Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 22(24)2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34948382

ABSTRACT

Prior studies show that glycogen synthase kinase 3ß (GSK3ß) contributes to cardiac ischemic injury and cardiac hypertrophy. GSK3ß is constitutionally active and phosphorylation of GSK3ß at serine 9 (S9) inactivates the kinase and promotes cellular growth. GSK3ß is also phosphorylated at serine 389 (S389), but the significance of this phosphorylation in the heart is not known. We analyzed GSK3ß S389 phosphorylation in diseased hearts and utilized overexpression of GSK3ß carrying ser→ala mutations at S9 (S9A) and S389 (S389A) to study the biological function of constitutively active GSK3ß in primary cardiomyocytes. We found that phosphorylation of GSK3ß at S389 was increased in left ventricular samples from patients with dilated cardiomyopathy and ischemic cardiomyopathy, and in hearts of mice subjected to thoracic aortic constriction. Overexpression of either GSK3ß S9A or S389A reduced the viability of cardiomyocytes subjected to hypoxia-reoxygenation. Overexpression of double GSK3ß mutant (S9A/S389A) further reduced cardiomyocyte viability. Determination of protein synthesis showed that overexpression of GSK3ß S389A or GSK3ß S9A/S389A increased both basal and agonist-induced cardiomyocyte growth. Mechanistically, GSK3ß S389A mutation was associated with activation of mTOR complex 1 signaling. In conclusion, our data suggest that phosphorylation of GSK3ß at S389 enhances cardiomyocyte survival and protects from cardiomyocyte hypertrophy.


Subject(s)
Cardiomegaly/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Myocardial Ischemia/metabolism , Myocytes, Cardiac/pathology , Animals , Cardiomegaly/pathology , Cell Proliferation , Cell Survival , Cells, Cultured , Humans , Male , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Myocardial Ischemia/pathology , Myocytes, Cardiac/metabolism , Phosphorylation , Rats, Sprague-Dawley
2.
Mol Ther Nucleic Acids ; 20: 589-605, 2020 Jun 05.
Article in English | MEDLINE | ID: mdl-32348937

ABSTRACT

Non-coding microRNAs (miRNAs) are powerful regulators of gene expression and critically involved in cardiovascular pathophysiology. The aim of the current study was to identify miRNAs regulating cardiac fibrosis. Cardiac samples of age-matched control subjects and sudden cardiac death (SCD) victims with primary myocardial fibrosis (PMF) were subjected to miRNA profiling. Old SCD victims with PMF and healthy aged human hearts showed increased expression of miR-1468-3p. In vitro studies in human cardiac fibroblasts showed that augmenting miR-1468-3p levels induces collagen deposition and cell metabolic activity and enhances collagen 1, connective tissue growth factor, and periostin expression. In addition, miR-1468-3p promotes cellular senescence with increased senescence-associated ß-galactosidase activity and increased expression of p53 and p16. AntimiR-1468-3p antagonized transforming growth factor ß1 (TGF-ß1)-induced collagen deposition and metabolic activity. Mechanistically, mimic-1468-3p enhanced p38 phosphorylation, while antimiR-1468-3p decreased TGF-ß1-induced p38 activation and abolished p38-induced collagen deposition. RNA sequencing analysis, a computational prediction model, and qPCR analysis identified dual-specificity phosphatases (DUSPs) as miR-1468-3p target genes, and regulation of DUSP1 by miR-1468-3p was confirmed with a dual-luciferase reporter assay. In conclusion, miR-1468-3p promotes cardiac fibrosis by enhancing TGF-ß1-p38 signaling. Targeting miR-1468-3p in the older population may be of therapeutic interest to reduce cardiac fibrosis.

3.
EBioMedicine ; 51: 102608, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31911272

ABSTRACT

BACKGROUND: Vascular endothelial zinc finger 1 (Vezf1) is a transcription factor previously shown to regulate vasculogenesis and angiogenesis. We aimed to investigate the role of Vezf1 in the postnatal heart. METHODS: The role of Vezf1 in regulating cardiac growth and contractile function was studied in zebrafish and in primary cardiomyocytes. FINDINGS: We find that expression of Vezf1 is decreased in diseased human myocardium and mouse hearts. Our experimental data shows that knockdown of zebrafish Vezf1 reduces cardiac growth and results in impaired ventricular contractile response to ß-adrenergic stimuli. However, Vezf1 knockdown is not associated with dysregulation of cardiomyocyte Ca2+ transient kinetics. Gene ontology enrichment analysis indicates that Vezf1 regulates cardiac muscle contraction and dilated cardiomyopathy related genes and we identify cardiomyocyte Myh7/ß-MHC as key target for Vezf1. We further identify a key role for an MCAT binding site in the Myh7 promoter regulating the response to Vezf1 knockdown and show that TEAD-1 is a binding partner of Vezf1. INTERPRETATION: We demonstrate a role for Vezf1 in regulation of compensatory cardiac growth and cardiomyocyte contractile function, which may be relevant in human cardiac disease.


