Abstract
Genetic studies have identified numerous factors linking β-adrenergic blockade to Parkinson’s disease (PD), including human leukocyte antigen genes, the renin–angiotensin system, poly(adenosine diphosphate-ribose) polymerase 1, nerve growth factor, vascular endothelial growth factor, and the reduced form of nicotinamide adenine dinucleotide phosphate. β-Adrenergic blockade has also been implicated in PD via its effects on matrix metalloproteinases, mitogen-activated protein kinase pathways, prostaglandins, cyclooxygenase 2, and nitric oxide synthase. β-Adrenergic blockade may have a significant role in PD; therefore, the characterization of β-adrenergic blockade in patients with PD is needed.
Keywords: β-adrenergic blocker, Parkinson’s disease, tremor, β-adrenergic antagonism
Introduction
Parkinson’s disease (PD) is the second most common form of neurodegeneration among elderly individuals. The PD is clinically characterized by tremors, rigidity, slowness of movement, and postural imbalance. Dopaminergic therapies in the form of l-dopa, dopamine (DA) agonists, or monoamine oxidase B inhibitors significantly improve the characteristic motor symptoms of bradykinesia and rigidity, with a beneficial effect upon tremor in a proportion of patients. 1 However, the relationship between β-adrenergic antagonism and PD has been well established. In the brain, β-adrenergic receptors are widely distributed in different regions, including the frontal, parietal, piriform, and retrosplenial cortices, medial septal nuclei, olfactory tubercle, midbrain, striatum, hippocampus, and thalamic nuclei. 2,3 The adrenergic receptors (or adrenoceptors) are a class of G-prote-in-coupled receptors that are targets of the catecholamines, especially norepinephrine (NE; noradrenaline) and epinephrine (adrenaline). Many cells possess these receptors, and the binding of a catecholamine to the receptor will generally stimulate the sympathetic nervous system. There are 2 main groups of adrenergic receptors, α and β. β receptors have the subtypes β1, β2 and β3. All 3 are linked to Gs proteins (although β2 also couples to Gi), which in turn are linked to adenylate cyclase. Agonist binding thus causes a rise in the intracellular concentration of the second messenger cyclic adenosine monophosphate (cAMP). Downstream effectors of cAMP include cAMP-dependent protein kinase A (PKA), which mediates some of the intracellular events following hormone binding. β-adrenergic receptors can participate in the modulation of neuronal activity of cerebral structures involved in motor control. Microiontophoretic administration of nonselective β-adrenergic receptors agonist isoproterenol significantly decreased firing rate and responses to superior cerebellar peduncle stimulation in 82% of studied neurons in a dose-dependent manner. Similar changes were induced by ejection of selective β1-adrenergic receptors agonist dobutamine, while fenoterol (selective β2-adrenergic receptors) increased or reduced firing rate in 32% and 19% of rat primary motor cortex (M1), respectively. Nonselective β-adrenergic receptors antagonist propranolol enhanced both the background and evoked activity in 84% of tested neurons. 4 The subthalamic nucleus (STN) is one of the key structures in idiopathic PD. 5 During the course of the disease, the nucleus develops a bursting 25 to 45 Hz firing pattern that is associated with the development of clinical motor symptoms, including akinesia and rigidity. The spiking activity of the STN was temporarily suppressed, after the application of the β-adrenergic blocker metoprolol. A transient reduction in PD symptoms (rigidity) was detected during the suppression of STN spiking activity in patients with PD. 6 Akathisia improved in 4 patients with idiopathic PD after low-dose propranolol treatment. 7 The NE has been suggested to modulate the expression of l-3,4-dihydroxyphenylalanine (l-dopa)-induced dyskinesia (LID). Targeting the NE system may provide relief from both PD and LID. Barnum et al 4 , 8 demonstrated that moderate NE loss reduced the development and expression of abnormal involuntary movements and rotations in hemiparkinsonian rats. These authors also showed that the β-adrenergic receptor blocker maintained its antidyskinetic effects in DA-treated NE-lesioned rats. The nonselective β-adrenergic receptor blocker, propranolol, significantly attenuated established LID in patients with PD. 9 The aberrant striatal signaling associated with LID was normalized after propranolol cotreatment, and intrastriatal propranolol acutely reduced LID in a 6-hydroxydopamine (6-OHDA)-lesioned rat model. 10 Improvement in tremor was observed within 2 to 4 weeks after the initiation of β-adrenergic receptor nipradilol therapy; the efficacy rate tended to be higher in the essential tremor group than in the PD group. 11 Arotinolol, a peripheral β-adrenergic receptor blocker, significantly suppressed postural tremor in a dose-dependent manner in monkeys with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced parkinsonism. 12 Nadolol, another peripheral β-adrenergic receptor blocker, yielded a 34% reduction in tremor distance but no change in tremor frequency in idiopathic PD. 13 However, a Cochran Collaboration review revealed that it is impossible to determine whether β-adrenergic receptor blocker therapy is effective and safe for the treatment of tremor in PD. 14 These findings suggest that β-adrenergic receptor blockade may play a role in PD; herein, we discuss the potential role of β-adrenergic receptor blockers in PD.
Genetic Factors Associated With β-adrenergic Inhibition and Parkinson’s Disease
Genetic studies provide an excellent opportunity to link molecular variations with epidemiological data. Variations in DNA sequences such as polymorphisms exert modest and subtle biological effects. Receptors play a crucial role in the regulation of cellular function, and small changes in their structure can influence intracellular signal transduction pathways.
Studies have suggested that human leukocyte antigen (HLA) genes are located in the major histocompatibility complex (MHC) class II loci and that several genes in the MHC region promote susceptibility to PD. Significantly increased MHC class II expression was detected in the cerebrospinal fluid (CSF) monocytes of patients with PD. 15 The HLA genes have also been implicated in PD, and large numbers of HLA-DR-positive reactive microglia have been detected in the substantia nigra (SN) and nigrostriatal tract of patients with PD. 16,17 The HLA-DR-positive microglia has also been detected in these regions in Parkinson’s-associated dementia patients in Guam. 17 The HLA-DRB1*03 was more common in patients with PD compared with controls. 18 A higher frequency of HLA-DRB1*0301 was demonstrated in the Chinese Han population, but a lower frequency of HLA-DRB1*0406 was observed in European populations compared with that in Asian populations. 19 The variant rs3129882 was identified in genome-wide association study of the HLA gene region, and this variant has been associated with an increased risk of PD in Chinese patients. 20 The highly polymorphic HLA-DRB1 locus contains the variant rs660895, which was also reported in patients with PD. 21 The HLA rs3129882 variant was detected in Chinese Han patients with late-onset sporadic PD. 22 Moreover, a correlation between HLA-DR and HLA-DQ gene polymorphisms and the anti-β-receptor antibodies in familial cardiomyopathy has been suggested. 23 Cardiac β-adrenergic receptors and adenylate cyclase activity in dilated cardiomyopathy have been shown to be modulated by circulating autoantibodies against the cardiac β1-adrenoceptor, the presence of which is regulated by the HLA-DR. 24 Propranolol abrogated interferon-γ-induced increases in HLA class II expression and interleukin 1β secretion. 25 Expression of HLA-DR was significantly reduced in the lymphocytes of carvedilol-treated patients with chronic heart failure (CHF). 26 These findings suggest that β-adrenergic blockers might affect PD via the suppression of MHC class II antigen expression.
Trace amines, including tyramine, tryptamine, octopamine, and β-phenylethylamine (β-PEA), exist in low concentrations in the brain. Metabolically, these trace amines are closely associated with DA, NE, and serotonin neurotransmitter systems in the brain. 27,28 Trace amines produce amphetamine-like responses, such as increases in alertness and euphoria and decreases in appetite, insomnia, and tremor. 29 Trace amines function independently of classical amine transmitters and mediate some of their effects via specific receptors, such as trace amine-associated receptors (TAARs). The receptors for trace amines belong to a family of G-protein-coupled receptors. 30 In humans, TAAR1 responds to tyramine, β-PEA, octopamine, and DA, and TAAR4 is activated by tyramine and β-PEA. 30 A relatively high level of tyramine and β-PEA is present in the nigrostriatal and mesolimbic regions of the brain. 31,32 Dopaminergic neurons synthesize β-PEA, and specific high-affinity binding sites for β-PEA and tyramine have been described in the striatum and other DA-rich areas. 33 –35 Both β-PEA and tyramine suppress inhibitory synaptic potentials in neurons of the central nervous system (CNS), supporting an emerging role for these trace amines as neuromodulators. 36 There is evidence suggesting that trace amines coexist with DA in dopaminergic cells 37 and activate motor activity through interactions with the dopaminergic system. 38 The β-PEA stimulates the release of DA from the cytoplasmic pool and behaves as a DA receptor agonist with a rapid and brief action. 39 Trace amines indirectly activate DA autoreceptors through an increased efflux of newly synthesized DA. 40 Numerous studies have indicated that trace amine dysregulation may play a role in PD. In vivo, β-PEA may exert behavioral effects on the caudate nucleus through the increased release of both DA and 5-hydroxydopamine (5-OHDA). 41 Both 5- and 6-OHDA, identified as naturally occurring amines in human urine, have been detected in patients with Parkinson’s treated with l-dopa in significantly higher concentrations than those detected in untreated patients and normal controls. 42 The expression of octopamine was below control levels in all groups of patients with PD, including patients in the early stages of the disease. 43 The major extracellular metabolite of DA, 3-methoxytyramine (3-MT), induces behavioral effects in a DA-independent manner, and these effects are partially mediated through TAAR1. In normal mice, the central administration of 3-MT induced a temporary mild hyperactivity with the concomitant onset of abnormal movements. 44 Trace amines such as tyramine and β-PEA reduce DA-induced responses in midbrain dopaminergic neurons via G-protein-independent signaling mechanisms and render dopaminergic neurons less sensitive to autoreceptor feedback inhibition, thereby enhancing their sensitivity to stimulation. 45 The TAAR1 knockout (KO) mice exhibited enhanced sensitivity to the psychomotor-stimulating effects of amphetamine, which were temporally correlated with significantly larger increases in the release of both DA and NE in the dorsal striatum and were associated with a 262% increase in the proportion of striatal high-affinity DA D2 receptors. 46 The TAAR1 is significantly involved in motor control and exerts an inhibitory effect on locomotion. 47 Moreover, numerous adrenergic ligands and 3-methylated metabolites of catecholamine neurotransmitters act as TAAR1 (rTAAR1) agonists in rats. These results suggest that trace amines and catecholamine metabolites may serve as endogenous ligands for a novel intercellular signaling system widely distributed throughout the vertebrate brain and periphery. 48 Tyramine and β-PEA are allosteric antagonists of β1-2-adrenergic receptors, whereas octopamine acts as an orthosteric β2-adrenergic receptor antagonist and is a β1-adrenergic receptor agonist. 49 However, β-adrenergic receptor blockers have endocrine-disrupting potential in aquatic organisms with tyramine and octopamine receptors. 50
