Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 636
Filter
Add more filters

Publication year range
1.
Circ Res ; 134(8): 1006-1022, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38506047

ABSTRACT

BACKGROUND: In heart failure, signaling downstream the ß2-adrenergic receptor is critical. Sympathetic stimulation of ß2-adrenergic receptor alters cAMP (cyclic adenosine 3',5'-monophosphate) and triggers PKA (protein kinase A)-dependent phosphorylation of proteins that regulate cardiac function. cAMP levels are regulated in part by PDEs (phosphodiesterases). Several AKAPs (A kinase anchoring proteins) regulate cardiac function and are proposed as targets for precise pharmacology. AKAP12 is expressed in the heart and has been reported to directly bind ß2-adrenergic receptor, PKA, and PDE4D. However, its roles in cardiac function are unclear. METHODS: cAMP accumulation in real time downstream of the ß2-adrenergic receptor was detected for 60 minutes in live cells using the luciferase-based biosensor (GloSensor) in AC16 human-derived cardiomyocyte cell lines overexpressing AKAP12 versus controls. Cardiomyocyte intracellular calcium and contractility were studied in adult primary cardiomyocytes from male and female mice overexpressing cardiac AKAP12 (AKAP12OX) and wild-type littermates post acute treatment with 100-nM isoproterenol (ISO). Systolic cardiac function was assessed in mice after 14 days of subcutaneous ISO administration (60 mg/kg per day). AKAP12 gene and protein expression levels were evaluated in left ventricular samples from patients with end-stage heart failure. RESULTS: AKAP12 upregulation significantly reduced total intracellular cAMP levels in AC16 cells through PDE8. Adult primary cardiomyocytes from AKAP12OX mice had significantly reduced contractility and impaired calcium handling in response to ISO, which was reversed in the presence of the selective PDE8 inhibitor (PF-04957325). AKAP12OX mice had deteriorated systolic cardiac function and enlarged left ventricles. Patients with end-stage heart failure had upregulated gene and protein levels of AKAP12. CONCLUSIONS: AKAP12 upregulation in cardiac tissue is associated with accelerated cardiac dysfunction through the AKAP12-PDE8 axis.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases , Heart Diseases , Receptors, Adrenergic , Animals , Female , Humans , Male , Mice , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , A Kinase Anchor Proteins/genetics , A Kinase Anchor Proteins/metabolism , Calcium/metabolism , Cell Cycle Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Heart Diseases/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Isoproterenol/pharmacology , Myocytes, Cardiac/metabolism , Receptors, Adrenergic/metabolism , Up-Regulation
2.
Mol Microbiol ; 119(4): 401-422, 2023 04.
Article in English | MEDLINE | ID: mdl-36760076

ABSTRACT

Cyclic AMP (cAMP) signaling is essential to Mycobacterium tuberculosis (Mtb) pathogenesis. However, the roles of phosphodiesterases (PDEs) Rv0805, and the recently identified Rv1339, in cAMP homeostasis and Mtb biology are unclear. We found that Rv0805 modulates Mtb growth within mice, macrophages and on host-associated carbon sources. Mycobacterium bovis BCG grown on a combination of propionate and glycerol as carbon sources showed high levels of cAMP and had a strict requirement for Rv0805 cNMP hydrolytic activity. Supplementation with vitamin B12 or spontaneous genetic mutations in the pta-ackA operon restored the growth of BCGΔRv0805 and eliminated propionate-associated cAMP increases. Surprisingly, reduction of total cAMP levels by ectopic expression of Rv1339 restored only 20% of growth, while Rv0805 complementation fully restored growth despite a smaller effect on total cAMP levels. Deletion of an Rv0805 localization domain also reduced BCG growth in the presence of propionate and glycerol. We propose that localized Rv0805 cAMP hydrolysis modulates activity of a specialized pathway associated with propionate metabolism, while Rv1339 has a broader role in cAMP homeostasis. Future studies will address the biological roles of Rv0805 and Rv1339, including their impacts on metabolism, cAMP signaling and Mtb pathogenesis.


Subject(s)
Mycobacterium tuberculosis , Phosphoric Diester Hydrolases , Animals , Mice , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Nucleotides, Cyclic/metabolism , Propionates/metabolism , Virulence , Hydrolysis , BCG Vaccine/metabolism , Glycerol/metabolism , Cyclic AMP/metabolism , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism
3.
Neurochem Res ; 49(8): 1993-2004, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38782837

