Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 244: 125328, 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37307967

ABSTRACT

Diabetes is a major public health problem due to morbidity and mortality associated with end organ complications. Uptake of fatty acids by Fatty Acid Transport Protein-2 (FATP2) contributes to hyperglycemia, diabetic kidney and liver disease pathogenesis. Because FATP2 structure is unknown, a homology model was constructed, validated by AlphaFold2 prediction and site-directed mutagenesis, and then used to conduct a virtual drug discovery screen. In silico similarity searches to two low-micromolar IC50 FATP2 inhibitors, followed by docking and pharmacokinetics predictions, narrowed a diverse 800,000 compound library to 23 hits. These candidates were further evaluated for inhibition of FATP2-dependent fatty acid uptake and apoptosis in cells. Two compounds demonstrated nanomolar IC50, and were further characterized by molecular dynamic simulations. The results highlight the feasibility of combining a homology model with in silico and in vitro screening, to economically identify high affinity inhibitors of FATP2, as potential treatment for diabetes and its complications.


Subject(s)
Diabetes Complications , Diabetes Mellitus , Humans , Fatty Acids , Drug Discovery , Biological Transport , Fatty Acid Transport Proteins , Molecular Docking Simulation , Molecular Dynamics Simulation
2.
Sci Rep ; 11(1): 20570, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34663875

ABSTRACT

Chronic kidney disease (CKD) affects more than 20 million people in the US, and it is associated with a significantly increased risk of sudden cardiac death (SCD). Despite the significance, the mechanistic relationship between SCD and CKD is not clear and there are few effective therapies. Using optical mapping techniques, we tested the hypothesis that mouse models of progressive diabetic kidney disease (DKD) exhibit enhanced ventricular arrhythmia incidence and underlying arrhythmia substrates. Compared to wild-type mice, both Leprdb/db eNOS-/- (2KO) and high fat diet plus low dose streptozotocin (HFD + STZ) mouse models of DKD experienced sudden death and greater arrhythmia inducibility, which was more common with isoproterenol than programmed electrical stimulation. 2KO mice demonstrated slowed conduction velocity, prolonged action potential duration (APD), and myocardial fibrosis; both 2KO and HFD + STZ mice exhibited arrhythmias and calcium dysregulation with isoproterenol challenge. Finally, circulating concentrations of the uremic toxin asymmetric dimethylarginine (ADMA) were elevated in 2KO mice. Incubation of human cardiac myocytes with ADMA prolonged APD, as also observed in 2KO mice hearts ex vivo. The present study elucidates an arrhythmia-associated mechanism of sudden death associated with DKD, which may lead to more effective treatments in the vulnerable DKD patient population.


Subject(s)
Arrhythmias, Cardiac/physiopathology , Diabetic Nephropathies/physiopathology , Action Potentials/physiology , Animals , Arrhythmias, Cardiac/pathology , Diabetes Complications/physiopathology , Diabetes Mellitus/physiopathology , Diabetic Nephropathies/pathology , Disease Models, Animal , Heart Rate/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Tachycardia, Ventricular/pathology , Tachycardia, Ventricular/physiopathology , Voltage-Sensitive Dye Imaging/methods
3.
JCI Insight ; 5(15)2020 08 06.
Article in English | MEDLINE | ID: mdl-32614804

ABSTRACT

Kidney disease is one of the most devastating complications of diabetes, and tubular atrophy predicts diabetic kidney disease (DKD) progression to end-stage renal disease. We have proposed that fatty acids bound to albumin contribute to tubular atrophy by inducing lipotoxicity, after filtration across damaged glomeruli, and subsequent proximal tubule reabsorption by a fatty acid transport protein-2-dependent (FATP2-dependent) mechanism. To address this possibility, genetic (Leprdb/db eNOS-/-) and induced (high-fat diet plus low-dose streptozotocin) mouse models of obesity and DKD were bred with global FATP2 gene-deleted mice (Slc27a2) and then phenotyped. DKD-prone mice with the Slc27a2-/- genotype demonstrated normalization of glomerular filtration rate, reduced albuminuria, improved kidney histopathology, and longer life span compared with diabetic Slc27a2+/+ mice. Genetic and induced DKD-prone Slc27a2-/- mice also exhibited markedly reduced fasting plasma glucose, with mean values approaching euglycemia, despite increased obesity and decreased physical activity. Glucose lowering in DKD-prone Slc27a2-/- mice was accompanied by ß cell hyperplasia and sustained insulin secretion. Together, our data indicate that FATP2 regulates DKD pathogenesis by a combined lipotoxicity and glucotoxicity (glucolipotoxicity) mechanism.


