produce detectable IFN-gamma with any of the stimuli, adherent cells were found to cooperate with non-adherent lymphocytes for maximal IFN-gamma production. However, IL-18, a monocyte-derived IFN-gamma-inducing cytokine able to synergize with IL-12, was undetectable in IFN-beta or IFN-beta+IL-12-stimulated cells. In conclusion, the ability of IFN-beta to synergize with IL-12 for IFN-gamma synthesis, without significant concomitant IL-10 production, suggest a strong boost to Th1 development, which seems to be IL-18-independent.
Subject(s)
Interferon Type I/pharmacology , Interleukin-12/metabolism , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Transforming Growth Factor beta/pharmacology , Cell Adhesion , Drug Interactions , Humans , In Vitro Techniques , Interferon Type I/administration & dosage , Interferon-gamma/biosynthesis , Interleukin-10/biosynthesis , Interleukin-12/administration & dosage , Leukocytes, Mononuclear/cytology , Recombinant Proteins , Th1 Cells/drug effects , Th1 Cells/immunology , Transforming Growth Factor beta/administration & dosage , Tumor Necrosis Factor-alpha/biosynthesisABSTRACT
The gene coding for bovine interferon-omega1 (BoIFN-omega1) was recently cloned and expressed at high levels in the yeast Pichia pastoris. The recombinant BoIFN-omega1 protein shows antiviral activity in different cell lines and has an antiluteolytic effect in cyclic ewes. In this article, we describe a method for purification of BoIFN-omega1 expressed in the methylotrophic yeast P. pastoris and characterization of its activity in vivo. The recombinant protein secreted to the culture medium had low activity because of self-aggregation. BoIFN-omega1 was solubilized using urea and desalting and finally purified by ion exchange chromatography on Q-Sepharose Fast Flow. The yield of purified product was approximately 300 mg/L of fermentation culture, with a specific antiviral activity of 10(8) IU/mg. Its purity was at least 80%. The biologic characterization of purified BoIFN-omega1 was determined by induction of an antiviral state on ewes challenged with 100 lethal doses (LD) of Aujeszky virus and by the extension of the corpus luteum life span and interestrous interval in cyclic cows. Ewes treated with 2 x 106 IU/kg BoIFN-omega1 were protected from Aujeszky virus infection. In cows receiving an intrauterine infusion of 1 mg BoIFN-omega1, equally distributed between the two uterine horns, twice daily from day 14 to day 22 of the experimental estrous cycle, the lifespan of the corpus luteum (25 vs. 19 days) and the interestrous intervals (26 vs. 21 days) were extended when compared with a control group (p < 0.05). We show that recombinant BoIFN-omega1 purified from P. pastoris has high antiviral activity and is an effective antiluteolytic agent in cattle.
Subject(s)
Interferon Type I/pharmacology , Animals , Antiviral Agents/pharmacology , Cattle , Cell Line , Corpus Luteum Maintenance/drug effects , Estrus/drug effects , Female , In Vitro Techniques , Interferon Type I/genetics , Interferon Type I/isolation & purification , Male , Pichia/genetics , Pregnancy , Pseudorabies/prevention & control , Recombinant Proteins , SheepABSTRACT
We compared the alpha/beta interferon (IFN-alpha/beta) sensitivities of the TC-83 vaccine strain and 24 enzootic and epizootic Venezuelan equine encephalitis (VEE) isolates. The IFN-resistant or -sensitive phenotype correlated well with epizootic or enzootic potential. IFN-alpha/beta resistance of Trinidad donkey (TRD) virus correlated with virulence determinants in the 5' noncoding region and glycoproteins. Infection of mice lacking a functional IFN system with the IFN-sensitive TC-83 virus resulted in disease equivalent to that produced by the virulent, IFN-resistant TRD virus, further demonstrating that IFN resistance contributes to VEE virus virulence and is a biological marker of epizootic potential.
Subject(s)
Encephalitis Virus, Venezuelan Equine/pathogenicity , Encephalomyelitis, Venezuelan Equine/veterinary , Horse Diseases/virology , Interferon Type I/pharmacology , Animals , Cell Line , Cytopathogenic Effect, Viral , Drug Resistance, Microbial/genetics , Encephalitis Virus, Venezuelan Equine/genetics , Encephalitis Virus, Venezuelan Equine/isolation & purification , Encephalomyelitis, Venezuelan Equine/virology , Glycoproteins/genetics , Horses , Humans , Mice , Virulence/genetics , Zoonoses/virologyABSTRACT
The gene coding for bIFN-omega 1 was isolated from bovine genomic DNA by polymerase chain reaction (PCR). Recombinant bIFN-omega 1 was expressed in the yeast Pichia pastoris and high levels of the recombinant protein (0.4 mg ml-1) were secreted to the culture media. The obtained bIFN-omega 1 showed a cross-species antiviral activity on four mammalian cell lines of calf, pig, hamster and human origin, but this activity was absent on Madin-Darby canine kidney (MDCK) cells. A delivery carrier was developed to permit a better release of bIFN-omega 1. When compared with a control group, an increase in 6 days in the corpus luteum lifespan was obtained in cyclic ewes following three interferon (IFN) intrauterine administrations on days 9, 10 and 11 post-estrus. In summary, these results demonstrated for the first time that biologically active recombinant bIFN-omega 1 was highly secreted by P. pastoris showing antiviral activity in different cell lines and an antiluteolytic effect in cyclic ewes, with no detrimental effects on the animals.