Subject(s)
DNA-Binding Proteins/metabolism , Myocardial Contraction , Myocardium/metabolism , Myocardium/pathology , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Adrenergic Agents/pharmacology , Animals , Binding Sites , Cardiomyopathies/genetics , Gene Expression Regulation/drug effects , Genes, Reporter , Humans , Luciferases/metabolism , Mice, Inbred C57BL , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism , Neovascularization, Physiologic/drug effects , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Rats, Sprague-Dawley , Zebrafish
4.
JACC Basic Transl Sci ; 4(1): 83-94, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30847422

ABSTRACT

Myocardial infarction (MI)-induced cardiac fibrosis attenuates cardiac contractile function, and predisposes to arrhythmias and sudden cardiac death. Expression of connective tissue growth factor (CTGF) is elevated in affected organs in virtually every fibrotic disorder and in the diseased human myocardium. Mice were subjected to treatment with a CTGF monoclonal antibody (mAb) during infarct repair, post-MI left ventricular (LV) remodeling, or acute ischemia-reperfusion injury. CTGF mAb therapy during infarct repair improved survival and reduced LV dysfunction, and reduced post-MI LV hypertrophy and fibrosis. Mechanistically, CTGF mAb therapy induced expression of cardiac developmental and/or repair genes and attenuated expression of inflammatory and/or fibrotic genes.

5.
Mol Ther ; 27(3): 600-610, 2019 03 06.
Article in English | MEDLINE | ID: mdl-30765322

ABSTRACT

Activin A and myostatin, members of the transforming growth factor (TGF)-ß superfamily of secreted factors, are potent negative regulators of muscle growth, but their contribution to myocardial ischemia-reperfusion (IR) injury is not known. The aim of this study was to investigate if activin 2B (ACVR2B) receptor ligands contribute to myocardial IR injury. Mice were treated with soluble ACVR2B decoy receptor (ACVR2B-Fc) and subjected to myocardial ischemia followed by reperfusion for 6 or 24 h. Systemic blockade of ACVR2B ligands by ACVR2B-Fc was protective against cardiac IR injury, as evidenced by reduced infarcted area, apoptosis, and autophagy and better preserved LV systolic function following IR. ACVR2B-Fc modified cardiac metabolism, LV mitochondrial respiration, as well as cardiac phenotype toward physiological hypertrophy. Similar to its protective role in IR injury in vivo, ACVR2B-Fc antagonized SMAD2 signaling and cell death in cardiomyocytes that were subjected to hypoxic stress. ACVR2B ligand myostatin was found to exacerbate hypoxic stress. In addition to acute cardioprotection in ischemia, ACVR2B-Fc provided beneficial effects on cardiac function in prolonged cardiac stress in cardiotoxicity model. By blocking myostatin, ACVR2B-Fc potentially reduces cardiomyocyte death and modifies cardiomyocyte metabolism for hypoxic conditions to protect the heart from IR injury.


Subject(s)
Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Smad2 Protein/metabolism , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Male , Mice , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myostatin/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Smad2 Protein/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Basic Res Cardiol ; 114(2): 7, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30635790