Subject(s)
Coenzyme A Ligases/physiology , Diabetes Mellitus, Experimental/complications , Diabetic Nephropathies/prevention & control , Glycemic Control , Nitric Oxide Synthase Type III/physiology , Receptors, Leptin/physiology , Albuminuria , Animals , Biomarkers/analysis , Diabetic Nephropathies/etiology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Disease Progression , Female , Glomerular Filtration Rate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese
4.
Invest Ophthalmol Vis Sci ; 58(1): 448-460, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28118669

ABSTRACT

Purpose: Joubert syndrome (JBTS) is an autosomal recessive ciliopathy with considerable phenotypic variability. In addition to central nervous system abnormalities, a subset of JBTS patients exhibit retinal dystrophy and/or kidney disease. Mutations in the AHI1 gene are causative for approximately 10% of all JBTS cases. The purpose of this study was to generate ahi1 mutant alleles in zebrafish and to characterize the retinal phenotypes. Methods: Zebrafish ahi1 mutants were generated using transcription activator-like effector nucleases (TALENs). Expression analysis was performed by whole-mount in situ hybridization. Anatomic and molecular characterization of photoreceptors was investigated by histology, electron microscopy, and immunohistochemistry. The optokinetic response (OKR) behavior assay was used to assess visual function. Kidney cilia were evaluated by whole-mount immunostaining. Results: The ahi1lri46 mutation in zebrafish resulted in shorter cone outer segments but did not affect visual behavior at 5 days after fertilization (dpf). No defects in rod morphology or rhodopsin localization were observed at 5 dpf. By 5 months of age, cone degeneration and rhodopsin mislocalization in rod photoreceptors was observed. The connecting cilium formed normally and Cc2d2a and Cep290 localized properly. Distal pronephric duct cilia were absent in mutant fish; however, only 9% of ahi1 mutants had kidney cysts by 5 dpf, suggesting that the pronephros remained largely functional. Conclusions: The results indicate that Ahi1 is required for photoreceptor disc morphogenesis and outer segment maintenance in zebrafish.


Subject(s)
Carrier Proteins/genetics , Cilia/ultrastructure , Ciliopathies/genetics , Morphogenesis , Mutation , Retinal Photoreceptor Cell Outer Segment/ultrastructure , Zebrafish Proteins/genetics , Animals , Carrier Proteins/metabolism , Cell Survival , Cilia/metabolism , Ciliopathies/metabolism , Ciliopathies/pathology , DNA Mutational Analysis , Genotype , Immunohistochemistry , In Situ Hybridization , Microscopy, Electron, Transmission , Proto-Oncogene Proteins , Retinal Photoreceptor Cell Outer Segment/metabolism , Zebrafish , Zebrafish Proteins/metabolism
5.
Invest Ophthalmol Vis Sci ; 57(10): 4517-26, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27571019

ABSTRACT

PURPOSE: Mutations in the gene ARL13B cause the classical form of Joubert syndrome, an autosomal recessive ciliopathy with variable degrees of retinal degeneration. As second-site modifier alleles can contribute to retinal pathology in ciliopathies, animal models provide a unique platform to test how genetic interactions modulate specific phenotypes. In this study, we analyzed the zebrafish arl13b mutant for retinal degeneration and for epistatic relationships with the planar cell polarity protein (PCP) component vangl2. METHODS: Photoreceptor and cilia structure was examined by light and electron microscopy. Immunohistochemistry was performed to examine ciliary markers. Genetic interactions were tested by pairwise crosses of heterozygous animals. Genetic mosaic animals were generated by blastula transplantation and analyzed by fluorescence microscopy. RESULTS: At 5 days after fertilization, photoreceptor outer segments were shorter in zebrafish arl13b-/- mutants compared to wild-type larvae, no overt signs of retinal degeneration were observed by light or electron microscopy. Starting at 14 days after fertilization (dpf) and continuing through 30 dpf, cells lacking Arl13b died following transplantation into wild-type host animals. Photoreceptors of arl13b-/-;vangl2-/- mutants were more compromised than the photoreceptors of single mutants. Finally, when grown within a wild-type retina, the vangl2-/- mutant cone photoreceptors displayed normal basal body positioning. CONCLUSIONS: We show that arl13b-/- mutants have shortened cilia and photoreceptor outer segments and exhibit a slow, progressive photoreceptor degeneration that occurs over weeks. The data suggest that loss of Arl13b leads to slow photoreceptor degeneration, but can be exacerbated by the loss of vangl2. Importantly, the data show that Arl13b can genetically and physically interact with Vangl2 and this association is important for normal photoreceptor structure. The loss of vangl2, however, does not affect basal body positioning.


Subject(s)
ADP-Ribosylation Factors/genetics , Cilia/metabolism , Mutation , Retinal Degeneration/diagnosis , Retinal Photoreceptor Cell Outer Segment/metabolism , Zebrafish Proteins/genetics , ADP-Ribosylation Factors/metabolism , Animals , Blotting, Western , Cells, Cultured , Cilia/ultrastructure , DNA , DNA Mutational Analysis , Disease Models, Animal , Immunohistochemistry , Immunoprecipitation , In Situ Hybridization , Larva , Microscopy, Electron , Retinal Degeneration/metabolism , Retinal Photoreceptor Cell Outer Segment/ultrastructure , Zebrafish , Zebrafish Proteins/metabolism
6.
Invest Ophthalmol Vis Sci ; 57(7): 2942-54, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27273592

