Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 2515: 17-28, 2022.
Article in English | MEDLINE | ID: mdl-35776343

ABSTRACT

Mitochondria are dynamic organelles that rely on a balance of opposing fission and fusion events to sustain mitochondrial function and efficiently meet the energy demands of a cell. As high-energy demanding cells, neurons rely heavily on optimally functional mitochondria with balanced mitochondrial dynamics, to ensure a sufficient energy supply required to maintain cell survival, establish membrane excitability and partake in processes of neurotransmission and plasticity. As such, many neurodegenerative diseases (e.g., Alzheimer's disease, Parkinson's disease) and stress conditions (e.g., stroke) leading to neuronal dysfunction or death are often associated with impaired mitochondrial function and dynamics, characterized by excessive mitochondrial fragmentation. For this reason, the assessment of mitochondrial morphology in neurons and within the brain can provide valuable information. The dynamic nature of mitochondria is not only observed in shape changes, but also changes in mitochondrial network connectivity and in cristae architecture. In this chapter, we will describe how mitochondrial morphology can be examined in vitro using hippocampal neuronal cultures and in vivo using mouse brain sections by immunocytochemistry, immunohistochemistry, and electron microscopy techniques.


Subject(s)
Mitochondria , Mitochondrial Membranes , Animals , Brain , Mice , Mitochondrial Dynamics , Neurons
2.
Cell Death Dis ; 13(4): 325, 2022 04 09.
Article in English | MEDLINE | ID: mdl-35397630

ABSTRACT

We previously showed that simvastatin (SV) restored memory in a mouse model of Alzheimer disease (AD) concomitantly with normalization in protein levels of memory-related immediate early genes in hippocampal CA1 neurons. Here, we investigated age-related changes in the hippocampal memory pathway, and whether the beneficial effects of SV could be related to enhanced neurogenesis and signaling in the Wnt/ß-catenin pathway. APP mice and wild-type (WT) littermate controls showed comparable number of proliferating (Ki67-positive nuclei) and immature (doublecortin (DCX)-positive) granule cells in the dentate gyrus until 3 months of age. At 4 months, Ki67 or DCX positive cells decreased sharply and remained less numerous until the endpoint (6 months) in both SV-treated and untreated APP mice. In 6 month-old APP mice, dendritic extensions of DCX immature neurons in the molecular layer were shorter, a deficit fully normalized by SV. Similarly, whereas mature granule cells (calbindin-immunopositive) were decreased in APP mice and not restored by SV, their dendritic arborizations were normalized to control levels by SV treatment. SV increased Prox1 protein levels (↑67.7%, p < 0.01), a Wnt/ß-catenin signaling target, while significantly decreasing (↓61.2%, p < 0.05) the upregulated levels of the ß-catenin-dependent Wnt pathway inhibitor DKK1 seen in APP mice. In APP mice, SV benefits were recapitulated by treatment with the Wnt/ß-catenin specific agonist WAY-262611, whereas they were fully abolished in mice that received the Wnt/ß-catenin pathway inhibitor XAV939 during the last month of SV treatment. Our results indicate that activation of the Wnt-ß-catenin pathway through downregulation of DKK1 underlies SV neuronal and cognitive benefits.


Subject(s)
Alzheimer Disease , Wnt Signaling Pathway , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Animals , Disease Models, Animal , Hippocampus/metabolism , Ki-67 Antigen/metabolism , Mice , Neurogenesis/physiology , Neurons/metabolism , Simvastatin/pharmacology , Simvastatin/therapeutic use , beta Catenin/metabolism
3.
Front Physiol ; 12: 611984, 2021.
Article in English | MEDLINE | ID: mdl-33584345