ABSTRACT

Sprouty1 (Spry1) is a negative modulator of receptor tyrosine kinase signaling, but its role in cardiomyocyte survival has not been elucidated. The aim of this study was to investigate the potential role of cardiomyocyte Spry1 in cardiac ischemia-reperfusion (I/R) injury. Infarct areas of mouse hearts showed an increase in Spry1 protein expression, which localized to cardiomyocytes. To investigate if cardiomyocyte Spry1 regulates I/R injury, 8-week-old inducible cardiomyocyte Spry1 knockout (Spry1 cKO) mice and control mice were subjected to cardiac I/R injury. Spry1 cKO mice showed reduction in release of cardiac troponin I and reduced infarct size after I/R injury compared to control mice. Similar to Spry1 knockdown in cardiomyocytes in vivo, RNAi-mediated Spry1 silencing in isolated cardiomyocytes improved cardiomyocyte survival following simulated ischemia injury. Mechanistically, Spry1 knockdown induced cardiomyocyte extracellular signal-regulated kinase (ERK) phosphorylation in healthy hearts and isolated cardiomyocytes, and enhanced ERK phosphorylation after I/R injury. Spry1-deficient cardiomyocytes showed better preserved mitochondrial membrane potential following ischemic injury and an increase in levels of phosphorylated ERK and phosphorylated glycogen synthase kinase-3ß (GSK-3ß) in mitochondria of hypoxic cardiomyocytes. Overexpression of constitutively active GSK-3ß abrogated the protective effect of Spry1 knockdown. Moreover, pharmacological inhibition of GSK-3ß protected wild-type cardiomyocytes from cell death, but did not further protect Spry1-silenced cardiomyocytes from hypoxia-induced injury. Cardiomyocyte Spry1 knockdown promotes ERK phosphorylation and offers protection from I/R injury. Our findings indicate that Spry1 is an important regulator of cardiomyocyte viability during ischemia-reperfusion injury.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Membrane Proteins/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Phosphoproteins/metabolism , Animals , Cell Survival/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats
7.
Basic Res Cardiol ; 111(1): 2, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26611206

ABSTRACT

The G protein-coupled apelin receptor regulates important processes of the cardiovascular homeostasis, including cardiac development, cardiac contractility, and vascular tone. Most recently, a novel endogenous peptide ligand for the apelin receptor was identified in zebrafish, and it was named apela/elabela/toddler. The peptide was originally considered as an exclusively embryonic regulator, and so far its function in the adult organism remains elusive. We show here that apela is predominantly expressed in the non-cardiomyocyte fraction in the adult rodent heart. We also provide evidence that apela binds to apelin receptors in the heart. Using isolated adult rat hearts, we demonstrate, that just like the fellow receptor agonist apelin, apela increases cardiac contractility and induces coronary vasodilation already in the nanomolar level. The inotropic effect, as revealed by Western blot analysis, is accompanied by a significant increase in extracellular signal-regulated kinase (ERK) 1/2 phosphorylation. Pharmacological inhibition of ERK1/2 activation markedly attenuates the apela-induced inotropy. Analysis of samples from infarcted mouse hearts showed that expression of both apela and apelin receptor is induced in failing mouse hearts and correlate with left ventricular ejection fraction. Hence, we conclude that apela is present in the adult heart, is upregulated in post-infarction cardiac remodeling, and increases cardiac contractility in an ERK1/2-dependent manner.


Subject(s)
Heart , Intercellular Signaling Peptides and Proteins/metabolism , Myocardium/metabolism , Receptors, G-Protein-Coupled/metabolism , Aging , Animals , Apelin Receptors , Blotting, Western , Disease Models, Animal , Male , Mice , Myocardial Infarction/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/physiology
9.
J Immunol ; 195(5): 2187-98, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26209629

ABSTRACT

Spontaneous preterm birth (SPTB) is a major factor associating with deaths and with lowered quality of life in humans. Environmental and genetic factors influence the susceptibility. Previously, by analyzing families with recurrent SPTB in linkage analysis, we identified a linkage peak close to the gene encoding CXCR3. Present objectives were to investigate the association of CXCR3 with SPTB in Finnish mothers (n = 443) and infants (n = 747), to analyze CXCR3 expression levels in human placenta and levels of its ligands in umbilical cord blood, and to verify the influence of Cxcr3 on SPTB-associating cytokines in mice. We detected an association between an intronic CXCR3 polymorphism, rs2280964, and SPTB in infants from families with recurrent preterm births (p = 0.009 versus term controls, odds ratio 0.52, 95% confidence interval 0.32-0.86). The minor allele was protective and undertransmitted to SPTB infants (p = 0.007). In the placenta and fetal membranes, the rs2280964 major allele homozygotes had higher expression levels than minor allele homozygotes; decidual trophoblasts showed strong CXCR3 immunoreactivity. Expression was higher in SPTB placentas compared with those from elective deliveries. Concentration of a CXCR3 ligand, CXCL9, was increased in cord blood from SPTB, and the protective rs2280964 allele was associated with low CXCL9. In CXCR3-deficient mice (Mus musculus), SPTB-associating cytokines were not acutely increased in amniotic fluid after preterm birth-inducing dose of maternal LPS. Our results indicate that CXCR3 contributes to SPTB. Activation of CXCR3 signaling may disturb the maternal-fetal tolerance, and this may promote labor.


