Skip to main content
British Journal of Pharmacology logoLink to British Journal of Pharmacology
. 2012 Mar;165(5):1223–1225. doi: 10.1111/j.1476-5381.2011.01663.x

What makes the α1A-adrenoceptor gene express the α1L-adrenoceptor functional phenotype?

S Ventura 1
PMCID: PMC3372710  PMID: 21913893

Abstract

The α1A-adrenoceptor is therapeutically exploited because of its prevalence in the lower urinary tract. The pharmacology shown by this lower urinary tract α1A-adrenoceptor is different from that shown by other α1A-adrenoceptors, which has led to it being subclassified as an α1L-adrenoceptor. Only in the last few years was it shown that this pharmacologically distinct α1L-adrenoceptor is a product of the α1A-adrenoceptor gene. In this issue of the BJP, Nishimune et al. review the literature on α1L-adrenoceptor pharmacology and discuss the possible molecular mechanisms by which the α1A-adrenoceptor gene is able to produce two pharmacologically distinct adrenoceptor subtypes. Based primarily from their own research using cell lines transfected with α1A-adrenoceptors, they conclude that a protein that interacts with the receptor is the most plausible explanation. The challenge remains to identify any such interacting protein and show how it is able to change the pharmacology of the receptor for different ligands.

LINKED ARTICLE

This article is a commentary on Nishimune et al., pp. 1226–1234 of this issue. To view this paper visit http://dx.doi.org/10.1111/j.1476-5381.2011.01591.x

Keywords: α-adrenoceptor, benign prostatic hyperplasia (BPH), cysteine-rich epidermal growth factor-like domain (CRELD), lower urinary tract, prostate, receptor interacting proteins, prazosin


The most effective and rapidly acting pharmacological treatments for benign prostatic hyperplasia (BPH) are the α1A-adrenoceptor antagonists, such as tamsulosin and alfuzosin (Miano et al., 2008; receptor nomenclature follows Alexander et al., 2011). This class of BPH therapeutic agents makes over US$ 3 billion in worldwide sales (Ventura et al., 2011). Previously, non-selective α1-adrenoceptor antagonists such as prazosin, doxazosin and terazosin were widely used, but these have now been largely superseded by tamsulosin and alfuzosin because of their greater selectivity for the α1A-adrenoceptor subtype over the α1B and α1D-adrenoceptor subtypes. The proportion of the α1A-adrenoceptor subtype expressed in the smooth muscle stroma of the prostate gland is greater than the proportion expressed in vascular smooth muscle, leading to a lower incidence of troublesome vascular side effects such as weakness, fatigue, postural hypotension and dizziness, which were commonplace with the use of the non-selective α1-adrenoceptor antagonists.

α1A-Adrenoceptors are abundant in the male lower urinary tract, and α1A-adrenoceptor antagonists are very effective in relieving lower urinary tract symptoms associated with urethral obstruction caused by prostate enlargement. Despite this, prostate and other lower urinary tract tissues, from all species, do not show typical α1A-adrenoceptor pharmacology (Nishimune et al., 2012). When used in functional isolated tissue experiments, isolated tissues from prostate gland, urethra and bladder, all exhibit a low affinity for prazosin when compared with other α1-adrenoceptor-expressing tissues. A corresponding change in affinity is not seen with tamsulosin. This pharmacological anomaly led to the postulate that a fourth α1-adrenoceptor existed, which was termed the α1L-adrenoceptor.

Only recently has it been demonstrated with the use of genetically modified adrenoceptor knockout mice that the prostatic α1L-adrenoceptor phenotype requires the expression of the α1A-adrenoceptor gene (Gray et al., 2008; Muramatsu et al., 2008). The term α1L-adrenoceptor is not currently recognized as an official nomenclature term. Rather, the latest edition of the Guide to Receptors and Channels states, ‘Some tissues possess α1A-adrenoceptors that display relatively low affinity in functional and binding assays for prazosin (pKi < 9) that might represent different receptor states (termed α1L-adrenoceptors)’ (Alexander et al., 2011). Further, investigation of this phenomenon is critical to developing a better treatment for BPH as it would seem that men suffering from urethral obstruction resulting from BPH would benefit more from a selective α1L-adrenoceptor antagonist rather than the selective α1A-adrenoceptor antagonists like tamsulosin, which are currently used and show no selectivity between α1A and α1L-adrenoceptors. At present, there are no antagonists showing higher affinity for α1L over α1A-adrenoceptors.

Early attempts to explain how the α1L-adrenoceptor phenotype could arise from the α1A-adrenoceptor gene concentrated on whether genetic polymorphisms or splice variants of this gene could give rise to the phenotype. However, α1A-adrenoceptors generated by known polymorphisms and splice variants in cell culture models all showed similar pharmacological characteristics to that of the α1A-adrenoceptor (Shibata et al., 1996; Suzuki et al., 2000; Ramsay et al., 2004), providing evidence that α1A-adrenoceptor polymorphisms and splice variants were not associated with generation of the α1L-adrenoceptor phenotype.

