Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Am J Physiol Renal Physiol ; 324(3): F267-F273, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36603001

ABSTRACT

The diverse functions of each nephron segment rely on the coordinated action of specialized cell populations that are uniquely defined by their transcriptional profile. In the collecting duct, there are two critical and distinct cell populations: principal cells and intercalated cells. Principal cells play key roles in the regulation of water, Na+, and K+, whereas intercalated cells are best known for their role in acid-base homeostasis. Currently, there are no in vitro systems that recapitulate the heterogeneity of the collecting ducts, which limits high-throughput and replicate investigations of genetic and physiological phenomena. Here, we demonstrated that the transcription factor Foxi1 is sufficient to alter the transcriptional identity of M-1 cells, a murine cortical collecting duct cell line. Specifically, overexpression of Foxi1 induces the expression of intercalated cell transcripts including Gpr116, Atp6v1b1, Atp6v1g3, Atp6v0d2, Slc4a9, and Slc26a4. These data indicate that overexpression of Foxi1 differentiates M-1 cells toward a non-A, non-B type intercalated cell phenotype and may provide a novel in vitro tool to study transcriptional regulation and physiological function of the renal collecting duct.NEW & NOTEWORTHY Transfection of M-1 cells with the transcription factor Foxi1 generates cells that express V-ATPase and Gpr116 as well as other genes associated with renal intercalated cells. This straightforward and novel in vitro system could be used to study processes including transcriptional regulation and cell specification and differentiation in renal intercalated cells.


Subject(s)
Forkhead Transcription Factors , Receptors, G-Protein-Coupled , Vacuolar Proton-Translocating ATPases , Animals , Mice , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cell Differentiation , Chloride-Bicarbonate Antiporters/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Kidney/metabolism , Receptors, G-Protein-Coupled/metabolism , Transcription Factors/metabolism , Vacuolar Proton-Translocating ATPases/metabolism
2.
Nephron ; 146(3): 278-281, 2022.
Article in English | MEDLINE | ID: mdl-34261071

ABSTRACT

G protein-coupled receptors (GPCRs) are cell surface proteins which play a key role in allowing cells, tissues, and organs to respond to changes in the external environment in order to maintain homeostasis. Despite the fact that GPCRs are known to play key roles in a variety of tissues, there are a large subset of GPCRs that remain poorly studied. In this minireview, we will summarize what is known regarding the "understudied" GPCRs with respect to renal function, and in so doing will highlight the promise represented by studying this gene family.


Subject(s)
Kidney , Receptors, G-Protein-Coupled , Homeostasis , Humans , Kidney/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
3.
J Pharmacol Exp Ther ; 377(1): 39-50, 2021 04.
Article in English | MEDLINE | ID: mdl-33414131

ABSTRACT

Short-chain fatty acids (SCFAs) are metabolites produced almost exclusively by the gut microbiota and are an essential mechanism by which gut microbes influence host physiology. Given that SCFAs induce vasodilation, we hypothesized that they might have additional cardiovascular effects. In this study, novel mechanisms of SCFA action were uncovered by examining the acute effects of SCFAs on cardiovascular physiology in vivo and ex vivo. Acute delivery of SCFAs in conscious radiotelemetry-implanted mice results in a simultaneous decrease in both mean arterial pressure and heart rate (HR). Inhibition of sympathetic tone by the selective ß-1 adrenergic receptor antagonist atenolol blocks the acute drop in HR seen with acetate administration, yet the decrease in mean arterial pressure persists. Treatment with tyramine, an indirect sympathomimetic, also blocks the acetate-induced acute drop in HR. Langendorff preparations show that acetate lowers HR only after long-term exposure and at a smaller magnitude than seen in vivo. Pressure-volume loops after acetate injection show a decrease in load-independent measures of cardiac contractility. Isolated trabecular muscle preparations also show a reduction in force generation upon SCFA treatment, though only at supraphysiological concentrations. These experiments demonstrate a direct cardiac component of the SCFA cardiovascular response. These data show that acetate affects blood pressure and cardiac function through parallel mechanisms and establish a role for SCFAs in modulating sympathetic tone and cardiac contractility, further advancing our understanding of the role of SCFAs in blood pressure regulation. SIGNIFICANCE STATEMENT: Acetate, a short-chain fatty acid, acutely lowers heart rate (HR) as well as mean arterial pressure in vivo in radiotelemetry-implanted mice. Acetate is acting in a sympatholytic manner on HR and exerts negative inotropic effects in vivo. This work has implications for potential short-chain fatty acid therapeutics as well as gut dysbiosis-related disease states.