ABSTRACT

PURPOSE: Tail-anchored (TA) proteins contain a single hydrophobic domain at the C-terminus and are posttranslationally inserted into the ER membrane via the GET (guided entry of tail-anchored proteins) pathway. The role of the GET pathway in photoreceptors is unexplored. The goal of this study was to characterize the zebrafish pinball wizard mutant, which disrupts Wrb, a core component of the GET pathway. METHODS: Electroretinography, optokinetic response measurements (OKR), immunohistochemistry, and electron microscopy analyses were employed to assess ribbon synapse function, protein expression, and ultrastructure in 5-day-old zebrafish larvae. Expression of wrb was investigated with real-time qRT-PCR and in situ hybridization. RESULTS: Mutation of wrb abolished the OKR and greatly diminished the ERG b-wave, but not the a-wave. Ribeye and SV2 were partially mislocalized in both photoreceptors and hair cells of wrb mutants. Fewer contacts were seen between photoreceptors and bipolar cells in wrb-/- mutants. Expression of wrb was observed throughout the nervous system and Wrb localized to the ER and synaptic region of photoreceptors. Morpholino knockdown of the cytosolic ATPase trc40, which targets TA proteins to the ER, also diminished the OKR. Overexpression of wrb fully restored contrast sensitivity in mutants, while overexpression of mutant wrbR73A, which cannot bind Trc40, did not. CONCLUSIONS: Proteins Wrb and Trc40 are required for synaptic transmission between photoreceptors and bipolar cells, indicating that TA protein insertion by the TRC pathway is a critical step in ribbon synapse assembly and function.


Subject(s)
Mutation , Nuclear Proteins/genetics , Photoreceptor Cells/physiology , Protein Transport/physiology , Synaptic Transmission/physiology , Animals , Electroretinography , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Protein Processing, Post-Translational/physiology , Qa-SNARE Proteins/genetics , Qa-SNARE Proteins/metabolism , Synapses/metabolism , Synapses/pathology , Synapses/physiology , Zebrafish , Zebrafish Proteins/genetics
7.
Biochem Biophys Res Commun ; 425(1): 25-32, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22820184

ABSTRACT

SH3BP2 activating mutations lead to an unique clinical condition in which patients develop symmetrical bone resorptive lesions of the jaw, a condition termed cherubism. Due to this specific temporal sequence and location of bone resorption, we investigated the transcriptional regulation of SH3BP2 expression. Analyses of 5'- and 3'-serial promoter deletions defined the core promoter/regulatory elements, including two repressor sites (from -1,200 to -1,000 and from +86 to +115, respectively) and two activator sites (a PARP1 binding site from -44 to -21 and a second activator site from +57 to +86). We identified that PARP1 binds to DNA from -44 to -21 by Streptavidin-biotin purification and confirmed this binding by electrophoretic mobility shift assay (EMSA). Mutagenesis of the PARP1 binding site on the SH3BP2 promoter showed that this binding site is essential for SH3BP2 expression. EMSA and chromatin immunoprecipitation (ChIP) assays confirmed that PARP1 was able to bind to the SH3BP2 promoter in vitro and in vivo. Indeed, knockout of Parp1 in mice BMMs reduced expression of SH3BP2. These results demonstrate that PARP1 regulates expression of SH3BP2.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Poly(ADP-ribose) Polymerases/metabolism , Promoter Regions, Genetic , Transcriptional Activation , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Base Sequence , Binding Sites , Cell Line , Chromatin Immunoprecipitation , Cloning, Molecular , Electrophoretic Mobility Shift Assay , Humans , Mice , Molecular Sequence Data , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/genetics , Transcription Initiation Site , Transcription, Genetic
8.
J Orthop Res ; 29(10): 1521-7, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21448930

ABSTRACT

Germline mutations in SH3BP2 gene have been identified in patients with cherubism, a skeletal disorder characterized by excessive osteoclastic bone resorption that is limited to the mandible and maxilla. We previously demonstrated that SH3BP2 overexpression in Raw264.7 cells increased RANKL-induced osteoclastogenesis. Here, we examine the effect of decreased SH3BP2 on osteoclastogenesis. shRNA knockdown of SH3BP2 decreased PLCγ2 phosphorylation and NFATc1 expression, and reduced the expression of osteoclast-specific genes. In BMMs knockdown of SH3BP2 led to reductions in both the number and the surface area of TRAP positive and multinucleated osteoclasts. Bone resorptive activity was also dramatically blocked by shRNA knockdown of SH3BP2. Similarly Sh3bp2(-/-) deficient mice BMMs formed smaller osteoclasts that stained less with TRAP than wild-type mice. Taken together, this study demonstrates that SH3BP2 knockdown significantly decreases osteoclast differentiation and function. These results suggest that SH3BP2 plays a critical role in osteoclastogenesis and is a potential target for suppression of pathologic bone resorption.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Cell Differentiation , Macrophages/physiology , Osteoclasts/cytology , Acid Phosphatase/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Bone Resorption , Cell Line , Gene Knockdown Techniques , Isoenzymes/metabolism , Mice , Mice, Knockout , NFATC Transcription Factors/metabolism , Phospholipase C gamma/metabolism , Phosphorylation , RANK Ligand/metabolism , RNA, Small Interfering , Tartrate-Resistant Acid Phosphatase
9.
Mol Pharmacol ; 76(2): 314-26, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19487244

