Top ▲

α1D-adrenoceptor

Click here for help

Target id: 24

Nomenclature: α1D-adrenoceptor

Family: Adrenoceptors

Gene and Protein Information Click here for help
class A G protein-coupled receptor
Species TM AA Chromosomal Location Gene Symbol Gene Name Reference
Human 7 572 20p13 ADRA1D adrenoceptor alpha 1D 8
Mouse 7 562 2 63.5 cM Adra1d adrenergic receptor, alpha 1d 3
Rat 7 561 3q36 Adra1d adrenoceptor alpha 1D 63
Previous and Unofficial Names Click here for help
ADRA1 | Adra-1 | ADRA1A | ADRA1R | Adrd1 | adrenergic receptor delta1 | α1A-adrenoceptor | α1A/D | α1a/d-adrenoceptor | alpha 1D-adrenoceptor | alpha 1D-adrenoreceptor | alpha1D-AR | Gpcr8
Database Links Click here for help
Specialist databases
GPCRdb ada1d_human (Hs), ada1d_mouse (Mm), ada1d_rat (Rn)
Other databases
Alphafold
ChEMBL Target
DrugBank Target
Ensembl Gene
Entrez Gene
Human Protein Atlas
KEGG Gene
OMIM
Pharos
RefSeq Nucleotide
RefSeq Protein
UniProtKB
Wikipedia
Natural/Endogenous Ligands Click here for help
(-)-adrenaline
(-)-noradrenaline

Download all structure-activity data for this target as a CSV file go icon to follow link

