Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Trends Pharmacol Sci ; 45(3): 197-209, 2024 03.
Article in English | MEDLINE | ID: mdl-38360510

ABSTRACT

Alzheimer's disease (AD), similar to AD-related dementias, is characterized by impaired/lost neuronal structures and functions due to a long progression of neurodegeneration. Derailed endogenous signal pathways and disease processes have critical roles in neurodegeneration and are pharmacological targets in inducing neuroregeneration. Pharmacologically switching/shifting the brain status from neurodegeneration to neuroregeneration is emerging as a new therapeutic concept, one that is not only achievable, but also essential for effective therapy for AD. The results of the pharmacological-induced shift from neurodegeneration to neuroregeneration are twofold: arresting cognitive deterioration (and directing the brain toward cognitive recovery) in established AD, and preventing neurodegeneration through building up cognitive resilience in patients with preclinical or probable AD. In this review, we discuss these new developments in AD pharmacology and relevant clinical trials.


Subject(s)
Alzheimer Disease , Cognition Disorders , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Brain/metabolism , Neurons/metabolism , Nerve Regeneration
2.
J Alzheimers Dis ; 96(2): 759-766, 2023.
Article in English | MEDLINE | ID: mdl-37742657

ABSTRACT

BACKGROUND: In pre-clinical studies, Bryostatin, MW (molecular weight) 904, has demonstrated synaptogenic, anti-apoptotic, anti-amyloid, and anti-tau tangle efficacies. OBJECTIVE: To identify AD patients who show significant cognitive benefit versus placebo when treated in a trial with chronic Bryostatin dosing. METHODS: In this 6-month 122 AD patient Bryostatin trial, there were two cohorts: the Moderate Cohort (MMSE, Mini-Mental Status Exam: 15-18) and the Moderately Severe Cohort (MMSE 10-14) as pre-specified secondary endpoints. Patient randomization was stratified by baseline SIB to insure balance in baseline cognitive ability between treatment arms. RESULTS: With no safety events noted by the data safety and monitoring board, the Moderately Severe (MMSE 10-14) Bryostatin-treated patients were significantly improved above the placebo patients for Weeks #13 through Week #42. After two cycles of 7 x i.v. Bryostatin doses over a 26-week period, the 10-14 Cohort Severe Impairment Battery (SIB), measured every 2 weeks, showed significant benefit using a Mixed Model Repeated Measures model (MMRM, 2-tailed, p < 0.05) for Weeks #13 through #42, even 16 weeks after dosing completion by Week #26. Placebo 10-14 patients showed no benefit, declining to negative 12.8 points by Week #42. Trend analyses confirmed the MMRM data for this Cohort, with a significant downward slope (equivalent to Cognitive Decline) for the placebo group, p < 0.001, 2-tailed, but no significant decline for the Bryostatin-treated group (p = 0.409, NS), treatment versus placebo p < 0.007. The Moderate Cohort patients showed no significant benefit. CONCLUSIONS: The Bryostatin-treated MMSE 10-14 patients showed no significant cognitive decline throughout the 10-month trial, versus placebo patients' decline of -12.8 SIB points.


Subject(s)
Alzheimer Disease , Cognition Disorders , Humans , Alzheimer Disease/drug therapy , Alzheimer Disease/complications , Bryostatins/adverse effects , Cognition Disorders/drug therapy , Double-Blind Method , Treatment Outcome
3.
Sci Rep ; 10(1): 18058, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093534

ABSTRACT

Fragile X syndrome (FXS), an X-chromosome linked intellectual disability, is the leading monogenetic cause of autism spectrum disorder (ASD), a neurodevelopmental condition that currently has no specific drug treatment. Building upon the demonstrated therapeutic effects on spatial memory of bryostatin-1, a relatively specific activator of protein kinase C (PKC)ε, (also of PKCα) on impaired synaptic plasticity/maturation and spatial learning and memory in FXS mice, we investigated whether bryostatin-1 might affect the autistic phenotypes and other behaviors, including open field activity, activities of daily living (nesting and marble burying), at the effective therapeutic dose for spatial memory deficits. Further evaluation included other non-spatial learning and memory tasks. Interestingly, a short period of treatment (5 weeks) only produced very limited or no therapeutic effects on the autistic and cognitive phenotypes in the Fmr1 KO2 mice, while a longer treatment (13 weeks) with the same dose of bryostatin-1 effectively rescued the autistic and non-spatial learning deficit cognitive phenotypes. It is possible that longer-term treatment would result in further improvement in these fragile X phenotypes. This effect is clearly different from other treatment strategies tested to date, in that the drug shows little acute effect, but strong long-term effects. It also shows no evidence of tolerance, which has been a problem with other drug classes (mGluR5 antagonists, GABA-A and -B agonists). The results strongly suggest that, at appropriate dosing and therapeutic period, chronic bryostatin-1 may have great therapeutic value for both ASD and FXS.


