Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Alzheimers Dement ; 18(6): 1248-1259, 2022 06.
Article in English | MEDLINE | ID: mdl-34569707

ABSTRACT

INTRODUCTION: Tumor necrosis factor (TNF) inhibitors are widely used to treat rheumatoid arthritis (RA) and their potential to retard Alzheimer's disease (AD) progression has been reported. However, their long-term effects on the dementia/AD risk remain unknown. METHODS: A propensity scored matched retrospective cohort study was conducted among 40,207 patients with RA within the US Veterans Affairs health-care system from 2000 to 2020. RESULTS: A total of 2510 patients with RA prescribed TNF inhibitors were 1:2 matched to control patients. TNF inhibitor use was associated with reduced dementia risk (hazard ratio [HR]: 0.64, 95% confidence interval [CI]: 0.52-0.80), which was consistent as the study period increased from 5 to 20 years after RA diagnosis. TNF inhibitor use also showed a long-term effect in reducing the risk of AD (HR: 0.57, 95% CI: 0.39-0.83) during the 20 years of follow-up. CONCLUSION: TNF inhibitor use is associated with lower long-term risk of dementia/AD among US veterans with RA.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Dementia , Veterans , Antirheumatic Agents/adverse effects , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/epidemiology , Dementia/chemically induced , Dementia/epidemiology , Dementia/prevention & control , Humans , Propensity Score , Retrospective Studies , Tumor Necrosis Factor Inhibitors
2.
Alzheimers Res Ther ; 13(1): 177, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34670619

ABSTRACT

BACKGROUND: Interactions between the gut microbiota, microglia, and aging may modulate Alzheimer's disease (AD) pathogenesis but the precise nature of such interactions is not known. METHODS: We developed an integrated multi-dimensional, knowledge-driven, systems approach to identify interactions among microbial metabolites, microglia, and AD. Publicly available datasets were repurposed to create a multi-dimensional knowledge-driven pipeline consisting of an integrated network of microbial metabolite-gene-pathway-phenotype (MGPPN) consisting of 34,509 nodes (216 microbial metabolites, 22,982 genes, 1329 pathways, 9982 mouse phenotypes) and 1,032,942 edges. RESULTS: We evaluated the network-based ranking algorithm by showing that abnormal microglia function and physiology are significantly associated with AD pathology at both genetic and phenotypic levels: AD risk genes were ranked at the top 6.4% among 22,982 genes, P < 0.001. AD phenotypes were ranked at the top 11.5% among 9982 phenotypes, P < 0.001. A total of 8094 microglia-microbial metabolite-gene-pathway-phenotype-AD interactions were identified for top-ranked AD-associated microbial metabolites. Short-chain fatty acids (SCFAs) were ranked at the top among prioritized AD-associated microbial metabolites. Through data-driven analyses, we provided evidence that SCFAs are involved in microglia-mediated gut-microbiota-brain interactions in AD at both genetic, functional, and phenotypic levels. CONCLUSION: Our analysis produces a novel framework to offer insights into the mechanistic links between gut microbial metabolites, microglia, and AD, with the overall goal to facilitate disease mechanism understanding, therapeutic target identification, and designing confirmatory experimental studies.


Subject(s)
Alzheimer Disease , Gastrointestinal Microbiome , Alzheimer Disease/genetics , Animals , Brain , Mice , Microglia , Phenotype
3.
Neurobiol Dis ; 159: 105485, 2021 11.
Article in English | MEDLINE | ID: mdl-34411704

ABSTRACT

Fragile X Syndrome (FXS) is caused by silencing the FMR1 gene which results in intellectual disability, hyperactivity, sensory hypersensitivity, autistic-like behavior, and susceptibility to seizures. This X-linked disorder is also associated with reduced cAMP levels in humans as well as animal models. We assessed the therapeutic and neurochemical effects of chronic administration of the phosphodiesterase-4D negative allosteric modulator, BPN14770, in a mouse model of FXS (Fmr1 KO). Groups of male Fmr1 KO mice and control littermates were treated with dietary BPN14770 commencing postnatal day 21. A dose-response effect was investigated. At 90 days of age, mice underwent behavior tests including open field, novel object recognition, three chambered sociability and social novelty tests, passive avoidance, and sleep duration analysis. These tests were followed by in vivo measurement of regional rates of cerebral protein synthesis (rCPS) with the autoradiographic L-[1-14C]leucine method. BPN14770 treatment had positive effects on the behavioral phenotype in Fmr1 KO mice. Some effects such as increased sleep duration and increased social behavior occurred in both genotypes. In the open field, the hyperactivity response in Fmr1 KO mice was ameliorated by BPN14770 treatment at low and intermediate doses. BPN14770 treatment tended to increase rCPS in a dose-dependent manner in WT mice, whereas in Fmr1 KO mice effects on rCPS were less apparent. Results indicate BPN14770 treatment improves some behavior in Fmr1 KO mice. Results also suggest a genotype difference in the regulation of translation via a cAMP-dependent pathway.