Subject(s)
Genetic Predisposition to Disease/genetics , Polymorphism, Single Nucleotide , Premature Birth/genetics , Receptors, CXCR3/genetics , Alleles , Animals , Blotting, Western , Case-Control Studies , Cytokines/blood , Female , Fetal Blood/metabolism , Gene Expression , Gene Frequency , Humans , Immunohistochemistry , Infant, Newborn , Male , Mice, Inbred C57BL , Mice, Knockout , Placenta/metabolism , Pregnancy , Receptors, CXCR3/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Hypertension ; 63(6): 1235-40, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24688123

ABSTRACT

Connective tissue growth factor (CTGF) is involved in the pathogenesis of various fibrotic disorders. However, its role in the heart is not clear. To investigate the role of CTGF in regulating the development of cardiac fibrosis and heart failure, we subjected mice to thoracic aortic constriction (TAC) or angiotensin II infusion, and antagonized the function of CTGF with CTGF monoclonal antibody (mAb). After 8 weeks of TAC, mice treated with CTGF mAb had significantly better preserved left ventricular (LV) systolic function and reduced LV dilatation compared with mice treated with control immunoglobulin G. CTGF mAb-treated mice exhibited significantly smaller cardiomyocyte cross-sectional area and reduced expression of hypertrophic marker genes. CTGF mAb treatment reduced the TAC-induced production of collagen 1 but did not significantly attenuate TAC-induced accumulation of interstitial fibrosis. Analysis of genes regulating extracellular matrix proteolysis showed decreased expression of plasminogen activator inhibitor-1 and matrix metalloproteinase-2 in mice treated with CTGF mAb. In contrast to TAC, antagonizing the function of CTGF had no effect on LV dysfunction or LV hypertrophy in mice subjected to 4-week angiotensin II infusion. Further analysis showed that angiotensin II-induced expression of hypertrophic marker genes or collagens was not affected by treatment with CTGF mAb. In conclusion, CTGF mAb protects from adverse LV remodeling and LV dysfunction in hearts subjected to pressure overload by TAC. Antagonizing the function of CTGF may offer protection from cardiac end-organ damage in patients with hypertension.


Subject(s)
Antibodies, Monoclonal/pharmacology , Connective Tissue Growth Factor/antagonists & inhibitors , Heart Failure/complications , Ventricular Dysfunction, Left/prevention & control , Ventricular Remodeling/drug effects , Angiotensin II/pharmacology , Animals , Antibodies, Monoclonal/immunology , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Collagen Type I/genetics , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/immunology , Constriction, Pathologic/physiopathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Gene Expression/drug effects , Heart/drug effects , Heart/physiopathology , Heart Failure/physiopathology , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Plasminogen Activator Inhibitor 1/genetics , Pressure , Reverse Transcriptase Polymerase Chain Reaction , Ventricular Dysfunction, Left/etiology , Weight-Bearing/physiology
11.
J Mol Cell Cardiol ; 67: 86-93, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24361238

ABSTRACT

cAMP-dependent protein kinase (PKA) regulates the L-type calcium channel, the ryanodine receptor, and phospholamban (PLB) thereby increasing inotropy. Cardiac contractility is also regulated by p38 MAPK, which is a negative regulator of cardiac contractile function. The aim of this study was to identify the mechanism mediating the positive inotropic effect of p38 inhibition. Isolated adult and neonatal cardiomyocytes and perfused rat hearts were utilized to investigate the molecular mechanisms regulated by p38. PLB phosphorylation was enhanced in cardiomyocytes by chemical p38 inhibition, by overexpression of dominant negative p38α and by p38α RNAi, but not with dominant negative p38ß. Treatment of cardiomyocytes with dominant negative p38α significantly decreased Ca(2+)-transient decay time indicating enhanced sarco/endoplasmic reticulum Ca(2+)-ATPase function and increased cardiomyocyte contractility. Analysis of signaling mechanisms involved showed that inhibition of p38 decreased the activity of protein phosphatase 2A, which renders protein phosphatase inhibitor-1 phosphorylated and thereby inhibits PP1. In conclusion, inhibition of p38α enhances PLB phosphorylation and diastolic Ca(2+) uptake. Our findings provide evidence for novel mechanism regulating cardiac contractility upon p38 inhibition.


