54 results on '"Dianne M. Perez"'
Search Results
2. Adrenoceptors in GtoPdb v.2021.3
- Author
-
Sarah Gora, Rory Sleno, Peter Zylbergold, Dianne M. Perez, Katrin Altosaar, Poornima Balaji, Sergio Parra, Terry Hébert, Dominic Devost, Susanna Cotecchia, Kenneth P. Minneman, Rebecca Hills, Van A. Doze, Richard A. Bond, Douglas C. Eikenburg, Shahriar Kan, Martin C. Michel, David B. Bylund, J. Paul Hieble, Roger J. Summers, Gayane Machkalyan, Robert M. Graham, and Eugénie Goupil
- Subjects
Agonist ,business.industry ,medicine.drug_class ,Rauwolscine ,Propranolol ,Pharmacology ,Atenolol ,chemistry.chemical_compound ,Terazosin ,chemistry ,Sympatholytic ,Bupranolol ,Prazosin ,Medicine ,business ,medicine.drug - Abstract
The nomenclature of the Adrenoceptors has been agreed by the NC-IUPHAR Subcommittee on Adrenoceptors [60, 186]. Adrenoceptors, α1 The three α1-adrenoceptor subtypes α1A, α1B and α1D are activated by the endogenous agonists (-)-adrenaline and (-)-noradrenaline. -(-)phenylephrine, methoxamine and cirazoline are agonists and prazosin and doxazosin antagonists considered selective for α1- relative to α2-adrenoceptors. [3H]prazosin and [125I]HEAT (BE2254) are relatively selective radioligands. S(+)-niguldipine also has high affinity for L-type Ca2+ channels. Fluorescent derivatives of prazosin (Bodipy FLprazosin- QAPB) are used to examine cellular localisation of α1-adrenoceptors. α1-Adrenoceptor agonists are used as nasal decongestants; antagonists to treat symptoms of benign prostatic hyperplasia (alfuzosin, doxazosin, terazosin, tamsulosin and silodosin, with the last two compounds being α1A-adrenoceptor selective and claiming to relax bladder neck tone with less hypotension); and to a lesser extent hypertension (doxazosin, terazosin). The α1- and β2-adrenoceptor antagonist carvedilol is used to treat congestive heart failure, although the contribution of α1-adrenoceptor blockade to the therapeutic effect is unclear. Several anti-depressants and anti-psychotic drugs are α1-adrenoceptor antagonists contributing to side effects such as orthostatic hypotension. Adrenoceptors, α2 The three α2-adrenoceptor subtypes α2A, α2B and α2C are activated by (-)-adrenaline and with lower potency by (-)-noradrenaline. brimonidine and talipexole are agonists and rauwolscine and yohimbine antagonists selective for α2- relative to α1-adrenoceptors. [3H]rauwolscine, [3H]brimonidine and [3H]RX821002 are relatively selective radioligands. There are species variations in the pharmacology of the α2A-adrenoceptor. Multiple mutations of α2-adrenoceptors have been described, some associated with alterations in function. Presynaptic α2-adrenoceptors regulate many functions in the nervous system. The α2-adrenoceptor agonists clonidine, guanabenz and brimonidine affect central baroreflex control (hypotension and bradycardia), induce hypnotic effects and analgesia, and modulate seizure activity and platelet aggregation. clonidine is an anti-hypertensive (relatively little used) and counteracts opioid withdrawal. dexmedetomidine (also xylazine) is increasingly used as a sedative and analgesic in human [31] and veterinary medicine and has sympatholytic and anxiolytic properties. The α2-adrenoceptor antagonist mirtazapine is used as an anti-depressant. The α2B subtype appears to be involved in neurotransmission in the spinal cord and α2C in regulating catecholamine release from adrenal chromaffin cells. Although subtype-selective antagonists have been developed, none are used clinically and they remain experimental tools. Adrenoceptors, β The three β-adrenoceptor subtypes β1, β2 and β3 are activated by the endogenous agonists (-)-adrenaline and (-)-noradrenaline. Isoprenaline is selective for β-adrenoceptors relative to α1- and α2-adrenoceptors, while propranolol (pKi 8.2-9.2) and cyanopindolol (pKi 10.0-11.0) are relatively selective antagonists for β1- and β2- relative to β3-adrenoceptors. (-)-noradrenaline, xamoterol and (-)-Ro 363 show selectivity for β1- relative to β2-adrenoceptors. Pharmacological differences exist between human and mouse β3-adrenoceptors, and the 'rodent selective' agonists BRL 37344 and CL316243 have low efficacy at the human β3-adrenoceptor whereas CGP 12177 (low potency) and L 755507 activate human β3-adrenoceptors [88]. β3-Adrenoceptors are resistant to blockade by propranolol, but can be blocked by high concentrations of bupranolol. SR59230A has reasonably high affinity at β3-adrenoceptors, but does not discriminate between the three β- subtypes [320] whereas L-748337 is more selective. [125I]-cyanopindolol, [125I]-hydroxy benzylpindolol and [3H]-alprenolol are high affinity radioligands that label β1- and β2- adrenoceptors and β3-adrenoceptors can be labelled with higher concentrations (nM) of [125I]-cyanopindolol together with β1- and β2-adrenoceptor antagonists. Fluorescent ligands such as BODIPY-TMR-CGP12177 can be used to track β-adrenoceptors at the cellular level [8]. Somewhat selective β1-adrenoceptor agonists (denopamine, dobutamine) are used short term to treat cardiogenic shock but, chronically, reduce survival. β1-Adrenoceptor-preferring antagonists are used to treat cardiac arrhythmias (atenolol, bisoprolol, esmolol) and cardiac failure (metoprolol, nebivolol) but also in combination with other treatments to treat hypertension (atenolol, betaxolol, bisoprolol, metoprolol and nebivolol) [507]. Cardiac failure is also treated with carvedilol that blocks β1- and β2-adrenoceptors, as well as α1-adrenoceptors. Short (salbutamol, terbutaline) and long (formoterol, salmeterol) acting β2-adrenoceptor-selective agonists are powerful bronchodilators used to treat respiratory disorders. Many first generation β-adrenoceptor antagonists (propranolol) block both β1- and β2-adrenoceptors and there are no β2-adrenoceptor-selective antagonists used therapeutically. The β3-adrenoceptor agonist mirabegron is used to control overactive bladder syndrome. There is evidence to suggest that β-adrenoceptor antagonists can reduce metastasis in certain types of cancer [189].
- Published
- 2021
- Full Text
- View/download PDF
3. Current Developments on the Role of α1-Adrenergic Receptors in Cognition, Cardioprotection, and Metabolism
- Author
-
Dianne M. Perez
- Subjects
cognition ,0301 basic medicine ,Sympathetic nervous system ,Adrenergic receptor ,QH301-705.5 ,Biology ,03 medical and health sciences ,Norepinephrine ,0302 clinical medicine ,medicine ,Biology (General) ,Receptor ,adrenergic receptor ,G protein-coupled receptor ,Cell Biology ,030104 developmental biology ,Epinephrine ,medicine.anatomical_structure ,cardioprotection ,G-protein coupled receptor ,Signal transduction ,Vascular smooth muscle contraction ,metabolism ,Neuroscience ,030217 neurology & neurosurgery ,Developmental Biology ,medicine.drug - Abstract
The α1-adrenergic receptors (ARs) are G-protein coupled receptors that bind the endogenous catecholamines, norepinephrine, and epinephrine. They play a key role in the regulation of the sympathetic nervous system along with β and α2-AR family members. While all of the adrenergic receptors bind with similar affinity to the catecholamines, they can regulate different physiologies and pathophysiologies in the body because they couple to different G-proteins and signal transduction pathways, commonly in opposition to one another. While α1-AR subtypes (α1A, α1B, α1C) have long been known to be primary regulators of vascular smooth muscle contraction, blood pressure, and cardiac hypertrophy, their role in neurotransmission, improving cognition, protecting the heart during ischemia and failure, and regulating whole body and organ metabolism are not well known and are more recent developments. These advancements have been made possible through the development of transgenic and knockout mouse models and more selective ligands to advance their research. Here, we will review the recent literature to provide new insights into these physiological functions and possible use as a therapeutic target.
- Published
- 2021
- Full Text
- View/download PDF
4. α1-Adrenergic Receptors in Neurotransmission, Synaptic Plasticity, and Cognition
- Author
-
Dianne M. Perez
- Subjects
0301 basic medicine ,cognition ,Sympathetic nervous system ,Adrenergic receptor ,Biology ,Neurotransmission ,03 medical and health sciences ,Norepinephrine ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Pharmacology (medical) ,neurotransmission ,Neurotransmitter ,Receptor ,G protein-coupled receptor ,adrenergic receptor ,Pharmacology ,synaptic plasticity ,lcsh:RM1-950 ,030104 developmental biology ,medicine.anatomical_structure ,lcsh:Therapeutics. Pharmacology ,chemistry ,030220 oncology & carcinogenesis ,Synaptic plasticity ,G-protein coupled receptor ,Neuroscience ,medicine.drug - Abstract
α1-adrenergic receptors are G-Protein Coupled Receptors that are involved in neurotransmission and regulate the sympathetic nervous system through binding and activating the neurotransmitter, norepinephrine, and the neurohormone, epinephrine. There are three α1-adrenergic receptor subtypes (α1A, α1B, α1D) that are known to play various roles in neurotransmission and cognition. They are related to two other adrenergic receptor families that also bind norepinephrine and epinephrine, the β- and α2-, each with three subtypes (β1, β2, β3, α2A, α2B, α2C). Previous studies assessing the roles of α1-adrenergic receptors in neurotransmission and cognition have been inconsistent. This was due to the use of poorly-selective ligands and many of these studies were published before the characterization of the cloned receptor subtypes and the subsequent development of animal models. With the availability of more-selective ligands and the development of animal models, a clearer picture of their role in cognition and neurotransmission can be assessed. In this review, we highlight the significant role that the α1-adrenergic receptor plays in regulating synaptic efficacy, both short and long-term synaptic plasticity, and its regulation of different types of memory. We will also present evidence that the α1-adrenergic receptors, and particularly the α1A-adrenergic receptor subtype, are a potentially good target to treat a wide variety of neurological conditions with diminished cognition.