Subject(s)
Acetates/pharmacology , Blood Pressure , Fatty Acids, Volatile/pharmacology , Heart Rate , Heart/drug effects , Myocardial Contraction , Acetates/administration & dosage , Animals , Fatty Acids, Volatile/administration & dosage , Female , Heart/physiology , Male , Mice , Mice, Inbred C57BL , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/physiology
4.
Proc Natl Acad Sci U S A ; 117(42): 26470-26481, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33004624

ABSTRACT

The diversity and near universal expression of G protein-coupled receptors (GPCR) reflects their involvement in most physiological processes. The GPCR superfamily is the largest in the human genome, and GPCRs are common pharmaceutical targets. Therefore, uncovering the function of understudied GPCRs provides a wealth of untapped therapeutic potential. We previously identified an adhesion-class GPCR, Gpr116, as one of the most abundant GPCRs in the kidney. Here, we show that Gpr116 is highly expressed in specialized acid-secreting A-intercalated cells (A-ICs) in the kidney using both imaging and functional studies, and we demonstrate in situ receptor activation using a synthetic agonist peptide unique to Gpr116. Kidney-specific knockout (KO) of Gpr116 caused a significant reduction in urine pH (i.e., acidification) accompanied by an increase in blood pH and a decrease in pCO2 compared to WT littermates. Additionally, immunogold electron microscopy shows a greater accumulation of V-ATPase proton pumps at the apical surface of A-ICs in KO mice compared to controls. Furthermore, pretreatment of split-open collecting ducts with the synthetic agonist peptide significantly inhibits proton flux in ICs. These data suggest a tonic inhibitory role for Gpr116 in the regulation of V-ATPase trafficking and urinary acidification. Thus, the absence of Gpr116 results in a primary excretion of acid in KO mouse urine, leading to mild metabolic alkalosis ("renal tubular alkalosis"). In conclusion, we have uncovered a significant role for Gpr116 in kidney physiology, which may further inform studies in other organ systems that express this GPCR, such as the lung, testes, and small intestine.


Subject(s)
Kidney/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Biochemical Phenomena , Biological Transport , Cell Movement/physiology , Epithelial Cells/metabolism , Female , Homeostasis , Humans , Kidney Tubules/metabolism , Male , Mice , Mice, Knockout
5.
Am J Physiol Cell Physiol ; 314(5): C627-C639, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29365273

ABSTRACT

The objective of this study was to determine the molecular identity of ion channels involved in K+ secretion by the mammary epithelium and to examine their regulation by purinoceptor agonists. Apical membrane voltage-clamp experiments were performed on human mammary epithelial cells where the basolateral membrane was exposed to the pore-forming antibiotic amphotericin B dissolved in a solution with intracellular-like ionic composition. Addition of the Na+ channel inhibitor benzamil reduced the basal current, consistent with inhibition of Na+ uptake across the apical membrane, whereas the KCa3.1 channel blocker TRAM-34 produced an increase in current resulting from inhibition of basal K+ efflux. Treatment with two-pore potassium (K2P) channel blockers quinidine, bupivacaine and a selective TASK1/TASK3 inhibitor (PK-THPP) all produced concentration-dependent inhibition of apical K+ efflux. qRT-PCR experiments detected mRNA expression for nine K2P channel subtypes. Western blot analysis of biotinylated apical membranes and confocal immunocytochemistry revealed that at least five K2P subtypes (TWIK1, TREK1, TREK2, TASK1, and TASK3) are expressed in the apical membrane. Apical UTP also increased the current, but pretreatment with the PKC inhibitor GF109203X blocked the response. Similarly, direct activation of PKC with phorbol 12-myristate 13-acetate produced a similar increase in current as observed with UTP. These results support the conclusion that the basal level of K+ secretion involves constitutive activity of apical KCa3.1 channels and multiple K2P channel subtypes. Apical UTP evoked a transient increase in KCa3.1 channel activity, but over time caused persistent inhibition of K2P channel function leading to an overall decrease in K+ secretion.