ABSTRACT

The understanding of the function of alpha(1)-adrenergic receptors in the brain has been limited due to a lack of specific ligands and antibodies. We circumvented this problem by using transgenic mice engineered to overexpress either wild-type receptor tagged with enhanced green fluorescent protein or constitutively active mutant alpha(1)-adrenergic receptor subtypes in tissues in which they are normally expressed. We identified intriguing alpha(1A)-adrenergic receptor subtype-expressing cells with a migratory morphology in the adult subventricular zone that coexpressed markers of neural stem cell and/or progenitors. Incorporation of 5-bromo-2-deoxyuridine in vivo increased in neurogenic areas in adult alpha(1A)-adrenergic receptor transgenic mice or normal mice given the alpha(1A)-adrenergic receptor-selective agonist, cirazoline. Neonatal neurospheres isolated from normal mice expressed a mixture of alpha(1)-adrenergic receptor subtypes, and stimulation of these receptors resulted in increased expression of the alpha(1B)-adrenergic receptor subtype, proneural basic helix-loop-helix transcription factors, and the differentiation and migration of neuronal progenitors for catecholaminergic neurons and interneurons. alpha(1)-Adrenergic receptor stimulation increased the apoptosis of astrocytes and regulated survival of neonatal neurons through phosphatidylinositol 3-kinase signaling. However, in adult normal neurospheres, alpha(1)-adrenergic receptor stimulation increased the expression of glial markers at the expense of neuronal differentiation. In vivo, S100-positive glial and betaIII tubulin neuronal progenitors colocalized with either alpha(1)-adrenergic receptor subtype in the olfactory bulb. Our results indicate that alpha(1)-adrenergic receptors can regulate both neurogenesis and gliogenesis that may be developmentally dependent. Our findings may lead to new therapies to treat neurodegenerative diseases.


Subject(s)
Neurogenesis , Neuroglia/metabolism , Neurons/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Adrenergic alpha-1 Receptor Agonists , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Movement/genetics , Cell Movement/physiology , Green Fluorescent Proteins/metabolism , Imidazoles/pharmacology , Immunohistochemistry , Interneurons/cytology , Interneurons/metabolism , Mice , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Adrenergic, alpha-1/genetics , Spheroids, Cellular/metabolism
10.
J Recept Signal Transduct Res ; 27(1): 27-45, 2007.
Article in English | MEDLINE | ID: mdl-17365508

ABSTRACT

The function and distribution of alpha1-adrenergic receptor (AR) subtypes in prostate cancer cells is well characterized. Previous studies have used RNA localization or low-avidity antibodies in tissue or cell lines to determine the alpha1-AR subtype and suggested that the alpha1A-AR is dominant. Two androgen-insensitive, human metastatic cancer cell lines DU145 and PC3 were used as well as the mouse TRAMP C1-C3 primary and clonal cell lines. The density of alpha1-ARs was determined by saturation binding and the distribution of the different alpha1-AR subtypes was examined by competition-binding experiments. In contrast to previous studies, the major alpha1-AR subtype in DU145, PC3 and all of the TRAMP cell lines is the alpha1B-AR. DU145 cells contained 100% of the alpha1B-AR subtype, whereas PC3 cells were composed of 21% alpha1 A-AR and 79% alpha1B-AR. TRAMP cell lines contained between 66% and 79% of the alpha1B-AR with minor fractions of the other two subtypes. Faster doubling time in the TRAMP cell lines correlated with decreasing alpha 1B-AR and increasing alpha1 A- and alpha1D-AR densities. Transfection with EGFP-tagged alpha1B-ARs revealed that localization was mainly intracellular, but the majority of the receptors translocated to the cell surface after extended preincubation (18 hr) with either agonist or antagonist. Localization was confirmed by ligand-binding studies and inositol phosphate assays where prolonged preincubation with either agonist and/or antagonist increased the density and function of alpha 1-ARs, suggesting that the native receptors were mostly intracellular and nonfunctional. Our studies indicate that alpha1B-ARs are the major alpha1-AR subtype expressed in DU145, PC3, and all TRAMP cell lines, but most of the receptor is localized in intracellular compartments in a nonfunctional state, which can be rescued upon prolonged incubation with any ligand.


Subject(s)
Prostatic Neoplasms/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Adrenergic Agonists/pharmacology , Adrenergic alpha-Antagonists/pharmacokinetics , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Epinephrine/pharmacology , Humans , Male , Mice , Piperazines/pharmacokinetics , Protein Binding
11.
Am J Physiol Cell Physiol ; 291(5): C946-56, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16760267

ABSTRACT

Transactivation of EGF receptors by G protein-coupled receptors is a well-known phenomenon. This process involves the ectodomain shedding of growth factors in the EGF family by matrix metalloproteinases. However, many of these studies employ transformed and/or cultured cells that overexpress labeled growth factors. In addition, few studies have shown that EGF itself is the growth factor that is shed and is responsible for transactivation of the EGF receptor. In this study, we show that freshly isolated, nontransformed lacrimal gland acini express two of the three known alpha(1)-adrenergic receptors (ARs), namely, alpha(1B)- and alpha(1D)-ARs. Alpha(1D)-ARs mediate phenylephrine (an alpha(1)-adrenergic agonist)-induced protein secretion and activation of p42/p44 MAPK, because the alpha(1D)-AR inhibitor BMY-7378, but not the alpha(1A)-AR inhibitor 5-methylurapidil, inhibits these processes. Activation of p42/p44 MAPK occurs through transactivation of the EGF receptor, which is inhibited by the matrix metalloproteinase ADAM17 inhibitor TAPI-1. In addition, phenylephrine caused the shedding of EGF from freshly isolated acini into the buffer. Incubation of freshly isolated cells with conditioned buffer from cells treated with phenylephrine resulted in activation of the EGF receptor and p42/p44 MAPK. The EGF receptor inhibitor AG1478 and an EGF-neutralizing antibody blocked this activation of p42/p44 MAPK. We conclude that in freshly isolated lacrimal gland acini, alpha(1)-adrenergic agonists activate the alpha(1D)-AR to stimulate protein secretion and the ectodomain shedding of EGF to transactivate the EGF receptor, potentially via ADAM17, which activates p42/p44 MAPK to negatively modulate protein secretion.