Subject(s)
Behavior, Animal , Cerebrum , Fragile X Syndrome , Phosphodiesterase 4 Inhibitors , Protein Biosynthesis , Sleep , Animals , Mice , Allosteric Regulation , Autoradiography , Behavior, Animal/drug effects , Cerebrum/drug effects , Cerebrum/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Disease Models, Animal , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/metabolism , Mice, Knockout , Phosphodiesterase 4 Inhibitors/pharmacology , Protein Biosynthesis/drug effects , Sleep/drug effects , Social Behavior
4.
Nat Med ; 27(5): 862-870, 2021 05.
Article in English | MEDLINE | ID: mdl-33927413

ABSTRACT

The goal of this study was to determine whether a phosphodiesterase-4D (PDE4D) allosteric inhibitor (BPN14770) would improve cognitive function and behavioral outcomes in patients with fragile X syndrome (FXS). This phase 2 trial was a 24-week randomized, placebo-controlled, two-way crossover study in 30 adult male patients (age 18-41 years) with FXS. Participants received oral doses of BPN14770 25 mg twice daily or placebo. Primary outcomes were prespecified as safety and tolerability with secondary efficacy outcomes of cognitive performance, caregiver rating scales and physician rating scales (ClinicalTrials.gov identifier: NCT03569631 ). The study met the primary outcome measure since BPN14770 was well tolerated with no meaningful differences between the active and placebo treatment arms. The study also met key secondary efficacy measures of cognition and daily function. Cognitive benefit was demonstrated using the National Institutes of Health Toolbox Cognition Battery assessments of Oral Reading Recognition (least squares mean difference +2.81, P = 0.0157), Picture Vocabulary (+5.81, P = 0.0342) and Cognition Crystallized Composite score (+5.31, P = 0.0018). Benefit as assessed by visual analog caregiver rating scales was judged to be clinically meaningful for language (+14.04, P = 0.0051) and daily functioning (+14.53, P = 0.0017). Results from this study using direct, computer-based assessment of cognitive performance by adult males with FXS indicate significant cognitive improvement in domains related to language with corresponding improvement in caregiver scales rating language and daily functioning.


Subject(s)
Cognition/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Fragile X Syndrome/drug therapy , Phosphodiesterase 4 Inhibitors/therapeutic use , Adolescent , Adult , Cross-Over Studies , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Humans , Language Tests , Male , Placebos/administration & dosage , Psychometrics/statistics & numerical data , Treatment Outcome , Young Adult
5.
Alzheimers Dement ; 17(8): 1297-1306, 2021 08.
Article in English | MEDLINE | ID: mdl-33559975

ABSTRACT

INTRODUCTION: At present, there is limited data on the risks, disparity, and outcomes for COVID-19 in patients with dementia in the United States. METHODS: This is a retrospective case-control analysis of patient electronic health records (EHRs) of 61.9 million adult and senior patients (age ≥ 18 years) in the United States up to August 21, 2020. RESULTS: Patients with dementia were at increased risk for COVID-19 compared to patients without dementia (adjusted odds ratio [AOR]: 2.00 [95% confidence interval (CI), 1.94-2.06], P < .001), with the strongest effect for vascular dementia (AOR: 3.17 [95% CI, 2.97-3.37], P < .001), followed by presenile dementia (AOR: 2.62 [95% CI, 2.28-3.00], P < .001), Alzheimer's disease (AOR: 1.86 [95% CI, 1.77-1.96], P < .001), senile dementia (AOR: 1.99 [95% CI, 1.86-2.13], P < .001) and post-traumatic dementia (AOR: 1.67 [95% CI, 1.51-1.86] P < .001). Blacks with dementia had higher risk of COVID-19 than Whites (AOR: 2.86 [95% CI, 2.67-3.06], P < .001). The 6-month mortality and hospitalization risks in patients with dementia and COVID-19 were 20.99% and 59.26%, respectively. DISCUSSION: These findings highlight the need to protect patients with dementia as part of the strategy to control the COVID-19 pandemic.


Subject(s)
COVID-19/complications , Dementia/complications , Adolescent , Adult , Aged , Aged, 80 and over , Alzheimer Disease/complications , Alzheimer Disease/epidemiology , Black People , Brain Injuries, Traumatic/complications , Brain Injuries, Traumatic/epidemiology , COVID-19/epidemiology , Case-Control Studies , Dementia/epidemiology , Dementia, Vascular/complications , Dementia, Vascular/epidemiology , Demography , Electronic Health Records , Female , Healthcare Disparities , Hospitalization/statistics & numerical data , Humans , Male , Middle Aged , Retrospective Studies , Treatment Outcome , United States/epidemiology , White People , Young Adult
6.
Front Cell Dev Biol ; 8: 599389, 2020.
Article in English | MEDLINE | ID: mdl-33363155