Subject(s)
Interferon Type I/genetics , Pichia/genetics , Amino Acid Sequence , Animals , Antiviral Agents/pharmacology , Cattle , Cell Line , Corpus Luteum/physiology , Cricetinae , Dogs , Electrophoresis, Polyacrylamide Gel , Female , Gene Expression , Humans , Interferon Type I/metabolism , Interferon Type I/pharmacology , Molecular Sequence Data , Polymerase Chain Reaction , Progesterone/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Sheep , Swine , Uterus/metabolismABSTRACT
Changes in chromatin supraorganization defined in terms of patterns of chromatin texture were studied by video image analysis in Feulgen-stained revertants of LTR-ras-transformed NIH 3T3 cells and in cell lines obtained by transfection of these revertants with sense and antisense constructs of the lysyl oxidase gene (also named Lox or "ras recision gene"). The objective was to determine whether changes in expression of the Lox gene, which have been assumed to modulate cell transformation by ras, could also affect the chromatin supraorganization changes known to be elicited in NIH 3T3 cells by ras transformation. The image analysis results revealed that, although a nuclear phenotype visually similar to the most frequent one (III) in ras-transformed NIH 3T3 cells also appeared in the revertant, it contained a remarkably less tight chromatin packing state. This situation was also found in the revertant transfected with the sense construct of the Lox gene, but in the revertant transfected with the Lox antisense constructs the chromatin texture of the III phenotype was equal to or close to that of the ras-transformed cells. With regard to the nuclear phenotype characterized by abundant loosely packed chromatin and less represented in the transformed cell lines (I'), changes in the various cell lines, although detectable, were not as drastic as those reported for the III phenotype. The enhancement in chromatin condensation of the type III nuclei, which affects euchromatin, is probably associated with a limited transcription of the genome. Although the image analysis results are mostly in agreement with previously published data on the molecular biology and tumorigenicity of the same cell lines, it appears that the phenomenon of chromatin condensation once established in NIH 3T3 cells by LTR-ras transformation could not be totally reverted by simply affecting Lox expression.
Subject(s)
Cell Transformation, Neoplastic , Chromatin/physiology , Genes, ras , Rosaniline Dyes , 3T3 Cells , Animals , Cell Line , Cell Nucleus/ultrastructure , Chromatin/ultrastructure , Coloring Agents , Humans , Interferon Type I/pharmacology , Mice , Phenotype , Protein-Lysine 6-Oxidase/biosynthesis , Protein-Lysine 6-Oxidase/genetics , Repetitive Sequences, Nucleic Acid , TransfectionABSTRACT
Mice infected with the gastrointestinal nematode parasite Nippostrongylus brasiliensis (Nb) develop responses associated with enhanced production of IL-4 (increased serum IgE levels and intestinal mucosal mastocytosis) and IL-5 (tissue and peripheral blood eosinophilia). The antagonistic effects of IFN on IL-4-mediated responses prompted an examination of the effects of IFN on the host response to Nb. Treatment with rIFN-alpha and rIFN-gamma induced a marked increase in parasite egg production (fecundity) in BALB/c mice infected with Nb and delayed intestinal expulsion of adult worms. Treatment with rIFN-alpha or rIFN-gamma also inhibited the rise in peripheral blood eosinophilia that follows inoculation with Nb, and the intensity of pulmonary perivascular tissue eosinophilia. However, Nb-induced increases in serum IgG levels and intestinal mastocytosis were only temporarily delayed by IFN. Induction of endogenous IFN production by injection of fixed Brucella abortus into mice infected with Nb also resulted in an increased worm fecundity and delayed adult worm expulsion. These effects were ablated when mice given Brucella abortus also received injections of neutralizing anti-IFN antibodies. Thus, IFN inhibit host protective immunity to Nb, perhaps by interfering with the production and effects of Th2 cytokines.