Subject(s)
Cell Separation/methods , Epidermal Growth Factor/metabolism , Epithelial Cells/metabolism , Lacrimal Apparatus/cytology , Receptors, Adrenergic, alpha-1/metabolism , Adrenergic alpha-1 Receptor Agonists , Adrenergic alpha-1 Receptor Antagonists , Animals , Binding, Competitive/drug effects , Culture Media, Conditioned , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Epithelial Cells/drug effects , ErbB Receptors/metabolism , Male , Matrix Metalloproteinase Inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Phenylephrine/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha-1/genetics , Transcriptional Activation/drug effects
12.
J Comp Neurol ; 497(2): 209-22, 2006 Jul 10.
Article in English | MEDLINE | ID: mdl-16705673

ABSTRACT

alpha(1)-Adrenergic receptors (ARs) are not well defined in the central nervous system. The particular cell types and areas that express these receptors are uncertain because of the lack of high avidity antibodies and selective ligands. We have developed transgenic mice that either systemically overexpress the human alpha(1A)-AR subtype fused with the enhanced green fluorescent protein (EGFP) or express the EGFP protein alone under the control of the mouse alpha(1A)-AR promoter. We confirm our transgenic model against the alpha(1A)-AR knockout mouse, which expresses the LacZ gene in place of the coding region for the alpha(1A)-AR. By using these models, we have now determined cellular localization of the alpha(1A)-AR in the brain, at the protein level. The alpha(1A)-AR or the EGFP protein is expressed prominently in neuronal cells in the cerebral cortex, hippocampus, hypothalamus, midbrain, pontine olivary nuclei, trigeminal nuclei, cerebellum, and spinal cord. The types of neurons were diverse, and the alpha(1A)-AR colocalized with markers for glutamic acid decarboxylase (GAD), gamma-aminobutyric acid (GABA), and N-methyl-D-aspartate (NMDA) receptors. Recordings from alpha(1A)-AR EGFP-expressing cells in the stratum oriens of the hippocampal CA1 region confirmed that these cells were interneurons. We could not detect expression of the alpha(1A)-AR in mature astrocytes, oligodendrocytes, or cerebral blood vessels, but we could detect the alpha(1A)-AR in oligodendrocyte progenitors. We conclude that the alpha(1A)-AR is abundant in the brain, expressed in various types of neurons, and may regulate the function of oligodendrocyte progenitors, interneurons, GABA, and NMDA receptor containing neurons.


Subject(s)
Antigens/metabolism , Brain/cytology , Neurons/physiology , Oligodendroglia/metabolism , Proteoglycans/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Stem Cells , gamma-Aminobutyric Acid/metabolism , Adrenergic alpha-1 Receptor Agonists , Animals , Brain/metabolism , Cell Differentiation/physiology , Gene Expression/physiology , Green Fluorescent Proteins/metabolism , Immunohistochemistry/methods , In Vitro Techniques , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Transgenic , Neurons/classification , Neurons/drug effects , Norepinephrine/analogs & derivatives , Norepinephrine/pharmacology , Patch-Clamp Techniques/methods , Radioligand Assay/methods , Receptors, Adrenergic, alpha-1/deficiency , beta-Galactosidase/metabolism
13.
Mol Pharmacol ; 70(1): 129-42, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16617165

ABSTRACT

alpha1-Adrenergic receptor (alpha1-ARs) subtypes (alpha1A, alpha1B, and alpha1D) regulate multiple signal pathways, such as phospholipase C, protein kinase C (PKC), and mitogen-activated protein kinases. We employed oligonucleotide microarray technology to explore the effects of both short- (1 h) and long-term (18 h) activation of the alpha1A-AR to enable RNA changes to occur downstream of earlier well characterized signaling pathways, promoting novel couplings. Polymerase chain reaction (PCR) studies confirmed that PKC was a critical regulator of alpha1A-AR-mediated gene expression, and secreted interleukin (IL)-6 also contributed to gene expression alterations. We next focused on two novel signaling pathways that might be mediated through alpha1A-AR stimulation because of the clustering of gene expression changes for cell adhesion/motility (syndecan-4 and tenascin-C) and hyaluronan (HA) signaling. We confirmed that alpha1-ARs induced adhesion in three cell types to vitronectin, an interaction that was also integrin-, FGF7-, and PKC-dependent. alpha1-AR activation also inhibited cell migration, which was integrin- and PKC-independent but still required secretion of FGF7. alpha1-AR activation also increased the expression and deposition of HA, a glycosaminoglycan, which displayed two distinct structures: pericellular coats and long cable structures, as well as increasing expression of the HA receptor, CD44. Long cable structures of HA can bind leukocytes, which this suggests that alpha1-ARs may be involved in proinflammatory responses. Our results indicate alpha1-ARs induce the secretion of factors that interact with the extracellular matrix to regulate cell adhesion, motility and proinflammatory responses through novel signaling pathways.