The primary function of the renin–angiotensin system (RAS) is to maintain fluid homeostasis and regulate blood pressure. Several components and receptors of the RAS have been identified in the CNS, 51 –54 suggesting that the RAS might be involved in brain activity. Garrido-Gil et al 55 demonstrated the presence of major RAS components in dopaminergic neurons, astrocytes and microglia in both the monkey and human substantia nigra compacta (SNc). Angiotensin types 1 and 2 (Ang 1 and 2) and renin–prorenin receptors are located at the surface of dopaminergic neurons and glial cells and have also been observed in the cytoplasm and nucleus, which suggests the presence of an intracrine or intracellular RAS in the monkey and human SNc. Levels of angiotensin-converting enzyme (ACE) activity are reduced in the CSF of patients with PD and increase with dopaminergic treatments. 56,57 Grammatopoulos et al 58 observed a maximal reduction in α-synuclein-induced toxicity of 85% and a 19% reduction in inclusion formation when cultures were treated with Ang 2 in the presence of the AT1 receptor antagonist losartan and AT2 receptor antagonist PD123319. These data suggest that agents acting on the RAS may be useful for preventing and/or treating of PD. In the striatum and nigra, the depletion of DA by reserpine induced a significant increase in the expression of AT1 and AT2, which diminished with the increasing restoration of DA function. Similarly, 6-OHDA-induced chronic dopaminergic denervation resulted in a significant increase in AT1 and AT2 expression, which diminished with l-dopa administration. A significant reduction in AT1 messenger RNA (mRNA) expression was also observed after the administration of DA to cultures of microglial cells. 59 These findings suggest an important interaction between the dopaminergic and the local RAS in the basal ganglia, which may be a major factor in the progression of PD. The AT1 receptor antagonist candesartan protected dopaminergic neurons in the nigrostriatal tract against neurotoxin-induced cell death. 60 Moreover, catecholamines alter the release of AT II. Ming et al 61 demonstrated that isoproterenol enhances the stimulatory effect of dexamethasone on AT gene expression via β2-adrenergic receptors in mouse hepatoma cells. Isoproterenol promoted an increase in the release of AT II from isolated perfused mesenteric arteries, and this release was blocked by propranolol treatment. 62 In other studies, isoproterenol increased the secretion of AT II in neuronal cultures, cultured bovine aortic endothelial cells, and the brachial arteries of patients with hypertension. 63 –65 Propranolol treatment reduced plasma renin activity (PRA) and AT I, AT II, and AT-(1-7) expression in the portal vein and periphery of cirrhotic patients compared with nontreated patients. 66 Carvedilol inhibited basal and stimulated ACE production in human endothelial cells 67 and exhibited beneficial effects on ACE activity and PRA levels in patients with CHF. 68 In addition, proliferating infantile hemangiomas express 2 essential components of the RAS, namely ACE and the AT II receptor, which are responsible for the propranolol-induced accelerated involution of large proliferating infantile hemangioma. 69 –71 Taken together, these findings suggest that the RAS is activated in patients with PD and that β-adrenergic blockers may play a role in PD by modulating the RAS.
Poly(adenosine diphosphate-ribose) polymerase 1 (PARP-1) is a nuclear protein that promotes either neuronal death or survival under certain stress conditions. The overexpression of PARP-1 has been reported in the dopaminergic neurons of the SN in PD. 72 Poly(adenosine diphosphate-ribose) polymerase 1 is also implicated in MPTP-induced neurotoxicity in vivo. 73 The MPTP is a neurotoxin that induces parkinsonian symptoms in human and animals, but mice lacking the PARP gene are spared from MPTP neurotoxicity. 74 In vitro studies have shown that PARP-1 participates in the regulation of α-synuclein expression through binding to the Rep1 polymorphic site upstream of the SNCA gene. 75 The PARP inhibitors attenuate neuronal death after MPPT-induced neurotoxicity in mice. 76,77 The pharmacological inhibition of PARP-1 reduces α-synuclein- and MPTP-induced cytotoxicity in in vitro models of PD. 78 The PARP-1 variants are protective against PD. 79 Moreover, rabbits treated with ketamine exhibited reduced left ventricular ejection fractions and ventricular conduction velocity and increased susceptibility to ventricular arrhythmia. Metoprolol treatment prevented these pathophysiological alterations. The expression of PARP-1 and apoptosis-inducing factor were increased after ketamine treatment and sharply reduced after metoprolol administration. 80 Propranolol treatment markedly suppressed PARP activation in skeletal muscle biopsies from pediatric patients with burn. 81 Propranolol also protected against staurosporine-induced DNA fragmentation and PARP cleavage in SH-SY5Y neuroblastoma cells. 82 The nonselective β-blocker carvedilol significantly inhibited apoptosis and suppressed activated PARP-1 cleavage in human cardiac tissue. 83 Carvedilol significantly reduced ischemia–reperfusion-induced poly- and mono-adenosine diphosphate-ribosylation in heart perfusion and rheological models. 84 Carvedilol also reduced PARP activity in the hippocampus and protected neurons against death after transient forebrain ischemia. 85 Metipranolol reduced the sodium nitroprusside-induced breakdown of PARP-1 in the eyes and retinas of rats. 86 These findings suggest that PARP-1 is activated in patients with PD and that β-adrenergic antagonists may affect PD through the suppression of PARP-1.
Nerve growth factor (NGF) is a small secreted protein that is important for the growth, maintenance, and survival of certain target neurons. The NGF has been implicated in maintaining and regulating the septohippocampal pathway, which is involved in learning and memory. 87 –89 The NGF is also present in the human SN 90 and the adrenal gland. 91 The NGF concentrations are reduced in the SN of patients with PD and in rat models of PD. 92,93 The NGF levels showed greater reductions during the early stages of PD, 89 implying that reduced NGF may be involved in the pathogenesis of the disease. The NGF protects the DA neurotoxicity induced by MPTP, rotenone, and 6-OHDA via different pathways. 94 –96 The chronic infusion of NGF into the rat striatum resulted in cholinergic hyperinnervation and the reduced spontaneous activity of striatal neurons. 97 Moreover, NGF increased the survival of dopaminergic grafts, rescued nigral dopaminergic neurons and restored motor dysfunction in a rat model of PD. 98,99 l-dopa induces the synthesis of NGF and growth hormone in patients with PD. 100 Clenbuterol, a long-acting β2-adrenergic agonist, significantly increased both NGF mRNA and protein expression in Swiss mouse 3T3 cells. 101 The exposure of nerve cells to isoproterenol, a β-adrenergic agonist, increased NGF mRNA expression, and this effect was blocked by propranolol. 102,103 Interestingly, NGF acts with the β2-adrenoceptor to induce spontaneous nociceptive behavior during temporomandibular joint inflammatory hyperalgesia. However, the coadministration of carrageenan with the β2-adrenoceptor antagonist ICI 118.55 significantly reduced NGF-induced nociception. 104 The neuroprotective effects of β-adrenergic receptor antagonists during cerebral ischemia may be associated with nociceptive pain. 105 –107 Brain injury is reduced and neurological outcomes are improved after middle cerebral artery occlusion in mice lacking the β2-adrenergic receptor agonist and in wild-type mice pretreated with a β2-adrenergic receptor antagonist. 108 Taken together, β-adrenergic receptor antagonists may play a role in PD through the regulation of NGF secretion.
Angiogenesis is a complex process that involves coordinated endothelial cell activation, proliferation, migration, tube formation, and capillary sprouting. In addition, angiogenesis requires the participation of numerous intracellular signaling pathways. Vascular endothelial growth factor (VEGF) is a key mediator of angiogenesis and has been shown to have neuroprotective effects on DA neurons in models of 6-OHDA-induced toxicity, reducing amphetamine-induced rotational behavior and preserving tyrosine hydroxylase-positive neurons and fibers. 109 In rat midbrain cultures, increased levels of VEGF-B transcription have been reported following the addition of the neurotoxin rotenone. 110 An increase in the number of VEGF-positive neurons and blood vessels has also been demonstrated in the SN of mice with MPTP-induced neurotoxicity. 111 Wada et al 112 further demonstrated the upregulated expression of VEGF in the SN of patients with PD. In PD animal models, the neuroprotective effects of VEGF are dose dependent. Indeed, low doses of VEGF have been shown to have a neuroprotective effect on DA neurons and result in behavioral improvement, whereas high doses of VEGF induced angiogenesis and glial proliferation. 113 Chronic treatment with l-dopa dose dependently induced the expression of VEGF in the basal ganglia nuclei. However, when coadministered with l-dopa, a small molecule inhibitor of VEGF signaling significantly attenuated the development of dyskinesia and completely blocked the angiogenic response and associated increase in blood–brain barrier permeability induced by the treatment. 114 These findings suggest that VEGF plays a role in the pathophysiology of l-dopa-induced dyskinesia. Moreover, the β-adrenergic receptor agonist isoproterenol significantly increased protein levels of VEGF in human choroidal endothelial cells. 115 The NE treatment increased VEGF levels in cultured nasopharyngeal carcinoma (NPC) tumor cells, and this increase was inhibited by propranolol treatment. The NE also induced invasiveness in all NPC cell lines in a dose-dependent manner, and this induction was blocked by propranolol treatment. 116 Propranolol significantly reduced VEGF activity in a phorbol myristate acetate (PMA)-activated human leukemic cell line. 117 This drug also repressed gastric cancer cell growth through its downstream effects on VEGF. 118,119 Alternatively, NE increased the expression of VEGF, and these effects were inhibited by propranolol treatment in pancreatic cancer cells. 120,121 In addition, epinephrine enhanced the expression of VEGF in colon adenocarcinoma cells. The stimulatory action of epinephrine on colon cancer growth was blocked by treatment with atenolol and ICI 118.551, which are β1- and β2-selective antagonists, respectively. 122 β2-Adrenergic receptor blockade regulated VEGF production in a mouse model of oxygen-induced retinopathy. 123 Hypoxia-inducible factor 1α (HIF-1α) and VEGF mRNA and protein expression were upregulated in a rat model of volume-overload heart failure; these abnormalities were reversed with carvedilol treatment. 124 These findings suggest that β-adrenergic antagonists modulate VEGF expression in PD.
