Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Apr 11.
Published in final edited form as: J Recept Signal Transduct Res. 2011 Feb 21;31(2):98–110. doi: 10.3109/10799893.2010.550008

Cardiac and neuroprotection regulated by α1-adrenergic receptor subtypes

Dianne M Perez 1, Van A Doze 2
PMCID: PMC3623295  NIHMSID: NIHMS454479  PMID: 21338248

Abstract

Sympathetic nervous system regulation by the α1-adrenergic receptor (AR) subtypes (α1A, α1B, α1D) 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 α1-AR subtypes in the context of cardiac hypertrophy, heart failure, apoptosis, ischemic preconditioning, neurogenesis, locomotion, neurodegeneration, cognition, neuroplasticity, depression, anxiety, epilepsy, and mental illness.

Keywords: Adrenergic, cardiac, cognition, epilepsy, heart failure, ischemia, receptor, neurodegeneration, neurogenesis, neurological, norepinephrine, preconditioning, protection, stem Cells

Nomenclature, initial tissue characterization, and cloning

α1-Adrenergic receptor (AR) subtypes (α1A, α1B, and α1D) are G-protein-coupled receptors (GPCRs) that mediate the sympathetic nervous system by binding the endogenous catecholamines, epinephrine, and norepinephrine (NE) (1). Raymond Ahlquist (2) introduced the initial concept of different subtypes of ARs (α and β), and all nine of the adrenergic subtypes (α1A, α1B, α1D, α2A, α2B, α2C, β1, β2, and β3,) are activated by the same catecholamines.

After further characterization in tissue, α1-ARs were subdivided into the α1A- and α1B-AR subtypes in the late 1980s based upon experimental data of two-site competition binding curves in rat brain to the antagonists WB4101 and phentolamine. The α1A-AR subtype was characterized as having a 10–100-fold higher affinity for these ligands than the α1B-AR subtype (3). During this same time period, the α1B-AR was cloned utilizing oligonucleotide probes made by peptide fragments of purified receptor (4). This receptor was correctly classified as the α1B-AR because of the cloned receptor’s lower affinity for WB4101 and phentolamine. The next α1-AR cloned was designated the α1C-AR, considered a newly discovered α1-AR subtype, because it did not neatly fit into previous pharmacological criteria for the α1A-AR subtype (5). This expressed receptor did have high affinity for typical α1A-AR ligands. However, because it was isolated from a bovine cDNA library, which had high sequence variation to the rat gene, its expression could not be detected in rat tissues. In addition, the receptor was insensitive to chloroethyl clonidine (an α1A-AR criteria). Thus, not having α1A-AR-like tissue distribution and having sensitivity to chloroethyl clonidine, an alkylating agent mistakenly thought at the time to be selective for the α1B-AR, led to this receptor being misclassified. A few years later, two groups recloned the same receptor from a rat cDNA library, whose tissues were previously characterized for α1A-AR pharmacology and thus allowing for a more accurate comparison. Both groups demonstrated that the misclassified α1C-AR really represented the tissue-defined α1A-AR (6,7). Before the publication of this corrective work for the α1C-AR misclassification, another receptor was cloned and was designated the α1A-AR because this receptor had high affinity for WB4101 (8). In reality, this clone was the real novel receptor subtype, not being previously pharmacologically described in tissues, and was independently cloned and more extensively characterized to reveal its novel pharmacology and was named the α1D-AR (9). This classification was accepted by the IUPHAR Adrenergic Receptor Subcommittee (10). Therefore, the α1C-AR subtype designation does not exist anymore and three α1-AR subtypes have now been fully characterized in both expressed systems and native tissues: the α1A, α1B, and α1D-ARs (10). With the sequencing of mammalian genomes, there does not appear to be additional AR subtypes.

Cardiac physiology

Myocardial α1-ARs

Of the ARs, myocardium contains β- and α1-ARs. Of the α1-ARs, the myocyte contains both the α1A- and α1B-AR subtypes. There are no α2-ARs present in the myocyte as well as little if any expression of the α1D-AR, despite the present of its mRNA in PCR studies (11,12). There is evidence to suggest that α1-ARs are also present in the cardiac fibroblast (13,14) and may regulate protein synthesis and secretion that are also needed for cardiac function. The ARs regulate both the contractility as well as the growth of the myocardium. While the β-ARs predominate in the regulation of heart function under normal physiological conditions, the α1-ARs are thought to become more important during pathological conditions and disease, such as hypertrophy, heart failure, and ischemic disease. For example, α1-ARs are generally thought to be more important in preserving or increasing myocardial contractility in the setting of heart failure and β-AR downregulation (1517). Since α1-ARs are GPCRs, the major signaling pathways utilize Gq, coupling to phospholipase Cβ and resulting in the membrane release of inositol 1,4,5- trisphosphate (IP3) and diacylglycerol. These second messengers activate the release of intracellular calcium and activation of PKC, respectively. However, there is evidence that α1A- and α1B-AR subtypes are differentially coupled to different G-proteins and signaling pathways in myocytes that may mediate the potential differences in cardiac physiology (18,19) as reviewed here.

α1-AR-mediated cardiac hypertrophy

Early studies (20,21) indicated that incubation of myocytes with catecholamines causes cellular hypertrophy by activation of α1-ARs. Subsequent reports established that the α1-AR-stimulated hypertrophy in myocytes progresses through a series of genetic events with induction of immediate–early genes followed by expression of embryonic genes, and increased contractile proteins (22). The end result of these changes in gene expression is an increased size of the myocyte. Cardiac hypertrophy initially has beneficial effects in terms of muscular economy by normalizing wall stress (i.e. adaptive hypertrophy). However, several studies have demonstrated that chronic hypertrophy is associated with a significant increase in the risk of heart failure, ischemic heart disease, and apoptosis (i.e. maladaptive hypertrophy; reviewed in ref. 23). We believed that the differential outcomes of hypertrophy can depend upon the α1-AR subtype, with the α1A-AR mediating adaptive and compensatory hypertrophy, whereas the α1B-AR mediates hypertrophy that is maladaptive and cardiac damaging.

Most cellular studies indicated that the α1A-AR subtype is the mediator of hypertrophy in neonatal myocytes (24,25). Knowlton et al. (25) showed that stimulation of the α1A-AR subtype in neonatal myocytes caused phosphoinositide hydrolysis and was responsible for cardiac hypertrophy, whereas stimulation of the α1B-AR subtype did not mediate hypertrophy. Similar results were obtained by Autelitano and Woodcock (24) using subtype-selective agonists. In vivo, an α1A-AR transgenic mouse model with constitutively active mutations (CAM) in the receptor and under the control of the native promoter to achieve systemic expression demonstrated cardiac hypertrophy independent of changes in blood pressure (manuscript in preparation), corroborating earlier cellular studies. This hypertrophy appears to be adaptive as these mice are protected against ischemia (26). In contrast, myocyte-targeted wild type (WT) α1A-AR do not display hypertrophy (27), even with vast amounts of receptor over expression. This same mouse model limits postinfarct ventricular remodeling and dysfunction and improves survival due to heart failure after myocardial infarction and thus appears to be cardiac adaptive (28). However, long-term effects of this heightened contractility eventually become pathological (29). Discrepancies between the mouse models could be due to the CAM receptor coupling promiscuously to signaling pathways not associated with a WT receptor. On the other hand, the CAM α1A-AR mouse also expresses the CAM receptor in cells other than the myocyte, and displays high secreted serum levels of interleukin (IL)-6 (manuscript in preparation), which may promote adaptive cardiac hypertrophy through the gp130 and STAT3 pathways (3033).

In contrast, myocyte-targeted CAM α1B-AR mice were shown to have hypertrophy ex vivo, although mild (34), and displayed a hastened time to heart failure with pressure overload (35). However, cardiac overexpression of the WT α1B-AR, while displaying ventricular dysfunction, did not display hypertrophy (36). However, this same mouse displayed elevated activation of signaling pathways associated with cardiac hypertrophy, such as calcineurin activity (37). A similar but different myocytetargeted WT α1B-AR mouse model (38) did not have basal hypertrophy, but developed a severe maladaptive hypertrophy with cardiac abnormalities when subjected to a 14-day treatment of phenylephrine (39). Both CAM and WT α1B-AR mice under the control of the endogenous promoter demonstrated both cardiac hypertrophy and cardiac dysfunction (40). Therefore, both overexpressed α1B-AR mouse models suggest a maladaptive response to cardiac hypertrophy. Consistent with the observation that the α1B-AR mediates cardiac hypertrophy is that the α1B-AR knockout (KO) mouse do not display NE-mediated hypertrophy (41).

α1-ARs in heart failure

Chronic heart failure is associated with prolonged stimulation of the adrenergic and sympathetic nervous system and increased plasma levels of catecholamines, resulting in β1-AR down-regulation and myocardial apoptosis (4244). This increased sympathetic activity is first initiated as an adaptive process and the heart hypertrophies to compensate for the decreased cardiac contractility. However, sustained cardiac contractility cannot be maintained indefinitely and this compensatory process becomes maladaptive, contributing significantly to disease progression by wall thinning, dilation, and finally heart failure.

Previous studies during the 1980–1990s on the roles of α1-ARs in human heart failure is controversial. Some studies indicated they were protective, whereas other studies indicated that α1-AR activation was detrimental that progressed heart failure (45). However many of these studies were performed before we knew the existence of the α1-AR subtypes and had the tools to define them, so these disparate reports or findings could be due to subtype-specific signaling. α1-AR antagonists were initially thought to be useful in treating heart failure due to decreasing sympathetic overload. However, in the Antihypertensive and Lipid-Lowering Treatment to Prevent Heart Attack Trial, the use of a nonselective α1-AR antagonist increased the risk of heart failure and mortality (46). In contrast, carvedilol, an antagonist of α1- and β-ARs but with higher affinity for the α1B-AR subtype (47) (and thus promoting α1A-AR signaling and inotropism), provided an effective treatment for chronic heart failure, suggesting that α1-AR subtypes may contribute to these differential effects on heart failure.

α1-ARs can direct either positive or negative inotropism depending upon the species or tissue preparation studied (48,49). One study indicated that in mouse trabecular tissue, the inotropism changes from positive to negative when analyzing right versus left trabeculae (50). However, this effect may also be dependent upon the α1-AR subtype. In rats, the right ventricular (RV) inotropic response to α1-ARs was switched from negative to positive in heart failure, through a pathway involving increased myofilament calcium sensitivity. This study suggested that increased α1-AR inotropic responses in the RV myocardium may be adaptive in heart failure by helping the failing RV respond to increased pulmonary pressures (51). In corroboration with this study, α1A-AR density increased compared with non-failing hearts when specifically analyzed in the failing human RV although overall α1-AR density did not change (52). In addition, mechanical unloading of the failing human heart with a left ventricular-assisted device significantly increased α1-AR density than before explantation (53). α1AB double KO mice had increased morbidity due to heart failure and a maladaptive cardiac phenotype from pressure overload (54), opposite from α1A-AR-mediated cardiac protection. The hypercontractile myocyte-targeted WT α1A-AR transgenic mouse also protected against pressure overload (55). Therefore, all of these studies suggest that α1A-AR supplementation or stimulation may be cardioprotective.

In Langendorff ex vivo heart studies, CAM α1B-AR mice under the control of the endogenous promoter were found to have an impaired cardiac inotropy when stimulated with phenylephrine (56). Similar results were obtained when adult myocytes were isolated from this mouse model. This mouse also displayed impaired cardiac relaxation times and decreased cardiac output (40). Myocyte-targeted CAM α1B-AR mice displayed hastened time to heart failure with pressure overload (35). In addition, myocyte-targeted WT α1B-AR mice also showed development of dilated cardiomyopathy (57). These mice had systolic dysfunction and progressed to heart failure and died prematurely. Recent evidence also suggests that these mice regulate pathological cardiac arrhythmias due to the down-regulation of potassium channels (58) and may also contribute to their premature death. The α1B-AR KO mice did not display any difference from controls when subjected to pressure overload (59). These results suggest that the α1B-AR may hasten cardiac pathological and maladaptive conditions such as cardiac arrhythmias and heart failure due to pressure overload.