Subject(s)
Receptors, Adrenergic, alpha-1/physiology , Signal Transduction/physiology , Adrenergic alpha-1 Receptor Antagonists , Adrenergic alpha-Antagonists/pharmacology , Alkaloids , Animals , Benzophenanthridines , Blotting, Western , Cell Adhesion/drug effects , Cell Adhesion/genetics , Cell Line, Transformed , Cell Movement/drug effects , Cell Movement/genetics , Extracellular Matrix/metabolism , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibroblast Growth Factor 7/genetics , Fibroblast Growth Factor 7/metabolism , Gene Expression Profiling , Interleukin-6/genetics , Interleukin-6/metabolism , Models, Biological , Oligonucleotide Array Sequence Analysis/methods , Phenanthridines/pharmacology , Prazosin/pharmacology , Protein Binding , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Rats , Receptors, Adrenergic, alpha-1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Time Factors , Vitronectin/genetics , Vitronectin/metabolism
14.
J Mol Cell Cardiol ; 39(5): 777-84, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16171811

ABSTRACT

Adrenergic receptors (ARs) play an important role in the regulation of cardiac function. Cardiac inotropy is primarily regulated by beta(1)-ARs. However, alpha(1)-ARs may play an important role in inotropy during heart failure. Previous work has suggested that the alpha(1B)-AR modulates beta(1)-AR function in the heart. The potential role of the alpha(1A)-AR has not been previously studied. We used transgenic mice that express constitutively active mutant (CAM) forms of the alpha(1A)-AR or alpha(1B)-AR regulated by their endogenous promoters. Expression of the CAM alpha(1A)-AR or CAM alpha(1B)-AR had no effect on basal cardiac function (developed pressure, +dP/dT, -dP/dT, heart rate, flow rate). However, both alpha(1)-AR subtypes significantly decreased isoproterenol-stimulated +dP/dT. Pertussis toxin had no effect on +dP/dT in CAM alpha(1A)-AR hearts but restored +dP/dT to non-transgenic values in CAM alpha(1B)-AR hearts. Radioligand binding indicated a selective decrease in the density of beta(1)-ARs in both CAM mice. However, G-proteins, cAMP, or the percentage of high and low affinity states were unchanged in either transgenic compared with control. These data demonstrate that CAM alpha(1A)- and alpha(1B)-ARs both down regulate beta(1)-AR-mediated inotropy in the mouse heart. However, alpha(1)-AR subtypes are coupled to different beta-AR mediated signaling pathways with the alpha(1B)-AR being pertussis toxin sensitive.


Subject(s)
Down-Regulation , Myocardium/metabolism , Pertussis Toxin/pharmacology , Receptor Cross-Talk/physiology , Receptors, Adrenergic, alpha-1/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction/drug effects , Animals , Cardiotonic Agents , Heterotrimeric GTP-Binding Proteins/metabolism , Isoproterenol/pharmacology , Mice , Mice, Transgenic , Mutation/genetics , Receptor Cross-Talk/drug effects , Receptors, Adrenergic, alpha-1/genetics , Receptors, Adrenergic, beta/genetics
15.
Cardiovasc Res ; 65(2): 436-45, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15639483

ABSTRACT

OBJECTIVE: Brief periods of ischemia stimulate an endogenous mechanism in the heart that protects the myocardium from subsequent ischemic injury. alpha1-Adrenergic receptors (ARs) have been implicated in this process. However, the lack of sufficiently selective antagonists has made it difficult to determine which alpha1-AR subtype protects the heart from ischemic injury. The goal of this study was to identify the alpha1-AR subtype that is involved in ischemic preconditioning. METHODS: We developed transgenic mice that express constitutively active mutant (CAM) forms of the alpha1A-AR or the alpha1B-AR regulated by their endogenous promoters. Hearts isolated from transgenic and non-transgenic mice were perfused by the Langendorff method using an ischemic preconditioning perfusion protocol or a non-preconditioning perfusion protocol prior to 30-min ischemia and 40-min reperfusion. Contractile function was continuously monitored through an intraventricular balloon. RESULTS: The contractile function of non-transgenic hearts perfused according to the ischemic preconditioning protocol completely recovered from 30-min ischemia. However, non-transgenic hearts perfused according to the non-preconditioning protocol recovered only 60% of their contractile function. The contractile function of CAM alpha1A-AR hearts, but not CAM alpha1B-AR hearts, completely recovered from 30-min ischemia even though they were perfused according to the non-preconditioning protocol. Thus, CAM alpha1A-AR hearts, but not CAM alpha1B-AR hearts, were inherently preconditioned against ischemic injury. Staurosporine, but not chelerythrine, completely reversed the preconditioning effect of CAM alpha1A-ARs. CONCLUSIONS: These data demonstrate that alpha1A-ARs protect the heart from ischemic injury through a staurosporine-sensitive signaling pathway that is independent of protein kinase C.