The reduced form of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) enzyme complex mediates critical physiological and pathological processes including cell signaling, inflammation and mitogenesis, through the generation of reactive oxygen species (ROS) from molecular oxygen. The NADPH oxidase is widely expressed in various immune cells, including microglia, macrophages, and neutrophils. Expression of Nox was observed in the nuclei of DA neurons in the SN of patients with PD and animals with 6-OHDA-induced neurotoxicity. 125 In the striatum and nigra, the depletion of DA with reserpine induced a significant increase in the expression of the NADPH subunit p47phox; expression of this subunit decreased with the increasing restoration of DA function. Similarly, 6-OHDA-induced chronic dopaminergic denervation resulted in a significant increase in the expression of p47phox, which was reduced after l-dopa administration. 59 The Nox activation increased zinc-induced DA neurodegeneration and MPPT-, rotenone-, angiotensin-, and paraquat-induced neurotoxicity in animal models of PD 126 –130 The inhibition and knockdown of Nox reduced paraquat-induced ROS generation and DA cell death, 130 and Nox inhibitors conferred protection against lipopolysaccharide (LPS)-induced toxicity, MPPT-induced oxidative stress, and apoptosis in mesencephalic DA neuronal cells. 131,132 NADPH oxidase 1 plays a crucial role in modulating the behavior of α-synuclein expression and aggregation in dopaminergic neurons. 133 The knockdown of cytosolic NADP+-dependent isocitrate dehydrogenase enhanced MPP+-induced oxidative injury in PC12 cells, a model system for neuronal differentiation. 134 Moreover, nebivolol, a third-generation selective β1-adrenoceptor, improved left ventricle dysfunction and survival immediately after myocardial ischemia and inhibited cardiac Nox activation. 135 Nebivolol treatment has been associated with improvements in insulin resistance, reduced proteinuria and reduced Nox activity, and the production of ROS in the kidneys and skeletal muscle tissue of transgenic TG(mRen2)27 rats (Ren2). 136,137 Nebivolol also improved diastolic relaxation, fibrosis, and remodeling in obese Zucker rats and reduced Nox-dependent superoxide production. 138 Carvedilol attenuated the increased expression of Nox subunits in the hearts and kidneys of rats after daunorubicin-induced cardiotoxicity and nephrotoxicity. 139 Activity of Nox in whole blood and isolated neutrophils was dose dependently inhibited by nebivolol, whereas atenolol, metoprolol, and carvedilol were markedly less effective in Watanabe heritable hyperlipidemic rabbits. 140 Celiprolol, a specific β1-antagonist with weak β2-agonistic activity, suppressed Nox p22phox, p47phox, gp91phox, and Nox-1 expression in the left ventricle of deoxycorticosterone acetate-salt hypertensive rats. 141 Taken together, these findings suggest that β-adrenergic antagonists play a role in PD through the suppression of NADPH expression.
The Role of β-Adrenergic Blockers in PD
Matrix metalloproteinases (MMPs) are proteolytic enzymes that are responsible for both extracellular matrix (ECM) remodeling and the regulation of leukocyte migration through the ECM, which is an important step in inflammatory processes. Neuroinflammation significantly contributes to progressive dopaminergic neurodegeneration in PD. The MMPs have been implicated in the degeneration of dopaminergic neurons. Expression of MMP-3 is increased during LPS-induced DA neurotoxicity. 142 The MMP-9 is also elevated in MPTP-induced parkinsonism in mice. 143 The application of dopaminergic neurotoxins to 2 human neuroblastoma cell lines downregulatedthe transcription and translation of tissue inhibitor of metalloproteinase (TIMP)-2, effectively enhancing MMP activity. 144 Exendin 4, which is an analog of glucagon-like peptide 1, significantly attenuated the loss of SN neurons and the striatal dopaminergic fibers in an MPTP-induced PD model and inhibited the expression of MMP-3. 145 Moreover, propranolol inhibited human brain endothelial cell tubulogenesis and MMP-9 secretion. 146 A selective β3-adrenoceptor agonist prevented human myometrial remodeling and MMP-2 and MMP-9 activation in an in vitro model of chorioamnionitis. 147 The NE treatment increased MMP-2 and MMP-9 levels in cultured NPC cells, and these increases were inhibited by propranolol treatment. The NE also induced the invasiveness of all NPC cell lines in a dose-dependent manner; this effect could be blocked with an MMP inhibitor and propranolol treatment. 116 Propranolol significantly reduced MMP-2 activity in a PMA-activated human leukemic cell line. 117 Propranolol-induced growth inhibition has been associated with arrest at both G0/G1 and G2/M and repressed gastric cancer cell growth through the downstream inhibition of MMP-2 and MMP-9. 118 The NE increased MMP-2 and MMP-9 expression, and these effects were inhibited by propranolol treatment in pancreatic cancer cells. 120,121 Epinephrine upregulated MMP-9 activity in human colon adenocarcinoma HT-29 cells, and this effect was blocked by the respective β1- and β2-selective antagonists atenolol and ICI 118.551. 122 These studies suggest that β-adrenergic antagonists might play an important role in the pathological process of PD through the regulation of TIMP levels and the downregulation of MMPs.
The mitogen-activated protein kinase (MAPK) pathways provide a key link between the membrane-bound receptors that receive cues from signaling molecules and changes in the patterns of gene expression, which include the extracellular signal-regulated kinase (ERK) cascade, the stress-activated protein kinases/c-Jun N-terminal kinase cascade, and the p38 MAPK/RK/HOG cascade. 148 Increased cytoplasmic ERK1/2 activity has been observed in the brains of human patients with PD, 149 and degenerating SN neurons typically display phosphorylated-ERK1/2 granules. 150 The activation of ERK1/2 was induced by the neurotoxin 6-OHDA, and the inhibition of ERK activation enhanced neuronal survival. 151,152 The mitochondrial localization of ERK2 activity suggests an effect of 6-OHDA on mitophagy and autophagic cell death in PD. 153 Dysregulation of the autophagy pathway has been observed in the brains of patients with PD and in animal models of PD. 154,155 In addition, the activation of p38 MAPK has been demonstrated in the SN of MPTP-treated mouse models of PD. 156 Moreover, vulnerability to glutamate-induced toxicity in DA neurons is dependent on endogenous DA and MAPK activation. 156 Interestingly, a genetic deficiency in MAPK kinase 2−/ − prevented MPTP-induced neurotoxicity in mouse models of PD. 157 Moreover, β-adrenoceptors stimulation has been shown to activate cAMP/PKA and MAPK pathways in pancreatic cancer cells. β2-Adrenergic antagonists suppressed invasion and proliferation through inhibition of both cAMP/PKA and Ras, which regulate MAPK pathway activation. 121 The NE stimulated pancreatic cancer cell proliferation, migration and invasion via β-adrenergic receptor-dependent activation of the p38/MAPK pathway. These stimulatory effects were completely abolished by propranolol or the p38/MAPK inhibitor SB203580 158 Propranolol was shown to exert its suppressive effects on hemangiomas through the HIF-1α-VEGF-A angiogenesis axis, with effects mediated by the PI3/Akt and p38/MAPK pathways. 159 β-Adrenergic antagonists may play a role in PD through suppression of the MAPK pathway.
Prostaglandins (PGs) play a role in inflammatory processes. 160 Cyclooxygenase (COX) participates in the conversion of arachidonic acid into PGs. Prostaglandin E2 (PGE2) is a key product of COX-2 and is increased in the SN of both patients with PD and animal models of MPTP-induced PD. 161,162 The lack of COX-2 activity attenuated MPTP-related DNA damage. 163 These findings suggest that the loss of genomic integrity can be induced through the concerted actions of COX-2, further supporting a potential contribution of DNA damage to the neurodegeneration in PD. The PGE2 is also directly and selectively toxic to dopaminergic neurons. 164 The PGE2 receptors have been detected on dopaminergic neurons in the rat SN. 164 Overexpression of COX-2 has been reported in PD and an MPTP-animal model. 165 , 166 The COX inhibitors provide neuroprotection in the MPTP-mouse model of PD. 167 Similarly, the regular use of COX-2 inhibiting nonsteroidal anti-inflammatory drugs (NSAIDs), such as ibuprofen, has been associated with a reduced incidence of PD. 168 Moreover, epinephrine increased the release of PGE2 in human colon adenocarcinoma HT-29 cells, which could be blocked by COX-2 inhibitors or atenolol and ICI 118.551 (β1- and β2-selective adrenergic antagonists, respectively). 122 β2-Adrenergic antagonists suppressed COX-2 expression in pancreatic cancer cells. 123 Propranolol inhibited cell proliferation and repressedgastric cancer cell growth through the downstream COX-2 pathway. 118,119 In addition, the administration of propranolol and a COX-2 inhibitor, which can be applied perioperatively in most patients with cancer, with minimal risk and low cost, counteracted several immunologic and endocrinological perturbations and improved recurrence-free survival rates in mice undergoing primary tumor excision. 169,170 These findings suggest that β-adrenergic antagonists may play a role in modulating the inflammatory process in PD.
The ROS play a major role in various cell-signaling pathways. The ROS activates various transcription factors and increases the expression of proteins that control cellular transformation, tumor cell survival, tumor cell proliferation and invasion, angiogenesis, and metastasis. Oxidative stress has been suggested to contribute to the pathogenesis of PD. Lymphocytes from untreated patients with PD exhibit increased oxidative stress. 171 Postmortem analyses of the brains of patients with PD have revealed evidence of increased lipid peroxidation in the SN. 172 –174 Superoxide dismutase is also increased in the SN of patients with PD. 174 Moreover, myocardial tissue sections display increased ROS levels after traumatic brain injuries. Treatment with propranolol reduced cardiac ROS levels. 175 Carvedilol modulated ROS-induced signaling. Carvedilol significantly reduced ischemia–reperfusion-induced free radical production and NAD+ catabolism, lipid peroxidation and red blood cell membrane damage, as determined by free malondialdehyde production in heart perfusion and rheological models. 80 Nebivolol improved diastolic dysfunction and myocardial remodeling through reductions in oxidative stress in the transgenic (mRen2) rat. 176 These findings suggest that β-adrenergic antagonists modulate oxidative stress in PD.
