Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
BMC Anesthesiol ; 19(1): 241, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31881996

ABSTRACT

BACKGROUND: The impact of general anesthesia on cognitive impairment is controversial and complex. A large body of evidence supports the association between exposure to surgery under general anesthesia and development of delayed neurocognitive recovery in a subset of patients. Existing literature continues to debate whether these short-term effects on cognition can be attributed to anesthetic agents themselves, or whether other variables are causative of the observed changes in cognition. Furthermore, there is conflicting data on the relationship between anesthesia exposure and the development of long-term neurocognitive disorders, or development of incident dementia in the patient population with normal preoperative cognitive function. Patients with pre-existing cognitive impairment present a unique set of anesthetic considerations, including potential medication interactions, challenges with cooperation during assessment and non-general anesthesia techniques, and the possibility that pre-existing cognitive impairment may impart a susceptibility to further cognitive dysfunction. MAIN BODY: This review highlights landmark and recent studies in the field, and explores potential mechanisms involved in perioperative cognitive disorders (also known as postoperative cognitive dysfunction, POCD). Specifically, we will review clinical and preclinical evidence which implicates alterations to tau protein, inflammation, calcium dysregulation, and mitochondrial dysfunction. As our population ages and the prevalence of Alzheimer's disease and other forms of dementia continues to increase, we require a greater understanding of potential modifiable factors that impact perioperative cognitive impairment. CONCLUSIONS: Future research should aim to further characterize the associated risk factors and determine whether certain anesthetic approaches or other interventions may lower the potential risk which may be conferred by anesthesia and/or surgery in susceptible individuals.


Subject(s)
Anesthesia, General/adverse effects , Cognitive Dysfunction/etiology , Postoperative Cognitive Complications/epidemiology , Anesthesia, General/methods , Anesthesiology , Cognitive Dysfunction/epidemiology , Humans , Postoperative Cognitive Complications/etiology , Risk Factors
2.
Sci Rep ; 7(1): 812, 2017 04 11.
Article in English | MEDLINE | ID: mdl-28400560

ABSTRACT

Fundamental cell signaling mechanisms that regulate dynamic remodeling of the extracellular matrix (ECM) in mechanically loaded tissues are not yet clearly understood. Trabecular meshwork (TM) tissue in the eye is under constant mechanical stress and continuous remodeling of ECM is crucial to maintain normal aqueous humor drainage and intraocular pressure (IOP). However, excessive ECM remodeling can cause fibrosis of the TM as in primary open-angle glaucoma (POAG) patients, and is characterized by increased resistance to aqueous humor drainage, elevated IOP, optic nerve degeneration and blindness. Increased levels of active transforming growth factor-ß2 (TGF-ß2) in the aqueous humor is the main cause of fibrosis of TM in POAG patients. Herein, we report a novel finding that, in TM cells, TGF-ß-induced increase in collagen expression is associated with phosphorylation of phosphatase and tensin homolog (PTEN) at residues Ser380/Thr382/383. Exogenous overexpression of a mutated form of PTEN with enhanced phosphatase activity prevented the TGF-ß-induced collagen expression by TM cells. We propose that rapid alteration of PTEN activity through changes in its phosphorylation status could uniquely regulate the continuous remodeling of ECM in the normal TM. Modulating PTEN activity may have high therapeutic potential to alleviating the fibrosis of TM in POAG patients.


Subject(s)
Glaucoma, Open-Angle/metabolism , PTEN Phosphohydrolase/metabolism , Protein Processing, Post-Translational , Trabecular Meshwork/pathology , Transforming Growth Factor beta/pharmacology , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Fibrosis , Glaucoma, Open-Angle/pathology , Humans , Phosphorylation , Trabecular Meshwork/metabolism , Transforming Growth Factor beta/metabolism
3.
J Neurosci ; 35(45): 15157-69, 2015 Nov 11.
Article in English | MEDLINE | ID: mdl-26558786