Cardiac apoptosis

Long-term exposure to catecholamines is toxic to cardiac myocytes (60), a process mediated primarily through β-AR stimulation (61). In contrast, while stimulation of α1-ARs does not appear to mediate cardiac myocyte apoptosis, it may promote protection against cell death. In neonatal rat myocytes, α1-AR stimulation inhibited apoptosis caused by cAMP (44), and was abolished by a MEK inhibitor suggesting a role for ERK1/2. Inhibition of protein phosphatase 1 by okadaic acid, which induces apoptosis in rat neonatal myocytes, was blocked by α1-AR stimulation (62). Ischemic preconditioning, which we demonstrated to be regulated through the α1A-AR subtype (26), limited apoptotic cell death through an increased bclx/bax ratio in the rabbit heart when stimulated by phenylephrine (63). In more recent studies, the α1A-AR subtype was shown to protect myocytes from apoptosis also through an ERK-mediated mechanism (64). These results suggest that α1A-ARs may mediate survival antiapoptotic signaling in cardiac myocytes.

As it appears that the α1A-AR promotes anti-apoptotic signaling, the α1B-AR may promote apoptosis. However, myocyte-targeted WT α1B-AR mice do not show increased apoptosis, whereas overexpression of Gq results in hypertrophy and/or apoptosis depending on the level of expression (65). Since α1-ARs in adult mouse myocytes may not activate the Gq-phospholipase C pathway (66), this may explain why α1B-AR mice do not promote apoptosis. There is also evidence that the α1B-AR may be coupled to Gi in the heart (67,68).

α1-AR ischemic preconditioning

Ischemic preconditioning is an endogenous protective mechanism in which brief episodes of cardiac ischemia protect the heart from damage caused by a subsequent episode of prolonged ischemia. Ischemia and hypoxia have been shown to increase the number of myocardial α1-ARs in both acute and chronic conditions (69). Ischemic preconditioning is mimicked by the α1-AR agonist phenylephrine and blocked by α1-AR antagonists (70,71). However, studies to determine which α1-AR subtype mediates this effect have produced conflicting results which were likely due to the use of nonselective ligands. Several studies looking at the early phase of preconditioning using 5-methylurapidil and chloroethyl clonidine have concluded that ischemic preconditioning is mediated by the α1B-AR, but not the α1A-AR (7274). However, subsequently, it was shown that chloroethyl clonidine was not differentially selective for α1-AR subtypes (75).

In mouse models, the CAM α1A-AR mice under the control of the endogenous promoter demonstrated inherent preconditioning (26). However, CAM α1B-AR mice, which utilize the endogenous promoter, did not display not only protection but also any worsening of the ischemic damage (26). In agreement, the myocyte-targeted WT α1A-AR limited postinfarct ventricular remodeling and dysfunction (28) and suppressed ischemia-induced IP3 generation (76). These studies are consistent with the study of Gao et al. (77), who found that heart-targeted CAM α1B-AR mice were not protected from ischemic injury. Therefore, data using transgenic mice provide clear evidence that ischemic preconditioning is mediated by α1A-ARs but not α1B-ARs.

α1-AR cardiac protective signaling pathways

The low-molecular-weight GTPase RhoA has been shown to be involved in the α1-AR growth pathways in myocytes (78,79). In addition, the AKAP-Lbc, an A-kinase-anchoring protein with an intrinsic Rho-specific guanine nucleotide exchange factor activity, was shown to be critical for activating RhoA and transducing hypertrophic signals downstream of α1-ARs (80). However, these pathways have not been shown to be either adaptive or maladaptive. Other pathways associated with α1-AR-mediated hypertrophy are Ras (81) and Gαq (78). While α1-ARs couple to Gαq, which can mediate hypertrophic signaling, Gq-mediated hypertrophy ultimately leads to its decompensation and apoptosis as evidenced by the Gq transgenic mouse model (65,82). Gq overexpressing hearts also had a lack of ERK activation (83). In contrast, Ras-mediated events convey characteristic features of hypertrophy but with normal contractile functions (84,85). α1A-AR-mediated ERK anti-apoptotic effects will be downstream of Ras and ERK can also be pro-hypertrophic (86,87). Thus, Ras/ERK signaling appears to be adaptive. Therefore, we suggest that α1A-AR-mediated adaptive hypertrophy may be mediated through Ras/ERK downstream signals, whereas the α1B-AR-mediated maladaptive hypertrophy is mediated through Gαq.

In addition to ERK signaling in α1-AR mediation of cardiac apoptosis and protection (64), the transcriptional coactivator p300 or histone deacetylase activity was also shown to mediate hypertrophy and protection from cell death by α1-ARs due to ischemia and reperfusion (8890). Inhibition of apoptosis by α1-ARs could also be part of the protective mechanism in delayed preconditioning (63). However, α1-ARs were also found to activate the forkhead box family of proapoptotic transcription factors (FOXO1) in cardiomyocytes (91) via nuclear translocation. Therefore, this pathway is associated with potential role of α1-ARs in mediating apoptosis in the heart and its maladaptive processes that we speculate are associated with α1B-AR-mediated signaling.

In addition, the cardioprotective pathways of α1-AR-mediated preconditioning have been linked to PKCε involvement (26,92,93). Preferential activation of PKCε, but not PKCα, has also been observed in phenylephrine-treated isolated neonatal and adult rat myocytes (94). PKC isoforms are also predicted to act as molecular switches that regulate the balance of signaling via proapoptotic JNK and anti-apoptotic PDK-1/AKT α1-AR-mediated pathways in myocytes (95). α1-AR-mediated late phase of ischemic protection has also been linked to heat shock protein (HSP) 70 (96) and α1-AR signaling mediates induction of the heat shock 70 promoter (97). In addition, another HSP27 and PKCε were both associated with ischemic protection via α1-AR signaling in myocytes (98). Interestingly, one study suggests that the PKC mediation of ischemic preconditioning is through factors secreted from the heart (99). Therefore, at least part of the cardioprotection mediated by the α1A-AR may be through PKCε signaling pathways.

Cardiac summary

Heart failure and other pathological cardiac effects are increased by sympathetic overdrive and provided the rationale for the successful use of β-blockers to treat heart failure (100). However, the adverse effects observed for α1-AR antagonists in heart failure (46) might be explained through cardioprotective pathways mediated by stimulation of the α1A-AR subtype summarized here. Therapeutic strategies to only activate the α1A-AR subtype during infarction or in the setting of heart failure may be a viable treatment option.

Neurophysiology

The noradrenergic (NA) system in the mammalian central nervous system (CNS) originates primarily in the locus coeruleus. From this nucleus in the brain stem, a highly divergent and diffuse projection of NE-containing axons radiate throughout the CNS innervating the spinal cord, cerebellum, thalamus, amygdala, hippocampus, and cortex (reviewed in ref. 101). The wide distribution of afferents is congruent with a global regulatory role in CNS function. Indeed, the central NE system has been shown to regulate a number of behavioral states including sleep and arousal, cognitive functions such as learning and memory, affective states such as anxiety and depression, changes in neuroplasticity, embryonic brain development, and adult neurogenesis (reviewed in ref. 102). The central NE system has also been implicated in a number of neurological disorders including Alzheimer’s disease, attention-deficit/hyperactivity (ADHD), depression, epilepsy, mania, Parkinson’s disease, posttraumatic stress disorder (PTSD), and schizophrenia (reviewed in ref. 103). Specificity in the central NE system arises from its diversity of nine AR subtypes with discrete expression patterns. Although all AR subtypes occur in the brain, the α1-ARs may be the most abundant. α1-ARs have a role in the numerous physiological states and neurological disorders that are associated with the central NE system.

Expression of α1-ARs in brain

The specific localization of the α1-AR subtypes in the brain has been hindered by the lack of subtype-selective antibodies and ligands. Early receptor binding or autoradiography studies indicated that α1-ARs are located throughout the brain (104). Later, in situ hybridization studies suggested that the α1-AR subtypes may be differentially expressed in the brain (105); however, these studies, while more accurate, are not quantitative due to mRNA transportation. Unfortunately, the lack of high affinity antibodies to the α1-AR subtypes has prevented reliable immunohistochemistry findings (106).

The pharmacology of the α1-ARs is very similar between human and rodents; however, the distribution is different (107). While there are comparable densities of α1-ARs in the thalamus and cortex of rodents and humans, the density of α1-ARs is much higher in the hippocampus of humans compared with rodents, especially in the CA3 region and dentate gyrus (107). Moreover, α1A- and α1B-ARs are differentially distributed in the human hippocampus, with the α1A-AR concentrated in the CA3 and the α1B-AR concentrated in the dentate gyrus (108).

To circumvent the lack of selective antibodies and ligands to determine the cellular localization of the α1-ARs, transgenic mice were developed, which overexpress either the α1A- or α1B-ARs, fused with an enhanced green fluorescent protein (EGFP). Using this transgenic tagged-GPCR approach, it was determined that α1A- and α1B-ARs exhibit a similar expression pattern in the CNS (109,110). Both receptors are expressed in the amygdala, cerebellum, cerebral cortex, hippocampus, hypothalamus, midbrain, pontine olivary nuclei, trigeminal nuclei, and spinal cord. Both receptors were also found in the same cell types including neurons, GABAergic interneurons, and NG2 oligodendrocyte progenitors. Interestingly, neither α1A- nor α1B-ARs were found in astrocytes or cerebral vascular smooth muscle, cells previously thought to express α1-ARs. Using transgenic KO mice for the individual α1-AR subtypes, the distribution of α1-ARs in the brain has been determined to be ~55% α1A (111), 35% α1B (112), and 10% α1D (113).

Neural actions of α1-ARs

Identification of the effects mediated by α1-AR subtype has also been hindered by the lack of subtype-selective ligands. The α1-ARs are generally stimulatory and found on postsynaptic cell bodies. α1-ARs can increase the excitation mediated by glutamate or acetylcholine (114) and prime excitatory synapses (115). Furthermore, they have been found to directly enhance neurotransmitter release from presynaptic terminals (116), as well as to modulate GABAergic (117) and glutamatergic inputs (118). Their stimulatory actions have often been attributed to a decrease in cellular resting conductance (119,120), but may also involve an increase in calcium. α1-ARs may also affect many CNS functions via non-neuronal mechanisms as they are also expressed in glia. α1-AR activation has been found to increase calcium release in Bergmann glial cells (121).

α1-ARs in neurogenesis

Recent evidence suggests that α1-ARs are involved in the neurogenic effects of NE. Increasing brain NE levels has been found to enhance the proliferation of neural progenitor cells (NPCs) (122), while depleting NE decreases NPC proliferation in vivo (123). Stimulating α1-ARs has also been shown to induce the proliferation and migration of embryonic NPCs in vitro (124,125). Using transgenic EGFP-tagged and CAM mouse models, we found that α1A-AR are expressed in neural stem cells (NSCs) and/or transient amplifying progenitors (TAPs) in vivo and that the chronic stimulation of these receptors increases neurogenesis (126). Moreover, we have found that treating adult normal (nontransgenic) mice with the mildly selective α1A-AR agonist, cirazoline, also increases neurogenesis. Interestingly, although we found that α1B - ARs also appear to be expressed on NSCs, chronic activation of these receptors did not increase neurogenesis. Indeed, CAM α1B-AR mice display an age-dependent neurodegeneration (127). Our finding that stimulating α1A-AR increases neurogenesis may have significant implications for many normal and pathological neurological processes.

α1-ARs in locomotion

Although the dopaminergic-striatal pathways are considered the primary mechanism for controlling locomotion and motor activity in the CNS, α1-ARs activated by NE may also be involved (128,129). Postsynaptic excitatory α1-ARs have been shown to modulate midbrain dopamine cell activity (130). α1-ARs have also been found to increase the tonic firing of principal neurons in the substantia nigra pars reticulate (131). The output of the basal ganglia and motorrelated behaviors may be significantly impacted by this increase in excitability. In vivo pharmacological evidence suggests that α1B-ARs are involved in the motor activity and spontaneous movement (132). Consistent with this finding, α1B-AR KO mice do not display fear-motivated exploratory behavior (133). Mice lacking α1B-AR also exhibit decreased psychostimulant-induced locomotor hyperactivity (134) that correlated with reduced psychostimulant-induced dopamine release in the nucleus accumbens (135). In contrast, the α1A-AR may mediate the facilitation of α-motor neuron activity in the rat spinal cord (136). In summary, the α1B-AR may be involved in controlling locomotion and motor activity, which is relevant to locomotor disorders.

α1-ARs in neurodegeneration

In contrast to long-term α1A-AR activation, chronic α1B - ARs stimulation appears to cause an age-progressive apoptotic neurodegeneration (127). Transgenic mice that systemically overexpress the α1B-AR display an age-dependent neurological disorder similar to multiple system atrophy (127) characterized by autonomic dysfunction, Parkinsonism, and ataxia. Histological examination reveals that α1B-AR overexpression causes a synucleinopathy marked by the aggregation/formation of α-synuclein inclusion bodies and interneuron loss in the cerebellum and hippocampus, as well as degeneration of spinal cord cell columns (137). Consistent with the finding that α1B-AR activation induces a Parkinson-like syndrome, mice lacking α1B-ARs (KO) do not show this neurodegenerative phenotype. In fact, α1B-AR KO mice appear to live a normal, albeit longer life than the α1A-AR KO mice (138).