ABSTRACT

Phosphodiesterase 8 (PDE8), as a member of PDE superfamily, specifically promotes the hydrolysis and degradation of intracellular cyclic adenosine monophosphate (cAMP), which may be associated with pathogenesis of Alzheimer's disease (AD). However, little is currently known about potential role in the central nervous system (CNS). Here we investigated the distribution and expression of PDE8 in brain of mouse, which we believe can provide evidence for studying the role of PDE8 in CNS and the relationship between PDE8 and AD. Here, C57BL/6J mice were used to observe the distribution patterns of two subtypes of PDE8, PDE8A and PDE8B, in different sexes in vivo by western blot (WB). Meanwhile, C57BL/6J mice were also used to demonstrate the distribution pattern of PDE8 in selected brain regions and localization in neural cells by WB and multiplex immunofluorescence staining. Furthermore, the triple transgenic (3×Tg-AD) mice and wild type (WT) mice of different ages were used to investigate the changes of PDE8 expression in the hippocampus and cerebral cortex during the progression of AD. PDE8 was found to be widely expressed in multiple tissues and organs including heart, kidney, stomach, brain, and liver, spleen, intestines, and uterus, with differences in expression levels between the two subtypes of PDE8A and PDE8B, as well as two sexes. Meanwhile, PDE8 was widely distributed in the brain, especially in areas closely related to cognitive function such as cerebellum, striatum, amygdala, cerebral cortex, and hippocampus, without differences between sexes. Furthermore, PDE8A was found to be expressed in neuronal cells, microglia and astrocytes, while PDE8B is only expressed in neuronal cells and microglia. PDE8A expression in the hippocampus of both female and male 3×Tg-AD mice was gradually increased with ages and PDE8B expression was upregulated only in cerebral cortex of female 3×Tg-AD mice with ages. However, the expression of PDE8A and PDE8B was apparently increased in both cerebral cortex and hippocampus in both female and male 10-month-old 3×Tg-AD mice compared WT mice. These results suggest that PDE8 may be associated with the progression of AD and is a potential target for its prevention and treatment in the future.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases , Alzheimer Disease , Mice, Inbred C57BL , Mice, Transgenic , Animals , Female , Male , Mice , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Alzheimer Disease/metabolism , Brain/metabolism , Hippocampus/metabolism
4.
Transgenic Res ; 32(6): 575-582, 2023 12.
Article in English | MEDLINE | ID: mdl-37851308

ABSTRACT

M-LP/Mpv17L (Mpv17-like protein) is an atypical cyclic nucleotide phosphodiesterase (PDE) without the molecular structure characteristic of the PDE family. Deficiency of M-LP/Mpv17L in mice has been found to result in development of ß-cell hyperplasia and improved glucose tolerance. Here, we report another phenotype observed in M-LP/Mpv17L-knockout (KO) mice: afferent cardiac hypertrophy. Although the hearts of M-LP/Mpv17L-KO mice did not differ in size from those of wild-type mice, there was marked narrowing of the left ventricular lumen and thickening of the ventricular wall. The diameter and cross-sectional area of cardiomyocytes in 8-month-old M-LP/Mpv17L-KO mice were increased 1.16-fold and 1.35-fold, respectively, relative to control mice, but showed no obvious abnormalities of cell structure, fibrosis or impaired cardiac function. In 80-day-old KO mice, the expression of hypertrophic marker genes, brain natriuretic peptide (BNF), actin alpha cardiac muscle 1 (ACTC1) and actin alpha 1 skeletal muscle (ACTA1), as well as the Wnt/ß-catenin pathway target genes, lymphoid enhancer-binding factor-1 (LEF1), axis inhibition protein 2 (AXIN2) and transcription factor 7 (TCF7), was significantly up-regulated relative to control mice, whereas fibrosis-related genes such as fibronectin 1 (FN1) and connective tissue growth factor (CTGF) were down-regulated. Western blot analysis revealed increased phosphorylation of molecules downstream of the cAMP/PKA signaling pathway, such as ß-catenin, ryanodine receptor 2 (RyR2), phospholamban (PLN) and troponin I (cTnI), as well as members of the MEK1-ERK1/2 signaling pathway, which is strongly involved in afferent cardiac hypertrophy. Taken together, these findings indicate that M-LP/Mpv17L is one of the PDEs actively functioning in the heart and that deficiency of M-LP/Mpv17L in mice promotes physiological cardiac hypertrophy.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases , Cardiomegaly , Animals , Mice , Actins/metabolism , beta Catenin/metabolism , Cardiomegaly/genetics , Cardiomegaly/metabolism , Fibrosis , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism
5.
Cell Physiol Biochem ; 55(6): 784-804, 2021 Dec 23.
Article in English | MEDLINE | ID: mdl-34936285