The enzyme nitric oxide synthase (NOS) is involved in the synthesis of nitric oxide (NO), which has also been implicated in PD. Inducible NOS (iNOS), endothelial NOS (eNOS), and neuronal NOS (nNOS) are present in microglia, endothelial cells, and neurons, respectively. 177 Reciprocal DA-glutamate interactions play a critical role in stimulating striatal nNOS activity. 178 The electrical and chemical stimulation of the SN and systemic DA D1/5 receptor agonist administration robustly increases striatal NO efflux 179 , 180 NO signaling exerts multiple effects on local striatal circuits and projection neurons involved in regulating basal ganglia output and nigrostriatal DA neuron activity 181 High levels of NOS expression have been detected postmortem in the nigrostriatal region and basal ganglia of the brains of patients with PD. 182 A significant increase in nitrite content has been observed in the polymorphonuclear leukocytes of patients with PD. 183 Both iNO and nNO are increased in both 6-OHDA and MPTP animal models. 183 –185 A lack of nNOS but not iNOS activity attenuated MPTP-related DNA damage. 163 These results suggest that a loss of genomic integrity can be triggered through the concerted actions of nNOS and further supports the view that DNA damage may contribute to neurodegeneration in PD. Moreover, studies with NOS inhibitors and NOS KO animals have also confirmed the role of NOS in neurodegeneration. 185 –187 Reduced and oxidized glutathione have been detected in the SN of patients with PD. 188 Moreover, metipranolol suppressed nitric oxide-induced lipid peroxidation in the eyes and retinas of rats. 86 Nebivolol prevented vascular NOS III uncoupling in experimental hyperlipidemia. 140 Propranolol suppressed hemangioma growth through the inhibition of eNOS protein expression and the subsequent production of nitric oxide. 189 Celiprolol activated eNOS through the PI3K-Akt pathway via oxidative stress-induced NF-ĸB activity. 141 These findings suggest that β-adrenergic antagonists may play a role in PD via the inhibition of NOS expression.
Conclusion
β-adrenergic receptor blockade may play a role in PD. Genetic studies have identified proteins that link β-adrenergic receptor antagonism to the pathology of PD, including HLA genes, the RAS, PARP-1, NGF, VEGF, and the NADPH. β-adrenergic receptor inhibition also affects PD via nongenomic mechanisms, including MMPs, MAPK pathways, PGs, COX-2, and NOS. The β-adrenergic receptor blockades are contradicted in asthma and patients with CHF and cautioned to nursing women. Depression has been associated with lipophilic β-adrenergic receptor blockades, such as propranolol. Serious CNS adverse effects, including agitation, confusion, and hallucinations, are rare. However, the most interesting side effect of β-adrenergic receptor blockade is hypotension or symptoms associated with hypotension. Thus, further examination of the relationship between β-adrenergic antagonists and PD is required.
Footnotes
The authors declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The authors received no financial support for the research, authorship, and/or publication of this article.
References
- 1. Schapira AH. Future directions in the treatment of Parkinson’s disease. Mov Disord. 2007;22(suppl 17):S385-S391. [DOI] [PubMed] [Google Scholar]
- 2. Asanuma M, Ogawa N, Mizukawa K, Haba K, Hirata H, Mori A. Distribution of the beta-2 adrenergic receptor messenger RNA in the rat brain by in situ hybridization histochemistry: effects of chronic reserpine treatment. Neurochem Res. 1991;16(12):1253-1256. [DOI] [PubMed] [Google Scholar]
- 3. Nicholas AP, Pieribone VA, Hökfelt T. Cellular localization of messenger RNA for beta-1 and beta-2 adrenergic receptors in rat brain: an in situ hybridization study. Neuroscience. 1993;56(4):1023-1039. [DOI] [PubMed] [Google Scholar]
- 4. Lukhanina EP, Kolomiets BP, Pilkevich NA. Effect of stimulation of beta-adrenergic receptors on neuronal activity in primary motor cortex of the rat. Neurosci Lett. 2003;340(2):99-102. [DOI] [PubMed] [Google Scholar]
- 5. Hassani OK, Mouroux M, Feger J. Increased subthalamic neuronal activity after nigral dopaminergic lesion independent of disinhibition via the globus pallidus. Neuroscience. 1996;72(1):105-115. [DOI] [PubMed] [Google Scholar]
- 6. Coenen VA, Gielen FL, Castro-Prado F, Abdel Rahman A, Honey CR. Noradrenergic modulation of subthalamic nucleus activity in human: metoprolol reduces spiking activity in microelectrode recordings during deep brain stimulation surgery for Parkinson’s disease. Acta Neurochir (Wien). 2008;150(8):757-762. [DOI] [PubMed] [Google Scholar]
- 7. Adler LA, Angrist B, Weinreb H, Rotrosen J. Studies on the time course and efficacy of beta-blockers in neuroleptic-induced akathisia and the akathisia of idiopathic Parkinson’s disease. Psychopharmacol Bull. 1991;27(2):107-111. [PubMed] [Google Scholar]
- 8. Barnum CJ, Bhide N, Lindenbach D, et al. Effects of noradrenergic denervation on l-DOPA-induced dyskinesia and its treatment by α- and β-adrenergic receptor antagonists in hemiparkinsonian rats. Pharmacol Biochem Behav. 2012;100(3):607-615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Carpentier AF, Bonnet AM, Vidailhet M, Agid Y. Improvement of levodopa-induced dyskinesia by propranolol in Parkinson’s disease. Neurology. 1996;46(6):1548-1551. [DOI] [PubMed] [Google Scholar]
- 10. Lindenbach D, Ostock CY, Eskow Jaunarajs KL, et al. Behavioral and cellular modulation of l-DOPA-induced dyskinesia by beta-adrenoceptor blockade in the 6-hydroxydopamine-lesioned rat. J Pharmacol Exp Ther. 2011;337(3):755-765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Yoshii F, Shinohara Y, Takeoka T, Kitagawa Y, Akiyama K, Yazaki K. Treatment of essential and parkinsonian tremor with nipradilol. Intern Med. 1996;35(11):861-865. [DOI] [PubMed] [Google Scholar]
- 12. Kuno S, Mizuta E, Nishida J, Takechi M. Therapeutic effects of arotinolol, a beta-adrenergic blocker, on tremor in MPTP-induced parkinsonian monkeys. Clin Neuropharmacol. 1992;15(5):381-386. [DOI] [PubMed] [Google Scholar]
- 13. Foster NL, Newman RP, LeWitt PA, Gillespie MM, Larsen TA, Chase TN. Peripheral beta-adrenergic blockade treatment of parkinsonian tremor. Ann Neurol. 1984;16(4):505-508. [DOI] [PubMed] [Google Scholar]
- 14. Crosby NJ, Deane KH, Clarke CE. Beta-blocker therapy for tremor in Parkinson’s disease. Cochrane Database Syst Rev. 2003;(1):CD003361. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Fiszer U, Mix E, Fredrikson S, Kostulas V, Link H. Parkinson’s disease and immunological abnormalities: increase of HLA-DR expression on monocytes in cerebrospinal fluid and of CD45RO+ T cells in peripheral blood. Acta Neurol Scand. 1994;90(3):160-166. [DOI] [PubMed] [Google Scholar]
- 16. McGeer PL, Itagaki S, Boyes BE, McGeer EG. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer’s disease brains. Neurology. 1988;38(8):1285-1291. [DOI] [PubMed] [Google Scholar]
- 17. McGeer PL, Itagaki S, McGeer EG. Expression of the histocompatibility glycoprotein HLA-DR in neurological disease. Acta Neuropathol. 1988;76(6):550-557. [DOI] [PubMed] [Google Scholar]
- 18. Saiki M, Baker A, Williams-Gray CH, et al. Association of the human leucocyte antigen region with susceptibility to Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2010;81(8):890-891. [DOI] [PubMed] [Google Scholar]
- 19. Sun C, Wei L, Luo F, et al. HLA-DRB1 alleles are associated with the susceptibility to sporadic parkinson’s disease in Chinese Han population. PLoS One. 2012;7(11):e48594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Zhao Y, Gopalai A, Ahmad-Annuar A, et al. Association of HLA locus variant in Parkinson’s disease [published online October 20, 2012]. Clin Genet. 2012. [DOI] [PubMed] [Google Scholar]
- 21. Ahmed I, Tamouza R, Delord M, et al. Association between Parkinson’s disease and the HLA-DRB1 locus. Mov Disord. 2012;27(9):1104-1110. [DOI] [PubMed] [Google Scholar]
- 22. Guo Y, Deng X, Zheng W, et al. HLA rs3129882 variant in Chinese Han patients with late-onset sporadic Parkinson disease. Neurosci Lett. 2011;501(3):185-187. [DOI] [PubMed] [Google Scholar]
- 23. Limas C, Limas CJ, Boudoulas H, et al. Anti-beta-receptor antibodies in familial cardiomyopathy: correlation with HLA-DR and HLA-DQ gene polymorphisms. Am Heart J. 1994;127(2):382-386. [DOI] [PubMed] [Google Scholar]
- 24. Limas CJ, Goldenberg IF, Limas C. Influence of anti-beta-receptor antibodies on cardiac adenylate cyclase in patients with idiopathic dilated cardiomyopathy. Am Heart J. 1990;119(6):1322-1328. [DOI] [PubMed] [Google Scholar]
- 25. Li Q, Milo R, Panitch H, Bever CT, Jr. Effect of propranolol and IFN-beta on the induction of MHC class II expression and cytokine production by IFN-gamma IN THP-1 human monocytic cells. Immunopharmacol Immunotoxicol. 1998;20(1):39-61. [DOI] [PubMed] [Google Scholar]
- 26. Shaw SM, Coppinger T, Waywell C, et al. The effect of beta-blockers on the adaptive immune system in chronic heart failure. Cardiovasc Ther. 2009;27(3):181-186. [DOI] [PubMed] [Google Scholar]
- 27. Philips SR, Davis BA, Durden DA, Boulton AA. Identification and distribution of m-tyramine in the rat. Can J Biochem. 1975;53(1):65-69. [DOI] [PubMed] [Google Scholar]