Subject(s)
Epithelial Cells/metabolism , Mammary Glands, Human/metabolism , Potassium Channels, Tandem Pore Domain/metabolism , Potassium/metabolism , Receptors, Purinergic P2Y/metabolism , Cell Line, Transformed , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Sodium Channels/metabolism , Female , Humans , Mammary Glands, Human/cytology , Mammary Glands, Human/drug effects , Membrane Potentials , Potassium Channel Blockers/pharmacology , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Potassium Channels, Tandem Pore Domain/genetics , Protein Kinase C/metabolism , Purinergic P2Y Receptor Agonists/pharmacology , Receptors, Purinergic P2Y/drug effects , Secretory Pathway , Sodium/metabolism , Uridine Triphosphate/pharmacology
6.
J Physiol ; 595(14): 4631-4645, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28481415

ABSTRACT

KEY POINTS: Hydrocortisone (HC) is required for activation of large-conductance Ca2+ -activated K+ current (BK) by purinergic receptor agonists. HC reduces insertion of the stress-regulated exon (STREX) in the KCNMA1 gene, permitting protein kinase C (PKC)-dependent channel activation. Overlapping and unique purinergic signalling regions exist at the apical border of differentiated surface cells. BK channels localize in the cilia of surface cells. ABSTRACT: In the present study we investigated the role of hydrocortisone (HC) on uridine-5'-triphosphate (UTP)-stimulated ion transport in differentiated, pseudostratified epithelia derived from normal human bronchial basal cells. The presence of a UTP-stimulated, paxilline-sensitive large-conductance Ca2+ -activated K+ (BK) current was demonstrated in control epithelia but was not stimulated in epithelia differentiated in the absence of HC (HC0). Addition of the BK channel opener NS11021 directly activated channels in control epithelia; however, under HC0 conditions, activation only occurred when UTP was added after NS11021. The PKC inhibitors GF109203x and Gö6983 blocked BK activation by UTP in control epithelia, suggesting that PKC-mediated phosphorylation plays a permissive role in purinoceptor-stimulated BK activation. Moreover, HC0 epithelia expressed significantly more KCNMA1 containing the stress-regulated exon (STREX), a splice-variant of the α-subunit that displays altered channel regulation by phosphorylation, compared to control epithelia. Furthermore, BK channels as well as purinergic receptors were shown to localize in unique and overlapping domains at the apical membrane of ciliated surface cells. These results establish a previously unrecognized role for glucocorticoids in regulation of BK channels in airway epithelial cells.


Subject(s)
Bronchi/physiology , Epithelial Cells/drug effects , Hydrocortisone/pharmacology , Large-Conductance Calcium-Activated Potassium Channels/physiology , Purinergic P2Y Receptor Agonists/pharmacology , Respiratory Mucosa/physiology , Adenosine Triphosphate/pharmacology , Cell Differentiation , Cell Line , Epithelial Cells/physiology , Humans , Indoles/pharmacology , Maleimides/pharmacology , Potassium Channel Blockers/pharmacology , Protein Kinase C/physiology , Protein Kinase Inhibitors/pharmacology , Receptors, Purinergic P2Y/physiology , Respiratory Mucosa/cytology , Uridine Triphosphate/pharmacology
7.
Am J Physiol Cell Physiol ; 313(1): C68-C79, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28446427