Subject(s)
Muscle Contraction/physiology , Myocytes, Cardiac/physiology , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Calcium/metabolism , Calcium-Binding Proteins/metabolism , Enzyme Activation/drug effects , Myocytes, Cardiac/drug effects , Phosphorylation , RNA Interference , Rats , p38 Mitogen-Activated Protein Kinases/pharmacology
12.
Mol Pharmacol ; 83(1): 129-41, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23066091

ABSTRACT

The ß(1)-adrenergic receptor (ß(1)AR) is the predominant ßAR in the heart and is the main target for ß-adrenergic antagonists, widely used in the treatment of cardiovascular diseases. Previously, we have shown that the human (h) ß(1)AR is cleaved in its N terminus by a metalloproteinase, both constitutively and in a receptor activation-dependent manner. In this study, we investigated the specific events involved in ß(1)AR regulation, focusing on the effects of long-term treatment with ß-adrenergic ligands on receptor processing in stably transfected human embryonic kidney 293(i) cells. The key findings were verified using the transiently transfected hß(1)AR and the endogenously expressed receptor in neonatal rat cardiomyocytes. By using flow cytometry and Western blotting, we demonstrated that isoproterenol, S-propranolol, CGP-12177 [4-[3-[(1,1-dimethylethyl)amino]2-hydroxypropoxy]-1,3-dihydro-2H-benzimidazol-2-one], pindolol, and timolol, which displayed agonistic properties toward the ß(1)AR in either the adenylyl cyclase or the mitogen-activated protein kinase signaling pathways, induced cleavage of the mature cell-surface receptor. In contrast, metoprolol, bisoprolol, and CGP-20712 [1-[2-((3-carbamoyl-4-hydroxy)phenoxy)ethylamino]-3-[4-(1-methyl-4-trifluoromethyl-2-imidazolyl)phenoxy]-2-propanol], which showed no agonistic activity, had only a marginal or no effect. Importantly, the agonists also stabilized intracellular receptor precursors, possibly via their pharmacological chaperone action, and they stabilized the receptor in vitro. The opposing effects on the two receptor forms thus led to an increase in the amount of cleaved receptor fragments at the plasma membrane. The results underscore the pluridimensionality of ß-adrenergic ligands and extend this property from receptor activation and signaling to the regulation of ß(1)AR levels. This phenomenon may contribute to the exceptional resistance of ß(1)ARs to downregulation and tendency toward upregulation following long-term ligand treatments.


Subject(s)
Adrenergic beta-1 Receptor Agonists/pharmacology , Receptors, Adrenergic, beta-1/metabolism , Adrenergic beta-1 Receptor Antagonists/pharmacology , Animals , Animals, Newborn , Cell Membrane/drug effects , Cell Membrane/metabolism , Cyclic AMP/biosynthesis , HEK293 Cells , Humans , Ligands , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phosphorylation , Protein Precursors/genetics , Protein Precursors/metabolism , Protein Stability , Radioligand Assay , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta-1/genetics , Signal Transduction , Transfection , Up-Regulation
13.
PLoS One ; 7(12): e51378, 2012.
Article in English | MEDLINE | ID: mdl-23227263

ABSTRACT

Preterm birth is the major cause of neonatal mortality and morbidity. In many cases, it has severe life-long consequences for the health and neurological development of the newborn child. More than 50% of all preterm births are spontaneous, and currently there is no effective prevention. Several studies suggest that genetic factors play a role in spontaneous preterm birth (SPTB). However, its genetic background is insufficiently characterized. The aim of the present study was to perform a linkage analysis of X chromosomal markers in SPTB in large northern Finnish families with recurrent SPTBs. We found a significant linkage signal (HLOD = 3.72) on chromosome locus Xq13.1 when the studied phenotype was being born preterm. There were no significant linkage signals when the studied phenotype was giving preterm deliveries. Two functional candidate genes, those encoding the androgen receptor (AR) and the interleukin-2 receptor gamma subunit (IL2RG), located near this locus were analyzed as candidates for SPTB in subsequent case-control association analyses. Nine single-nucleotide polymorphisms (SNPs) within these genes and an AR exon-1 CAG repeat, which was previously demonstrated to be functionally significant, were analyzed in mothers with preterm delivery (n = 272) and their offspring (n = 269), and in mothers with exclusively term deliveries (n = 201) and their offspring (n = 199), all originating from northern Finland. A replication study population consisting of individuals born preterm (n = 111) and term (n = 197) from southern Finland was also analyzed. Long AR CAG repeats (≥ 26) were overrepresented and short repeats (≤ 19) underrepresented in individuals born preterm compared to those born at term. Thus, our linkage and association results emphasize the role of the fetal genome in genetic predisposition to SPTB and implicate AR as a potential novel fetal susceptibility gene for SPTB.