ABSTRACT

In Alzheimer's disease, accumulation of soluble oligomers of ß-amyloid peptide is known to be highly toxic, causing disturbances in synaptic activity and neuronal death. Multiple studies relate these effects to increased oxidative stress and aberrant activity of calcium-permeable cation channels leading to calcium imbalance. The transient receptor potential melastatin 2 (TRPM2) channel, a Ca(2+)-permeable nonselective cation channel activated by oxidative stress, has been implicated in neurodegenerative diseases, and more recently in amyloid-induced toxicity. Here we show that the function of TRPM2 is augmented by treatment of cultured neurons with ß-amyloid oligomers. Aged APP/PS1 Alzheimer's mouse model showed increased levels of endoplasmic reticulum stress markers, protein disulfide isomerase and phosphorylated eukaryotic initiation factor 2α, as well as decreased levels of the presynaptic marker synaptophysin. Elimination of TRPM2 in APP/PS1 mice corrected these abnormal responses without affecting plaque burden. These effects of TRPM2 seem to be selective for ß-amyloid toxicity, as ER stress responses to thapsigargin or tunicamycin in TRPM2(-/-) neurons was identical to that of wild-type neurons. Moreover, reduced microglial activation was observed in TRPM2(-/-)/APP/PS1 hippocampus compared with APP/PS1 mice. In addition, age-dependent spatial memory deficits in APP/PS1 mice were reversed in TRPM2(-/-)/APP/PS1 mice. These results reveal the importance of TRPM2 for ß-amyloid neuronal toxicity, suggesting that TRPM2 activity could be potentially targeted to improve outcomes in Alzheimer's disease. SIGNIFICANCE STATEMENT: Transient receptor potential melastatin 2 (TRPM2) is an oxidative stress sensing calcium-permeable channel that is thought to contribute to calcium dysregulation associated with neurodegenerative diseases, including Alzheimer's disease. Here we show that oligomeric ß-amyloid, the toxic peptide in Alzheimer's disease, facilitates TRPM2 channel activation. In mice designed to model Alzheimer's disease, genetic elimination of TRPM2 normalized deficits in synaptic markers in aged mice. Moreover, the absence of TRPM2 improved age-dependent spatial memory deficits observed in Alzheimer's mice. Our results reveal the importance of TRPM2 for neuronal toxicity and memory impairments in an Alzheimer's mouse model and suggest that TRPM2 could be targeted for the development of therapeutic agents effective in the treatment of dementia.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Memory Disorders/metabolism , TRPM Cation Channels/deficiency , Alzheimer Disease/chemically induced , Amyloid beta-Peptides/toxicity , Animals , Cells, Cultured , Humans , Male , Memory Disorders/chemically induced , Memory Disorders/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Plaque, Amyloid/metabolism , TRPM Cation Channels/genetics
4.
Orbit ; 34(4): 220-2, 2015.
Article in English | MEDLINE | ID: mdl-26121166

ABSTRACT

We report a case of a 44-year-old man who presented with a left medial canthal mass and epiphora. Imaging was suggestive of a mass continuous with the nasolacrimal sac. Subsequent surgical exploration revealed a mass adherent to bone with invasion of the lacrimal system. Histological examination revealed a squamous/transitional cell papilloma overlying a low-grade mucoepidermoid carcinoma (MEC). Complete surgical resection was completed and pathology confirmed the diagnosis. This is the first case in which a MEC has been reported concurrently with an overlying papilloma, providing support for the hypothesis that MECs arise from papillomas in the lacrimal sac. Additionally, the tissue stained positive for p63, which is congruent with MEC immunoreactivity in the salivary gland. The description of these unique histopathological findings may assist in definitive diagnosis and improve our understanding of the pathophysiology underlying lacrimal sac MEC tumors.