ABSTRACT

Preeclampsia is a common hypertensive disorder in pregnant women and whose causes and consequences have focused primarily on cardiovascular outcomes on the mother and offspring, often without taking into consideration the possible effects on the brain. One possible cause of preeclampsia has been attributed to alterations in the renin-angiotensin system, which has also been linked to cognitive decline. In this pilot study, we use a transgenic mouse model that chronically overexpresses human angiotensinogen and renin (R+A+ mice) that displayed characteristics of preeclampsia such as proteinuria during gestation. Offspring of these mothers as well as from control mothers were also examined. We were primarily interested in detecting whether cognitive deficits were present in the mothers and offspring in the long term and used a spatial learning and memory task as well as an object recognition task at three timepoints: 3, 8, and 12 months post-partum or post-natal, while measuring blood pressure and performing urine analysis after each timepoint. While we did not find significant deficits in preeclamptic mothers at the later timepoints, we did observe negative consequences in the pups of R+A+ mice that coincided with hemodynamic alterations whereby pups had higher whisker-evoked oxygenated hemoglobin levels and increased cerebral blood flow responses compared to control pups. Our study provides validation of this preeclampsia mouse model for future studies to decipher the underlying mechanisms of long-term cognitive deficits found in offspring.

4.
Geroscience ; 42(5): 1237-1256, 2020 10.
Article in English | MEDLINE | ID: mdl-32700176

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder that is multifactorial in nature. Yet, despite being the most common form of dementia in the elderly, AD's primary cause remains unknown. As such, there is currently little to offer AD patients as the vast majority of recently tested therapies have either failed in well-controlled clinical trials or inadequately treat AD. Recently, emerging preclinical and clinical evidence has associated the brain renin angiotensin system (RAS) to AD pathology. Accordingly, various components of the brain RAS were shown to be altered in AD patients and mouse models, including the angiotensin II type 1 (AT1R), angiotensin IV receptor (AT4R), and Mas receptors. Collectively, the changes observed within the RAS have been proposed to contribute to many of the neuropathological hallmarks of AD, including the neuronal, cognitive, and vascular dysfunctions. Accumulating evidence has additionally identified antihypertensive medications targeting the RAS, particularly angiotensin receptor blockers (ARBs) and angiotensin-converting enzyme inhibitors (ACEIs), to delay AD onset and progression. In this review, we will discuss the emergence of the RAS's involvement in AD and highlight putative mechanisms of action underlying ARB's beneficial effects that may explain their ability to modify the risk of developing AD or AD progression. The RAS may provide novel molecular targets for recovering memory pathways, cerebrovascular function, and other pathological landmarks of AD.


Subject(s)
Alzheimer Disease/drug therapy , Angiotensin Receptor Antagonists/pharmacology , Brain/drug effects , Receptors, Angiotensin , Aged , Alzheimer Disease/prevention & control , Angiotensin II , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Humans , Renin-Angiotensin System
5.
Hypertension ; 75(6): 1464-1474, 2020 06.
Article in English | MEDLINE | ID: mdl-32362228

ABSTRACT

Antihypertensive medications targeting the renin-angiotensin system have lowered the incidence and progression of Alzheimer disease. Understanding how these medications function could lead to novel therapeutic strategies. AT4Rs (angiotensin IV receptors) have been associated with angiotensin receptor blockers' cognitive, cerebrovascular, and neuroinflammatory rescue in Alzheimer disease models. Yet, whether AT4Rs act alone or with AT2Rs remains unknown. Here, we investigated whether AT2Rs contribute to losartan's benefits and whether chronic AT2R activation could mimic angiotensin receptor blocker benefits in transgenic mice overexpressing familial Alzheimer disease mutations of the human APP (amyloid precursor protein). Losartan-treated mice (10 mg/kg per day, drinking water, 7 months) received intracerebroventricular (1 month) administration of vehicle or AT2R antagonist PD123319 (1.6 nmol/day). PD123319 countered losartan's benefits on spatial learning and memory, neurovascular coupling, and hampered those on oxidative stress and nitric oxide bioavailability. PD123319 did not oppose losartan's benefits on short-term memory and vasodilatory function and had no benefit on neuroinflammation or Aß (amyloid ß) pathology. Mice receiving either vehicle or selective AT2R agonist compound 21 (intracerebroventricular: 1 nmol/day, 1 month or drinking water: 10 mg/kg per day, 7 months), showed no improvement in memory, vasodilatory function, or nitric oxide bioavailability. Compound 21 treatment normalized neurovascular coupling, reduced astrogliosis independent of persisting microgliosis, and exacerbated oxidative stress in APP mice. Compound 21 reduced dense core Aß plaques, but not diffuse plaques or Aß species. Our findings suggest that targeting AT2Rs is not an ideal strategy for restoring Aß-related cognitive and cerebrovascular deficits.