Subject(s)
Ischemic Preconditioning, Myocardial , Myocardial Ischemia/metabolism , Myocardium/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Staurosporine/pharmacology , Alkaloids , Animals , Benzophenanthridines , Enzyme Inhibitors/pharmacology , Female , Male , Mice , Mice, Transgenic , Myocardial Contraction , Perfusion , Phenanthridines/pharmacology , Protein Kinase C/antagonists & inhibitors
16.
Recept Channels ; 10(3-4): 109-16, 2004.
Article in English | MEDLINE | ID: mdl-15512845

ABSTRACT

The alpha1A-adrenergic receptor (AR) has a higher affinity for several agonists and antagonists compared to alpha1B or alpha1D ARs. Mutagenesis studies were used to determine residues potentially responsible for this subtype selectivity. Oxymetazoline has a 50-fold lower affinity for alpha1D ARs compared to alpha1A ARs and also displayed a significant loss of affinity for an alpha1A Leu-290 to Phe mutant. It was concluded that steric interactions between the alpha1D ARs Phe-360 and the bulkytert-butyl group of oxymetazoline partially accounts for this lower affinity. Thus, the alpha1A AR binding pocket may more easily accommodate bulk at the paraposition of the phenyl ring than the alpha1D AR.


Subject(s)
Oxymetazoline/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Adrenergic alpha-1 Receptor Agonists , Adrenergic alpha-1 Receptor Antagonists , Amino Acid Sequence , Animals , COS Cells , Chlorocebus aethiops , Models, Molecular , Molecular Sequence Data , Mutation , Rats , Receptors, Adrenergic, alpha-1/genetics , Sequence Alignment
17.
J Comp Neurol ; 478(1): 1-10, 2004 Oct 04.
Article in English | MEDLINE | ID: mdl-15334645

ABSTRACT

alpha1-Adrenergic receptors (ARs) are well-known mediators of the sympathetic nervous system, are highly abundant in the brain, but are the least understood in the central nervous system. The particular cell types in the brain that contain these receptors or their functions are not known because of the lack of high avidity antibodies and selective ligands. We developed transgenic mice that endogenously overexpress the alpha1B-AR subtype fused with the enhanced green fluorescent protein (EGFP). Endogenous expression was obtained by using a 3.4 kb fragment of the mouse alpha1B-AR promoter. Using this model, we determined cellular localization of the alpha1B-AR throughout the brain. The alpha1B-AR-EGFP fusion protein is expressed in neurons throughout the brain and in the Purkinje cells of the cerebellum. The alpha1B-AR is also expressed in NG2 oligodendrocyte precursor cells in both neonatal cell cultures and in the adult cerebral cortex, but is weakly expressed in mature oligodendrocytes. The alpha1B-AR was not observed in astrocytes or in cerebral vascular smooth muscle, cell types previously suggested to contain alpha1-ARs. We conclude that the alpha1B-AR is highly abundant throughout the brain, predominately in neurons, and may be involved in the development of the oligodendrocyte. In adult NG2 cells, implicated in stem cell-like functions, the alpha1B-AR may also play a role. This is the first report of a transgenic tagged-GPCR approach to determine in vivo localization of a receptor.


Subject(s)
Antigens/metabolism , Brain/metabolism , Neurons/metabolism , Oligodendroglia/metabolism , Proteoglycans/metabolism , Receptors, Adrenergic, alpha-1/biosynthesis , Animals , Cells, Cultured , Green Fluorescent Proteins , Immunohistochemistry , Luminescent Proteins , Mice , Mice, Transgenic , Microscopy, Confocal , Stem Cells/metabolism
18.
Brain ; 126(Pt 12): 2667-81, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12937073

ABSTRACT

The alpha1-adrenergic receptors (alpha1ARs) play an important role in mediating sympathetic neurotransmission in peripheral organ systems; however, central alpha1ARs are not well characterized. Additionally, due to the lack of sufficiently subtype-selective drugs or high avidity antibodies, the contribution of each alpha1AR subtype to various central functions is currently unclear. Transcription regulation through alpha1AR subtypes in the CNS is also unknown. Of interest, transgenic mice that systemically overexpress the alpha1BAR show central symptoms that include age-progressive impaired mobility, neurodegeneration and susceptibility to epileptic seizure. To investigate the molecular basis of this phenotype, oligonucleotide microarray studies of whole brains of various ages were carried out to compare gene expression profiles between transgenic and normal brains. The results indicated changes in expression of apoptotic, calcium regulatory, neurodegenerative and genes involved in neurotransmission. Defects in regulation of intracellular calcium are known to play a role in cell death; thus, these genes may provide clues as to the molecular basis of alpha1BAR-induced neurodegeneration. Epilepsy is a disorder that can be caused by an imbalance between excitatory (e.g. glutamate) and inhibitory (e.g. GABA) signals. Microarray analysis of transgenic brains showed increased N-methyl-d-aspartate (NMDA) receptors and decreased GABAA, which were confirmed with immunohistochemistry, western blot and radioligand binding studies. The alpha1BAR also co-localized with the glutamatergic distribution, suggesting a glutamate imbalance as a molecular rationale for the epileptic seizures.