ABSTRACT

BACKGROUND/AIMS: Cystic fibrosis transmembrane conductance regulator (CFTR), the anion channel that is defective in cystic fibrosis (CF), is phosphorylated and activated by cAMP-dependent protein kinase (PKA). cAMP levels are downregulated by a large family of phosphodiesterases that have variable expression in different cell types. We have previously observed high levels of PDE8A expression in well-differentiated primary human bronchial epithelial (pHBE) cells and thus aimed to assess whether it played a role in cAMP-dependent regulation of CFTR activity. METHODS: We assessed the effect of the selective PDE8 inhibitor PF-04957325 (PF) on intracellular cAMP levels ([cAMP]i) in well differentiated pHBE cells from non-CF or CF donors and also in CFBE41o- cells that stably express wild-type CFTR (CFBE41o- WT) using ELISA and FRET-FLIM microscopy. CFTR channel function was also measured using electrophysiological recordings from pHBE and CFBE41o- WT cells mounted in Ussing Chambers. RESULTS: PDE8 inhibition elevated [cAMP]i in well-differentiated pHBE cells and stimulated wild-type CFTR-dependent ion transport under basal conditions or after cells had been pre-stimulated with physiological cAMP-elevating agents. The response to PDE8 inhibition was larger than to PDE3 or PDE5 inhibition but smaller and synergistic with that elicited by PDE4 inhibition. CRISPR Cas9-mediated knockdown of PDE8A enhanced CFTR gene and protein expression yet reduced the effect of PDE8 inhibition. Acute pharmacological inhibition PDE8 increased CFTR activity in CF pHBE cells (F508del/F508del and F508del/R117H-5T) treated with clinically-approved CFTR modulators. CONCLUSION: These results provide the first evidence that PDE8A regulates CFTR and identifies PDE8A as a potential target for adjunct therapies to treat CF.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis/metabolism , Epithelial Cells/metabolism , Respiratory Mucosa/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Cell Line , Cricetinae , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/pathology , Humans , Respiratory Mucosa/pathology
6.
BMC Cancer ; 21(1): 577, 2021 May 20.
Article in English | MEDLINE | ID: mdl-34016083

ABSTRACT

BACKGROUND: PDE9A (Phosphodiesterase 9A) plays an important role in proliferation of cells, their differentiation and apoptosis via intracellular cGMP (cyclic guanosine monophosphate) signaling. The expression pattern of PDE9A is associated with diverse tumors and carcinomas. Therefore, PDE9A could be a prospective candidate as a therapeutic target in different types of carcinoma. The study presented here was designed to carry out the prognostic value as a biomarker of PDE9A in Colorectal cancer (CRC). The present study integrated several cancer databases with in-silico techniques to evaluate the cancer prognosis of CRC. RESULTS: The analyses suggested that the expression of PDE9A was significantly down-regulated in CRC tissues than in normal tissues. Moreover, methylation in the DNA promoter region might also manipulate PDE9A gene expression. The Kaplan-Meier curves indicated that high level of expression of PDE9A gene was associated to higher survival in OS, RFS, and DSS in CRC patients. PDE9A demonstrated the highest positive correlation for rectal cancer recurrence with a marker gene CEACAM7. Furtheremore, PDE9A shared consolidated pathways with MAPK14 to induce survival autophagy in CRC cells and showed interaction with GUCY1A2 to drive CRPC. CONCLUSIONS: Overall, the prognostic value of PDE9A gene could be used as a potential tumor biomarker for CRC.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/genetics , Biomarkers, Tumor/genetics , Colorectal Neoplasms/mortality , Gene Expression Regulation, Neoplastic , Neoplasm Recurrence, Local/epidemiology , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Computer Simulation , Datasets as Topic , Disease-Free Survival , Humans , Kaplan-Meier Estimate , Neoplasm Recurrence, Local/genetics , Prognosis , Progression-Free Survival , Signal Transduction/genetics
8.
Nature ; 519(7544): 472-6, 2015 Mar 26.
Article in English | MEDLINE | ID: mdl-25799991

ABSTRACT

Cyclic guanosine monophosphate (cGMP) is a second messenger molecule that transduces nitric-oxide- and natriuretic-peptide-coupled signalling, stimulating phosphorylation changes by protein kinase G. Enhancing cGMP synthesis or blocking its degradation by phosphodiesterase type 5A (PDE5A) protects against cardiovascular disease. However, cGMP stimulation alone is limited by counter-adaptions including PDE upregulation. Furthermore, although PDE5A regulates nitric-oxide-generated cGMP, nitric oxide signalling is often depressed by heart disease. PDEs controlling natriuretic-peptide-coupled cGMP remain uncertain. Here we show that cGMP-selective PDE9A (refs 7, 8) is expressed in the mammalian heart, including humans, and is upregulated by hypertrophy and cardiac failure. PDE9A regulates natriuretic-peptide- rather than nitric-oxide-stimulated cGMP in heart myocytes and muscle, and its genetic or selective pharmacological inhibition protects against pathological responses to neurohormones, and sustained pressure-overload stress. PDE9A inhibition reverses pre-established heart disease independent of nitric oxide synthase (NOS) activity, whereas PDE5A inhibition requires active NOS. Transcription factor activation and phosphoproteome analyses of myocytes with each PDE selectively inhibited reveals substantial differential targeting, with phosphorylation changes from PDE5A inhibition being more sensitive to NOS activation. Thus, unlike PDE5A, PDE9A can regulate cGMP signalling independent of the nitric oxide pathway, and its role in stress-induced heart disease suggests potential as a therapeutic target.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Cardiomegaly/enzymology , Cardiomegaly/metabolism , Cyclic GMP/metabolism , Nitric Oxide , 3',5'-Cyclic-AMP Phosphodiesterases/antagonists & inhibitors , 3',5'-Cyclic-AMP Phosphodiesterases/deficiency , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Aortic Valve Stenosis/complications , Cardiomegaly/drug therapy , Cardiomegaly/etiology , Humans , Male , Mice , Mice, Inbred C57BL , Muscle Cells/enzymology , Myocardium/enzymology , Natriuretic Peptides/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase , Phosphodiesterase Inhibitors/pharmacology , Phosphodiesterase Inhibitors/therapeutic use , Pressure , Signal Transduction/drug effects , Stress, Physiological , Up-Regulation
9.
Microbiology (Reading) ; 166(2): 129-140, 2020 02.
Article in English | MEDLINE | ID: mdl-31730032