Subject(s)
Alzheimer Disease , Imidazoles/pharmacology , Neurovascular Coupling/drug effects , Plaque, Amyloid , Pyridines/pharmacology , Receptor, Angiotensin, Type 2/metabolism , Receptors, Angiotensin/metabolism , Renin-Angiotensin System/drug effects , Sulfonamides/pharmacology , Thiophenes/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Cognition/drug effects , Disease Models, Animal , Losartan/pharmacology , Mice , Neuroimmunomodulation , Plaque, Amyloid/immunology , Plaque, Amyloid/pathology , Vasodilation/drug effects
6.
Neurobiol Dis ; 134: 104644, 2020 02.
Article in English | MEDLINE | ID: mdl-31669735

ABSTRACT

Angiotensin II type 1 receptor antagonists like losartan have been found to lower the incidence and progression to Alzheimer's disease (AD), as well as rescue cognitive and cerebrovascular deficits in AD mouse models. We previously found that co-administration of an angiotensin IV (AngIV) receptor (AT4R) antagonist prevented losartan's benefits, identifying AT4Rs as a possible target to counter AD pathogenesis. Therein, we investigated whether directly targeting AT4Rs could counter AD pathogenesis in a well-characterized mouse model of AD. Wild-type and human amyloid precursor protein (APP) transgenic (J20 line) mice (4.5 months old) received vehicle or AngIV (~1.3 nmol/day, 1 month) intracerebroventricularly via osmotic minipumps. AngIV restored short-term memory, spatial learning and memory in APP mice. AngIV normalized hippocampal AT4R levels, increased hippocampal subgranular zone cellular proliferation and dendritic arborization, and reduced oxidative stress. AngIV rescued whisker-evoked neurovascular coupling, endothelial- and smooth muscle cell-mediated cerebral vasodilatory responses, and cerebrovascular nitric oxide bioavailability. AngIV did not alter blood pressure, neuroinflammation or amyloid-ß (Aß) pathology. These preclinical findings identify AT4R as a promising target to counter Aß-related cognitive and cerebrovascular deficits in AD.


Subject(s)
Alzheimer Disease/pathology , Angiotensin II/analogs & derivatives , Hippocampus/drug effects , Memory/drug effects , Neurovascular Coupling/drug effects , Amyloid beta-Protein Precursor/genetics , Angiotensin II/pharmacology , Animals , Disease Models, Animal , Humans , Infusions, Intraventricular , Mice , Mice, Inbred C57BL , Mice, Transgenic
7.
FASEB J ; 33(12): 13280-13293, 2019 12.
Article in English | MEDLINE | ID: mdl-31557051