- 28. Axelrod J, Saavedra JM. Octopamine. Nature. 1977;265(5594):501-504. [DOI] [PubMed] [Google Scholar]
- 29. Zucchi R, Chiellini G, Scanlan TS, Grandy DK. Trace amine-associated receptors and their ligands. Br J Pharmacol. 2006;149(8):967-978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Borowsky B, Adham N, Jones KA, et al. Trace amines: identification of a family of mammalian G protein-coupled receptors. Proc Natl Acad Sci USA. 2001;98(16):8966-8971. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Boulton AA. Identification, distribution, metabolism, and function of meta and para tyramine, phenylethylamine and tryptamine in brain. Adv Biochem Psychopharmacol. 1976;15:57-67. [PubMed] [Google Scholar]
- 32. Paterson IA, Juorio AV, Boulton AA. 2-Phenylethylamine: a modulator of catecholamine transmission in the mammalian central nervous system? J Neurochem. 1990;55(6):1827-1837. [DOI] [PubMed] [Google Scholar]
- 33. Greenshaw A, Juorio AV, Nguyen TV. Depletion of striatal β-phenylethylamine following dopamine but not 5-HT denervation. Brain Res Bull. 1986;17(4):477-484. [DOI] [PubMed] [Google Scholar]
- 34. Richard L, Hauger T, Philskolnick Steven MP. Specific [3H]IS-phenylethylamine binding sited in rat brain. Eur J Pharmacol. 1982;83(1-2):147-148. [DOI] [PubMed] [Google Scholar]
- 35. Vaccari A. High affinity binding of[3H]-tyramine in the central nervous system. Br J Pharmacol. 1986;89(1):15-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Federici M, Geracitano R, Tozzi A, et al. Trace amines depress GABAB response in dopaminergic neurons by inhibiting G-βγ-gated inwardly rectifying potassium channels. Mol Pharmacol. 2005;67(4):1283-1290. [DOI] [PubMed] [Google Scholar]
- 37. Juorio AV, Paterson IA, Zhu MY, Matte G. Electrical stimulation of the substantia nigra and changes of 2-phenylethylamine synthesis in the rat striatum. J Neurochem. 1991;56 (1 ):213-220. [DOI] [PubMed] [Google Scholar]
- 38. Durden DA, Philips SR. Kinetic measurements of the turnover rates of phenylethylamine and tryptamine in vivo in the rat brain. J Neurochem. 1980;34(6):1725-1732. [DOI] [PubMed] [Google Scholar]
- 39. Barroso N, Rodriguez M. Action of beta-phenylethylamine and related amines on nigrostriatal dopamine neurotransmission. Eur J Pharmacol. 1996;297(3):195-203. [DOI] [PubMed] [Google Scholar]
- 40. Geracitano R, Federici M, Prisco S, Bernardi G, Mercuri NB. Inhibitory effects of trace amines on rat midbrain dopaminergic neurons. Neuropharmacology. 2004;46(6):807-814. [DOI] [PubMed] [Google Scholar]
- 41. Bailey BA, Philips SR, Boulton AA. In vivo release of endogenous dopamine, 5-hydroxytryptamine and some of their metabolites from rat caudate nucleus by phenylethylamine. Neurochem Res. 1987;12(2):173-178. [DOI] [PubMed] [Google Scholar]
- 42. Andrew R, Watson DG, Best SA, Midgley JM, Wenlong H, Petty RK. The determination of hydroxydopamines and other trace amines in the urine of parkinsonian patients and normal controls. Neurochem Res. 1993;18(11):1175-1177. [DOI] [PubMed] [Google Scholar]
- 43. D’Andrea G, Nordera G, Pizzolato G, et al. Trace amine metabolism in Parkinson’s disease: low circulating levels of octopamine in early disease stages. Neurosci Lett. 2010;469(3):348-351. [DOI] [PubMed] [Google Scholar]
- 44. Sotnikova TD, Beaulieu JM, Espinoza S, et al. The dopamine metabolite 3-methoxytyramine is a neuromodulator. PLoS One. 2010;5(10):e13452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Ledonne A, Federici M, Giustizieri M, et al. Trace amines depress D2-autoreceptor-mediated responses on midbrain dopaminergic cells. Br J Pharmacol. 2010;160(6):1509-1520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Wolinsky TD, Swanson CJ, Smith KE, et al. The Trace Amine 1 receptor knockout mouse: an animal model with relevance to schizophrenia. Genes Brain Behav. 2007;6(7):628-639. [DOI] [PubMed] [Google Scholar]
- 47. Sotnikova TD, Zorina OI, Ghisi V, Caron MG, Gainetdinov RR. Trace amine associated receptor 1 and movement control. Parkinsonism Relat Disord. 2008;14(suppl 2):S99-S102. [DOI] [PubMed] [Google Scholar]
- 48. Bunzow JR, Sonders MS, Arttamangkul S, et al. Amphetamine, 3,4-methylenedioxymethamphetamine, lysergic acid diethylamide, and metabolites of the catecholamine neurotransmitters are agonists of a rat trace amine receptor. Mol Pharmacol. 2001;60(6):1181-1188. [DOI] [PubMed] [Google Scholar]
- 49. Kleinau G, Pratzka J, Nürnberg D, et al. Differential modulation of Beta-adrenergic receptor signaling by trace amine-associated receptor 1 agonists. PLoS One. 2011;6(10):e27073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Massarsky A, Trudeau VL, Moon TW. β-blockers as endocrine disruptors: the potential effects of human β-blockers on aquatic organisms. J Exp Zool A Ecol Genet Physiol. 2011;315(5):251-265. [DOI] [PubMed] [Google Scholar]
- 51. Changaris DG, Keil LC, Severs WB. Angiotensin II immunohistochemistry of the rat brain. Neuroendocrinol. 1978;25(5):257-274. [DOI] [PubMed] [Google Scholar]
- 52. Healy DP, Printz MP. Distribution of immunoreactive angiotensin II, angiotensin I, angiotensinogen, and renin in the central nervous system of intact and nephrectomized rats. Hypertension. 1984;6(suppl 1):130-136. [DOI] [PubMed] [Google Scholar]
- 53. Ganten D, Hermann K, Bayer D, Unger T, Lang RR. Angiotensin synthesis in the brain and increased turnover in hypertensive rats. Science. 1983;221(4613):869-871. [DOI] [PubMed] [Google Scholar]
- 54. Dzau VJ, Ingelfinger J, Pratt RE, Ellison KE. Identification of renin and angiotensinogen messenger RNA sequences in rat brain. Hypertension. 1986;8(6):544-548. [DOI] [PubMed] [Google Scholar]
- 55. Garrido-Gil P, Valenzuela R, Villar-Cheda B, Lanciego JL, Labandeira-Garcia JL. Expression of angiotensinogen and receptors for angiotensin and prorenin in the monkey and human substantia nigra: an intracellular renin–angiotensin system in the nigra [published online March 11, 2012]. Brain Struct Funct. 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Zubenko GS, Volicer L, Direnfeld LK, Freeman M, Langlais PJ, Nixon RA. Cerebrospinal fluid levels of angiotensin-converting enzyme in Alzheimer’s disease, Parkinson’s disease and progressive supranuclear palsy. Brain Res. 1985;328(2):215-221. [DOI] [PubMed] [Google Scholar]
- 57. Konings CH, Kuiper MA, Bergmans PL, Grijpma AM, van Kamp GJ, Wolters EC. Increased angiotensin-converting enzyme activity in cerebrospinal fluid of treated patients with Parkinson’s disease. Clin Chim Acta. 1994;231(1):101-106. [DOI] [PubMed] [Google Scholar]
- 58. Grammatopoulos TN, Outeiro TF, Hyman BT, Standaert DG. Angiotensin II protects against alpha-synuclein toxicity and reduces protein aggregation in vitro. Biochem Biophys Res Commun. 2007;363(3):846-851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Villar-Cheda B, Rodríguez-Pallares J, Valenzuela R, et al. Nigral and striatal regulation of angiotensin receptor expression by dopamine and angiotensin in rodents: implications for progression of Parkinson’s disease. Eur J Neurosci. 2010;32(10):1695-1706. [DOI] [PubMed] [Google Scholar]
- 60. Mertens B, Varcin M, Michotte Y, Sarre S. The neuroprotective action of candesartan is related to interference with the early stages of 6-hydroxydopamine-induced dopaminergic cell death. Eur J Neurosci. 2011;34(7):1141-1148. [DOI] [PubMed] [Google Scholar]
- 61. Ming M, Wu J, Lachance S, Delalandre A, Carrière S, Chan JS. β-adrenergic receptors and angiotensinogen gene expression in mouse hepatoma cells in vitro. Hypertension. 1995;25(1):105-109. [DOI] [PubMed] [Google Scholar]
- 62. Nakamaru M, Jackson EK, Inagami T. Beta-adrenoceptor-mediated release of angiotensin II from mesenteric arteries. Am J Physiol. 1986;250(1 pt 2):H144-H148. [DOI] [PubMed] [Google Scholar]
- 63. Richards EM, Hermann K, Sumners C, Raizada MK, Phillips MI. Release of immunoreactive angiotensin II from neuronal cultures: adrenergic influences. Am J Physiol. 1989;257:C588-C595. [DOI] [PubMed] [Google Scholar]
- 64. Tang SS, Stevenson L, Dzau VJ. Endothelial renin-angiotensin pathway. Adrenergic regulation of angiotensin secretion. Circ Res. 1990;66(1):103-108. [DOI] [PubMed] [Google Scholar]
- 65. Taddei S, Favilla S, Duranti P, Simonini N, Salvetti A. Vascular renin–angiotensin system and neurotransmission in hypertensive persons. Hypertension. 1991;18(3):266-277. [DOI] [PubMed] [Google Scholar]
- 66. Vilas-Boas WW, Ribeiro-Oliveira A, Jr, Ribeiro Rda C, et al. Effect of propranolol on the splanchnic and peripheral renin–angiotensin system in cirrhotic patients. World J Gastroenterol. 2008;14(44):6824-6830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Saijonmaa O, Nyman T, Fyhrquist F. Carvedilol inhibits basal and stimulated ACE production in human endothelial cells. J Cardiovasc Pharmacol. 2004;43(5):616-621. [DOI] [PubMed] [Google Scholar]
- 68. Cohen Solal A, Jondeau G, Beauvais F, Berdeaux A. Beneficial effects of carvedilol on angiotensin-converting enzyme activity and renin plasma levels in patients with chronic heart failure. Eur J Heart Fail. 2004;6(4):463-466. [DOI] [PubMed] [Google Scholar]
- 69. Itinteang T, Brasch HD, Tan ST, Day DJ. Expression of components of the renin–angiotensin system in proliferating infantile haemangioma may account for the propranolol-induced accelerated involution. J Plast Reconstr Aesthet Surg. 2011;64(4):759-765. [DOI] [PubMed] [Google Scholar]
