Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
JCI Insight ; 7(23)2022 12 08.
Article in English | MEDLINE | ID: mdl-36301667

ABSTRACT

The folding and trafficking of transmembrane glycoproteins are essential for cellular homeostasis and are compromised in many diseases. In Niemann-Pick type C disease, a lysosomal disorder characterized by impaired intracellular cholesterol trafficking, the transmembrane glycoprotein NPC1 misfolds due to disease-causing missense mutations. While mutant NPC1 has emerged as a robust target for proteostasis modulators, drug development efforts have been unsuccessful in mouse models. Here, we demonstrated unexpected differences in trafficking through the medial Golgi between mouse and human I1061T-NPC1, a common disease-causing mutant. We established that these distinctions are governed by differences in the NPC1 protein sequence rather than by variations in the endoplasmic reticulum-folding environment. Moreover, we demonstrated direct effects of mutant protein trafficking on the response to small molecules that modulate the endoplasmic reticulum-folding environment by affecting Ca++ concentration. Finally, we developed a panel of isogenic human NPC1 iNeurons expressing WT, I1061T-, and R934L-NPC1 and demonstrated their utility in testing these candidate therapeutics. Our findings identify important rules governing mutant NPC1's response to proteostatic modulators and highlight the importance of species- and mutation-specific responses for therapy development.


Subject(s)
Niemann-Pick C1 Protein , Niemann-Pick Disease, Type C , Humans , Animals , Mice , Niemann-Pick Disease, Type C/drug therapy , Niemann-Pick Disease, Type C/genetics , Protein Transport
2.
Int J Mol Sci ; 22(5)2021 Mar 07.
Article in English | MEDLINE | ID: mdl-33799993

ABSTRACT

microRNAs (miRNAs) regulate messenger RNA (mRNA) abundance and translation during key developmental processes including muscle differentiation. Assessment of miRNA targets can provide insight into muscle biology and gene expression profiles altered by disease. mRNA and miRNA libraries were generated from C2C12 myoblasts during differentiation, and predicted miRNA targets were identified based on presence of miRNA binding sites and reciprocal expression. Seventeen miRNAs were differentially expressed at all time intervals (comparing days 0, 2, and 5) of differentiation. mRNA targets of differentially expressed miRNAs were enriched for functions related to calcium signaling and sarcomere formation. To evaluate this relationship in a disease state, we evaluated the miRNAs differentially expressed in human congenital myotonic dystrophy (CMD) myoblasts and compared with normal control. Seventy-four miRNAs were differentially expressed during healthy human myocyte maturation, of which only 12 were also up- or downregulated in CMD patient cells. The 62 miRNAs that were only differentially expressed in healthy cells were compared with differentiating C2C12 cells. Eighteen of the 62 were conserved in mouse and up- or down-regulated during mouse myoblast differentiation, and their C2C12 targets were enriched for functions related to muscle differentiation and contraction.


Subject(s)
MicroRNAs/genetics , Muscle, Skeletal/cytology , Myoblasts/cytology , Myotonic Dystrophy/genetics , RNA, Messenger/genetics , Animals , Calcium Signaling/genetics , Cell Differentiation , Cell Line , Genes, X-Linked , Humans , Mice , Myoblasts/physiology , Myotonic Dystrophy/pathology , Sarcomeres/genetics , Transcriptome
3.
Stem Cell Reports ; 9(3): 725-731, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28781079

ABSTRACT

Specifically ablating genes in human induced pluripotent stem cells (iPSCs) allows for studies of gene function as well as disease mechanisms in disorders caused by loss-of-function (LOF) mutations. While techniques exist for engineering such lines, we have developed and rigorously validated a method of simultaneous iPSC reprogramming while generating CRISPR/Cas9-dependent insertions/deletions (indels). This approach allows for the efficient and rapid formation of genetic LOF human disease cell models with isogenic controls. The rate of mutagenized lines was strikingly consistent across experiments targeting four different human epileptic encephalopathy genes and a metabolic enzyme-encoding gene, and was more efficient and consistent than using CRISPR gene editing of established iPSC lines. The ability of our streamlined method to reproducibly generate heterozygous and homozygous LOF iPSC lines with passage-matched isogenic controls in a single step provides for the rapid development of LOF disease models with ideal control lines, even in the absence of patient tissue.