α1-ARs in cognitive function and neuroplasticity

The function of α1-ARs in learning and memory has not been clearly defined and is controversial. Most in vivo studies suggest that activating α1-ARs improves memory while blocking α1-ARs impairs cognition (139141). Other studies have reported that α1-ARs inhibit memory (142144). These differences may be due to species-specific differences or to differential regulation by α1-AR subtypes. Recently, we found that chronic α1A-AR stimulation improves cognitive function (145; manuscript submitted), whereas long-term α1B-AR activation impaired learning and memory (unpublished findings). Furthermore, we found that treating normal mice with a selective agonist increased both spatial and declarative memory processes, while α1A-AR KO mice have diminished cognitive function. The mechanism for these actions is not known for certain. However, neurogenesis has been shown to enhance certain forms of learning and memory, while neurodegeneration generally impairs cognitive function.

α1-AR dysfunction is implicated in the pathogenesis of Alzheimer’s disease (AD). For example, polymorphisms in the α1A-AR are associated with AD susceptibility (146). Brain tissue from aged transgenic AD mice displays increased α1-ARs in cerebral cortices (147). Increasing α1-AR activation in the medial prefrontal cortex improves cognitive function of rats (148). In AD patients, α1-AR binding is significantly reduced in the prefrontal cortex (149,150). Furthermore, α1A-AR mRNA expression is decreased in the prefrontal cortex in patients with dementia (151). These results suggest that increasing α1A-AR activity may delay or alleviate AD symptoms.

Given that α1-ARs affect cognitive function, it is not surprising then that numerous studies have shown that α1-ARs also affect plasticity in the hippocampus, a cortical structure critical for learning and memory. NE through α1-AR activation has been shown to promote hippocampal long-term potentiation (LTP), an important synaptic mechanism underlying learning (140,152,153). α1-ARs may also play a role in long-term depression (LTD) in the hippocampus (154). The role of the various α1-AR subtypes in neuroplasticity is unknown.

α1-ARs in depression and anxiety

The involvement of NE in depression is well-established. Although several studies have suggested that α1-ARs are involved in the antidepressant effects of NE (155), the role of individual α1-AR subtypes in mood is not well-defined. Recently, using transgenic mice, we showed that chronic α1A-AR was associated with a significant decrease in depression-like behavior, whereas chronic α1B-AR stimulation was prodepressant (156,157). Subsequently, we have shown that chronic α1A-AR activation reduces anxiety and obsessive–compulsive-like behavior (158; manuscript submitted). The mechanism for these actions is not known. However, since considerable evidence links depression to neurogenesis, enhanced α1A-AR-stimulated neurogenesis deserves serious attention as the mechanism. Increased α1A-AR stimulated GABA release in the amygdala and other cortical structures could be involved in the anti-anxiety actions (159).

α1A-ARs in epilepsy

NE has long been known to be potently anti- epileptogenic (see reviews in refs. 160,161). Numerous studies using different models of epilepsy have shown that the anticonvulsant effects are mediated in part by α1-ARs (162168). The underlying mechanism has not been clearly established. However, the activation of α1-ARs has been shown to enhance tonic GABA-mediated inhibition in several regions of the brain including the piriform cortex (169), amygdala (170), medial septum (171), hippocampus (120), and frontal cortex (172). The increase in GABA release resulted from a direct α1-AR-mediated decrease in a potassium conductance resulting in a depolarization of inhibitory GABAergic interneurons (120,172). This increase in GABA-mediated inhibitory tone, in turn, reduces epileptiform bursting in the hippocampus (173). This physiological response appears to be mediated by α1A-ARs in the hippocampus (174,175) and amygdala (159), and may be impaired by stress (170), which reduces α1-AR numbers.

The α1-AR subtypes have both positive and negative roles in regulating seizure activity. Both CAM α1A-AR and WT α1A-AR mice under the control of the endogenous promoter demonstrate an antiepileptic phenotype with increased seizure thresholds to flurothyl, a chemoconvulsant (176; manuscript in preparation). In contrast, the CAM α1B-AR mice exhibit decreased seizure thresholds to flurothyl. Moreover, CAM α1B-AR mice develop a grand mal seizure disorder that appears to be a multifocal epilepsy (177). The mechanism underlying the seizures in the CAM α1B-AR mice is not known, but may be due to NMDA/GABAA receptor dysregulation and apoptosis (178). Consistent with these findings, we have found that α1A-AR KO mice often display spontaneous seizures, whereas α1B-AR KO mice do not show this phenotype. Indeed, the lack of α1B-ARs appears to confer some resistance to the neurotoxicity produced by seizures (179). Furthermore, α1B-AR KO mice do not appear to develop the neurodegeneration associated with prolonged and severe seizures (179). We also have evidence that chronic activation of the α1A-AR, but not the α1B-AR, protects the brain from apoptosis (178). Using transgenic mice, we found that the hippocampus expresses the highest amount of the α1A-AR compared with other areas in the brain (110). We also discovered that both the overexpressed WT α1A-AR and the CAM α1A-AR mice have an increased density of hippocampal interneurons (180) and demonstrate protection from anoxia/trauma and hyperexcitability (145). Taken together, these results suggest that α1A-AR activation is not only anti-epileptogenic, but neuroprotective.

α1-AR subtypes in mental illness

Heightened or excessive NE activity has been associated with schizophrenia, mania, and PTSD (reviewed in 142). The α1-AR subtypes may have both positive and negative roles in these mental illnesses. α1-AR antagonism has been recognized as a potential mechanism of antipsychotic action since the discovery that many antipsychotics block α1-ARs (181). For example, the atypical antipsychotic clozapine has a high affinity toward these receptors (182). Many sources cite the antagonism of the α1A-AR subtype as mediating this antipsychotic action though the data may not support this claim sufficiently, as the conclusions were based on studies performed in the peripheral nervous system while studying side effects (183) or not actually focused on the α1-AR subtypes (184). The binding affinities of clozapine for α1A- and α1B -ARs are not significantly different (185,186); however, the observed α1-AR up-regulation that occurs with clozapine treatment is mainly in areas of high α1B-AR expression (187). In addition, the atypical antipsychotic risperidone has 120-fold antagonist selectivity for the α1B-AR in the hippocampus (185). Furthermore, risperidone increases survival of hippocampal NSCs in a ketamine-induced model of schizophrenia (188). These results suggest that α1B-ARs are involved in schizophrenia and antagonism of the α1B-AR, but not the α1A-AR, may be mediating part of atypical antipsychotic action. Interestingly, single nucleotide polymorphisms in the α1A-AR promoter region have been found in an isolated population with schizophrenia (189).

Antagonism of the α1-ARs is also used to treat the nightmares and sleep disturbances associated with PTSD. Recent evidence suggests that excessive PKC activation in the prefrontal cortex may be involved in PTSD as well as bipolar disorder (190). Antagonists and/or antipsychotics that block α1-AR-mediated PKC activity have shown some benefit in treating these disorders. The level of RGS4, a small protein that inhibits PKC activity, is reduced in schizophrenia (191). RGS4 has also been linked genetically to schizophrenia and bipolar disorder (192195). Interestingly, RGS4 is suggested to interact with α1B-ARs, whereas RGS2 proteins will directly interact with α1A-ARs, but not with α1B-ARs or α1D-ARs (196). Taken together, these findings suggest that α1A-ARs and α1B-ARs may play different roles in these mental illnesses. Studies are currently being conducted to determine the effects of chronic α1A-AR and α1B-AR activation on these mental illnesses.

Neuro summary

Although the role of α1-ARs in the CNS has been the least understood historically, recent evidence suggests that many of their actions are mediated differentially by α1A - ARs and α1B-ARs. Chronic α1A-AR stimulation increases neurogenesis, enhances learning and memory, and improves mood. It may also protect the brain from anoxia and traumatic injury, seizures, and age-dependent neurodegeneration. Therapeutic strategies to selectively activate the α1A-AR subtype and/or only block α1B-ARs may be neuroprotective, and therefore may be useful for treating a number of neurological disorders.

Epilogue

Our latest research indicates that chronic activation of the α1A-AR, but not the α1B-AR, increases life span (138; manuscript submitted). Using our transgenic α1-AR and KO mice, we found that both the CAM α1A-AR and overexpressed α1A-AR EGFP mice live significantly longer (>10%) than their normal littermates. Unlike many other mouse models of longevity, these mice are of normal body size and mobility. In contrast, the CAM α1B-AR and overexpressed α1B-AR EGFP mice have a shorter life span than their normal littermates. Furthermore, we found that α1A-AR KO mice do not live as long as α1B-AR KO mice. Taken together, these results suggest that the cardiac and neuroprotection afforded by the α1A-AR translates into increased longevity.

Footnotes

Declaration of interest

The authors report no declarations of interest.