ABSTRACT

Mitogen-activated protein kinase (MAPK) regulation of cAMP-specific phosphodiesterase function has been demonstrated in mammalian cells and suspected to occur in other eukaryotes. Epistasis analysis in the soil amoeba Dictyostelium discoideum suggests the atypical MAPK Erk2 downregulates the function of the cAMP-specific phosphodiesterase RegA to regulate progression of the developmental life cycle. A putative MAPK docking motif located near a predicted MAPK phosphorylation site was characterized for contributions to RegA function and binding to Erk2 because a similar docking motif has been previously characterized in the mammalian PDE4D phosphodiesterase. The overexpression of RegA with alterations to this docking motif (RegAD-) restored RegA function to regA- cells based on developmental phenotypes, but low-level expression of RegAD- from the endogenous regA promoter failed to rescue wild-type morphogenesis. Co-immunoprecipitation analysis indicated that Erk2 associates with both RegA and RegAD-, suggesting the docking motif is not required for this association. Epistasis analysis between regA and the only other Dictyostelium MAPK, erk1, suggests Erk1 and RegA can function in different pathways but that some erk1- phenotypes may require cAMP signalling. These results imply that MAPK downregulation of RegA in Dictyostelium is accomplished through a different mechanism than MAPK regulation of cAMP-specific phosphodiesterases in mammalian cells and that the regulation in Dictyostelium does not require a proximal MAPK docking motif.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Dictyostelium/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protozoan Proteins/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Binding Sites , Dictyostelium/genetics , Dictyostelium/growth & development , Dictyostelium/metabolism , Models, Biological , Morphogenesis , Mutation , Phosphorylation , Protein Binding , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Signal Transduction
10.
Microb Cell Fact ; 19(1): 224, 2020 Dec 07.
Article in English | MEDLINE | ID: mdl-33287814

ABSTRACT

BACKGROUND: Monascus azaphilone pigments (MonAzPs), which were produced by Monascus species, have been used as important food colorant and food supplements for more than one billion people during their daily life. Moreover, MonAzPs recently have received more attention because of their diverse physiological activities. However, the high microbial production of MonAzPs is still not always guaranteed. Herein, the aim of this study was to develop an efficient biotechnological process for MonAzPs production. RESULTS: In this study, exogenous cyclic adenosine monophosphate (cAMP) treatment not only induced MonAzPs production, but also stimulated the expression of a cAMP phosphodiesterase gene, named as mrPDE, in M. purpureus HJ11. Subsequently, MrPDE was identified as a cAMP phosphodiesterase by in vitro enzymatic reaction with purified enzyme. Further, a gene knockout mutant of mrPDE was constructed to verify the activation of cAMP signalling pathway. Deletion of mrPDE in M. purpureus HJ11 improved cAMP concentration by 378% and enhanced PKA kinase activity 1.5-fold, indicating that activation of cAMP signalling pathway was achieved. The ΔmrPDE strain produced MonAzPs at 8563 U/g, with a 2.3-fold increase compared with the WT strain. Moreover, the NAPDH/NADP+ ratio of the ΔmrPDE strain was obviously higher than that of the wild type strain, which led to a higher proportion of yellow MonAzPs. With fed-batch fermentation of the ΔmrPDE strain, the production and yield of MonAzPs achieved 332.1 U/mL and 8739 U/g. CONCLUSIONS: A engineered M. purpureus strain for high MonAzPs production was successfully developed by activating the cAMP signalling pathway. This study not only describes a novel strategy for development of MonAzPs-producing strain, but also provides a roadmap for engineering efforts towards the production of secondary metabolism in other filamentous fungi.


Subject(s)
Cyclic AMP/metabolism , Metabolic Engineering , Monascus/metabolism , Pigments, Biological/biosynthesis , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Benzopyrans , Fermentation , Gene Knockout Techniques , Genes, Fungal , Monascus/genetics , NADP/metabolism , Secondary Metabolism , Signal Transduction
11.
J Neurovirol ; 25(6): 825-836, 2019 12.
Article in English | MEDLINE | ID: mdl-31332697