Subsequently, a ‘interacting protein’ hypothesis to explain the generation of α1L-adrenoceptors from α1A-adrenocepors has been postulated, following observations from radioligand binding studies. The basis for this hypothesis is that radioligand binding studies of lower urinary tract tissues are almost always carried out using membrane homogenates and yield ligand affinities that fit the pharmacological profile of α1A-adrenoceptor pharmacology. This is despite the findings that isolated intact preparations of prostate, urethra and bladder tissue display α1L-adrenoceptor pharmacology when they have been used in functional studies. In an earlier review, Nishimune et al., (2010a) suggested that this discrepancy was caused by the homogenization process disrupting the cell membrane and thus separating α1A-adrenoceptors from the putative ‘interacting protein’. They hypothesized that only when the α1A-adrenoceptor is bound to the interacting protein does it display α1L-adrenoceptor pharmacology. This idea is supported by their earlier paper that demonstrated that radioligand binding studies of tissue segments from lower urinary tract tissues produced a α1L-adrenoceptor ligand affinity profile, while crude homogenized membrane fractions from these tissues yielded a α1A-adrenoceptor profile (Muramatsu et al., 2005). The presumption is that the more intact tissue segments maintain a more complete membrane with little or no disruption to the α1A-adrenoceptor – interacting protein complex.

This interacting protein theory is a plausible and logical explanation for the occurrence of α1L-adrenoceptor pharmacology in lower urinary tract tissues expressing abundant α1A-adrenoceptors. Indeed, the interaction of proteins also covers the possibility that a receptor heteromer may be the cause of the changed pharmacology of α1A-adrenoceptors in the lower urinary tract. However, it is arguable whether tissue homogenization would disrupt the cell membrane sufficiently to destroy protein–protein interactions at the molecular level. Nevertheless, Nishimune et al. (2010b) identified cysteine-rich epidermal growth factor-like domain 1α (CRELD1α) as a novel down-regulating protein and therefore a protein that interacts with the α1A-adrenoceptor. CRELD1α was identified using a yeast two-hybrid approach, with the entire open reading frame of the human α1A-adrenoceptor gene used as bait (Nishimune et al., 2010b). Subsequent transfection of cDNA for the α1A-adrenoceptor gene alone into CHO cells yielded cells expressing α1A-adrenoceptors with the typical α1A-adrenoceptor pharmacological profile, as well as a low proportion of α1L-adrenoceptor sites. The small number of α1L-adrenoceptors was presumably due to endogenous CRELD1α as knockdown of CRELD1α enhanced the expression of α1A-adrenoceptors while over-expression of CRELD1α reduced α1A-adrenoceptor expression (Nishimune et al., 2010b). Following this, they were able to produce α1A-adrenoceptor-enhanced and α1L-adrenoceptor-dominant cell lines that were used in ligand binding, and functional agonist and antagonist profile studies. Results for α1A-adrenoceptor-enhanced and α1L-adrenoceptor-dominant CHO cells were in agreement with the published profiles for α1A and α1L-adrenoceptor phenotypes, respectively, as seen in intact tissues.

Although the evidence presented in this paper is persuasive (Nishimune et al., 2010b), there are still questions to be answered before the story is truly convincing. For instance, CRELD1α over-expression yielded α1L-adrenoceptor dominant cells expressing a higher proportion of α1L-adrenoceptors; however, this was because of a reduction in α1A-adrenoceptor binding sites rather than their conversion to α1L-adrenoceptors as would be expected if CRELD1α were a true α1A-adrenoceptor interacting protein. Consequently, the expression of α1L-adrenoceptor binding sites does not appear to change regardless of CRELD1α expression. Alternatively, this observation itself could be interpreted as the CRELD protein inhibiting radioligand binding in some way, perhaps by internalization of the α1A-adrenoceptor rather than changing its binding affinity to that of the phenotype of the α1L-adrenoceptor. Experiments using membrane-permeable and membrane-impermeable agonists and antagonists would go a long way towards a clearer understanding of these results.

Other observations from this research throw up questions that need answers. For instance, the efficacy of agonists seen in cells with different levels of CRELD1α expression differs when their activity is compared in functional assays. This introduces the possibility of ligand-biased signalling, which needs to be addressed and could further confound progress in this area. Similar agonist efficacy differences have been observed in functional experiments with intact tissues expressing the different phenotypes. Furthermore, over-expression of CRELD1α (α1L-dominant) also seemed to introduce an element of irreversible antagonism to prazosin at low concentrations, when compared with CRELD1α knockdown (α1A-abundant) cells in functional assays.