ABSTRACT

A global, quantitative proteomics/systems-biology analysis of the selective pharmacological inhibition of phosphodiesterase-4D (PDE4D) revealed the differential regulation of pathways associated with neuroplasticity in memory-associated brain regions. Subtype selective inhibitors of PDE4D bind in an allosteric site that differs between mice and humans in a single amino acid (tyrosine vs. phenylalanine, respectively). Therefore to study selective inhibition of PDE4D by BPN14770, a subtype selective allosteric inhibitor of PDE4D, we utilized a line of mice in which the PDE4D gene had been humanized by mutating the critical tyrosine to phenylalanine. Relatively low doses of BPN14770 were effective at reversing scopolamine-induced memory and cognitive deficits in humanized PDE4D mice. Inhibition of PDE4D alters the expression of protein kinase A (PKA), Sirt1, Akt, and Bcl-2/Bax which are components of signaling pathways for regulating endocrine response, stress resistance, neuronal autophagy, and apoptosis. Treatment with a series of antagonists, such as H89, sirtinol, and MK-2206, reversed the effect of BPN14770 as shown by behavioral tests and immunoblot analysis. These findings suggest that inhibition of PDE4D enhances signaling through the cAMP-PKA-SIRT1-Akt -Bcl-2/Bax pathway and thereby may provide therapeutic benefit in neurocognitive disorders.

7.
Oncogene ; 39(19): 3791-3802, 2020 05.
Article in English | MEDLINE | ID: mdl-32203163

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDE) break down cyclic nucleotides such as cAMP and cGMP, reducing the signaling of these important intracellular second messengers. Several unique families of phosphodiesterases exist, and certain families are clinically important modulators of vasodilation. In the current work, we have summarized the body of literature that describes an emerging role for the PDE4 subfamily of phosphodiesterases in malignancy. We have systematically investigated PDE4A, PDE4B, PDE4C, and PDE4D isoforms and found evidence associating them with several cancer types including hematologic malignancies and lung cancers, among others. In this review, we compare the evidence examining the functional role of each PDE4 subtype across malignancies, looking for common signaling themes, signaling pathways, and establishing the case for PDE4 subtypes as a potential therapeutic target for cancer treatment.


Subject(s)
Hematologic Neoplasms/genetics , Lung Neoplasms/genetics , Protein Isoforms/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Hematologic Neoplasms/classification , Hematologic Neoplasms/pathology , Humans , Lung Neoplasms/classification , Lung Neoplasms/pathology , Protein Isoforms/classification , Signal Transduction/genetics
8.
PLoS One ; 15(3): e0229819, 2020.
Article in English | MEDLINE | ID: mdl-32203525

ABSTRACT

This large, retrospective case-control study of electronic health records from 56 million unique adult patients examined whether or not treatment with a Tumor Necrosis Factor (TNF) blocking agent is associated with lower risk for Alzheimer's disease (AD) in patients with rheumatoid arthritis (RA), psoriasis, and other inflammatory diseases which are mediated in part by TNF and for which a TNF blocker is an approved treatment. The analysis compared the diagnosis of AD as an outcome measure in patients receiving at least one prescription for a TNF blocking agent (etanercept, adalimumab, and infliximab) or for methotrexate. Adjusted odds ratios (AORs) were estimated using the Cochran-Mantel-Haenszel (CMH) method and presented with 95% confidence intervals (CIs) and p-values. RA was associated with a higher risk for AD (Adjusted Odds Ratio (AOR) = 2.06, 95% Confidence Interval: (2.02-2.10), P-value <0.0001) as did psoriasis (AOR = 1.37 (1.31-1.42), P <0.0001), ankylosing spondylitis (AOR = 1.57 (1.39-1.77), P <0.0001), inflammatory bowel disease (AOR = 2.46 (2.33-2.59), P < 0.0001), ulcerative colitis (AOR = 1.82 (1.74-1.91), P <0.0001), and Crohn's disease (AOR = 2.33 (2.22-2.43), P <0.0001). The risk for AD in patients with RA was lower among patients treated with etanercept (AOR = 0.34 (0.25-0.47), P <0.0001), adalimumab (AOR = 0.28 (0.19-0.39), P < 0.0001), or infliximab (AOR = 0.52 (0.39-0.69), P <0.0001). Methotrexate was also associated with a lower risk for AD (AOR = 0.64 (0.61-0.68), P <0.0001), while lower risk was found in patients with a prescription history for both a TNF blocker and methotrexate. Etanercept and adalimumab also were associated with lower risk for AD in patients with psoriasis: AOR = 0.47 (0.30-0.73 and 0.41 (0.20-0.76), respectively. There was no effect of gender or race, while younger patients showed greater benefit from a TNF blocker than did older patients. This study identifies a subset of patients in whom systemic inflammation contributes to risk for AD through a pathological mechanism involving TNF and who therefore may benefit from treatment with a TNF blocking agent.