- Published
- 2020
- Full Text
- View/download PDF
5. Adrenoceptors (version 2019.4) in the IUPHAR/BPS Guide to Pharmacology Database
- Author
-
Rory Sleno, Shahriar Kan, Peter Zylbergold, Dominic Devost, Richard A. Bond, Sarah Gora, Rebecca Hills, Dianne M. Perez, J. Paul Hieble, Kenneth P. Minneman, Terry Hébert, Katrin Altosaar, Martin C. Michel, David B. Bylund, Sergio Parra, Roger J. Summers, Gayane Machkalyan, Eugénie Goupil, Van A. Doze, Poornima Balaji, Douglas C. Eikenburg, Robert M. Graham, and Susanna Cotecchia
- Subjects
Agonist ,medicine.drug_class ,business.industry ,Rauwolscine ,Propranolol ,Pharmacology ,Atenolol ,Yohimbine ,chemistry.chemical_compound ,chemistry ,Bisoprolol ,Sympatholytic ,medicine ,Prazosin ,business ,medicine.drug - Abstract
The nomenclature of the Adrenoceptors has been agreed by the NC-IUPHAR Subcommittee on Adrenoceptors [58], see also [180]. Adrenoceptors, α1α1-Adrenoceptors are activated by the endogenous agonists (-)-adrenaline and (-)-noradrenaline. phenylephrine, methoxamine and cirazoline are agonists and prazosin and cirazoline antagonists considered selective for α1- relative to α2-adrenoceptors. [3H]prazosin and [125I]HEAT (BE2254) are relatively selective radioligands. S(+)-niguldipine also has high affinity for L-type Ca2+ channels. Fluorescent derivatives of prazosin (Bodipy PLprazosin- QAPB) are used to examine cellular localisation of α1-adrenoceptors. Selective α1-adrenoceptor agonists are used as nasal decongestants; antagonists to treat hypertension (doxazosin, prazosin) and benign prostatic hyperplasia (alfuzosin, tamsulosin). The α1- and β2-adrenoceptor antagonist carvedilol is used to treat congestive heart failure, although the contribution of α1-adrenoceptor blockade to the therapeutic effect is unclear. Several anti-depressants and anti-psychotic drugs are α1-adrenoceptor antagonists contributing to side effects such as orthostatic hypotension and extrapyramidal effects.Adrenoceptors, α2 α2-Adrenoceptors are activated by (-)-adrenaline and with lower potency by (-)-noradrenaline. brimonidine and talipexole are agonists and rauwolscine and yohimbine antagonists selective for α2- relative to α1-adrenoceptors. [3H]rauwolscine, [3H]brimonidine and [3H]RX821002 are relatively selective radioligands. There is species variation in the pharmacology of the α2A-adrenoceptor. Multiple mutations of α2-adrenoceptors have been described, some associated with alterations in function. Presynaptic α2-adrenoceptors regulate many functions in the nervous system. The α2-adrenoceptor agonists clonidine, guanabenz and brimonidine affect central baroreflex control (hypotension and bradycardia), induce hypnotic effects and analgesia, and modulate seizure activity and platelet aggregation. clonidine is an anti-hypertensive and counteracts opioid withdrawal. dexmedetomidine (also xylazine) is used as a sedative and analgesic in human and veterinary medicine with sympatholytic and anxiolytic properties. The α2-adrenoceptor antagonist yohimbine has been used to treat erectile dysfunction and mirtazapine as an anti-depressant. The α2B subtype appears to be involved in neurotransmission in the spinal cord and α2C in regulating catecholamine release from adrenal chromaffin cells.Adrenoceptors, ββ-Adrenoceptors are activated by the endogenous agonists (-)-adrenaline and (-)-noradrenaline. Isoprenaline is selective for β-adrenoceptors relative to α1- and α2-adrenoceptors, while propranolol (pKi 8.2-9.2) and cyanopindolol (pKi 10.0-11.0) are relatively β1 and β2 adrenoceptor-selective antagonists. (-)-noradrenaline, xamoterol and (-)-Ro 363 show selectivity for β1- relative to β2-adrenoceptors. Pharmacological differences exist between human and mouse β3-adrenoceptors, and the 'rodent selective' agonists BRL 37344 and CL316243 have low efficacy at the human β3-adrenoceptor whereas CGP 12177 and L 755507 activate human β3-adrenoceptors [88]. β3-Adrenoceptors are resistant to blockade by propranolol, but can be blocked by high concentrations of bupranolol. SR59230A has reasonably high affinity at β3-adrenoceptors, but does not discriminate well between the three β- subtypes whereas L 755507 is more selective. [125I]-cyanopindolol, [125I]-hydroxy benzylpindolol and [3H]-alprenolol are high affinity radioligands that label β1- and β2- adrenoceptors and β3-adrenoceptors can be labelled with higher concentrations (nM) of [125I]-cyanopindolol together with β1- and β2-adrenoceptor antagonists. [3H]-L-748337 is a β3-selective radioligand [474]. Fluorescent ligands such as BODIPY-TMR-CGP12177 can be used to track β-adrenoceptors at the cellular level [8]. Somewhat selective β1-adrenoceptor agonists (denopamine, dobutamine) are used short term to treat cardiogenic shock but, chronically, reduce survival. β1-Adrenoceptor-preferring antagonists are used to treat hypertension (atenolol, betaxolol, bisoprolol, metoprolol and nebivolol), cardiac arrhythmias (atenolol, bisoprolol, esmolol) and cardiac failure (metoprolol, nebivolol). Cardiac failure is also treated with carvedilol that blocks β1- and β2-adrenoceptors, as well as α1-adrenoceptors. Short (salbutamol, terbutaline) and long (formoterol, salmeterol) acting β2-adrenoceptor-selective agonists are powerful bronchodilators used to treat respiratory disorders. Many first generation β-adrenoceptor antagonists (propranolol) block both β1- and β2-adrenoceptors and there are no β2-adrenoceptor-selective antagonists used therapeutically. The β3-adrenoceptor agonist mirabegron is used to control overactive bladder syndrome.
- Published
- 2019
- Full Text
- View/download PDF
6. Targeting Adrenergic Receptors in Metabolic Therapies for Heart Failure
- Author
-
Dianne M. Perez
- Subjects
0301 basic medicine ,Chronotropic ,Cardiac function curve ,Sympathetic nervous system ,Sympathetic Nervous System ,Adrenergic receptor ,QH301-705.5 ,Adrenergic beta-Antagonists ,Adrenergic ,Review ,heart ,Oxidative phosphorylation ,030204 cardiovascular system & hematology ,Pharmacology ,Catalysis ,Inorganic Chemistry ,03 medical and health sciences ,0302 clinical medicine ,Receptors, Adrenergic, alpha-1 ,Receptors, Adrenergic, beta ,Animals ,Humans ,Medicine ,Molecular Targeted Therapy ,Biology (General) ,Physical and Theoretical Chemistry ,QD1-999 ,Molecular Biology ,Beta oxidation ,Spectroscopy ,adrenergic receptor ,myocyte ,Heart Failure ,business.industry ,Organic Chemistry ,General Medicine ,medicine.disease ,Computer Science Applications ,Chemistry ,030104 developmental biology ,medicine.anatomical_structure ,Heart failure ,business ,Oxidation-Reduction ,metabolism ,Metabolic Networks and Pathways - Abstract
The heart has a reduced capacity to generate sufficient energy when failing, resulting in an energy-starved condition with diminished functions. Studies have identified numerous changes in metabolic pathways in the failing heart that result in reduced oxidation of both glucose and fatty acid substrates, defects in mitochondrial functions and oxidative phosphorylation, and inefficient substrate utilization for the ATP that is produced. Recent early-phase clinical studies indicate that inhibitors of fatty acid oxidation and antioxidants that target the mitochondria may improve heart function during failure by increasing compensatory glucose oxidation. Adrenergic receptors (α1 and β) are a key sympathetic nervous system regulator that controls cardiac function. β-AR blockers are an established treatment for heart failure and α1A-AR agonists have potential therapeutic benefit. Besides regulating inotropy and chronotropy, α1- and β-adrenergic receptors also regulate metabolic functions in the heart that underlie many cardiac benefits. This review will highlight recent studies that describe how adrenergic receptor-mediated metabolic pathways may be able to restore cardiac energetics to non-failing levels that may offer promising therapeutic strategies.
- Published
- 2021
- Full Text
- View/download PDF
7. The role of α1-adrenergic receptors in regulating metabolism: increased glucose tolerance, leptin secretion and lipid oxidation
- Author
-
Dianne M. Perez, Robert S. Papay, and Ting Shi
- Subjects
0301 basic medicine ,Genetically modified mouse ,medicine.medical_specialty ,Glucose uptake ,Leptin ,Lipid metabolism ,Cell Biology ,White adipose tissue ,Metabolism ,Biology ,medicine.disease ,Biochemistry ,Impaired glucose tolerance ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Lipid oxidation ,Internal medicine ,medicine ,Molecular Biology ,030217 neurology & neurosurgery - Abstract
The role of α1-adrenergic receptors (α1-ARs) and their subtypes in metabolism is not well known. Most previous studies were performed before the advent of transgenic mouse models and utilized transformed cell lines and poorly selective antagonists. We have now studied the metabolic regulation of the α1A- and α1B-AR subtypes in vivo using knock-out (KO) and transgenic mice that express a constitutively active mutant (CAM) form of the receptor, assessing subtype-selective functions. CAM mice increased glucose tolerance while KO mice display impaired glucose tolerance. CAM mice increased while KO decreased glucose uptake into white fat tissue and skeletal muscle with the CAM α1A-AR showing selective glucose uptake into the heart. Using indirect calorimetry, both CAM mice demonstrated increased whole body fatty acid oxidation, while KO mice preferentially oxidized carbohydrate. CAM α1A-AR mice displayed significantly decreased fasting plasma triglycerides and glucose levels while α1A-AR KO displayed increased levels of triglycerides and glucose. Both CAM mice displayed increased plasma levels of leptin while KO mice decreased leptin levels. Most metabolic effects were more efficacious with the α1A-AR subtype. Our results suggest that stimulation of α1-ARs results in a favorable metabolic profile of increased glucose tolerance, cardiac glucose uptake, leptin secretion and increased whole body lipid metabolism that may contribute to its previously recognized cardioprotective and neuroprotective benefits.
- Published
- 2016
- Full Text
- View/download PDF
8. α1A-Adrenergic Receptors Regulate Cardiac Hypertrophy In Vivo Through Interleukin-6 Secretion
- Author
-
Ting Shi, Robert S. Papay, Michael T. Piascik, Dianne M. Perez, and Sathyamangla V. Naga Prasad
- Subjects
Genetically modified mouse ,medicine.medical_specialty ,Adrenergic receptor ,Transgene ,Cardiomegaly ,Mice, Transgenic ,Biology ,p38 Mitogen-Activated Protein Kinases ,Muscle hypertrophy ,Mice ,Receptors, Adrenergic, alpha-1 ,Internal medicine ,medicine ,Animals ,Myocytes, Cardiac ,Receptor ,Mice, Knockout ,Pharmacology ,Interleukin-6 ,NF-kappa B ,Articles ,Glycoprotein 130 ,Endocrinology ,STAT protein ,Molecular Medicine ,Signal transduction ,Signal Transduction - Abstract
The role of α₁-adrenergic receptors (ARs) in the regulation of cardiac hypertrophy is still unclear, because transgenic mice demonstrated hypertrophy or the lack of it despite high receptor overexpression. To further address the role of the α₁-ARs in cardiac hypertrophy, we analyzed unique transgenic mice that overexpress constitutively active mutation (CAM) α₁A-ARs or CAM α₁B-ARs under the regulation of large fragments of their native promoters. These constitutively active receptors are expressed in all tissues that endogenously express their wild-type counterparts as opposed to only myocyte-targeted transgenic mice. In this study, we discovered that CAM α₁A-AR mice in vivo have cardiac hypertrophy independent of changes in blood pressure, corroborating earlier studies, but in contrast to myocyte-targeted α₁A-AR mice. We also found cardiac hypertrophy in CAM α₁B-AR mice, in agreement with previous studies, but hypertrophy only developed in older mice. We also discovered unique α₁-AR-mediated hypertrophic signaling that was AR subtype-specific with CAM α₁A-AR mice secreting atrial naturietic factor and interleukin-6 (IL-6), whereas CAM α₁B-AR mice expressed activated nuclear factor-κB (NF-κB). These particular hypertrophic signals were blocked when the other AR subtype was coactivated. We also discovered that crossbreeding the two CAM models (double CAM α₁A/B-AR) inhibited the development of hypertrophy and was reversible with single receptor activation, suggesting that coactivation of the receptors can lead to novel antagonistic signal transduction. This was confirmed by demonstrating antagonistic signals that were even lower than normal controls in the double CAM α₁A/B-AR mice for p38, NF-κB, and the IL-6/glycoprotein 130/signal transducer and activator of transcription 3 pathway. Because α₁A/B double knockout mice fail to develop hypertrophy in response to IL-6, our results suggest that IL-6 is a major mediator of α₁A-AR cardiac hypertrophy.
- Published
- 2013
- Full Text
- View/download PDF
9. A unique microRNA profile in end-stage heart failure indicates alterations in specific cardiovascular signaling networks
- Author
-
Edward F. Plow, Ashwin Kotwal, Dianne M. Perez, Manveen K Gupta, Venkata Suresh K Surampudi, Christine S. Moravec, Chang Gong Liu, Sadashiva S. Karnik, Randall C. Starling, Subha Sen, Zhong-Hui Duan, Qingyu Wu, and Sathyamangla V. Naga Prasad
- Subjects
0301 basic medicine ,Male ,Microarray ,Microarrays ,Gene Identification and Analysis ,Gene Expression ,lcsh:Medicine ,Genetic Networks ,030204 cardiovascular system & hematology ,Bioinformatics ,Biochemistry ,Cardiovascular System ,Mice ,0302 clinical medicine ,Gene expression ,Medicine and Health Sciences ,Gene Regulatory Networks ,lcsh:Science ,Cells, Cultured ,Multidisciplinary ,Heart ,Transfection ,Genomics ,Middle Aged ,Nucleic acids ,Bioassays and Physiological Analysis ,Female ,Signal transduction ,DNA microarray ,Anatomy ,Network Analysis ,Algorithms ,Research Article ,Signal Transduction ,Computer and Information Sciences ,Cardiology ,Computational biology ,Biology ,Research and Analysis Methods ,03 medical and health sciences ,microRNA ,medicine ,Genetics ,Animals ,Humans ,Non-coding RNA ,Heart Failure ,Biology and life sciences ,Gene Expression Profiling ,lcsh:R ,medicine.disease ,Gene regulation ,Signaling Networks ,MicroRNAs ,030104 developmental biology ,Heart failure ,Cardiovascular Anatomy ,RNA ,lcsh:Q ,NODAL - Abstract
It is well established that the gene expression patterns are substantially altered in cardiac hypertrophy and heart failure, however, less is known about the reasons behind such global differences. MicroRNAs (miRNAs) are short non-coding RNAs that can target multiple molecules to regulate wide array of proteins in diverse pathways. The goal of the study was to profile alterations in miRNA expression using end-stage human heart failure samples with an aim to build signaling network pathways using predicted targets for the altered miRNA and to determine nodal molecules regulating individual networks. Profiling of miRNAs using custom designed microarray and validation with an independent set of samples identified eight miRNAs that are altered in human heart failure including one novel miRNA yet to be implicated in cardiac pathology. To gain an unbiased perspective on global regulation by top eight altered miRNAs, functional relationship of predicted targets for these eight miRNAs were examined by network analysis. Ingenuity Pathways Analysis network algorithm was used to build global signaling networks based on the targets of altered miRNAs which allowed us to identify participating networks and nodal molecules that could contribute to cardiac pathophysiology. Majority of the nodal molecules identified in our analysis are targets of altered miRNAs and known regulators of cardiovascular signaling. Cardio-genomics heart failure gene expression public data base was used to analyze trends in expression pattern for target nodal molecules and indeed changes in expression of nodal molecules inversely correlated to miRNA alterations. We have used NF kappa B network as an example to show that targeting other molecules in the network could alter the nodal NF kappa B despite not being a miRNA target suggesting an integrated network response. Thus, using network analysis we show that altering key functional target proteins may regulate expression of the myriad signaling pathways underlying the cardiac pathology.