Subject(s)
Cellular Reprogramming/genetics , Gene Editing , Induced Pluripotent Stem Cells/metabolism , Loss of Function Mutation/genetics , CRISPR-Cas Systems/genetics , Cell Line , Genetic Heterogeneity , Genomic Instability , Genotype , Humans , INDEL Mutation/genetics , RNA, Guide, Kinetoplastida/metabolism , Reproducibility of Results
4.
Cell Metab ; 23(3): 505-16, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26853750

ABSTRACT

Glucagon drives hepatic gluconeogenesis and maintains blood glucose levels during fasting. The mechanism that attenuates glucagon action following refeeding is not understood. The present study demonstrates an increase in perivenous liver hypoxia immediately after feeding, which stabilizes hypoxia-inducible factor 2α (HIF2α) in liver. The transient postprandial increase in hepatic HIF2α attenuates glucagon signaling. Hepatocyte-specific disruption of HIF2α increases postprandial blood glucose and potentiates the glucagon response. Independent of insulin/AKT signaling, activation of hepatic HIF2α resulted in lower blood glucose, improved glucose tolerance, and decreased gluconeogenesis due to blunted hepatic glucagon action. Mechanistically, HIF2α abrogated glucagon-PKA signaling by activating cAMP-phosphodiesterases in a MEK/ERK-dependent manner. Repression of glucagon signaling by HIF2α ameliorated hyperglycemia in streptozotocin-induced diabetes and acute insulin-resistant animal models. This study reveals that HIF2α is essential for the acute postprandial regulation of hepatic glucagon signaling and suggests HIF2α as a potential therapeutic target in the treatment of diabetes.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Glucagon/metabolism , Insulin/physiology , Liver/metabolism , Animals , Blood Glucose , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Experimental/metabolism , Gluconeogenesis , Hepatocytes/metabolism , Hyperglycemia , Mice, Inbred C57BL , Mice, Transgenic , Postprandial Period , Primary Cell Culture , Signal Transduction , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
5.
J Mol Neurosci ; 55(3): 684-705, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25189318

ABSTRACT

As members of the proneural basic-helix-loop-helix (bHLH) family of transcription factors, Ascl1 and Neurog2 direct the differentiation of specific populations of neurons at various times and locations within the developing nervous system. In order to characterize the mechanisms employed by these two bHLH factors, we generated stable, doxycycline-inducible lines of P19 embryonic carcinoma cells that express comparable levels of Ascl1 and Neurog2. Upon induction, both Ascl1 and Neurog2 directed morphological and immunocytochemical changes consistent with initiation of neuronal differentiation. Comparison of Ascl1- and Neurog2-regulated genes by microarray analyses showed both shared and distinct transcriptional changes for each bHLH protein. In both Ascl1- and Neurog2-differentiating cells, repression of Oct4 mRNA levels was accompanied by increased Oct4 promoter methylation. However, DNA demethylation was not detected for genes induced by either bHLH protein. Neurog2-induced genes included glutamatergic marker genes while Ascl1-induced genes included GABAergic marker genes. The Neurog2-specific induction of a gene encoding a protein phosphatase inhibitor, Ppp1r14a, was dependent on distinct, canonical E-box sequences within the Ppp1r14a promoter and the nucleotide sequences within these E-boxes were partially responsible for Neurog2-specific regulation. Our results illustrate multiple novel mechanisms by which Ascl1 and Neurog2 regulate gene repression during neuronal differentiation in P19 cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Nerve Tissue Proteins/metabolism , Neurogenesis , Promoter Regions, Genetic , Animals , Cell Line, Tumor , Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins , Mice , Muscle Proteins/genetics , Muscle Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Octamer Transcription Factor-3/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Transcription, Genetic
6.
J Neurochem ; 120(5): 667-83, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21623794