Subject(s)
Eye Neoplasms/surgery , Lacrimal Apparatus/surgery , Mucoepidermoid Tumor/surgery , Papilloma/surgery , Adult , Diagnosis, Differential , Eye Neoplasms/diagnosis , Humans , Lacrimal Apparatus/pathology , Male , Membrane Proteins/analysis , Mucoepidermoid Tumor/diagnosis , Papilloma/diagnosis , Tomography, X-Ray Computed
5.
Retin Cases Brief Rep ; 9(2): 173-6, 2015.
Article in English | MEDLINE | ID: mdl-25764315

ABSTRACT

PURPOSE: To describe the clinical findings in a patient demonstrating recovery from nonparaneoplastic autoimmune retinopathy after a minimal course of steroid treatment. METHODS: Clinical presentation was documented, and paraclinical tests were obtained using Humphrey automated perimetry for visual fields, Western blotting for antiretinal antibodies, and electroretinography for evaluation of rod and cone function. RESULTS: Initial presentation revealed marked visual field deficits, electroretinographic dysfunction, and the presence of α-enolase autoantibodies. After a brief course of oral corticosteroids, the patient demonstrated improvement in visual fields, disappearance of α-enolase autoantibodies, partial recovery of the cone on-response, and complete recovery of the rod response. CONCLUSION: This case is distinguished from previous reports by the rapidity and degree of recovery, the brevity of treatment required, and the unique electroretinographic recovery pattern with concomitant disappearance of α-enolase autoantibodies. These findings suggest a pathologic role for α-enolase autoantibodies in autoimmune rod bipolar cell dysfunction. Identification of other cases exhibiting such improvements and associated autoantibody activity may expand our understanding of nonparaneoplastic autoimmune retinopathy disease pathogenesis.


Subject(s)
Autoantibodies/immunology , Autoimmune Diseases/drug therapy , Prednisone/administration & dosage , Retinal Diseases/drug therapy , Administration, Oral , Autoimmune Diseases/diagnosis , Autoimmune Diseases/immunology , Biopsy , Dose-Response Relationship, Drug , Electroretinography , Glucocorticoids/administration & dosage , Humans , Male , Middle Aged , Paraneoplastic Syndromes , Remission Induction , Retinal Diseases/diagnosis , Retinal Diseases/immunology , Tomography, Optical Coherence , Visual Fields
6.
Neurobiol Aging ; 35(4): 926-33, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24112788

ABSTRACT

Alterations in the expression and signaling of brain-derived neurotrophic factor (BDNF) and the precursor to nerve growth factor (NGF), proNGF, play a role in the neuronal and cognitive dysfunction of Alzheimer's disease. Aggregated amyloid-ß has been shown to down-regulate specific BDNF transcripts in Alzheimer's disease, but the role of tau pathology in neurotrophin dysregulation has not been investigated. We measured levels of BDNF mRNA and protein using real-time quantitative reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay and proNGF protein using Western blotting in parietal cortex of subjects with tauopathies, neurodegenerative diseases exhibiting tau pathology without amyloid-ß accumulation. We observed a significant increase in the level of proNGF protein in Pick's disease and a significant decrease in BDNF mRNA and protein levels in Pick's disease and corticobasal degeneration, but no neurotrophin alterations in progressive supranuclear palsy. The decrease in total BDNF mRNA levels in these tauopathies was predominantly due to down-regulation of transcript IV. These findings implicate tau pathology in neurotrophin dysregulation, which may represent a mechanism through which tau confers toxicity in Alzheimer's disease and related non-Alzheimer's dementias.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Gene Expression , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Tauopathies/genetics , Tauopathies/metabolism , tau Proteins/metabolism , tau Proteins/toxicity , Aged , Aged, 80 and over , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Down-Regulation , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged , Pick Disease of the Brain/genetics , Pick Disease of the Brain/metabolism , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
J Neurosci ; 33(42): 16552-64, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24133259

