Skip to main content
American Journal of Alzheimer's Disease and Other Dementias logoLink to American Journal of Alzheimer's Disease and Other Dementias
. 2013 May 20;28(5):427–439. doi: 10.1177/1533317513488924

The Role of Beta-Adrenergic Receptor Blockers in Alzheimer’s Disease

Potential Genetic and Cellular Signaling Mechanisms

Khanh vinh quốc Lương 1,, Lan Thi Hoàng Nguyễn 1
PMCID: PMC10852699  PMID: 23689075

Abstract

According to genetic studies, Alzheimer’s disease (AD) is linked to beta-adrenergic receptor blockade through numerous factors, 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. Beta-adrenergic receptor blockade is also implicated in AD due to its effects on matrix metalloproteinases, mitogen-activated protein kinase pathways, prostaglandins, cyclooxygenase-2, and nitric oxide synthase. Beta-adrenergic receptor blockade may also have a significant role in AD, although the role is controversial. Behavioral symptoms, sex, or genetic factors, including Beta 2-adrenergic receptor variants, apolipoprotein E, and cytochrome P450 CYP2D6, may contribute to beta-adrenergic receptor blockade modulation in AD. Thus, the characterization of beta-adrenergic receptor blockade in patients with AD is needed.

Keywords: beta-adrenergic receptor blocker, Alzheimer’s disease, dementia, beta-adrenergic receptor antagonism

Introduction

Alzheimer’s disease (AD) is the most common form of dementia in the elderly individualsand is associated with progressive memory loss and cognitive dysfunction. The AD is associated with beta-adrenergic receptors. In the brain, beta-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. 1,2 The adrenergic receptors (or adrenoceptors) are a class of G-protein-coupled receptors that are targets of the catecholamines, especially norepinephrine (noradrenaline) and epinephrine (adrenaline). Many cells possess adrenergic receptors, and the binding of a catecholamine to these receptors will generally stimulate the sympathetic nervous system. There are 2 main groups of adrenergic receptors, α and β. Beta receptors have the subtypes beta1, beta2, and beta3. All 3 beta subtype receptors are linked to Gs proteins (although beta2 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 (PKA), which mediates some of the intracellular events following hormone binding. Amyloid beta peptide (Aβ) induces subtle alterations in the synaptic function in AD. The Aβ interacts with beta2 adrenergic receptors in the central noradrenergic system to regulate synaptic functions in the prefrontal cortical neurons and induces the internalization and degradation of the beta2-adrenergic receptor that results in the impairment of adrenergic and glutamatergic activities. 3,4 Beta2-adrenergic receptors play an important role in AD. Compared with the thalamus of control brains, the thalamus of the brains with dementia had a lower total concentration of beta-adrenergic receptors. Compared with the control brains, brain with dementia have significantly lower concentrations of beta1-adrenergic receptor in the hippocampus and higher concentrations in the nucleus basalis of Meynert (NbM) and cerebellar hemisphere, whereas brains wirh dementia have lower concentrations of beta2-adrenergic receptor concentrations in the thalamus, NbM, and cerebellar hemispheres and higher concentrations in the hippocampus and putamen. 5 Compared with non-AD patients, patients with AD have lymphocytes that have lower beta2-adrenergic receptor levels and lower levels of beta2-adrenergic-stimulated cAMP. 6 Fibroblasts isolated from patients with AD have a reduced beta2-adrenergic receptor response. 7 Karczewski et al 8 demonstrated the presence of agonistic autoantibodies directed at adrenergic receptors in the circulation of patients with mild-to-moderate Alzheimer’s and vascular dementia. Beta-adrenoceptors mediate the ability of norepinephrine (NE) to differentially modulate Aβ1-42-induced immune responses. The NE suppresses Aβ1-42-mediated cytotoxicity and monocytic chemotactic protein 1 secretion but enhances Aβ-mediated IL-1β secretion via beta-adrenoceptor activity combined with the activating of cAMP/protein kinase A pathway and cAMP response element binding in human microglia-like THP-1 cells. 9 In addition, reduced levels of NE are associated with behavioral phenotypes observed in a TgCRND8 mouse model of AD. 10 The NE promotes murine microglial uptake and degradation of Aβ. 11 Moreover, the beta3-adrenergic receptor agonist (CL316243), but not the beta2-adrenergic receptor agonist, rescued this Aβ-induced memory loss. 12 The beta2-adrenergic agonist clenbuterol improved the performance of many of the young and aged rats and monkeys that had performed poorly under control conditions. 13 The degeneration of locus ceruleus neurons and reduced levels of NE potentiated Aβ-induced cortical inflammation. 14 Moreover, patients with cognitive impairment who were on beta2-adrenergic receptor blockers had poorer delayed memory retrieval. 15 However, compared with both nonaggessive patients with AD and control participants, agrressive patients with AD had small but significant increases (approximately 25%) in beta1- and beta2-adrenergic receptors of the cerebellar cortex. 16 Patients with AD have larger total numbers of beta2- and beta1-adrenoceptors in the hippocampus. By contrast, in the AD putamen, where beta1-receptors were highly expressed, the total numbers of beta- and beta1-receptors were significantly reduced with no consistent change in the number of beta2-receptors. 17 Furthermore, compared with cerebellar AD and control tissues, the hippocampal has higher total beta adrenoceptor density. 18 The AD has significantly higher total number of beta receptors of the cerebral microvessels and numbers of beta2-receptors, which is the type that is predominately expressed in microvessels. 19 Activation of the beta2-adrenergic receptor stimulates γ-secretase activity and accelerates amyloid plaque formation. The beta2-adrenergic receptor-selective antagonist ICI 118,551 reduced Aβ peptide production, 20,21 suggesting that blockade of beta2-adrenergic receptor function might be effective in the prevention and treatment of AD. The use of beta2-adrenergic receptor antagonists correlated with a decreased incidence of AD among patients with hypertension. 2224 Propranolol reduced aggression and agitation in patients with senile dementia. 2528 Propranolol also restored cognitive deficits and improved amyloid and tau pathologies in a senescence-accelerated mouse model. 29,30 Carvedilol, a nonselective beta-adrenergic receptor blocker, demonstrated a neuroprotective effect in colchicine- and aluminum chloride-induced cognitive dysfunction and oxidative damage. 31,32 Carvedilol also significantly attenuated brain oligomeric beta-amyloid content and cognitive deterioration in 2 independent AD mouse models. 33 In addition, nebivolol is highly tolerable and safe and can significantly reduce amyloid neuropathology in the brain, which could be one of the most important parameters for primary prevention of AD. 34 These findings suggested that beta-adrenergic receptor blockade may play a role in AD. Thus, we discuss the potential role of beta-adrenergic receptor blockers in AD.

Genetic Factors Associated With Beta-Adrenergic Inhibition and AD

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.

Previous 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 AD. Previous studies showed that HLA-DR1 was associated with enhanced cumulative recall ability, and conversely, HLA-DR5 was associated with a diminished delayed verbal recall and spatial recall abilities on cognitive abilities in an older nondemented population. 35 Brains with AD have increased MHC class II glycoprotein expression on microglial cells. 3639 Furthermore, the AD retina has a significantly increased level of MHC class II expression. 40 HLA-DR was abnormally expressed in the neutrophils and monocytes of patients with AD. 41 Moreover, the postmortem brains of patients with AD had increased numbers of HLA-DR and interleukin 2 (IL-2)-receptor-positive cells, which were correlated with the number of senile plaques. 42 Shalit et al 43 observed a slight increase in HLA-DR levels in the mild stage of AD without changes in CD4, CD8, and IL-2 levels. In the moderately severe stage of AD, however, HLA-DR and CD4 levels increased, and CD8 levels slightly increased, suggesting that the peripheral immune reaction in AD may be correlated with the clinical stage of the disease. Furthermore, following long-term therapeutic immunization of an AD mouse model carring the DRB1*1501 allele, Aβ were effectively cleared from the brain parenchyma, and brain microglial activation was reduced. 44 These results suggested that HLA-DR alleles are directly associated with specific Aβ T-cell epitopes with highly immunogenic properties of the abundant DRB1*1501 allele in this mouse model of AD. Moreover, HLA-DR and HLA-DQ gene polymorphisms may be correlated with the anti-beta-receptor antibodies in familial cardiomyopathy. 45 Cardiac beta-adrenergic receptors and adenylate cyclase activity in dilated cardiomyopathy are modulated by circulating autoantibodies against the cardiac beta1-adrenoceptor, the presence of which is regulated by the HLA-DR. 46 In addition, propranolol abrogated the interferon-gamma-induced increases in HLA class II expression and interleukin-1beta (IL-1β) secretion. 47 The lymphocytes of carvedilol-treated chronic heart failure (CHF) patients have significantly reduced HLA-DR expression. 48 These findings suggested that beta-adrenergic receptor blockers might affect AD via the suppression of MHC class II antigen expression.