Subject(s)
Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Nippostrongylus , Strongylida Infections/immunology , Animals , Brucella abortus/immunology , Eosinophilia/etiology , Eosinophilia/prevention & control , Female , Immunoglobulin E/blood , Interferon Inducers/pharmacology , Interleukin-4/biosynthesis , Interleukin-5/biosynthesis , Lung/immunology , Lung/pathology , Mastocytosis/immunology , Mice , Mice, Inbred BALB C , Nippostrongylus/immunology , Nippostrongylus/isolation & purification , Recombinant Proteins , Strongylida Infections/etiology , Strongylida Infections/parasitologyABSTRACT
El factor de cremiento epidérmico (EGF) es capaz de inhibir el crecimiento in vitro de líneas celulares tumorales humanas con una alta expresión de receptores (más de 1 x 105 sitios/células). El INF-gamma ( 1000 U/mL)potencia este efecto inhibidor, desplazando la concentración umbral de EGF capaz de producir inhibición del crecimiento, a concentraciones fisiológicas (10 ng/mL). Este efecto es específico, por cuanto el IFN-alfa no lo produce y no es mediado por una transmodulación del receptor de EGF, ya que el tratamiento de las células tumorales con IFN-gamma no cambia la afinidad ni el número de receptores de EGF. Estos resultados sugieren que la combinación EGF más IFN-gamma pudiera ser útil en el tratamiento de algunos tumores humanos (AU)
Subject(s)
In Vitro Techniques , Epidermal Growth Factor/pharmacology , Carcinoma, Squamous Cell , Vulvar Neoplasms , Cell Line , Interferon-gamma/pharmacology , Interferon Type I/pharmacologyABSTRACT
El factor de cremiento epidérmico (EGF) es capaz de inhibir el crecimiento in vitro de líneas celulares tumorales humanas con una alta expresión de receptores (más de 1 x 105 sitios/células). El INF-gamma ( 1000 U/mL)potencia este efecto inhibidor, desplazando la concentración umbral de EGF capaz de producir inhibición del crecimiento, a concentraciones fisiológicas (10 ng/mL). Este efecto es específico, por cuanto el IFN-alfa no lo produce y no es mediado por una transmodulación del receptor de EGF, ya que el tratamiento de las células tumorales con IFN-gamma no cambia la afinidad ni el número de receptores de EGF. Estos resultados sugieren que la combinación EGF más IFN-gamma pudiera ser útil en el tratamiento de algunos tumores humanos
Subject(s)
Carcinoma, Squamous Cell , Epidermal Growth Factor/pharmacology , In Vitro Techniques , Interferon Type I/pharmacology , Interferon-gamma/pharmacology , Cell Line , Vulvar NeoplasmsABSTRACT
Bovine interferon-alpha I1 (bIFN-alpha) may be useful for enhancing fertility in sheep and cattle because it has extensive sequence homology with ovine and bovine trophoblast protein-1 and, like those proteins, extends corpus luteum lifespan. To test the effectiveness of bIFN-alpha to enhance fertility, several experiments were performed in which inseminated heifers were given i.m. injections of bIFN-alpha approximately at the time of embryo-mediated signals that result in maintenance of the corpus luteum. In Exp. 1, heifers given 20 mg of bIFN-alpha daily from d 14 to 17 tended (P less than .07) to have lower pregnancy rates at d 110 to 112 of gestation (36/75; 48% vs 43/72; 60%). Similar results were obtained in Exp. 2 when heifers received a single injection of 40 mg of bIFN-alpha or placebo at d 13 after estrus; pregnancy rates at d 42 were 39/104 (38%) for bIFN-alpha and 47/98 (48%) for placebo. In Exp. 3, heifers were given gradually increasing doses of bIFN-alpha or placebo from d 11 to 19, because such a regimen had been shown to reduce the number of heifers experiencing hyperthermia after bIFN-alpha injection. Pregnancy rates were 42/95 (44%) for bIFN-alpha and 62/111 (56%) for placebo. Across all three experiments, pregnancy rates were lower (P less than .01) for heifers treated with bIFN-alpha (117/274; 43%) than for heifers treated with placebo (152/281; 54%). In conclusion, these results demonstrate that, under the administration systems used, bIFN-alpha does not increase pregnancy rate, but rather tends to reduce it.
Subject(s)
Cattle/physiology , Estrus/drug effects , Fertility/drug effects , Fertilization/drug effects , Interferon Type I/pharmacology , Animals , Body Temperature/drug effects , Double-Blind Method , Female , Pregnancy , Random Allocation , Recombinant ProteinsABSTRACT
Experiments were performed to determine the mechanism by which recombinant bovine interferon-alpha I1 (rbIFN-alpha) causes an acute reduction in plasma concentrations of progesterone. In experiment 1, administration of a prostaglandin synthesis inhibitor blocked rbIFN-alpha-induced hyperthermia but did not prevent the decline in plasma concentrations of progesterone. The decline in progesterone concentrations caused by rbIFN-alpha was, therefore, not a direct consequence of the associated hyperthermia or of pathways mediated through prostaglandin synthesis. It is also unlikely that rbIFN-alpha acts to increase the clearance of progesterone since injection of rbIFN-alpha did not decrease plasma concentrations of progesterone in ovariectomized cows given an intravaginal implant of progesterone (experiment 2). In experiment 3, rbIFN-alpha did not affect basal and LH-induced release of progesterone from cultured luteal slices, indicating that rbIFN-alpha is unlikely to affect luteal function directly. Injection of rbIFN-alpha did, however, cause a decrease in plasma concentrations of LH in ovariectomized cows (experiment 4) that coincided temporally with the decrease in progesterone concentrations seen in cows having a functional corpus luteum. The present results strongly suggest that rbIFN-alpha acts to reduce secretion of progesterone by interfering with pituitary support for luteal synthesis of progesterone. The finding that rbIFN-alpha can inhibit LH secretion implies that interferon-alpha molecules should be considered among the cytokines that can regulate hypothalamic or pituitary function.