ABSTRACT

In Alzheimer's disease (AD), soluble amyloid-ß oligomers (AßOs) trigger neurotoxic signaling, at least partially, via the cellular prion protein (PrP(C)). However, it is unknown whether other ligands of PrP(C) can regulate this potentially toxic interaction. Stress-inducible phosphoprotein 1 (STI1), an Hsp90 cochaperone secreted by astrocytes, binds to PrP(C) in the vicinity of the AßO binding site to protect neurons against toxic stimuli. Here, we investigated a potential role of STI1 in AßO toxicity. We confirmed the specific binding of AßOs and STI1 to the PrP and showed that STI1 efficiently inhibited AßO binding to PrP in vitro (IC50 of ∼70 nm) and also decreased AßO binding to cultured mouse primary hippocampal neurons. Treatment with STI1 prevented AßO-induced synaptic loss and neuronal death in mouse cultured neurons and long-term potentiation inhibition in mouse hippocampal slices. Interestingly, STI1-haploinsufficient neurons were more sensitive to AßO-induced cell death and could be rescued by treatment with recombinant STI1. Noteworthy, both AßO binding to PrP(C) and PrP(C)-dependent AßO toxicity were inhibited by TPR2A, the PrP(C)-interacting domain of STI1. Additionally, PrP(C)-STI1 engagement activated α7 nicotinic acetylcholine receptors, which participated in neuroprotection against AßO-induced toxicity. We found an age-dependent upregulation of cortical STI1 in the APPswe/PS1dE9 mouse model of AD and in the brains of AD-affected individuals, suggesting a compensatory response. Our findings reveal a previously unrecognized role of the PrP(C) ligand STI1 in protecting neurons in AD and suggest a novel pathway that may help to offset AßO-induced toxicity.


Subject(s)
Amyloid beta-Peptides/metabolism , Heat-Shock Proteins/metabolism , Neurons/metabolism , PrPC Proteins/metabolism , Alzheimer Disease/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Cells, Cultured , Hippocampus/metabolism , Mice , Protein Binding , Signal Transduction/physiology , alpha7 Nicotinic Acetylcholine Receptor/metabolism
8.
Clin Invest Med ; 36(1): E50-6, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23374600

ABSTRACT

In 2011, members of the Clinician Investigator Trainee Association of Canada - Association des cliniciens-chercheurs en formation du Canada (CITAC-ACCFC) and the Canadian Society for Clinician Investigators (CSCI) held a joint Annual General Meeting (AGM) and Young Investigator Forum (YIF) September 12-14 in Ottawa, ON, Canada. The theme of the meeting was "The Role of Government and Regulatory Organizations in Shaping the Environment of the Clinician Scientist". The meeting was well attended by established clinician scientists and clinician investigator trainees from across Canada. The aim of this scientific overview is to highlight the research presented by trainees at both the oral plenary session as well as the poster presentation sessions of this meeting. This work covers a wide variety of medical disciplines, focusing on translational medicine, from the basic sciences to clinical application.


Subject(s)
Biomedical Research , Research Personnel , Canada , Congresses as Topic , Humans
9.
Adv Exp Med Biol ; 961: 433-47, 2013.
Article in English | MEDLINE | ID: mdl-23224901

ABSTRACT

Stroke is a very strong risk factor for dementia. Furthermore, ischemic stroke and Alzheimer's disease (AD) share a number of overlapping mechanisms of neuron loss and dysfunction, including those induced by the inappropriate activation of N-methyl-D-aspartate receptors (NMDARs). These receptors form a major subtype of excitatory glutamate receptor. They are nonselective cation channels with appreciable Ca(2+) permeability, and their overactivation leads to neurotoxicity in the cortex and hippocampus. NMDARs have therefore been therapeutic targets in both conditions, but they have failed in the treatment of stroke, and there is limited rationale for using them in treating AD. In this chapter, we discuss current understanding of subtypes of NMDARs and their potential roles in -ischemic stroke and AD. We also discuss the properties of several other nonselective cation channels, transient receptor potential melastatin 2 and 7 channels, and their implications in linking these conditions.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Brain Ischemia/metabolism , Hippocampus/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , TRPM Cation Channels/metabolism , Aging/pathology , Alzheimer Disease/pathology , Animals , Brain Ischemia/pathology , Calcium/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Hippocampus/pathology , Humans , Protein Serine-Threonine Kinases , Stroke/metabolism , Stroke/pathology
10.
Mol Brain ; 5: 11, 2012 Apr 09.
Article in English | MEDLINE | ID: mdl-22487454