Subject(s)
Apoptosis/genetics , Neurodegenerative Diseases/genetics , Receptors, Adrenergic, alpha-1/physiology , Animals , Cerebral Cortex/metabolism , Female , Gene Expression , Gene Expression Profiling , Hippocampus/metabolism , Male , Mice , Mice, Transgenic , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Oligonucleotide Array Sequence Analysis , Receptors, Adrenergic, alpha-1/genetics , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism
19.
Mol Pharmacol ; 63(5): 1104-16, 2003 May.
Article in English | MEDLINE | ID: mdl-12695539

ABSTRACT

Alpha(1)-adrenoceptor subtypes (alpha(1A)-, alpha(1B)-, alpha(1D)-) are known to couple to similar signaling pathways, although differences among the subtypes do exist. As a more sensitive assay, we used oligonucleotide microarrays to identify gene expression changes in Rat-1 fibroblasts stably expressing each individual subtype. We report the gene expressions that change by at least a factor of 2 or more. Gene expression profiles significantly changed equally among all three subtypes, despite the unequal efficacy of the inositol phosphate response. Gene expressions were clustered into cytokines/growth factors, transcription factors, enzymes, and extracellular matrix proteins. There were also a number of individual subtype-specific changes in gene expression, suggesting a link to independent pathways. In addition, all three alpha(1)-AR subtypes robustly stimulated the transcription of the prohypertrophic cytokine interleukin (IL)-6, but differentially altered members of the IL-6 signaling pathway (gp-130 and STAT3). This was confirmed by measurement of secreted IL-6, activated STAT3, and gp-130 levels. Activation of STAT3 Tyr705 phosphorylation by the alpha(1)-ARs was not through IL-6 activation but was synergistic with IL-6, suggesting direct effects. Interestingly, alpha(1B)-AR stimulation caused the dimerization-dependent phosphorylation of Tyr705 on STAT3 but did not activate the transcriptional-dependent phosphorylation of Ser727. The alpha(1B)-AR also constitutively down-regulated the protein levels of gp-130. These results suggest that the alpha(1B)-AR has differential effects on the phosphorylation status of the STAT3 pathway and may not be as prohypertrophic as the other two subtypes.


Subject(s)
Antigens, CD/metabolism , DNA-Binding Proteins/metabolism , Interleukin-6/metabolism , Membrane Glycoproteins/metabolism , Receptors, Adrenergic, alpha-1/genetics , Trans-Activators/metabolism , Animals , Binding, Competitive , Blotting, Northern , Cells, Cultured , Cytokine Receptor gp130 , Epinephrine/pharmacology , Fibroblasts/metabolism , Gene Expression , Gene Expression Profiling , Humans , Inositol Phosphates/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation , Rats , Receptors, Adrenergic, alpha-1/classification , STAT3 Transcription Factor , Serine/metabolism , Signal Transduction/physiology , Tritium
20.
Cardiovasc Res ; 57(2): 443-55, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12566117

ABSTRACT

OBJECTIVE: Cardiac hypertrophy is closely associated with the development of cardiomyopathies that lead to heart failure. The alpha(1B) adrenergic receptor (alpha(1)-AR) is an important regulator of the hypertrophic process. Cardiac hypertrophy induced by systemic overexpression of the alpha(1b)-AR in a mouse model does not progress to heart failure. We wanted to explore potential gene expression differences that characterize this type of hypertrophy that may identify genes that prevent progression to heart failure. METHODS: Transgenic and normal mice (B6CBA) representing two time points were compared; one at 2-3 months of age before disease manifests and the other at 12 months when the hypertrophy is significant. Age-matched hearts were removed, cRNA prepared and biotinylated. Aliquots of the cRNA was subjected to hybridization with Affymetrix chips representing 12,656 murine genes. Gene expression profiles were compared with normal age-matched controls as the baseline and confirmed by Northern and Western analysis. RESULTS: The non-EST genes could be grouped into five functional classifications: embryonic, proliferative, inflammatory, cardiac-related, and apoptotic. Growth response genes involved primarily Src-related receptors and signaling pathways. Transgenic hearts also had a 60% higher Src protein content. There was an inflammatory response that was verified by an increase in IgG and kappa-chained immunoglobulins by western analysis. Apoptosis may be regulated by cell cycle arrest through a p53-dependent mechanism. Cardiac gene expression was decreased for common hypertrophy-inducing proteins such as actin, collagen and GP130 pathways. CONCLUSIONS: Our results suggest a profile of gene expression in a case of atypical cardiac hypertrophy that does not progress to heart failure. Since many of these altered gene expressions have not been linked to heart failure models, our findings may provide a novel insight into the particular role that the alpha(1B)AR plays in its overall progression or regression.


Subject(s)
Cardiomegaly/genetics , Gene Expression Regulation , Receptors, Adrenergic, alpha-1/genetics , Animals , Antigens, CD/metabolism , Apoptosis/genetics , Blotting, Western , Cardiomegaly/metabolism , Cytokine Receptor gp130 , Disease Progression , Gene Expression Profiling/methods , Genes, src , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred CBA , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Phenotype , Protein-Tyrosine Kinases/physiology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...