References

  • 1.Graham RM, Lanier SM. Identification and characterization of alpha- adrenergic receptors chapter 50. In: Fozzard HA, et al., editors. The Heart, and Cardiovascular System. New York: Raven Press; 1986. pp. 1059–1095. [Google Scholar]
  • 2.Ahlquist RP. A study of the adrenotropic receptors. Am J Physiol. 1948;153:586–600. doi: 10.1152/ajplegacy.1948.153.3.586. [DOI] [PubMed] [Google Scholar]
  • 3.Morrow AL, Creese I. Characterization of alpha 1-adrenergic receptor subtypes in rat brain: a reevaluation of [3H]WB4104 and [3H]prazosin binding. Mol Pharmacol. 1986;29:321–330. [PubMed] [Google Scholar]
  • 4.Cotecchia S, Schwinn DA, Randall RR, Lefkowitz RJ, Caron MG, Kobilka BK. Molecular cloning and expression of the cDNA for the hamster alpha 1-adrenergic receptor. Proc Natl Acad Sci USA. 1988;85:7159–7163. doi: 10.1073/pnas.85.19.7159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Schwinn DA, Lomasney JW, Lorenz W, Szklut PJ, Fremeau RT, Jr, Yang-Feng TL, Caron MG, Lefkowitz RJ, Cotecchia S. Molecular cloning and expression of the cDNA for a novel alpha 1-adrenergic receptor subtype. J Biol Chem. 1990;265:8183–8189. [PubMed] [Google Scholar]
  • 6.Perez DM, Piascik MT, Malik N, Gaivin R, Graham RM. Cloning, expression, and tissue distribution of the rat homolog of the bovine alpha 1C-adrenergic receptor provide evidence for its classification as the alpha 1A subtype. Mol Pharmacol. 1994;46:823–831. [PubMed] [Google Scholar]
  • 7.Laz TM, Forray C, Smith KE, Bard JA, Vaysse PJ, Branchek TA, Weinshank RL. The rat homologue of the bovine alpha 1c-adrenergic receptor shows the pharmacological properties of the classical alpha 1A subtype. Mol Pharmacol. 1994;46:414–422. [PubMed] [Google Scholar]
  • 8.Lomasney JW, Cotecchia S, Lorenz W, Leung WY, Schwinn DA, Yang-Feng TL, Brownstein M, Lefkowitz RJ, Caron MG. 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. 1991;266:6365–6369. [PubMed] [Google Scholar]
  • 9.Perez DM, Piascik MT, Graham RM. Solution-phase library screening for the identification of rare clones: isolation of an alpha 1D-adrenergic receptor cDNA. Mol Pharmacol. 1991;40:876–883. [PubMed] [Google Scholar]
  • 10.Hieble JP, Bylund DB, Clarke DE, Eikenburg DC, Langer SZ, Lefkowitz RJ, Minneman KP, Ruffolo RR., Jr International Union of Pharmacology. X. Recommendation for nomenclature of alpha 1-adrenoceptors: consensus update. Pharmacol Rev. 1995;47:267–270. [PubMed] [Google Scholar]
  • 11.Rokosh DG, Bailey BA, Stewart AF, Karns LR, Long CS, Simpson PC. Distribution of alpha 1C-adrenergic receptor mRNA in adult rat tissues by RNase protection assay and comparison with alpha 1B and alpha 1D. Biochem Biophys Res Commun. 1994;200:1177–1184. doi: 10.1006/bbrc.1994.1575. [DOI] [PubMed] [Google Scholar]
  • 12.Scofield MA, Liu F, Abel PW, Jeffries WB. Quantification of steady state expression of mRNA for alpha-1 adrenergic receptor subtypes using reverse transcription and a competitive polymerase chain reaction. J Pharmacol Exp Ther. 1995;275:1035–1042. [PubMed] [Google Scholar]
  • 13.Bürger A, Benicke M, Deten A, Zimmer HG. Catecholamines stimulate interleukin-6 synthesis in rat cardiac fibroblasts. Am J Physiol Heart Circ Physiol. 2001;281:H14–H21. doi: 10.1152/ajpheart.2001.281.1.H14. [DOI] [PubMed] [Google Scholar]
  • 14.Lai KB, Sanderson JE, Yu CM. Suppression of collagen production in norepinephrine stimulated cardiac fibroblasts culture: differential effect of alpha and beta-adrenoreceptor antagonism. Cardiovasc Drugs Ther. 2009;23:271–280. doi: 10.1007/s10557-009-6183-6. [DOI] [PubMed] [Google Scholar]
  • 15.Böhm M, Diet F, Feiler G, Kemkes B, Erdmann E. Alpha-adrenoceptors and alpha-adrenoceptor-mediated positive inotropic effects in failing human myocardium. J Cardiovasc Pharmacol. 1988;12:357–364. doi: 10.1097/00005344-198809000-00015. [DOI] [PubMed] [Google Scholar]
  • 16.Landzberg JS, Parker JD, Gauthier DF, Colucci WS. Effects of myocardial alpha 1-adrenergic receptor stimulation and blockade on contractility in humans. Circulation. 1991;84:1608–1614. doi: 10.1161/01.cir.84.4.1608. [DOI] [PubMed] [Google Scholar]
  • 17.Skomedal T, Borthne K, Aass H, Geiran O, Osnes JB. Comparison between alpha-1 adrenoceptor-mediated and beta adrenoceptor-mediated inotropic components elicited by norepinephrine in failing human ventricular muscle. J Pharmacol Exp Ther. 1997;280:721–729. [PubMed] [Google Scholar]
  • 18.Wang H, Yang B, Zhang Y, Han H, Wang J, Shi H, Wang Z. Different subtypes of alpha1-adrenoceptor modulate different K+ currents via different signaling pathways in canine ventricular myocytes. J Biol Chem. 2001;276:40811–40816. doi: 10.1074/jbc.M105572200. [DOI] [PubMed] [Google Scholar]
  • 19.O-Uchi J, Sasaki H, Morimoto S, Kusakari Y, Shinji H, Obata T, Hongo K, Komukai K, Kurihara S. Interaction of α1-adrenoceptor subtypes with different G proteins induces opposite effects on cardiac L-type Ca2+ channel. Circ Res. 2008;102:1378–1388. doi: 10.1161/CIRCRESAHA.107.167734. [DOI] [PubMed] [Google Scholar]
  • 20.Simpson P. Norepinephrine-stimulated hypertrophy of cultured rat myocardial cells is an alpha 1 adrenergic response. J Clin Invest. 1983;72:732–738. doi: 10.1172/JCI111023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Meidell RS, Sen A, Henderson SA, Slahetka MF, Chien KR. Alpha 1-adrenergic stimulation of rat myocardial cells increases protein synthesis. Am J Physiol. 1986;251:H1076–H1084. doi: 10.1152/ajpheart.1986.251.5.H1076. [DOI] [PubMed] [Google Scholar]
  • 22.Long CS, Kariya K, Karns L, Simpson PC. Sympathetic modulation of the cardiac myocyte phenotype: studies with a cell-culture model of myocardial hypertrophy. Basic Res Cardiol. 1992;87(Suppl 2):19–31. doi: 10.1007/978-3-642-72477-0_3. [DOI] [PubMed] [Google Scholar]
  • 23.Selvetella G, Hirsch E, Notte A, Tarone G, Lembo G. Adaptive and maladaptive hypertrophic pathways: points of convergence and divergence. Cardiovasc Res. 2004;63:373–380. doi: 10.1016/j.cardiores.2004.04.031. [DOI] [PubMed] [Google Scholar]
  • 24.Autelitano DJ, Woodcock EA. Selective activation of alpha1A-adrenergic receptors in neonatal cardiac myocytes is sufficient to cause hypertrophy and differential regulation of alpha1- adrenergic receptor subtype mRNAs. J Mol Cell Cardiol. 1998;30:1515–1523. doi: 10.1006/jmcc.1998.0717. [DOI] [PubMed] [Google Scholar]
  • 25.Knowlton KU, Michel MC, Itani M, Shubeita HE, Ishihara K, Brown JH, Chien KR. The alpha 1A-adrenergic receptor subtype mediates biochemical, molecular, and morphologic features of cultured myocardial cell hypertrophy. J Biol Chem. 1993;268:15374–15380. [PubMed] [Google Scholar]
  • 26.Rorabaugh BR, Ross SA, Gaivin RJ, Papay RS, McCune DF, Simpson PC, Perez DM. alpha1A— but not alpha1B—adrenergic receptors precondition the ischemic heart by a staurosporinesensitive, chelerythrine-insensitive mechanism. Cardiovasc Res. 2005;65:436–445. doi: 10.1016/j.cardiores.2004.10.009. [DOI] [PubMed] [Google Scholar]
  • 27.Lin F, Owens WA, Chen S, Stevens ME, Kesteven S, Arthur JF, Woodcock EA, Feneley MP, Graham RM. Targeted alpha(1A)-adrenergic receptor overexpression induces enhanced cardiac contractility but not hypertrophy. Circ Res. 2001;89:343–350. doi: 10.1161/hh1601.095912. [DOI] [PubMed] [Google Scholar]
  • 28.Du XJ, Gao XM, Kiriazis H, Moore XL, Ming Z, Su Y, Finch AM, Hannan RA, Dart AM, Graham RM. Transgenic alpha1A-adrenergic activation limits post-infarct ventricular remodeling and dysfunction and improves survival. Cardiovasc Res. 2006;71:735–743. doi: 10.1016/j.cardiores.2006.06.015. [DOI] [PubMed] [Google Scholar]
  • 29.Chaulet H, Lin F, Guo J, Owens WA, Michalicek J, Kesteven SH, Guan Z, Prall OW, Mearns BM, Feneley MP, Steinberg SF, Graham RM. Sustained augmentation of cardiac alpha1A-adrenergic drive results in pathological remodeling with contractile dysfunction, progressive fibrosis and reactivation of matricellular protein genes. J Mol Cell Cardiol. 2006;40:540–552. doi: 10.1016/j.yjmcc.2006.01.015. [DOI] [PubMed] [Google Scholar]
  • 30.Kunisada K, Tone E, Fujio Y, Matsui H, Yamauchi-Takihara K, Kishimoto T. Activation of gp130 transduces hypertrophic signals via STAT3 in cardiac myocytes. Circulation. 1998;98:346–352. doi: 10.1161/01.cir.98.4.346. [DOI] [PubMed] [Google Scholar]
  • 31.Hirota H, Chen J, Betz UA, Rajewsky K, Gu Y, Ross J, Jr, Müller W, Chien KR. Loss of a gp130 cardiac muscle cell survival pathway is a critical event in the onset of heart failure during biomechanical stress. Cell. 1999;97:189–198. doi: 10.1016/s0092-8674(00)80729-1. [DOI] [PubMed] [Google Scholar]
  • 32.Kunisada K, Negoro S, Tone E, Funamoto M, Osugi T, Yamada S, Okabe M, Kishimoto T, Yamauchi-Takihara K. Signal transducer and activator of transcription 3 in the heart transduces not only a hypertrophic signal but a protective signal against doxorubicininduced cardiomyopathy. Proc Natl Acad Sci USA. 2000;97:315–319. doi: 10.1073/pnas.97.1.315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Jacoby JJ, Kalinowski A, Liu MG, Zhang SS, Gao Q, Chai GX, Ji L, Iwamoto Y, Li E, Schneider M, Russell KS, Fu XY. Cardiomyocyterestricted knockout of STAT3 results in higher sensitivity to inflammation, cardiac fibrosis, and heart failure with advanced age. Proc Natl Acad Sci USA. 2003;100:12929–12934. doi: 10.1073/pnas.2134694100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Milano CA, Dolber PC, Rockman HA, Bond RA, Venable ME, Allen LF, Lefkowitz RJ. Myocardial expression of a constitutively active alpha 1B-adrenergic receptor in transgenic mice induces cardiac hypertrophy. Proc Natl Acad Sci USA. 1994;91:10109–10113. doi: 10.1073/pnas.91.21.10109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Wang BH, Du XJ, Autelitano DJ, Milano CA, Woodcock EA. Adverse effects of constitutively active alpha(1B)-adrenergic receptors after pressure overload in mouse hearts. Am J Physiol Heart Circ Physiol. 2000;279:H1079–H1086. doi: 10.1152/ajpheart.2000.279.3.H1079. [DOI] [PubMed] [Google Scholar]
  • 36.Grupp IL, Lorenz JN, Walsh RA, Boivin GP, Rindt H. Overexpression of alpha1B-adrenergic receptor induces left ventricular dysfunction in the absence of hypertrophy. Am J Physiol. 1998;275:H1338–H1350. doi: 10.1152/ajpheart.1998.275.4.H1338. [DOI] [PubMed] [Google Scholar]
  • 37.Benoit MJ, Rindt H, Allen BG. Cardiac-specific transgenic overexpression of alpha1B-adrenergic receptors induce chronic activation of ERK MAPK signalling. Biochem Cell Biol. 2004;82:719–727. doi: 10.1139/o04-123. [DOI] [PubMed] [Google Scholar]
  • 38.Akhter SA, Milano CA, Shotwell KF, Cho MC, Rockman HA, Lefkowitz RJ, Koch WJ. Transgenic mice with cardiac overexpression of alpha1B-adrenergic receptors. In vivo alpha1- adrenergic receptor-mediated regulation of beta-adrenergic signaling. J Biol Chem. 1997;272:21253–21259. doi: 10.1074/jbc.272.34.21253. [DOI] [PubMed] [Google Scholar]