ABSTRACT

Treatment-emergent depression is a common complication in patients with chronic hepatitis C virus (HCV) infection undergoing antiviral combination therapy with IFN-α and ribavirin. It has recently been shown that changes in A-to-I RNA editing rates are associated with various pathologies such as inflammatory disorders, depression and suicide. Interestingly, IFN-α induces gene expression of the RNA editing enzyme ADAR1-1 (ADAR1a-p150) and alters overall RNA editing activity. In this study, we took advantage of the high prevalence of pharmacologically induced depression in patients treated with IFN-α and ribavirin to test the interest of RNA editing-related biomarkers in white blood cells of patients. In this 16-week longitudinal study, a small cohort of patients was clinically evaluated using standard assessment methods prior to and during antiviral therapy and blood samples were collected to analyse RNA editing modifications. A-I RNA editing activity on the phosphodiesterase 8A (PDE8A) gene, a previously identified RNA editing hotspot in the context of lupus erythematosus, was quantified by using an ultra-deep next-generation sequencing approach. We also monitored gene expression levels of the ADAR enzymes and the PDE8A gene during treatment by qPCR. As expected, psychiatric evaluation could track treatment-emergent depression, which occurred in 30% of HCV patients. We show that PDE8A RNA editing is increased in all patients following interferon treatment, but differently in 30% of patients. This effect was mimicked in a cellular model using SHSY-5Y neuroblastoma cells. By combining the data of A-I RNA editing and gene expression, we generated an algorithm that allowed discrimination between the group of patients who developed a treatment-emergent depression and those who did not. The current model of drug-induced depression identified A-I RNA editing biomarkers as useful tools for the identification of individuals at risk of developing depression in an objective, quantifiable biological blood test.


Subject(s)
Antiviral Agents/adverse effects , Biomarkers/blood , Depression/blood , Depression/chemically induced , Hepatitis C, Chronic/drug therapy , RNA Editing/drug effects , 3',5'-Cyclic-AMP Phosphodiesterases/blood , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Adenosine Deaminase/blood , Adenosine Deaminase/genetics , Adult , Aged , Female , Hepacivirus , Humans , Interferon-alpha/adverse effects , Longitudinal Studies , Male , Middle Aged , Polyethylene Glycols/adverse effects , RNA Editing/physiology , Recombinant Proteins/adverse effects , Ribavirin/adverse effects
12.
PLoS Genet ; 12(8): e1006174, 2016 08.
Article in English | MEDLINE | ID: mdl-27560698

ABSTRACT

The human face is a complex assemblage of highly variable yet clearly heritable anatomic structures that together make each of us unique, distinguishable, and recognizable. Relatively little is known about the genetic underpinnings of normal human facial variation. To address this, we carried out a large genomewide association study and two independent replication studies of Bantu African children and adolescents from Mwanza, Tanzania, a region that is both genetically and environmentally relatively homogeneous. We tested for genetic association of facial shape and size phenotypes derived from 3D imaging and automated landmarking of standard facial morphometric points. SNPs within genes SCHIP1 and PDE8A were associated with measures of facial size in both the GWAS and replication cohorts and passed a stringent genomewide significance threshold adjusted for multiple testing of 34 correlated traits. For both SCHIP1 and PDE8A, we demonstrated clear expression in the developing mouse face by both whole-mount in situ hybridization and RNA-seq, supporting their involvement in facial morphogenesis. Ten additional loci demonstrated suggestive association with various measures of facial shape. Our findings, which differ from those in previous studies of European-derived whites, augment understanding of the genetic basis of normal facial development, and provide insights relevant to both human disease and forensics.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/genetics , Carrier Proteins/genetics , Face/anatomy & histology , Genome-Wide Association Study , Maxillofacial Development/genetics , Adolescent , Animals , Black People , Female , Humans , Male , Mice , Morphogenesis/genetics , Phenotype , Polymorphism, Single Nucleotide , Tanzania
13.
Am J Respir Cell Mol Biol ; 58(4): 530-541, 2018 04.
Article in English | MEDLINE | ID: mdl-29262264

ABSTRACT

Two cAMP signaling compartments centered on adenylyl cyclase (AC) exist in human airway smooth muscle (HASM) cells, one containing ß2-adrenergic receptor AC6 and another containing E prostanoid receptor AC2. We hypothesized that different PDE isozymes selectively regulate cAMP signaling in each compartment. According to RNA-sequencing data, 18 of 24 PDE genes were expressed in primary HASM cells derived from age- and sex-matched donors with and without asthma. PDE8A was the third most abundant of the cAMP-degrading PDE genes, after PDE4A and PDE1A. Knockdown of PDE8A using shRNA evoked twofold greater cAMP responses to 1 µM forskolin in the presence of 3-isobutyl-1-methylxanthine. Overexpression of AC2 did not alter this response, but overexpression of AC6 increased cAMP responses an additional 80%. We examined cAMP dynamics in live HASM cells using a fluorescence sensor. PF-04957325, a PDE8-selective inhibitor, increased basal cAMP concentrations by itself, indicating a significant basal level of cAMP synthesis. In the presence of an AC inhibitor to reduce basal signaling, PF-04957325 accelerated cAMP production and increased the inhibition of cell proliferation induced by isoproterenol, but it had no effect on cAMP concentrations or cell proliferation regulated by prostaglandin E2. Lipid raft fractionation of HASM cells revealed PDE8A immunoreactivity in buoyant fractions containing caveolin-1 and AC5/6 immunoreactivity. Thus, PDE8 is expressed in lipid rafts of HASM cells, where it specifically regulates ß2-adrenergic receptor AC6 signaling without effects on signaling by the E prostanoid receptors 2/4-AC2 complex. In airway diseases such as asthma and chronic obstructive pulmonary disease, PDE8 may represent a novel therapeutic target to modulate HASM responsiveness and airway remodeling.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adenylyl Cyclases/metabolism , Asthma/enzymology , Cyclic AMP/metabolism , Muscle, Smooth/enzymology , Myocytes, Smooth Muscle/enzymology , Receptors, Adrenergic, beta-2/metabolism , Respiratory System/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Adenylyl Cyclases/genetics , Airway Remodeling , Asthma/genetics , Asthma/pathology , Asthma/physiopathology , Case-Control Studies , Cell Proliferation , Cells, Cultured , Humans , Membrane Microdomains/enzymology , Membrane Microdomains/pathology , Muscle, Smooth/pathology , Muscle, Smooth/physiopathology , Myocytes, Smooth Muscle/pathology , Receptors, Adrenergic, beta-2/genetics , Respiratory System/pathology , Respiratory System/physiopathology , Second Messenger Systems , Time Factors
14.
J Biol Chem ; 292(18): 7531-7541, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28302718