- Published
- 2017
10. A unique microRNA profile in end-stage heart failure indicates alterations in specific cardiovascular signaling networks
- Author
-
Qingyu Wu, Sathyamangla V. Naga Prasad, Carlo M. Croce, Ashwin Kotwal, George A. Calin, Subha Sen, Dianne M. Perez, Edward F. Plow, Randall C. Starling, Sadashiva S. Karnik, Christine S. Moravec, Stefano Volinia, Zong Hui Duan, Manveen K Gupta, Chang Gong Liu, and Venkata Suresh K Surampudi
- Subjects
Cardiomyopathy, Dilated ,Male ,Immunoblotting ,Cardiomyopathy ,Biomolecular Networks ,Computational biology ,Biology ,Bioinformatics ,Biochemistry ,Cardiovascular System ,Cell Line ,Mice ,Gene expression ,microRNA ,medicine ,Animals ,Humans ,Molecular Biology ,Oligonucleotide Array Sequence Analysis ,Regulation of gene expression ,Heart Failure ,Computational Biology ,Nucleic Acid Hybridization ,Reproducibility of Results ,Cell Biology ,MicroRNA Profile ,Middle Aged ,medicine.disease ,MicroRNAs ,Gene Expression Regulation ,Heart failure ,Female ,Additions and Corrections ,Signal transduction ,NODAL ,Signal Transduction - Abstract
It is well established that gene expression patterns are substantially altered in cardiac hypertrophy and heart failure, but the reasons for such differences are not clear. MicroRNAs (miRNAs) are short noncoding RNAs that provide a novel mechanism for gene regulation. The goal of this study was to comprehensively test for alterations in miRNA expression using human heart failure samples with an aim to build signaling pathway networks using predicted targets for the miRNAs and to identify nodal molecules that control these networks. Genome-wide profiling of miRNAs was performed using custom-designed miRNA microarray followed by validation on an independent set of samples. Eight miRNAs are significantly altered in heart failure of which we have identified two novel miRNAs that are yet to be implicated in cardiac pathophysiology. To gain an unbiased global perspective on regulation by altered miRNAs, predicted targets of eight miRNAs were analyzed using the Ingenuity Pathways Analysis network algorithm to build signaling networks and identify nodal molecules. The majority of nodal molecules identified in our analysis are targets of altered miRNAs and are known regulators of cardiovascular signaling. A heart failure gene expression data base was used to analyze changes in expression patterns for these target nodal molecules. Indeed, expression of nodal molecules was altered in heart failure and inversely correlated to miRNA changes validating our analysis. Importantly, using network analysis we have identified a limited number of key functional targets that may regulate expression of the myriad proteins in heart failure and could be potential therapeutic targets.
- Published
- 2016
11. α1A-Adrenergic receptor prevents cardiac ischemic damage through PKCδ/GLUT1/4-mediated glucose uptake
- Author
-
Ting Shi, Robert S. Papay, and Dianne M. Perez
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Glucose uptake ,Myocardial Ischemia ,Stimulation ,Apoptosis ,Mice, Transgenic ,Deoxyglucose ,Biochemistry ,Article ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,Internal medicine ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,Myocytes, Cardiac ,Hypoxia ,Molecular Biology ,Protein kinase C ,Glucose Transporter Type 1 ,Glucose Transporter Type 4 ,biology ,Cell Membrane ,Glucose transporter ,Biological Transport ,Cell Biology ,Mice, Inbred C57BL ,Protein Kinase C-delta ,030104 developmental biology ,Endocrinology ,Glucose ,chemistry ,Animals, Newborn ,Cytoprotection ,biology.protein ,GLUT1 ,Mutant Proteins ,Rottlerin ,GLUT4 ,hormones, hormone substitutes, and hormone antagonists ,Signal Transduction - Abstract
While α(1)-adrenergic receptors (ARs) have been previously shown to limit ischemic cardiac damage, the mechanisms remain unclear. Most previous studies utilized low oxygen conditions in addition to ischemic buffers with glucose deficiencies, but we discovered profound differences if the two conditions are separated. We assessed both mouse neonatal and adult myocytes and HL-1 cells in a series of assays assessing ischemic damage under hypoxic or low glucose conditions. We found that α(1)-AR stimulation protected against increased lactate dehydrogenase release or Annexin V(+) apoptosis under conditions that were due to low glucose concentration not to hypoxia. The α(1)-AR antagonist prazosin or nonselective protein kinase C (PKC) inhibitors blocked the protective effect. α(1)-AR stimulation increased (3)H-deoxyglucose uptake that was blocked with either an inhibitor to glucose transporter 1 or 4 (GLUT1 or GLUT4) or small interfering RNA (siRNA) against PKCδ. GLUT1/4 inhibition also blocked α(1)-AR-mediated protection from apoptosis. The PKC inhibitor rottlerin or siRNA against PKCδ blocked α(1)-AR stimulated GLUT1 or GLUT4 plasma membrane translocation. α(1)-AR stimulation increased plasma membrane concentration of either GLUT1 or GLUT4 in a time-dependent fashion. Transgenic mice overexpressing the α(1A)-AR but not α(1B)-AR mice displayed increased glucose uptake and increased GLUT1 and GLUT4 plasma membrane translocation in the adult heart while α(1A)-AR but not α(1B)-AR knockout mice displayed lowered glucose uptake and GLUT translocation. Our results suggest that α(1)-AR activation is anti-apoptotic and protective during cardiac ischemia due to glucose deprivation and not hypoxia by enhancing glucose uptake into the heart via PKCδ-mediated GLUT translocation that may be specific to the α(1A)-AR subtype.
- Published
- 2016
12. Localization of the mouse α1A-adrenergic receptor (AR) in the brain: α1AAR is expressed in neurons, GABAergic interneurons, and NG2 oligodendrocyte progenitors
- Author
-
Dan F. McCune, John C. McGrath, Manoj C. Rodrigo, Dianne M. Perez, Paul C. Simpson, Robert S. Papay, Robert J. Gaivin, Van A. Doze, and Archana Jha
- Subjects
Cerebellum ,Patch-Clamp Techniques ,Green Fluorescent Proteins ,Glutamate decarboxylase ,Central nervous system ,Gene Expression ,Alpha (ethology) ,Mice, Transgenic ,In Vitro Techniques ,Biology ,Membrane Potentials ,Mice ,Norepinephrine ,Radioligand Assay ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,Antigens ,Receptor ,gamma-Aminobutyric Acid ,Cellular localization ,Neurons ,Stem Cells ,General Neuroscience ,Brain ,Cell Differentiation ,beta-Galactosidase ,Immunohistochemistry ,Oligodendrocyte ,Cell biology ,Oligodendroglia ,medicine.anatomical_structure ,nervous system ,GABAergic ,Proteoglycans ,Adrenergic alpha-1 Receptor Agonists ,Neuroscience - 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.
- Published
- 2006
- Full Text
- View/download PDF
13. Both α- and α-adrenergic receptors crosstalk to downregulate β-ARs in mouse heart: coupling to differential PTX-sensitive pathways
- Author
-
Dianne M. Perez, Robert J. Gaivin, Ting Shi, Paul C. Simpson, Boyd R. Rorabaugh, and Robert S. Papay
- Subjects
Genetically modified mouse ,medicine.medical_specialty ,Adrenergic receptor ,Chemistry ,Adrenergic ,Endogeny ,Pertussis toxin ,Endocrinology ,Downregulation and upregulation ,Internal medicine ,medicine ,Signal transduction ,Cardiology and Cardiovascular Medicine ,Receptor ,Molecular Biology - 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.
- Published
- 2005
- Full Text
- View/download PDF
14. Mouse ?1B-adrenergic receptor is expressed in neurons and NG2 oligodendrocytes
- Author
-
Dan F. McCune, Robert S. Papay, John C. McGrath, Robert J. Gaivin, Wendy B. Macklin, Boyd R. Rorabaugh, and Dianne M. Perez
- Subjects
Genetically modified mouse ,Cell type ,Cerebellum ,Adrenergic receptor ,Green Fluorescent Proteins ,Central nervous system ,Mice, Transgenic ,Biology ,Mice ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,Antigens ,Receptor ,Cells, Cultured ,Cellular localization ,Neurons ,Microscopy, Confocal ,Stem Cells ,General Neuroscience ,Brain ,Immunohistochemistry ,Oligodendrocyte ,Cell biology ,Luminescent Proteins ,Oligodendroglia ,medicine.anatomical_structure ,Proteoglycans ,Neuroscience - 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.
- Published
- 2004
- Full Text
- View/download PDF
15. The α1B-adrenergic receptor decreases the inotropic response in the mouse Langendorff heart model
- Author
-
Pedro J. Gonzalez-Cabrera, Dan F. McCune, Michael T. Piascik, Dan Chalothorn, Dianne M. Perez, Boyd R. Rorabaugh, June Yun, and Sean A. Ross
- Subjects
Male ,Cardiac function curve ,Inotrope ,medicine.medical_specialty ,Langendorff heart ,Heart disease ,Physiology ,Gene Expression ,Mice, Transgenic ,Biology ,Piperazines ,Contractility ,Mice ,Phenylephrine ,Receptors, Adrenergic, alpha-1 ,Physiology (medical) ,Internal medicine ,Heart rate ,medicine ,Animals ,Myocytes, Cardiac ,Adrenergic alpha-Antagonists ,Heart ,medicine.disease ,Myocardial Contraction ,Stimulation, Chemical ,Perfusion ,Endocrinology ,Circulatory system ,Female ,Cardiology and Cardiovascular Medicine ,medicine.drug - Abstract
alpha(1)-Adrenergic receptors (ARs) are known mediators of a positive inotropy in the heart, which may play even more important roles in heart disease. Due to a lack of sufficiently selective ligands, the contribution of each of the three alpha(1)-AR subtypes (alpha(1A), alpha(1B) and alpha(1D)) to cardiac function is not clearly defined. In this study, we used a systemically expressing mouse model that overexpresses the alpha(1B)-AR to define the role of this subtype in cardiac function.We used the mouse Langendorff heart model to assess changes in contractility under basal and phenylephrine-induced conditions.We find that a 50% increase of the alpha(1B)-AR in the heart does not change basal cardiac parameters compared to age-matched normals (heart rate, +/-dP/dT and coronary flow). However, the inotropic response to phenylephrine is blunted. The same results were obtained in isolated adult myocytes. The difference in inotropy could be blocked by the selective alpha(1A)-AR antagonist, 5-methylurapidil, which correlated with decreases in alpha(1A)-AR density, suggesting that the alpha(1B)-AR had caused a compensatory downregulation of the alpha(1A)-AR.These results suggest that the alpha(1B)-AR does not have a major role in the positive inotropic response in the mouse myocardium but may negatively modulate the response of the alpha(1A)-AR.
- Published
- 2003
- Full Text
- View/download PDF
16. Gene expression profile of neurodegeneration induced by 1B-adrenergic receptor overactivity: NMDA/GABAA dysregulation and apoptosis
- Author
-
Dianne M. Perez, Zhong Ying, Dan F. McCune, Robert J. Gaivin, June Yun, Robert S. Papay, Attaporn Boongird, Imad Najm, and Pedro J. Gonzalez-Cabrera
- Subjects
Male ,Gene Expression ,Apoptosis ,Mice, Transgenic ,Biology ,Neurotransmission ,Hippocampus ,Receptors, N-Methyl-D-Aspartate ,gamma-Aminobutyric acid ,Mice ,Glutamatergic ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,Oligonucleotide Array Sequence Analysis ,Cerebral Cortex ,GABAA receptor ,Gene Expression Profiling ,Neurodegeneration ,Glutamate receptor ,Neurodegenerative Diseases ,Receptors, GABA-A ,medicine.disease ,Gene expression profiling ,NMDA receptor ,Female ,Neurology (clinical) ,Neuroscience ,medicine.drug - 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.