ABSTRACT

Aeroallergens produced by Alternaria alternata can elicit life-threatening exacerbations of asthma in patients sensitized to this fungus. In this study, the effect of Alternaria on ion transport mechanisms underlying mucociliary clearance and airway epithelial barrier function was investigated in human airway epithelial cells. Apical exposure to Alternaria induced an increase in anion secretion that was inhibited by blockers of CFTR and Ca2+-activated Cl- channels. Stimulation of anion secretion was dependent on Ca2+ uptake from the apical solution. Alternaria exposure also produced an increase in reactive oxygen species (ROS) that was blocked by pretreatment with the oxidant scavenger glutathione (GSH). GSH and the NADPH oxidase inhibitor/complex 1 electron transport inhibitor diphenylene iodonium chloride (DPI) blocked ATP release and the increase in intracellular [Ca2+] evoked by AlternariaAlternaria also decreased transepithelial resistance, and a portion of this effect was dependent on the increase in ROS. However, the Alternaria-induced increase in unidirectional dextran (molecular mass = 4,000 Da) flux across the epithelium could not be accounted for by increased oxidative stress. These results support the conclusion that oxidative stress induced by Alternaria was responsible for regulating Ca2+-dependent anion secretion and tight junction electrical resistance that would be expected to affect mucociliary clearance.


Subject(s)
Allergens/pharmacology , Alternaria/chemistry , Calcium/metabolism , Epithelial Cells/drug effects , Oxidative Stress/drug effects , Adenosine Triphosphate/metabolism , Alternaria/immunology , Bronchi , Cell Line, Transformed , Cell Polarity , Complex Mixtures/pharmacology , Dextrans/metabolism , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Epithelial Cells/immunology , Glutathione/pharmacology , Humans , Ion Transport/drug effects , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Onium Compounds/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism
8.
Mol Metab ; 6(1): 148-158, 2017 01.
Article in English | MEDLINE | ID: mdl-28123945

ABSTRACT

OBJECTIVES: Obesity is characterized by excessive fat mass and is associated with serious diseases such as type 2 diabetes. Targeting excess fat mass by sustained lipolysis has been a major challenge for anti-obesity therapies due to unwanted side effects. TLQP-21, a neuropeptide encoded by the pro-peptide VGF (non-acronymic), that binds the complement 3a receptor 1 (C3aR1) on the adipocyte membrane, is emerging as a novel modulator of adipocyte functions and a potential target for obesity-associated diseases. The molecular mechanism is still largely uncharacterized. METHODS: We used a combination of pharmacological and genetic gain and loss of function approaches. 3T3-L1 and mature murine adipocytes were used for in vitro experiments. Chronic in vivo experiments were conducted on diet-induced obese wild type, ß1, ß2, ß3-adrenergic receptor (AR) deficient and C3aR1 knockout mice. Acute in vivo lipolysis experiments were conducted on Sprague Dawley rats. RESULTS: We demonstrated that TLQP-21 does not possess lipolytic properties per se. Rather, it enhances ß-AR activation-induced lipolysis by a mechanism requiring Ca2+ mobilization and ERK activation of Hormone Sensitive Lipase (HSL). TLQP-21 acutely potentiated isoproterenol-induced lipolysis in vivo. Finally, chronic peripheral TLQP-21 treatment decreases body weight and fat mass in diet induced obese mice by a mechanism involving ß-adrenergic and C3a receptor activation without associated adverse metabolic effects. CONCLUSIONS: In conclusion, our data identify an alternative pathway modulating lipolysis that could be targeted to diminish fat mass in obesity without the side effects typically observed when using potent pro-lipolytic molecules.


Subject(s)
Adipocytes/drug effects , Peptide Fragments/metabolism , Receptors, Complement/drug effects , 3T3-L1 Cells , Adipocytes/metabolism , Adrenergic Agents/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Extracellular Signal-Regulated MAP Kinases , Lipolysis/drug effects , Lipolysis/physiology , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Obese , Mitogen-Activated Protein Kinase Kinases , Neuropeptides/metabolism , Obesity/chemically induced , Obesity/metabolism , Peptide Fragments/physiology , Rats , Rats, Sprague-Dawley , Receptors, Complement/metabolism , Signal Transduction , Sterol Esterase/adverse effects
9.
Am J Physiol Cell Physiol ; 311(2): C225-36, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27306366