  • 39.Iaccarino G, Keys JR, Rapacciuolo A, Shotwell KF, Lefkowitz RJ, Rockman HA, Koch WJ. Regulation of myocardial betaARK1 expression in catecholamine-induced cardiac hypertrophy in transgenic mice overexpressing alpha1B-adrenergic receptors. J Am Coll Cardiol. 2001;38:534–540. doi: 10.1016/s0735-1097(01)01396-1. [DOI] [PubMed] [Google Scholar]
  • 40.Zuscik MJ, Chalothorn D, Hellard D, Deighan C, McGee A, Daly CJ, Waugh DJ, Ross SA, Gaivin RJ, Morehead AJ, Thomas JD, Plow EF, McGrath JC, Piascik MT, Perez DM. Hypotension, autonomic failure, and cardiac hypertrophy in transgenic mice overexpressing the alpha 1B-adrenergic receptor. J Biol Chem. 2001;276:13738–13743. doi: 10.1074/jbc.M008693200. [DOI] [PubMed] [Google Scholar]
  • 41.Vecchione C, Fratta L, Rizzoni D, Notte A, Poulet R, Porteri E, Frati G, Guelfi D, Trimarco V, Mulvany MJ, Agabiti-Rosei E, Trimarco B, Cotecchia S, Lembo G. Cardiovascular influences of alpha1b-adrenergic receptor defect in mice. Circulation. 2002;105:1700–1707. doi: 10.1161/01.cir.0000012750.08480.55. [DOI] [PubMed] [Google Scholar]
  • 42.Bristow MR, Ginsburg R, Gilbert EM, Hershberger RE. Heterogeneous regulatory changes in cell surface membrane receptors coupled to a positive inotropic response in the failing human heart. Basic Res Cardiol. 1987;82(Suppl 2):369–376. doi: 10.1007/978-3-662-11289-2_36. [DOI] [PubMed] [Google Scholar]
  • 43.Steinfath M, Danielsen W, von der Leyen H, Mende U, Meyer W, Neumann J, Nose M, Reich T, Schmitz W, Scholz H. Reduced alpha 1- and beta 2-adrenoceptor-mediated positive inotropic effects in human end-stage heart failure. Br J Pharmacol. 1992;105:463–469. doi: 10.1111/j.1476-5381.1992.tb14276.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Iwai-Kanai E, Hasegawa K, Araki M, Kakita T, Morimoto T, Sasayama S. alpha- and beta-adrenergic pathways differentially regulate cell type-specific apoptosis in rat cardiac myocytes. Circulation. 1999;100:305–311. doi: 10.1161/01.cir.100.3.305. [DOI] [PubMed] [Google Scholar]
  • 45.Brodde OE. Beta- and alpha-adrenoceptor-agonists and -antagonists in chronic heart failure. Basic Res Cardiol. 1990;85(Suppl 1):57–66. doi: 10.1007/978-3-662-11038-6_5. [DOI] [PubMed] [Google Scholar]
  • 46.ALLHAT Collaborative Research Group. Major cardiovascular events in hypertensive patients randomized to doxazosin vs chlorthalidone: the antihypertensive and lipid-lowering treatment to prevent heart attack trial (ALLHAT) JAMA. 2000;283:1967–1975. [PubMed] [Google Scholar]
  • 47.Koshimizu TA, Tsujimoto G, Hirasawa A, Kitagawa Y, Tanoue A. Carvedilol selectively inhibits oscillatory intracellular calcium changes evoked by human alpha1D- and alpha1B-adrenergic receptors. Cardiovasc Res. 2004;63:662–672. doi: 10.1016/j.cardiores.2004.05.014. [DOI] [PubMed] [Google Scholar]
  • 48.Varma DR, Rindt H, Chemtob S, Mulay S. Mechanism of the negative inotropic effects of alpha 1-adrenoceptor agonists on mouse myocardium. Can J Physiol Pharmacol. 2003;81:783–789. doi: 10.1139/y03-071. [DOI] [PubMed] [Google Scholar]
  • 49.Varma DR, Deng XF. Cardiovascular alpha1-adrenoceptor subtypes: functions and signaling. Can J Physiol Pharmacol. 2000;78:267–292. [PubMed] [Google Scholar]
  • 50.Wang GY, McCloskey DT, Turcato S, Swigart PM, Simpson PC, Baker AJ. Contrasting inotropic responses to alpha1-adrenergic receptor stimulation in left versus right ventricular myocardium. Am J Physiol Heart Circ Physiol. 2006;291:H2013–H2017. doi: 10.1152/ajpheart.00167.2006. [DOI] [PubMed] [Google Scholar]
  • 51.Wang GY, Yeh CC, Jensen BC, Mann MJ, Simpson PC, Baker AJ. Heart failure switches the RV alpha1-adrenergic inotropic response from negative to positive. Am J Physiol Heart Circ Physiol. 2010;298:H913–H920. doi: 10.1152/ajpheart.00259.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Jensen BC, Swigart PM, De Marco T, Hoopes C, Simpson PC. {alpha}1-Adrenergic receptor subtypes in nonfailing and failing human myocardium. Circ Heart Fail. 2009;2:654–663. doi: 10.1161/CIRCHEARTFAILURE.108.846212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Grigore A, Poindexter B, Vaughn WK, Nussmeier N, Frazier OH, Cooper JR, Gregoric ID, Buja LM, Bick RJ. Alterations in alpha adrenoreceptor density and localization after mechanical left ventricular unloading with the Jarvik flowmaker left ventricular assist device. J Heart Lung Transplant. 2005;24:609–613. doi: 10.1016/j.healun.2004.03.005. [DOI] [PubMed] [Google Scholar]
  • 54.O’Connell TD, Swigart PM, Rodrigo MC, Ishizaka S, Joho S, Turnbull L, Tecott LH, Baker AJ, Foster E, Grossman W, Simpson PC. Alpha1-adrenergic receptors prevent a maladaptive cardiac response to pressure overload. J Clin Invest. 2006;116:1005–1015. doi: 10.1172/JCI22811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Du XJ, Fang L, Gao XM, Kiriazis H, Feng X, Hotchkin E, Finch AM, Chaulet H, Graham RM. Genetic enhancement of ventricular contractility protects against pressure-overload-induced cardiac dysfunction. J Mol Cell Cardiol. 2004;37:979–987. doi: 10.1016/j.yjmcc.2004.07.010. [DOI] [PubMed] [Google Scholar]
  • 56.Ross SA, Rorabaugh BR, Chalothorn D, Yun J, Gonzalez-Cabrera PJ, McCune DF, Piascik MT, Perez DM. The alpha(1B)-adrenergic receptor decreases the inotropic response in the mouse Langendorff heart model. Cardiovasc Res. 2003;60:598–607. doi: 10.1016/j.cardiores.2003.09.020. [DOI] [PubMed] [Google Scholar]
  • 57.Lemire I, Ducharme A, Tardif JC, Poulin F, Jones LR, Allen BG, Hébert TE, Rindt H. Cardiac-directed overexpression of wild-type alpha1B-adrenergic receptor induces dilated cardiomyopathy. Am J Physiol Heart Circ Physiol. 2001;281:H931–H938. doi: 10.1152/ajpheart.2001.281.2.H931. [DOI] [PubMed] [Google Scholar]
  • 58.Rivard K, Trépanier-Boulay V, Rindt H, Fiset C. Electrical remodeling in a transgenic mouse model of alpha1B-adrenergic receptor overexpression. Am J Physiol Heart Circ Physiol. 2009;296:H704–H718. doi: 10.1152/ajpheart.00337.2008. [DOI] [PubMed] [Google Scholar]
  • 59.Du XJ. Distinct role of adrenoceptor subtypes in cardiac adaptation to chronic pressure overload. Clin Exp Pharmacol Physiol. 2008;35:355–360. doi: 10.1111/j.1440-1681.2007.04871.x. [DOI] [PubMed] [Google Scholar]
  • 60.Rona G. Catecholamine cardiotoxicity. J Mol Cell Cardiol. 1985;17:291–306. doi: 10.1016/s0022-2828(85)80130-9. [DOI] [PubMed] [Google Scholar]
  • 61.Mann DL, Kent RL, Parsons B, Cooper G., 4th Adrenergic effects on the biology of the adult mammalian cardiocyte. Circulation. 1992;85:790–804. doi: 10.1161/01.cir.85.2.790. [DOI] [PubMed] [Google Scholar]
  • 62.Singh K, Communal C, Colucci WS. Inhibition of protein phosphatase 1 induces apoptosis in neonatal rat cardiac myocytes: role of adrenergic receptor stimulation. Basic Res Cardiol. 2000;95:389–396. doi: 10.1007/s003950070038. [DOI] [PubMed] [Google Scholar]
  • 63.Baghelai K, Graham LJ, Wechsler AS, Jakoi ER. Delayed myocardial preconditioning by alpha1-adrenoceptors involves inhibition of apoptosis. J Thorac Cardiovasc Surg. 1999;117:980–986. doi: 10.1016/s0022-5223(99)70379-x. [DOI] [PubMed] [Google Scholar]
  • 64.Huang Y, Wright CD, Merkwan CL, Baye NL, Liang Q, Simpson PC, O’Connell TD. An alpha1A-adrenergic-extracellular signalregulated kinase survival signaling pathway in cardiac myocytes. Circulation. 2007;115:763–772. doi: 10.1161/CIRCULATIONAHA.106.664862. [DOI] [PubMed] [Google Scholar]
  • 65.Adams JW, Sakata Y, Davis MG, Sah VP, Wang Y, Liggett SB, Chien KR, Brown JH, Dorn GW., 2nd Enhanced Galphaq signaling: a common pathway mediates cardiac hypertrophy and apoptotic heart failure. Proc Natl Acad Sci USA. 1998;95:10140–10145. doi: 10.1073/pnas.95.17.10140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hilal-Dandan R, Kanter JR, Brunton LL. Characterization of G-protein signaling in ventricular myocytes from the adult mouse heart: differences from the rat. J Mol Cell Cardiol. 2000;32:1211–1221. doi: 10.1006/jmcc.2000.1156. [DOI] [PubMed] [Google Scholar]
  • 67.Steinberg SF, Drugge ED, Bilezikian JP, Robinson RB. Acquisition by innervated cardiac myocytes of a pertussis toxin-specific regulatory protein linked to the alpha 1-receptor. Science. 1985;230:186–188. doi: 10.1126/science.2994230. [DOI] [PubMed] [Google Scholar]
  • 68.Rorabaugh BR, Gaivin RJ, Papay RS, Shi T, Simpson PC, Perez DM. Both alpha(1A)- and alpha(1B)-adrenergic receptors crosstalk to down regulate beta(1)-ARs in mouse heart: coupling to differential PTX-sensitive pathways. J Mol Cell Cardiol. 2005;39:777–784. doi: 10.1016/j.yjmcc.2005.07.015. [DOI] [PubMed] [Google Scholar]
  • 69.Kurz T, Yamada KA, DaTorre SD, Corr PB. Alpha 1-adrenergic system and arrhythmias in ischaemic heart disease. Eur Heart J. 1991;12(Suppl F):88–98. doi: 10.1093/eurheartj/12.suppl_f.88. [DOI] [PubMed] [Google Scholar]
  • 70.Bankwala Z, Hale SL, Kloner RA. Alpha-adrenoceptor stimulation with exogenous norepinephrine or release of endogenous catecholamines mimics ischemic preconditioning. Circulation. 1994;90:1023–1028. doi: 10.1161/01.cir.90.2.1023. [DOI] [PubMed] [Google Scholar]
  • 71.Banerjee A, Locke-Winter C, Rogers KB, Mitchell MB, Brew EC, Cairns CB, Bensard DD, Harken AH. Preconditioning against myocardial dysfunction after ischemia and reperfusion by an alpha 1-adrenergic mechanism. Circ Res. 1993;73:656–670. doi: 10.1161/01.res.73.4.656. [DOI] [PubMed] [Google Scholar]
  • 72.Hu K, Nattel S. Mechanisms of ischemic preconditioning in rat hearts. Involvement of alpha 1B-adrenoceptors, pertussis toxinsensitive G proteins, and protein kinase C. Circulation. 1995;92:2259–2265. doi: 10.1161/01.cir.92.8.2259. [DOI] [PubMed] [Google Scholar]
  • 73.Kariya T, Minatoguchi S, Ohno T, Yamashita K, Uno Y, Arai M, Koshiji M, Fujiwara T, Fujiwara H. Infarct size-reducing effect of ischemic preconditioning is related to alpha1b-adrenoceptors but not to alpha1a-adrenoceptors in rabbits. J Cardiovasc Pharmacol. 1997;30:437–445. doi: 10.1097/00005344-199710000-00006. [DOI] [PubMed] [Google Scholar]
  • 74.Gao H, Chen L, Yang HT. Activation of alpha1B-adrenoceptors alleviates ischemia/reperfusion injury by limitation of mitochondrial Ca2+ overload in cardiomyocytes. Cardiovasc Res. 2007;75:584–595. doi: 10.1016/j.cardiores.2007.04.008. [DOI] [PubMed] [Google Scholar]
  • 75.Xiao L, Jeffries WB. Kinetics of alkylation of cloned rat alpha1-adrenoceptor subtypes by chloroethylclonidine. Eur J Pharmacol. 1998;347:319–327. doi: 10.1016/s0014-2999(98)00109-5. [DOI] [PubMed] [Google Scholar]