The idea of a α1A-adrenoceptor interacting protein is a logical and plausible argument to explain the transition from α1A-adrenoceptor gene to the α1L-adrenoceptor functional phenotype which is abundant in male lower urinary tract tissue. CRELD1α appears to go part of the way to fulfilling the criteria that one would expect of an interacting protein for this receptor, but further experimental challenges remain to strengthen its case. Such experiments might include radioligand binding of whole cells versus membrane preparations using α1A-adrenoceptor-expressing CHO cells with and without simultaneous CRELD1α transfection. This would show whether the α1A-adrenoceptor – CRELD1α complex can be disrupted during membrane homogenization, leading to a change in pharmacological profile. Furthermore, demonstration of a direct interaction of the α1A-adrenoceptor and CRELD1α using resonance energy transfer techniques (FRET/BRET) in transfected CHO cells or immunoprecipitation techniques in native tissue could provide significant support for this hypothesis.

Glossary

BPH

benign prostatic hyperplasia

BRET

bioluminescence resonance energy transfer

CRELD

cysteine-rich epidermal growth factor-like domain

FRET

fluorescence resonance energy transfer

Conflicts of interest

The author states no conflict of interest.

References

  1. Alexander SPH, Mathie A, Peters JA. Guide to Receptors and Channels (GRAC), 5th edn. Br J Pharmacol. 2011;164(Suppl.1):S1–S324. doi: 10.1111/j.1476-5381.2011.01649_1.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Gray KT, Short JL, Ventura S. The α1A-adrenoceptor gene is required for the α1L-adrenoceptor mediated response in isolated preparations of the mouse prostate. Br J Pharmacol. 2008;155:103–109. doi: 10.1038/bjp.2008.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Miano R, De Nunzio C, Asimakopoulos AD, Germani S, Tubaro A. Treatment options for benign prostatic hyperplasia in older men. Med Sci Monit. 2008;14:RA94–R102. [PubMed] [Google Scholar]
  4. Muramatsu I, Tanaka T, Suzuki F, Li Z, Hiraizumi-Hiraoka Y, Anisuzzaman ASM, et al. Quantifying receptor properties: the tissue segment binding method – a powerful tool for the pharmacome analysis of native receptors. J Pharmacol Sci. 2005;98:331–339. doi: 10.1254/jphs.cpj05001x. [DOI] [PubMed] [Google Scholar]
  5. Muramatsu I, Morishima S, Suzuki F, Yoshiki H, Anisuzzaman AS, Tanaka T, et al. Identification of α1L-adrenoceptor in mice and its abolition by α1A-adrenoceptor gene knockout. Br J Pharmacol. 2008;155:1224–1234. doi: 10.1038/bjp.2008.360. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Nishimune A, Suzuki F, Yoshiki H, Morishima S, Muramatsu I. α1A-Adrenoceptor pharmacome: α1L-Adrenoceptor and α1A-adrenoceptor in the lower urinary tract. Int J Urol. 2010a;17:31–37. doi: 10.1111/j.1442-2042.2009.02368.x. [DOI] [PubMed] [Google Scholar]
  7. Nishimune A, Suzuki F, Yoshiki H, Morishima S, Muramatsu I. Identification of cysteine-rich epidermal growth factor-like domain 1α (CRELD1α) as a novel α1A-adrenoceptor-down-regulating protein and establishment of an α1L-adrenoceptor-expressing cell line. J Pharmacol Sci. 2010b;113:169–181. doi: 10.1254/jphs.10093fp. [DOI] [PubMed] [Google Scholar]
  8. Nishimune A, Yoshiki J, Uwada J, Anisuzzaman ASM, Umada H, Muramatsu I. Phenotype pharmacology of lower urinary tract α1-adrenoceptors. Br J Pharmacol. 2012;165:1226–1234. doi: 10.1111/j.1476-5381.2011.01591.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Ramsay D, Carr IC, Pediani J, Lopez-Gimenez JF, Thurlow R, Fidock M, et al. High-affinity interactions between human α1A-adrenoceptor C-terminal splice variants produce homo- and heterodimers but do not generate the α1L-adrenoceptor. Mol Pharmacol. 2004;66:228–239. doi: 10.1124/mol.66.2.228. [DOI] [PubMed] [Google Scholar]
  10. Shibata K, Hirasawa A, Moriyama N, Kawabe K, Ogawa S, Tsujimoto G. α1a-adrenoceptor polymorphism: pharmacological characterization and association with benign prostatic hypertrophy. Br J Pharmacol. 1996;118:1403–1408. doi: 10.1111/j.1476-5381.1996.tb15552.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Suzuki F, Taniguchi T, Takauji R, Murata S, Muramatsu I. Splice isoforms of α1a-adrenoceptor in rabbit. Br J Pharmacol. 2000;129:1569–1576. doi: 10.1038/sj.bjp.0703242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Ventura S, Oliver VL, White CW, Xie JH, Haynes JM, Exintaris B. Novel drug targets for the pharmacotherapy of benign prostatic hyperplasia (BPH) Br J Pharmacol. 2011;163:891–907. doi: 10.1111/j.1476-5381.2011.01332.x. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from British Journal of Pharmacology are provided here courtesy of The British Pharmacological Society

RESOURCES