Agonists
Key to terms and symbols View all chemical structures Click column headers to sort
Ligand Sp. Action Value Parameter Reference
cirazoline Small molecule or natural product Rn Full agonist 6.9 pKi 73
pKi 6.9 [73]
clonidine Small molecule or natural product Approved drug Ligand has a PDB structure Rn Full agonist 6.9 pKi 73
pKi 6.9 [73]
St 587 Small molecule or natural product Rn Agonist 6.5 pKi 73
pKi 6.5 [73]
(-)-noradrenaline Small molecule or natural product Approved drug Click here for species-specific activity table Ligand is endogenous in the given species Ligand has a PDB structure Hs Full agonist 5.5 – 7.4 pKi 45,86,98
pKi 5.5 – 7.4 [45,86,98]
(-)-adrenaline Small molecule or natural product Approved drug Ligand is endogenous in the given species Ligand has a PDB structure Immunopharmacology Ligand Rn Full agonist 6.3 pKi 73
pKi 6.3 [73]
(-)-noradrenaline Small molecule or natural product Approved drug Click here for species-specific activity table Ligand is endogenous in the given species Ligand has a PDB structure Rn Full agonist 6.3 pKi 73
pKi 6.3 [73]
(-)-adrenaline Small molecule or natural product Approved drug Primary target of this compound Click here for species-specific activity table Ligand is endogenous in the given species Ligand has a PDB structure Immunopharmacology Ligand Hs Full agonist 5.3 – 7.2 pKi 45,86,98
pKi 5.3 – 7.2 [45,86,98]
oxymetazoline Small molecule or natural product Approved drug Ligand has a PDB structure Rn Partial agonist 6.2 pKi 73
pKi 6.2 [73]
SKF 89748 Small molecule or natural product Rn Full agonist 6.1 pKi 73
pKi 6.1 [73]
6-fluoro-noradrenaline Small molecule or natural product Rn Full agonist 6.0 pKi 73
pKi 6.0 [73]
xylometazoline Small molecule or natural product Approved drug Rn Agonist 6.0 pKi 73
pKi 6.0 [73]
(+)-adrenaline Small molecule or natural product Click here for species-specific activity table Ligand has a PDB structure Hs Full agonist 6.0 pKi 98
pKi 6.0 [98]
phenylephrine Small molecule or natural product Approved drug Rn Full agonist 5.9 pKi 73
pKi 5.9 [73]
oxymetazoline Small molecule or natural product Approved drug Click here for species-specific activity table Ligand has a PDB structure Hs Partial agonist 5.3 – 6.4 pKi 81,86,98
pKi 5.3 – 6.4 [81,86,98]
corbadrine Small molecule or natural product Approved drug Rn Full agonist 5.6 pKi 73
pKi 5.6 [73]
clonidine Small molecule or natural product Approved drug Click here for species-specific activity table Ligand has a PDB structure Hs Agonist 5.6 pKi 86
pKi 5.6 [86]
indanidine Small molecule or natural product Rn Full agonist 5.5 pKi 73
pKi 5.5 [73]
cirazoline Small molecule or natural product Click here for species-specific activity table Hs Full agonist 5.5 pKi 86
pKi 5.5 [86]
NS-49 Small molecule or natural product Click here for species-specific activity table Hs Partial agonist 5.4 pKi 81
pKi 5.4 [81]
phenylephrine Small molecule or natural product Approved drug Click here for species-specific activity table Hs Full agonist 4.7 pKi 86
pKi 4.7 [86]
methoxamine Small molecule or natural product Approved drug Rn Full agonist 4.5 pKi 73
pKi 4.5 [73]
methoxamine Small molecule or natural product Approved drug Click here for species-specific activity table Hs Full agonist 3.8 – 4.9 pKi 86,98
pKi 3.8 – 4.9 [86,98]
amidephrine Small molecule or natural product Rn Full agonist 4.2 pKi 73
pKi 4.2 [73]
(-)-adrenaline Small molecule or natural product Approved drug Click here for species-specific activity table Ligand has a PDB structure Immunopharmacology Ligand Hs Full agonist 6.7 – 7.7 pEC50 86
pEC50 6.7 – 7.7 [86]
(-)-noradrenaline Small molecule or natural product Approved drug Click here for species-specific activity table Ligand has a PDB structure Hs Full agonist 6.6 – 7.8 pEC50 86
pEC50 6.6 – 7.8 [86]
phenylephrine Small molecule or natural product Approved drug Click here for species-specific activity table Hs Full agonist 6.2 – 7.2 pEC50 86
pEC50 6.2 – 7.2 [86]
oxymetazoline Small molecule or natural product Approved drug Click here for species-specific activity table Ligand has a PDB structure Hs Partial agonist 5.6 – 7.3 pEC50 86
pEC50 5.6 – 7.3 [86]
cirazoline Small molecule or natural product Click here for species-specific activity table Hs Full agonist 5.4 – 6.9 pEC50 86
pEC50 5.4 – 6.9 [86]
methoxamine Small molecule or natural product Approved drug Click here for species-specific activity table Hs Full agonist 5.1 – 5.4 pEC50 86
pEC50 5.1 – 5.4 [86]
View species-specific agonist tables
Agonist Comments
Non catecholamine agonists, such as methoxamine and amidephrine, have both low affinity and low intrinsic activity at the α1D- adrenoceptor [73]. There are no selective agonists currently available for α1D-AR.
As the endogenous ligand, (-)-adrenaline has intrinsic activity across the adrenoceptor family, but α1D and α2A subtypes have been identified as primary drug targets as the agonist has highest affinity at these isoforms. A range of pEC50 values are given to reflect findings in different studies and across different signalling assays. Potency of agonists is generally higher for intracellular Ca2+ release than for ERK1/2 phosphorylation. In contrast to α1A- and α1B-AR, no agonists act at α1D-AR to cause cAMP generation [86]. In some cases higher potency for ERK1/2 phosphorylation can reflect off target effects of agonists such as oxymetazoline that also activates 5-HT1B receptors endogenously expressed in CHO cells [19].
Antagonists
Key to terms and symbols View all chemical structures Click column headers to sort
Ligand Sp. Action Value Parameter Reference
olanzapine Small molecule or natural product Approved drug Click here for species-specific activity table Rn Antagonist 6.4 pA2 80
pA2 6.4 [80]
Description: Measured as antagonism of phenylephrine-induced contraction of endothelium-denuded rat aorta.
[125I]HEAT (BE2254) Small molecule or natural product Click here for species-specific activity table Ligand is labelled Ligand is radioactive Hs Antagonist 9.5 – 9.9 pKd 95,98
pKd 9.5 – 9.9 [95,98]
HEAT (BE2254) Small molecule or natural product Click here for species-specific activity table Ligand is labelled Ligand is radioactive Hs Antagonist 9.5 pKd 98
pKd 9.5 [98]
RX18 Small molecule or natural product Hs Antagonist 10.3 pKi 90
pKi 10.3 [90]
A-123189 Small molecule or natural product Click here for species-specific activity table Rn Antagonist 9.8 pKi 10
pKi 9.8 [10]
tamsulosin Small molecule or natural product Approved drug Primary target of this compound Click here for species-specific activity table Ligand has a PDB structure Hs Antagonist 9.2 – 10.2 pKi 27,87,98,112
pKi 9.2 – 10.2 [27,87,98,112]
A-119637 Small molecule or natural product Click here for species-specific activity table Rn Antagonist 9.7 pKi 10
pKi 9.7 [10]
prazosin Small molecule or natural product Approved drug Primary target of this compound Click here for species-specific activity table Ligand has a PDB structure Hs Inverse agonist 9.1 – 10.2 pKi 27,87,98,112
pKi 9.1 – 10.2 [27,87,98,112]
A-119637 Small molecule or natural product Click here for species-specific activity table Hs Antagonist 9.6 pKi 10
pKi 9.6 [10]
A-123189 Small molecule or natural product Click here for species-specific activity table Hs Antagonist 9.5 pKi 10
pKi 9.5 [10]
(S)-41 Small molecule or natural product Hs Antagonist 9.5 pKi 93
pKi 9.5 [93]
NAN 190 Small molecule or natural product Click here for species-specific activity table Hs Antagonist 9.2 pKi 113
pKi 9.2 [113]
(+)-cyclazosin Small molecule or natural product Click here for species-specific activity table Ligand has a PDB structure Hs Inverse agonist 8.5 – 9.9 pKi 32,87
pKi 8.5 – 9.9 [32,87]
WB 4101 Small molecule or natural product Click here for species-specific activity table Hs Antagonist 8.6 – 9.6 pKi 27,87,98
pKi 8.6 – 9.6 [27,87,98]
BMY-7378 Small molecule or natural product Click here for species-specific activity table Rn Antagonist 9.0 pKi 10
pKi 9.0 [10]
BMY-7378 Small molecule or natural product Click here for species-specific activity table Hs Antagonist 8.6 – 9.1 pKi 10,87,113
pKi 8.6 – 9.1 [10,87,113]
doxazosin Small molecule or natural product Approved drug Primary target of this compound Click here for species-specific activity table Hs Antagonist 8.3 – 9.1 pKi 39,87
pKi 8.3 – 9.1 (Ki 8.13x10-10 M) [39,87]
[3H]prazosin Small molecule or natural product Click here for species-specific activity table Ligand is labelled Ligand is radioactive Ligand has a PDB structure Hs Antagonist 8.7 pKi 87
pKi 8.7 [87]
dapiprazole Small molecule or natural product Approved drug Primary target of this compound Hs Antagonist 8.4 pKi 7,87
pKi 8.4 (Ki 4.09x10-9 M) [7,87]
phenoxybenzamine Small molecule or natural product Approved drug Click here for species-specific activity table Hs Antagonist 8.4 pKi 87
pKi 8.4 non-competitive antagonist [87]
terazosin Small molecule or natural product Approved drug Primary target of this compound Click here for species-specific activity table Hs Antagonist 7.7 – 9.1 pKi 69,87
pKi 7.7 – 9.1 (Ki 8.5x10-10 M) [69,87]
spiperone Small molecule or natural product Click here for species-specific activity table Ligand has a PDB structure Hs Antagonist 8.1 pKi 113
pKi 8.1 [113]
alfuzosin Small molecule or natural product Approved drug Primary target of this compound Click here for species-specific activity table Hs Antagonist 7.7 – 8.4 pKi 41,87
pKi 7.7 – 8.4 [41,87]
spiroxatrine Small molecule or natural product Click here for species-specific activity table Hs Antagonist 7.9 pKi 87,113
pKi 7.9 [87,113]
ketanserin Small molecule or natural product Approved drug Click here for species-specific activity table Ligand has a PDB structure Hs Antagonist 7.8 pKi 113
pKi 7.8 [113]
ritanserin Small molecule or natural product Click here for species-specific activity table Ligand has a PDB structure Hs Antagonist 7.8 pKi 113
pKi 7.8 [113]
silodosin Small molecule or natural product Approved drug Click here for species-specific activity table Hs Antagonist 6.9 – 8.7 pKi 87,98
pKi 6.9 – 8.7 [87,98]
upidosin Small molecule or natural product Click here for species-specific activity table Hs Antagonist 7.8 pKi 27
pKi 7.8 [27]
RS-100329 Small molecule or natural product Click here for species-specific activity table Hs Antagonist 7.6 – 7.9 pKi 87,112
pKi 7.6 – 7.9 [87,112]
clozapine Small molecule or natural product Approved drug Click here for species-specific activity table Hs Antagonist 7.7 pKi 113
pKi 7.7 [113]
KMUP-1 Small molecule or natural product Click here for species-specific activity table Rn Antagonist 7.7 pKi 61
pKi 7.7 [61]
(+)-cyclazosin Small molecule or natural product Ligand has a PDB structure Rn Inverse agonist 7.6 pKi 32
pKi 7.6 [32]
mianserin Small molecule or natural product Approved drug Click here for species-specific activity table Hs Antagonist 7.5 pKi 113
pKi 7.5 [113]
phentolamine Small molecule or natural product Approved drug Primary target of this compound Click here for species-specific activity table Hs Inverse agonist 6.8 – 8.2 pKi 87,98
pKi 6.8 – 8.2 [87,98]
risperidone Small molecule or natural product Approved drug Click here for species-specific activity table Ligand has a PDB structure Hs Antagonist 7.4 pKi 113
pKi 7.4 [113]
dibenamine Small molecule or natural product Hs Antagonist 7.4 pKi 87
pKi 7.4 non-competitive antagonist [87]
RS-17053 Small molecule or natural product Click here for species-specific activity table Hs Antagonist 6.8 – 7.8 pKi 27,87
pKi 6.8 – 7.8 [27,87]
Ro-70-0004 Small molecule or natural product Click here for species-specific activity table Hs Antagonist 7.2 pKi 112
pKi 7.2 [112]
SKF 105854 Small molecule or natural product Hs Antagonist 7.1 pKi 42
pKi 7.1 [42]
cyproheptadine Small molecule or natural product Approved drug Click here for species-specific activity table Ligand has a PDB structure Hs Antagonist 6.9 pKi 113
pKi 6.9 [113]
5-methylurapidil Small molecule or natural product Click here for species-specific activity table Hs Antagonist 5.6 – 8.0 pKi 27,87,98,113
pKi 5.6 – 8.0 [27,87,98,113]
S(+)-niguldipine Small molecule or natural product Click here for species-specific activity table Hs Antagonist 5.9 – 7.4 pKi 27,87,98
pKi 5.9 – 7.4 [27,87,98]
indoramin Small molecule or natural product Approved drug Click here for species-specific activity table Hs Antagonist 6.3 – 6.7 pKi 27,87
pKi 6.3 – 6.7 [27,87]
labetalol Small molecule or natural product Approved drug Primary target of this compound Click here for species-specific activity table Hs Antagonist 6.1 – 6.6 pKi 7,87
pKi 6.1 – 6.6 [7,87]
SNAP5089 Small molecule or natural product Click here for species-specific activity table Hs Antagonist 5.7 pKi 87
pKi 5.7 [87]
RX18 Small molecule or natural product Rn Antagonist 9.1 pEC50 90
pEC50 9.1 [90]
Description: Antagonist potency determined in isolated rat thoracic aorta
View species-specific antagonist tables
Antagonist Comments
Although cyclazosin does not show selectivity in radioligand binding assays with recombinant α1-ARs, functional selectivity for the α1D-AR is observed in functional assays using isolated tissue measuring affinity for native α1- subtypes [72]. SNAP5089 is >1000-fold and RS-100329 and Ro-70-0004 are both 50-fold selective for α1A-ARs over the α1B- and α1D-AR subtypes [87,112]. BMY-7378 is the only α1D-AR selective antagonist currently available [87] and has some weak partial agonist actions at the α1A, α1B and 5-HT1A receptor [34,86]. Differentiation between neutral antagonists and inverse agonists at the α1D-AR has not been studied extensively. Prazosin, cyclazosin, doxazosin and terazosin are approved drugs that have similar affinity for all α1-AR subtypes. Phenoxybenzamine is an irreversible α1A-AR antagonist used to block the pressor effects of catecholamines prior to surgery for phaeochromocytoma. Labetalol is predominantly a β-AR antagonist but also has some α-AR blocking properties and is considered safe for use in pregnancy to treat eclampsia and pre-eclampsia. It also behaves as a partial agonist in some systems.
Allosteric Modulator Comments
Lorazepam and midazolam have been shown to increase the maximum response to phenylephrine in cells expressing the human α1D-AR [111].
Primary Transduction Mechanisms Click here for help
Transducer Effector/Response
Gq/G11 family Phospholipase C stimulation
Calcium channel
Other - See Comments
Comments:  The α1D-adrenoceptor is coupled to calcium release and inositol phosphate production less efficiently than either the α1A- or α1B-adrenoceptor.
References:  35,71
Secondary Transduction Mechanisms Click here for help
Transducer Effector/Response
Phospholipase D stimulation
Other - See Comments
Comments:  α1-ARs (all subtypes) can also activate protein Kinase C and mitogen activated protein kinases. Some agonists acting at the α1D-AR can weakly activate adenylate cyclase in the presence of forskolin. α1D-AR couples to adenylate cyclase much less efficiently than either the α1A- or α1B-AR and agonists generally have no effect.
References:  35,71,87
Tissue Distribution Click here for help
Epicardial coronary arteries, prefrontal cortex, hippocampus, bladder.
Species:  Human
Technique:  RT-PCR.
References:  52,99-101
α1D- adrenoceptor message and protein is predominant in human bladder.
Species:  Human
Technique:  RNase protection assay, RT-PCR.
References:  67
The α1D-adrenoceptor was the predominant α1 subtype in the human aorta, but either had the lowest expression of the three subtypes, or was not detectable, in other arteries and veins. However, another report showed high expression of α1D-adrenoceptor in blood vessels of human prostate.
Species:  Human
Technique:  RNAse Protection, immunohistochemistry.
References:  91,109
Lymphocytes.
Species:  Human
Technique:  In situ hybridisation.
References:  105
Prefrontal cortex, reticular thalmic nucleus, hippocampus, cingulate cortex, spinal cord.
Species:  Mouse
Technique:  In situ hybridisation.
References:  40,94
Leydig cells.
Species:  Mouse
Technique:  RT-PCR.
References:  48
Concomitant activation of all α1 subtypes in the nucleus accumbens is required for α1adrenergic inhibition of accumbal dopaminergic activity.
Species:  Rat
Technique:  In vivo microdialysis.
References:  4
Activation of the bladder mechanosensory fibres.
Species:  Rat
Technique:  Single-unit afferent nerve fibre activity (SAA) of primary bladder afferent nerves and their relationship with bladder microcontractions in rats.
References:  2
In the rat brain, highest levels of α1D-adrenoceptor message are found in the olfactory bulb, cerebral cortex, hippocampus, dentate gyrus, reticular thalamic nucleus, motor neurons and the inferior olivary complex. In the thalamus, the α1B and α1D-adrenoceptors have a complimentary distribution.
Species:  Rat
Technique:  In situ hybridisation, RT-PCR.
References:  21,78,97
Expression Datasets Click here for help