  • 76.Amirahmadi F, Turnbull L, Du XJ, Graham RM, Woodcock EA. Heightened alpha1A-adrenergic receptor activity suppresses ischaemia/reperfusion-induced Ins(1,4,5)P3 generation in the mouse heart: a comparison with ischaemic preconditioning. Clin Sci. 2008;114:157–164. doi: 10.1042/CS20070110. [DOI] [PubMed] [Google Scholar]
  • 77.Gao XM, Wang BH, Woodcock E, Du XJ. Expression of active alpha(1B)-adrenergic receptors in the heart does not alleviate ischemic reperfusion injury. J Mol Cell Cardiol. 2000;32:1679–1686. doi: 10.1006/jmcc.2000.1201. [DOI] [PubMed] [Google Scholar]
  • 78.Sah VP, Hoshijima M, Chien KR, Brown JH. Rho is required for Galphaq and alpha1-adrenergic receptor signaling in cardiomyocytes. Dissociation of Ras and Rho pathways. J Biol Chem. 1996;271:31185–31190. doi: 10.1074/jbc.271.49.31185. [DOI] [PubMed] [Google Scholar]
  • 79.Hines WA, Thorburn A. Ras and rho are required for Galphaqinduced hypertrophic gene expression in neonatal rat cardiac myocytes. J Mol Cell Cardiol. 1998;30:485–494. doi: 10.1006/jmcc.1997.0613. [DOI] [PubMed] [Google Scholar]
  • 80.Appert-Collin A, Cotecchia S, Nenniger-Tosato M, Pedrazzini T, Diviani D. The A-kinase anchoring protein (AKAP)-Lbcsignaling complex mediates alpha1 adrenergic receptor-induced cardiomyocyte hypertrophy. Proc Natl Acad Sci USA. 2007;104:10140–10145. doi: 10.1073/pnas.0701099104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ramirez MT, Sah VP, Zhao XL, Hunter JJ, Chien KR, Brown JH. The MEKK–JNK pathway is stimulated by alpha1-adrenergic receptor and ras activation and is associated with in vitro and in vivo cardiac hypertrophy. J Biol Chem. 1997;272:14057–14061. doi: 10.1074/jbc.272.22.14057. [DOI] [PubMed] [Google Scholar]
  • 82.Sakata Y, Hoit BD, Liggett SB, Walsh RA, Dorn GW., 2nd Decompensation of pressure-overload hypertrophy in G alpha q-overexpressing mice. Circulation. 1998;97:1488–1495. doi: 10.1161/01.cir.97.15.1488. [DOI] [PubMed] [Google Scholar]
  • 83.D’Angelo DD, Sakata Y, Lorenz JN, Boivin GP, Walsh RA, Liggett SB, Dorn GW., 2nd Transgenic Galphaq overexpression induces cardiac contractile failure in mice. Proc Natl Acad Sci USA. 1997;94:8121–8126. doi: 10.1073/pnas.94.15.8121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Gottshall KR, Hunter JJ, Tanaka N, Dalton N, Becker KD, Ross J, Jr, Chien KR. Ras-dependent pathways induce obstructive hypertrophy in echo-selected transgenic mice. Proc Natl Acad Sci USA. 1997;94:4710–4715. doi: 10.1073/pnas.94.9.4710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Hunter JJ, Tanaka N, Rockman HA, Ross J, Jr, Chien KR. Ventricular expression of a MLC-2v-ras fusion gene induces cardiac hypertrophy and selective diastolic dysfunction in transgenic mice. J Biol Chem. 1995;270:23173–23178. doi: 10.1074/jbc.270.39.23173. [DOI] [PubMed] [Google Scholar]
  • 86.Xiao L, Pimental DR, Amin JK, Singh K, Sawyer DB, Colucci WS. MEK1/2–ERK1/2 mediates alpha1-adrenergic receptor-stimulated hypertrophy in adult rat ventricular myocytes. J Mol Cell Cardiol. 2001;33:779–787. doi: 10.1006/jmcc.2001.1348. [DOI] [PubMed] [Google Scholar]
  • 87.Wang L, Proud CG. Ras/Erk signaling is essential for activation of protein synthesis by Gq protein-coupled receptor agonists in adult cardiomyocytes. Circ Res. 2002;91:821–829. doi: 10.1161/01.res.0000041029.97988.e9. [DOI] [PubMed] [Google Scholar]
  • 88.Wenham D, Rahmatullah RJ, Rahmatullah M, Hansen CA, Robishaw JD. Differential coupling of alpha1-adrenoreceptor subtypes to phospholipase C and mitogen activated protein kinase in neonatal rat cardiac myocytes. Eur J Pharmacol. 1997;339:77–86. doi: 10.1016/s0014-2999(97)01359-9. [DOI] [PubMed] [Google Scholar]
  • 89.Davidson SM, Townsend PA, Carroll C, Yurek-George A, Balasubramanyam K, Kundu TK, Stephanou A, Packham G, Ganesan A, Latchman DS. The transcriptional coactivator p300 plays a critical role in the hypertrophic and protective pathways induced by phenylephrine in cardiac cells but is specific to the hypertrophic effect of urocortin. Chembiochem. 2005;6:162–170. doi: 10.1002/cbic.200400246. [DOI] [PubMed] [Google Scholar]
  • 90.Rojas Gomez DM, Schulte JS, Mohr FW, Dhein S. Alpha-1-adrenoceptor subtype selective regulation of connexin 43 expression in rat cardiomyocytes. Naunyn Schmiedebergs Arch Pharmacol. 2008;377:77–85. doi: 10.1007/s00210-007-0244-9. [DOI] [PubMed] [Google Scholar]
  • 91.Morris JB, Kenney B, Huynh H, Woodcock EA. Regulation of the proapoptotic factor FOXO1 (FKHR) in cardiomyocytes by growth factors and alpha1-adrenergic agonists. Endocrinology. 2005;146:4370–4376. doi: 10.1210/en.2005-0162. [DOI] [PubMed] [Google Scholar]
  • 92.Zhou H-Z, Karliner JS, Zhu P, Mochly-Rosen D, Messing RO, Gray MO. Cardioprotection caused by ischemic preconditioning or by an α1-adrenergic receptor agonist is blocked in hearts from epsilon protein kinase C knockout mice. J Am Coll Cardiol. 2002;39:299. [Google Scholar]
  • 93.Wang Y, Ashraf M. Activation of alpha1-adrenergic receptor during Ca2+ pre-conditioning elicits strong protection against Ca2+ overload injury via protein kinase C signaling pathway. J Mol Cell Cardiol. 1998;30:2423–2435. doi: 10.1006/jmcc.1998.0802. [DOI] [PubMed] [Google Scholar]
  • 94.Pucéat M, Hilal-Dandan R, Strulovici B, Brunton LL, Brown JH. Differential regulation of protein kinase C isoforms in isolated neonatal and adult rat cardiomyocytes. J Biol Chem. 1994;269:16938–16944. [PubMed] [Google Scholar]
  • 95.Guo J, Sabri A, Elouardighi H, Rybin V, Steinberg SF. Alpha1- adrenergic receptors activate AKT via a Pyk2/PDK-1 pathway that is tonically inhibited by novel protein kinase C isoforms in cardiomyocytes. Circ Res. 2006;99:1367–1375. doi: 10.1161/01.RES.0000252830.01581.fd. [DOI] [PubMed] [Google Scholar]
  • 96.Meng X, Brown JM, Ao L, Banerjee A, Harken AH. Norepinephrine induces cardiac heat shock protein 70 and delayed cardioprotection in the rat through alpha 1 adrenoceptors. Cardiovasc Res. 1996;32:374–383. doi: 10.1016/0008-6363(96)00078-8. [DOI] [PubMed] [Google Scholar]
  • 97.Lacoste A, De Cian MC, Cueff A, Poulet SA. Noradrenaline and alpha-adrenergic signaling induce the hsp70 gene promoter in mollusc immune cells. J Cell Sci. 2001;114:3557–3564. doi: 10.1242/jcs.114.19.3557. [DOI] [PubMed] [Google Scholar]
  • 98.Takahashi Y, Takemura S, Minamiyama Y, Shibata T, Hirai H, Sasaki Y, Sakaguchi M, Suehiro S. Landiolol has cardioprotective effects against reperfusion injury in the rat heart via the PKCepsilon signaling pathway. Free Radic Res. 2007;41:757–769. doi: 10.1080/10715760701338810. [DOI] [PubMed] [Google Scholar]
  • 99.Serejo FC, Rodrigues LF, Jr, da Silva Tavares KC, de Carvalho AC, Nascimento JH. Cardioprotective properties of humoral factors released from rat hearts subject to ischemic preconditioning. J Cardiovasc Pharmacol. 2007;49:214–220. doi: 10.1097/FJC.0b013e3180325ad9. [DOI] [PubMed] [Google Scholar]
  • 100.Foody JM, Farrell MH, Krumholz HM. beta-Blocker therapy in heart failure: scientific review. JAMA. 2002;287:883–889. doi: 10.1001/jama.287.7.883. [DOI] [PubMed] [Google Scholar]
  • 101.Iversen LL, Iversen SD, Bloom FE, Roth RH. Introduction to Neuropsychopharmacology. New York: Oxford University Press; 2009. Catecholamines; pp. 150–213. [Google Scholar]
  • 102.Ordway GA, Schwartz MA, Frazer A, editors. Brain Norepinephrine. Cambridge: Cambridge University Press; 2007. Part III. Norepinephrine and behavioral; pp. 157–298. [Google Scholar]
  • 103.Ordway GA, Schwartz MA, Frazer A, editors. Brain Norepinephrine. Cambridge: Cambridge University Press; 2007. Part III. The biology of norepinephrine in CNS pathology; pp. 299–514. [Google Scholar]
  • 104.Unnerstall JR, Fernandez I, Orensanz LM. The alpha-adrenergic receptor: radiohistochemical analysis of functional characteristics and biochemical differences. Pharmacol Biochem Behav. 1985;22:859–874. doi: 10.1016/0091-3057(85)90538-6. [DOI] [PubMed] [Google Scholar]
  • 105.Domyancic AV, Morilak DA. Distribution of alpha1A adrenergic receptor mRNA in the rat brain visualized by in situ hybridization. J Comp Neurol. 1997;386:358–378. doi: 10.1002/(sici)1096-9861(19970929)386:3<358::aid-cne3>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
  • 106.Jensen BC, Swigart PM, Simpson PC. Ten commercial antibodies for alpha-1-adrenergic receptor subtypes are nonspecific. Naunyn Schmiedebergs Arch Pharmacol. 2009;379:409–412. doi: 10.1007/s00210-008-0368-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Palacios JM, Hoyer D, Cortés R. alpha 1-Adrenoceptors in the mammalian brain: similar pharmacology but different distribution in rodents and primates. Brain Res. 1987;419:65–75. doi: 10.1016/0006-8993(87)90569-5. [DOI] [PubMed] [Google Scholar]
  • 108.Zilles K, Gross G, Schleicher A, Schildgen S, Bauer A, Bahro M, Schwendemann G, Zech K, Kolassa N. Regional and laminar distributions of alpha 1-adrenoceptors and their subtypes in human and rat hippocampus. Neuroscience. 1991;40:307–320. doi: 10.1016/0306-4522(91)90122-5. [DOI] [PubMed] [Google Scholar]
  • 109.Papay R, Gaivin R, McCune DF, Rorabaugh BR, Macklin WB, McGrath JC, Perez DM. Mouse alpha1B-adrenergic receptor is expressed in neurons and NG2 oligodendrocytes. J Comp Neurol. 2004;478:1–10. doi: 10.1002/cne.20215. [DOI] [PubMed] [Google Scholar]
  • 110.Papay R, Gaivin R, Jha A, McCune DF, McGrath JC, Rodrigo MC, Simpson PC, Doze VA, Perez DM. Localization of the mouse alpha1A-adrenergic receptor (AR) in the brain: alpha1AAR is expressed in neurons, GABAergic interneurons, and NG2 oligodendrocyte progenitors. J Comp Neurol. 2006;497:209–222. doi: 10.1002/cne.20992. [DOI] [PubMed] [Google Scholar]
  • 111.Rokosh DG, Simpson PC. Knockout of the alpha 1A/C-adrenergic receptor subtype: the alpha 1A/C is expressed in resistance arteries and is required to maintain arterial blood pressure. Proc Natl Acad Sci USA. 2002;99:9474–9479. doi: 10.1073/pnas.132552699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Cavalli A, Lattion AL, Hummler E, Nenniger M, Pedrazzini T, Aubert JF, Michel MC, Yang M, Lembo G, Vecchione C, Mostardini M, Schmidt A, Beermann F, Cotecchia S. Decreased blood pressure response in mice deficient of the alpha1b-adrenergic receptor. Proc Natl Acad Sci USA. 1997;94:11589–11594. doi: 10.1073/pnas.94.21.11589. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Tanoue A, Nasa Y, Koshimizu T, Shinoura H, Oshikawa S, Kawai T, Sunada S, Takeo S, Tsujimoto G. The alpha(1D)-adrenergic receptor directly regulates arterial blood pressure via vasoconstriction. J Clin Invest. 2002;109:765–775. doi: 10.1172/JCI14001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Mouradian RD, Sessler FM, Waterhouse BD. Noradrenergic potentiation of excitatory transmitter action in cerebrocortical slices: evidence for mediation by an alpha 1 receptor-linked second messenger pathway. Brain Res. 1991;546:83–95. doi: 10.1016/0006-8993(91)91162-t. [DOI] [PubMed] [Google Scholar]