Subject(s)
Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/therapy , Bryostatins/administration & dosage , Bryostatins/physiology , Cognition Disorders/genetics , Cognition Disorders/therapy , Fragile X Syndrome/genetics , Fragile X Syndrome/therapy , Animals , Behavior, Animal , Bryostatins/pharmacology , Learning , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Protein Kinase C/metabolism , Spatial Memory
4.
Trends Pharmacol Sci ; 40(9): 655-668, 2019 09.
Article in English | MEDLINE | ID: mdl-31402121

ABSTRACT

Alzheimer's disease (AD), the leading disorder of memory impairment in our aging population, is increasing at an alarming rate. AD is currently identified by three 'gold standard criteria': (i) dementia in life, (ii) amyloid plaques at autopsy, and (iii) neurofibrillary tangles at autopsy. Several autopsy studies have indicated that dementia in life is a consequence of lost synaptic networks in the brain, while many clinical trials targeting neurotoxic amyloid beta (Aß) have consistently failed to produce therapeutic effects on memory function in AD patients. Restoring cognitive function(s) by activating endogenous repairing/regenerating mechanisms that are synaptogenic and antiapoptotic (preventing neuronal death), however, is emerging as a necessary disease-modifying therapeutic strategy against AD and possibly for other degenerative dementias, such as Parkinson's disease and multi-infarct dementia.


Subject(s)
Alzheimer Disease/pathology , Alzheimer Disease/therapy , Nerve Regeneration , Animals , Cognition Disorders/pathology , Humans , Neural Stem Cells/pathology , Neurons/pathology , Synapses/pathology , Transcranial Direct Current Stimulation/methods
5.
Behav Pharmacol ; 29(7): 592-604, 2018 10.
Article in English | MEDLINE | ID: mdl-30179884

ABSTRACT

Executive functioning is a high-level cognitive ability, regulating other abilities and behaviors to achieve desired goals. A typical executive task can be defined as the capacity to maintain one's attention on the current task, that is, responding only to the correct but not to distractive stimuli. Impairments of executive functions, or executive dysfunctions, have a growing impact on everyday life and academic achievement and are usually an early feature, and one of the core features, in brain injury and memory and behavioral disorders. Furthermore, emerging evidence indicates that memory therapeutics cannot achieve their clinical benefits in cognition if executive dysfunction is not effectively and simultaneously treated. Improvement of executive functions might be achieved through targeting some signaling pathways in the brain, including the brain-derived neurotrophic factor signaling pathways. These agents may be useful either as stand-alone interventions for patients with executive dysfunction and/or psychiatric and memory disorders or as essential adjuncts to drugs that target the underlying pathology in various brain injury and memory and behavioral disorders.


Subject(s)
Cognition Disorders/drug therapy , Cognition Disorders/metabolism , Executive Function/drug effects , Nerve Growth Factors/metabolism , Pharmacology/methods , Animals , Brain Injuries/complications , Cognition Disorders/etiology , Humans
6.
Neural Regen Res ; 13(3): 406-407, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29623916
7.
Curr Neuropharmacol ; 16(7): 1036-1044, 2018.
Article in English | MEDLINE | ID: mdl-29046153