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 central nervous system (CNS), 4952 suggesting that the RAS might be involved in brain activity. Increasing evidence suggests that specific components of the RAS may have a crucial role in learning and memory processes. Angiotensin-converting enzyme (ACE) activity was reported in the homogenates of postmortem brain tissue from patients with AD and was correlated with Aβ plaque load. 53 The increased binding of radioactively labeled ACE inhibitor to ACE was demonstrated in AD temporal cortices. 54 Another report also demonstrated elevated neuronal and perivascular ACE immune-reactivity in AD parietal cortices. 55 Recently, ACE activity was found to be increased in the peripheral blood of patients with late-onset AD; however, ACE activity was not correlated with the level of Aβ in peripheral blood. 56 Thus, the role of ACE in AD remains controversial as well as the role of beta-adrenergic receptor antagonisms in AD; ACE inhibits Aβ aggregation and lowers the levels of secreted Aβ in living cells, an effect that is blocked with ACE inhibitor. 57,58 By contrast, in another study, the ACE inhibitor did not have an effect on cerebral Aβ levels and plaque deposition in vivo. 59 Although short-term treatment with ACE inhibitors failed to increase Aβ formation in the brain, long-term treatment enhanced the Aβ deposition in aged amyloid precursor protein (APP) transgenic mice. 60 Moreover, treatment with RAS blockers modulated serum adipocytokines and glucose homeostasis, thereby potentially slowing the cognitive decline in patients with AD. 61 The angiotensin receptor blocker losartan also exerted direct neuroprotective effects via its Aβ-reducing and anti-inflammatory effects in the CNS. 62 Furthermore, the renin inhibitor aliskiren conferred neuronal resistance to Aβ toxicity in primary rat cortical cultures. 63 The ACE I/I genotype and I allele showed an increased risk of AD, 64,65 but the D/D genotype was associated with a reduced risk. 66 Compared with the D/D genotype, the I/I genotype is linked to smaller volumes of the hippocampus and the amygdala 67 and has increased brain Aβ42 load. 68 Moreover, catecholamines altered the release of AT II (Angiotensinogen II). Ming et al 69 demonstrated that isoproterenol enhanced the stimulatory effect of dexamethasone on AT gene expression via β2-adrenergic receptors in mouse hepatoma cells. In addition, isoproterenol promoted an increase in the release of AT II from isolated perfused mesenteric arteries, and this release was blocked by propranolol treatment. 70 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. 7173 Compared with nontreated patients, patients with cirrhosis had reduced plasma renin activity (PRA) and AT I, AT II, and AT-(1-7) expression in the portal vein and periphery due to propranolol treatment. 74 Prevention or modification of certain vascular risk factors and proper management of cardiovascular disease may prevent the development or progression of dementia, including AD. 75 Protein homeostasis plays a role in the development of numerous disorders. Misfolded proteins are central in the pathophysiology of neurodegenerative diseases, such as AD, and play a role in the pathophysiology of common human cardiac diseases such as pathologic cardiac hypertrophy and dilated and ischemic cardiomyopathies. 76 In addition, cardiac surgery with cardiopulmonary bypass caused a profound cerebral inflammatory response, which was accompanied by increased postoperative cerebrospinal fluid (CSF) levels of the AD biomarker Aβ 142 . 77 Carvedilol inhibited basal and stimulated ACE production in human endothelial cells 78 and exhibited beneficial effects on ACE activity and PRA levels in patients with CHF . 79 In addition, proliferating infantile hemangiomas expressed 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 hemangiomas. 8082 Taken together, the RAS is activated in AD and the impact of beta-adrenergic receptor blockade on this system will affect AD.

Poly(ADP-ribose) polymerase 1 (PARP-1) is a nuclear protein that contributes to both neuronal death and survival under stressful conditions. The residual PARP activity found in PARP-1-deficient cells has been recently attributed to a novel DNA damage-dependent PARP. 83 The PARP cleavage is enhanced in the peripheral blood mononuclear cells of patients with mild cognitive impairment. 84 Enhanced PARP activity has been reported in AD and may be a marker for AD. 85 Poly-ADP-ribose polymers increase with age in the brains of an Alzheimer’s mouse model, and Aβ-activated poly-ADP-ribose polymers induced astrocytic metabolic failure and neuronal death in response to oxidative stress. Inhibition of either PARP or the nicotinamide adenine dinucleotide phosphate oxidase prevented the appearance of poly-ADP-ribose polymers and the mitochondrial depolarization. 86 The PARP-1 polymorphism modified the risk of AD in both an independent manner and through an interaction with the proinflammatory factor IL-1A. 87 The PARP-1 gene is also highly associated with AD susceptibility. Both Ht3-TT and Ht4-CC, which are the PARP haplotypes, were significantly associated with an increased risk of AD, whereas the Ht1-TC haplotype showed a protective effect against AD when compared with control participants. 88 Moreover, rabbits treated with ketamine exhibited reduced left ventricular ejection fractions, ventricular conduction velocity, and increased susceptibility to ventricular arrhythmia, which were prevented by metoprolol treatment. The expression of Parp-1 and apoptosis-inducing factor increased after ketamine treatment and sharply decreased after metoprolol administration. 89 Propranolol treatment markedly suppressed PARP activation in skeletal muscle biopsies from pediatric patients with burn. 90 Propranolol also protected against staurosporine-induced DNA fragmentation and PARP cleavage in SH-SY5Y neuroblastoma cells. 91 Furthermore, the nonselective β-receptor blocker carvedilol significantly inhibited apoptosis and suppressed activated PARP-1 cleavage in human cardiac tissue. 92 Carvedilol significantly reduced ischemia–reperfusion-induced poly- and mono-ADP-ribosylation in heart perfusion and rheological models. 93 Carvedilol also reduced PARP activity in the hippocampus and protected neurons against death after transient forebrain ischemia. 94 Metipranolol reduced the sodium nitroprusside-induced breakdown of PARP-1 in the eyes and retinas of rats. 95 These findings suggested that PARP-1 is activated in patients with AD and that beta-adrenergic receptor antagonists may affect AD through the suppression of PARP-1.

Angiogenesis is a complex process that involves the coordinated steps of endothelial cell activation, proliferation, migration, tube formation and capillary sprouting. In addition, angiogenesis requires the participation of several intracellular signaling pathways. Vascular endothelial growth factor (VEGF) is a key mediator of angiogenesis. Pathological angiogenesis may be a key event in the pathogenesis of AD. The abnormal regulation of VEGF expression has been reported in AD pathogenesis. Brain homogenates of APP23 mice, a transgenic model of AD, induced the formation of new vessels during in vivo angiogenesis and was blocked by a VEGF antagonist. 96 Compared with control participants, patients with AD had higher expression levels of angiopoietin 2 and VEGF in the micro-circulatory system. 97 Clusters of reactive astrocytes showed enhanced VEGF immunoreactivity in the neocortex of patients with AD but not in elderly control participants. 98 Increases in the VEGF levels in the CSF were also observed in patients with AD and vascular dementia but not in healthy controls. 99 These findings suggested that angiogenic changes occur in the micro-circulation of the brain with AD and may contribute to disease pathogenesis. The VEGF interacts with Aβ and co-localizes with Aβ in the brains of patients with AD. 100 The Aβ also inhibits VEGF-induced migration of endothelial cells as well as VEGF-induced permeability in an in vitro model of the blood–brain barrier. 101 The VEGF gene variability may be a genetic factor that influences lifespan in a cohort of Italian patients. 102 The VEGF polymorphisms are associated with AD in Italian, Han Chinese, and Tunisian populations. 103106 A number of mechanisms might link cancer with AD and other neurodegenerative diseases. 107 Roe et al 108 used population-based data from the Cardiovascular Health Cognition study to confirm the negative correlation between cancer and AD but not vascular dementia in Caucasian adults. Results from the Framingham Heart Study also had the same conclusion. 109 Moreover, the beta-adrenergic receptor agonist isoproterenol significantly increased VEGF protein levels in human choroidal endothelial cells. 110 The NE treatment increased VEGF levels in cultured nasopharyngeal carcinoma (NPC) tumor cells, and this increase was inhibited by propranolol treatment. Norepinephrine also induced invasiveness in all NPC cell lines in a dose-dependent manner, and this induction was blocked by propranolol treatment. 111 Moreover, propranolol significantly reduced VEGF activity in a phorbol myristate acetate (PMA)-activated human leukemic cell line. 112 This drug also repressed gastric cancer cell growth via its downstream effects on VEGF. 113,114 Alternatively, NE increased VEGF expression, and these effects were inhibited by propranolol treatment in pancreatic cancer cells. 115,116 In addition, epinephrine enhanced the VEGF expression in colon adenocarcinoma cells, and the stimulatory action of epinephrine on colon cancer growth was blocked by treatment with atenolol and ICI 118 551, which are beta1- and beta2-selective antagonists, respectively. 117 Beta2-adrenergic receptor blockade regulated VEGF production in a mouse model of oxygen-induced retinopathy. 118 Hypoxia-inducible factor 1α and VEGF messenger RNA and protein expression were both upregulated in a rat model of volume-overload heart failure; carvediol treatment reversed these abnormalities. 119 These findings suggested that beta-adrenergic receptor antagonists modulated VEGF expression in AD.

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, by generating reactive oxygen species (ROS) from molecular oxygen. The NOX is widely expressed in various immune cells, including microglia, macrophages, and neutrophils. In AD, NOX is activated in microglia, resulting in the formation of ROS that are toxic to neighboring neurons. 120 The Aβ induces mitochondrial dysfunction and oxidative stress in astrocytes and neural death via NOX activation. 121,122 The NOX expression and activity are specifically upregulated in vulnerable brain regions of mildly cognitive impaired patients. 123 The NOX is also upregulated in the frontal and temporal cortices and contributes to AD progression. 124 The inhibition of NOX or the gene deletion of its functional p47phox (phox: phagocyte oxidation) subunit promotes alternative and anti-inflammatory microglial activation during neuroinflammation. 125 Moreover, nebivolol, a third-generation selective beta1-adrenoceptor, improved left ventricle dysfunction and survival immediately after myocardial ischemia and inhibited cardiac NOX activation. 126 Nebivolol treatment has been associated with improvements in insulin resistance, reduced proteinuria, and reduced NOX activity as well as the production of ROS in the kidneys and skeletal muscle tissue of transgenic TG(mRen2)27 rats (Ren2). 127,128 Moreover, nebivolol also improved diastolic relaxation, fibrosis, and remodeling in obese Zucker rats and also reduced NOX-dependent superoxide production. 129 Carvedilol attenuated the increased expression of NOX subunits in the hearts and kidneys of rats after daunorubicin-induced cardiotoxicity and nephrotoxicity. 130 Activity of NOX in whole blood and isolated neutrophils was inhibited by nebivolol in a dose-dependent manner, whereas atenolol, metoprolol, and carvedilol were markedly less effective in Watanabe heritable hyperlipidemic rabbits. 131 Celiprolol, a specific beta1-receptor antagonist with weak beta2-receptor agonistic activity, suppressed NOX p22phox, p47phox, gp91phox, and Nox1 expression in the left ventricle of deoxycorticosterone acetate-salt hypertensive rats. 132 Taken together, these findings suggested that beta-adrenergic receptor antagonists play a role in AD through the suppression of NADPH expression.