Subject(s)
Chromosomes, Human, X/genetics , Genetic Association Studies , Genetic Linkage , Genetic Predisposition to Disease , Premature Birth/genetics , Receptors, Androgen/genetics , Case-Control Studies , Family , Female , Fetus/pathology , Finland , Genetic Markers , Haplotypes/genetics , Humans , Interleukin Receptor Common gamma Subunit/genetics , Lod Score , Logistic Models , Male , Models, Genetic , Pedigree , Phenotype , Polymorphism, Single Nucleotide/genetics , Reproducibility of Results , Trinucleotide Repeat Expansion/genetics
14.
PLoS One ; 7(5): e37153, 2012.
Article in English | MEDLINE | ID: mdl-22606343

ABSTRACT

Drosophila is a well-established model organism for studying innate immunity because of its high resistance against microbial infections and lack of adaptive immunity. In addition, the immune signaling cascades found in Drosophila are evolutionarily conserved. Upon infection, activation of the immune signaling pathways, Toll and Imd, leads to the expression of multiple immune response genes, such as the antimicrobial peptides (AMPs). Previously, we identified an uncharacterized gene edin among the genes, which were strongly induced upon stimulation with Escherichia coli in Drosophila S2 cells. Edin has been associated with resistance against Listeria monocytogenes, but its role in Drosophila immunity remains elusive. In this study, we examined the role of Edin in the immune response of Drosophila both in vitro and in vivo. We report that edin expression is dependent on the Imd-pathway NF-κB transcription factor Relish and that it is expressed upon infection both in vitro and in vivo. Edin encodes a pro-protein, which is further processed in S2 cells. In our experiments, Edin did not bind microbes, nor did it possess antimicrobial activity to tested microbial strains in vitro or in vivo. Furthermore, edin RNAi did not significantly affect the expression of AMPs in vitro or in vivo. However, edin RNAi flies showed modestly impaired resistance to E. faecalis infection. We conclude that Edin has no potent antimicrobial properties but it appears to be important for E. faecalis infection via an uncharacterized mechanism. Further studies are still required to elucidate the exact role of Edin in the Drosophila immune response.


Subject(s)
Drosophila Proteins/genetics , Drosophila Proteins/immunology , Drosophila melanogaster/genetics , Drosophila melanogaster/immunology , Amino Acid Sequence , Animals , Animals, Genetically Modified , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Base Sequence , Cell Line , DNA Primers/genetics , Drosophila Proteins/antagonists & inhibitors , Drosophila melanogaster/microbiology , Genes, Insect , Gram-Negative Bacterial Infections/genetics , Gram-Negative Bacterial Infections/immunology , Immunity, Innate , Molecular Sequence Data , RNA Interference , Sequence Homology, Amino Acid , Signal Transduction , Species Specificity , Transcription Factors/genetics , Transcription Factors/immunology
15.
APMIS ; 119(10): 651-62, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21917002

ABSTRACT

In mammals, phagocytosis coordinates host defence on two levels: It acts both as an effector of the innate immunity, as well as an initiator of the adaptive immunity. The fruit fly Drosophila melanogaster (D. melanogaster) lacks the adaptive immune response, and the role of Drosophila plasmatocytes, cells that resemble phagocytosing mammalian macrophages, is limited to innate immune responses. During the past years, several studies have shed light on the role of phagocytosis in the Drosophila host defence. At least in some infection models, the systemic production of potent antimicrobial peptides (AMPs) does not completely compensate for the need for cellular immune responses. As a model, Drosophila offers powerful tools for studying phagocytosis including, large-scale RNA interference (RNAi) based in vitro screens that can be combined with classical Drosophila genetics. These kinds of approaches have led to important discoveries related especially to microbial recognition by Drosophila plasmatocytes. Events following initial recognition, however, have remained more elusive. This review summarizes the current knowledge on Drosophila phagocytosis focusing on the most recent advancements in the field, and highlighting the benefits the Drosophila system has to offer for research on phagocytosis.


Subject(s)
Drosophila melanogaster/immunology , Immunity, Innate/immunology , Phagocytosis/immunology , Animals , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Hemocytes/immunology , Host-Pathogen Interactions/immunology , Phagocytosis/genetics , Signal Transduction/immunology
16.
J Leukoc Biol ; 89(5): 649-59, 2011 May.
Article in English | MEDLINE | ID: mdl-21208897

ABSTRACT

Phagocytosis is an ancient cellular process that plays an important role in host defense. In Drosophila melanogaster phagocytic, macrophage-like hemocytes recognize and ingest microbes. We performed an RNAi-based in vitro screen in the Drosophila hemocyte cell line S2 and identified Abi, cpa, cofilin regulator 14-3-3ζ, tlk, CG2765, and CG15609 as mediators of bacterial phagocytosis. Of these identified genes, 14-3-3ζ had an evolutionarily conserved role in phagocytosis: bacterial phagocytosis was compromised when 14-3-3ζ was targeted with RNAi in primary Drosophila hemocytes and when the orthologous genes Ywhab and Ywhaz were silenced in zebrafish and mouse RAW 264.7 cells, respectively. In Drosophila and zebrafish infection models, 14-3-3ζ was required for resistance against Staphylococcus aureus. We conclude that 14-3-3ζ is essential for phagocytosis and microbial resistance in insects and vertebrates.