- Published
- 2003
- Full Text
- View/download PDF
17. Gene expression profiling of α1b-adrenergic receptor-induced cardiac hypertrophy by oligonucleotide arrays
- Author
-
Sean A. Ross, Pedro J. Gonzalez-Cabrera, Michael T. Piascik, Dan F. McCune, Robert J. Gaivin, June Yun, Michael J. Zuscik, and Dianne M. Perez
- Subjects
medicine.medical_specialty ,Physiology ,Transgene ,Blotting, Western ,Cardiomyopathy ,Apoptosis ,Cardiomegaly ,Mice, Transgenic ,Biology ,Muscle hypertrophy ,Mice ,Antigens, CD ,Receptors, Adrenergic, alpha-1 ,Physiology (medical) ,Internal medicine ,Gene expression ,Cytokine Receptor gp130 ,medicine ,Animals ,Receptor ,Oligonucleotide Array Sequence Analysis ,Membrane Glycoproteins ,Gene Expression Profiling ,Protein-Tyrosine Kinases ,medicine.disease ,Gene expression profiling ,Genes, src ,Phenotype ,Endocrinology ,Gene Expression Regulation ,Heart failure ,Disease Progression ,Mice, Inbred CBA ,Signal transduction ,Cardiology and Cardiovascular Medicine ,Signal Transduction - Abstract
Objective: Cardiac hypertrophy is closely associated with the development of cardiomyopathies that lead to heart failure. The α1B adrenergic receptor (α1-AR) is an important regulator of the hypertrophic process. Cardiac hypertrophy induced by systemic overexpression of the α1b-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 κ-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 α1BAR plays in its overall progression or regression.
- Published
- 2003
- Full Text
- View/download PDF
18. The effects of alpha1A adrenergic receptor knockout on learning, memory, and seizure incidence (845.2)
- Author
-
Mariaha J. Lyons, Dianne M. Perez, Van A. Doze, Paul C. Simpson, Arthur Thorsen, Brianna Goldenstein, Albertine Cooper, Katie M. Collette, and Sarah G. Wood
- Subjects
medicine.medical_specialty ,Endocrinology ,Adrenergic receptor ,business.industry ,Internal medicine ,Incidence (epidemiology) ,Genetics ,medicine ,Learning memory ,business ,Molecular Biology ,Biochemistry ,Biotechnology - Published
- 2014
- Full Text
- View/download PDF
19. Hypotension, Autonomic Failure, and Cardiac Hypertrophy in Transgenic Mice Overexpressing the α1B-Adrenergic Receptor
- Author
-
Edward F. Plow, Robert J. Gaivin, Craig J. Daly, Michael J. Zuscik, David Waugh, John C. McGrath, Clare Deighan, Ann McGee, Annitta J. Morehead, Sean A. Ross, Dianne M. Perez, David Hellard, James D. Thomas, Dan Chalothorn, and Michael T. Piascik
- Subjects
Male ,Cardiac output ,medicine.medical_specialty ,Time Factors ,Epinephrine ,Hydrocortisone ,Adrenergic receptor ,Transgene ,Blood Pressure ,Cardiomegaly ,Mice, Transgenic ,Inositol 1,4,5-Trisphosphate ,Biology ,Kidney ,Biochemistry ,Mice ,Norepinephrine ,Phenylephrine ,Organ Culture Techniques ,Heart Rate ,Receptors, Adrenergic, alpha-1 ,Internal medicine ,Bradycardia ,Heart Septum ,medicine ,Animals ,Humans ,Promoter Regions, Genetic ,Pure autonomic failure ,Receptor ,Molecular Biology ,Mice, Knockout ,Dose-Response Relationship, Drug ,Body Weight ,Organ Size ,Cell Biology ,medicine.disease ,Femoral Artery ,Phenotype ,medicine.anatomical_structure ,Blood pressure ,Endocrinology ,Echocardiography ,Catecholamine ,Hypotension ,Artery ,medicine.drug - Abstract
alpha(1)-Adrenergic receptors (alpha(1A), alpha(1B), and alpha(1D)) are regulators of systemic arterial blood pressure and blood flow. Whereas vasoconstrictory action of the alpha(1A) and alpha(1D) subtypes is thought to be mainly responsible for this activity, the role of the alpha(1B)-adrenergic receptor (alpha(1B)AR) in this process is controversial. We have generated transgenic mice that overexpress either wild type or constitutively active alpha(1B)ARs. Transgenic expression was under the control of the isogenic promoter, thus assuring appropriate developmental and tissue-specific expression. Cardiovascular phenotypes displayed by transgenic mice included myocardial hypertrophy and hypotension. Indicative of cardiac hypertrophy, transgenic mice displayed an increased heart to body weight ratio, which was confirmed by the echocardiographic finding of an increased thickness of the interventricular septum and posterior wall. Functional deficits included an increased isovolumetric relaxation time, a decreased heart rate, and cardiac output. Transgenic mice were hypotensive and exhibited a decreased pressor response. Vasoconstrictory regulation by alpha(1B)AR was absent as shown by the lack of phenylephrine-induced contractile differences between ex vivo mesenteric artery preparations. Plasma epinephrine, norepinephrine, and cortisol levels were also reduced in transgenic mice, suggesting a loss of sympathetic nerve activity. Reduced catecholamine levels together with basal hypotension, bradycardia, reproductive problems, and weight loss suggest autonomic failure, a phenotype that is consistent with the multiple system atrophy-like neurodegeneration that has been reported previously in these mice. These results also suggest that this receptor subtype is not involved in the classic vasoconstrictory action of alpha(1)ARs that is important in systemic regulation of blood pressure.
- Published
- 2001
- Full Text
- View/download PDF
20. Phe-308 and Phe-312 in Transmembrane Domain 7 Are Major Sites of α1-Adrenergic Receptor Antagonist Binding
- Author
-
June Yun, Robert J. Gaivin, Pedro J. Gonzalez-Cabrera, Sean A. Ross, David Waugh, Dianne M. Perez, and Michael J. Zuscik
- Subjects
Agonist ,Chemistry ,medicine.drug_class ,Niguldipine ,Antagonist ,Cell Biology ,Biochemistry ,Partial agonist ,Cirazoline ,HYDIA ,Transmembrane domain ,chemistry.chemical_compound ,Competitive antagonist ,medicine ,Biophysics ,Molecular Biology - Abstract
Although agonist binding in adrenergic receptors is fairly well understood and involves residues located in transmembrane domains 3 through 6, there are few residues reported that are involved in antagonist binding. In fact, a major docking site for antagonists has never been reported in any G-protein coupled receptor. It has been speculated that antagonist binding is quite diverse depending upon the chemical structure of the antagonist, which can be quite different from agonists. We now report the identification of two phenylalanine residues in transmembrane domain 7 of the α1a-adrenergic receptor (Phe-312 and Phe-308) that are a major site of antagonist affinity. Mutation of either Phe-308 or Phe-312 resulted in significant losses of affinity (4–1200-fold) for the antagonists prazosin, WB4101, BMY7378, (+) niguldipine, and 5-methylurapidil, with no changes in affinity for phenethylamine-type agonists such as epinephrine, methoxamine, or phenylephrine. Interestingly, both residues are involved in the binding of all imidazoline-type agonists such as oxymetazoline, cirazoline, and clonidine, confirming previous evidence that this class of ligand binds differently than phenethylamine-type agonists and may be more antagonist-like, which may explain their partial agonist properties. In modeling these interactions with previous mutagenesis studies and using the current backbone structure of rhodopsin, we conclude that antagonist binding is docked higher in the pocket closer to the extracellular surface than agonist binding and appears skewed toward transmembrane domain 7.
- Published
- 2001
- Full Text
- View/download PDF
21. Characteristics for a Salt-bridge Switch Mutation of the α1b Adrenergic Receptor
- Author
-
Dianne M. Perez and James E. Porter
- Subjects
Agonist ,medicine.drug_class ,Mutant ,Cell Biology ,Alpha-1B adrenergic receptor ,Pharmacology ,Biology ,Biochemistry ,Transmembrane domain ,Aspartic acid ,medicine ,5-HT5A receptor ,Salt bridge ,Receptor ,Molecular Biology - Abstract
Agonist-dependent activation of the α1-adrenergic receptor is postulated to be initiated by disruption of an interhelical salt-bridge constraint between an aspartic acid (Asp-125) and a lysine residue (Lys-331) in transmembrane domains three and seven, respectively. Single point mutations that disrupt the charges of either of these residues results in constitutive activity. To validate this hypothesis, we used site-directed mutagenesis to switch the position of these amino acids to observe, if possible, regeneration of the salt-bridge reverses that the constitutive activity of the single point mutations. The transiently expressed switch mutant receptor displayed an altered pharmacological profile. The affinity of selective α1b-adrenergic receptor antagonists for the switch mutant (D125K/K331D) was no different from the wild-type α1b-adrenergic receptor, suggesting that both receptors are maintaining similar tertiary structures in the cell membrane. However, there was a significant 4–6-fold decrease in the affinity of protonated amine receptor agonists and a 3–6-fold increase in the affinity of carboxylated catechol derivatives for the switch mutant compared with the wild-type α1b-adrenergic receptor. This pharmacology is consistent with a reversed charge at position 125 in transmembrane domain three. Interestingly, the ability of either a negatively or positively charged agonist to generate soluble inositol phosphates was similar for both types of receptors. Finally, the switch mutant (D125K/K331D) displayed similar basal signaling activity as the wild-type receptor, reversing the constitutive activity of the single point mutations (D125K and K331D). This suggests an ionic constraint has been reformed in the switch mutant analogous to the restraint previously described for the wild-type α1b-adrenergic receptor. These results strongly establish the disruption of an electrostatic interaction as an initial step in the agonist-dependent activation of α1-adrenergic receptors.
- Published
- 1999
- Full Text
- View/download PDF
22. The Third Extracellular Loop of the β2-Adrenergic Receptor Can Modulate Receptor/G Protein Affinity
- Author
-
Ming Ming Zhao, Dianne M. Perez, and Robert J. Gaivin
- Subjects
Pharmacology ,Beta-3 adrenergic receptor ,Agonist ,Adrenergic receptor ,G protein ,medicine.drug_class ,Molecular Sequence Data ,Guanosine ,Biology ,chemistry.chemical_compound ,Biochemistry ,chemistry ,GTP-Binding Proteins ,Guanosine 5'-O-(3-Thiotriphosphate) ,COS Cells ,Cyclic AMP ,Biophysics ,medicine ,Animals ,Molecular Medicine ,Amino Acid Sequence ,Receptors, Adrenergic, beta-2 ,Signal transduction ,Receptor ,Cyclase activity - Abstract
Chimeric receptors of the beta2-adrenergic receptor in which the extracellular loops were replaced with the corresponding amino acids of the alpha1a-adrenergic receptor were generated to measure changes in alpha1-antagonist affinity. Although no changes in alpha1-antagonist affinity were measured in the beta2/alpha1a chimeras, a decreased IC50 (10-fold) for agonists as compared with wild type beta2 control was found because of the replacement of the third extracellular loop (EX3). These agonist high affinity changes were because of a greater proportion of high affinity sites (2-fold) that were convertible to low affinity sites with guanosine 5'-3-O-(thio)triphosphate. Adenylate cyclase activity evoked by the EX3 chimera showed commensurate increases in the basal signal transduction as well as the isoproterenol-stimulated potency, suggesting constitutive activity. However, unlike other constitutively active adrenergic receptor mutants in which the mutation causes G protein-independent changes, the mechanism of the EX3 chimera seems to be attributable to a greater ease with which the active ternary complex is formed because of a higher affinity/coupling of the G protein. Although the changes because of EX3 are indirect and most likely affect helical packing, they support an emerging hypothesis that G protein-coupled receptors have evolved their structure-function relationships to constrain the receptor in an inactive state.