The Role of Beta-Adrenergic Blockers in Alzheimer’s Disease

Matrix metalloproteinases (MMPs) are proteolytic enzymes that are responsible for remodeling the extracellular matrix and regulating leukocyte migration through the extracellular matrix. This migration is an important step in inflammatory and infectious pathophysiology. The MMPs are produced by many cell types, including lymphocytes, granulocytes, astrocytes, and activated macrophages. 133 Increasing evidence suggests that MMPs play an important role in the pathogenesis of AD. Leake et al. 134 identified an approximately 50% increase in the cortical levels of MMP-1 in AD. This finding is consistent with the presence of an inflammatory state within the brain in AD and contributes to the blood–brain barrier dysfunction observed in AD. Plasma MMP-3 was also significantly elevated in patients with AD. 135 The MMP-3 was expressed predominantly in the brain white matter and was also expressed in senile plaques in the cortices of patients with AD. 136 Compared with the control participants, patients with AD had significantly elevated plasma MMP-9 levels. 137 In the brain tissue of patients with AD, MMP-9 expression was found in the cytoplasm of neurons, neurofibrillary tangles, senile plaques, and vascular walls. 138 In addition, there were inverse correlations between the Global Cognitive and Mini-Mental State Examination scores and MMP-9 activity. 139 The Aβ is a potent stimulator of MMP-9 and MMP-2 activity in mixed hippocampal astrocyte cultures. 140 The interaction of Aβ and RAGEs induces MMP-2 expression in brain endothelial cells. 141 The MMP genotypes may influence the risk of dementia, and MMP gene polymorphisms have been reported to associate with vascular dementia and AD. 142 The MMP-3 variants are associated with changes in the Aβ levels in humans and an increased risk of dementia. 143,144 Treatment with an MMP-9 inhibitor improved the Aβ-mediated cognitive impairment and neurotoxicity in mice. 145 These findings further suggested that MMP-9 plays a causal role in Aβ-induced cognitive impairment and neurotoxicity. Moreover, propranolol inhibited human brain endothelial cell tubulogenesis and MMP-9 secretion. 146 A selective beta3-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 the NPC cell lines in a dose-dependent manner, and this effect could be blocked with an MMP inhibitor and propranolol treatment. 111 Propranolol significantly reduced MMP-2 activity in a PMA-activated human leukemic cell line. 112 Propranolol-induced growth inhibition has been associated with arrest at both G0/G1 and G2/M and repressed gastric cancer cell growth via the downstream inhibition of MMP-2 and MMP-9. 113 NE increased MMP-2 and MMP-9 expression, and these effects were inhibited by propranolol treatment in pancreatic cancer cells. 115,116 Epinephrine upregulated MMP-9 activity in human colon adenocarcinoma HT-29 cells, and this effect was blocked by beta1- and beta2-selective receptor antagonists, atenolol, and ICI 118,551. 117 These studies suggested that beta-adrenergic receptor antagonists might play an important role in the pathological process of PD via the regulation of tissue inhibitor of mettaloproteinase levels and the downregulation of MMPs.

The mitogen-activated protein kinase (MAPK) signaling pathways provide a key link between membrane-bound receptors that receive cues from signaling molecules and changes in the patterns of gene expression, which include the extracellular signal-regulated kinases cascade, the stress-activated protein kinases/c-Jun N-terminal kinase (SAPK/JNK) cascade, and the p38 MAPK/RK/HOG cascade. 148 An increase in the activation and expression levels of MKK6, one of the upstream activators of p38 MAPK, has been observed in AD brain tissue. 149 Patients with AD had increased levels of p38 MAPK phosphorylation associated with Aβ plaques and neurofibrillary tangle-bearing neurons. 150 . 151152 There is a link between Aβ-induced oxidative stress, activation of stress kinases SAPK/JNK and p38, and tau hyperphosphorylation, which was suggested in neurites surrounding amyloid plaque. 151 Strong protein kinase of 38-kDa (p38-P) immunereactivity was observed in about 50% to 70% of neurons with neurofibrillary tangles and in dystrophic neurites of senile plaques in AD. 152 In vitro activation of MKK6-p38 MAPK pathway resulted in tau phosphorylation at Ser-396, which suggested that MAPK pathway has a functional role in microtubule binding. Abnormal phosphorylation at Ser-396 was demonstrated in AD brain but not in normal functioning adult brain. 153 In addition, AD hippocampal isolated from post-mortem human brains showed co-immunopreciptate of MKK6 and phosphorylated tau protein, and such studies also showed that APP co-immunoprecipitated with both ASK-1 and MKK6. 154 Moreover, Aβ stimulated glial cell cultures and activate p38 MAPK, 155 contributing to the loss of neurons observed in neurodegenerative disease. A novel p38 α-MAPK inhibitor (MW01-2-069A-SRM) suppressed brain proinflammatory cytokine upregulation and attenuated synaptic dysfunction and behavioral deficits in an AD mouse model. 156 Inhibition of p38 MAPK with SB203580 decreased IL-1β-induced tau phosphorylation in vitro in neuronal cultures, 157 thus highlighting the importance of p38 MAPK as a target for combating neuro-inflammation. Moreover, beta-adrenoceptor stimulation activated the cAMP/PKA and MAPK pathways in pancreatic cancer cells. Beta2-adrenergic receptor antagonists suppressed invasion and proliferation via the inhibition of both cAMP/PKA and Ras, which regulate MAPK pathway activation. 116 The NE stimulated pancreatic cancer cell proliferation, migration, and invasion via the beta-adrenergic receptor-dependent activation of the p38/MAPK pathway. These stimulatory effects were completely abolished by treatment with propranolol or the p38/MAPK inhibitor SB203580. 158 Propranolol exerts its suppressive effects on hemangiomas via the hypoxia-inducible factor-1α-VEGF-A angiogenesis axis, with effects mediated by the PI3K/Akt and p38/MAPK pathways. 159 Taken together, these findings suggested that beta-adrenergic receptor antagonists may play a role in AD via suppression of the MAPK pathway.

Inflammation is thought to be integral to the pathogenesis of AD. Prostaglandins (PGs) play a role in inflammatory processes. 160 Cyclooxygenase (COX) participates in the conversion of arachidonic acid (AA) into PGs. The AA and its various metabolites, including PGs, thromboxanes, and leukotriene B4, induce a significantly higher secretion of both Aβ40 and Aβ42 peptides. 161 COX-2 and PGE2 synthesis are induced by Aβ1-42 in astrocytic cells via a nuclear factor-κB-dependent mechanism. 162 The PG receptors are expressed in the hypothalamus, thalamus, and limbic system, 163 and COX-2 is expressed by excitatory neurons at postsynaptic sites in the rat cerebral cortex. 164 Overexpression of COX-2 is observed in the perinuclear, dendritic, and axonal areas of pyramidal neurons as well as in subregions of the hippocampal formation in AD. 165,166 Moreover, COX-2 potentiated Aβ protein generation via mechanisms that involve γ-secretase activity. 167,168 Long-term treatment with nonsteroidal anti-inflammatory drugs has shown beneficial effects, including the improvement in AD progression. 169,170 The COX-2 was abnormally expressed in neutrophils and monocytes in patients with AD. 41 The COX-2 G/G genotype is associated with AD. 171 Moreover, epinephrine increased the release of PGE2 in human colon adenocarcinoma HT-29 cells, and this release was blocked by treatment with COX-2 inhibitors or atenolol and ICI 118 551 (beta1- and beta2-selective adrenergic antagonists, respectively). 117 Beta2-adrenergic receptor antagonists suppressed COX-2 expression in pancreatic cancer cells. 116 Propranolol inhibited cell proliferation and repressed gastric cancer cell growth via the downstream COX-2 pathway. 113,114 In addition, the administration of propranolol and a COX-2 inhibitor, applied perioperatively in most patients with cancer with minimal risk and low cost, counteracted several immunological and endocrinological perturbations and improved recurrence-free survival rates in mice undergoing primary tumor excision. 172,173 These findings suggested that beta-adrenergic receptor antagonists play a role in modulating the inflammatory process in AD.