- 70. Léauté-Labrèze C, Dumas de la Roque E, Hubiche T, Boralevi F, Thambo JB, Taïeb A. Propranolol for severe hemangiomas of infancy. N Engl J Med. 2008;358(24):2649-2651. [DOI] [PubMed] [Google Scholar]
- 71. Sans V, de la Roque ED, Berge J, et al. Propranolol for severe infantile hemangiomas: follow-up report. Pediatrics. 2009;124(3):e423-e431. [DOI] [PubMed] [Google Scholar]
- 72. Soós JJ, Engelhardt JI, Siklós L, Havas L, Majtényi K. The expression of PARP, NF-kappa B and parvalbumin is increased in Parkinson disease. Neuroreport. 2004;15(11):1715-1718. [DOI] [PubMed] [Google Scholar]
- 73. Wang H, Shimoji M, Yu SW, Dawson TM, Dawson VL. Apoptosis inducing factor and PARP-mediated injury in the MPTP mouse model of Parkinson’s disease. Ann N Y Acad Sci. 2003;991:132-139. [DOI] [PubMed] [Google Scholar]
- 74. Mandir AS, Przedborski S, Jackson-Lewis V, et al. Poly(ADP-ribose) polymerase activation mediates 1-methyl-4-phenyl-1, 2,3,6-tetrahydropyridine (MPTP)-induced parkinsonism. Proc Natl Acad Sci USA. 1999;96(10):5774-5779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Chiba-Falek O, Kowalak JA, Smulson ME, Nussbaum RL. Regulation of alphasynuclein expression by poly (ADP ribose) polymerase-1 (PARP-1) binding to the NACP-Rep1 polymorphic site upstream of the SNCA gene. Am J Hum Genet. 2005;76(3):478-492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Iwashita A, Mihara K, Yamazaki S, et al. A new Poly(ADP-ribose) polymerase inhibitor, FR261529 [2-(4-chlorophenyl)-5-quinoxalinecarboxamide], ameliorates methamphetamine-induced dopaminergic neurotoxicity in mice. J Pharmacol Exp Ther. 2004;310(3):1114-1124. [DOI] [PubMed] [Google Scholar]
- 77. Yokoyama H, Kuroiwa H, Tsukada T, Uchida H, Kato H, Araki T. Poly(ADP-ribose) polymerase inhibitor can attenuate the neuronal death after 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced neurotoxicity in mice. J Neurosci Res. 2010;88(7):1522-1536. [DOI] [PubMed] [Google Scholar]
- 78. Outeiro TF, Grammatopoulos TN, Altmann S, et al. Pharmacological inhibition of PARP-1 reduces alpha-synuclein- and MPP+-induced cytotoxicity in Parkinson’s disease in vitro models. Biochem Biophys Res Commun. 2007;357(3):596-602. [DOI] [PubMed] [Google Scholar]
- 79. Infante J, Sánchez-Juan P, Mateo I, et al. Poly(ADP-ribose) polymerase-1 (PARP-1) genetic variants are protective against Parkinson’s disease. J Neurol Sci. 2007;256(1-2):68-70. [DOI] [PubMed] [Google Scholar]
- 80. Li Y, Shi J, Yang BF, et al. Ketamine-induced ventricular structural, sympathetic and electrophysiological remodelling: pathological consequences and protective effects of metoprolol. Br J Pharmacol. 2012;165(6):1748-1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Oláh G, Finnerty CC, Sbrana E, et al. Increased poly(ADP-ribosyl)ation in skeletal muscle tissue of pediatric patients with severe burn injury: prevention by propranolol treatment. Shock. 2011;36(1):18-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Mikami M, Goubaeva F, Song JH, Lee HT, Yang J. beta-Adrenoceptor blockers protect against staurosporine-induced apoptosis in SH-SY5Y neuroblastoma cells. Eur J Pharmacol. 2008;589(1-3):14-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Usta E, Mustafi M, Straub A, Ziemer G. The nonselective beta-blocker carvedilol suppresses apoptosis in human cardiac tissue: a pilot study. Heart Surg Forum. 2010;13(4):E218-E222. [DOI] [PubMed] [Google Scholar]
- 84. Habon T, Szabados E, Kesmarky G, et al. The effect of carvedilol on enhanced ADP-ribosylation and red blood cell membrane damage caused by free radicals. Cardiovasc Res. 2001;52(1):153-160. [DOI] [PubMed] [Google Scholar]
- 85. Strosznajder RP, Jesko H, Dziewulska J. Effect of carvedilol on neuronal survival and poly(ADP-ribose) polymerase activity in hippocampus after transient forebrain ischemia. Acta Neurobiol Exp (Wars). 2005;65(2):137-143. [DOI] [PubMed] [Google Scholar]
- 86. Osborne NN, Wood JP. Metipranolol blunts nitric oxide-induced lipid peroxidation and death of retinal photoreceptors: a comparison with other anti-glaucoma drugs. Invest Ophthalmol Vis Sci. 2004;45(10):3787-3795. [DOI] [PubMed] [Google Scholar]
- 87. Wu CK, Yeh HH. Nerve growth factor rapidly increases muscarinic tone in mouse medial septum/diagonal band of broca. J Neurosci. 2005;25(17):4232-4242. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Poucet B, Herrmann T. Septum and medial frontal cortex contribution to spatial problem-solving. Behav Brain Res. 1990;37(3):269-280. [DOI] [PubMed] [Google Scholar]
- 89. Olton DS, Walker Gage FH. Hippocampal connections and spatial discrimination. Brain Res. 1978;139(2):295-308. [DOI] [PubMed] [Google Scholar]
- 90. Nishio T, Furukawa S, Akiguchi I, Sunohara N. Medial nigral dopamine neurons have rich neurotrophin support in humans. Neuroreport. 1998;9(12):2847-2851. [DOI] [PubMed] [Google Scholar]
- 91. Salini V, Pizzuti A, Falini A, et al. β-nerve growth factor (βNGF) mRNA expression in the parkinsonian adrenal gland. Exp Neurol. 1991;113(2):166-170. [DOI] [PubMed] [Google Scholar]
- 92. Mogi M, Togari A, Kondo T, et al. Brain-derived growth factor and nerve growth factor concentrations are decreased in the substantia nigra in Parkinson’s disease. Neurosci Lett. 1999;270(1):45-48. [DOI] [PubMed] [Google Scholar]
- 93. Pedre LL, Fuentes NP, González LA, et al. Nerve growth factor levels in parkinson disease and experimental parkinsonian rats. Brain Res. 2002;952(1):122-127. [DOI] [PubMed] [Google Scholar]
- 94. Shimoke K, Chiba H. Nerve growth factor prevents 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-induced cell death via the Akt pathway by suppressing caspase-3-like activity using PC12 cell: Relevance to therapeutical application for Parkinson’s disease. J Neurosci Res. 2001;63(5):402-409. [DOI] [PubMed] [Google Scholar]
- 95. Hirata Y, Meguro T, Kiuchi K. Differential effect of nerve growth factor on dopaminergic neurotoxin-induced apoptosis. J Neurochem. 2006;99(2):416-425. [DOI] [PubMed] [Google Scholar]
- 96. Salinas M, Diaz R, Abraham NG, Ruiz de Galarreta CM, Cuadrado A. Nerve growth factor protects against 6-hydroxydopamine-induced oxidative stress by increasing expression of Heme oxygenase-1 in a phosphatidylinositol 3-kinase-dependent manner. J Biol Chem. 2003;278(16):13898-13904. [DOI] [PubMed] [Google Scholar]
- 97. Förander P, Söderström S, Humpel C, Strömberg I. Chronic infusion of nerve growth factor into rat striatum increases cholinergic markers and inhibits striatal neuronal discharge rate. Uro J Neurosci. 1996;8(9):1822-1832. [DOI] [PubMed] [Google Scholar]
- 98. Chaturvedi RK, Shukla S, Seth K, Agrawal AK. Nerve growth factor increases survival of dopaminergic graft, rescue nigral dopaminergic neurons and restores functional deficits in rat model of Parkinson’s disease. Neurosci Lett. 2006;398(1-2):44-49. [DOI] [PubMed] [Google Scholar]
- 99. Li M, Zhang SZ, Guo YW, et al. Human umbilical vein-derived dopaminergic-like cell transplantation with nerve growth factor ameliores motor dysfunction in a rat model of Parkinson’s disease. Neurochem Res. 2010;35(10):1522-1529. [DOI] [PubMed] [Google Scholar]
- 100. Müller T, Hellwig R, Muhlack S. Levodopa induces synthesis of nerve growth factor and growth hormone in patients with Parkinson disease. Clin Neuropharmacol. 2011;34(3):101-103. [DOI] [PubMed] [Google Scholar]
- 101. Samina Riaz S, Tomlinson DR. Pharmacological modulation of nerve growth factor synthesis: a mechanistic comparison of vitamin D receptor and beta2-adrenoceptor agonists. Brain Res Mol Brain Res. 2000;85(1-2):179-188. [DOI] [PubMed] [Google Scholar]
- 102. Schwartz JP. Stimulation of nerve growth factor mRNA content in C6 glioma cells by a beta-adrenergic receptor and by cyclic AMP. Glia. 1988;1(4):282-285. [DOI] [PubMed] [Google Scholar]
- 103. Dal Toso R, De Bernardi MA, Brooker G, Costa E, Mocchetti I. Beta adrenergic and prostaglandin receptor activation increases nerve growth factor mRNA content in C6-2B rat astrocytoma cells. J Pharmacol Exp Ther. 1988;246(3):1190-1193. [PubMed] [Google Scholar]
- 104. Pelegrini-da-Silva A, Oliveira MC, Parada CA, Tambeli CH. Nerve growth factor acts with the beta2-adrenoceptor to induce spontaneous nociceptive behavior during temporomandibular joint inflammatory hyperalgesia. Life Sci. 2008;83(223-24):780-785. [DOI] [PubMed] [Google Scholar]
- 105. Latchaw JP, Little JR, Slugg RM, Lesser RP, Stowe N. Treatment of acute focal cerebral ischemia and recirculation with d-propranolol. Neurosurgery 1985;16(1):18-22. [DOI] [PubMed] [Google Scholar]
- 106. Savitz SI, Erhardt JA, Anthony JV, et al. The novel beta-blocker, carvedilol, provides neuroprotection in transient focal stroke. J Cereb Blood Flow Metab. 2000;20(8):1197-1204. [DOI] [PubMed] [Google Scholar]
- 107. Goyagi T, Kimura T, Nishikawa T, Tobe Y, Masaki Y. Beta-adrenoreceptor antagonists attenuate brain injury after transient focal ischemia in rats. Anesth Analg. 2006;103(3):658- 663. [DOI] [PubMed] [Google Scholar]
- 108. Han RQ, Ouyang YB, Xu L, Agrawal R, Patterson AJ, Giffard RG. Postischemic brain injury is attenuated in mice lacking the beta2-adrenergic receptor. Anesth Analg. 2009;108(1):280-287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Yasuhara T, Shingo T, Kobayashi K, et al. Neuroprotective effects of vascular endothelial growth factor (VEGF) upon dopaminergic neurons in a rat model of Parkinson’s disease. Eur J Neurosci. 2004;19(6):1494-1504. [DOI] [PubMed] [Google Scholar]