Subject(s)
14-3-3 Proteins/genetics , Actin Depolymerizing Factors/genetics , Biological Evolution , Drosophila melanogaster/genetics , Drosophila melanogaster/microbiology , Escherichia coli/pathogenicity , Phagocytosis/physiology , 14-3-3 Proteins/antagonists & inhibitors , Animals , Cells, Cultured , Drosophila melanogaster/immunology , Escherichia coli Infections/genetics , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Female , Gene Silencing , Genes, Insect , Hemocytes/metabolism , Male , Mice , RNA, Small Interfering/genetics , Zebrafish/genetics , Zebrafish/immunology , Zebrafish/microbiology
17.
J Mol Cell Cardiol ; 50(4): 634-41, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21223972

ABSTRACT

Parthenolide has shown promise in treatment of various cancers via inhibition of the transcription factor signal transducer and activator of transcription 3 (STAT3). Activation of STAT3 has been observed in left ventricular hypertrophy (LVH); however, its exact role is not known. The aim of the study was to examine the effects of parthenolide on pressure overload-induced LVH in rats. Pressure overload was induced by angiotensin II (Ang II) infusion (33 µg/kg/h) for 1 week in the presence or absence of parthenolide (0.5mg/kg/day, i.p.). Ang II infusion resulted in LVH associated with increased phosphorylation of STAT3 at Tyr705 and Ser727. Parthenolide treatment had no effect on ejection fraction, but abolished the activation of STAT3 and reduced the Ang II-induced LVH (LV posterior wall thickness in end-diastole: 2.28 ± 0.12 mm vs. 1.80 ± 0.06 mm, P<0.001). Importantly, parthenolide treatment had no effect on heart rate or blood pressure. Parthenolide treatment almost completely abolished the Ang II-induced increase in the number of cells positive for prolyl-4-hydroxylase, a marker for collagen-synthesizing cells, as well as Ang II-induced interstitial fibrosis in the left ventricles. This was associated with significant attenuation of Ang II-induced increase in mRNA levels of type 1 collagen and fibronectin. Moreover, parthenolide attenuated the Ang II-induced expression of interleukin-6, a potent pro-hypertrophic fibroblast-derived factor. We conclude that pharmacological inhibition of STAT3 signaling by parthenolide has favorable effects on pressure overload-induced LVH through attenuation of fibroblast activation. Therefore parthenolide may prove as a useful therapy for certain cardiovascular disease.


Subject(s)
Angiotensin II/therapeutic use , Fibroblasts/drug effects , Fibroblasts/metabolism , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/drug therapy , STAT3 Transcription Factor/metabolism , Sesquiterpenes/pharmacology , Animals , Blotting, Western , Echocardiography , Electrophoretic Mobility Shift Assay , Hemodynamics/drug effects , Hypertrophy, Left Ventricular/metabolism , Immunohistochemistry , Male , Phosphorylation/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
18.
J Immunol ; 184(11): 6188-98, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20421637

ABSTRACT

Because NF-kappaB signaling pathways are highly conserved in evolution, the fruit fly Drosophila melanogaster provides a good model to study these cascades. We carried out an RNA interference (RNAi)-based genome-wide in vitro reporter assay screen in Drosophila for components of NF-kappaB pathways. We analyzed 16,025 dsRNA-treatments and identified 10 novel NF-kappaB regulators. Of these, nine dsRNA-treatments affect primarily the Toll pathway. G protein-coupled receptor kinase (Gprk)2, CG15737/Toll pathway activation mediating protein, and u-shaped were required for normal Drosomycin response in vivo. Interaction studies revealed that Gprk2 interacts with the Drosophila IkappaB homolog Cactus, but is not required in Cactus degradation, indicating a novel mechanism for NF-kappaB regulation. Morpholino silencing of the zebrafish ortholog of Gprk2 in fish embryos caused impaired cytokine expression after Escherichia coli infection, indicating a conserved role in NF-kappaB signaling. Moreover, small interfering RNA silencing of the human ortholog GRK5 in HeLa cells impaired NF-kappaB reporter activity. Gprk2 RNAi flies are susceptible to infection with Enterococcus faecalis and Gprk2 RNAi rescues Toll(10b)-induced blood cell activation in Drosophila larvae in vivo. We conclude that Gprk2/GRK5 has an evolutionarily conserved role in regulating NF-kappaB signaling.