ABSTRACT

BACKGROUND: As the human lifespan increases, the number of people affected by agerelated dementia is growing at an epidemic pace. Vascular pathology dramatically affects cognitive profiles, resulting in dementia and cognitive impairment. While vascular dementia itself constitutes a medical challenge, hypo-perfusion/vascular risk factors enhance amyloid toxicity and other memory- damaging factors and hasten Alzheimer's disease (AD) and other memory disorders' progression, as well as negatively affect treatment outcome. METHODS: Research and online content related to vascular cognitive impairment and dementia is reviewed, specifically focusing on the potential treatment of the disorder. RESULTS: Few therapeutic options are currently available to improve the prognosis of patients with vascular dementia and cognitive impairment, mixed AD dementia with vascular pathology, or other memory disorders. Emerging evidence, however, indicates that, like AD and other memory disorders, synaptic impairment underlies much of the memory impairment in the cognitive decline of vascular cognitive impairment and vascular dementia. CONCLUSION: Effective rescues of the memory functions might be achieved through synaptic and memory therapeutics, targeting distinct molecular signaling pathways that support the formation of new synapses and maintaining their connections. Potential therapeutic agents include: 1) memory therapeutic agents that rescue synaptic and memory functions after the brain insults; 2) antipathologic therapeutics and an effective management of vascular risk factors; and 3) preventative therapeutic agents that achieve memory therapy through functional enhancement. These therapeutic agents are also likely to benefit patients with AD and/or other types of memory disorders.


Subject(s)
Cognitive Dysfunction/therapy , Dementia, Vascular/therapy , Animals , Humans , Nootropic Agents/pharmacology , Nootropic Agents/therapeutic use
8.
Eur J Pharmacol ; 807: 71-74, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28472666

ABSTRACT

Despite over a half-century's intensive research worldwide, the currently available antidepressants remain sub-optimal. Therapeutic options for treatment-resistant depression, for instance, are rather limited. Here, we found that rats exhibited a lasting treatment-resistant depressive immobility in response to open space swim test at a high intensity of induction. The induced depressive behavior is associated with a dramatic impairment in spatial learning and memory. Both the depressive immobility and impairment in spatial learning and memory are sensitive to a period of chronic treatment with bryopstatin-1, a relatively selective activator of protein kinase Cε. Bryostatin-1-like analogues therefore might have therapeutic values for the treatment of treatment-resistant depression.


Subject(s)
Bryostatins/pharmacology , Depressive Disorder, Treatment-Resistant/drug therapy , Animals , Behavior, Animal/drug effects , Bryostatins/therapeutic use , Depressive Disorder, Treatment-Resistant/physiopathology , Male , Rats , Rats, Wistar , Spatial Learning/drug effects , Time Factors
9.
J Alzheimers Dis ; 58(2): 521-535, 2017.
Article in English | MEDLINE | ID: mdl-28482641

ABSTRACT

Bryostatin 1, a potent activator of protein kinase C epsilon (PKCɛ), has been shown to reverse synaptic loss and facilitate synaptic maturation in animal models of Alzheimer's disease (AD), Fragile X, stroke, and other neurological disorders. In a single-dose (25 µg/m2) randomized double-blind Phase IIa clinical trial, bryostatin levels reached a maximum at 1-2 h after the start of infusion. In close parallel with peak blood levels of bryostatin, an increase of PBMC PKCɛ was measured (p = 0.0185) within 1 h from the onset of infusion. Of 9 patients with a clinical diagnosis of AD, of which 6 received drug and 3 received vehicle within a double-blind protocol, bryostatin increased the Mini-Mental State Examination (MMSE) score by +1.83±0.70 unit at 3 h versus -1.00±1.53 unit for placebo. Bryostatin was well tolerated in these AD patients and no drug-related adverse events were reported. The 25 µg/m2 administered dose was based on prior clinical experience with three Expanded Access advanced AD patients treated with bryostatin, in which return of major functions such as swallowing, vocalization, and word recognition were noted. In one Expanded Access patient trial, elevated PKCɛ levels closely tracked cognitive benefits in the first 24 weeks as measured by MMSE and ADCS-ADL psychometrics. Pre-clinical mouse studies showed effective activation of PKCɛ and increased levels of BDNF and PSD-95. Together, these Phase IIa, Expanded Access, and pre-clinical results provide initial encouragement for bryostatin 1 as a potential treatment for AD.