- 110. Falk T, Zhang S, Sherman SJ. Vascular endothelial growth factor B (VEGF-B) is up-regulated and exogenous VEGF-B is neuroprotective in a culture model of Parkinson’s disease. Mol Neurodegener. 2009;4:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Barcia C, Bautista V, Sánchez-Bahillo A, et al. Changes in vascularization in substantia nigra pars compacta of monkeys rendered parkinsonian. J Neural Transm. 2005;112(9):1237-1348. [DOI] [PubMed] [Google Scholar]
- 112. Wada K, Arai H, Takanashi M, et al. Expression levels of vascular endothelial growth factor and its receptors in Parkinson’s disease. Neuroreport. 2006;17(7):705-709. [DOI] [PubMed] [Google Scholar]
- 113. Yasuhara T, Shingo T, Muraoka K, et al. The differences between high and low-dose administration of VEGF to dopaminergic neurons of in vitro and in vivo Parkinson’s disease model. Brain Res. 2005;1038(1):1-10. [DOI] [PubMed] [Google Scholar]
- 114. Ohlin KE, Francardo V, Lindgren HS, et al. Vascular endothelial growth factor is upregulated by l-dopa in the parkinsonian brain: implications for the development of dyskinesia. Brain. 2011;134(pt 8):2339-2357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Steinle JJ, Cappocia FC, Jr, Jiang Y. Beta-adrenergic receptor regulation of growth factor protein levels in human choroidal endothelial cells. Growth Factors. 2008;26(6):325-330. [DOI] [PubMed] [Google Scholar]
- 116. Yang EV, Sood AK, Chen M, et al. Norepinephrine up-regulates the expression of vascular endothelial growth factor, matrix metalloproteinase (MMP)-2, and MMP-9 in nasopharyngeal carcinoma tumor cells. Cancer Res. 2006;66(21):10357-10364. [DOI] [PubMed] [Google Scholar]
- 117. Hajighasemi F, Hajighasemi S. Effect of propranolol on angiogenic factors in human hematopoietic cell lines in vitro. Iran Biomed J. 2009;13(4):223-228. [PubMed] [Google Scholar]
- 118. Liao X, Che X, Zhao W, Zhang D, Bi T, Wang G. The β-adrenoceptor antagonist, propranolol, induces human gastric cancer cell apoptosis and cell cycle arrest via inhibiting nuclear factor κB signaling. Oncol Rep. 2010;24(6):1669-1676. [DOI] [PubMed] [Google Scholar]
- 119. Liao X, Che X, Zhao W, et al. Effects of propranolol in combination with radiation on apoptosis and survival of gastric cancer cells in vitro. Radiat Oncol. 2010;5:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Guo K, Ma Q, Wang L, et al. Norepinephrine-induced invasion by pancreatic cancer cells is inhibited by propranolol. Oncol Rep. 2009;22(4):825-830. [DOI] [PubMed] [Google Scholar]
- 121. Zhang D, Ma QY, Hu HT, Zhang M. β2-adrenergic antagonists suppress pancreatic cancer cell invasion by inhibiting CREB, NFκB and AP-1. Cancer Biol Ther. 2010;10(1):19-29. [DOI] [PubMed] [Google Scholar]
- 122. Wong HP, Ho JW, Koo MW, et al. Effects of adrenaline in human colon adenocarcinoma HT-29 cells. Life Sci. 2011;88(25-26):1108-1112. [DOI] [PubMed] [Google Scholar]
- 123. Martini D, Monte MD, Ristori C, et al. Antiangiogenic effects of β2-adrenergic receptor blockade in a mouse model of oxygen-induced retinopathy. J Neurochem. 2011;119(6):1317-1329. [DOI] [PubMed] [Google Scholar]
- 124. Shyu KG, Lu MJ, Chang H, Sun HY, Wang BW, Kuan P. Carvedilol modulates the expression of hypoxia-inducible factor-1alpha and vascular endothelial growth factor in a rat model of volume-overload heart failure. J Card Fail. 2005;11(2):152-159. [DOI] [PubMed] [Google Scholar]
- 125. Choi DH, Cristóvão AC, Guhathakurta S, et al. NADPH oxidase 1-mediated oxidative stress leads to dopamine neuron death in Parkinson’s disease. Antioxid Redox Signal. 2012;16(10):1033-1045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Kumar A, Singh BK, Ahmad I, et al. Involvement of NADPH oxidase and glutathione in zinc-induced dopaminergic neurodegeneration in rats: similarity with paraquat neurotoxicity. Brain Res. 2012;1438:48-64. [DOI] [PubMed] [Google Scholar]
- 127. Zawada WM, Banninger GP, Thornton J, et al. Generation of reactive oxygen species in 1-methyl-4-phenylpyridinium (MPP+) treated dopaminergic neurons occurs as an NADPH oxidase-dependent two-wave cascade. J Neuroinflammation. 2011;8:129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Zhou H, Zhang F, Chen SH, et al. Rotenone activates phagocyte NADPH oxidase by binding to its membrane subunit gp91phox. Free Radic Biol Med. 2012;52(2):303-313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Rodriguez-Pallares J, Rey P, Parga JA, Muñoz A, Guerra MJ, Labandeira-Garcia JL. Brain angiotensin enhances dopaminergic cell death via microglial activation and NADPH-derived ROS. Neurobiol Dis. 2008;31(1):58-73. [DOI] [PubMed] [Google Scholar]
- 130. Cristóvão AC, Choi DH, Baltazar G, Beal MF, Kim YS. The role of NADPH oxidase 1-derived reactive oxygen species in paraquat-mediated dopaminergic cell death. Antioxid Redox Signal. 2009;11(9):2105-2118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Anantharam V, Kaul S, Song C, Kanthasamy A, Kanthasamy AG. Pharmacological inhibition of neuronal NADPH oxidase protects against 1-methyl-4-phenylpyridinium (MPP+)-induced oxidative stress and apoptosis in mesencephalic dopaminergic neuronal cells. Neurotoxicology. 2007;28(5):988-997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Zhang D, Hu X, Wei SJ, et al. Squamosamide derivative FLZ protects dopaminergic neurons against inflammation-mediated neurodegeneration through the inhibition of NADPH oxidase activity. J Neuroinflammation. 2008;5:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Cristóvão AC, Guhathakurta S, Bok E, et al. NADPH Oxidase 1 Mediates α-synucleinopathy in Parkinson’s disease. J Neurosci. 2012;32(42):14465-14477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Yang ES, Park JW. Knockdown of cytosolic NADP+-dependent isocitrate dehydrogenase enhances MPP+-induced oxidative injury in PC12 cells. BMB Rep. 2011;44(5):312-316. [DOI] [PubMed] [Google Scholar]
- 135. Sorrentino SA, Doerries C, Manes C, et al. Nebivolol exerts beneficial effects on endothelial function, early endothelial progenitor cells, myocardial neovascularization, and left ventricular dysfunction early after myocardial infarction beyond conventional β1-blockade. J Am Coll Cardiol. 2011;57(5):601-611. [DOI] [PubMed] [Google Scholar]
- 136. Manrique C, Lastra G, Habibi J, et al. Nebivolol improves insulin sensitivity in the TGR(Ren2)27 rat. Metabolism. 2011;60(12):1757-1766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Whaley-Connell A, Habibi J, Johnson M, et al. Nebivolol reduces proteinuria and renal NADPH oxidase-generated reactive oxygen species in the transgenic Ren2 rat. Am J Nephrol. 2009;30(4):354-360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138. Zhou X, Ma L, Habibi J, et al. Nebivolol improves diastolic dysfunction and myocardial remodeling through reductions in oxidative stress in the Zucker obese rat. Hypertension. 2010;55(4):880-888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Arozal W, Watanabe K, Veeraveedu PT, et al. Protective effect of carvedilol on daunorubicin-induced cardiotoxicity and nephrotoxicity in rats. Toxicology. 2010;274(1-3):18-26. [DOI] [PubMed] [Google Scholar]
- 140. Mollnau H, Schulz E, Daiber A, et al. Nebivolol prevents vascular NOS III uncoupling in experimental hyperlipidemia and inhibits NADPH oxidase activity in inflammatory cells. Arterioscler Thromb Vasc Biol. 2003;23(4):615-621. [DOI] [PubMed] [Google Scholar]
- 141. Kobayashi N, Mita S, Yoshida K, et al. Celiprolol activates eNOS through the PI3K-Akt pathway and inhibits VCAM-1 Via NF-kappaB induced by oxidative stress. Hypertension. 2003;42(5):1004-1013. [DOI] [PubMed] [Google Scholar]
- 142. McClain JA, Phillips LL, Fillmore HL. Increased MMP-3 and CTGF expression during lipopolysaccharide-induced dopaminergic neurodegeneration. Neurosci Lett. 2009;460(1):27-31. [DOI] [PubMed] [Google Scholar]
- 143. Lorenzl S, Caligasan N, Yang L, et al. Matrix metalloproteinase-9 is elevated in 1-methyl-4-1,2,3,6-tetrahydropyridine-induced parkinsonism in mice. Neuromolecular Med. 2004;5(2):119-132. [DOI] [PubMed] [Google Scholar]
- 144. Kim SY, Woo MS, Park JS, Huyn JW, Kim YS, Kim HS. The neuroprotective role of tissue inhibitor of metalloproteinase-2 in MMP+- or 6-OHDA-treated SK-N-BE(2)C and SH-SY5Y human neuroblastoma cells. Neurosci Lett. 2010;468(2):136-140. [DOI] [PubMed] [Google Scholar]
- 145. Kim S, Moon M, Park S. Exendin-4 protects dopaminergic neurons by inhibition of microglial activation and matrix metalloproteinase-3 expression in an animal model of Parkinson’s disease. J Endocrinol. 2009;202(3):431-439. [DOI] [PubMed] [Google Scholar]
- 146. Annabi B, Lachambre MP, Plouffe K, Moumdjian R, Béliveau R. Propranolol adrenergic blockade inhibits human brain endothelial cells tubulogenesis and matrix metalloproteinase-9 secretion. Pharmacol Res. 2009;60(5):438-445. [DOI] [PubMed] [Google Scholar]