The ROS have been implicated in the pathogenesis of neuronal death in AD. Increased levels of ROS have been reported in AD. 174,175 Oxygen-free radical injury causes some AD-type molecular abnormalities in human neuronal cells. 176 Oxidative stress is a sign of AD pathology and may be an early event in the progression of the mild cognitive impairment that leads to AD. 177 Cultured skin fibroblasts from patients with AD exhibited increased superoxide dismutase activity, and these cells were more susceptible to free radical damage. 178,179 Mitochondria-derived ROS resulted in enhanced amyloidogenic amyloid precursor protein (APP) processing, and Aβ itself led to mitochondrial dysfunction and increased ROS levels. 180 Moreover, myocardial tissue sections displayed increased ROS levels after traumatic brain injuries. Treatment with propranolol lowered cardiac ROS levels. 181 d-propranolol attenuated lysosomal iron accumulation and oxidative injury in endothelial cells. 182 Carvedilol modulated ROS-induced signaling. Carvedilol also 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. 93 Carvedilol also protected against colcichine- and aluminum-induced neurotoxicity in rats by attenuating oxidative stress, including lipid peroxidation, nitrite concentration and restored reduced glutathione, superoxide dismutase, catalase, and glutathione S-transferase activity. Carvediol also improved the memory of rats in the Morris water maze test. 31,32 Furthermore, nebivolol improved diastolic dysfunction and myocardial remodeling through reductions in oxidative stress in transgenic (mRen2) rats. 183 These findings suggested that beta-adrenergic receptor antagonists modulate oxidative stress in AD.

Nitric oxide synthase (NOS) generates nitric oxide (NO), which is a critical signaling molecule involved in synaptic plasticity and memory. 184,185 Endothelial NO (eNO) plays an important role in modulating APP expression and processing within the brain and cerebro-vasculature. Brain tissue from eNOS-/- mice had statistically higher APP and BACE1 protein levels, as well as increased BACE1, beta-site APP-cleaving enzyme1, enzyme activity and Aβ1-42 wild-type control. 186 Prolonged NO treatment resulted in tau aggregation in SH-SY5Y cells. 187 Studies have reported that the leukocytes and brain microvessels of patients with AD had significantly increased NOS activity. 188,189 Moreover, NOS may contribute to the pathogenesis of AD. In AD and APP transgenic mice, astrocytes with high NOS levels were associated with Aβ protein deposits. 190 In AD-like mice, NOS deficiency also protected against premature mortality, cerebral plaque formation, increased Aβ protein levels, astrocytosis, and microgliosis. 191 Moreover, metipranolol suppressed NO-induced lipid peroxidation in the eyes and retinas of rats. 95 Nebivolol prevented vascular NOS III uncoupling in experimental hyperlipidemia, 131 and propranolol suppressed hemangioma growth via inhibition of eNOS protein expression and the subsequent production of NO. 192 Furthermore, celiprolol activated eNOS through the PI3K-Akt pathway via oxidative stress-induced NF-kB activity. 132 These findings suggested that beta-adrenergic receptor antagonists play a role in AD via the inhibition of NOS expression.

Conclusion

Beta-adrenergic receptor blockade may play a role in AD. Genetic studies have identified proteins that link beta-adrenergic receptor antagonism to the pathology of AD, including HLA genes, the RAS, PARP-1, NGF, VEGF, and the reduced form of NADP. Beta-adrenergic receptor inhibition also affects AD via nongenomic mechanisms, including MMPs, MAPK pathways, PGs, COX-2, and NOS. The beta-adrenergic receptor blockades are contradicted in patients with asthma and CHF and cautioned to nursing women. Depression has been associated with lipophilic beta-adrenergic receptor blockades, such as propranolol. Serious CNS adverse effects, including agitation, confusion, and hallucinations, are rare. However, the most interesting side effect of beta-adrenergic receptor blockade is hypotension or symptoms associated with hypotension. The role of beta-adrenergic receptor blockade in AD is still controversial. It is still unclear whether behavioral symptoms, sex, or genetic factors, including beta2-adrenergic receptor variants, apoliporotein E (apoE), and cytochrome P450 CYP2D6 participate in the beta-adrenergic receptor blockade modulation in AD. Various behavioral abnormalities appear to be present in subgroups of patients with AD. 193,194 Compared with both nonaggressive patients with AD and control participants, aggressive patients with AD had small but significant (approximately 25%) increases in concentrations of beta1- and beta2-adrenergic receptors in the cerebellar cortex. 16 There was also an apparent sex difference in cerebral amyloid plaque formation. Compared with the males, transgenic female Tg2576 mice had more Aβ40 and Aβ42 in the brain. 195 Ni et al 20 reported that female mice had more amyloid plaques than age-matched males among the control mice. These authors also revealed that female mice appeared to be more sensitive to chronic treatment with beta-adrenergic receptor agonist than the male mice. Beta2-adrenergic receptor polymorphisms contributed to AD pathology. 196,197 The ApoE is a major cholesterol carrier that supports lipid transport and injury repair in the brain. The ApoE polymorphic alleles are the main genetic determinants of AD risk; individuals carrying the ε4 allele are at increased risk of AD compared with those carrying the more common ε3 allele, whereas the ε2 allele decreases risk. 198 The apoE ε4 allele frequency was significantly higher in the AD groups compared with the control group. 199 Carvedilol reduces the severity of atherosclerosis in apoE-deficient mice via reducing superoxide production. 200 The CYP2D6B allele is associated with AD. 201,202 The CYP2D6 allele frequency is known to vary among racial/ethnic groups. In general, the frequency of the functional group of predominant alleles in European caucasians is 71%. In Asians, the functional alleles represent only ∼50% of the frequency of CYP2D6 alleles. 203 Moreover, genetic polymorphism of CYP2D6 results in altered pharmacokinetics of beta-adrenergic receptor antagonistic medications. 204207 However, substantial reservation regarding these findings need to be noted. It is not entirely clear whether the direct action of beta-adrenergic receptor antagonists in brain has been separated from the impact of beta drugs on the cardiovascular system, which, in turn, affects AD. Thus, further studies on the relationship between beta-adrenergic receptor antagonists and AD are warranted.