ABSTRACT

cAMP-dependent protein kinase (PKA) plays a critical role in nervous system development by modulating sonic hedgehog and bone morphogenetic protein signaling. In the current studies, P19 embryonic carcinoma cells were neuronally differentiated by expression of the proneural basic helix-loop-helix transcription factor Ascl1. After expression of Ascl1, but prior to expression of neuronal markers such as microtubule associated protein 2 and neuronal ß-tubulin, P19 cells demonstrated a large, transient increase in both mRNA and protein for the endogenous protein kinase inhibitor (PKI)ß. PKIß-targeted shRNA constructs both reduced the levels of PKIß expression and blocked the neuronal differentiation of P19 cells. This inhibition of differentiation was rescued by transfection of a shRNA-resistant expression vector for the PKIß protein, and this rescue required the PKA-specific inhibitory sequence of the PKIß protein. PKIß played a very specific role in the Ascl1-mediated differentiation process as other PKI isoforms were unable to rescue the deficit conferred by shRNA-mediated knockdown of PKIß. Our results define a novel requirement for PKIß and its inhibition of PKA during neuronal differentiation of P19 cells.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Neoplastic/physiology , Neurons/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , CREB-Binding Protein/metabolism , Carcinoma/pathology , Cell Differentiation/drug effects , Cell Line, Transformed , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Neurons/drug effects , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Signal Transduction/drug effects , Thionucleotides/pharmacology , Transfection , Tubulin/metabolism
7.
Dev Biol ; 361(1): 103-15, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22037235

ABSTRACT

Regulated proliferation and cell cycle exit are essential aspects of neurogenesis. The Yap transcriptional coactivator controls proliferation in a variety of tissues during development, and this activity is negatively regulated by kinases in the Hippo signaling pathway. We find that Yap is expressed in mitotic mouse retinal progenitors and it is downregulated during neuronal differentiation. Forced expression of Yap prolongs proliferation in the postnatal mouse retina, whereas inhibition of Yap by RNA interference (RNAi) decreases proliferation and increases differentiation. We show Yap is subject to post-translational inhibition in the retina, and also downregulated at the level of mRNA expression. Using a cell culture model, we find that expression of the proneural basic helix-loop-helix (bHLH) transcription factors Neurog2 or Ascl1 downregulates Yap mRNA levels, and simultaneously inhibits Yap protein via activation of the Lats1 and/or Lats2 kinases. Conversely, overexpression of Yap prevents proneural bHLH proteins from initiating cell cycle exit. We propose that mutual inhibition between proneural bHLH proteins and Yap is an important regulator of proliferation and cell cycle exit during mammalian neurogenesis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Neurogenesis/physiology , Neurons/cytology , Phosphoproteins/metabolism , Retina/cytology , Animals , Cell Cycle Proteins , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Gene Expression Regulation, Developmental/genetics , Mice , Nerve Tissue Proteins/metabolism , Neurons/physiology , RNA Interference , Stem Cells/metabolism , YAP-Signaling Proteins
8.
Methods Mol Biol ; 706: 41-52, 2011.
Article in English | MEDLINE | ID: mdl-21104053

ABSTRACT

Cell-based microarrays have been used for a wide variety of assays including gain-of-function, loss-of-function and compound screening. Many of these assays have employed fluorescent proteins as reporters. These fluorescent reporter proteins can be monitored in living cells but have low sensitivity of detection compared to enzymatic reporters. Here we have described a novel transcriptional reporter assay using the alkaline phosphatase reporter enzyme and a fluorescent substrate (ELF-97) to screen for gain-of-function mutations in the type-I cGMP-dependent protein kinase (PRKG1). We have identified a constitutively active mutant of this enzyme in which a conserved Glu at position 81 was mutated to Lys.