- Published
- 1998
- Full Text
- View/download PDF
23. Identification of a Conserved Switch Residue Responsible for Selective Constitutive Activation of the β2-Adrenergic Receptor
- Author
-
James E. Porter, Michael J. Zuscik, Robert J. Gaivin, and Dianne M. Perez
- Subjects
Sodium-Hydrogen Exchangers ,Intracellular pH ,Molecular Sequence Data ,Mutant ,Stimulation ,Biology ,medicine.disease_cause ,Biochemistry ,GTP-Binding Proteins ,Cyclic AMP ,medicine ,Animals ,Amino Acid Sequence ,Receptor ,Molecular Biology ,Conserved Sequence ,Mutation ,Sequence Homology, Amino Acid ,Isoproterenol ,Cell Biology ,Transfection ,Adrenergic beta-Agonists ,Hydrogen-Ion Concentration ,Molecular biology ,Kinetics ,Transmembrane domain ,COS Cells ,Mutagenesis, Site-Directed ,Receptors, Adrenergic, beta-2 ,Efflux - Abstract
A cysteine-to-phenylalanine mutation of residue 116 in the third transmembrane domain of the beta2-adrenergic receptor caused selective constitutive activation of Na+/H+ exchange through a pathway not involving cAMP. This selectivity was identified by comparing binding and signaling characteristics of wild-type (WT) versus C116F mutant receptors transiently transfected into COS-1 cells. Indicating constitutive activity, ligand binding to the C116F mutant showed a 78-fold higher than WT affinity for isoproterenol and a 40-fold lower than WT affinity for ICI 118551. Although agonist-independent activation of cAMP production was not exhibited by the C116F mutant, a constitutive stimulation of the Na+/H+ exchanger (NHE1) was observed. This was identified by measuring either basal intracellular pH (pHi) or rate of pHi recovery from cellular acid load. Due to a higher rate of H+ efflux through NHE1, C116F transfectants exhibited a significantly higher pHi (7.42) than did WT transfectants (7.1). Furthermore, the rate of pHi recovery from acid load facilitated by NHE1 was 2.1-fold faster in mutant transfectants than in WT transfectants. The lower rate seen in the WT case was stimulated by epinephrine, and the higher rate seen in the mutant case was inhibited by ICI 118551. These findings, which show that a C116F mutation of the beta2-adrenergic receptor evokes selective constitutive coupling to NHE1 over cAMP, form the basis of our prediction that multiple and distinct activation states can exist in G protein-coupled receptors.
- Published
- 1998
- Full Text
- View/download PDF
24. Long-term α1B-adrenergic receptor activation shortens lifespan, while α1A-adrenergic receptor stimulation prolongs lifespan in association with decreased cancer incidence
- Author
-
Donald A. Sens, Katie M. Collette, Robert S. Papay, Mariaha J. Lyons, Haley M. Amoth, Dianne M. Perez, Xu Dong Zhou, and Van A. Doze
- Subjects
Male ,medicine.medical_specialty ,Aging ,Time Factors ,Adrenergic receptor ,Transgene ,Central nervous system ,Longevity ,Adrenergic ,Stimulation ,Mice, Transgenic ,Biology ,Article ,Mice ,Internal medicine ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,Receptor ,Cancer ,General Medicine ,Neoplasms, Experimental ,medicine.disease ,Molecular medicine ,Gene Expression Regulation, Neoplastic ,Endocrinology ,medicine.anatomical_structure ,Female ,Geriatrics and Gerontology ,Follow-Up Studies ,Signal Transduction - Abstract
The α1-adrenergic receptor (α1AR) subtypes, α1AAR and α1BAR, have differential effects in the heart and central nervous system. Long-term stimulation of the α1AAR subtype prolongs lifespan and provides cardio- and neuro-protective effects. We examined the lifespan of constitutively active mutant (CAM)-α1BAR mice and the incidence of cancer in mice expressing the CAM form of either the α1AAR (CAM-α1AAR mice) or α1BAR. CAM-α1BAR mice have a significantly shortened lifespan when compared with wild-type (WT) animals; however, the effect was sex dependent. Female CAM-α1BAR mice lived significantly shorter lives, while the median lifespan of male CAM-α1BAR mice was not different when compared with that of WT animals. There was no difference in the incidence of cancer in either sex of CAM-α1BAR mice. The incidence of cancer was significantly decreased in CAM-α1AAR mice when compared with that in WT, and no sex-dependent effects were observed. Further study is warranted on cancer incidence after activation of each α1AR subtype and the effect of sex on lifespan following activation of the α1BAR. The implications of a decrease in cancer incidence following long-term α1AAR stimulation could lead to improved treatments for cancer.
- Published
- 2014
25. Novel proteins associated with human dilated cardiomyopathy: selective reduction in α(1A)-adrenergic receptors and increased desensitization proteins
- Author
-
Ting Shi, Christine S. Moravec, and Dianne M. Perez
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,medicine.medical_specialty ,Adrenergic receptor ,Proteome ,Adrenergic ,Biology ,Proteomics ,Biochemistry ,Article ,Receptors, G-Protein-Coupled ,Internal medicine ,Receptors, Adrenergic, alpha-1 ,medicine ,Humans ,Receptor ,Molecular Biology ,Heart Failure ,Dilated cardiomyopathy ,Cell Biology ,Middle Aged ,medicine.disease ,Cell biology ,Endocrinology ,Heart failure ,Signal transduction ,Signal Transduction - Abstract
Therapeutics to treat human heart failure and the identification of proteins associated with heart failure are still limited. We analyzed α1-adrenergic receptor subtypes in human heart failure and performed proteomic analysis on more uniform samples to identify novel proteins associated with human heart failure. Six failing hearts with end-stage dilated cardiomyopathy and four non-failing heart controls were subjected to proteomic analysis. Out of 48 identified proteins, 26 proteins were redundant between samples. 10 of these 26 proteins were previously reported to be associated with heart failure. Of the newly identified proteins, we found several muscle proteins and mitochondrial/electron transport proteins, while novel were functionally similar to previous reports. However, we also found novel proteins involved in functional classes such as β-oxidation and G-protein coupled receptor signaling and desensitization not previously associated with heart failure. We also performed radioligand-binding studies on the heart samples and confirmed a large loss of β1-adrenergic receptors in end-stage dilated cardiomyopathy but also found a selective decrease in the α1A-adrenergic receptor subtype not previously reported. We have identified new proteins and functional categories associated with end-stage dilated cardiomyopathy. We also report that similar to the previously characterized loss of β1-adrenergic receptors in heart failure, there is also a concomitant loss of α1A-adrenergic receptors, which are considered cardioprotective proteins.
- Published
- 2013
26. α 1 -Adrenergic Receptor Subtypes
- Author
-
Dianne M. Perez, Robert M. Graham, John Hwa, and Michael T. Piascik
- Subjects
Sympathetic nervous system ,Physiology ,Molecular Sequence Data ,Population ,Norepinephrine ,medicine ,Animals ,Humans ,Amino Acid Sequence ,RNA, Messenger ,Receptor ,education ,G protein-coupled receptor ,education.field_of_study ,Molecular Structure ,biology ,Receptors, Adrenergic, alpha ,Membrane glycoproteins ,medicine.anatomical_structure ,Epinephrine ,Immunology ,biology.protein ,Signal transduction ,Cardiology and Cardiovascular Medicine ,Neuroscience ,Signal Transduction ,medicine.drug - Abstract
The α1ARs are important mediators of sympathetic nervous system responses, particularly those involved in cardiovascular homeostasis, such as arteriolar smooth muscle constriction and cardiac contraction.1 2 In addition, α1ARs have more recently been implicated in the pathogenesis of cardiac hypertrophy, in ischemia-induced cardiac arrhythmias, and in ischemic preconditioning.1 3 Like other ARs, α1ARs are activated by the catecholamines, norepinephrine and epinephrine. They are intrinsic membrane glycoproteins and are members of the GPCR superfamily. Over the past 10 to 15 years, data initially based on functional, radioligand, and biochemical studies have accumulated, indicating that the α1ARs are a heterogeneous group of distinct but related proteins. This conclusion has been confirmed with the molecular cloning of three distinct α1-receptor subtypes, although until recently discrepancies between the properties of the cloned expressed receptors and those characterized pharmacologically and biochemically have led to confusion in the classification of α1-receptor subtypes and their coupled effector responses. As detailed in the present review, much of this confusion has now been clarified for the three cloned α1ARs. These and other recent insights into the molecular structure, function, and signaling of α1ARs, the control of α1AR-gene expression, and pharmacological evidence for additional α1AR subtypes will be reviewed here. For additional information, the reader is also referred to several previous reviews of α1ARs.4 5 6 7 Functional studies of AR responses, particularly from the laboratories of McGrath8 and Ruffolo,9 provided the initial evidence that there may be subtypes of α1ARs. These studies indicated that postjunctional responses mediated by α1ARs could not be explained adequately on the basis of a single population of receptors. This concept was further advanced …
- Published
- 1996
- Full Text
- View/download PDF
27. Chimeras of α1-Adrenergic Receptor Subtypes Identify Critical Residues That Modulate Active State Isomerization
- Author
-
Robert M. Graham, Dianne M. Perez, and John Hwa
- Subjects
Agonist ,Epinephrine ,medicine.drug_class ,Inositol Phosphates ,Recombinant Fusion Proteins ,Inositol 1,4,5-Trisphosphate ,Transfection ,Biochemistry ,Structure-Activity Relationship ,Phospholipase A2 ,Cricetinae ,Chlorocebus aethiops ,medicine ,Animals ,Receptor ,Molecular Biology ,Cells, Cultured ,Binding selectivity ,chemistry.chemical_classification ,Arachidonic Acid ,Phospholipase C ,biology ,Wild type ,Membrane Proteins ,Cell Biology ,Receptors, Adrenergic, alpha ,Transmembrane protein ,Rats ,Amino acid ,chemistry ,biology.protein ,Signal Transduction - Abstract
We have identified previously two amino acids, one in each of the fifth and sixth transmembrane segments of both the alpha1a-adrenergic receptor and the alpha1b-adrenergic receptor (AR), that account almost entirely for the selectivity of agonist binding by these receptor subtypes (Hwa, J., Graham, R. M., and Perez, D. M. (1995) J. Biol. Chem. 270, 23189-23195). Thus reversal of these two residues, from those found in the native receptor of one subtype to those in the other subtype, produces complementary changes in subtype selectivity of agonist binding. Here we show that mutating only one of these residues in either the alpha1b-AR or the alpha1a-AR to the corresponding residue in the other subtype (Ala204 --Val for the alpha1b; Met292 --Leu for the alpha1a-AR) results in chimeras that are constitutively active for signaling by both the phospholipase C and phospholipase A2 pathways. This is evident by an increased affinity for agonists, increased basal phospholipase C and phospholipase A2 activation, and increased agonist potency. Although mutation of the other residue involved in agonist binding selectivity, to the corresponding residue in the other subtype (Leu314 --Met for the alpha1b-AR; Val185 --Ala for the alpha1a-AR) does not alter receptor binding or signaling, per se, when combined with the corresponding constitutively activating mutations, the resulting chimeras, Ala204 --Val/Leu314 --Met ( alpha1b-AR) and Val185 --Ala/Met292 --Leu ( alpha1a-AR), display wild type ligand binding and signaling. A simple interpretation of these results is that the alpha1a- and alpha1b-ARs possess residues that critically modulate isomerization from the basal state, R, to the active state R*, and that the native receptor structures have evolved to select residues that repress active state isomerization. It is likely that the residues identified here modulate important interhelical interactions between the fifth and sixth transmembrane segments that inhibit or promote receptor signaling.
- Published
- 1996
- Full Text
- View/download PDF
28. Alpha1A Adrenergic Receptor Stimulation Improves Mood in Mice
- Author
-
Shelby L. Poitra, Van A. Doze, Anna H. Fossen, Elizabeth Luger, Lee N. Wilkie, Bethany A. Davis, Dianne M. Perez, Jim R. Haselton, and Katie M. Collette
- Subjects
0303 health sciences ,medicine.medical_specialty ,Adrenergic receptor ,business.industry ,Stimulation ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Mood ,Endocrinology ,Internal medicine ,Genetics ,Medicine ,business ,Molecular Biology ,030217 neurology & neurosurgery ,030304 developmental biology ,Biotechnology - Published
- 2012
- Full Text
- View/download PDF
29. Long-Term α1A-Adrenergic Receptor Stimulation Improves Synaptic Plasticity, Cognitive Function, Mood, and Longevity
- Author
-
Mariaha J. Lyons, Dianne M. Perez, Bethany A. Davis, Brian Nelson, Elizabeth Luger, Manveen K Gupta, Paul C. Simpson, Sarah G. Wood, James Haselton, Katie M. Collette, Robert S. Papay, Van A. Doze, and Brianna Goldenstein
- Subjects
Agonist ,Male ,medicine.medical_specialty ,medicine.drug_class ,Long-Term Potentiation ,Longevity ,Hippocampus ,Stimulation ,Mice, Transgenic ,Biology ,chemistry.chemical_compound ,Mice ,Cognition ,Organ Culture Techniques ,Internal medicine ,Receptors, Adrenergic, alpha-1 ,Neuroplasticity ,medicine ,Animals ,Pharmacology ,Mice, Knockout ,Neuronal Plasticity ,Neurogenesis ,Long-term potentiation ,Articles ,Cirazoline ,Mice, Inbred C57BL ,Affect ,Endocrinology ,chemistry ,Synaptic plasticity ,Synapses ,Mice, Inbred CBA ,Molecular Medicine ,Female ,Adrenergic alpha-1 Receptor Agonists - Abstract
The role of α(1)-adrenergic receptors (α(1)ARs) in cognition and mood is controversial, probably as a result of past use of nonselective agents. α(1A)AR activation was recently shown to increase neurogenesis, which is linked to cognition and mood. We studied the effects of long-term α(1A)AR stimulation using transgenic mice engineered to express a constitutively active mutant (CAM) form of the α(1A)AR. CAM-α(1A)AR mice showed enhancements in several behavioral models of learning and memory. In contrast, mice that have the α(1A)AR gene knocked out displayed poor cognitive function. Hippocampal brain slices from CAM-α(1A)AR mice demonstrated increased basal synaptic transmission, paired-pulse facilitation, and long-term potentiation compared with wild-type (WT) mice. WT mice treated with the α(1A)AR-selective agonist cirazoline also showed enhanced cognitive functions. In addition, CAM-α(1A)AR mice exhibited antidepressant and less anxious phenotypes in several behavioral tests compared with WT mice. Furthermore, the lifespan of CAM-α(1A)AR mice was 10% longer than that of WT mice. Our results suggest that long-term α(1A)AR stimulation improves synaptic plasticity, cognitive function, mood, and longevity. This may afford a potential therapeutic target for counteracting the decline in cognitive function and mood associated with aging and neurological disorders.