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. 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]
  • 2. 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]
  • 3. Wang D, Govindaiah G, Liu R, De Arcangelis V, Cox CL, Xiang YK. Binding of amyloid beta peptide to beta2 adrenergic receptor induces PKA-dependent AMPA receptor hyperactivity. FASEB J. 2010;24(9):3511–3521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Wang D, Yuen EY, Zhou Y, Yan Z, Xiang YK. Amyloid beta peptide-(1-42) induces internalization and degradation of β2 adrenergic receptors in prefrontal cortical neurons. J Biol Chem. 2011;286(36):31852–31863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Shimohama S, Taniguchi T, Fujiwara M, Kameyama M. Changes in beta-adrenergic receptor subtypes in Alzheimer-type dementia. J Neurochem. 1987;48(4):1215–1221. [DOI] [PubMed] [Google Scholar]
  • 6. Oppenheim G, Mintzer J, Halperin Y, Eliakim R, Stessman J, Ebstein RP. Acute desensitization of lymphocyte beta-adrenergic-stimulated adenylate cyclase in old age and Alzheimer’s disease. Life Sci. 1984;35(17):1795–1802. [DOI] [PubMed] [Google Scholar]
  • 7. Huang HM, Gibson GE. Altered beta-adrenergic receptor-stimulated cAMP formation in cultured skin fibroblasts from Alzheimer donors. J Biol Chem. 1993;268(20):14616–14621. [PubMed] [Google Scholar]
  • 8. Karczewski P, Hempel P, Kunze R, Bimmler M. Agonistic autoantibodies to the α1-adrenergic receptor and the β2-adrenergic receptor in Alzheimer’s and vascular dementia. Scand J Immunol. 2012;75(5):524–530. [DOI] [PubMed] [Google Scholar]
  • 9. Yang JH, Lee EO, Kim SE, Suh YH, Chong YH. Norepinephrine differentially modulates the innate inflammatory response provoked by amyloid-β peptide via action at β-adrenoceptors and activation of cAMP/PKA pathway in human THP-1 macrophages. Exp Neurol. 2012;236(2):199–206. [DOI] [PubMed] [Google Scholar]
  • 10. Francis BM, Yang J, Hajderi E, et al. Reduced tissue levels of noradrenaline are associated with behavioral phenotypes of the TgCRND8 mouse model of Alzheimer’s disease. Neuropsychopharmacology. 2012;37(8):1934–1944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Kong Y, Ruan L, Qian L, Liu X, Le Y. Norepinephrine promotes microglia to uptake and degrade amyloid beta peptide through upregulation of mouse formyl peptide receptor 2 and induction of insulin-degrading enzyme. J Neurosci. 2010;30(35):11848–11857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Gibbs ME, Maksel D, Gibbs Z, Hou X, Summers RJ, Small DH. Memory loss caused by beta-amyloid protein is rescued by a β3-adrenoceptor agonist. Neurobiol Aging. 2010;31(4):614–624 [DOI] [PubMed] [Google Scholar]
  • 13. Ramos BP, Colgan LA, Nou E, Arnsten AF. Beta2 adrenergic agonist, clenbuterol, enhances working memory performance in aging animals. Neurobiol Aging. 2008;29(7):1060–1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Heneka MT, Galea E, Gavriluyk V, et al. Noradrenergic depletion potentiates beta -amyloid-induced cortical inflammation: implications for Alzheimer’s disease. J Neurosci. 2002;22(7):2434–2442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Gliebus G, Lippa CF. The influence of beta-blockers on delayed memory function in people with cognitive impairment. Am J Alzheimers Dis Other Demen. 2007;22(1):57–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Russo-Neustadt A, Cotman CW. Adrenergic receptors in Alzheimer’s disease brain: selective increases in the cerebella of aggressive patients. J Neurosci. 1997;17(14):5573–5580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Kalaria RN, Andorn AC, Tabaton M, Whitehouse PJ, Harik SI, Unnerstall JR. Adrenergic receptors in aging and Alzheimer’s disease: increased β2-receptors in prefrontal cortex and hippocampus. J Neurochem. 1989;53(6):1772–1781. [DOI] [PubMed] [Google Scholar]
  • 18. Lemmer B, Langer L, Ohm T, Bohl J. Beta-adrenoceptor density and subtype distribution in cerebellum and hippocampus from patients with Alzheimer’s disease. Naunyn Schmiedebergs Arch Pharmacol. 1993;347(2):214–219. [DOI] [PubMed] [Google Scholar]
  • 19. Kalaria RN, Harik SI. Increased α2- and β2-adrenergic receptors in cerebral microvessels in Alzheimer disease. Neurosci Lett. 1989;106(1-2):233–238. [DOI] [PubMed] [Google Scholar]
  • 20. Ni Y, Zhao X, Bao G, et al. Activation of β2-adrenergic receptor stimulates gamma-secretase activity and accelerates amyloid plaque formation. Nat Med. 2006;12(12):1390–1396. [DOI] [PubMed] [Google Scholar]
  • 21. Yu NN, Wang XX, Yu JT, et al. Blocking β2-adrenergic receptor attenuates acute stress-induced amyloid beta peptides production. Brain Res. 2010;1317:305–310 [DOI] [PubMed] [Google Scholar]
  • 22. Hajjar I, Catoe H, Sixta S, et al. Cross-sectional and longitudinal association between antihypertensive medications and cognitive impairment in an elderly population. J Gerontol A Biol Sci Med Sci. 2005;60(1):67–73. [DOI] [PubMed] [Google Scholar]
  • 23. Khachaturian AS, Zandi PP, Lyketsos CG, et al. Antihypertensive medication use and incident Alzheimer disease: the Cache County Study. Arch Neurol. 2006;63(5):686–692. [DOI] [PubMed] [Google Scholar]
  • 24. Rosenberg PB, Mielke MM, Tschanz J, et al. Effects of cardiovascular medications on rate of functional decline in Alzheimer disease. Am J Geriatr Psychiatry. 2008;16(11):883–892. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Weiler PG, Mungas D, Bernick C. Propranolol for the control of disruptive behavior in senile dementia. J Geriatr Psychiatry Neurol. 1988;1(4):226–230. [DOI] [PubMed] [Google Scholar]
  • 26. Shankle WR, Nielson KA, Cotman CW. Low-dose propranolol reduces aggression and agitation resembling that associated with orbitofrontal dysfunction in elderly demented patients. Alzheimer Dis Assoc Disord. 1995;9(4):233–237. [PubMed] [Google Scholar]
  • 27. Peskind ER, Tsuang DW, Bonner LT, et al. Propranolol for disruptive behaviors in nursing home residents with probable or possible Alzheimer disease: a placebo-controlled study. Alzheimer Dis Assoc Disord. 2005;19(1):23–28. [DOI] [PubMed] [Google Scholar]
  • 28. Summers WK. The management of agitation in demented patients with propranolol. J Alzheimers Dis. 2006;9(1):69–75. [DOI] [PubMed] [Google Scholar]
  • 29. Dobarro M, Orejana L, Aguirre N, Ramírez MJ. Propranolol reduces cognitive deficits, amyloid β levels, tau phosphorylation and insulin resistance in response to chronic corticosterone administration [published online November 30, 2012]. Int J Neuropsychopharmacol. 2012:1–10. [DOI] [PubMed] [Google Scholar]
  • 30. Dobarro M, Orejana L, Aguirre N, Ramírez MJ. Propranolol restores cognitive deficits and improves amyloid and Tau pathologies in a senescence-accelerated mouse model. Neuropharmacology. 2013;64:137–144. [DOI] [PubMed] [Google Scholar]
  • 31. Kumar A, Dogra S. Neuroprotective effect of carvedilol, an adrenergic antagonist against colchicine induced cognitive impairment and oxidative damage in rat. Pharmacol Biochem Behav. 2009;92(1):25–31. [DOI] [PubMed] [Google Scholar]
  • 32. Kumar A, Prakash A, Dogra S. Neuroprotective effect of carvedilol against aluminium induced toxicity: possible behavioral and biochemical alterations in rats. Pharmacol Rep. 2011;63(4):915–923. [DOI] [PubMed] [Google Scholar]
  • 33. Wang J, Ono K, Dickstein DL, et al. Carvedilol as a potential novel agent for the treatment of Alzheimer’s disease. Neurobiol Aging. 2011;32(12):2321.e1–e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Wang J, Wright HM, Vempati P, et al. Investigation of Nebivolol as a Novel Therapeutic Agent for the Treatment of Alzheimer’s Disease. J Alzheimers Dis. 2012;33(4):1147–1156. [DOI] [PubMed] [Google Scholar]
  • 35. Payton A, van den Boogerd E, Davidson Y, et al. Influence and interactions of cathepsin D, HLA-DRB1 and APOE on cognitive abilities in an older non-demented population. Genes Brain Behav. 2006;5(suppl 1):23–31. [DOI] [PubMed] [Google Scholar]
  • 36. McGeer PL, Itagaki S, Tago H, McGeer EG. Reactive microglia in patients with senile dementia of the Alzheimer type are positive for the histocompatibility glycoprotein HLA-DR. Neurosct Lett. 1987;79(1-2):195–200. [DOI] [PubMed] [Google Scholar]
  • 37. Styren SD, Civin WH, Rogers J. Molecular, cellular, and pathologic characterization of HLA-DR immunoreactivity in normal elderly and Alzheimer’s disease brain. Exp Neurol. 1990;110(1):93–104. [DOI] [PubMed] [Google Scholar]
  • 38. Tooyama I, Kimura H, Akiyama H, McGeer PL. Reactive microglia express class I and class II antigens in Alzheimer’s disease. Brain Res. 1990;523(2):273–280. [DOI] [PubMed] [Google Scholar]
  • 39. Xiang Z, Haroutunian V, Ho L, Purohit D, Pasinetti GM. Microglia activation in the brain as inflammatory biomarker of Alzheimer’s disease neuropathology and clinical dementia. Dis Markers. 2006;22(1-2):95–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Liew SCK, Penfold PL, Provis JM, Madigan MC, Billson FA. Modulation of MHC class II expression in the absence of lymphocytic infiltrates in Alzheimer’s retinae. J Neurophathol Exp Neurol. 1994;53(2):150–157. [DOI] [PubMed] [Google Scholar]
  • 41. Fiala M, Lin J, Ringman J, et al. Ineffective phagocytosis of amyloid-beta by macrophages of Alzheimer’s disease patients. J Alzheimers Dis. 2005;7(3):221–232. [DOI] [PubMed] [Google Scholar]
  • 42. Rogers J, Luber-Narod J, Styren SD, Civin WH. Expression of immune system associated antigens by cells of the human central nervous system: relationship to the pathology of Alzheimer’s disease. Neurobiol Aging. 1988;9(4):339–349. [DOI] [PubMed] [Google Scholar]