Subject(s)
Alzheimer Disease/epidemiology , Antirheumatic Agents/therapeutic use , Arthritis, Psoriatic/drug therapy , Arthritis, Rheumatoid/drug therapy , Tumor Necrosis Factor Inhibitors/therapeutic use , Adalimumab/therapeutic use , Adolescent , Adult , Aged , Arthritis, Psoriatic/epidemiology , Arthritis, Rheumatoid/epidemiology , Case-Control Studies , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/epidemiology , Etanercept/therapeutic use , Female , Humans , Infliximab/therapeutic use , Male , Methotrexate/therapeutic use , Middle Aged , Retrospective Studies , Risk , Young Adult
9.
ACS Chem Neurosci ; 11(9): 1311-1323, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32212718

ABSTRACT

We aimed to develop radioligands for PET imaging of brain phosphodiesterase subtype 4D (PDE4D), a potential target for developing cognition enhancing or antidepressive drugs. Exploration of several chemical series gave four leads with high PDE4D inhibitory potency and selectivity, optimal lipophilicity, and good brain uptake. These leads featured alkoxypyridinyl cores. They were successfully labeled with carbon-11 (t1/2 = 20.4 min) for evaluation with PET in monkey. Whereas two of these radioligands did not provide PDE4D-specific signal in monkey brain, two others, [11C]T1660 and [11C]T1650, provided sizable specific signal, as judged by pharmacological challenge using rolipram or a selective PDE4D inhibitor (BPN14770) and subsequent biomathematical analysis. Specific binding was highest in prefrontal cortex, temporal cortex, and hippocampus, regions that are important for cognitive function. [11C]T1650 was progressed to evaluation in humans with PET, but the output measure of brain enzyme density (VT) increased with scan duration. This instability over time suggests that radiometabolite(s) were accumulating in the brain. BPN14770 blocked PDE4D uptake in human brain after a single dose, but the percentage occupancy was difficult to estimate because of the unreliability of measuring VT. Overall, these results show that imaging of PDE4D in primate brain is feasible but that further radioligand refinement is needed, most likely to avoid problematic radiometabolites.


Subject(s)
Brain , Positron-Emission Tomography , Brain/diagnostic imaging , Brain/metabolism , Carbon Radioisotopes , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Radiopharmaceuticals , Rolipram/pharmacology
10.
J Pharmacol Exp Ther ; 371(2): 250-259, 2019 11.
Article in English | MEDLINE | ID: mdl-31488603

ABSTRACT

Recent imaging studies of amyloid and tau in cognitively normal elderly subjects imply that Alzheimer's pathology can be tolerated by the brain to some extent due to compensatory mechanisms operating at the cellular and synaptic levels. The present study investigated the effects of an allosteric inhibitor of phosphodiesterase-4D (PDE4D), known as BPN14770 (2-(4-((2-(3-Chlorophenyl)-6-(trifluoromethyl)pyridin-4-yl)methyl)phenyl)acetic Acid), on impairment of memory, dendritic structure, and synaptic proteins induced by bilateral microinjection of oligomeric amyloid beta (Aß 1-42 into the hippocampus of humanized PDE4D (hPDE4D) mice. The hPDE4D mice provide a unique and powerful genetic tool for assessing PDE4D target engagement. Behavioral studies showed that treatment with BPN14770 significantly improved memory acquisition and retrieval in the Morris water maze test and the percentage of alternations in the Y-maze test in the model of Aß impairment. Microinjection of oligomeric Aß 1-42 caused decreases in the number of dendrites, dendritic length, and spine density of pyramid neurons in the hippocampus. These changes were prevented by BPN14770 in a dose-dependent manner. Furthermore, molecular studies showed that BPN14770 prevented Aß-induced decreases in synaptophysin, postsynaptic density protein 95, phosphorylated cAMP-response element binding protein (CREB)/CREB, brain-derived neurotrophic factor, and nerve growth factor inducible protein levels in the hippocampus. The protective effects of BPN14770 against Aß-induced memory deficits, synaptic damage, and the alteration in the cAMP-meditated cell signaling cascade were blocked by H-89 (N-[2-(p-Bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide dihydrochloride), an inhibitor of protein kinase A. These results suggest that BPN14770 may activate compensatory mechanisms that support synaptic health even with the onset of amyloid pathology in Alzheimer's disease. SIGNIFICANCE STATEMENT: This study demonstrates that a phosphodiesterase-4D allosteric inhibitor, BPN14770, protects against memory loss and neuronal atrophy induced by oligomeric Aß 1-42. The study provides useful insight into the potential role of compensatory mechanisms in Alzheimer's disease in a model of oligomeric Aß 1-42 neurotoxicity.