ABSTRACT

Glucocorticoids strongly influence the mucosal-defense functions performed by the bronchial epithelium, and inhaled corticosteroids are critical in the treatment of patients with inflammatory airway diseases such as asthma, chronic obstructive pulmonary disease, and cystic fibrosis. A common pathology associated with these diseases is reduced mucociliary clearance, a defense mechanism involving the coordinated transport of salt, water, and mucus by the bronchial epithelium, ultimately leading to retention of pathogens and particles in the airways and to further disease progression. In the present study we investigated the role of hydrocortisone (HC) in differentiation and development of the ion transport phenotype of normal human bronchial epithelial cells under air-liquid interface conditions. Normal human bronchial epithelial cells differentiated in the absence of HC (HC0) showed significantly less benzamil-sensitive short-circuit current than controls, as well as a reduced response after stimulation with the selective ß2-adrenergic receptor agonist salbutamol. Apical membrane localization of epithelial Na(+) channel α-subunits was similarly reduced in HC0 cells compared with controls, supporting a role of HC in the trafficking and density of Na(+) channels in the plasma membrane. Additionally, glucocorticoid exposure during differentiation regulated the transcription of cystic fibrosis transmembrane conductance regulator and ß2-adrenergic receptor mRNAs and appeared to be necessary for the expression of cystic fibrosis transmembrane conductance regulator-dependent anion secretion in response to ß2-agonists. HC had no significant effect on surface cell differentiation but did modulate the expression of mucin mRNAs. These findings indicate that glucocorticoids support mucosal defense by regulating critical transport pathways essential for effective mucociliary clearance.


Subject(s)
Bronchi/physiology , Cell Differentiation/physiology , Epithelial Cells/physiology , Hydrocortisone/metabolism , Ion Transport/physiology , Mucociliary Clearance/physiology , Amiloride/analogs & derivatives , Amiloride/pharmacology , Bronchi/drug effects , Bronchi/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Cystic Fibrosis/metabolism , Cystic Fibrosis/physiopathology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Humans , Ion Transport/drug effects , Mucins/metabolism , Mucociliary Clearance/drug effects , Receptors, Adrenergic, beta-2/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/physiology , Sodium/metabolism
10.
Am J Physiol Lung Cell Mol Physiol ; 310(1): L50-8, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26566905

ABSTRACT

Carvedilol functions as a nonselective ß-adrenergic receptor (AR)/α1-AR antagonist that is used for treatment of hypertension and heart failure. Carvedilol has been shown to function as an inverse agonist, inhibiting G protein activation while stimulating ß-arrestin-dependent signaling and inducing receptor desensitization. In the present study, short-circuit current (Isc) measurements using human airway epithelial cells revealed that, unlike ß-AR agonists, which increase Isc, carvedilol decreases basal and 8-(4-chlorophenylthio)adenosine 3',5'-cyclic monophosphate-stimulated current. The decrease in Isc resulted from inhibition of the cystic fibrosis transmembrane conductance regulator (CFTR). The carvedilol effect was abolished by pretreatment with the ß2-AR antagonist ICI-118551, but not the ß1-AR antagonist atenolol or the α1-AR antagonist prazosin, indicating that its inhibitory effect on Isc was mediated through interactions with apical ß2-ARs. However, the carvedilol effect was blocked by pretreatment with the microtubule-disrupting compound nocodazole. Furthermore, immunocytochemistry experiments and measurements of apical CFTR expression by Western blot analysis of biotinylated membranes revealed a decrease in the level of CFTR protein in monolayers treated with carvedilol but no significant change in monolayers treated with epinephrine. These results demonstrate that carvedilol binding to apical ß2-ARs inhibited CFTR current and transepithelial anion secretion by a mechanism involving a decrease in channel expression in the apical membrane.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Carbazoles/pharmacology , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Epithelial Cells/drug effects , Propanolamines/pharmacology , Receptors, Adrenergic, beta-2/drug effects , Anions/metabolism , Arrestins/metabolism , Carvedilol , Cells, Cultured , Cyclic AMP/metabolism , Epithelial Cells/metabolism , Humans , Signal Transduction , beta-Arrestins
11.
Am J Physiol Cell Physiol ; 309(12): C847-55, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26491049