Show »

Log average relative transcript abundance in mouse tissues measured by qPCR from Regard, J.B., Sato, I.T., and Coughlin, S.R. (2008). Anatomical profiling of G protein-coupled receptor expression. Cell, 135(3): 561-71. [PMID:18984166] [Raw data: website]

There should be a chart of expression data here, you may need to enable JavaScript!
Functional Assays Click here for help
Contraction of isolated rat aortic ring and skeletal muscle arteriole.
Species:  Rat
Tissue:  Vasculature.
Response measured:  Contraction.
References:  43,60
Tamsulosin treatment upregulates both α1A- and α1D-AR mRNA.
Species:  Rat
Tissue:  Prostate.
Response measured:  Receptor mRNA expression.
References:  56
Regulation of hippocampal α1D-AR mRNA by corticosterone in adrenalectomized rats. Corticosteroids prevent the adrenalectomized decrease in hippocampal α1D-AR.
Species:  Rat
Tissue:  Brain.
Response measured:  Receptor expression.
References:  21
The α1D-AR is intracellular but still mediates increases in intracellular calcium and reactive oxygen species.
Species:  Human
Tissue:  Aortic smooth muscle, transfected HEK 293 cells.
Response measured:  Measurement of intracellular calcium and reactive oxygen species.
References:  29,54,110
Amino acids Asp176 in the third transmembrane domain (TMD), Glu237 in TMD IV, and Ser258 in TMD V of α1D-AR are involved in binding prazosin and tamsulosin.
Species:  Human
Tissue:  Transfected HEK 293 cells.
Response measured:  Receptor binding.
References:  77
Alteration of α1D-AR density, signal transduction and blood pressure by syntrophins (α-syntrophin increases α1D-AR density; β2-syntrophin increases signaling efficacy of inositol phosphates and ERK).
Species:  Human
Tissue:  Transfected HEK 293 cells.
Response measured:  Receptor density and signalling.
References:  14,64-65
α1D-AR releases ATP, which induces P2X7 receptors to increase [Ca2+](i) but not to stimulate protein secretion. P2X7 receptors in turn activate α1D-AR to increase [Ca2+](i) but not to stimulate protein secretion.
Species:  Rat
Tissue:  Lacrimal gland.
Response measured:  [Ca2+](i).
References:  20
Function and phosphorylation state of α1D-AR is modulated by activation of receptor tyrosine kinases, PKC, insulin, IGF-I and EGF.
Species:  Rat
Tissue:  Transfected Rat-1 fibroblasts.
Response measured:  Phosphorylation and desensitization.
References:  30-31,89
The α1D-AR induces vascular smooth muscle apoptosis via a p53-dependent mechanism.
Species:  Human
Tissue:  Aortic smooth muscle cells.
Response measured:  Apoptosis.
References:  28
α1D-AR promotes trophic effects(pseudocapillary formation, proliferation and migration) in fragments of human mature vessels and is potentiated with hypoxia.
Species:  Human
Tissue:  Endothelial cells.
Response measured:  Cell trophism.
References:  108
α1D-AR stimulates protein secretion and ectodomain shedding of EGF to transactivate the EGF receptor, potentially via ADAM17, which activates p42/p44 MAPK to negatively modulate protein secretion.
Species:  Rat
Tissue:  Lacrimal gland epithelial cells.
Response measured:  Measurement of EGF release and receptor activation.
References:  12,44
Addition of a signal peptide sequence (16 amino acids) to, or N-terminal truncation of the α1D-AR gene increases expression of binding sites but not protein.
Species:  Human
Tissue:  Transfected neuro2A and COS-1 cells
Response measured:  [3H]-prazosin binding to α1D-AR.
References:  83,85
Carvedilol selectively inhibits oscillatory intracellular calcium changes evoked by α1D- and α1B-AR.
Species:  Human
Tissue:  Transfected HEK 293 cells.
Response measured:  Measurement of intracellular calcium.
References:  58
In a renal artery stenosis model, GRK2 gene knockout or GRKct peptide treated mice enhance α1D-AR vasoconstriction.
Species:  Mouse
Tissue:  Kidney.
Response measured:  Vasoconstriction.
References:  16
Cell surface expression of α1D-AR is controlled by heterodimerization with α1B-or β2-ARs; Angiotensin I receptor hetrodimerizes with α1D-AR in preeclamptic rats.
Species:  Human
Tissue:  Transfected HEK 293 cells, rat aorta.
Response measured:  Receptor dimerization.
References:  14,33,36-37,106
Methylation-dependent disruption of Sp1 binding in promoter region in a cell-specific manner results in repression of basal α1D-AR expression
Species:  Human
Tissue:  SK-N-MC and DU145 cells.
Response measured:  DNA methylation, receptor expression.
References:  70
Regulation of α1D-AR signal complex signalosome.
Species:  Human
Tissue:  Transfected HEK 293 cells.
Response measured:  Coimmunoprecipitation and blot overlay assays.
References:  66
The selectivity of α-AR agonists for the human α1A, α1B, and α1D-ARs.
Species:  Human
Tissue:  CHO-K1 cell stably expressing α1D-AR.
Response measured:  Whole cell [3H]prazosin binding, intracellular Ca2+ release, ERK1/2 phosphorylation and cAMP accumulation.
References:  86
The affinity and selectivity of α-AR antagonists, antidepressants, and antipsychotics for the human α1A, α1B, and α1D-ARs.
Species:  Human
Tissue:  CHO-K1 cell stably expressing α1D-AR.
Response measured:  Whole cell [3H]prazosin binding.
References:  87
Role of α1D-AR phosphorylation sites in 3rd IC loop and C-terminus in Ca2+ signalling, ERK modulation, plasma membrane localisation and β-arrestin interaction.
Species:  Human
Tissue:  Transfected HEK 293 cells.
Response measured:  Receptor phosphorylation, Ca2+ concentration, ERK1/2 activation, receptor internalisation and interaction with β-arrestin.
References:  9
α1D-ARs are responsible for the greater contractile sensitivity, slower time-course and post-activation contraction to an adrenoceptor-mediated stimulus in conductance vessels.
Species:  Rat
Tissue:  Aorta and tail artery.
Response measured:  Contractile response, inositol phosphate accumulation, ERK1/2 phosphorylation.
References:  26
Scribble binds multiple α1D-AR C-terminal PDZ ligands.
Species:  Human
Tissue:  Transfected HEK 293 cells.
Response measured:  DMR, SNAP GST pulldown, co-IP, biolayer interferometry, x-ray crystallography.
References:  50
N-glycosylation of α1D-AR N-terminal domain is required for correct trafficking, function, and biogenesis.
Species:  Human
Tissue:  Transfected HEK 293 cells.
Response measured:  DMR, sucrose density centrifugation, confocal microscopy, co-localisation analysis.
References:  51
Endogenous N-terminal domain cleavage modulates α1D-AR pharmacodynamics.
Species:  Human
Tissue:  HEK 293, SW480, HEPG2, MCF7, and A375 cells.
Response measured:  DMR, SNAP tagged proteins, immunoblotting, mass spec, PI hydrolysis, radioligand binding, live cell imaging and cell surface localisation.
References:  59
Cross-talk between α1D-AR and TRP-V1 receptors triggers prostate cancer cell proliferation.
Species:  Human
Tissue:  PC3 and DU145 cells.
Response measured:  Western blotting, ECAR, siRNA, qRT-PCR, Ca2+, BrdU, IP3 assays.
References:  76
Cross-talk with β2-ARs enhances ligand affinity properties from endothelial α1D-AR that mediates carotid relaxation.
Species:  Rat
Tissue:  Carotid artery.
Response measured:  Relaxation and contraction.
References:  84
α1D-AR transactivates EGFR.
Species:  Human
Tissue:  CHO cells stably expressing α1D-AR and transiently expressing EGFR.
Response measured:  Calcium increases and activation of CaMKII, PI3K, and Src, but not ERK1/2 and Akt.
References:  107
Physiological Functions Click here for help
Contraction of mesenteric resistance arteries.
Species:  Rat
Tissue:  Vasculature.
References:  71
α1D-adrenoceptors mediate nerve stimulated contraction of corpus cavernosa.
Species:  Rat
Tissue:  Corpus Cavernosa.
References:  75
Coronary artery vasoconstriction.
Species:  Mouse
Tissue:  Vasculature.
References:  11
Femoral artery vasoconstriction.
Species:  Rat
Tissue:  Vasculature.
References:  47
Locomotor activity in response to environmental stimulation.
Species:  Mouse
Tissue:  Brain.
References:  92
Reflex evoked urethral contraction.
Species:  Rat
Tissue:  Urethra.
References:  17
Endothelium dependent vasodilation of mesenteric vascular bed.
Species:  Rat
Tissue:  Vasculature.
References:  25
Vasopressor nerve responses in the pithed rat, previously identified as α2-ARr mediated, may be α1D-AR mediated.
Species:  Rat
Tissue:  Carotid artery.
References:  23-24
Management of distal ureteral stone by α1D-AR antagonist naftopidil.
Species:  Human
Tissue:  Kidney, ureter, bladder.
References:  115
Control of carotid and mesenteric vasoconstriction by α1D-AR as revealed in α1A/B double knockout mice.
Species:  Mouse
Tissue:  Carotid and mesenteric vasculature.
References:  6,15,68
Role of α1D-AR in the renal vascular response to high-fructose feeding.
Species:  Rat
Tissue:  Kidney.
References:  1
Role of α1A- and α1D-ARs in renal vasoconstriction and haemodynamics in diabetes.
Species:  Rat
Tissue:  Kidney.
References:  5
Role of α1D-AR in choice of α1A-AR antagonist for treatment of prostate hyperplasia.
Species:  Human
Tissue:  Prostate.
References:  55
Impairment of α1D-AR-induced relaxation of rat carotid artery during endothelial dysfunction.
Species:  Rat
Tissue:  Carotid artery.
References:  22
α1D-AR in the urothelium facilitate the micturition reflex and storage.
Species:  Mouse
Tissue:  Bladder.
References:  13,49,96
Role of α1D-AR in contraction of femoral resistance arteries.
Species:  Mouse
Tissue:  Femoral artery.
References:  114
Prostate cancer cell proliferation.
Species:  Human
Tissue:  Prostate cancer PC3 cells.
References:  76
Pathogenesis of hypertension.
Species:  Rat
Tissue:  Blood pressure and cardiac function.
References:  88
Proliferation of pulmonary artery smooth muscle.
Species:  Rat
Tissue:  Pulmonary artery smooth muscle cells.
References:  62
Food intake.
Species:  Rat
Tissue:  Median raphe nucleus.
References:  18
α1D-ARs and high sensitivity slow time-course contraction in conductance arteries; rat and α1D-AR knockout mice.
Species:  Rat
Tissue:  Aorta and tail artery.
References:  26
Physiological Consequences of Altering Gene Expression Click here for help
α1D knockout mice had hypotension, a decreased pressor and decreased coronary vasoconstrictor response to phenylephrine and resistance to salt induced hypertension.
Species:  Mouse
Tissue: 
Technique:  Transgenesis.
References:  11,103-104
α1D knockout mice have delayed tail-flick and hindpaw-licking responses to thermal stimuli.
Species:  Mouse
Tissue: 
Technique:  Gene knockout.
References:  40
α1D-AR knockout mice have lower levels of basal systolic and mean arterial BP, and lower levels of circulating catecholamines than wild-type mice; effects of salt-loading.
Species:  Mouse
Tissue:  Vasculature.
Technique:  Gene knockout.
References:  46,103
α1D-AR has a role in auditory sensory function, attention or working memory rather than reference memory, and the sensorimotor gating deficits induced by the NMDA receptor antagonist. Mice show sensory, attention and memory abnormalities of behavior.
Species:  Mouse
Tissue:  Brain, behavior.
Technique:  Gene knockout.
References:  74
α1D-AR knockout mice lose the slow decay of response in aorta following agonist removal.
Species:  Mouse
Tissue:  Aorta.
Technique:  Gene knockout.
References:  26
Xenobiotics Influencing Gene Expression Click here for help
Peroxynitrite generated through septic shock (bacterial infection) can inhibit maximum binding and signal transduction (intracellular calcium) of the α1A- and α1D-AR. This may be due to modification of these receptor subtypes by peroxynitrite and represents a possible mechanism contributing to systemic hypotension in sepsis.
Species:  Human
Tissue:  CHO cells transfected with the human α1A-, α1B- and α1D-ARs.
Technique:  Ligand binding and measurement of intracellular calcium.
References:  102
Peroxynitrite generated through septic shock (bacterial infection) can inhibit noradrenaline-induced contraction in rat endothelium-denuded aorta strips which contain 1A- and 1D-AR subtypes and represents a possible contributory mechanism underlying systemic hypotension in sepsis.
Species:  Rat
Tissue:  Endothelium-denuded aorta strips.
Technique:  Recording of tension changes in organ bath culture.
References:  102
Erythropoietin reverses sepsis-induced vasoplegia to norepinephrine through preservation of α1D-AR mRNA expression in mouse aorta.
Species:  Mouse
Tissue:  Thoracic aorta.
Technique:  Survival time, contractile response, NO level, iNOS, eNOS, GRK2 and α1D-AR expression (mRNA and protein).
References:  53
Phenotypes, Alleles and Disease Models Click here for help Mouse data from MGI