- Published
- 2011
30. Cardiac and neuroprotection regulated by α(1)-adrenergic receptor subtypes
- Author
-
Van A. Doze and Dianne M. Perez
- Subjects
Sympathetic nervous system ,Context (language use) ,Protective Agents ,Biochemistry ,Neuroprotection ,Article ,Norepinephrine ,Receptors, Adrenergic, alpha-1 ,Neuroplasticity ,Medicine ,Animals ,Humans ,Molecular Biology ,Neurons ,business.industry ,Myocardium ,Neurogenesis ,Cell Biology ,medicine.disease ,medicine.anatomical_structure ,Cytoprotection ,Heart failure ,Anesthesia ,Ischemic preconditioning ,business ,Neuroscience ,medicine.drug - Abstract
Sympathetic nervous system regulation by the α(1)-adrenergic receptor (AR) subtypes (α(1A), α(1B), α(1D)) is complex, whereby chronic activity can be either detrimental or protective for both heart and brain function. This review will summarize the evidence that this dual regulation can be mediated through the different α(1)-AR subtypes in the context of cardiac hypertrophy, heart failure, apoptosis, ischemic preconditioning, neurogenesis, locomotion, neurodegeneration, cognition, neuroplasticity, depression, anxiety, epilepsy, and mental illness.
- Published
- 2011
31. alpha1-Adrenergic receptors regulate neurogenesis and gliogenesis
- Author
-
Dianne M. Perez, Ting Shi, Manveen K Gupta, Chris Jurgens, Robert S. Papay, Robert J. Gaivin, and Van A. Doze
- Subjects
medicine.medical_specialty ,Cellular differentiation ,Neurogenesis ,Green Fluorescent Proteins ,Subventricular zone ,Stimulation ,Mice, Transgenic ,Biology ,Mice ,Phosphatidylinositol 3-Kinases ,Cell Movement ,Interneurons ,Neurosphere ,Internal medicine ,Receptors, Adrenergic, alpha-1 ,Spheroids, Cellular ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Receptor ,Gliogenesis ,Pharmacology ,Neurons ,Imidazoles ,Cell Differentiation ,Articles ,Immunohistochemistry ,Neural stem cell ,Cell biology ,Endocrinology ,medicine.anatomical_structure ,Animals, Newborn ,Molecular Medicine ,Adrenergic alpha-1 Receptor Agonists ,Neuroglia ,Biomarkers - 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.
- Published
- 2009
32. α 1 Adrenergic receptor regulation of interneuron function
- Author
-
Dianne M. Perez, Pat A Carr, Chris Jurgens, Van A. Doze, and Chris A. Knudson
- Subjects
medicine.anatomical_structure ,Interneuron ,Chemistry ,Genetics ,medicine ,Molecular Biology ,Biochemistry ,Alpha-1A adrenergic receptor ,Function (biology) ,Biotechnology ,Alpha-1 adrenergic receptor ,Cell biology - Published
- 2009
- Full Text
- View/download PDF
33. Alpha‐1A adrenergic receptor regulation of learning and memory in mice
- Author
-
Sarah Boese, Brianna Goldenstein, Danielle Schlosser, Van A. Doze, James Haselton, Dianne M. Perez, Patrick A. Carr, and Chris A. Knudson
- Subjects
Beta-3 adrenergic receptor ,medicine.medical_specialty ,Endocrinology ,Chemistry ,Internal medicine ,Genetics ,medicine ,Alpha-1B adrenergic receptor ,Molecular Biology ,Biochemistry ,Alpha-1A adrenergic receptor ,Biotechnology - Published
- 2009
- Full Text
- View/download PDF
34. Alpha‐1 adrenergic receptor regulation of seizures and neurodegeneration
- Author
-
Dianne M. Perez, Patrick A. Carr, Brianna Goldenstein, Chris Jurgens, Chris A. Knudson, Jessica Lichter, and Van A. Doze
- Subjects
medicine.medical_specialty ,Endocrinology ,business.industry ,Internal medicine ,Neurodegeneration ,Genetics ,medicine ,medicine.disease ,business ,Molecular Biology ,Biochemistry ,Biotechnology ,Alpha-1 adrenergic receptor - Published
- 2008
- Full Text
- View/download PDF
35. Beta 1-adrenergic receptor autoantibodies mediate dilated cardiomyopathy by agonistically inducing cardiomyocyte apoptosis
- Author
-
Vincent K. Tuohy, Qing Wang, Cengiz Z. Altuntas, Peter J. Wickley, Dianne M. Perez, Pamela Clark, Justin M. Johnson, Sandro L. Yong, Derek S. Damron, Zoran B. Popović, Daniel Jane-wit, and Marc S. Penn
- Subjects
Cardiomyopathy, Dilated ,Male ,Programmed cell death ,Cardiomyopathy ,Apoptosis ,medicine.disease_cause ,Autoimmunity ,Autoimmune Diseases ,Mice ,Physiology (medical) ,Idiopathic dilated cardiomyopathy ,medicine ,Animals ,Humans ,Myocytes, Cardiac ,Protein kinase A signaling ,Immunoadsorption ,Receptor ,Cells, Cultured ,Autoantibodies ,business.industry ,Autoantibody ,Adrenergic beta-Agonists ,medicine.disease ,Immunology ,Receptors, Adrenergic, beta-1 ,Cardiology and Cardiovascular Medicine ,business - Abstract
Background— Antibodies to the β 1 -adrenergic receptor (β 1 AR) are detected in a substantial number of patients with idiopathic dilated cardiomyopathy (DCM). The mechanism whereby these autoantibodies exert their pathogenic effect is unknown. Here, we define a causal mechanism whereby β 1 AR-specific autoantibodies mediate noninflammatory cardiomyocyte cell death during murine DCM. Methods and Results— We used the β 1 AR protein as an immunogen in SWXJ mice and generated a polyclonal battery of autoantibodies that showed selective binding to the β 1 AR. After transfer into naive male hosts, β 1 AR antibodies elicited fulminant DCM at high frequency. DCM was attenuated after immunoadsorption of β 1 AR IgG before transfer and by selective pharmacological antagonism of host β 1 AR but not β 2 AR. We found that β 1 AR autoantibodies shifted the β 1 AR into the agonist-coupled high-affinity state and activated the canonical cAMP-dependent protein kinase A signaling pathway in cardiomyocytes. These events led to functional alterations in intracellular calcium handling and contractile function. Sustained agonism by β 1 AR autoantibodies elicited caspase-3 activation, cardiomyocyte apoptosis, and DCM in vivo, and these processes were prevented by in vivo treatment with the pan-caspase inhibitor Z-VAD-FMK. Conclusions— Our data show how β 1 AR-specific autoantibodies elicit DCM by agonistically inducing cardiomyocyte apoptosis.
- Published
- 2007
36. Dominance of the alpha1B-adrenergic receptor and its subcellular localization in human and TRAMP prostate cancer cell lines
- Author
-
Robert J. Gaivin, Manveen K Gupta, Ting Shi, Dianne M. Perez, and Dan F. McCune
- Subjects
Agonist ,Male ,medicine.medical_specialty ,Epinephrine ,medicine.drug_class ,Cell ,Biology ,urologic and male genital diseases ,Biochemistry ,Piperazines ,Mice ,DU145 ,Internal medicine ,Cell Line, Tumor ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,Humans ,Receptor ,Molecular Biology ,Adrenergic alpha-Antagonists ,Dose-Response Relationship, Drug ,Prostatic Neoplasms ,Cell Biology ,Transfection ,Molecular biology ,Adrenergic Agonists ,Endocrinology ,medicine.anatomical_structure ,Cell culture ,Intracellular ,Tramp ,Protein Binding - 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.
- Published
- 2007
37. Alpha‐1A Adrenergic Receptor Overexpression Protects Hippocampal Interneurons
- Author
-
Dianne M. Perez, Chris A. Knudson, Van A. Doze, and Patrick A. Carr
- Subjects
medicine.medical_specialty ,Endocrinology ,Chemistry ,Internal medicine ,Genetics ,medicine ,Hippocampal formation ,Molecular Biology ,Biochemistry ,Alpha-1A adrenergic receptor ,Biotechnology - Published
- 2007
- Full Text
- View/download PDF
38. Effects of alpha1D-adrenergic receptors on shedding of biologically active EGF in freshly isolated lacrimal gland epithelial cells
- Author
-
Robin R. Hodges, Chika Funaki, Robert J. Gaivin, Darlene A. Dartt, L. Chen, Driss Zoukhri, and Dianne M. Perez
- Subjects
Male ,Transcriptional Activation ,medicine.medical_specialty ,Adrenergic receptor ,Physiology ,Lacrimal gland ,Cell Separation ,Biology ,Matrix Metalloproteinase Inhibitors ,Lacrimal apparatus ,Binding, Competitive ,Rats, Sprague-Dawley ,Transactivation ,Phenylephrine ,Epidermal growth factor ,Internal medicine ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,RNA, Messenger ,Receptor ,Mitogen-Activated Protein Kinase 3 ,Epidermal Growth Factor ,Lacrimal Apparatus ,Epithelial Cells ,Cell Biology ,Cell biology ,Rats ,Enzyme Activation ,ErbB Receptors ,Endocrinology ,medicine.anatomical_structure ,Ectodomain ,Culture Media, Conditioned ,Adrenergic alpha-1 Receptor Antagonists ,Adrenergic alpha-1 Receptor Agonists ,Signal transduction ,hormones, hormone substitutes, and hormone antagonists - 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 α1-adrenergic receptors (ARs), namely, α1B- and α1D-ARs. α1D-ARs mediate phenylephrine (an α1-adrenergic agonist)-induced protein secretion and activation of p42/p44 MAPK, because the α1D-AR inhibitor BMY-7378, but not the α1A-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, α1-adrenergic agonists activate the α1D-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.