Subject(s)
Genes, Reporter , Organophosphorus Compounds , Quinazolinones , Tissue Array Analysis/methods , Alkaline Phosphatase/genetics , Amino Acid Substitution , Animals , COS Cells , Chlorocebus aethiops , Coloring Agents , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Feasibility Studies , HEK293 Cells , Humans , Indicators and Reagents , Lysine/metabolism , Substrate Specificity
9.
Mol Cell Neurosci ; 44(3): 282-96, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20382226

ABSTRACT

The basic helix-loop-helix transcription factor Ascl1 plays a critical role in the intrinsic genetic program responsible for neuronal differentiation. Here, we describe a novel model system of P19 embryonic carcinoma cells with doxycycline-inducible expression of Ascl1. Microarray hybridization and real-time PCR showed that these cells demonstrated increased expression of many neuronal proteins in a time- and concentration-dependent manner. Interestingly, the gene encoding the cell cycle regulator Gadd45gamma was increased earliest and to the greatest extent following Ascl1 induction. Here, we provide the first evidence identifying Gadd45gamma as a direct transcriptional target of Ascl1. Transactivation and chromatin immunoprecipitation assays identified two E-box consensus sites within the Gadd45gamma promoter necessary for Ascl1 regulation, and demonstrated that Ascl1 is bound to this region within the Gadd45gamma promoter. Furthermore, we found that overexpression of Gadd45gamma itself is sufficient to initiate some aspects of neuronal differentiation independent of Ascl1.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Gene Expression Regulation , Intracellular Signaling Peptides and Proteins/metabolism , Neurons/physiology , Transcription, Genetic , Animals , Anti-Bacterial Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Differentiation/drug effects , Doxycycline/pharmacology , Gene Expression Regulation/drug effects , Helix-Loop-Helix Motifs , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mice , Microarray Analysis , Neurons/cytology , Neurons/drug effects , Patch-Clamp Techniques , Promoter Regions, Genetic , Tumor Cells, Cultured , GADD45 Proteins
10.
Genomics ; 93(2): 159-68, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18955127

ABSTRACT

In an effort to define novel transcriptional regulatory elements, microarray cotransfection was used to functionally characterize conserved non-coding sequences (CNSs) of three immediate early genes: c-fos, JunB and EGR-1. Cotransfection of fluorescent CNS reporter constructs and expression vectors for constitutively active signaling proteins demonstrated that many of the CNSs alter both the basal and regulated expressions of reporter constructs, but the effects of these CNSs were usually specific for their homologous promoter. One CNS located in the first intron of the c-fos gene conferred regulation by cAMP-dependent protein kinase (PKA), cGMP-dependent protein kinase (PKG) and Raf. Mutagenesis and cotransfection experiments showed that PKA regulation of this c-fos intronic element was mediated by two adjacent CRE-like sequences and the transcription factor CREB. In the context of a reporter containing previously characterized regulatory elements, the novel intronic sequence contributed 50% of the transcriptional response to PKA. These studies suggest that microarray transfection studies may be useful in functional characterization of conserved genomic sequences on a larger scale.


Subject(s)
Genes, Immediate-Early/genetics , Genome, Human , Microarray Analysis , Transfection , Animals , Cell Line , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/metabolism , Exons , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/metabolism , Humans , Introns , Kidney/cytology , Luciferases, Firefly/metabolism , Mice , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism , Time Factors
11.
FASEB J ; 18(13): 1544-6, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15319368

ABSTRACT

Insulin-like growth factor-I (IGF-I) protects neurons of the peripheral nervous system from apoptosis, but the underlying signaling pathways are not well understood. We studied IGF-I mediated signaling in embryonic dorsal root ganglia (DRG) neurons. DRG neurons express IGF-I receptors (IGF-IR), and IGF-I activates the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. High glucose exposure induces apoptosis, which is inhibited by IGF-I through the PI3K/Akt pathway. IGF-I stimulation of the PI3K/Akt pathway phosphorylates three known Akt effectors: the survival transcription factor cyclic AMP response element binding protein (CREB) and the pro-apoptotic effector proteins glycogen synthase kinase-3beta (GSK-3beta) and forkhead (FKHR). IGF-I regulates survival at the nuclear level through accumulation of phospho-Akt in DRG neuronal nuclei, increased CREB-mediated transcription, and nuclear exclusion of FKHR. High glucose increases expression of the pro-apoptotic Bcl protein Bim (a transcriptional target of FKHR). However, IGF-I does not regulate Bim or anti-apoptotic Bcl-xL protein expression levels, which suggests that IGF-I neuroprotection is not through regulation of their expression. High glucose also induces loss of the initiator caspase-9 and increases caspase-3 cleavage, effects blocked by IGF-I. These data suggest that IGF-I prevents apoptosis in DRG neurons by regulating PI3K/Akt pathway effectors, including GSK-3beta, CREB, and FKHR, and by blocking caspase activation.