ABSTRACT

BACKGROUND: Glutathione (GSH) plays an important role in neuronal oxidant defence. Depletion of cellular GSH is observed in neurodegenerative diseases and thereby contributes to the associated oxidative stress and Ca2+ dysregulation. Whether depletion of cellular GSH, associated with neuronal senescence, directly influences Ca2+ permeation pathways is not known. Transient receptor potential melastatin type 2 (TRPM2) is a Ca2+ permeable non-selective cation channel expressed in several cell types including hippocampal pyramidal neurons. Moreover, activation of TRPM2 during oxidative stress has been linked to cell death. Importantly, GSH has been reported to inhibit TRPM2 channels, suggesting they may directly contribute to Ca2+ dysregulation associated with neuronal senescence. Herein, we explore the relation between cellular GSH and TRPM2 channel activity in long-term cultures of hippocampal neurons. RESULTS: In whole-cell voltage-clamp recordings, we observe that TRPM2 current density increases in cultured pyramidal neurons over time in vitro. The observed increase in current density was prevented by treatment with NAC, a precursor to GSH synthesis. Conversely, treatment of cultures maintained for 2 weeks in vitro with L-BSO, which depletes GSH by inhibiting its synthesis, augments TRPM2 currents. Additionally, we demonstrate that GSH inhibits TRPM2 currents through a thiol-independent mechanism, and produces a 3.5-fold shift in the dose-response curve generated by ADPR, the intracellular agonist for TRPM2. CONCLUSION: These results indicate that GSH plays a physiologically relevant role in the regulation of TRPM2 currents in hippocampal pyramidal neurons. This interaction may play an important role in aging and neurological diseases associated with depletion of GSH.


Subject(s)
Cellular Senescence , Glutathione/metabolism , Homeostasis , Ion Channel Gating , Neurons/cytology , Pyramidal Cells/metabolism , TRPM Cation Channels/metabolism , Acetylcysteine/pharmacology , Adenosine Diphosphate Ribose/pharmacology , Animals , Cells, Cultured , Cellular Senescence/drug effects , Gene Expression Regulation/drug effects , Glutathione/pharmacology , HEK293 Cells , Homeostasis/drug effects , Humans , Ion Channel Gating/drug effects , Mice , Neurons/drug effects , Neurons/metabolism , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Sulfhydryl Compounds/metabolism , TRPM Cation Channels/genetics , Time Factors
11.
Mol Brain ; 4: 44, 2011 Dec 21.
Article in English | MEDLINE | ID: mdl-22188973

ABSTRACT

Transient receptor potential melastatin 2 (TRPM2) is a calcium permeable non-selective cation channel that functions as a sensor of cellular redox status. Highly expressed within the CNS, we have previously demonstrated the functional expression of these channels in CA1 pyramidal neurons of the hippocampus. Although implicated in oxidative stress-induced neuronal cell death, and potentially in neurodegenerative disease, the physiological role of TRPM2 in the central nervous system is unknown. Interestingly, we have shown that the activation of these channels may be sensitized by co-incident NMDA receptor activation, suggesting a potential contribution of TRPM2 to synaptic transmission. Using hippocampal cultures and slices from TRPM2 null mice we demonstrate that the loss of these channels selectively impairs NMDAR-dependent long-term depression (LTD) while sparing long-term potentiation. Impaired LTD resulted from an inhibition of GSK-3ß, through increased phosphorylation, and a reduction in the expression of PSD95 and AMPARs. Notably, LTD could be rescued in TRPM2 null mice by recruitment of GSK-3ß signaling following dopamine D2 receptor stimulation. We propose that TRPM2 channels play a key role in hippocampal synaptic plasticity.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Hippocampus/cytology , N-Methylaspartate/metabolism , Neuronal Plasticity/physiology , Synapses/metabolism , TRPM Cation Channels/metabolism , Animals , Cells, Cultured , Glycogen Synthase Kinase 3 beta , Hippocampus/metabolism , Mice , Mice, Knockout , Neurons/cytology , Neurons/physiology , Patch-Clamp Techniques , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission/physiology , TRPM Cation Channels/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...