Show »

Allele Composition & genetic background Accession Phenotype Id Phenotype Reference
Adra1dtm1Gzt Adra1dtm1Gzt/Adra1dtm1Gzt
involves: 129/Sv * C57BL/6J
MGI:106673  MP:0001544 abnormal cardiovascular system physiology PMID: 11901185 
Adra1dtm1Jabl Adra1dtm1Jabl/Adra1dtm1Jabl
involves: 129S6/SvEvTac
MGI:106673  MP:0003313 abnormal locomotor activation PMID: 12874602 
Adra1dtm1Gzt Adra1dtm1Gzt/Adra1dtm1Gzt
involves: 129X1/SvJ * C57BL/6
MGI:106673  MP:0004142 abnormal muscle tone PMID: 15196805 
Adra1dtm1Gzt Adra1dtm1Gzt/Adra1dtm1Gzt
involves: 129X1/SvJ * C57BL/6
MGI:106673  MP:0003088 abnormal prepulse inhibition PMID: 15196805 
Adra1dtm1Gzt Adra1dtm1Gzt/Adra1dtm1Gzt
involves: 129X1/SvJ * C57BL/6
MGI:106673  MP:0008428 abnormal spatial working memory PMID: 15196805 
Adra1dtm1Gzt Adra1dtm1Gzt/Adra1dtm1Gzt
involves: 129X1/SvJ * C57BL/6
MGI:106673  MP:0001489 decreased startle reflex PMID: 15196805 
Adra1dtm1Gzt Adra1dtm1Gzt/Adra1dtm1Gzt
involves: 129/Sv * C57BL/6J
MGI:106673  MP:0002843 decreased systemic arterial blood pressure PMID: 11901185 
Adra1dtm1Gzt Adra1dtm1Gzt/Adra1dtm1Gzt
involves: 129/Sv * C57BL/6J
MGI:106673  MP:0006264 decreased systemic arterial systolic blood pressure PMID: 11901185 
Adra1dtm1Jabl Adra1dtm1Jabl/Adra1dtm1Jabl
involves: 129S6/SvEvTac
MGI:106673  MP:0002757 decreased vertical activity PMID: 12874602 
Adra1dtm1Gzt Adra1dtm1Gzt/Adra1dtm1Gzt
involves: 129X1/SvJ * C57BL/6
MGI:106673  MP:0003858 enhanced coordination PMID: 15196805 
Adra1dtm1Gzt Adra1dtm1Gzt/Adra1dtm1Gzt
involves: 129/Sv * C57BL/6J
MGI:106673  MP:0001596 hypotension PMID: 11901185 
Adra1dtm1Jabl Adra1dtm1Jabl/Adra1dtm1Jabl
involves: 129S6/SvEvTac
MGI:106673  MP:0009750 impaired behavioral response to addictive substance PMID: 12874602 
Biologically Significant Variants Click here for help
Type:  Single nucleotide polymorphisms
Species:  Human
Description:  ADRA1D T1848A (P=0.023) and ADRA1D A1905G (P=0.029) SNPs are associated with the improvement of left ventricular fractional shortening by β-blockers in chronic heart failure.
SNP accession: 
References:  79
Type:  Single nucleotide polymorphisms
Species:  Human
Description:  The side effects of domperidone to treat gastroparesis are associated with SNPs in the α1D-AR promoter region.
SNP accession: 
References:  82
General Comments
α1D-AR message and protein is predominant in human bladder [67]. In bladder tissue from normal rats, only 25% of α1-AR mRNA was of the α1D-subtype; however following bladder obstruction, this percentage increased to 75% [38].
When recombinant α1D-ARs are expressed in fibroblast cell lines, most of the expression is intracellular, as opposed to the cell surface expression of the other α1 subtypes [57].