Subject(s)
Alzheimer Disease , Antipsychotic Agents/therapeutic use , Bryostatins/therapeutic use , Cognition Disorders , Protein Kinase C-epsilon/metabolism , Adult , Aged , Aged, 80 and over , Alzheimer Disease/complications , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Cognition Disorders/drug therapy , Cognition Disorders/enzymology , Cognition Disorders/etiology , Disks Large Homolog 4 Protein/metabolism , Double-Blind Method , Female , Humans , Male , Mental Status Schedule , Mice , Mice, Inbred C57BL , Middle Aged , Neuropsychological Tests , Phosphopyruvate Hydratase/metabolism , Psychometrics , Synaptophysin/metabolism , Time Factors
10.
J Pharmacol Exp Ther ; 357(2): 300-10, 2016 May.
Article in English | MEDLINE | ID: mdl-26941170

ABSTRACT

Fragile X syndrome (FXS) is characterized by synaptic immaturity, cognitive impairment, and behavioral changes. The disorder is caused by transcriptional shutdown in neurons of thefragile X mental retardation 1gene product, fragile X mental retardation protein. Fragile X mental retardation protein is a repressor of dendritic mRNA translation and its silencing leads to dysregulation of synaptically driven protein synthesis and impairments of intellect, cognition, and behavior, and FXS is a disorder that currently has no effective therapeutics. Here, young fragile X mice were treated with chronic bryostatin-1, a relatively selective protein kinase Cεactivator, which induces synaptogenesis and synaptic maturation/repair. Chronic treatment with bryostatin-1 rescues young fragile X mice from the disorder phenotypes, including normalization of most FXS abnormalities in 1) hippocampal brain-derived neurotrophic factor expression, 2) postsynaptic density-95 levels, 3) transformation of immature dendritic spines to mature synapses, 4) densities of the presynaptic and postsynaptic membranes, and 5) spatial learning and memory. The therapeutic effects were achieved without downregulation of metabotropic glutamate receptor (mGluR) 5 in the hippocampus and are more dramatic than those of a late-onset treatment in adult fragile X mice. mGluR5 expression was in fact lower in fragile X mice and its expression was restored with the bryostatin-1 treatment. Our results show that synaptic and cognitive function of young FXS mice can be normalized through pharmacological treatment without downregulation of mGluR5 and that bryostatin-1-like agents may represent a novel class of drugs to treat fragile X mental retardation at a young age and in adults.


Subject(s)
Fragile X Syndrome/drug therapy , Memory Disorders/drug therapy , Spatial Memory/drug effects , Synapses/drug effects , Animals , Brain-Derived Neurotrophic Factor/metabolism , Bryostatins/pharmacology , Dendritic Spines/drug effects , Disks Large Homolog 4 Protein , Enzyme Activators/pharmacology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/pathology , Fragile X Syndrome/psychology , Guanylate Kinases/genetics , Guanylate Kinases/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Maze Learning/drug effects , Membrane Proteins/genetics , Membrane Proteins/metabolism , Memory Disorders/etiology , Memory Disorders/psychology , Mice , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Protein Kinase C-epsilon/drug effects , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Presynaptic/drug effects , Synapses/pathology
12.
Trends Pharmacol Sci ; 36(6): 384-94, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25959522

ABSTRACT

Evidence is accumulating that many memory disorders, including those due to neurodegenerative diseases, traumatic brain injury (TBI), vascular disease, or abnormal brain development, share common features of memory-related pathology. Structural and functional deficits of synapses are at the core of the underlying pathophysiology, constituting a critical point of convergence in memory disorders. Memory therapeutics that target synaptic loss and dysfunction - that is, to slow, halt, or reverse progression of the disorders at the level of synapses, via synaptogenic molecular cascades such as those of protein kinase C (PKC) and brain-derived neurotrophic factor (BDNF) - possess universal therapeutic value for many forms of memory disorder. They may be useful either as standalone interventions for patients with memory disorders or as adjuncts to drugs that target the underlying pathology.


Subject(s)
Memory Disorders/drug therapy , Mesenchymal Stem Cell Transplantation , Neuroprotective Agents/pharmacology , Synapses/metabolism , Synaptic Transmission , Animals , Humans , Memory Disorders/metabolism , Memory Disorders/therapy , Neuroprotective Agents/therapeutic use , Synapses/drug effects
13.
Behav Pharmacol ; 25(5-6): 410-24, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25028862

ABSTRACT

Stress in life is unavoidable, affecting everyone on a daily basis. Psychological stress in mammals triggers a rapidly organized response for survival, but it may also cause a variety of behavioral disorders and damage cognitive function. Stress is associated with biases in cognitive processing; some of the most enduring memories are formed by traumatic events. Our understanding of how cognition is shaped by stress is still relatively primitive; however, evidence is rapidly accumulating that the 'mature' brain has a great capacity for plasticity and that there are numerous ways through which pharmacological therapeutics could rescue cognitive function and regain cognitive balance. In this review, we discuss recent advances in our understanding of the interplay between stress and cognitive processes and potential therapeutic approaches to stress-related behavioral and cognitive disorders.