Subject(s)
Ganglia, Spinal/cytology , Insulin-Like Growth Factor I/pharmacology , Neurons/drug effects , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Apoptosis/drug effects , Caspase 3 , Caspase 9 , Caspases/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , DNA-Binding Proteins/metabolism , Forkhead Transcription Factors , Ganglia, Spinal/embryology , Glucose/antagonists & inhibitors , Glucose/pharmacology , Mitogen-Activated Protein Kinases/metabolism , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/enzymology , Neurons/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt , Rats , Signal Transduction/drug effects
12.
Mol Cell Proteomics ; 3(8): 770-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15118071

ABSTRACT

A wide variety of bioinformatic tools have been described to characterize potential transcriptional regulatory mechanisms based on genomic sequence analysis and microarray hybridization studies. However, these regulatory mechanisms are still experimentally verified using transient transfection methods. Current transfection methods are limited both by their large scale and by the low level of efficiency for certain cell types. Our goals were to develop a microarray-based transfection method that could be optimized for different cell types and that would be useful in reporter assays of transcriptional regulation. Here we describe a novel transfection method, termed STEP (surface transfection and expression protocol), which employs microarray-based DNA transfection of adherent cells in the functional analysis of transcriptional regulation. In STEP, recombinant proteins with biological activities designed to enhance transfection are complexed with expression vector DNAs prior to spotting on microscope slides. The recombinant proteins used in STEP complexes can be varied to increase the efficiency for different cell types. We demonstrate that STEP efficiently transfects both supercoiled plasmids and PCR-generated linear expression cassettes. A co-transfection assay using effector expression vectors encoding the cAMP-dependent protein kinase (PKA), as well as reporter vectors containing PKA-regulated promoters, showed that STEP transfection allows detection and quantitation of transcriptional regulation by this protein kinase. Because bioinformatic studies often result in the identification of many putative regulatory elements and signaling pathways, this approach should be of utility in high-throughput functional genomic studies of transcriptional regulation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Recombinant Proteins/genetics , Transfection/methods , Animals , Cell Line, Tumor , Gene Products, tat/genetics , Gene Products, tat/metabolism , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , Oligonucleotide Array Sequence Analysis/methods , Recombinant Proteins/metabolism , Transcription, Genetic , Tumor Cells, Cultured
13.
Mol Cell Biol ; 24(5): 2169-80, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14966294

ABSTRACT

Transforming growth factor beta (TGFbeta) interacts with cell surface receptors to initiate a signaling cascade critical in regulating growth, differentiation, and development of many cell types. TGFbeta signaling involves activation of Smad proteins which directly regulate target gene expression. Here we show that Smad proteins also regulate gene expression by using a previously unrecognized pathway involving direct interaction with protein kinase A (PKA). PKA has numerous effects on growth, differentiation, and apoptosis, and activation of PKA is generally initiated by increased cellular cyclic AMP (cAMP). However, we found that TGFbeta activates PKA independent of increased cAMP, and our observations support the conclusion that there is formation of a complex between Smad proteins and the regulatory subunit of PKA, with release of the catalytic subunit from the PKA holoenzyme. We also found that the activation of PKA was required for TGFbeta activation of CREB, induction of p21(Cip1), and inhibition of cell growth. Taken together, these data indicate an important and previously unrecognized interaction between the TGFbeta and PKA signaling pathways.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Signal Transduction/physiology , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cell Line , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/genetics , Enzyme Activation , Enzyme Inhibitors/metabolism , I-kappa B Proteins/metabolism , Macromolecular Substances , Male , Mice , Mice, Knockout , Pancreas/cytology , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Smad3 Protein , Smad4 Protein , Trans-Activators/genetics
14.
J Biol Chem ; 277(40): 37242-53, 2002 Oct 04.
Article in English | MEDLINE | ID: mdl-12093798