  • 43. Shalit F, Sregni B, Brodie C, Kott E, Huberman M. T lymphocyte subpopulations and activation markers correlate with severity of Alzheimer’s disease. Clin Immunol Immunopathol. 1995;75(3):246–250. [DOI] [PubMed] [Google Scholar]
  • 44. Zota V, Nemirovsky A, Baron R, et al. HLA-DR alleles in amyloid β-peptide autoimmunity: a highly immunologic role for the DRB1*1501 allele. J Immunol. 2009;183(5):3522–3530. [DOI] [PubMed] [Google Scholar]
  • 45. 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]
  • 46. 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]
  • 47. 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]
  • 48. 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]
  • 49. Changaris DG, Keil LC, Severs WB. Angiotensin II immunohistochemistry of the rat brain. Neuroendocrinol. 1978;25(5):257–274. [DOI] [PubMed] [Google Scholar]
  • 50. 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]
  • 51. 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]
  • 52. 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]
  • 53. Arregui A, Perry E, Rossor M, Tomlinson BE. Angiotensin converting enzyme in Alzheimer’s disease: increased activity in caudate nucleus and cortical areas. J Neurochem. 1982;38(5):1490–1492. [DOI] [PubMed] [Google Scholar]
  • 54. Barnes NM, Cheng CH, Costall B, Naylor RJ, Williams TJ, Wischik CM. Angiotensin converting enzyme density is increased in temporal cortex from patients with Alzheimer’s disease. Eur J Pharmacol. 1991;200(2-3):289–292. [DOI] [PubMed] [Google Scholar]
  • 55. Savaskan E, Hock C, Olivieri G, et al. Cortical alterations of angiotensin converting enzyme, angiotensin II and AT1 receptor in Alzheimer’s dementia. Neurobiol Aging. 2001;22(4):541–546. [DOI] [PubMed] [Google Scholar]
  • 56. Akatsu H, Ogawa N, Kanesaka T, et al. High activity of peripheral blood angiotensin-converting enzyme is associated with later-onset of Alzheimer’s disease. J Neurol Sci. 2011;300(1-2):67–73. [DOI] [PubMed] [Google Scholar]
  • 57. Hu J, Igarashi A, Kamata M, Nakagawa H. Angiotensin converting enzyme degrades Alzheimer amyloid β-peptide (Aβ); retards Aβ aggregation, deposition, fibril formation; and inhibits cytotoxicity. J Biol Chem. 2001;276(51):47863–47868. [DOI] [PubMed] [Google Scholar]
  • 58. Heming ML, Selkoe DJ. Amyloid β-protein is degraded by cellular angiotensin-converting enzyme (ACE) and elevated by an ACE inhibitor. J Biol Chem. 2005;280(45):37644–37650. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Hemming ML, Selkoe DJ, Farris W. Effects of prolonged angiotensin-converting enzyme inhibitor treatment on amyloid β-protein metabolism in mouse models of Alzheimer disease. Neurobiol Dis. 2007;26(1):273–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Zou K, Yamaguchi H, Akatsu H, et al. Angiotensin-converting enzyme converts amyloid beta-protein 1-42 (Aβ1-42) to Aβ1-40, and its inhibition enhances brain Aβ deposition. J Neurosci. 2007;27(32):8628–8635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Furiya Y, Ryo M, Kawahara M, Kiriyama T, Morikawa M, Ueno S. Renin-angiotensin system blockers affect cognitive decline and serum adipocytokines in Alzheimer’s disease [published on November 8, 2012]. Alzheimers Dement. 2012; pii: S1552–5260(12) 02382–5. [DOI] [PubMed] [Google Scholar]
  • 62. Danielyan L, Klein R, Hanson LR, et al. Protective effects of intranasal losartan in the APP/PS1 transgenic mouse model of Alzheimer disease. Rejuvenation Res. 2010;13(2-3):195–201. [DOI] [PubMed] [Google Scholar]
  • 63. Chen SD, Wu CL, Lin TK, Chuang YC, Yang DI. Renin inhibitor aliskiren exerts neuroprotection against amyloid beta-peptide toxicity in rat cortical neurons. Neurochem Int. 2012;61(3):369–377. [DOI] [PubMed] [Google Scholar]
  • 64. Kölsch H, Jessen F, Freymann N, et al. ACE I/D polymorphism is a risk factor of Alzheimer’s disease but not of vascular dementia. Neurosci Lett. 2005;377(1):37–39. [DOI] [PubMed] [Google Scholar]
  • 65. Elkins JS, Douglas VC, Johnston SC. Alzheimer disease risk and genetic variation in ACE: a meta-analysis. Neurology. 2004;62(3):363–368. [DOI] [PubMed] [Google Scholar]
  • 66. Lehmann DJ, Cortina-Borja M, Warden DR, et al. Large meta-analysis establishes the ACE insertion-deletion polymorphism as a marker of Alzheimer’s disease. Am J Epidemiol. 2005;162(4):305–317. [DOI] [PubMed] [Google Scholar]
  • 67. Sleegers K, den Heijer T, van Dijk EJ, et al. ACE gene is associated with Alzheimer’s disease and atrophy of hippocampus and amygdala. Neurobiol Aging. 2005;26(8):1153–1159. [DOI] [PubMed] [Google Scholar]
  • 68. Lendon CL, Thaker U, Harris JM, et al. The angiotensin 1-converting enzyme insertion (I)/deletion (D) polymorphism does not influence the extent of amyloid or tau pathology in patients with sporadic Alzheimer’s disease. Neurosci Lett. 2002;328(3):314–318. [DOI] [PubMed] [Google Scholar]
  • 69. 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]
  • 70. 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]
  • 71. 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(3 pt 1):C588–C595. [DOI] [PubMed] [Google Scholar]
  • 72. 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]
  • 73. 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]
  • 74. 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]
  • 75. Muqtadar H, Testai FD, Gorelick PB. The dementia of cardiac disease. Curr Cardiol Rep. 2012;14(6):732–740. [DOI] [PubMed] [Google Scholar]
  • 76. Willis MS, Patterson C. Proteotoxicity and cardiac dysfunction--Alzheimer’s disease of the heart? N Engl J Med. 2013;368(5):455–464. [DOI] [PubMed] [Google Scholar]
  • 77. Reinsfelt B, Westerlind A, Blennow K, Zetterberg H, Ricksten SE. Open-heart surgery increases cerebrospinal fluid levels of Alzheimer-associated amyloid β. Acta Anaesthesiol Scand. 2013;57(1):82–88. [DOI] [PubMed] [Google Scholar]
  • 78. 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]
  • 79. 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]
  • 80. 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(6):759–765. [DOI] [PubMed] [Google Scholar]
  • 81. 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]
  • 82. 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]
  • 83. Shall S, de Murcia G. Poly(ADP-ribose) polymerase-1: what have we learned from the deficient mouse model? Mutat Res. 2000;460(1):1–15. [DOI] [PubMed] [Google Scholar]
  • 84. Love S, Barber R, Wilcock GK. Increased poly(ADP-ribosyl)ation of nuclear proteins in Alzheimers’s disease. Brain. 1999;122(pt 2):247–253. [DOI] [PubMed] [Google Scholar]
  • 85. Kassner SS, Bonaterra GA, Kaiser E, et al. Novel systemic markers for patients with Alzheimer disease?–A pilot study. Current Alzheimer Res. 2008;5(4):358–366. [DOI] [PubMed] [Google Scholar]
  • 86. Abeti R, Abramov AY, Duchen MR. β-amyloid activates PARP causing astrocytic metabolic failure and neuronal death. Brain. 2011;134(pt 6):1658–1672. [DOI] [PubMed] [Google Scholar]
  • 87. Infante J, Llorca J, Mateo I, et al. Interaction between Poly(ADPRibose) Polymerase 1 and Interleukin 1A genes is associated with Alzheimer’s disease risk. Dement Geriatr Cogn Disord. 2007;23(4):215–218. [DOI] [PubMed] [Google Scholar]
  • 88. Liu HP, Lin WY, Wu BT, et al. Evaluation of the poly(ADP-ribose) polymerase-1 gene variants in Alzheimer’s disease. J Clin Lab Anal. 2010;24(3):182–186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89. 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]
  • 90. 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]
  • 91. 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]
  • 92. 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]
  • 93. 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]
  • 94. 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]
  • 95. 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]
  • 96. Schultheiss C, Blechert B, Gaertner FC, Drecoll E, Mueller J. In vivo characterization of endothelial cell activation in a transgenic mouse model of Alzheimer’s disease. Angiogenesis. 2006;9(2):59–65. [DOI] [PubMed] [Google Scholar]
  • 97. Thirumangalakudi L, Samany PG, Owoso A, Wiskar B, Grammas P. Angiogenic proteins are expressed by brain blood vessels in Alzheimer’s disease. J Alzheimers Dis. 2006;10(1):111–118. [DOI] [PubMed] [Google Scholar]
  • 98. Kalaria RN, Cohen DL, Premkumar DR, Nag S, LaManna JC, Lust WD. Vascular endothelial growth factor in Alzheimer’s disease and experimental cerebral ischemia. Brain Res Mol Brain Res. 1998;62(1):101–105. [DOI] [PubMed] [Google Scholar]
  • 99. Tarkowski E, Issa R, Sjögren M, et al. Increased intrathecal levels of the angiogenic factors VEGF and TGF-beta in Alzheimer’s disease and vascular dementia. Neurobiol Aging. 2002;23(2):237–243. [DOI] [PubMed] [Google Scholar]
  • 100. Yang SP, Bae DG, Kang HJ, Gwag BJ, Gho YS, Chae CB. Co-accumulation of vascular endothelial growth factor with beta-amyloid in the brain of patients with Alzheimer’s disease. Neurobiol Aging. 2004;25(3):283–290. [DOI] [PubMed] [Google Scholar]
  • 101. Patel NS, Mathura VS, Bachmeier C, et al. Alzheimer’s beta-amyloid peptide blocks vascular endothelial growth factor mediated signaling via direct interaction with VEGFR-2. J Neurochem. 2010;112(1):66–76. [DOI] [PubMed] [Google Scholar]
  • 102. Del Bo R, Ghezzi S, Scarlato M, et al. Role of VEGF gene variability in longevity: a lesson from the Italian population. Neurobiol Aging. 2008;29(12):1917–1922. [DOI] [PubMed] [Google Scholar]