ABSTRACT

Aerobic physical exercise (EX) and controlling cardiovascular risk factors in midlife can improve and protect cognitive function in healthy individuals and are considered to be effective at reducing late-onset dementia incidence. By investigating commonalities between these preventative approaches, we sought to identify possible targets for effective interventions. We compared the efficacy of EX and simvastatin (SV) pharmacotherapy to counteract cognitive deficits induced by a high-cholesterol diet (2%, HCD) in mice overexpressing TGF-ß1 (TGF mice), a model of vascular cognitive impairment and dementia. Cognitive deficits were found in hypercholesterolemic mice for object recognition memory, and both SV and EX prevented this decline. EX improved stimulus-evoked cerebral blood flow responses and was as effective as SV in normalizing endothelium-dependent vasodilatory responses in cerebral arteries. The up-regulation of galectin-3-positive microglial cells in white matter (WM) of HCD-fed TGF mice with cognitive deficits was significantly reduced by both SV and EX concurrently with cognitive recovery. Altered hippocampal neurogenesis, gray matter astrogliosis, or microgliosis did not correlate with cognitive deficits or benefits. Overall, results indicate that SV and EX prevented cognitive decline in hypercholesterolemic mice and that they share common sites of action in preventing endothelial cell dysfunction and reducing WM inflammation.-Trigiani, L. J., Royea, J., Tong, X.-K., Hamel, E. Comparative benefits of simvastatin and exercise in a mouse model of vascular cognitive impairment and dementia.


Subject(s)
Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/therapy , Dementia/drug therapy , Dementia/therapy , Physical Conditioning, Animal/methods , Simvastatin/therapeutic use , Animals , Cerebrovascular Circulation/drug effects , Disease Models, Animal , Inflammation/drug therapy , Inflammation/therapy , Mice , Microglia/drug effects , Microglia/metabolism
8.
Hypertension ; 72(5): 1217-1226, 2018 11.
Article in English | MEDLINE | ID: mdl-30354809

ABSTRACT

The angiotensin receptor blocker losartan mitigated cerebrovascular and cognitive deficits in mouse models of Alzheimer disease, in line with some clinical evidence of reduced onset and progression to Alzheimer disease. We investigated whether these benefits apply to another angiotensin receptor blocker, namely candesartan. Adult transgenic mice overexpressing a mutated form of the human APP (amyloid precursor protein) and wild-type controls were treated with vehicle or candesartan (cohort 1: 2 months, 1 mg/kg per day, osmotic subcutaneous minipumps; cohort 2: 5 months, 10 mg/kg per day in drinking water). Candesartan largely restored endothelial and smooth muscle function and reduced neuroinflammation in both cohorts, without improving sensory evoked cerebral blood flow responses. Candesartan exerted restorative effects on the reduced number of Ki67-immunopositive proliferating cells in the granule cell layer of the hippocampus but not on that of DCX (doublecortin)-positive immature granule cells, despite normalizing the length of their dendritic projections in the molecular layer. Amyloid plaque load and impaired cognitive function were unaltered by candesartan, and blood pressure was decreased in treated APP and wild-type mice. Overall, findings show that candesartan shared several advantages reported previously for losartan, but it exhibited limited cognitive benefits and stronger blood pressure lowering effects. The choice of angiotensin receptor blocker may thus be critical for therapeutic efficacy in patients with vascular diseases at high risk of developing Alzheimer disease.


Subject(s)
Alzheimer Disease/drug therapy , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Benzimidazoles/therapeutic use , Brain/drug effects , Tetrazoles/therapeutic use , Alzheimer Disease/genetics , Alzheimer Disease/physiopathology , Amyloid beta-Protein Precursor/genetics , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Blood Pressure/drug effects , Brain/physiopathology , Cell Proliferation/drug effects , Cognition/drug effects , Disease Models, Animal , Doublecortin Protein , Male , Mice , Mice, Transgenic , Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Tetrazoles/pharmacology
9.
J Neurosci ; 37(22): 5562-5573, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28476949