References

Show »

1. Abdulla MH, Sattar MA, Johns EJ, Abdullah NA, Hye Khan MA, Rathore HA. (2012) High-fructose feeding impacts on the adrenergic control of renal haemodynamics in the rat. Br J Nutr, 107 (2): 218-28. [PMID:21733307]

2. Aizawa N, Sugiyama R, Ichihara K, Fujimura T, Fukuhara H, Homma Y, Igawa Y. (2016) Functional roles of bladder α1-adrenoceptors in the activation of single-unit primary bladder afferent activity in rats. BJU Int, 117 (6): 993-1001. [PMID:26332379]

3. Alonso-Llamazares A, Zamanillo D, Casanova E, Ovalle S, Calvo P, Chinchetru MA. (1995) Molecular cloning of alpha 1d-adrenergic receptor and tissue distribution of three alpha 1-adrenergic receptor subtypes in mouse. J Neurochem, 65: 2387-2392. [PMID:7595531]

4. Aono Y, Taguchi H, Saigusa T, Uchida T, Takada K, Takiguchi H, Shirakawa T, Shimizu N, Koshikawa N, Cools AR. (2015) Simultaneous activation of the α1A-, α1B- and α1D-adrenoceptor subtypes in the nucleus accumbens reduces accumbal dopamine efflux in freely moving rats. Behav Pharmacol, 26 (1-2): 73-80. [PMID:25438092]

5. Armenia, Sattar MA, Abdullah NA, Khan MA, Johns EJ. (2008) Alpha1A- and alpha1D-adrenoceptors are the major functional subtypes of renal alpha1-adrenoceptors in streptozotocin-induced diabetic and normal Sprague-Dawley rats. Auton Autacoid Pharmacol, 28 (1): 1-10. [PMID:18257746]

6. Arévalo-León LE, Gallardo-Ortíz IA, Urquiza-Marín H, Villalobos-Molina R. (2003) Evidence for the role of alpha1D- and alpha1A-adrenoceptors in contraction of the rat mesenteric artery. Vascul Pharmacol, 40 (2): 91-6. [PMID:12646397]

7. Auerbach SS, DrugMatrix® and ToxFX® Coordinator National Toxicology Program. National Toxicology Program: Dept of Health and Human Services. Accessed on 02/05/2014. Modified on 02/05/2014. DrugMatrix, https://ntp.niehs.nih.gov/drugmatrix/index.html

8. Bruno JF, Whittaker J, Song JF, Berelowitz M. (1991) Molecular cloning and sequencing of a cDNA encoding a human alpha 1A adrenergic receptor. Biochem Biophys Res Commun, 179 (3): 1485-90. [PMID:1656955]

9. Carmona-Rosas G, Hernández-Espinosa DA, Alcántara-Hernández R, Alfonzo-Méndez MA, García-Sainz JA. (2019) Distinct phosphorylation sites/clusters in the carboxyl terminus regulate α1D-adrenergic receptor subcellular localization and signaling. Cell Signal, 53: 374-389. [PMID:30419287]

10. Carroll WA, Sippy KB, Esbenshade TA, Buckner SA, Hancock AA, Meyer MD. (2001) Two novel and potent 3-[(o-methoxyphenyl)piperazinylethyl]-5-phenylthien. Bioorg Med Chem Lett, 11 (9): 1119-21. [PMID:11354357]

11. Chalothorn D, McCune DF, Edelmann SE, Tobita K, Keller BB, Lasley RD, Perez DM, Tanoue A, Tsujimoto G, Post GR et al.. (2003) Differential cardiovascular regulatory activities of the alpha 1B- and alpha 1D-adrenoceptor subtypes. J Pharmacol Exp Ther, 305 (3): 1045-53. [PMID:12649302]

12. Chen L, Hodges RR, Funaki C, Zoukhri D, Gaivin RJ, Perez DM, Dartt DA. (2006) Effects of alpha1D-adrenergic receptors on shedding of biologically active EGF in freshly isolated lacrimal gland epithelial cells. Am J Physiol, Cell Physiol, 291 (5): C946-56. [PMID:16760267]

13. Chen Q, Takahashi S, Zhong S, Hosoda C, Zheng HY, Ogushi T, Fujimura T, Ohta N, Tanoue A, Tsujimoto G et al.. (2005) Function of the lower urinary tract in mice lacking alpha1d-adrenoceptor. J Urol, 174 (1): 370-4. [PMID:15947692]

14. Chen Z, Hague C, Hall RA, Minneman KP. (2006) Syntrophins regulate alpha1D-adrenergic receptors through a PDZ domain-mediated interaction. J Biol Chem, 281 (18): 12414-20. [PMID:16533813]

15. Chiba S, Tsukada M. (2002) Existence of functional alpha1A- and alpha1D- but no alpha1B-adrenoceptor subtypes in rat common carotid arteries. Jpn J Pharmacol, 88 (2): 146-50. [PMID:11928714]

16. Cohn HI, Harris DM, Pesant S, Pfeiffer M, Zhou RH, Koch WJ, Dorn GW, Eckhart AD. (2008) Inhibition of vascular smooth muscle G protein-coupled receptor kinase 2 enhances alpha1D-adrenergic receptor constriction. Am J Physiol Heart Circ Physiol, 295 (4): H1695-704. [PMID:18723764]

17. Conley RK, Williams TJ, Ford AP, Ramage AG. (2001) The role of alpha(1)-adrenoceptors and 5-HT(1A) receptors in the control of the micturition reflex in male anaesthetized rats. Br J Pharmacol, 133 (1): 61-72. [PMID:11325795]

18. da Silva ES, Flores RA, Cella EC, Levone BR, Taschetto AP, Kochenborger L, Terenzi MG, Faria MS, Paschoalini MA. (2014) Blockade of median raphe nucleus α1-adrenoceptor subtypes increases food intake in rats. Pharmacol Biochem Behav, 124: 350-5. [PMID:24955865]

19. da Silva Junior ED, Sato M, Merlin J, Broxton N, Hutchinson DS, Ventura S, Evans BA, Summers RJ. (2017) Factors influencing biased agonism in recombinant cells expressing the human α1A -adrenoceptor. Br J Pharmacol, 174 (14): 2318-2333. [PMID:28444738]

20. Dartt DA, Hodges RR. (2011) Interaction of alpha1D-adrenergic and P2X(7) receptors in the rat lacrimal gland and the effect on intracellular [Ca2+] and protein secretion. Invest Ophthalmol Vis Sci, 52 (8): 5720-9. [PMID:21685341]

21. Day HE, Kryskow EM, Watson SJ, Akil H, Campeau S. (2008) Regulation of hippocampal alpha1d adrenergic receptor mRNA by corticosterone in adrenalectomized rats. Brain Res, 1218: 132-40. [PMID:18534559]

22. de Andrade CR, Fukada SY, Olivon VC, de Godoy MA, Haddad R, Eberlin MN, Cunha FQ, de Souza HP, Laurindo FR, de Oliveira AM. (2006) Alpha1D-adrenoceptor-induced relaxation on rat carotid artery is impaired during the endothelial dysfunction evoked in the early stages of hyperhomocysteinemia. Eur J Pharmacol, 543 (1-3): 83-91. [PMID:16828078]

23. Docherty JR. (2011) Vasopressor nerve responses in the pithed rat, previously identified as α2-adrenoceptor mediated, may be α1D-adrenoceptor mediated. Eur J Pharmacol, 658 (2-3): 182-6. [PMID:21376031]

24. Docherty JR. (2012) Yohimbine antagonises α1A- and α1D-adrenoceptor mediated components in addition to the α2A-adrenoceptor component to pressor responses in the pithed rat. Eur J Pharmacol, 679 (1-3): 90-4. [PMID:22290390]

25. Filippi S, Parenti A, Donnini S, Granger HJ, Fazzini A, Ledda F. (2001) alpha(1D)-adrenoceptors cause endothelium-dependent vasodilatation in the rat mesenteric vascular bed. J Pharmacol Exp Ther, 296 (3): 869-75. [PMID:11181918]

26. Flacco N, Parés J, Serna E, Segura V, Vicente D, Pérez-Aso M, Noguera MA, Ivorra MD, McGrath JC, D'Ocon P. (2013) α1D-Adrenoceptors are responsible for the high sensitivity and the slow time-course of noradrenaline-mediated contraction in conductance arteries. Pharmacol Res Perspect, 1 (1): e00001. [PMID:25505555]

27. Ford AP, Daniels DV, Chang DJ, Gever JR, Jasper JR, Lesnick JD, Clarke DE. (1997) Pharmacological pleiotropism of the human recombinant alpha1A-adrenoceptor: implications for alpha1-adrenoceptor classification. Br J Pharmacol, 121 (6): 1127-35. [PMID:9249248]

28. García-Cazarín ML, Smith JL, Clair DK, Piascik MT. (2008) The alpha1D-adrenergic receptor induces vascular smooth muscle apoptosis via a p53-dependent mechanism. Mol Pharmacol, 74 (4): 1000-7. [PMID:18628404]

29. García-Cazarín ML, Smith JL, Olszewski KA, McCune DF, Simmerman LA, Hadley RW, Kraner SD, Piascik MT. (2008) The alpha1D-adrenergic receptor is expressed intracellularly and coupled to increases in intracellular calcium and reactive oxygen species in human aortic smooth muscle cells. J Mol Signal, 3: 6. [PMID:18304336]

30. García-Sáinz JA, Rodríguez-Pérez CE, Romero-Avila MT. (2004) Human alpha1D-adrenoceptor phosphorylation and desensitization. Biochem Pharmacol, 67 (10): 1853-8. [PMID:15130762]

31. García-Sáinz JA, Vázquez-Cuevas FG, Romero-Avila MT. (2001) Phosphorylation and desensitization of alpha1d-adrenergic receptors. Biochem J, 353 (Pt 3): 603-10. [PMID:11171057]