ABSTRACT

In the absence of cyclic nucleotides, the cAMP-dependent protein kinase and cGMP-dependent protein kinases (cGKs) suppress phosphotransfer activity at the catalytic cleft by competitive inhibition of substrate binding with a pseudosubstrate sequence within the holoenzyme. The magnitude of inhibition can be diminished by autophosphorylation near this pseudosubstrate sequence. Activation of type I cGK (cGKI) and type II cGK (cGKII) are differentially regulated by their cyclic nucleotide-binding sites. To address the possibility that the distinct activation mechanisms of cGKII and cGKI result from differences in the autophosphorylation of the inhibitory domain, we investigated the effects of autophosphorylation on the kinetics of activation. Unlike the type I cGKs (cGKIalpha and Ibeta), cGKII autophosphorylation did not alter the basal activity, nor the sensitivity of the enzyme to cyclic nucleotide activation. To determine residues responsible for autoinhibition of cGKII, Ala was substituted for basic residues (Lys(122), Arg(118), and Arg(119)) or a hydrophobic residue (Val(125)) within the putative pseudosubstrate domain of cGKII. The integrity of these residues was essential for full cGKII autoinhibition. Furthermore, a cGKII truncation mutant containing this autoinhibitory region demonstrated a nanomolar IC(50) toward a constitutively active form of cGKII. Finally, we present evidence that the dominant negative properties of this truncation mutant are specific to cGKII when compared with cAMP-dependent protein kinase Calpha and cGKIbeta. These findings extend the known differences in the activation mechanisms among cGK isoforms and allow the design of an isoform-specific cGKII inhibitor.


Subject(s)
Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Isoenzymes/antagonists & inhibitors , Alanine , Amino Acid Sequence , Binding Sites , Cell Line , Cyclic AMP/pharmacology , Cyclic GMP/pharmacology , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinase Type II , Cyclic GMP-Dependent Protein Kinases/chemistry , Cyclic GMP-Dependent Protein Kinases/genetics , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/chemistry , Kinetics , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Fragments/pharmacology , Phosphorylation , Polymerase Chain Reaction , Protein Conformation , Recombinant Proteins/antagonists & inhibitors , Transfection
15.
Am J Physiol Cell Physiol ; 282(1): C59-66, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11742798

ABSTRACT

The expression and function of the endogenous inhibitor of cAMP-dependent protein kinase (PKI) in endothelial cells are unknown. In this study, overexpression of rabbit muscle PKI gene into endothelial cells inhibited the cAMP-mediated increase and exacerbated thrombin-induced decrease in endothelial barrier function. We investigated PKI expression in human pulmonary artery (HPAECs), foreskin microvessel (HMECs), and brain microvessel endothelial cells (HBMECs). RT-PCR using specific primers for human PKI alpha, human PKI gamma, and mouse PKI beta sequences detected PKI alpha and PKI gamma mRNA in all three cell types. Sequencing and BLAST analysis indicated that forward and reverse DNA strands for PKI alpha and PKI gamma were of >96% identity with database sequences. RNase protection assays showed protection of the 542 nucleotides in HBMEC and HPAEC PKI alpha mRNA and 240 nucleotides in HBMEC, HPAEC, and HMEC PKI gamma mRNA. Western blot analysis indicated that PKI gamma protein was detected in all three cell types, whereas PKI alpha was found in HBMECs. In summary, endothelial cells from three different vascular beds express PKI alpha and PKI gamma, which may be physiologically important in endothelial barrier function.


Subject(s)
Carrier Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Endothelium, Vascular/enzymology , Intracellular Signaling Peptides and Proteins , Isoenzymes/genetics , Adaptor Proteins, Signal Transducing , Animals , Brain/blood supply , Capillary Permeability/physiology , Cells, Cultured , Cyclic AMP/metabolism , Dermis/blood supply , Endothelium, Vascular/cytology , Gene Expression/physiology , Gene Expression Regulation, Enzymologic/physiology , Humans , Muscle, Skeletal/enzymology , Pulmonary Artery/cytology , RNA, Messenger/analysis , Rabbits , Vascular Resistance/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...