ABSTRACT

Photoactivated adenylyl cyclase (PAC) and guanylyl cyclase rhodopsin increase the concentrations of intracellular cyclic nucleotides upon illumination, serving as promising second-generation tools in optogenetics. To broaden the arsenal of such tools, it is desirable to have light-activatable enzymes that can decrease cyclic nucleotide concentrations in cells. Here, we report on an unusual microbial rhodopsin that may be able to meet the demand. It is found in the choanoflagellate Salpingoeca rosetta and contains a C-terminal cyclic nucleotide phosphodiesterase (PDE) domain. We examined the enzymatic activity of the protein (named Rh-PDE) both in HEK293 membranes and whole cells. Although Rh-PDE was constitutively active in the dark, illumination increased its hydrolytic activity 1.4-fold toward cGMP and 1.6-fold toward cAMP, as measured in isolated crude membranes. Purified full-length Rh-PDE displayed maximal light absorption at 492 nm and formed the M intermediate with the deprotonated Schiff base upon illumination. The M state decayed to the parent spectral state in 7 s, producing long-lasting activation of the enzyme domain with increased activity. We discuss a possible mechanism of the Rh-PDE activation by light. Furthermore, Rh-PDE decreased cAMP concentration in HEK293 cells in a light-dependent manner and could do so repeatedly without losing activity. Thus, Rh-PDE may hold promise as a potential optogenetic tool for light control of intracellular cyclic nucleotides (e.g. to study cyclic nucleotide-associated signal transduction cascades).


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Choanoflagellata/enzymology , Light , Protozoan Proteins/metabolism , Rhodopsin/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Choanoflagellata/genetics , HEK293 Cells , Humans , Protein Domains , Protozoan Proteins/genetics , Rhodopsin/genetics
15.
Microbiology (Reading) ; 164(3): 410-419, 2018 03.
Article in English | MEDLINE | ID: mdl-29458560

ABSTRACT

A variety of bacteria, including Escherichia coli, are known to enter the viable but non-culturable (VBNC) state under various stress conditions. During this state, cells lose colony-forming activities on conventional agar plates while retaining signs of viability. Diverse environmental stresses including starvation induce the VBNC state. However, little is known about the genetic mechanism inducing this state. Here, we aimed to reveal the genetic determinants of the VBNC state of E. coli. We hypothesized that the VBNC state is a process wherein specific gene products important for colony formation are depleted during the extended period of stress conditions. If so, higher expression of these genes would maintain colony-forming activities, thereby restraining cells from entering the VBNC state. From an E. coli plasmid-encoded ORF library, we identified genes that were responsible for maintaining high colony-forming activities after exposure to starvation condition. Among these, cpdA encoding cAMP phosphodiesterase exhibited higher performance in the maintenance of colony-forming activities. As cpdA overexpression decreases intracellular cAMP, cAMP or its complex with cAMP-receptor protein (CRP) may negatively regulate colony-forming activities under stress conditions. We confirmed this using deletion mutants lacking adenylate cyclase or CRP. These mutants fully maintained colony-forming activities even after a long period of starvation, while wild-type cells lost most of this activity. Thus, we concluded that the lack of cAMP-CRP effectively retains high colony-forming activities, indicating that cAMP-CRP acts as a positive regulator necessary for the induction of the VBNC state in E. coli.