32. Giardinà D, Crucianelli M, Romanelli R, Leonardi A, Poggesi E, Melchiorre C. (1996) Synthesis and biological profile of the enantiomers of [4-(4-amino-6,7-dimethoxyquinazolin-2-yl)-cis-octahydroquinoxalin- 1-yl]furan-2-ylmethanone (cyclazosin), a potent competitive alpha 1B- adrenoceptor antagonist. J Med Chem, 39 (23): 4602-7. [PMID:8917649]

33. González-Hernández Mde L, Godínez-Hernández D, Bobadilla-Lugo RA, López-Sánchez P. (2010) Angiotensin-II type 1 receptor (AT1R) and alpha-1D adrenoceptor form a heterodimer during pregnancy-induced hypertension. Auton Autacoid Pharmacol, 30 (3): 167-72. [PMID:20102360]

34. Greuel JM, Glaser T. (1992) The putative 5-HT1A receptor antagonists NAN-190 and BMY 7378 are partial agonists in the rat dorsal raphe nucleus in vitro. Eur J Pharmacol, 211 (2): 211-9. [PMID:1535319]

35. Hague C, Chen Z, Uberti M, Minneman KP. (2003) Alpha(1)-adrenergic receptor subtypes: non-identical triplets with different dancing partners?. Life Sci, 74 (4): 411-8. [PMID:14609720]

36. Hague C, Lee SE, Chen Z, Prinster SC, Hall RA, Minneman KP. (2006) Heterodimers of alpha1B- and alpha1D-adrenergic receptors form a single functional entity. Mol Pharmacol, 69 (1): 45-55. [PMID:16195468]

37. Hague C, Uberti MA, Chen Z, Hall RA, Minneman KP. (2004) Cell surface expression of alpha1D-adrenergic receptors is controlled by heterodimerization with alpha1B-adrenergic receptors. J Biol Chem, 279 (15): 15541-9. [PMID:14736874]

38. Hampel C, Dolber PC, Smith MP, Savic SL, Th roff JW, Thor KB, Schwinn DA. (2002) Modulation of bladder alpha1-adrenergic receptor subtype expression by bladder outlet obstruction. J Urol, 167 (3): 1513-21. [PMID:11832780]

39. Hancock AA, Buckner SA, Brune ME, Katwala S, Milicic I, Ireland LM, Morse PA, Knepper SM, Meyer MD,Chapple CR et al.. (1998) Pharmacological characterization of A-131701, a novel R 1 -adrenoceptor antagonist selective for R 1A - and R 1D - compared to R 1B -adrenoceptors. Drug Development Research, 44: 140-162.

40. Harasawa I, Honda K, Tanoue A, Shinoura H, Ishida Y, Okamura H, Murao N, Tsujimoto G, Higa K, Kamiya HO et al.. (2003) Responses to noxious stimuli in mice lacking alpha(1d)-adrenergic receptors. Neuroreport, 14 (14): 1857-60. [PMID:14534435]

41. Hieble JP. (2000) Adrenoceptor subclassification: an approach to improved cardiovascular therapeutics. Pharm Acta Helv, 74 (2-3): 163-71. [PMID:10812954]

42. Hieble JP, Bylund DB, Clarke DE, Eikenburg DC, Langer SZ, Lefkowitz RJ, Minneman KP, Ruffolo Jr RR. (1995) International Union of Pharmacology. X. Recommendation for nomenclature of alpha 1-adrenoceptors: consensus update. Pharmacol Rev, 47 (2): 267-70. [PMID:7568329]

43. Hieble JP, Ruffolo Jr RR. (1996) Subclassification and nomenclature of alpha 1- and alpha 2-adrenoceptors. Prog Drug Res, 47: 81-130. [PMID:8961765]

44. Hodges RR, Shatos MA, Tarko RS, Vrouvlianis J, Gu J, Dartt DA. (2005) Nitric oxide and cGMP mediate alpha1D-adrenergic receptor-Stimulated protein secretion and p42/p44 MAPK activation in rat lacrimal gland. Invest Ophthalmol Vis Sci, 46 (8): 2781-9. [PMID:16043851]

45. Horie K, Obika K, Foglar R, Tsujimoto G. (1995) Selectivity of the imidazoline alpha-adrenoceptor agonists (oxymetazoline and cirazoline) for human cloned alpha 1-adrenoceptor subtypes. Br J Pharmacol, 116 (1): 1611-8. [PMID:8564227]

46. Hosoda C, Koshimizu TA, Tanoue A, Nasa Y, Oikawa R, Tomabechi T, Fukuda S, Shinoura H, Oshikawa S, Takeo S et al.. (2005) Two alpha1-adrenergic receptor subtypes regulating the vasopressor response have differential roles in blood pressure regulation. Mol Pharmacol, 67 (3): 912-22. [PMID:15598970]

47. Hrometz SL, Edelmann SE, McCune DF, Olges JR, Hadley RW, Perez DM, Piascik MT. (1999) Expression of multiple alpha1-adrenoceptors on vascular smooth muscle: correlation with the regulation of contraction. J Pharmacol Exp Ther, 290 (1): 452-63. [PMID:10381812]

48. Huo S, Zhong X, Wu X, Li Y. (2012) Effects of norepinephrine and acetylcholine on the development of cultured Leydig cells in mice. J Biomed Biotechnol, 2012: 503093. [PMID:23093848]

49. Ishihama H, Momota Y, Yanase H, Wang X, de Groat WC, Kawatani M. (2006) Activation of alpha1D adrenergic receptors in the rat urothelium facilitates the micturition reflex. J Urol, 175 (1): 358-64. [PMID:16406942]

50. Janezic EM, Harris DA, Dinh D, Lee KS, Stewart A, Hinds TR, Hsu PL, Zheng N, Hague C. (2019) Scribble co-operatively binds multiple α1D-adrenergic receptor C-terminal PDZ ligands. Sci Rep, 9 (1): 14073. [PMID:31575922]

51. Janezic EM, Lauer SM, Williams RG, Chungyoun M, Lee KS, Navaluna E, Lau HT, Ong SE, Hague C. (2020) N-glycosylation of α1D-adrenergic receptor N-terminal domain is required for correct trafficking, function, and biogenesis. Sci Rep, 10 (1): 7209. [PMID:32350295]

52. Jensen BC, Swigart PM, Laden ME, DeMarco T, Hoopes C, Simpson PC. (2009) The alpha-1D Is the predominant alpha-1-adrenergic receptor subtype in human epicardial coronary arteries. J Am Coll Cardiol, 54 (13): 1137-45. [PMID:19761933]

53. Kandasamy K, Choudhury S, Singh V, Addison MP, Darzi SA, Kasa JK, Thangamalai R, Dash JR, Kumar T, Sultan F et al.. (2016) Erythropoietin Reverses Sepsis-Induced Vasoplegia to Norepinephrine Through Preservation of α1D-Adrenoceptor mRNA Expression and Inhibition of GRK2-Mediated Desensitization in Mouse Aorta. J Cardiovasc Pharmacol Ther, 21 (1): 100-13. [PMID:26025460]

54. Khattar SK, Bora RS, Priyadarsiny P, Gautam A, Gupta D, Tiwari A, Nanda K, Singh R, Chugh A, Bansal V et al.. (2006) Molecular cloning, stable expression and cellular localization of human alpha1-adrenergic receptor subtypes: effect of charcoal/dextran treated serum on expression and localization of alpha1D -adrenergic receptor. Biotechnol Lett, 28 (21): 1731-9. [PMID:16912925]

55. Kojima Y, Sasaki S, Kubota Y, Hayase M, Hayashi Y, Shinoura H, Tsujimoto G, Kohri K. (2008) Expression of alpha1-adrenoceptor subtype mRNA as a predictor of the efficacy of subtype selective alpha1-adrenoceptor antagonists in the management of benign prostatic hyperplasia. J Urol, 179 (3): 1040-6. [PMID:18206918]

56. Kojima Y, Sasaki S, Kubota Y, Imura M, Oda N, Kiniwa M, Hayashi Y, Kohri K. (2011) Up-regulation of α1a and α1d-adrenoceptors in the prostate by administration of subtype selective α1-adrenoceptor antagonist tamsulosin in patients with benign prostatic hyperplasia. J Urol, 186 (4): 1530-6. [PMID:21855934]

57. Koshimizu TA, Tanoue A, Hirasawa A, Yamauchi J, Tsujimoto G. (2003) Recent advances in alpha1-adrenoceptor pharmacology. Pharmacol Ther, 98 (2): 235-44. [PMID:12725871]

58. Koshimizu TA, Tsujimoto G, Hirasawa A, Kitagawa Y, Tanoue A. (2004) Carvedilol selectively inhibits oscillatory intracellular calcium changes evoked by human alpha1D- and alpha1B-adrenergic receptors. Cardiovasc Res, 63 (4): 662-72. [PMID:15306222]

59. Kountz TS, Lee KS, Aggarwal-Howarth S, Curran E, Park JM, Harris DA, Stewart A, Hendrickson J, Camp ND, Wolf-Yadlin A et al.. (2016) Endogenous N-terminal Domain Cleavage Modulates α1D-Adrenergic Receptor Pharmacodynamics. J Biol Chem, 291 (35): 18210-21. [PMID:27382054]