  • 115.Gordon GR, Bains JS. Priming of excitatory synapses by alpha1 adrenoceptor-mediated inhibition of group III metabotropic glutamate receptors. J Neurosci. 2003;23:6223–6231. doi: 10.1523/JNEUROSCI.23-15-06223.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Marek GJ, Aghajanian GK. Alpha 1B-adrenoceptor-mediated excitation of piriform cortical interneurons. Eur J Pharmacol. 1996;305:95–100. doi: 10.1016/0014-2999(96)00158-6. [DOI] [PubMed] [Google Scholar]
  • 117.Hirono M, Obata K. Alpha-adrenoceptive dual modulation of inhibitory GABAergic inputs to Purkinje cells in the mouse cerebellum. J Neurophysiol. 2006;95:700–708. doi: 10.1152/jn.00711.2005. [DOI] [PubMed] [Google Scholar]
  • 118.Chen Q, Li DP, Pan HL. Presynaptic alpha1 adrenergic receptors differentially regulate synaptic glutamate and GABA release to hypothalamic presympathetic neurons. J Pharmacol Exp Ther. 2006;316:733–742. doi: 10.1124/jpet.105.094797. [DOI] [PubMed] [Google Scholar]
  • 119.McCormick DA, Pape HC, Williamson A. Actions of norepinephrine in the cerebral cortex and thalamus: implications for function of the central noradrenergic system. Prog Brain Res. 1991;88:293–305. doi: 10.1016/s0079-6123(08)63817-0. [DOI] [PubMed] [Google Scholar]
  • 120.Bergles DE, Doze VA, Madison DV, Smith SJ. Excitatory actions of norepinephrine on multiple classes of hippocampal CA1 interneurons. J Neurosci. 1996;16:572–585. doi: 10.1523/JNEUROSCI.16-02-00572.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Kulik A, Haentzsch A, Lückermann M, Reichelt W, Ballanyi K. Neuron-glia signaling via alpha(1) adrenoceptor-mediated Ca(2+) release in Bergmann glial cells in situ. J Neurosci. 1999;19:8401–8408. doi: 10.1523/JNEUROSCI.19-19-08401.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Malberg JE, Eisch AJ, Nestler EJ, Duman RS. Chronic antidepressant treatment increases neurogenesis in adult rat hippocampus. J Neurosci. 2000;20:9104–9110. doi: 10.1523/JNEUROSCI.20-24-09104.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Kulkarni VA, Jha S, Vaidya VA. Depletion of norepinephrine decreases the proliferation, but does not influence the survival and differentiation, of granule cell progenitors in the adult rat hippocampus. Eur J Neurosci. 2002;16:2008–2012. doi: 10.1046/j.1460-9568.2002.02268.x. [DOI] [PubMed] [Google Scholar]
  • 124.Hiramoto T, Ihara Y, Watanabe Y. alpha-1 Adrenergic receptors stimulation induces the proliferation of neural progenitor cells in vitro. Neurosci Lett. 2006;408:25–28. doi: 10.1016/j.neulet.2006.02.052. [DOI] [PubMed] [Google Scholar]
  • 125.Hiramoto T, Satoh Y, Takishima K, Watanabe Y. Induction of cell migration of neural progenitor cells in vitro by alpha-1 adrenergic receptor and dopamine D1 receptor stimulation. Neuroreport. 2008;19:793–797. doi: 10.1097/WNR.0b013e3282fd1270. [DOI] [PubMed] [Google Scholar]
  • 126.Gupta MK, Papay RS, Jurgens CW, Gaivin RJ, Shi T, Doze VA, Perez DM. alpha1-Adrenergic receptors regulate neurogenesis and gliogenesis. Mol Pharmacol. 2009;76:314–326. doi: 10.1124/mol.109.057307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Zuscik MJ, Sands S, Ross SA, Waugh DJ, Gaivin RJ, Morilak D, Perez DM. Overexpression of the alpha1B-adrenergic receptor causes apoptotic neurodegeneration: multiple system atrophy. Nat Med. 2000;6:1388–1394. doi: 10.1038/82207. [DOI] [PubMed] [Google Scholar]
  • 128.Stone EA, Zhang Y, Rosengarten H, Yeretsian J, Quartermain D. Brain alpha 1-adrenergic neurotransmission is necessary for behavioral activation to environmental change in mice. Neuroscience. 1999;94:1245–1252. doi: 10.1016/s0306-4522(99)00394-2. [DOI] [PubMed] [Google Scholar]
  • 129.Stone EA, Rosengarten H, Lin Y, Quartermain D. Pharmacological blockade of brain alpha1-adrenoceptors as measured by ex vivo [3H]prazosin binding is correlated with behavioral immobility. Eur J Pharmacol. 2001;420:97–102. doi: 10.1016/s0014-2999(01)01003-2. [DOI] [PubMed] [Google Scholar]
  • 130.Grenhoff J, Svensson TH. Prazosin modulates the firing pattern of dopamine neurons in rat ventral tegmental area. Eur J Pharmacol. 1993;233:79–84. doi: 10.1016/0014-2999(93)90351-h. [DOI] [PubMed] [Google Scholar]
  • 131.Berretta N, Bernardi G, Mercuri NB. Alpha(1)-adrenoceptormediated excitation of substantia nigra pars reticulata neurons. Neuroscience. 2000;98:599–604. doi: 10.1016/s0306-4522(00)00135-4. [DOI] [PubMed] [Google Scholar]
  • 132.Stone EA, Lin Y, Itteera A, Quartermain D. Pharmacological evidence for the role of central alpha 1B-adrenoceptors in the motor activity and spontaneous movement of mice. Neuropharmacology. 2001;40:254–261. doi: 10.1016/s0028-3908(00)00151-9. [DOI] [PubMed] [Google Scholar]
  • 133.Knauber J, Müller WE. Decreased exploratory activity and impaired passive avoidance behaviour in mice deficient for the alpha(1b)-adrenoceptor. Eur Neuropsychopharmacol. 2000;10:423–427. doi: 10.1016/s0924-977x(00)00100-0. [DOI] [PubMed] [Google Scholar]
  • 134.Drouin C, Darracq L, Trovero F, Blanc G, Glowinski J, Cotecchia S, Tassin JP. Alpha1b-adrenergic receptors control locomotor and rewarding effects of psychostimulants and opiates. J Neurosci. 2002;22:2873–2884. doi: 10.1523/JNEUROSCI.22-07-02873.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Auclair A, Cotecchia S, Glowinski J, Tassin JP. D-amphetamine fails to increase extracellular dopamine levels in mice lacking alpha 1b-adrenergic receptors: relationship between functional and nonfunctional dopamine release. J Neurosci. 2002;22:9150–9154. doi: 10.1523/JNEUROSCI.22-21-09150.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Wada T, Hasegawa Y, Ono H. Characterization of alpha1- adrenoceptor subtypes in facilitation of rat spinal motoneuron activity. Eur J Pharmacol. 1997;340:45–52. doi: 10.1016/s0014-2999(97)01406-4. [DOI] [PubMed] [Google Scholar]
  • 137.Papay R, Zuscik MJ, Ross SA, Yun J, McCune DF, Gonzalez-Cabrera P, Gaivin R, Drazba J, Perez DM. Mice expressing the alpha(1B)-adrenergic receptor induces a synucleinopathy with excessive tyrosine nitration but decreased phosphorylation. J Neurochem. 2002;83:623–634. doi: 10.1046/j.1471-4159.2002.01170.x. [DOI] [PubMed] [Google Scholar]
  • 138.Collette KM, Hurtt CC, Perez DM, Doze VA. Chronic α1A-adrenergic receptor stimulation increases lifespan in mice. FASEB J. 2011 in press. [Google Scholar]
  • 139.Pussinen R, Nieminen S, Koivisto E, Haapalinna A, Riekkinen P, Sr, Sirvio J. Enhancement of intermediate-term memory by an alpha-1 agonist or a partial agonist at the glycine site of the NMDA receptor. Neurobiol Learn Mem. 1997;67:69–74. doi: 10.1006/nlme.1996.3738. [DOI] [PubMed] [Google Scholar]
  • 140.Pussinen R, Sirviö J. Minor role for alpha1-adrenoceptors in the facilitation of induction and early maintenance of long-term potentiation in the CA1 field of the hippocampus. J Neurosci Res. 1998;51:309–315. doi: 10.1002/(SICI)1097-4547(19980201)51:3<309::AID-JNR4>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
  • 141.Miyashita T, Williams CL. Enhancement of noradrenergic neurotransmission in the nucleus of the solitary tract modulates memory storage processes. Brain Res. 2003;987:164–175. doi: 10.1016/s0006-8993(03)03323-7. [DOI] [PubMed] [Google Scholar]
  • 142.Arnsten AFT. Norepinephrine and cognitive disorders. In: Ordway GA, Schwartz MA, Frazer A, editors. Brain Norepinephrine. Cambridge: Cambridge University Press; 2007. pp. 408–435. [Google Scholar]
  • 143.Mao ZM, Arnsten AF, Li BM. Local infusion of an alpha-1 adrenergic agonist into the prefrontal cortex impairs spatial working memory performance in monkeys. Biol Psychiatry. 1999;46:1259–1265. doi: 10.1016/s0006-3223(99)00139-0. [DOI] [PubMed] [Google Scholar]
  • 144.Gibbs ME, Summers RJ. Stimulation of alpha1-adrenoceptors inhibits memory consolidation in the chick. Eur J Neurosci. 2001;14:1369–1376. doi: 10.1046/j.0953-816x.2001.01742.x. [DOI] [PubMed] [Google Scholar]
  • 145.Goldenstein B, Jurgens C, Knudson C, Lichter J, Carr P, Perez D, Doze V. α1-Adrenergic receptor regulation of seizures and neurodegeneration. FASEB J. 2008;22 748.12. [Google Scholar]
  • 146.Hong CJ, Wang YC, Liu TY, Liu HC, Tsai SJ. A study of alphaadrenoceptor gene polymorphisms and Alzheimer disease. J Neural Transm. 2001;108:445–450. doi: 10.1007/s007020170065. [DOI] [PubMed] [Google Scholar]
  • 147.Klingner M, Apelt J, Kumar A, Sorger D, Sabri O, Steinbach J, Scheunemann M, Schliebs R. Alterations in cholinergic and noncholinergic neurotransmitter receptor densities in transgenic Tg2576 mouse brain with beta-amyloid plaque pathology. Int J Dev Neurosci. 2003;21:357–369. doi: 10.1016/j.ijdevneu.2003.08.001. [DOI] [PubMed] [Google Scholar]
  • 148.Lapiz MD, Morilak DA. Noradrenergic modulation of cognitive function in rat medial prefrontal cortex as measured by attentional set shifting capability. Neuroscience. 2006;137:1039–1049. doi: 10.1016/j.neuroscience.2005.09.031. [DOI] [PubMed] [Google Scholar]
  • 149.Kalaria RN. Characterization of [125I]HEAT binding to alpha 1-receptors in human brain: assessment in aging and Alzheimer’s disease. Brain Res. 1989;501:287–294. doi: 10.1016/0006-8993(89)90645-8. [DOI] [PubMed] [Google Scholar]
  • 150.Shimohama S, Taniguchi T, Fujiwara M, Kameyama M. Biochemical characterization of alpha-adrenergic receptors in human brain and changes in Alzheimer-type dementia. J Neurochem. 1986;47:1295–1301. [PubMed] [Google Scholar]
  • 151.Szot P, White SS, Greenup JL, Leverenz JB, Peskind ER, Raskind MA. Changes in adrenoreceptors in the prefrontal cortex of subjects with dementia: evidence of compensatory changes. Neuroscience. 2007;146:471–480. doi: 10.1016/j.neuroscience.2007.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Izumi Y, Zorumski CF. Norepinephrine promotes long-term potentiation in the adult rat hippocampus in vitro. Synapse. 1999;31:196–202. doi: 10.1002/(SICI)1098-2396(19990301)31:3<196::AID-SYN4>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
  • 153.Katsuki H, Izumi Y, Zorumski CF. Noradrenergic regulation of synaptic plasticity in the hippocampal CA1 region. J Neurophysiol. 1997;77:3013–3020. doi: 10.1152/jn.1997.77.6.3013. [DOI] [PubMed] [Google Scholar]
  • 154.Scheiderer CL, Dobrunz LE, McMahon LL. Novel form of longterm synaptic depression in rat hippocampus induced by activation of alpha 1 adrenergic receptors. J Neurophysiol. 2004;91:1071–1077. doi: 10.1152/jn.00420.2003. [DOI] [PubMed] [Google Scholar]
  • 155.Stone EA, Quartermain D. Alpha-1-noradrenergic neurotransmission, corticosterone, and behavioral depression. Biol Psychiatry. 1999;46:1287–1300. doi: 10.1016/s0006-3223(99)00234-6. [DOI] [PubMed] [Google Scholar]