- Published
- 2006
39. The Adrenergic Receptors
- Author
-
Dianne M. Perez
- Subjects
Beta-3 adrenergic receptor ,medicine.medical_specialty ,Endocrinology ,Adrenergic receptor ,Chemistry ,Internal medicine ,medicine ,Adrenergic ,Alpha-1B adrenergic receptor ,Alpha-1A adrenergic receptor - Published
- 2006
- Full Text
- View/download PDF
40. Fentanyl attenuates alpha1B-adrenoceptor-mediated pulmonary artery contraction
- Author
-
Daniel F. McCune, Paul A. Murray, Dianne M. Perez, Ju Tae Sohn, and Xueqin Ding
- Subjects
Agonist ,Contraction (grammar) ,Adrenergic receptor ,medicine.drug_class ,Rauwolscine ,Pharmacology ,In Vitro Techniques ,Pulmonary Artery ,Clonidine ,Piperazines ,Fentanyl ,chemistry.chemical_compound ,Phenylephrine ,Dogs ,Chloroethylclonidine ,Receptors, Adrenergic, alpha-1 ,medicine ,Prazosin ,Animals ,Dose-Response Relationship, Drug ,business.industry ,Hydrogen-Ion Concentration ,Anesthesiology and Pain Medicine ,chemistry ,Vasoconstriction ,business ,Anesthetics, Intravenous ,medicine.drug - Abstract
Background The authors tested the hypothesis that the intravenous anesthetic fentanyl would attenuate the pulmonary vasoconstrictor response to alpha1-adrenoceptor activation. They also investigated the alpha1-adrenoceptor subtypes that could potentially mediate this effect of fentanyl. Methods Endothelium-denuded canine pulmonary arterial rings were suspended for isometric tension recording. Dose-response curves for the alpha1-adrenoceptor agonist phenylephrine were generated in the absence and presence of fentanyl. The effects of inhibiting alpha2 (rauwolscine), alpha1 (prazosin), alpha1A (5-methylurapidil), alpha1B (chloroethylclonidine), and alpha1D (BMY 7378) adrenoceptors on phenylephrine contraction were also investigated. Receptor "protection" studies were performed to investigate the specific role of alpha1B adrenoceptors in mediating fentanyl-induced changes in phenylephrine contraction. Finally, competition binding studies were performed in rat-1 fibroblasts stably transfected with human alpha1-adrenoceptor complementary DNAs corresponding to the alpha1A-, alpha1B-, or alpha1D-adrenoceptor subtypes to directly assess whether fentanyl can compete for the alpha1-adrenoceptor activation pocket. Results Fentanyl attenuated phenylephrine contraction in a dose-dependent fashion. Rauwolscine had no effect on phenylephrine contraction. Phenylephrine contraction was inhibited by prazosin and abolished by chloroethylclonidine but was relatively resistant to inhibition by 5-methylurapidil and BMY 7378. Pretreatment with fentanyl before exposure to chloroethylclonidine increased the maximal contractile response to phenylephrine compared to chloroethylclonidine pretreatment alone. Competition binding studies revealed that fentanyl binds to all three alpha1-adrenoceptor subtypes, with a fivefold greater affinity for the alpha1B-adrenoceptor compared with the alpha1D-adrenoceptor subtype. Conclusion Phenylephrine-induced contraction is primarily mediated by alpha1B-adrenoceptor activation in canine pulmonary artery. Fentanyl attenuates phenylephrine contraction by binding to alpha1B adrenoceptors.
- Published
- 2005
41. Multiple signaling states of G-protein-coupled receptors
- Author
-
Sadashiva S. Karnik and Dianne M. Perez
- Subjects
Agonist ,Models, Molecular ,Rhodopsin ,medicine.drug_class ,Protein Conformation ,media_common.quotation_subject ,Plasma protein binding ,Pharmacology ,Receptors, G-Protein-Coupled ,Desensitization (telecommunications) ,medicine ,Animals ,Humans ,Internalization ,Receptor ,G protein-coupled receptor ,media_common ,biology ,Chemistry ,Mutation ,biology.protein ,Molecular Medicine ,Signal transduction ,Neuroscience ,Protein Binding ,Signal Transduction - Abstract
Studies have been amassed in the past several years indicating that an agonist can conform a receptor into an activation state that is dependent upon an intrinsic property of the agonist usually based upon its chemical composition. Theoretically, each different agonist could impart its own unique activation state. Evidence for multiple signaling states for the G-protein-coupled receptors will be reviewed and is derived from many different pharmacological behaviors: efficacy, kinetics, protean agonism, differential desensitization and internalization, inverse agonism, and fusion chimeras. A recent extension of the ternary complex model is suggested by evidence that the different processes that govern deactivation, such as desensitization and internalization, is also regulated by conformers specific to the agonist. Rhodopsin may serve as a primer for the study of multiple activation states. Therapeutic implications that utilize multiple signaling states hold vast promise in the rationale design of drugs.
- Published
- 2005
42. A Mouse Model for Multiple System Atrophy
- Author
-
Dianne M. Perez
- Subjects
Sympathetic nervous system ,Cerebellar ataxia ,Adrenergic receptor ,business.industry ,Parkinsonism ,Central nervous system ,medicine.disease ,Norepinephrine ,medicine.anatomical_structure ,Atrophy ,Epinephrine ,Medicine ,medicine.symptom ,business ,Neuroscience ,medicine.drug - Abstract
Multiple system atrophy (MSA) is an adult-onset sporadic progressive neurodegenerative disorder of unknown etiology that is clinically characterized by the variable combination of signs of autonomic failure parkinsonism, cerebellar ataxia, and pyramidal signs. MSA affects both men and women in their 6th decade and progresses relentlessly until death after an average of nine years' survival from the onset of symptoms. Currently available therapeutic strategies for MSA are to keep the patient comfortable and to provide some respite for the patient's symptoms. To date, there are no treatment strategies that arrest or delay the progression of MSA. This chapter focuses on a transgenic mouse model that systemically overexpresses the α 1s -adrenergic receptor. Adrenergic receptors mediate the sympathetic nervous system by binding epinephrine and norepinephrine. The role of this receptor in the central nervous system is unknown but is thought to be stimulatory in nature, to affect the release of other neurotransmitters, and to be involved in locomotion. Nevertheless, this mouse model is useful in deciphering the neurotransmission pathways involved with MSA and may lead to some therapeutic recourse for patients.
- Published
- 2005
- Full Text
- View/download PDF
43. Differential regulation of the cell cycle by alpha1-adrenergic receptor subtypes
- Author
-
Dianne M. Perez, Pedro J. Gonzalez-Cabrera, Ting Shi, June Yun, Dan F. McCune, and Boyd R. Rorabaugh
- Subjects
medicine.medical_specialty ,Cell cycle checkpoint ,Epinephrine ,Transcription, Genetic ,Myocytes, Smooth Muscle ,Stimulation ,Cell Count ,Cell Cycle Proteins ,Cycloheximide ,Biology ,PC12 Cells ,S Phase ,chemistry.chemical_compound ,Endocrinology ,Internal medicine ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,Humans ,Receptor ,Cyclin ,Kinase ,Tumor Suppressor Proteins ,G1 Phase ,Cell cycle ,Fibroblasts ,Flow Cytometry ,Adrenergic Agonists ,Cyclin-Dependent Kinases ,Cell biology ,Rats ,chemistry ,Cell Division ,Cyclin-Dependent Kinase Inhibitor p27 ,medicine.drug - Abstract
Alpha(1)-Adrenergic receptors have been implicated in growth-promoting pathways. A microarray study of individual alpha(1)-adrenergic receptor subtypes (alpha(1A), alpha(1B), and alpha(1D)) expressed in Rat-1 fibroblasts revealed that epinephrine altered the transcription of several cell cycle regulatory genes in a direction consistent with the alpha(1A)- and alpha(1D)-adrenergic receptors mediating G(1)-S cell cycle arrest and the alpha(1B-)mediating cell-cycle progression. A time course indicated that in alpha(1A) cells, epinephrine stimulated a G(1)-S arrest, which began after 8 h of stimulation and maximized at 16 h, at which point was completely blocked with cycloheximide. The alpha(1B)-adrenergic receptor profile also showed unchecked cell cycle progression, even under low serum conditions and induced foci formation. The G(1)-S arrest induced by alpha(1A)- and alpha(1D)-adrenergic receptors was associated with decreased cyclin-dependent kinase-6 and cyclin E-associated kinase activities and increased expression of the cyclin-dependent kinase inhibitor p27(Kip1), all of which were blocked by prazosin. There were no differences in kinase activities and/or expression of p27(Kip1) in epinephrine alpha(1B)-AR fibroblasts, although the microarray did indicate differences in p27(Kip1) RNA levels. Cell counts proved the antimitotic effect of epinephrine in alpha(1A) and alpha(1D) cells and indicated that alpha(1B)-adrenergic receptor subtype expression was sufficient to cause proliferation of Rat-1 fibroblasts independent of agonist stimulation. Analysis in transfected PC12 cells also confirmed the alpha(1A)- and alpha(1B)-adrenergic receptor effect. The alpha(1B)-subtype native to DDT1-MF2 cells, a smooth muscle cell line, caused progression of the cell cycle. These results indicate that the alpha(1A)- and alpha(1D)-adrenergic receptors mediate G(1)-S cell-cycle arrest, whereas alpha(1B)-adrenergic receptor expression causes a cell cycle progression and may induce transformation in sensitive cell lines.
- Published
- 2004
44. Differential cardiovascular regulatory activities of the alpha 1B- and alpha 1D-adrenoceptor subtypes
- Author
-
Gozoh Tsujimoto, Bradley B. Keller, Akito Tanoue, Dan Chalothorn, Ginell R. Post, Michael T. Piascik, Kimimasa Tobita, Robert D. Lasley, Dianne M. Perez, Stephanie E. Edelmann, and Dan F. McCune
- Subjects
Genetically modified mouse ,medicine.medical_specialty ,Mice, Transgenic ,Electrocardiography ,Mice ,Phenylephrine ,Internal medicine ,Receptors, Adrenergic, alpha-1 ,medicine ,Extracellular ,Cyclic AMP ,Animals ,Receptor ,Aorta ,Pharmacology ,Mice, Knockout ,Dose-Response Relationship, Drug ,Chemistry ,Kinase ,Myocardium ,Heart ,Smooth muscle contraction ,Adrenergic Agonists ,Perfusion ,Endocrinology ,Vasoconstriction ,Knockout mouse ,Molecular Medicine ,Mitogen-Activated Protein Kinases ,Vascular smooth muscle contraction ,medicine.drug - Abstract
The regulation of cardiac and vascular function by the alpha 1B- and alpha 1D-adrenoceptors (ARs) has been assessed in two lines of transgenic mice, one over-expressing a constitutively active alpha 1B-AR mutation (alpha 1B-ARC128F) and the other an alpha 1D-AR knockout line. The advantage of using mice expressing a constitutively active alpha 1B-AR is that the receptor is tonically active, thus avoiding the use of nonselective agonists that can activate all subtypes. In hearts from animals expressing alpha 1B-ARC128F, the activities of the mitogen-activated protein kinases, extracellular signal-regulated kinase, and c-Jun N-terminal kinase were significantly elevated compared with nontransgenic control animals. Mice over-expressing the alpha 1B-ARC128F had echocardiographic evidence of contractile dysfunction and increases in chamber dimensions. In isolated-perfused hearts or left ventricular slices from alpha 1B-ARC128F-expressing animals, the ability of isoproterenol to increase contractile force or increase cAMP levels was significantly decreased. In contrast to the prominent effects on the heart, constitutive activation of the alpha 1B-AR had little effect on the ability of phenylephrine to induce vascular smooth muscle contraction in the isolated aorta. The ability of phenylephrine to stimulate coronary vasoconstriction was diminished in alpha 1D-AR knockout mice. In alpha 1D-AR knockout animals, no negative effects on cardiac contractile function were noted. These results show that the alpha1-ARs regulate distinctly different physiologic processes. The alpha 1B-AR appears to be involved in the regulation of cardiac growth and contractile function, whereas the alpha 1D-AR is coupled to smooth muscle contraction and the regulation of systemic arterial blood pressure.
- Published
- 2003
45. Overexpression of the alpha1B-adrenergic receptor causes apoptotic neurodegeneration: multiple system atrophy
- Author
-
Robert J. Gaivin, Dianne M. Perez, Sean A. Ross, David Waugh, David A. Morilak, Michael J. Zuscik, and Scott A. Sands
- Subjects
Genetically modified mouse ,medicine.medical_specialty ,Adrenergic receptor ,Central nervous system ,Substantia nigra ,Apoptosis ,Mice, Transgenic ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Mice ,Atrophy ,Seizures ,Internal medicine ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,Receptor ,Cerebral Cortex ,Neurodegeneration ,Age Factors ,Neurodegenerative Diseases ,Parkinson Disease ,General Medicine ,medicine.disease ,Hindlimb ,Substantia Nigra ,Endocrinology ,medicine.anatomical_structure ,Phenotype ,Cerebral cortex - Abstract
Progress toward elucidating the function of alpha1B-adrenergic receptors (alpha1BARs) in the central nervous system has been constrained by a lack of agonists and antagonists with adequate alpha1B-specificity. We have obviated this constraint by generating transgenic mice engineered to overexpress either wild-type or constitutively active alpha1BARs in tissues that normally express the receptor, including the brain. All transgenic lines showed granulovacular neurodegeneration, beginning in alpha1B-expressing domains of the brain and progressing with age to encompass all areas. The degeneration was apoptotic and did not occur in non-transgenic mice. Correspondingly, transgenic mice showed an age-progressive hindlimb disorder that was parkinsonian-like, as demonstrated by rescue of the dysfunction by 3, 4-dihydroxyphenylalanine and considerable dopaminergic-neuronal degeneration in the substantia nigra. Transgenic mice also had a grand mal seizure disorder accompanied by a corresponding dysplasia and neurodegeneration of the cerebral cortex. Both behavioral phenotypes (locomotor impairment and seizure) could be partially rescued with the alpha1AR antagonist terazosin, indicating that alpha1AR signaling participated directly in the pathology. Our results indicate that overstimulation of alpha1BAR leads to apoptotic neurodegeneration with a corresponding multiple system atrophy indicative of Shy-Drager syndrome, a disease whose etiology is unknown.