  • 103. Del Bo R, Scarlato M, Ghezzi S, et al. Vascular endothelial growth factor gene variability is associated with increased risk for AD. Ann Neurol. 2005;57(3):373–380. [DOI] [PubMed] [Google Scholar]
  • 104. Chiappelli M, Borroni B, Archetti S, et al. VEGF gene and phenotype relation with Alzheimer’s disease and mild cognitive impairment. Rejuvenation Res. 2006;9(4):485–493. [DOI] [PubMed] [Google Scholar]
  • 105. Yuan Q, Zuo X, Jia J. Association between promoter polymorphisms of vascular endothelial growth factor gene and sporadic Alzheimer’s disease among Northern Chinese Han. Neurosci Lett. 2009;457(3):133–136. [DOI] [PubMed] [Google Scholar]
  • 106. Smach MA, Charfeddine B, Othman LB, et al. -1154G/A and -2578C/A polymorphisms of the vascular endothelial growth factor gene in Tunisian Alzheimer patients in relation to beta-amyloid (1-42) and total tau protein. Neurosci Lett. 2010;472(2):139–142. [DOI] [PubMed] [Google Scholar]
  • 107. Tavares AR, Jr, de Melo AC, Sternberg C. Cancer linked to Alzheimer disease but not vascular dementia. Neurology. 2010;75(13):1215–1216. [PubMed] [Google Scholar]
  • 108. Roe CM, Fitzpatrick AL, Xiong C, et al. Cancer linked to Alz heimer disease but not vascular dementia. Neurology. 2010;74(2):106–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109. Driver JA, Beiser A, Au R, et al. Inverse association between cancer and Alzheimer’s disease: results from the Framingham Heart Study. BMJ. 2012;344:e1442. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. 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]
  • 111. 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]
  • 112. 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]
  • 113. 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]
  • 114. 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]
  • 115. 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]
  • 116. 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]
  • 117. 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]
  • 118. 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]
  • 119. 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]
  • 120. Shimohama S, Tanino H, Kawakami N, et al. Activation of NADPH oxidase in Alzheimer’s disease brains. Biochem Biophys Res Commun. 2000;273(1):5–9. [DOI] [PubMed] [Google Scholar]
  • 121. Jana A, Pahan K. Fibrillar amyloid-beta-activated human astroglia kill primary human neurons via neutral sphingomyelinase: implications for Alzheimer’s disease. J Neurosci. 2010;30(38):12676–12689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Abramov AY, Canevari L, Duchen MR. Beta-amyloid peptides induce mitochondrial dysfunction and oxidative stress in astrocytes and death of neurons through activation of NADPH oxidase. J Neurosci. 2004;24(2):565–575. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Bruce-Keller AJ, Gupta S, Parrino TE, et al. NOX activity is increased in mild cognitive impairment. Antioxid Redox Signal. 2010;12(12):1371–1382 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. Ansari MA, Scheff SW. NADPH-oxidase activation and cognition in Alzheimer disease progression. Free Radic Biol Med. 2011;51(1):171–178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Choi SH, Aid S, Kim HW, Jackson SH, Bosetti F. Inhibition of NADPH oxidase promotes alternative and anti-inflammatory microglial activation during neuroinflammation. J Neurochem. 2012;120(2):292–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. 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]
  • 127. 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]
  • 128. 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]
  • 129. Zhou X, Ma L, Habibi J, Whaley-Connell A, Hayden MR, 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]
  • 130. 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]
  • 131. 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]
  • 132. 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]
  • 133. Yong VW, Krekoski CA, Forsyth PA, Bell R, Edwards DR. Matrix metalloproteinases and diseases of the CNS. Trends Neurosci. 1998;21(2):75–80. [DOI] [PubMed] [Google Scholar]
  • 134. Leake A, Morris CM, Whateley J. Brain matrix metalloproteinase 1 levels are elevated in Alzheimer’s disease. Neurosci Lett. 2000;291(3):201–203. [DOI] [PubMed] [Google Scholar]
  • 135. Horstmann S, Budig L, Gardner H, et al. Matrix metalloproteinases in peripheral blood and cerebrospinal fluid in patients with Alzheimer’s disease. Int Psychogeriatr. 2010;22(6):966–972. [DOI] [PubMed] [Google Scholar]
  • 136. Yoshiyama Y, Asahina M, Hattori T. Selective distribution of matrix metalloproteinase-3 (MMP-3) in Alzheimer’s disease brain. Acta Neuropathol. 2000;99(2):91–95. [DOI] [PubMed] [Google Scholar]
  • 137. Lorenzl S, Albers DS, Relkin N, et al. Increased plasma levels of matrix metalloproteinase-9 in patients with Alzheimer’s disease. Neurochem Int. 2003;43(3):191–196. [DOI] [PubMed] [Google Scholar]
  • 138. Asahina M, Yoshiyama Y, Hattori T. Expression of matrix metalloproteinase-9 and urinary-type plasminogen activator in Alzheimer’s disease brain. Clin Neuropathol. 2001;20(2):60–63. [PubMed] [Google Scholar]
  • 139. Bruno MA, Mufson EJ, Wuu J, Cuello AC. Increased matrix metalloproteinase 9 activity in mild cognitive impairment. J Neuropathol Exp Neurol. 2009;68(12):1309–1318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140. Deb S, Gottschall PE. Increased production of matrix metalloproteinases in enriched astrocyte and mixed hippocampal cultures treated with beta-amyloid peptides. J Neurochem. 1996;66(4):1641–1647. [DOI] [PubMed] [Google Scholar]
  • 141. Du H, Li P, Wang J, Qing X, Li W. The interaction of amyloid β and the receptor for advanced glycation endproducts induces matrix metalloproteinase-2 expression in brain endothelial cells. Cell Mol Neurobiol. 2012;32(1):141–147. [DOI] [PubMed] [Google Scholar]
  • 142. Flex A, Gaetani E, Proia AS, et al. Analysis of functional polymorphisms of metalloproteinase genes in persons with vascular dementia and Alzheimer’s disease. J Gerontol A Biol Sci Med Sci. 2006;61(10):1065–1069. [DOI] [PubMed] [Google Scholar]
  • 143. Reitz C, van Rooij FJ, Soares HD, et al. Matrix metalloproteinase 3 haplotypes and plasma amyloid beta levels: the Rotterdam Study. Neurobiol Aging. 2010;31(4):715–718. [DOI] [PubMed] [Google Scholar]
  • 144. Helbecque N, Cottel D, Hermant X, Amouyel P. Impact of the matrix metalloproteinase MMP-3 on dementia. Neurobiol Aging. 2007;28(8):1215–1220. [DOI] [PubMed] [Google Scholar]
  • 145. Mizoguchi H, Takuma K, Fukuzaki E, et al. Matrix metalloprotease-9 inhibition improves amyloid beta-mediated cognitive impairment and neurotoxicity in mice. J Pharmacol Exp Ther. 2009;331(1):14–22. [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 X, Rottkamp CA, Hartzler A, Sun Z, Takeda A, et al. Activation of MKK6, an upstream activator of p38, in Alzheimer’s disease. J Neurochem. 2001;79(2):311–318. [DOI] [PubMed] [Google Scholar]
  • 150. Hensley K, Floyd RA, Zheng NY, et al. P38 kinase is activated in the Alzheimer’s disease brain. J Neurochem. 1999;72(5):2053–2058. [DOI] [PubMed] [Google Scholar]
  • 151. Puig B, Gómez-Isla T, Ribé E, et al. Expression of stress-activated kinases c-Jun N-terminal kinase (SAPK/JNK-P) and p38 kinase (p38-P), and tau hyperphosphorylation in neurites surrounding betaA plaques in APP Tg2576 mice. Neuropathol Appl Neurobiol. 2004;30(5):491–502. [DOI] [PubMed] [Google Scholar]
  • 152. Ferrer I, Blanco R, Carmona M, Puig B. Phosphorylated mitogen-activated protein kinase (MAPK/ERK-P), protein kinase of 38 kDa (p38-P), stress-activated protein kinase (SAPK/JNK-P), and calcium/calmodulin-dependent kinase II (CaM kinase II) are differentially expressed in tau deposits in neurons and glial cells in tauopathies. J Neural Transm. 2001;108(12):1397–1415. [DOI] [PubMed] [Google Scholar]
  • 153. Bramblett GT, Goedert M, Jakes R, Merrick SE, Trojanowski JQ, Lee VMY. Abnormal tau phosphorylation at Ser396 in Alzheimer’s disease recapitulates development and contributes to reduced microtubule binding. Neuron. 1993;10(6):1089–1099. [DOI] [PubMed] [Google Scholar]
  • 154. Peel AL, Sorscher N, Kim JY, Galvan V, Chen S, Bredesen DE. Tau phosphorylation in Alzheimer’s disease: potential involvement of an APP-MAP kinase complex. Neuromol Med. 2004;5(3):205–218. [DOI] [PubMed] [Google Scholar]
  • 155. Pyo H, Jou I, Jung S, Hong S, Joe E. Mitogen-activated protein kinases activated by lipopolysaccharide and β-amyloid in cultured rat microglia. Neuroreport. 1998;9(5):871–874. [DOI] [PubMed] [Google Scholar]
  • 156. Munoz Ralay L, Ranaivo H, Roy SM, et al. A novel p38 alpha MAPK inhibitor suppresses brain proinflammatory cytokine up-regulation and attenuates synaptic dysfunction and behavioral deficits in an Alzheimer’s disease mouse model. J Neuroinflammation. 2007;4:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157. Li Y, Liu L, Barger SW, Griffin WST. Interleukin-1 mediates pathological effects of microglia on tau phosphorylation and on synaptophysin synthesis in cortical neurons through a p38-MAPK pathway. J Neurosci. 2003;23(5):1605–1611. [DOI] [PMC free article] [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. Amtul Z, Uhrig M, Wang L, Rozmahel RF, Beyreuther K. Detrimental effects of arachidonic acid and its metabolites in cellular and mouse models of Alzheimer’s disease: structural insight. Neurobiol Aging. 2012;33(4):831.e21–e31. [DOI] [PubMed] [Google Scholar]