ABSTRACT

Human airway epithelial cells express ß-adrenergic receptors (ß-ARs), which regulate mucociliary clearance by stimulating transepithelial anion transport and ciliary beat frequency. Previous studies using airway epithelial cells showed that stimulation with isoproterenol increased cell migration and wound repair by a cAMP-dependent mechanism. In the present study, impedance-sensing arrays were used to measure cell migration and epithelial restitution following wounding of confluent normal human bronchial epithelial (NHBE) and Calu-3 cells by electroporation. Stimulation with epinephrine or the ß2-AR-selective agonist salbutamol significantly delayed wound closure and reduced the mean surface area of lamellipodia protruding into the wound. Treatment with the ß-AR bias agonist carvedilol or isoetharine also produced a delay in epithelial restitution similar in magnitude to epinephrine and salbutamol. Measurements of extracellular signal-regulated kinase phosphorylation following salbutamol or carvedilol stimulation showed no significant change in the level of phosphorylation compared with untreated control cells. However, inhibition of protein phosphatase 2A activity completely blocked the delay in wound closure produced by ß-AR agonists. In Calu-3 cells, where CFTR expression was inhibited by RNAi, salbutamol did not inhibit wound repair, suggesting that ß-AR agonist stimulation and loss of CFTR function share a common pathway leading to inhibition of epithelial repair. Confocal images of the basal membrane of Calu-3 cells labeled with anti-ß1-integrin (clone HUTS-4) antibody showed that treatment with epinephrine or carvedilol reduced the level of activated integrin in the membrane. These findings suggest that treatment with ß-AR agonists delays airway epithelial repair by a G protein- and cAMP-independent mechanism involving protein phosphatase 2A and a reduction in ß1-integrin activation in the basal membrane.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Cell Movement/physiology , Receptors, Adrenergic, beta/metabolism , Respiratory Mucosa/metabolism , Wound Healing/physiology , Cell Line , Cell Movement/drug effects , Electroporation , Enzyme-Linked Immunosorbent Assay , Humans , Immunohistochemistry , Microscopy, Confocal , Wound Healing/drug effects
12.
PLoS One ; 10(9): e0138704, 2015.
Article in English | MEDLINE | ID: mdl-26384331

ABSTRACT

Interferons (IFNs) have been shown to inhibit influenza A virus (IAV) replication and play an essential role in controlling viral infection. Here we studied the kinetics and magnitude of induction of type I and type III IFN transcripts by primary porcine airway epithelial cells (pAECs) in response to swine and human origin IAV. We observed that swine influenza viruses (SIV) replicate more efficiently than the human pandemic influenza A/California/2009 (pH1N1 CA/09) in pAECs. Interestingly, we also found significant difference in kinetics of IFN-ß, IFN-λ1 and IFN-λ3 gene expression by these viruses. While there was delay of up to 12 hours post infection (h p.i.) in induction of IFN genes in pAECs infected with swine IAV A/Sw/Illinois/2008 (H1N1 IL/08), human pH1N1 CA/09 rapidly induced IFN-ß, IFN-λ1 and IFN-λ3 gene expression as early as 4 h p.i. However, the magnitude of IFN-ß and IFN-λ3 induction at 24 h p.i. was not significantly different between the viral strains tested. Additionally, we found that swine H1N1 IL/08 was less sensitive to dsRNA induced antiviral response compared to human pH1N1 CA/09. Our data suggest that the human and swine IAVs differ in their ability to induce and respond to type I and type III interferons in swine cells. Swine origin IAV may have adapted to the pig host by subverting innate antiviral responses to viral infection.


Subject(s)
Bronchi/metabolism , Bronchi/virology , Influenza A Virus, H1N1 Subtype/physiology , Interferons/biosynthesis , Animals , Cells, Cultured , Dogs , Humans , Influenza A Virus, H1N1 Subtype/growth & development , Madin Darby Canine Kidney Cells , Swine
SELECTION OF CITATIONS
SEARCH DETAIL
...