  • 156.Doze VA, Handel EM, Jensen KA, Darsie B, Luger EJ, Haselton JR, Talbot JN, Rorabaugh BR. Alpha(1A)- and alpha(1B)-adrenergic receptors differentially modulate antidepressant-like behavior in the mouse. Brain Res. 2009;1285:148–157. doi: 10.1016/j.brainres.2009.06.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Collette K, Fagerlie R, Haselton J, Perez DM, Doze V. Norepinephrine, through activation of α1A-ARs, stimulates production of new neurons, leading to an alleviation of depression and anxiety. FASEB J. 2010;24:1058–1057. [Google Scholar]
  • 158.Wood S, Luger E, Bethany Davis B, Haselton J, Perez D, Doze V. Chronic α1A-adrenergic receptor stimulation reduces anxiety in mice. FASEB J. 2010;24 768.6. [Google Scholar]
  • 159.Aroniadou-Anderjaska V, Qashu F, Braga MF. Mechanisms regulating GABAergic inhibitory transmission in the basolateral amygdala: implications for epilepsy and anxiety disorders. Amino Acids. 2007;32:305–315. doi: 10.1007/s00726-006-0415-x. [DOI] [PubMed] [Google Scholar]
  • 160.Weinshenker D, Szot P. The role of catecholamines in seizure susceptibility: new results using genetically engineered mice. Pharmacol Ther. 2002;94:213–233. doi: 10.1016/s0163-7258(02)00218-8. [DOI] [PubMed] [Google Scholar]
  • 161.Giorgi FS, Pizzanelli C, Biagioni F, Murri L, Fornai F. The role of norepinephrine in epilepsy: from the bench to the bedside. Neurosci Biobehav Rev. 2004;28:507–524. doi: 10.1016/j.neubiorev.2004.06.008. [DOI] [PubMed] [Google Scholar]
  • 162.Löscher W, Czuczwar SJ. Comparison of drugs with different selectivity for central alpha 1-and alpha 2-adrenoceptors in animal models of epilepsy. Epilepsy Res. 1987;1:165–172. doi: 10.1016/0920-1211(87)90037-4. [DOI] [PubMed] [Google Scholar]
  • 163.Neuman RS. Suppression of penicillin-induced focal epileptiform activity by locus ceruleus stimulation: mediation by an alpha 1-adrenoceptor. Epilepsia. 1986;27:359–366. doi: 10.1111/j.1528-1157.1986.tb03554.x. [DOI] [PubMed] [Google Scholar]
  • 164.Wu HQ, Tullii M, Samanin R, Vezzani A. Norepinephrine modulates seizures induced by quinolinic acid in rats: selective and distinct roles of alpha-adrenoceptor subtypes. Eur J Pharmacol. 1987;138:309–318. doi: 10.1016/0014-2999(87)90468-7. [DOI] [PubMed] [Google Scholar]
  • 165.Stanton PK. Noradrenergic modulation of epileptiform bursting and synaptic plasticity in the dentate gyrus. Epilepsy Res Suppl. 1992;7:135–150. [PubMed] [Google Scholar]
  • 166.Ferry B, Magistretti PJ, Pralong E. Noradrenaline modulates glutamate-mediated neurotransmission in the rat basolateral amygdale in vitro. Eur J Neurosci. 1997;9:1356–1364. doi: 10.1111/j.1460-9568.1997.tb01490.x. [DOI] [PubMed] [Google Scholar]
  • 167.Yan QS, Dailey JW, Steenbergen JL, Jobe PC. Anticonvulsant effect of enhancement of noradrenergic transmission in the superior colliculus in genetically epilepsy-prone rats (GEPRs): a microinjection study. Brain Res. 1998;780:199–209. doi: 10.1016/s0006-8993(97)01139-6. [DOI] [PubMed] [Google Scholar]
  • 168.Weinshenker D, Szot P, Miller NS, Palmiter RD. Alpha(1) and beta(2) adrenoreceptor agonists inhibit pentylenetetrazoleinduced seizures in mice lacking norepinephrine. J Pharmacol Exp Ther. 2001;298:1042–1048. [PubMed] [Google Scholar]
  • 169.Gellman RL, Aghajanian GK. Pyramidal cells in piriform cortex receive a convergence of inputs from monoamine activated GABAergic interneurons. Brain Res. 1993;600:63–73. doi: 10.1016/0006-8993(93)90402-9. [DOI] [PubMed] [Google Scholar]
  • 170.Braga MF, Aroniadou-Anderjaska V, Manion ST, Hough CJ, Li H. Stress impairs alpha(1A) adrenoceptor-mediated noradrenergic facilitation of GABAergic transmission in the basolateral amygdala. Neuropsychopharmacology. 2004;29:45–58. doi: 10.1038/sj.npp.1300297. [DOI] [PubMed] [Google Scholar]
  • 171.Alreja M, Liu W. Noradrenaline induces IPSCs in rat medial septal/diagonal band neurons: involvement of septohippocampal GABAergic neurons. J Physiol (Lond) 1996;494(Pt 1):201–215. doi: 10.1113/jphysiol.1996.sp021485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Kawaguchi Y, Shindou T. Noradrenergic excitation and inhibition of GABAergic cell types in rat frontal cortex. J Neurosci. 1998;18:6963–6976. doi: 10.1523/JNEUROSCI.18-17-06963.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Hillman KL, Lei S, Doze VA, Porter JE. Alpha-1A adrenergic receptor activation increases inhibitory tone in CA1 hippocampus. Epilepsy Res. 2009;84:97–109. doi: 10.1016/j.eplepsyres.2008.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Hillman KL, Knudson CA, Carr PA, Doze VA, Porter JE. Adrenergic receptor characterization of CA1 hippocampal neurons using real time single cell RT-PCR. Brain Res Mol Brain Res. 2005;139:267–276. doi: 10.1016/j.molbrainres.2005.05.033. [DOI] [PubMed] [Google Scholar]
  • 175.Hillman KL, Doze VA, Porter JE. Alpha1A-adrenergic receptors are functionally expressed by a subpopulation of cornu ammonis-1 interneurons in rat hippocampus. J Pharmacol Exp Ther. 2007;321:1062–1068. doi: 10.1124/jpet.106.119297. [DOI] [PubMed] [Google Scholar]
  • 176.Jurgens CWD, Knudson CA, Carr PA, Perez DM, Doze VA. α1-Adrenergic receptor regulation of interneuron function. FASEB J. 2009;23 946.4. [Google Scholar]
  • 177.Kunieda T, Zuscik MJ, Boongird A, Perez DM, Lüders HO, Najm IM. Systemic overexpression of the alpha 1B-adrenergic receptor in mice: an animal model of epilepsy. Epilepsia. 2002;43:1324–1329. doi: 10.1046/j.1528-1157.2002.13202.x. [DOI] [PubMed] [Google Scholar]
  • 178.Yun J, Gaivin RJ, McCune DF, Boongird A, Papay RS, Ying Z, Gonzalez-Cabrera PJ, Najm I, Perez DM. Gene expression profile of neurodegeneration induced by alpha1B-adrenergic receptor overactivity: NMDA/GABAA dysregulation and apoptosis. Brain. 2003;126:2667–2681. doi: 10.1093/brain/awg277. [DOI] [PubMed] [Google Scholar]
  • 179.Pizzanelli C, Lazzeri G, Fulceri F, Giorgi FS, Pasquali L, Cifelli G, Murri L, Fornai F. Lack of alpha 1b-adrenergic receptor protects against epileptic seizures. Epilepsia. 2009;50(Suppl 1):59–64. doi: 10.1111/j.1528-1167.2008.01972.x. [DOI] [PubMed] [Google Scholar]
  • 180.Knudson CA, Carr PA, Perez DM, Doze VA. α1A-Adrenergic receptor overexpression protects hippocampal interneurons. FASEB J. 2007;21:A1209. (896.6) [Google Scholar]
  • 181.Cohen BM, Lipinski JF. In vivo potencies of antipsychotic drugs in blocking α1-noradrenergic and dopamine D2 receptors: implications for drug mechanisms of action. Life Sci. 1986;147:1194–1198. doi: 10.1016/0024-3205(86)90111-6. [DOI] [PubMed] [Google Scholar]
  • 182.Baldessarini RJ, Huston-Lyons D, Campbell A, Marsh E, Cohen BM. Do central antiadrenergic actions contribute to the atypical properties of clozapine? Br J Psychiatry Suppl. 1992:12–16. [PubMed] [Google Scholar]
  • 183.Saiz PA, Susce MT, Clark DA, Kerwin RW, Molero P, Arranz MJ, de Leon J. An investigation of the alpha1A-adrenergic receptor gene and antipsychotic-induced side-effects. Hum Psychopharmacol. 2008;23:107–114. doi: 10.1002/hup.903. [DOI] [PubMed] [Google Scholar]
  • 184.Ma J, Ye N, Cohen BM. Expression of noradrenergic alpha1, serotoninergic 5HT2a and dopaminergic D2 receptors on neurons activated by typical and atypical antipsychotic drugs. Prog Neuropsychopharmacol Biol Psychiatry. 2006;30:647–657. doi: 10.1016/j.pnpbp.2005.11.040. [DOI] [PubMed] [Google Scholar]
  • 185.Sleight AJ, Koek W, Bigg DC. Binding of antipsychotic drugs at alpha 1A- and alpha 1B-adrenoceptors: risperidone is selective for the alpha 1B-adrenoceptors. Eur J Pharmacol. 1993;238:407–410. doi: 10.1016/0014-2999(93)90876-j. [DOI] [PubMed] [Google Scholar]
  • 186.Cahir M, King DJ. Antipsychotics lack alpha 1A/B adrenoceptor subtype selectivity in the rat. Eur Neuropsychopharmacol. 2005;15:231–234. doi: 10.1016/j.euroneuro.2004.10.002. [DOI] [PubMed] [Google Scholar]
  • 187.Cahir M, Mawhinney T, King DJ. Differential region-specific regulation of central alpha 1-adrenoceptor binding following chronic haloperidol and clozapine administration in the rat. Psychopharmacology (Berl) 2004;172:196–201. doi: 10.1007/s00213-003-1639-z. [DOI] [PubMed] [Google Scholar]
  • 188.Keilhoff G, Grecksch G, Bernstein HG, Roskoden T, Becker A. Risperidone and haloperidol promote survival of stem cells in the rat hippocampus. Eur Arch Psychiatry Clin Neurosci. 2010;260:151–162. doi: 10.1007/s00406-009-0033-1. [DOI] [PubMed] [Google Scholar]
  • 189.Clark DA, Arranz MJ, Mata I, Lopéz-Ilundain J, Pérez-Nievas F, Kerwin RW. Polymorphisms in the promoter region of the alpha1A-adrenoceptor gene are associated with schizophrenia/ schizoaffective disorder in a Spanish isolate population. Biol Psychiatry. 2005;58:435–439. doi: 10.1016/j.biopsych.2005.04.051. [DOI] [PubMed] [Google Scholar]
  • 190.Manji HK, McNamara R, Chen G, Lenox RH. Signalling pathways in the brain: cellular transduction of mood stabilisation in the treatment of manic-depressive illness. Aust N Z J Psychiatry. 1999;(Suppl):S65–S83. doi: 10.1111/j.1440-1614.1999.00670.x. [DOI] [PubMed] [Google Scholar]
  • 191.Volk DW, Eggan SM, Lewis DA. Alterations in metabotropic glutamate receptor 1a and regulator of G protein signaling 4 in the prefrontal cortex in schizophrenia. Am J Psychiatry. 2010;167:1489–1498. doi: 10.1176/appi.ajp.2010.10030318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Mirnics K, Middleton FA, Stanwood GD, Lewis DA, Levitt P. Disease-specific changes in regulator of G-protein signaling-4 (RGS4) expression in schizophrenia. Mol Psychiatry. 2001;6:293–301. doi: 10.1038/sj.mp.4000866. [DOI] [PubMed] [Google Scholar]
  • 193.Prasad KM, Chowdari KV, Nimgaonkar VL, Talkowski ME, Lewis DA, Keshavan MS. Genetic polymorphisms of the RGS4 and dorsolateral prefrontal cortex morphometry among first episode schizophrenia patients. Mol Psychiatry. 2005;10:213–219. doi: 10.1038/sj.mp.4001562. [DOI] [PubMed] [Google Scholar]
  • 194.Erdely HA, Tamminga CA, Roberts RC, Vogel MW. Regional alterations in RGS4 protein in schizophrenia. Synapse. 2006;59:472–479. doi: 10.1002/syn.20265. [DOI] [PubMed] [Google Scholar]
  • 195.Ding L, Hegde AN. Expression of RGS4 splice variants in dorsolateral prefrontal cortex of schizophrenic and bipolar disorder patients. Biol Psychiatry. 2009;65:541–545. doi: 10.1016/j.biopsych.2008.10.026. [DOI] [PubMed] [Google Scholar]
  • 196.Hague C, Bernstein LS, Ramineni S, Chen Z, Minneman KP, Hepler JR. Selective inhibition of alpha1A-adrenergic receptor signaling by RGS2 association with the receptor third intracellular loop. J Biol Chem. 2005;280:27289–27295. doi: 10.1074/jbc.M502365200. [DOI] [PubMed] [Google Scholar]

RESOURCES