Subject(s)
Brain/drug effects , Brain/physiopathology , Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Stress, Psychological/drug therapy , Stress, Psychological/physiopathology , Animals , Cognition/drug effects , Cognition/physiology , Humans
14.
J Pharmacol Exp Ther ; 349(3): 393-401, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24659806

ABSTRACT

Fragile X syndrome (FXS) is caused by transcriptional silencing in neurons of the FMR1 gene product, fragile X mental retardation protein (FMRP), a repressor of dendritic mRNA translation. The lack of FMRP leads to dysregulation of synaptically driven protein synthesis and impairments of intellect, cognition, and behavior, a disorder that currently has no effective therapeutics. Fragile X mice were treated with chronic bryostatin-1, a relatively selective protein kinase ε activator with pharmacological profiles of rapid mGluR desensitization, synaptogenesis, and synaptic maturation/repairing. Differences in the major FXS phenotypes, synapses, and cognitive functions were evaluated and compared among the age-matched groups. Long-term treatment with bryostatin-1 rescues adult fragile X mice from the disorder phenotypes, including normalization of most FXS abnormalities in hippocampal brain-derived neurotrophic factor expression and secretion, postsynaptic density-95 levels, glycogen synthase kinase-3ß phosphorylation, transformation of immature dendritic spines to mature synapses, densities of the presynaptic and postsynaptic membranes, and spatial learning and memory. Our results show that synaptic and cognitive function of adult FXS mice can be normalized through pharmacologic treatment and that bryostatin-1-like agents may represent a novel class of drugs to treat fragile X mental retardation even after postpartum brain development has largely completed.


Subject(s)
Bryostatins/therapeutic use , Fragile X Syndrome/drug therapy , Hippocampus/drug effects , Maze Learning/drug effects , Memory/drug effects , Spatial Behavior/drug effects , Synapses/drug effects , Animals , Bryostatins/administration & dosage , Dendritic Spines/drug effects , Dendritic Spines/pathology , Disease Models, Animal , Fragile X Syndrome/metabolism , Fragile X Syndrome/pathology , Fragile X Syndrome/physiopathology , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Male , Mice , Mice, Inbred Strains , Microscopy, Confocal , Microscopy, Electron , Protein Kinase C-epsilon/metabolism , Synapses/metabolism , Synapses/physiology , Synapses/ultrastructure
15.
Prog Mol Biol Transl Sci ; 122: 31-59, 2014.
Article in English | MEDLINE | ID: mdl-24484697

ABSTRACT

The protein kinase C (PKC) isoforms, which play an essential role in transmembrane signal conduction, can be viewed as a family of "memory kinases." Evidence is emerging that they are critically involved in memory acquisition and maintenance, in addition to their involvement in other functions of cells. Deficits in PKC signal cascades in neurons are one of the earliest abnormalities in the brains of patients suffering from Alzheimer's disease. Their dysfunction is also involved in several other types of memory impairments, including those related to emotion, mental retardation, brain injury, and vascular dementia/ischemic stroke. Inhibition of PKC activity leads to a reduced capacity of many types of learning and memory, but may have therapeutic values in treating substance abuse or aversive memories. PKC activators, on the other hand, have been shown to possess memory-enhancing and antidementia actions. PKC pharmacology may, therefore, represent an attractive area for developing effective cognitive drugs for the treatment of many types of memory disorders and dementias.