Subject(s)
Amyloid beta-Peptides/toxicity , Hippocampus/drug effects , Memory Disorders/chemically induced , Memory Disorders/prevention & control , Neuroprotective Agents/therapeutic use , Peptide Fragments/toxicity , Phosphodiesterase 4 Inhibitors/therapeutic use , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Gene Knock-In Techniques , Hippocampus/pathology , Humans , Memory Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroprotective Agents/pharmacology , Phosphodiesterase 4 Inhibitors/pharmacology , Random Allocation
11.
J Med Chem ; 62(10): 4884-4901, 2019 05 23.
Article in English | MEDLINE | ID: mdl-31013090

ABSTRACT

Novel pyridine- and pyrimidine-based allosteric inhibitors are reported that achieve PDE4D subtype selectivity through recognition of a single amino acid difference on a key regulatory domain, known as UCR2, that opens and closes over the catalytic site for cAMP hydrolysis. The design and optimization of lead compounds was based on iterative analysis of X-ray crystal structures combined with metabolite identification. Selectivity for the activated, dimeric form of PDE4D provided potent memory enhancing effects in a mouse model of novel object recognition with improved tolerability and reduced vascular toxicity over earlier PDE4 inhibitors that lack subtype selectivity. The lead compound, 28 (BPN14770), has entered midstage, human phase 2 clinical trials for the treatment of Fragile X Syndrome.


Subject(s)
Brain Diseases/drug therapy , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Drug Design , Fragile X Syndrome/drug therapy , Phosphodiesterase 4 Inhibitors/chemical synthesis , Allosteric Regulation/drug effects , Animals , Behavior, Animal/drug effects , Brain Diseases/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Fragile X Syndrome/enzymology , Humans , Inhibitory Concentration 50 , Male , Mice, Inbred ICR , Molecular Structure , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/pharmacology , Structure-Activity Relationship
12.
Front Mol Neurosci ; 12: 22, 2019.
Article in English | MEDLINE | ID: mdl-30800055

ABSTRACT

Recent, large-scale, genome-wide association studies (GWAS) provide a first view of the genetic fine structure of cognitive performance in healthy individuals. These studies have pooled data from up to 1.1 million subjects based on simple measures of cognitive performance including educational attainment, self-reported math ability, highest math class taken, and pooled, normalized scores from cognitive tests. These studies now allow the genome-wide interrogation of genes and pathways for their potential impact on human cognitive performance. The phosphodiesterase (PDE) enzymes regulate key cyclic nucleotide signaling pathways. Many are expressed in the brain and have been the targets of CNS drug discovery. Genetic variation in PDE1C, PDE4B and PDE4D associates with multiple measures of human cognitive function. The large size of the human PDE4B and PDE4D genes allows genetic fine structure mapping to transcripts encoding dimeric (long) forms of the enzymes. Upstream and downstream effectors of the cAMP pathway modulated by PDE4D [adenylate cyclase 1 (ADCY1), ADCY8, PRKAR1A, CREB1, or CREBBP] did not show genetic association with cognitive performance, however, genetic association was seen with brain derived neurotrophic factor (BDNF), a gene whose expression is modulated by cAMP. Notably absent was genetic association in healthy subjects to targets of CNS drug discovery designed to improve cognition in disease states by the modulation of cholinergic [acetylcholinesterase (ACHE), choline acetyltransferase (CHAT), nicotinic alpha 7 acetylcholine receptor (CHRNA7)], serotonergic (HTR6), histaminergic (HRH3), or glutamatergic (GRM5) pathways. These new data provide a rationale for exploring the therapeutic benefit of selective inhibitors of PDE1C, PDE4B and PDE4D in CNS disorders affecting cognition.

13.
Neuropsychopharmacology ; 43(11): 2299-2309, 2018 10.
Article in English | MEDLINE | ID: mdl-30131563

ABSTRACT

Inhibitors of phosphodiesterase-4 (PDE4) have beneficial effects on memory in preclinical and clinical studies. Development of these drugs has stalled due to dose-limiting side effects of nausea and emesis. While use of subtype-selective inhibitors (i.e., for PDE4A, B, or D) could overcome this issue, conservation of the catalytic region, to which classical inhibitors bind, limits this approach. The present study examined the effects of BPN14770, an allosteric inhibitor of PDE4D, which binds to a primate-specific, N-terminal region. In mice engineered to express PDE4D with this primate-specific sequence, BPN14770 was 100-fold more potent for improving memory than in wild-type mice; meanwhile, it exhibited low potency in a mouse surrogate model for emesis. BPN14770 also antagonized the amnesic effects of scopolamine, increased cAMP signaling in brain, and increased BDNF and markers of neuronal plasticity associated with memory. These data establish a relationship between PDE4D target engagement and effects on memory for BPN14770 and suggest clinical potential for PDE4D-selective inhibitors.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Memory/drug effects , Phosphodiesterase 4 Inhibitors/metabolism , Phosphodiesterase 4 Inhibitors/pharmacology , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Crystallography, X-Ray , Dose-Response Relationship, Drug , Humans , Maze Learning/drug effects , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphodiesterase 4 Inhibitors/chemistry , Protein Binding/physiology
14.
Neurobiol Learn Mem ; 148: 38-49, 2018 02.
Article in English | MEDLINE | ID: mdl-29294383