- Published
- 2000
46. Novel aromatic residues in transmembrane domains IV and V involved in agonist binding at alpha(1a)-adrenergic receptors
- Author
-
Michael J. Zuscik, David Waugh, Dianne M. Perez, and Ming Ming Zhao
- Subjects
Agonist ,Adrenergic receptor ,Epinephrine ,Stereochemistry ,medicine.drug_class ,Protein Conformation ,Inositol Phosphates ,Phenylalanine ,Molecular Sequence Data ,Biochemistry ,chemistry.chemical_compound ,Structure-Activity Relationship ,Cricetinae ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,Phosphatidylinositol ,Amino Acid Sequence ,Receptor ,Molecular Biology ,Ephedrine ,Cell Membrane ,Wild type ,Cell Biology ,Rats ,Transmembrane domain ,chemistry ,Models, Chemical ,COS Cells ,Mutagenesis, Site-Directed ,Adrenergic alpha-Agonists ,Sequence Alignment ,Endogenous agonist - Abstract
We examined the role that aromatic residues located in the transmembrane helices of the alpha(1a)-adrenergic receptor play in promoting antagonist binding. Since alpha(1)-antagonists display low affinity binding at beta(2)-adrenergic receptors, two phenylalanine residues, Phe-163 and Phe-187, of the alpha(1a)-AR were mutated to the corresponding beta(2)-residue. Neither F163Q nor F187A mutations of the alpha(1a) had any effect on the affinity of the alpha(1)-antagonists. However, the affinity of the endogenous agonist epinephrine was reduced 12.5- and 8-fold by the F163Q and F187A mutations, respectively. An additive loss in affinity (150-fold) for epinephrine was observed at an alpha(1a) containing both mutations. The loss of agonist affinity scenario could be reversed by a gain of affinity with mutation of the corresponding residues in the beta(2) to the phenylalanine residues in the alpha(1a). We propose that both Phe-163 and Phe-187 are involved in independent aromatic interactions with the catechol ring of agonists. The potency but not the efficacy of epinephrine in stimulating phosphatidylinositol hydrolysis was reduced 35-fold at the F163Q/F187A alpha(1a) relative to the wild type receptor. Therefore, Phe-163 and Phe-187 represent novel binding contacts in the agonist binding pocket of the alpha(1a)-AR, but are not involved directly in receptor activation.
- Published
- 2000
47. The agonism and synergistic potentiation of weak partial agonists by triethylamine in alpha 1-adrenergic receptor activation: evidence for a salt bridge as the initiating process
- Author
-
James E. Porter, David Waugh, Dianne M. Perez, Stephanie E. Edelmann, and Michael T. Piascik
- Subjects
Agonist ,medicine.drug_class ,Stereochemistry ,Inositol Phosphates ,Population ,Sodium Chloride ,Partial agonist ,Protein Structure, Secondary ,Cell Line ,chemistry.chemical_compound ,Structure-Activity Relationship ,Cricetinae ,Receptors, Adrenergic, alpha-1 ,medicine ,Ethylamines ,Animals ,Humans ,Inositol ,education ,Triethylamine ,Pharmacology ,education.field_of_study ,Binding Sites ,Drug Synergism ,Rats ,Transmembrane domain ,chemistry ,Solubility ,Second messenger system ,COS Cells ,Mutagenesis, Site-Directed ,Molecular Medicine ,Salt bridge ,Adrenergic alpha-1 Receptor Agonists - Abstract
Alpha 1-adrenergic receptor (AR) activation is thought to be initiated by disruption of a constraining interhelical salt bridge (). Disruption of this salt bridge is achieved through a competition for the aspartic acid residue in transmembrane domain three by the protonated amine of the endogenous ligand norepinephrine and a lysine residue in transmembrane domain seven. To further test this hypothesis, we investigated the possibility that a simple amine could mimic an important functional group of the endogenous ligand and break this alpha 1-AR ionic constraint leading to agonism. Triethylamine (TEA) was able to generate concentration-dependent increases of soluble inositol phosphates in COS-1 cells transiently transfected with the hamster alpha 1b-AR and in Rat-1 fibroblasts stably transfected with the human alpha 1a-AR subtype. TEA was also able to synergistically potentiate the second messenger production by weak partial alpha 1-AR agonists and this effect was fully inhibited by the alpha 1-AR antagonist prazosin. However, this synergistic potentiation was not observed for full alpha 1-AR agonists. Instead, TEA caused a parallel rightward shift of the dose-response curve, consistent with the properties of competitive antagonism. TEA specifically bound to a single population of alpha 1-ARs with a Ki of 28.7 +/- 4.7 mM. In addition, the site of binding by TEA to the alpha 1-AR is at the conserved aspartic acid residue in transmembrane domain three, which is part of the constraining salt bridge. These results indicate a direct interaction of TEA in the receptor agonist binding pocket that leads to a disruption of the constraining salt bridge, thereby initiating alpha 1-AR activation.
- Published
- 1998
48. Molecular Mechanisms of Ligand Binding and Activation in α1-Adrenergic Receptors
- Author
-
James E. Porter, Ming Ming Zhao, John Hwa, and Dianne M. Perez
- Subjects
Agonist ,biology ,Adrenergic receptor ,Chemistry ,Stereochemistry ,medicine.drug_class ,Mutant ,Ligand (biochemistry) ,Rhodopsin ,Docking (molecular) ,biology.protein ,medicine ,Biophysics ,Transducin ,Receptor - Abstract
Publisher Summary To rationally design selective drugs, an understanding of subtype differences in the ligand binding pockets would be invaluable. It is hypothesized that only a few nonconserved residues are critical for the ligand-binding specificity of each α 1 AR subtype. Nothing is known about the agonist-dependent molecular mechanisms of α 1 AR stimulation. The activation mechanism for a related G-protein-coupled receptor, rhodopsin, has been studied. The ligand isomerization of retinal when exposed to light breaks a constraining salt-bridge between transmembranes (TMs) 3 and 7, allowing the rhodopsin receptor to adopt an active conformation that can now signal through transducin. A “constraining factor” has also been postulated for the α 1b AR subtype, holding the receptor in a basal configuration until bound by a receptor agonist. However, no molecular evidence for this α 1 BR constraining factor has been presented. In this chapter, the activational mechanism is also explored by testing the hypothesis that α 1 ARs conserved the activational mechanism of rhodopsin in which a saltbridge between TM 3 and TM 7 is disrupted. If the salt-bridge hypothesis is correct, eliminating the negative charge at position 125 should also generate constitutively active α 1b ARs. To investigate this possibility, the D 125 of the WT α 1b AR was changed to an A or a K. The binding properties for these D 125 mutations showed no significant changes in affinity for antagonists when compared with the WT (wild-type) receptor. However, there was a significantly lowered epinephrine affinity (threefold) for these D 125 mutant receptors when compared with the WT α 1b R. This decrease in the epinephrine-binding affinity is likely because of the elimination of the conserved negative charged at position 125, shown in the β AR system to be responsible for docking with the protonated amine of epinephrine.
- Published
- 1997
- Full Text
- View/download PDF
49. The unique nature of the serine interactions for alpha 1-adrenergic receptor agonist binding and activation
- Author
-
John Hwa and Dianne M. Perez
- Subjects
Agonist ,Models, Molecular ,Epinephrine ,medicine.drug_class ,Stereochemistry ,Mutant ,Molecular Sequence Data ,medicine.disease_cause ,Ligands ,Biochemistry ,Binding, Competitive ,Serine ,Phenylephrine ,Receptors, Adrenergic, alpha-2 ,Cricetinae ,Receptors, Adrenergic, alpha-1 ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Inositol phosphate ,Receptor ,Molecular Biology ,chemistry.chemical_classification ,Mutation ,Binding Sites ,Sequence Homology, Amino Acid ,Synephrine ,Wild type ,Hydrogen Bonding ,Cell Biology ,Rats ,Transmembrane domain ,chemistry ,Mutagenesis, Site-Directed ,Receptors, Adrenergic, beta-2 ,Adrenergic alpha-Agonists - Abstract
Activation of the beta2- and alpha2-adrenergic receptors (AR) involves hydrogen bonding of serine residues in the fifth transmembrane segment (TMV) to the catechol hydroxyls of the endogenous agonists, epinephrine and norepinephrine. With the beta2-AR both Ser204 and Ser207 but not a third TMV serine (Ser203) are required for binding and full agonist activity. However, with the alpha2a-AR only one of two TMV serines (Ser204, equivalent to Ser207 in the beta-AR) appears to contribute partially to agonist-binding and activation. Because the alpha1a-AR uniquely contains only two TMV serines, this subtype was used to systematically evaluate the role of hydrogen bonding in alpha1-AR activation. Binding of epinephrine or its monohydroxyl congeners, phenylephrine and synephrine, was not decreased when tested with alanine- substitution mutants that lacked either Ser188 (Ser188--Ala) or Ser192 (Ser192--Ala). With the substitution of both serines in the double mutant, Ser188/192--Ala, binding of all three ligands was significantly reduced (10- 100-fold) consistent with a single hydrogen bond interaction. However, receptor-mediated inositol phosphate production was markedly attenuated only with the Ser188--Ala mutation and not with Ser192--Ala. In support of the importance of Ser188, binding of phenylephrine (meta-hydroxyl only) by Ser192--Ala increased 7-fold over that observed with either the wild type receptor or the Ser188--Ala mutation. Binding of synephrine (para-hydroxyl only) was unchanged with the Ser192--Ala mutation. In addition, when combined with a recently described constitutively active alpha1a-AR mutation (Met292--Leu), only the Ser188--Ala mutation and not Ser192--Ala relieved the high affinity binding and increased agonist potency observed with the Met292--Leu mutation. A simple interpretation of these findings is that the meta-hydroxyl of the endogenous agonists preferentially binds to Ser188, and it is this hydrogen bond interaction, and not that between the para-hydroxyl and Ser192, that allows receptor activation. Furthermore, since Ser188 and Ser192 are separated by three residues on the TMV alpha-helix, whereas Ser204 and Ser207 of the beta2-AR are separated by only two residues, the orientation of the catechol ring in the alpha1-AR binding pocket appears to be unique and rotated approximately 120 degrees to that in the beta2-AR.
- Published
- 1996
50. Identification of critical determinants of alpha 1-adrenergic receptor subtype selective agonist binding
- Author
-
John Hwa, Dianne M. Perez, and Robert M. Graham
- Subjects
Agonist ,Models, Molecular ,medicine.drug_class ,Stereochemistry ,Mutant ,Molecular Sequence Data ,Kidney ,Ligands ,Transfection ,Biochemistry ,Protein Structure, Secondary ,Cell Line ,Cricetinae ,Receptors, Adrenergic, alpha-1 ,Chlorocebus aethiops ,medicine ,Animals ,Point Mutation ,Amino Acid Sequence ,Receptor ,Molecular Biology ,chemistry.chemical_classification ,Binding Sites ,Ligand ,Chemistry ,Cell Biology ,Transmembrane protein ,Recombinant Proteins ,Amino acid ,Transmembrane domain ,Mutagenesis, Site-Directed ,Adrenergic alpha-Agonists ,Macromolecule - Abstract
alpha 1-Adrenergic receptor (AR) subtypes mediate many effects of the sympathetic nervous system. The three cloned subtypes (alpha 1a-AR, alpha 1b-AR, alpha 1d-AR), although structurally similar, bind a series of ligands with different relative potencies. This is particularly true for the alpha 1a-AR, which recognizes a number of agonists and antagonists with 5-50-fold higher affinity than the alpha 1b- or alpha 1d- subtypes. Since ligands bind to receptor-residues that are located in the transmembrane spanning domains, we hypothesize that subtype differences in ligand recognition are due to differences in the binding properties of nonconserved transmembrane residues. Using site-directed mutagenesis, selected putative ligand-binding residues in the alpha 1b-AR were converted, either individually or in combination, to the corresponding residues in the alpha 1a-AR. Mutation of two such residues (of approximately 172 amino acids in the transmembrane domains) converted the agonist binding profile entirely to that of the alpha 1a-AR. Over 80% of this conversion was due to an Ala204--Val substitution; the remainder was due to the additional substitution of Leu314--Met. To confirm that Ala204 and Leu314 are indeed critical for agonist subtype-selectivity, the equivalent residues in the alpha 1a-AR (Val185 and Met293) were reversed of that of the alpha 1b-AR. Correspondingly, the agonist-binding profile of this double alpha 1a-AR mutant reverted to that of the alpha 1b-AR. From these data, in conjunction with macromolecular modeling of the ligand-binding pocket, a model has been developed, which indicates that the determinants of these two residues for agonist subtype-selectivity are due not only to interactions between their side chains and specific ligand moieties but also to a critical interaction between these two amino acids.
- Published
- 1995
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.