  • 162. Blanco A, Alvarez S, Fresno M, Muñoz-Fernández MÁ. Amyloid-β induces cyclooxygenase-2 and PGE2 release in human astrocytes in NF-κ B dependent manner. J Alzheimers Dis. 2010;22(2):493–505. [DOI] [PubMed] [Google Scholar]
  • 163. Watanabe Y, Watanabe Y, Hamada K, et al. Distinct localization of prostaglandin D2, E2, and F2α binding sites in monkey brain. Brain Res. 1989;478(1):142–148. [DOI] [PubMed] [Google Scholar]
  • 164. Kaufmann WE, Worley PF, Pegg J, Bremer M, Isakson P. COX-2, a synaptically induced enzyme, is expressed by excitatory neurons at postsynaptic sites in rat cerebral cortex. Proc Natl Acad Sci U S A. 1996;93(6):2317–2321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165. Yasojima K, Schwab C, McGeer EG, McGeer PL. Distribution of cyclooxygenase-1 and cyclooxygenase-2 mRNA and proteins in human brain and peripheral organs. Brain Res. 1999;830(2):226–236. [DOI] [PubMed] [Google Scholar]
  • 166. Ho L, Pieroni L, Winger D, Purohit DP, Aisen PS, Pasinetti GM. Regional distribution of cyclooxygenase-2 in the hippocampal formation in Alzheimer’s disease. J Neurosci Res. 1999;57(3):295–303. [DOI] [PubMed] [Google Scholar]
  • 167. Xiang Z, Ho L, Yemul S, et al. Cyclooxygenase-2 promotes amyloid plaque deposition in a mouse model of Alzheimer’s disease neuropathology. Gene Expr. 2002;10(5-6):271–278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168. Qin W, Ho L, Pompl PN, et al. Cyclooxygenase (COX)-2 and COX-1 potentiate beta-amyloid peptide generation through mechanisms that involve gamma-secretase activity. J Biol Chem. 2003;278(51):50970–50977. [DOI] [PubMed] [Google Scholar]
  • 169. Rich JB, Rasmusson DX, Folstein MF, Carson KA, Kawas C, Brandt J. Nonsteroidal anti-inflammatory drugs in Alzheimer’s disease. Neurology. 1995;45(1):51–55. [DOI] [PubMed] [Google Scholar]
  • 170. McGeer PL, Schulzer M, McGeer EG. Arthritis and anti-inflammatory agents as possible protective factors for Alzheimer’s disease; a review of 17 epidemiologic studies. Neurology. 1996;47(2):425–432. [DOI] [PubMed] [Google Scholar]
  • 171. Fehér A, Juhász A, Rimanóczy A, Kálmán J, Janka Z. Association study of interferon-γ, cytosolic phospholipase A2, and cyclooxygenase-2 gene polymorphisms in Alzheimer disease. Am J Geriatr Psychiatry. 2010;18(11):983–987. [DOI] [PubMed] [Google Scholar]
  • 172. 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]
  • 173. Benish M, Bartal I, Goldfarb Y, et al. 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]
  • 174. Bains JS, Shaw CA. Neurodegenerative disorders in humans: the role of glutathione in oxidative stress-mediated neuronal death. Brain Res Rev. 1997;25(3):335–358. [DOI] [PubMed] [Google Scholar]
  • 175. Butterfield DA, Howard B, Yatin S, et al. Elevated oxidative stress in models of normal brain aging and Alzheimer’s disease. Life Sci. 1999;65(18-19):1883–1892. [DOI] [PubMed] [Google Scholar]
  • 176. de la Monte SM, Ganju N, Feroz N, et al. Oxygen free radical injury is sufficient to cause some Alzheimer-type molecular abnormalities in human CNS neuronal cells. J Alzheimers Dis. 2000;2(3-4):261–281. [DOI] [PubMed] [Google Scholar]
  • 177. López N, Tormo C, De Blas I, Llinares I, Alom J. Oxidative stress in Alzheimer’s disease and mild cognitive impairment with high sensitivity and specificity. J Alzheimers Dis. 2013;33(3):823–829. [DOI] [PubMed] [Google Scholar]
  • 178. Zemlan FP, Thienhaus OJ, Bosmann HB. Superoxide dismutase activity in Alzheimer’s disease: possible mechanism for paired helical filament formation. Brain Res. 1989;476(1):160–162. [DOI] [PubMed] [Google Scholar]
  • 179. Tesco G, Latorraca S, Piersanti P, Sorbi S, Piacentii S, Amaducci L. Free radical injury in skin cultured fibroblasts from Alzheimer’s disease patients. Ann NY Acad Sci. 1992;673:149–153. [DOI] [PubMed] [Google Scholar]
  • 180. Leuner K, Schütt T, Kurz C, et al. Mitochondrion-derived reactive oxygen species lead to enhanced amyloid beta formation. Antioxid Redox Signal. 2012;16(12):1421–1433 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181. 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]
  • 182. Mak IT, Chmielinska JJ, Nedelec L, Torres A, Weglicki WB. D-propranolol attenuates lysosomal iron accumulation and oxidative injury in endothelial cells. J Pharmacol Exp Ther. 2006;317(2):522–528. [DOI] [PubMed] [Google Scholar]
  • 183. Ma L, Gul R, Habibi J, Yang M, Pulakat L, 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]
  • 184. Schuman EM, Madison DV. A requirement for the intercellular messenger nitric oxide in long-term potentiation. Science. 1991;254(5037):1503–1506. [DOI] [PubMed] [Google Scholar]
  • 185. Zorumski CF, Izumi Y. Nitric oxide and hippocampal synaptic plasticity. Biochem Pharmacol. 1993;46(5):777–785. [DOI] [PubMed] [Google Scholar]
  • 186. Austin SA, Santhanam AV, Katusic ZS. Endothelial nitric oxide modulates expression and processing of amyloid precursor protein. Circ Res. 2010;107(12):1498–1502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187. Takahashi M, Chin Y, Nonaka T, Hasegawa M, Watanabe N, Arai T. Prolonged nitric oxide treatment induces tau aggregation in SH-SY5Y cells. Neurosci Lett. 2012;510(1):48–52. [DOI] [PubMed] [Google Scholar]
  • 188. De Servi B, La Porta CAM, Bontempelli M, Comolli R. Decrease of TGF-β1 plasma levels and increase of nitric oxide synthase activity in leukocytes as potential biomarkers of Alzheimer’s disease. Exper Gerontol. 2002;37(6):813–821. [DOI] [PubMed] [Google Scholar]
  • 189. Dorheim MA, Tracey WR, Pollock JS, Grammas P. Nitric oxide synthase activity is elevated in brain microvessels in Alzheimer’s disease. Biochem Biophys Res Commun. 1994;205(1):659–665. [DOI] [PubMed] [Google Scholar]
  • 190. Lüth HJ, Holzer M, Gärtner U, Staufenbiel M, Arendt T. Expression of endothelial and inducible NOS-isoforms is increased in Alzheimer’s disease, in APP23 transgenic mice and after experimental brain lesion in rat: evidence for an induction by amyloid pathology. Brain Res. 2001;913(1):57–67. [DOI] [PubMed] [Google Scholar]
  • 191. Nathan C, Calingasan N, Nezezon J, et al. Protection from Alzheimer’s-like disease in the mouse by genetic ablation of inducible nitric oxide synthase. J Exp Med. 2005;202(9):1163–1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192. 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]
  • 193. Reisberg B, Borenstein J, Salob SP, Ferris SH, Franssen E, Georgotas A. Behavioral symptoms in Alzheimer’s disease: phenomenology and treatment. J Clin Psychiatry. 1987;48(suppl):9–15. [PubMed] [Google Scholar]
  • 194. Mega MS, Cummings JL, Fiorello T, Gornbein J. The spectrum of behavioral changes in Alzheimer’s disease. Neurology. 1996;46(1):130–135. [DOI] [PubMed] [Google Scholar]
  • 195. Callahan MJ, Lipinski WJ, Bian F, Durham RA, Pack A, Walker LC. Augmented senile plaque load in aged female beta-amyloid precursor protein-transgenic mice. Am J Pathol. 2001;158(3):1173–1177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196. Bullido MJ, Ramos MC, Ruiz-Gómez A, et al. Polymorphism in genes involved in adrenergic signaling associated with Alzheimer’s. Neurobiol Aging. 2004;25(7):853–859 [DOI] [PubMed] [Google Scholar]
  • 197. Yu JT, Tan L, Ou JR, et al. Polymorphisms at the beta2-adrenergic receptor gene influence Alzheimer’s disease susceptibility. Brain Res. 2008;1210:216–222. [DOI] [PubMed] [Google Scholar]
  • 198. Liu CC, Kanekiyo T, Xu H, Bu G. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol. 2013;9(2):106–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199. Tsuang D, Leverenz JB, Lopez OL, et al. APOE ε4 increases risk for dementia in pure synucleinopathies. JAMA Neurol. 2013;70(2):223–228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200. Shimada K, Hirano E, Kimura T, Fujita M, Kishimoto C. Carvedilol reduces the severity of atherosclerosis in apolipoprotein E-deficient mice via reducing superoxide production. Exp Biol Med (Maywood). 2012;237(9):1039–1044 [DOI] [PubMed] [Google Scholar]
  • 201. Chen X, Xia Y, Alford M, et al. The CYP2D6B allele is associated with a milder synaptic pathology in Alzheimer’s disease. Ann Neurol. 1995;38(4):653–658. [DOI] [PubMed] [Google Scholar]
  • 202. Saitoh T, Xia Y, Chen X, et al. The CYP2D6B mutant allele is overrepresented in the Lewy body variant of Alzheimer’s disease. Ann Neurol. 1995;37(1):110–112. [DOI] [PubMed] [Google Scholar]
  • 203. Bradford LD. CYP2D6 allele frequency in European Caucasians, Asians, Africans and their descendants. Pharmacogenomics. 2002;3(2):229–243 [DOI] [PubMed] [Google Scholar]
  • 204. Lai ML, Wang SL, Lai MD, Lin ET, Tse M, Huang JD. Propranolol disposition in Chinese subjects of different CYP2D6 genotypes. Clin Pharmacol Ther. 1995;58(3):264–268. [DOI] [PubMed] [Google Scholar]
  • 205. Taguchi M, Nozawa T, Mizumaki K, et al. Nonlinear mixed effects model analysis of the pharmacokinetics of metoprolol in routinely treated Japanese patients. Biol Pharm Bull. 2004;27(10):1642–1648. [DOI] [PubMed] [Google Scholar]
  • 206. Takekuma Y, Takenaka T, Kiyokawa M, et al. Evaluation of effects of polymorphism for metabolic enzymes on pharmacokinetics of carvedilol by population pharmacokinetic analysis. Biol Pharm Bull. 2007;30(3):537–542. [DOI] [PubMed] [Google Scholar]
  • 207. Jin SK, Chung HJ, Chung MW, et al. Influence of CYP2D6*10 on the pharmacokinetics of metoprolol in healthy Korean volunteers. J Clin Pharm Ther. 2008;33(5):567–573. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Alzheimer's Disease and Other Dementias are provided here courtesy of SAGE Publications

RESOURCES