ABSTRACT

Traumatic brain injury (TBI) significantly decreases cyclic AMP (cAMP) signaling which produces long-term synaptic plasticity deficits and chronic learning and memory impairments. Phosphodiesterase 4 (PDE4) is a major family of cAMP hydrolyzing enzymes in the brain and of the four PDE4 subtypes, PDE4D in particular has been found to be involved in memory formation. Although most PDE4 inhibitors target all PDE4 subtypes, PDE4D can be targeted with a selective, negative allosteric modulator, D159687. In this study, we hypothesized that treating animals with D159687 could reverse the cognitive deficits caused by TBI. To test this hypothesis, adult male Sprague Dawley rats received sham surgery or moderate parasagittal fluid-percussion brain injury. After 3 months of recovery, animals were treated with D159687 (0.3 mg/kg, intraperitoneally) at 30 min prior to cue and contextual fear conditioning, acquisition in the water maze or during a spatial working memory task. Treatment with D159687 had no significant effect on these behavioral tasks in non-injured, sham animals, but did reverse the learning and memory deficits in chronic TBI animals. Assessment of hippocampal slices at 3 months post-TBI revealed that D159687 reversed both the depression in basal synaptic transmission in area CA1 as well as the late-phase of long-term potentiation. These results demonstrate that a negative allosteric modulator of PDE4D may be a potential therapeutic to improve chronic cognitive dysfunction following TBI.


Subject(s)
Brain Injuries, Traumatic/complications , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/physiopathology , Hippocampus/drug effects , Learning/drug effects , Learning/physiology , Long-Term Potentiation/drug effects , Phosphodiesterase 4 Inhibitors/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Benzhydryl Compounds/pharmacology , Cognitive Dysfunction/etiology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Fear/physiology , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Phenylurea Compounds/pharmacology , Phosphodiesterase 4 Inhibitors/administration & dosage , Rats , Rats, Sprague-Dawley , Spatial Memory/drug effects , Spatial Memory/physiology
15.
JMIR Res Protoc ; 7(1): e20, 2018 Jan 24.
Article in English | MEDLINE | ID: mdl-29367186

ABSTRACT

BACKGROUND: This report provides data on the use of social media advertising as a clinical trial recruitment strategy targeting healthy volunteers aged 60 years and older. The social media advertising campaign focused on enrollment for a Phase 1 clinical trial. Traditional means of recruiting-billboards, newspaper advertising, word of mouth, personal referrals, and direct mail-were not producing enough qualified participants. OBJECTIVE: To demonstrate the effectiveness of using targeted advertising on the social networking site Facebook to recruit people aged 60 years and older for volunteer clinical trial participation. METHODS: The trial sponsor used a proactive approach to recruit participants using advertising on social media. The sponsor placed and monitored an Institutional Review Board-approved advertising campaign on Facebook to recruit potential candidates for a Phase 1 clinical trial. The clinical trial required a 10-day residential (overnight) stay at a clinic in Michigan, with one follow-up visit. The sponsor of the clinical trial placed the advertising, which directed interested respondents to a trial-specific landing page controlled by the Contract Research Organization (CRO). The CRO provided all follow-up consenting, prescreening, screening, and enrollment procedures. The campaign was waged over an 8-week period to supplement recruiting by the CRO. RESULTS: A total of 621 people responded to a Facebook advertising campaign by completing an online form or telephoning the CRO, and the clinical trial was fully enrolled at 45 subjects following an 8-week Facebook advertising campaign. CONCLUSIONS: An 8-week Facebook advertising campaign contributed to 868 inquiries made regarding a Phase 1 clinical trial seeking to enroll healthy elderly subjects. Over the initial 11 weeks of recruitment, 178 inquiries were received using traditional methods of outreach. Respondents to the Facebook advertising campaign described in this report engaged with the sponsored advertising at a higher rate than is typical for social media-based clinical trial recruitment strategies. The older adults' engagement rate of 4.92% was more than twice as high as click-through rates of younger adults engaged with social media advertising in other clinical trial recruitment studies. Advertising placed on the social media platform Facebook is effective with the healthy volunteer population aged 60 years and older. This approach can quickly and cost-effectively reach qualified candidates for clinical trial recruitment as a supplement to traditional means of recruiting. TRIAL REGISTRATION: ClinicalTrials.gov: NCT02840279; https://clinicaltrials.gov/ct2/show/NCT02840279 (Archived by WebCite at http://www.webcitation.org/6wamIWXAt).