- 147. Lirussi F, O’Brien M, Wendremaire M, et al. SAR150640, a selective beta3-adrenoceptor agonist, prevents human myometrial remodelling and activation of matrix metalloproteinase in an in vitro model of chorioamnionitis. Br J Pharmacol. 2010;159(6):1354-1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Hipskind RA, Bilbe G. MAP kinase signaling cascades and gene expression in osteoblasts. Front Biosci. 1998;3:d804-d816. [DOI] [PubMed] [Google Scholar]
- 149. Zhu JH, Kulich SM, Oury TD, Chu CT. Cytoplasmic aggregates of phosphorylated extracellular signal-regulated protein kinases in Lewy body diseases. Am J Pathol. 2002;161(6):2087-2098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Zhu JH, Guo F, Shelburne J, Watkins S, Chu CT. Localization of phosphorylated ERK/MAP kinases to mitochondria and autophagosomes in Lewy body diseases. Brain Pathol. 2003;13(4):473-481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. Kulich SM, Chu CT. Sustained extracellular signal-regulated kinase activation by 6-hydroxydopamine: implications for Parkinson’s disease. J Neurochem. 2001;77(4):1058-1066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Kulich SM, Horbinski C, Patel M, Chu CT. 6-Hydroxydopamine induces mitochondrial ERK activation. Free Radic Biol Med. 2007;43(3):372-383. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Dagda RK, Zhu J, Kulich SM, Chu CT. Mitochondrially localized ERK2 regulates mitophagy and autophagic cell stress: implications for Parkinson’s disease. Autophagy. 2008;4(6):770-782. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Lynch-Day MA, Mao K, Wang K, Zhao M, Klionsky DJ. The role of autophagy in Parkinson’s disease. Cold Spring Harb Perspect Med. 2012;2(4):a009357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Karunakaran S, Ravindranath V. Activation of p38 MAPK in the substantia nigra leads to nuclear translocation of NF-kappaB in MPTP-treated mice: implication in Parkinson’s disease. J Neurochem. 2009;109(6):17911-17919. [DOI] [PubMed] [Google Scholar]
- 156. Izumi Y, Yamamoto N, Matsuo T, et al. Vulnerability to glutamate toxicity of dopaminergic neurons is dependent on endogenous dopamine and MAPK activation. J Neurochem. 2009;110(2):745-755. [DOI] [PubMed] [Google Scholar]
- 157. Thomas T, Timmer M, Cesnulevicius K, Hitti E, Kotlyarov A, Gaestel M. MAPKAP kinase 2-deficiency prevents neurons from cell death by reducing neuroinflammation--relevance in a mouse model of Parkinson’s disease. J Neurochem. 2008;105(5):2039-2052. [DOI] [PubMed] [Google Scholar]
- 158. Huang XY, Wang HC, Yuan Z, Huang J, Zheng Q. Norepinephrine stimulates pancreatic cancer cell proliferation, migration and invasion via β-adrenergic receptor-dependent activation of P38/MAPK pathway. Hepatogastroenterology. 2012;59(115):889-893. [DOI] [PubMed] [Google Scholar]
- 159. Chim H, Armijo BS, Miller E, Gliniak C, Serret MA, Gosain AK. Propranolol induces regression of hemangioma cells through HIF-1α-mediated inhibition of VEGF-A. Ann Surg. 2012;256(1):146-156. [DOI] [PubMed] [Google Scholar]
- 160. Ricciotti E, Fitzgerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol. 2011;31(5):986-1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Mattammal MB, Strong R, Lakshmi VM, Chung HD, Stephenson AH. Prostaglandin H synthetase-mediated metabolism of dopamine: implication for Parkinson’s disease. J Neurochem. 1995;64(4):1645-1654. [DOI] [PubMed] [Google Scholar]
- 162. Wang T, Pei Z, Zhang W, et al. MMP-induced COX-2 activation and subsequent neurodegeneration. FASEB J. 2005;19(9):1134-1136. [DOI] [PubMed] [Google Scholar]
- 163. Hoang T, Choi DK, Nagai M, et al. Neuronal NOS and cyclooxygenase-2 contribute to DNA damage in a mouse model of Parkinson disease. Free Radic Biol Med. 2009;47(7):1049-1056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Carrasco E, Casper D, Werner P. PGE2 receptor EP1 renders dopaminergic neurons selectively vulnerable to low-level oxidative stress and direct PGE2 neurotoxicity. J Neurosci Res. 2007;85(14):3109-3117. [DOI] [PubMed] [Google Scholar]
- 164. Knott C, Stern G, Wilkin GP. Inflammatory regulators in Parkinson’s disease: iNOS, lipocortin-1, and cyclooxygenases-1 and -2. Mol Cell Neurosci. 2000;16(6):724-739. [DOI] [PubMed] [Google Scholar]
- 166. Teishmann P, Tieu K, Choi DK, et al. Cyclooxygenase-2 is instrumental in Parkinson’s disease neurodegeneration. Proc Natl Acad Sci U S A. 2003;100(9):5473-5478. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Teishmann P, Ferger B. Inhibiton of the cyclooxygenase isoenzymes COX-1 and COX-2 provide neuroprotection in the MPTP-mouse model of Parkinson’s disease. Synapse. 2001;39(2):167-174. [DOI] [PubMed] [Google Scholar]
- 168. Chen H, Zhang SM, Hernan MA, et al. Nonsteroidal anti-inflammatory drugs and the risk of Parkinson disease. Arch Neurol. 2003;60(8):1059-1064. [DOI] [PubMed] [Google Scholar]
- 169. Glasner A, Avraham R, Rosenne E, et al. Improving survival rates in two models of spontaneous postoperative metastasis in mice by combined administration of a beta-adrenergic antagonist and a cyclooxygenase-2 inhibitor. J Immunol. 2010;184(5):2449-2457. [DOI] [PubMed] [Google Scholar]
- 170. Benish M, Bartal I, Goldfarb Y, et L. Perioperative use of beta-blockers and COX-2 inhibitors may improve immune competence and reduce the risk of tumor metastasis. Ann Surg Oncol. 2008;15(7):2042-2052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Prigione A, Isaias IU, Galbussera A, Begni B, Pezzoli G, Ferrarese C. Increased oxidative stress in lymphocytes from untreated Parkinson’s disease. Parkinsonism Related Disord. 2009;15(4):327-328. [DOI] [PubMed] [Google Scholar]
- 172. Dexter DT, Carter CJ, Wells FR, et al. Basal lipid peroxidation in substantia nigra is increased in Parkinson’s disease. J Neurochem. 1989;52(2):381-389. [DOI] [PubMed] [Google Scholar]
- 173. Dexter DT, Holley AE, Flitter WD, et al. Increased levels of lipid hydroperoxides in the parkinsonian substantia nigra: an HPLC and ESR study. Mov Disord. 1994;9(1):92-97. [DOI] [PubMed] [Google Scholar]
- 174. Saggu H, Cooksey J, Dexter D, et al. A selective increase in particulate superoxide dismutase activity in parkinsonian substantia nigra. Neurochem. 1989;53(3):692-697. [DOI] [PubMed] [Google Scholar]
- 175. Larson BE, Stockwell DW, Boas S, et al. Cardiac reactive oxygen species after traumatic brain injury. J Surg Res. 2012;173(2): e73-e81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Ma L, Gul R, Habibi J, et al. Nebivolol improves diastolic dysfunction and myocardial remodeling through reductions in oxidative stress in the transgenic (mRen2) rat. Am J Physiol Heart Circ Physiol. 2012;302(11): H2341-H2351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Nathan C, Xie QW. Regulation of biosynthesis of nitric oxide. J Biol Chem. 1994;269(19):13725-13728. [PubMed] [Google Scholar]
- 178. Park DJ, West AR. Regulation of striatal nitric oxide synthesis by local dopamine and glutamate interactions. J Neurochem. 2009;111(6):1457-1465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Sammut S, Bray KE, West AR. Dopamine D2 receptor-dependent modulation of striatal NO synthase activity. Psychopharmacology (Berl). 2007;191(3):793-803. [DOI] [PubMed] [Google Scholar]
- 180. Sammut S, Park DJ, West AR. Frontal cortical afferents facilitate striatal nitric oxide transmission in vivo via a NMDA receptor and neuronal NOS-dependent mechanism. J Neurochem. 2007;103(3):1145-1156. [DOI] [PubMed] [Google Scholar]
- 181. West AR, Galloway MP, Grace AA. Regulation of striatal dopamine neurotransmission by nitric oxide: effector pathways and signaling mechanisms. Synapse. 2002;44(4):227-245. [DOI] [PubMed] [Google Scholar]
- 182. Hunot S, Boissiere F, Faucheux B, et al. Nitric oxide synthase and neuronal vulnerability in Parkinson’s disease. Neurosci. 1996;72(2):355-363. [DOI] [PubMed] [Google Scholar]
- 183. Barthwal MK, Srivastava N, Shukla R, et al. Polymorphonuclear leukocyte nitrate content and antioxidant enzymes in Parkinson’s disease. Acta Neurol Scand. 1999;100(5):300-304. [DOI] [PubMed] [Google Scholar]
- 184. Singh S, Das T, Ravindran A, et al. Involvement of nitric oxide in neurodegeneration: a study on the experimental models of Parkinson’s disease. Redox Rep. 2005;10(2):103-109. [DOI] [PubMed] [Google Scholar]
- 185. Muramatsu Y Kurosaki R, Wanatabe H, et al. cerebral alterations in a MPTP-mouse model of Parkinson’s disease-an immunocytochemical study. J Neural Transm. 2003;110(10):1129-1144. [DOI] [PubMed] [Google Scholar]
- 186. Schulz JB, Mathews RT, Muqit MM, Browne SE, Beal MF. Inhibition of neuronal nitric acid synthase by 7-nitroindazole protect against MPTP-induced neurotoxicity in mice. J Neurochem. 1995;64(2):936-939. [DOI] [PubMed] [Google Scholar]
- 187. Dehmer T, Lindenau J, Haid S, Dichgans J, Schulz JB. Deficiency of inducible nitric oxide synthase protects against MPTP toxicity in vivo. J Neurochem. 2000;74(5):2213-2216. [DOI] [PubMed] [Google Scholar]
- 188. Kaur D, Lee D, Ragapolan S, Andersen JK. Glutathione depletion in immortalized midbrain-derived dopaminergic neurons results in increases in the labile iron pool: implications for Parkinson’s disease. Free Radic Biol Med. 2009;46(5):593-598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189. Dai Y, Hou F, Buckmiller L, et al. Decreased eNOS protein expression in involuting and propranolol-treated hemangiomas. Arch Otolaryngol Head Neck Surg. 2012;138(2):177-182. [DOI] [PubMed] [Google Scholar]