Subject(s)
Memory/physiology , Protein Kinase C/metabolism , Signal Transduction/physiology , Animals , Humans , Isoenzymes
16.
Article in English | MEDLINE | ID: mdl-24291721

ABSTRACT

Bryostatin 1, a potential anti-Alzheimer drug, is effective at subnanomolar concentrations. Measurement is complicated by the formation of low m/z degradation products and the formation of adducts with various cations, which make accurate quantitation difficult. Adduct formation caused the sample matrix or mobile phase to partition bryostatin 1 into products of different mass. Degradation of the 927 [M+Na](+) ion to a 869m/z product was strongly influenced by ionization conditions. We validated a bryostatin 1 assay in biological tissues using capillary column HPLC with nanospray ionization (NSI) in a triple-quadrupole mass spectrometer in selected reaction monitoring (SRM) mode. Adduct formation was controlled by adding 1mM acetic acid and 0.1mM sodium acetate to the HPLC buffer, maximizing the formation of the [M+Na](+) ion. Efficient removal of contaminating cholesterol from the sample during solvent extraction was also critical. The increased sensitivity provided by NSI and capillary-bore columns and the elimination of signal partitioning due to adduct formation and degradation in the ionization source enabled a detection limit of 1×10(-18)mol of bryostatin 1 and a LLOQ of 3×10(-18)mol from 1µl of sample. Bryostatin 1 at low pmol/l concentrations enabled measurement in brain and other tissues without the use of radioactive labels. Despite bryostatin 1's high molecular weight, considerable brain access was observed, with peak brain concentrations exceeding 8% of the peak blood plasma concentrations. Bryostatin 1 readily crosses the blood-brain barrier, reaching peak concentrations of 0.2nM, and specifically activates and translocates brain PKCɛ.


Subject(s)
Bryostatins/analysis , Bryostatins/blood , Chromatography, Liquid/methods , Mass Spectrometry/methods , Animals , Brain Chemistry , Bryostatins/chemistry , Bryostatins/pharmacology , Cattle , Drug Stability , Humans , Limit of Detection , Male , Mice , Mice, Inbred C57BL , Protein Kinase C-epsilon/analysis , Protein Kinase C-epsilon/chemistry , Protein Kinase C-epsilon/metabolism , Reproducibility of Results
17.
Behav Pharmacol ; 24(3): 222-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23591125

ABSTRACT

The 'vascular depression' hypothesis has recently attracted significant research attention, although the causal relationship between vascular-related injuries and depression has not been established. Here, we show that one episode of cerebral ischemia was sufficient to greatly increase the sensitivity of rats to potentially depressogenic events, evaluated at below-threshold intensities in the open space swim test. The induced 'ischemic depression' was lasting and sensitive to an acute administration of brain-derived neurotrophic factor or bryostatin-1, a relatively selective activator of protein kinase Cε, during the induction phase. Chronic treatment with bryostatin-1 (5 weeks) after the induction of depressive behavior reversed the depressive immobility and produced a lasting therapeutic effect, which remained effective 3 weeks after discontinuation of the treatment. Similar treatment with alaproclate, a selective serotonin reuptake inhibitor, in contrast, produced temporary relief from the depressive symptoms, with the therapeutic effect disappearing soon after the end of the treatment. The results strongly suggest that cerebral ischemia has a direct role in shaping the sensitivity of an individual to depressogenic events and that bryostatin-1-like agents may be developed as therapeutics for treating ischemic depression in humans.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antidepressive Agents/therapeutic use , Bryostatins/therapeutic use , Depression/drug therapy , Depression/etiology , Hypoxia-Ischemia, Brain/complications , Analysis of Variance , Animals , Brain-Derived Neurotrophic Factor/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Food Preferences/drug effects , In Situ Nick-End Labeling , Injections, Intraventricular , Male , Rats , Rats, Wistar , Sucrose/administration & dosage , Sweetening Agents/administration & dosage , Time Factors
18.
Adv Pharmacol ; 64: 273-302, 2012.
Article in English | MEDLINE | ID: mdl-22840750

ABSTRACT

Memories are much more easily impaired than improved. Dementias, a lasting impairment of memory function, occur in a variety of cognitive disorders and become more clinically dominant as the population ages. Protein kinase C is one of the "cognitive kinases," and plays an essential role in both memory acquisition and maintenance. Deficits in protein kinase C (PKC) signal cascades in neurons represent one of the earliest changes in the brains of patients with Alzheimer's disease (AD) and other types of memory impairment, including those related to cerebral ischemia and ischemic stroke. Inhibition or impairment of PKC activity results in compromised learning and memory, whereas an appropriate activation of certain PKC isozymes leads to an enhancement of learning and memory and/or antidementic effects. In preclinical studies, PKC activators have been shown to increase the expression and activity of PKC isozymes, thereby restoring PKC signaling and downstream activity, including stimulation of neurotrophic activity, synaptic/structural remodeling, and synaptogenesis in the hippocampus and related cortical areas. PKC activators also reduce the accumulation of neurotoxic amyloid and tau protein hyperphosphorylation and support anti-apoptotic processes in the brain. These observations strongly suggest that PKC pharmacology may represent an attractive area for the development of effective cognition-enhancing therapeutics for the treatment of dementias.