16.
Sci Rep ; 7(1): 14653, 2017 11 07.
Article in English | MEDLINE | ID: mdl-29116166

ABSTRACT

Fragile-X syndrome (FXS) patients display intellectual disability and autism spectrum disorder due to silencing of the X-linked, fragile-X mental retardation-1 (FMR1) gene. Dysregulation of cAMP metabolism is a consistent finding in patients and in the mouse and fly FXS models. We therefore explored if BPN14770, a prototypic phosphodiesterase-4D negative allosteric modulator (PDE4D-NAM) in early human clinical trials, might provide therapeutic benefit in the mouse FXS model. Daily treatment of adult male fmr1 C57Bl6 knock-out mice with BPN14770 for 14 days reduced hyperarousal, improved social interaction, and improved natural behaviors such as nesting and marble burying as well as dendritic spine morphology. There was no decrement in behavioral scores in control C57Bl6 treated with BPN14770. The behavioral benefit of BPN14770 persisted two weeks after washout of the drug. Thus, BPN14770 may be useful for the treatment of fragile-X syndrome and other disorders with decreased cAMP signaling.


Subject(s)
Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Fragile X Syndrome/drug therapy , Phosphodiesterase 4 Inhibitors/therapeutic use , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Fragile X Mental Retardation Protein/metabolism , Fragile X Syndrome/psychology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype
17.
PLoS One ; 12(5): e0178013, 2017.
Article in English | MEDLINE | ID: mdl-28542295

ABSTRACT

Traumatic brain injury (TBI) initiates a deleterious inflammatory response that exacerbates pathology and worsens outcome. This inflammatory response is partially mediated by a reduction in cAMP and a concomitant upregulation of cAMP-hydrolyzing phosphodiesterases (PDEs) acutely after TBI. The PDE4B subfamily, specifically PDE4B2, has been found to regulate cAMP in inflammatory cells, such as neutrophils, macrophages and microglia. To determine if PDE4B regulates inflammation and subsequent pathology after TBI, adult male Sprague Dawley rats received sham surgery or moderate parasagittal fluid-percussion brain injury (2 ± 0.2 atm) and were then treated with a PDE4B - selective inhibitor, A33, or vehicle for up to 3 days post-surgery. Treatment with A33 reduced markers of microglial activation and neutrophil infiltration at 3 and 24 hrs after TBI, respectively. A33 treatment also reduced cortical contusion volume at 3 days post-injury. To determine whether this treatment paradigm attenuated TBI-induced behavioral deficits, animals were evaluated over a period of 6 weeks after surgery for forelimb placement asymmetry, contextual fear conditioning, water maze performance and spatial working memory. A33 treatment significantly improved contextual fear conditioning and water maze retention at 24 hrs post-training. However, this treatment did not rescue sensorimotor or working memory deficits. At 2 months after surgery, atrophy and neuronal loss were measured. A33 treatment significantly reduced neuronal loss in the pericontusional cortex and hippocampal CA3 region. This treatment paradigm also reduced cortical, but not hippocampal, atrophy. Overall, these results suggest that acute PDE4B inhibition may be a viable treatment to reduce inflammation, pathology and memory deficits after TBI.


Subject(s)
Brain Injuries, Traumatic/prevention & control , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Maze Learning/drug effects , Memory Disorders/drug therapy , Microglia/drug effects , Pyrimidines/pharmacology , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Cells, Cultured , Cyclic AMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Male , Microglia/metabolism , Rats , Rats, Sprague-Dawley
18.
Sci Rep ; 7: 40115, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28054669

ABSTRACT

Inhibition of cyclic AMP (cAMP)-specific phosphodiesterase 4 (PDE4) has been proposed as a potential treatment for a series of neuropsychological conditions such as depression, anxiety and memory loss. However, the specific involvement of each of the PDE4 subtypes (PDE4A, 4B and 4C) in different categories of behavior has yet to be elucidated. In the present study, we compared the possible pharmacological effects of PDE4B and PDE4D selective inhibitors, A-33 and D159687, in mediating neurological function in mice. Both compounds were equally potent in stimulating cAMP signaling in the mouse hippocampal cell line HT-22 leading to an increase in CREB phosphorylation. In contrast, A-33 and D159687 displayed distinct neuropharmacological effects in mouse behavioral tests. A-33 has an antidepressant-like profile as indicated by reduced immobility time in the forced swim and tail suspension tasks, as well as reduced latency to feed in the novelty suppressed feeding test. D159687, on the other hand, had a procognitive profile as it improved memory in the novel object recognition test but had no antidepressant or anxiolytic benefit. The present data suggests that inhibitors targeting specific subtypes of PDE4 may exhibit differential pharmacological effects and aid a more efficient pharmacotherapy towards neuropsychological conditions.