Subject(s)
Cyclic AMP Receptor Protein/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/physiology , Stress, Physiological/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Cyclic AMP/metabolism , Cyclic AMP Receptor Protein/genetics , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli Proteins/genetics , Gene Deletion , Gene Expression , Gene Library
16.
J Biol Chem ; 291(7): 3668-81, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26668313

ABSTRACT

The intracellular infections of Mycobacterium tuberculosis, which is the causative agent of tuberculosis, are regulated by many cyclic dinucleotide signaling. Rv2837c from M. tuberculosis is a soluble, stand-alone DHH-DHHA1 domain phosphodiesterase that down-regulates c-di-AMP through catalytic degradation and plays an important role in M. tuberculosis infections. Here, we report the crystal structure of Rv2837c (2.0 Å), and its complex with hydrolysis intermediate 5'-pApA (2.35 Å). Our structures indicate that both DHH and DHHA1 domains are essential for c-di-AMP degradation. Further structural analysis shows that Rv2837c does not distinguish adenine from guanine, which explains why Rv2837c hydrolyzes all linear dinucleotides with almost the same efficiency. We observed that Rv2837c degraded other c-di-NMPs at a lower rate than it did on c-di-AMP. Nevertheless, our data also showed that Rv2837c significantly decreases concentrations of both c-di-AMP and c-di-GMP in vivo. Our results suggest that beside its major role in c-di-AMP degradation Rv2837c could also regulate c-di-GMP signaling pathways in bacterial cell.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , 3',5'-Cyclic-GMP Phosphodiesterases/metabolism , Bacterial Proteins/metabolism , Exoribonucleases/metabolism , Models, Molecular , Mycobacterium tuberculosis/enzymology , 3',5'-Cyclic-AMP Phosphodiesterases/chemistry , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , 3',5'-Cyclic-GMP Phosphodiesterases/chemistry , 3',5'-Cyclic-GMP Phosphodiesterases/genetics , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Biocatalysis , Catalytic Domain , Conserved Sequence , Cyclic AMP/analogs & derivatives , Cyclic AMP/chemistry , Cyclic AMP/metabolism , Cyclic GMP/analogs & derivatives , Cyclic GMP/chemistry , Cyclic GMP/metabolism , Dinucleoside Phosphates/chemistry , Dinucleoside Phosphates/metabolism , Exoribonucleases/chemistry , Exoribonucleases/genetics , Molecular Sequence Data , Mutation , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Alignment , Substrate Specificity
17.
Pharmacogenet Genomics ; 27(10): 356-362, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28727628

ABSTRACT

OBJECTIVE: Replication of associations in genome-wide association studies is desirable to ensure that such signals are potentially clinically meaningful. This study aimed to replicate associations of selected single-nucleotide polymorphisms (SNPs) with hypothyroidism and serum thyroid-stimulating hormone (TSH) using electronic medical records (EMRs). PATIENTS AND METHODS: A cross-sectional study was carried out among patients of European Caucasian ethnicity from the Genetics of Diabetes Audit and Research Tayside recruited in Tayside (Scotland, UK). EMRs (biochemistry, prescribing, hospital admissions and demographics) were used to ascertain patients with hypothyroidism and their controls as well as average serum TSH concentration, and linked to genetic biobank data. Genetic tests of association were performed using logistic and linear regression models. RESULTS: We analysed 1703 cases of hypothyroidism and 9457 controls. All four SNPs located on chromosome 9 at FOXE1 were associated with hypothyroidism with similar effect estimates (odds ratio=0.75-0.76, P<5e-08). Also, loci on chromosomes 1 (PTPN22), six (HLA-E/HLA-C) and 12 (SH2B3) were replicated. For serum TSH, we confirmed 12 SNPs previously reported at PDE8B, CAPZB, PDE10A, LOC105371356, NR3C2, VEGFA, IGFBP5, INSR, PRDM11, NFIA, ITPK1 and ABO. Overall, these SNPs accounted for 6.8% of the serum TSH variation (P<1e-04). CONCLUSION: EMRs linked to genomic data in large populations enable validation of genome-wide association studies discoveries without additional genotyping costs. Our replication confirmed at genome-wide significance the association of loci at FOXE1 with hypothyroidism, and PDE8B, CAPZB and PDE10A with serum TSH. A total of 12 SNPs seemed to explain nearly 7% of the serum TSH variation.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/genetics , CapZ Actin Capping Protein/genetics , Diabetes Mellitus/genetics , Forkhead Transcription Factors/genetics , Phosphoric Diester Hydrolases/genetics , Thyrotropin/blood , Case-Control Studies , Cross-Sectional Studies , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide
18.
EMBO Rep ; 16(4): 520-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25648146

ABSTRACT

Compartmentalized cAMP signaling regulates mitochondrial dynamics, morphology, and oxidative phosphorylation. However, regulators of the mitochondrial cAMP pathway, and its broad impact on organelle function, remain to be explored. Here, we report that Drosophila Prune is a cyclic nucleotide phosphodiesterase that localizes to the mitochondrial matrix. Knocking down prune in cultured cells reduces mitochondrial transcription factor A (TFAM) and mitochondrial DNA (mtDNA) levels. Our data suggest that Prune stabilizes TFAM and promotes mitochondrial DNA (mtDNA) replication through downregulation of mitochondrial cAMP signaling. In addition, our work demonstrates the prevalence of mitochondrial cAMP signaling in metazoan and its new role in mitochondrial biogenesis.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/genetics , DNA Replication , DNA, Mitochondrial/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Mitochondria/genetics , Transcription Factors/genetics , 3',5'-Cyclic-AMP Phosphodiesterases/metabolism , Animals , Cells, Cultured , Cyclic AMP/metabolism , DNA, Mitochondrial/metabolism , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/metabolism , Drosophila melanogaster/enzymology , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mitochondria/enzymology , Mitochondria/ultrastructure , Organelle Biogenesis , Oxidative Phosphorylation , Protein Stability , RNA, Double-Stranded/genetics , RNA, Double-Stranded/metabolism , Signal Transduction , Transcription Factors/metabolism , Red Fluorescent Protein
19.
J Endocrinol Invest ; 40(10): 1061-1067, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28382505