Subject(s)
Dementia/drug therapy , Dementia/enzymology , Protein Kinase C/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Animals , Dementia/physiopathology , Enzyme Activation , Humans , Isoenzymes/metabolism , Memory
19.
J Neurosci ; 31(2): 630-43, 2011 Jan 12.
Article in English | MEDLINE | ID: mdl-21228172

ABSTRACT

Among the pathologic hallmarks of Alzheimer's disease (AD) neurodegeneration, only synaptic loss in the brains of AD patients closely correlates with the degree of dementia in vivo. Here, we describe a molecular basis for this AD loss of synapses: pathological reduction of synaptogenic PKC isozymes and their downstream synaptogenic substrates, such as brain-derived neurotrophic factor. This reduction, particularly of PKC α and ε, occurs in association with elevation of soluble ß amyloid protein (Aß), but before the appearance of the amyloid plaques or neuronal loss in the Tg2576 AD transgenic mouse strain. Conversely, treatment of the Tg2576 mouse brain with the PKC activator, bryostatin-1, restores normal or supranormal levels of PKC α and ε, reduces the level of soluble Aß, prevents and/or reverses the loss of hippocampal synapses, and prevents the memory impairment observed at 5 months postpartum. Similarly, the PKC ε-specific activator, DCP-LA, effectively prevents synaptic loss, amyloid plaques, and cognitive deficits (also prevented by bryostatin-1) in the much more rapidly progressing 5XFAD transgenic strain. These results suggest that synaptic loss and the resulting cognitive deficits depend on the balance between the lowering effects of Aß on PKC α and ε versus the lowering effects of PKC on Aß in AD transgenic mice.


Subject(s)
Alzheimer Disease , Amyloid beta-Peptides/metabolism , Cognition Disorders/psychology , Protein Kinase C-epsilon/metabolism , Synapses/pathology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/psychology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Bryostatins/pharmacology , Caprylates/pharmacology , Enzyme Activators/pharmacology , Hippocampus/pathology , Isoenzymes/metabolism , Male , Maze Learning , Memory , Mice , Mice, Transgenic , Plaque, Amyloid/pathology , Protein Kinase C-alpha/metabolism
20.
Pharmacol Ther ; 127(1): 66-77, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20382181

ABSTRACT

Evidence is accumulating indicating that some protein kinase C (PKC) isozymes play an essential role in various phases as well as types of learning and memory. Abnormal functions of PKC signal cascades in the brains have been found to represent one of the earliest changes in patients with Alzheimer's disease (AD) and other types of memory deficits, including those related to cerebral ischemic/stroke events. In preclinical studies, an inhibition or impairment of PKC activity leads to compromised learning and memory, whereas an appropriate activation of some PKC isozymes results in an enhancement of learning and memory and/or antidementic effects against memory disorders. PKC activators not only increase activity of PKC isozymes and thereby restore PKC signaling activity, including neurotrophic activity, synaptic/structural remodeling, and synaptogenesis in the hippocampus and related cortical areas, but also reduce the accumulation of neurotoxic amyloid and tau protein hyperphosphorylation in the brain. These observations strongly suggest that PKC isoform pharmacology may represent an attractive area for the development of cognition-enhancing agents and therapeutics against memory loss in the future.


Subject(s)
Cognition Disorders/drug therapy , Dementia/drug therapy , Enzyme Activators/pharmacology , Memory/drug effects , Nootropic Agents/pharmacology , Protein Kinase C/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Alzheimer Disease/etiology , Animals , Cattle , Cognition Disorders/enzymology , Dementia/enzymology , Enzyme Activation , Enzyme Activators/therapeutic use , Female , Humans , Male , Nootropic Agents/therapeutic use , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...