60. Leech CJ, Faber JE. (1996) Different alpha-adrenoceptor subtypes mediate constriction of arterioles and venules. Am J Physiol, 270 (2 Pt 2): H710-22. [PMID:8779849]

61. Liu CM, Lo YC, Wu BN, Wu WJ, Chou YH, Huang CH, An LM, Chen IJ. (2007) cGMP-enhancing- and alpha1A/alpha1D-adrenoceptor blockade-derived inhibition of Rho-kinase by KMUP-1 provides optimal prostate relaxation and epithelial cell anti-proliferation efficacy. Prostate, 67 (13): 1397-410. [PMID:17639498]

62. Liu R, Zhang Q, Luo Q, Qiao H, Wang P, Yu J, Cao Y, Lu B, Qu L. (2017) Norepinephrine stimulation of alpha1D-adrenoceptor promotes proliferation of pulmonary artery smooth muscle cells via ERK-1/2 signaling. Int J Biochem Cell Biol, 88: 100-112. [PMID:28476501]

63. Lomasney JW, Cotecchia S, Lorenz W, Leung WY, Schwinn DA, Yang-Feng TL, Brownstein M, Lefkowitz RJ, Caron MG. (1991) Molecular cloning and expression of the cDNA for the alpha 1A-adrenergic receptor. The gene for which is located on human chromosome 5. J Biol Chem, 266 (10): 6365-9. [PMID:1706716]

64. Lyssand JS, DeFino MC, Tang XB, Hertz AL, Feller DB, Wacker JL, Adams ME, Hague C. (2008) Blood pressure is regulated by an alpha1D-adrenergic receptor/dystrophin signalosome. J Biol Chem, 283 (27): 18792-800. [PMID:18468998]

65. Lyssand JS, Lee KS, DeFino M, Adams ME, Hague C. (2011) Syntrophin isoforms play specific functional roles in the α1D-adrenergic receptor/DAPC signalosome. Biochem Biophys Res Commun, 412 (4): 596-601. [PMID:21846462]

66. Lyssand JS, Whiting JL, Lee KS, Kastl R, Wacker JL, Bruchas MR, Miyatake M, Langeberg LK, Chavkin C, Scott JD et al.. (2010) Alpha-dystrobrevin-1 recruits alpha-catulin to the alpha1D-adrenergic receptor/dystrophin-associated protein complex signalosome. Proc Natl Acad Sci USA, 107 (50): 21854-9. [PMID:21115837]

67. Malloy BJ, Price DT, Price RR, Bienstock AM, Dole MK, Funk BL, Rudner XL, Richardson CD, Donatucci CF, Schwinn DA. (1998) Alpha1-adrenergic receptor subtypes in human detrusor. J Urol, 160 (3 Pt 1): 937-43. [PMID:9720591]

68. Methven L, Simpson PC, McGrath JC. (2009) Alpha1A/B-knockout mice explain the native alpha1D-adrenoceptor's role in vasoconstriction and show that its location is independent of the other alpha1-subtypes. Br J Pharmacol, 158 (7): 1663-75. [PMID:19888965]

69. Meyer MD, Altenbach RJ, Basha FZ, Carroll WA, Drizin I, Elmore SW, Ehrlich PP, Lebold SA, Tietje K, Sippy KB et al.. (1997) Synthesis and pharmacological characterization of 3-[2-((3aR,9bR)-cis-6-methoxy-2,3,3a,4,5,9b-hexahydro-1H-benz[e] isoindol-2-yl)ethyl]pyrido-[3',4':4,5]thieno[3,2-d]pyrimidine-2,4 (1H,3H)-dione (A-131701): a uroselective alpha 1A adrenoceptor antagonist for the symptomatic treatment of benign prostatic hyperplasia. J Med Chem, 40 (20): 3141-3. [PMID:9379432]

70. Michelotti GA, Brinkley DM, Morris DP, Smith MP, Louie RJ, Schwinn DA. (2007) Epigenetic regulation of human alpha1d-adrenergic receptor gene expression: a role for DNA methylation in Sp1-dependent regulation. FASEB J, 21 (9): 1979-93. [PMID:17384146]

71. Michelotti GA, Price DT, Schwinn DA. (2000) Alpha 1-adrenergic receptor regulation: basic science and clinical implications. Pharmacol Ther, 88 (3): 281-309. [PMID:11337028]

72. Minarini A, Budriesi R, Chiarini A, Leonardi A, Melchiorre C. (1998) Search for alpha 1-adrenoceptor subtypes selective antagonists: design, synthesis and biological activity of cystazosin, an alpha 1D-adrenoceptor antagonist. Bioorg Med Chem Lett, 8 (11): 1353-8. [PMID:9871765]

73. Minneman KP, Theroux TL, Hollinger S, Han C, Esbenshade TA. (1994) Selectivity of agonists for cloned alpha 1-adrenergic receptor subtypes. Mol Pharmacol, 46 (5): 929-36. [PMID:7969082]

74. Mishima K, Tanoue A, Tsuda M, Hasebe N, Fukue Y, Egashira N, Takano Y, Kamiya HO, Tsujimoto G, Iwasaki K et al.. (2004) Characteristics of behavioral abnormalities in alpha1d-adrenoceptors deficient mice. Behav Brain Res, 152 (2): 365-73. [PMID:15196805]

75. Mizusawa H, Hedlund P, Sjunnesson J, Brioni JD, Sullivan JP, Andersson KE. (2002) Enhancement of apomorphine-induced penile erection in the rat by a selective alpha(1D)-adrenoceptor antagonist. Br J Pharmacol, 136 (5): 701-8. [PMID:12086979]

76. Morelli MB, Amantini C, Nabissi M, Liberati S, Cardinali C, Farfariello V, Tomassoni D, Quaglia W, Piergentili A, Bonifazi A et al.. (2014) Cross-talk between alpha1D-adrenoceptors and transient receptor potential vanilloid type 1 triggers prostate cancer cell proliferation. BMC Cancer, 14: 921. [PMID:25481381]

77. Nagaoka Y, Ahmed M, Hossain M, Bhuiyan MA, Ishiguro M, Nakamura T, Watanabe M, Nagatomo T. (2008) Amino acids of the human alpha1d-adrenergic receptor involved in antagonist binding. J Pharmacol Sci, 106 (1): 114-20. [PMID:18187928]

78. Nicholas AP, Hökfelt T, Pieribone VA. (1996) The distribution and significance of CNS adrenoceptors examined with in situ hybridization. Trends Pharmacol Sci, 17 (7): 245-55. [PMID:8756183]

79. Nonen S, Okamoto H, Fujio Y, Takemoto Y, Yoshiyama M, Hamaguchi T, Matsui Y, Yoshikawa J, Kitabatake A, Azuma J. (2008) Polymorphisms of norepinephrine transporter and adrenergic receptor alpha1D are associated with the response to beta-blockers in dilated cardiomyopathy. Pharmacogenomics J, 8 (1): 78-84. [PMID:17404580]

80. Nourian Z, Mow T, Muftic D, Burek S, Pedersen ML, Matz J, Mulvany MJ. (2008) Orthostatic hypotensive effect of antipsychotic drugs in Wistar rats by in vivo and in vitro studies of alpha1-adrenoceptor function. Psychopharmacology (Berl.), 199 (1): 15-27. [PMID:18542932]

81. Obika K, Shibata K, Horie K, Foglar R, Kimura K, Tsujimoto G. (1995) NS-49, a novel alpha 1a-adrenoceptor-selective agonist characterization using recombinant human alpha 1-adrenoceptors. Eur J Pharmacol, 291 (3): 327-34. [PMID:8719417]

82. Parkman HP, Jacobs MR, Mishra A, Hurdle JA, Sachdeva P, Gaughan JP, Krynetskiy E. (2011) Domperidone treatment for gastroparesis: demographic and pharmacogenetic characterization of clinical efficacy and side-effects. Dig Dis Sci, 56 (1): 115-24. [PMID:21063774]

83. Paton DM. (1976) Adrenergic innervation of the oviduct in the regulation of ovum transport. Res Reprod, 8 (1): 3. [PMID:1251089]

84. Pernomian L, Gomes MS, Restini CB, Pupo AS, de Oliveira AM. (2013) Cross-talk with β2 -adrenoceptors enhances ligand affinity properties from endothelial alpha1 D -adrenoceptors that mediates carotid relaxation. J Pharm Pharmacol, 65 (9): 1337-46. [PMID:23927472]

85. Petrovska R, Kapa I, Klovins J, Schiöth HB, Uhlén S. (2005) Addition of a signal peptide sequence to the alpha1D-adrenoceptor gene increases the density of receptors, as determined by [3H]-prazosin binding in the membranes. Br J Pharmacol, 144 (5): 651-9. [PMID:15678090]

86. Proudman RGW, Baker JG. (2021) The selectivity of α-adrenoceptor agonists for the human α1A, α1B, and α1D-adrenoceptors. Pharmacol Res Perspect, 9 (4): e00799. [PMID:34355529]

87. Proudman RGW, Pupo AS, Baker JG. (2020) The affinity and selectivity of α-adrenoceptor antagonists, antidepressants, and antipsychotics for the human α1A, α1B, and α1D-adrenoceptors. Pharmacol Res Perspect, 8 (4): e00602. [PMID:32608144]