ABSTRACT

PURPOSE: Hashimoto's thyroiditis (HT) as a chronic autoimmune disease of the thyroid gland is the most common cause of hypothyroidism. Since HT and hypothyroidism are closely related, the main aim of this study was to explore the association of established hypothyroidism single-nucleotide polymorphisms (SNPs) with HT. METHODS: The case-control dataset included 200 HT cases and 304 controls. Diagnosis of HT cases was based on clinical examination, measurement of thyroid antibodies (TgAb, TPOAb), hormones (TSH and FT4) and ultrasound examination. We genotyped and analysed 11 known hypothyroidism-associated genetic variants. Case-control association analysis was performed in order to test each SNP for the association with HT using logistic regression model. Additionally, each SNP was tested for the association with thyroid-related quantitative traits (TPOAb levels, TgAb levels and thyroid volume) in HT cases only using linear regression. RESULTS: We identified two genetic variants nominally associated with HT rs3184504 in SH2B3 gene (P = 0.0135, OR = 0.74, 95% CI = 0.57-0.95) and rs4704397 in PDE8B gene (P = 0.0383, OR = 1.32, 95% CI = 1.01-1.74). The SH2B3 genetic variant also showed nominal association with TPOAb levels (P = 0.0163, ß = -0.46) and rs4979402 inside DFNB31 gene was nominally associated with TgAb levels (P = 0.0443, ß = 0.41). CONCLUSIONS: SH2B3 gene has previously been associated with susceptibility to several autoimmune diseases, whereas PDE8B has been associated with TSH levels and suggested to modulate thyroid physiology that may influence the manifestation of thyroid disease. Identified loci are novel and biologically plausible candidates for HT development and represent good basis for further exploration of HT susceptibility.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/genetics , Biomarkers/metabolism , Hashimoto Disease/genetics , Hypothyroidism/genetics , Polymorphism, Single Nucleotide , Proteins/genetics , Adaptor Proteins, Signal Transducing , Autoantibodies/blood , Case-Control Studies , Follow-Up Studies , Hashimoto Disease/complications , Hashimoto Disease/pathology , Humans , Hypothyroidism/etiology , Hypothyroidism/pathology , Intracellular Signaling Peptides and Proteins , Phenotype , Prognosis
20.
Proc Natl Acad Sci U S A ; 110(16): E1533-42, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23509299

ABSTRACT

V-raf-1 murine leukemia viral oncogene homolog 1 (Raf-1) is a key activator of the ERK pathway and is a target for cross-regulation of this pathway by the cAMP signaling system. The cAMP-activated protein kinase, PKA, inhibits Raf-1 by phosphorylation on S259. Here, we show that the cAMP-degrading phosphodiesterase-8A (PDE8A) associates with Raf-1 to protect it from inhibitory phosphorylation by PKA, thereby enhancing Raf-1's ability to stimulate ERK signaling. PDE8A binds to Raf-1 with high (picomolar) affinity. Mapping of the interaction domain on PDE8A using peptide array technology identified amino acids 454-465 as the main binding site, which could be disrupted by mutation. A cell-permeable peptide corresponding to this region disrupted the PDE8A/Raf-1 interaction in cells, thereby reducing ERK activation and the cellular response to EGF. Overexpression of a catalytically inactive PDE8A in cells displayed a dominant negative phenotype on ERK activation. These effects were recapitulated at the organism level in genetically modified (PDE8A(-/-)) mice. Similarly, PDE8 deletion in Drosophila melanogaster reduced basal ERK activation and sensitized flies to stress-induced death. We propose that PDE8A is a physiological regulator of Raf-1 signaling in some cells.


Subject(s)
3',5'-Cyclic-AMP Phosphodiesterases/metabolism , MAP Kinase Signaling System/physiology , Proto-Oncogene Proteins c-raf/metabolism , 3',5'-Cyclic-AMP Phosphodiesterases/genetics , Animals , Blotting, Western , DNA Primers/genetics , Drosophila melanogaster , Gene Deletion , HEK293 Cells , HeLa Cells , Humans , Immunoprecipitation , MAP Kinase Signaling System/genetics , Mass Spectrometry , Mice , Mice, Knockout , Mutagenesis, Site-Directed , Phosphorylation , Surface Plasmon Resonance
SELECTION OF CITATIONS
SEARCH DETAIL