Subject(s)
Drosophila Proteins/immunology , G-Protein-Coupled Receptor Kinase 2/immunology , G-Protein-Coupled Receptor Kinase 5/metabolism , Immunity, Innate , NF-kappa B/immunology , Signal Transduction/physiology , Animals , Blotting, Western , Drosophila , Drosophila Proteins/metabolism , G-Protein-Coupled Receptor Kinase 2/metabolism , G-Protein-Coupled Receptor Kinase 5/immunology , Gram-Negative Chemolithotrophic Bacteria/immunology , Gram-Negative Chemolithotrophic Bacteria/metabolism , Humans , Immunohistochemistry , Immunoprecipitation , NF-kappa B/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Zebrafish
19.
J Immunol ; 180(8): 5413-22, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18390723

ABSTRACT

NF-kappaB transcription factors are involved in evolutionarily conserved signaling pathways controlling multiple cellular processes including apoptosis and immune and inflammatory responses. Immune response of the fruit fly Drosophila melanogaster to Gram-negative bacteria is primarily mediated via the Imd (immune deficiency) pathway, which closely resembles the mammalian TNFR signaling pathway. Instead of cytokines, the main outcome of Imd signaling is the production of antimicrobial peptides. The pathway activity is delicately regulated. Although many of the Imd pathway components are known, the mechanisms of negative regulation are more elusive. In this study we report that a previously uncharacterized gene, pirk, is highly induced upon Gram-negative bacterial infection in Drosophila in vitro and in vivo. pirk encodes a cytoplasmic protein that coimmunoprecipitates with Imd and the cytoplasmic tail of peptidoglycan recognition protein LC (PGRP-LC). RNA interference-mediated down-regulation of Pirk caused Imd pathway hyperactivation upon infection with Gram-negative bacteria, while overexpression of pirk reduced the Imd pathway response both in vitro and in vivo. Furthermore, pirk-overexpressing flies were more susceptible to Gram-negative bacterial infection than wild-type flies. We conclude that Pirk is a negative regulator of the Imd pathway.


Subject(s)
Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster/immunology , Drosophila melanogaster/microbiology , Enterobacter cloacae/immunology , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/immunology , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila Proteins/immunology , Drosophila melanogaster/metabolism , Enterobacter cloacae/physiology , Genes, Insect , Molecular Sequence Data , NF-kappa B/immunology , NF-kappa B/metabolism , Oligonucleotide Array Sequence Analysis , RNA Interference , Sequence Alignment , Signal Transduction
20.
J Biol Chem ; 281(20): 14370-5, 2006 May 19.
Article in English | MEDLINE | ID: mdl-16531407

ABSTRACT

Double-stranded RNA (dsRNA) fragments are readily internalized and processed by Drosophila S2 cells, making these cells a widely used tool for the analysis of gene function by gene silencing through RNA interference (RNAi). The underlying mechanisms are insufficiently understood. To identify components of the RNAi pathway in S2 cells, we developed a screen based on rescue from RNAi-induced lethality. We identified Argonaute 2, a core component of the RNAi machinery, and three gene products previously unknown to be involved in RNAi in Drosophila: DEAD-box RNA helicase Belle, 26 S proteasome regulatory subunit 8 (Pros45), and clathrin heavy chain, a component of the endocytic machinery. Blocking endocytosis in S2 cells impaired RNAi, suggesting that dsRNA fragments are internalized by receptor-mediated endocytosis. Indeed, using a candidate gene approach, we identified two Drosophila scavenger receptors, SR-CI and Eater, which together accounted for more than 90% of the dsRNA uptake into S2 cells. When expressed in mammalian cells, SR-CI was sufficient to mediate internalization of dsRNA fragments. Our data provide insight into the mechanism of dsRNA internalization by Drosophila cells. These results have implications for dsRNA delivery into mammalian cells.


Subject(s)
Endocytosis , RNA, Double-Stranded/chemistry , Receptors, Scavenger/metabolism , Animals , Cell Line , Drosophila melanogaster , Gene Library , Gene Silencing , Genetic Techniques , Luciferases/metabolism , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL
...