88. Rodríguez JE, Saucedo-Campos AD, Vega AV, Ramírez-Hernández D, Martínez-Aguilar L, Jiménez-Flores JR, Andrade-Jorge E, Estrada-Soto SE, Villalobos-Molina R, Touyz RM et al.. (2020) The α1D-adrenoreceptor antagonist BMY 7378 reverses cardiac hypertrophy in spontaneously hypertensive rats. J Hypertens, 38 (8): 1496-1503. [PMID:32195823]

89. Rodríguez-Pérez CE, Calvo-Ochoa E, Kalashnikova EV, Reyes-Cruz G, Romero-Avila MT, García-Sáinz JA. (2009) Receptor tyrosine kinases regulate alpha1D-adrenoceptor signaling properties: phosphorylation and desensitization. Int J Biochem Cell Biol, 41 (6): 1276-83. [PMID:19038360]

90. Romeo G, Salerno L, Pittalà V, Modica MN, Siracusa MA, Materia L, Buccioni M, Marucci G, Minneman KP. (2014) High affinity ligands and potent antagonists for the α1D-adrenergic receptor. Novel 3,8-disubstituted [1]benzothieno[3,2-d]pyrimidine derivatives. Eur J Med Chem, 83: 419-32. [PMID:24992070]

91. Rudner XL, Berkowitz DE, Booth JV, Funk BL, Cozart KL, D'Amico EB, El-Moalem H, Page SO, Richardson CD, Winters B et al.. (1999) Subtype specific regulation of human vascular alpha(1)-adrenergic receptors by vessel bed and age. Circulation, 100 (23): 2336-43. [PMID:10587338]

92. Sadalge A, Coughlin L, Fu H, Wang B, Valladares O, Valentino R, Blendy JA. (2003) alpha 1d Adrenoceptor signaling is required for stimulus induced locomotor activity. Mol Psychiatry, 8 (7): 664-72. [PMID:12874602]

93. Sakauchi N, Furukawa H, Shirai J, Sato A, Kuno H, Saikawa R, Yoshida M. (2017) Identification of 3,4-dihydro-2H-thiochromene 1,1-dioxide derivatives with a phenoxyethylamine group as highly potent and selective α1D adrenoceptor antagonists. Eur J Med Chem, 139: 114-127. [PMID:28800452]

94. Santana N, Mengod G, Artigas F. (2013) Expression of α(1)-adrenergic receptors in rat prefrontal cortex: cellular co-localization with 5-HT(2A) receptors. Int J Neuropsychopharmacol, 16 (5): 1139-51. [PMID:23195622]

95. Schwinn DA, Johnston GI, Page SO, Mosley MJ, Wilson KH, Worman NP, Campbell S, Fidock MD, Furness LM, Parry-Smith DJ et al.. (1995) Cloning and pharmacological characterization of human alpha-1 adrenergic receptors: sequence corrections and direct comparison with other species homologues. J Pharmacol Exp Ther, 272 (1): 134-42. [PMID:7815325]

96. Schwinn DA, Michelotti GA. (2000) alpha1-adrenergic receptors in the lower urinary tract and vascular bed: potential role for the alpha1d subtype in filling symptoms and effects of ageing on vascular expression. BJU Int, 85 Suppl 2: 6-11. [PMID:10781179]

97. Segura V, Flacco N, Oliver E, Barettino D, D'Ocon P, Ivorra MD. (2010) Alpha1-adrenoceptors in the rat cerebral cortex: new insights into the characterization of alpha1L- and alpha1D-adrenoceptors. Eur J Pharmacol, 641 (1): 41-8. [PMID:20511116]

98. Shibata K, Foglar R, Horie K, Obika K, Sakamoto A, Ogawa S, Tsujimoto G. (1995) KMD-3213, a novel, potent, alpha 1a-adrenoceptor-selective antagonist: characterization using recombinant human alpha 1-adrenoceptors and native tissues. Mol Pharmacol, 48 (2): 250-8. [PMID:7651358]

99. Sigala S, Peroni A, Mirabella G, Fornari S, Palazzolo F, Pezzotti G, Simeone C, Cunico SC, Spano P. (2004) Alpha1 adrenoceptor subtypes in human urinary bladder: sex and regional comparison. Life Sci, 76 (4): 417-27. [PMID:15530504]

100. Szot P, White SS, Greenup JL, Leverenz JB, Peskind ER, Raskind MA. (2006) Compensatory changes in the noradrenergic nervous system in the locus ceruleus and hippocampus of postmortem subjects with Alzheimer's disease and dementia with Lewy bodies. J Neurosci, 26 (2): 467-78. [PMID:16407544]

101. Szot P, White SS, Greenup JL, Leverenz JB, Peskind ER, Raskind MA. (2007) Changes in adrenoreceptors in the prefrontal cortex of subjects with dementia: evidence of compensatory changes. Neuroscience, 146 (1): 471-80. [PMID:17324522]

102. Takakura K, Taniguchi T, Muramatsu I, Takeuchi K, Fukuda S. (2002) Modification of alpha1 -adrenoceptors by peroxynitrite as a possible mechanism of systemic hypotension in sepsis. Crit Care Med, 30 (4): 894-9. [PMID:11940765]

103. Tanoue A, Koba M, Miyawaki S, Koshimizu TA, Hosoda C, Oshikawa S, Tsujimoto G. (2002) Role of the alpha1D-adrenergic receptor in the development of salt-induced hypertension. Hypertension, 40 (1): 101-6. [PMID:12105146]

104. Tanoue A, Nasa Y, Koshimizu T, Shinoura H, Oshikawa S, Kawai T, Sunada S, Takeo S, Tsujimoto G. (2002) The alpha(1D)-adrenergic receptor directly regulates arterial blood pressure via vasoconstriction. J Clin Invest, 109 (6): 765-75. [PMID:11901185]

105. Tayebati SK, Bronzetti E, Morra Di Cella S, Mulatero P, Ricci A, Rossodivita I, Schena M, Schiavone D, Veglio F, Amenta F. (2000) In situ hybridization and immunocytochemistry of alpha1-adrenoceptors in human peripheral blood lymphocytes. J Auton Pharmacol, 20 (5-6): 305-12. [PMID:11350496]

106. Uberti MA, Hague C, Oller H, Minneman KP, Hall RA. (2005) Heterodimerization with beta2-adrenergic receptors promotes surface expression and functional activity of alpha1D-adrenergic receptors. J Pharmacol Exp Ther, 313 (1): 16-23. [PMID:15615865]

107. Ulu N, Henning RH, Guner S, Zoto T, Duman-Dalkilic B, Duin M, Gurdal H. (2013) Intracellular transactivation of epidermal growth factor receptor by α1A-adrenoceptor is mediated by phosphatidylinositol 3-kinase independently of activation of extracellular signal regulated kinases 1/2 and serine-threonine kinases in Chinese hamster ovary cells. J Pharmacol Exp Ther, 347 (1): 47-56. [PMID:23902938]

108. Vinci MC, Bellik L, Filippi S, Ledda F, Parenti A. (2007) Trophic effects induced by alpha1D-adrenoceptors on endothelial cells are potentiated by hypoxia. Am J Physiol Heart Circ Physiol, 293 (4): H2140-7. [PMID:17660397]

109. Walden PD, Gerardi C, Lepor H. (1999) Localization and expression of the alpha1A-1, alpha1B and alpha1D-adrenoceptors in hyperplastic and non-hyperplastic human prostate. J Urol, 161 (2): 635-40. [PMID:9915474]

110. Wang SY, Song Y, Xu M, He QH, Han QD, Zhang YY. (2007) Internalization and distribution of three alpha1-adrenoceptor subtypes in HEK293A cells before and after agonist stimulation. Acta Pharmacol Sin, 28 (3): 359-66. [PMID:17302998]

111. Waugh DJ, Gaivin RJ, Damron DS, Murray PA, Perez DM. (1999) Binding, partial agonism, and potentiation of alpha(1)-adrenergic receptor function by benzodiazepines: A potential site of allosteric modulation. J Pharmacol Exp Ther, 291 (3): 1164-71. [PMID:10565838]

112. Williams TJ, Blue DR, Daniels DV, Davis B, Elworthy T, Gever JR, Kava MS, Morgans D, Padilla F, Tassa S et al.. (1999) In vitro alpha1-adrenoceptor pharmacology of Ro 70-0004 and RS-100329, novel alpha1A-adrenoceptor selective antagonists. Br J Pharmacol, 127 (1): 252-8. [PMID:10369480]

113. Yoshio R, Taniguchi T, Itoh H, Muramatsu I. (2001) Affinity of serotonin receptor antagonists and agonists to recombinant and native alpha1-adrenoceptor subtypes. Jpn J Pharmacol, 86 (2): 189-95. [PMID:11459121]

114. Zacharia J, Hillier C, Tanoue A, Tsujimoto G, Daly CJ, McGrath JC, MacDonald A. (2005) Evidence for involvement of alpha1D-adrenoceptors in contraction of femoral resistance arteries using knockout mice. Br J Pharmacol, 146 (7): 942-51. [PMID:16170328]

115. Zhou SG, Lu JL, Hui JH. (2011) Comparing efficacy of α1D-receptor antagonist naftopidil and α1A/D-receptor antagonist tamsulosin in management of distal ureteral stones. World J Urol, 29 (6): 767-71. [PMID:21845472]

Contributors

Show »

How to cite this page