Subject(s)
Central Nervous System/drug effects , Central Nervous System/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Enzyme Inhibitors/pharmacology , Psychotropic Drugs/pharmacology , Animals , Behavior, Animal/drug effects , Cell Line , Cyclic AMP/metabolism , Enzyme Inhibitors/administration & dosage , Mice , Psychotropic Drugs/administration & dosage , Signal Transduction/drug effects
19.
J Neurosci ; 36(27): 7095-108, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27383587

ABSTRACT

UNLABELLED: Learning and memory impairments are common in traumatic brain injury (TBI) survivors. However, there are no effective treatments to improve TBI-induced learning and memory impairments. TBI results in decreased cAMP signaling and reduced cAMP-response-element binding protein (CREB) activation, a critical pathway involved in learning and memory. TBI also acutely upregulates phosphodiesterase 4B2 (PDE4B2), which terminates cAMP signaling by hydrolyzing cAMP. We hypothesized that a subtype-selective PDE4B inhibitor could reverse the learning deficits induced by TBI. To test this hypothesis, adult male Sprague-Dawley rats received sham surgery or moderate parasagittal fluid-percussion brain injury. At 3 months postsurgery, animals were administered a selective PDE4B inhibitor or vehicle before cue and contextual fear conditioning, water maze training and a spatial working memory task. Treatment with the PDE4B inhibitor significantly reversed the TBI-induced deficits in cue and contextual fear conditioning and water maze retention. To further understand the underlying mechanisms of these memory impairments, we examined hippocampal long-term potentiation (LTP). TBI resulted in a significant reduction in basal synaptic transmission and impaired expression of LTP. Treatment with the PDE4B inhibitor significantly reduced the deficits in basal synaptic transmission and rescued LTP expression. The PDE4B inhibitor reduced tumor necrosis factor-α levels and increased phosphorylated CREB levels after TBI, suggesting that this drug inhibited molecular pathways in the brain known to be regulated by PDE4B. These results suggest that a subtype-selective PDE4B inhibitor is a potential therapeutic to reverse chronic learning and memory dysfunction and deficits in hippocampal synaptic plasticity following TBI. SIGNIFICANCE STATEMENT: Currently, there are an estimated 3.2-5.3 million individuals living with disabilities from traumatic brain injury (TBI) in the United States, and 8 of 10 of these individuals report cognitive disabilities (Thurman et al., 1999; Lew et al., 2006; Zaloshnja et al., 2008). One of the molecular mechanisms associated with chronic cognitive disabilities is impaired cAMP signaling in the hippocampus. In this study, we report that a selective phosphodiesterase 4B (PDE4B) inhibitor reduces chronic cognitive deficits after TBI and rescues deficits in hippocampal long-term potentiation. These results suggest that PDE4B inhibition has the potential to improve learning and memory ability and overall functioning for people living with TBI.


Subject(s)
Brain Injuries, Traumatic/complications , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Phosphodiesterase 4 Inhibitors/therapeutic use , Animals , Conditioning, Classical/drug effects , Disease Models, Animal , Fear/drug effects , Hippocampus/drug effects , Hippocampus/physiology , Interleukin-1beta/metabolism , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Memory, Short-Term/drug effects , Nerve Tissue Proteins/metabolism , Phenylacetates/pharmacology , Phenylacetates/therapeutic use , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Thiophenes/pharmacology , Thiophenes/therapeutic use , Tumor Necrosis Factor-alpha/metabolism
20.
Neurotherapeutics ; 12(1): 49-56, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25371167

ABSTRACT

Between 20% and 25% of patients diagnosed with Alzheimer's disease (AD) do not have amyloid burden as assessed by positron emission tomography imaging. Thus, there is a need for nonamyloid-directed therapies for AD, especially for those patients with non-amyloid AD. The family of phosphodiesterase-4 (PDE4) enzymes are underexploited therapeutic targets for central nervous system indications. While the PDE4A, B, and D subtypes are expressed in brain, the strict amino acid sequence conservation of the active site across the four subtypes of PDE4 has made it difficult to discover subtype inhibitors. The recent elucidation of the structure of the PDE4 N- and C-terminal regulatory domains now makes it possible to design subtype-selective, negative allosteric modulators (PDE4-NAMs). These act through closing the N-terminal UCR2 or C-terminal CR3 regulatory domains, and thereby inhibit the enzyme by blocking access of cyclic adenosine monophosphate (cAMP) to the active site. PDE4B-NAMs have the potential to reduce neuroinflammation by dampening microglia cytokine production triggered by brain amyloid, while PDE4D-NAMs have potent cognitive benefit by augmenting signaling through the cAMP/protein kinase A/cAMP response element-binding protein (CREB) pathway for memory consolidation. The importance of PDE4D for human cognition is underscored by the recent discovery of PDE4D mutations in acrodysostosis (ACRDY2: MIM 600129), an ultra rare disorder associated with intellectual disability. Thus, the family of PDE4 enzymes provides rich opportunities for the development of mechanistically novel drugs to treat neuroinflammation or the cognitive deficits in AD.


Subject(s)
Alzheimer Disease/enzymology , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Alzheimer Disease/pathology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...