ABSTRACT

The use of angiotensin receptor blockers (ARBs) correlates with reduced onset and progression of Alzheimer's disease (AD). The mechanism depicting how ARBs such as losartan restore cerebrovascular and cognitive deficits in AD is unknown. Here, we propose a mechanism underlying losartan's benefits by selectively blocking the effects of angiotensin IV (AngIV) at its receptor (AT4R) with divalinal in mice overexpressing the AD-related Swedish and Indiana mutations of the human amyloid precursor protein (APP mice) and WT mice. Young (3-month-old) mice were treated with losartan (∼10 mg/kg/d, 4 months), followed by intracerebroventricular administration of vehicle or divalinal in the final month of treatment. Spatial learning and memory were assessed using Morris water mazes at 3 and 4 months of losartan treatment. Cerebrovascular reactivity and whisker-evoked neurovascular coupling responses were measured at end point (∼7 months of age), together with biomarkers related to neuronal and vascular oxidative stress (superoxide dismutase-2), neuroinflammation (astroglial and microglial activation), neurogenesis (BrdU-labeled newborn cells), and amyloidosis [soluble amyloid-ß (Aß) species and Aß plaque load]. Divalinal countered losartan's capacity to rescue spatial learning and memory and blocked losartan's benefits on dilatory function and baseline nitric oxide bioavailability. Divalinal reverted losartan's anti-inflammatory effects, but failed to modify losartan-mediated reductions in oxidative stress. Neither losartan nor divalinal affected arterial blood pressure or significantly altered the amyloid pathology in APP mice. Our findings identify activation of the AngIV/AT4R cascade as the underlying mechanism in losartan's benefits and a target that could restore Aß-related cognitive and cerebrovascular deficits in AD.SIGNIFICANCE STATEMENT Antihypertensive medications that target the renin angiotensin system, such as angiotensin receptor blockers (ARBs), have been associated with lower incidence and progression of Alzheimer's disease (AD) in cohort studies. However, the manner by which ARBs mediate their beneficial effects is unknown. Here, the angiotensin IV receptor (AT4R) was identified as mediating the cognitive and cerebrovascular rescue of losartan, a commonly prescribed ARB, in a mouse model of AD. The AT4R was further implicated in mediating anti-inflammatory benefits. AT4R-mediated effects were independent from changes in blood pressure, amyloidosis, and oxidative stress. Overall, our results implicate the angiotensin IV/AT4R cascade as a promising candidate for AD intervention.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/physiopathology , Cerebrovascular Circulation/drug effects , Cognition Disorders/drug therapy , Cognition Disorders/physiopathology , Losartan/administration & dosage , Receptors, Angiotensin/metabolism , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Cognition/drug effects , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Spatial Learning/drug effects , Spatial Memory/drug effects
10.
Cell Mol Neurobiol ; 36(2): 219-32, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26993506

ABSTRACT

Alzheimer's disease (AD) is a multifactorial and multifaceted disease for which we currently have very little to offer since there is no curative therapy, with only limited disease-modifying drugs. Recent studies in AD mouse models that recapitulate the amyloid-ß (Aß) pathology converge to demonstrate that it is possible to salvage cerebrovascular function with a variety of drugs and, particularly, therapies used to treat cardiovascular diseases such as hypercholesterolemia and hypertension. These drugs can reestablish dilatory function mediated by various endothelial and smooth muscle ion channels as well as nitric oxide availability, benefits that result in normalized brain perfusion. These cerebrovascular benefits would favor brain perfusion, which may help maintain neuronal function and, possibly, delay cognitive failure. However, restoring cerebrovascular function in AD mouse models was not necessarily accompanied by rescue of cognitive deficits related to spatial learning and memory. The results with cardiovascular therapies rather suggest that drugs originally designed to treat cardiovascular diseases that concurrently restore cerebrovascular and cognitive function do so through their pleiotropic effects. Specifically, recent findings suggest that these drugs act directly on brain cells and neuronal pathways involved in memory formation, hence, working simultaneously albeit independently on neuronal and vascular targets. These findings may help select medications for patients with cardiovascular diseases at risk of developing AD with increasing age. Further, they may identify molecular targets for recovering memory pathways that bear potential for new therapeutic avenues.


Subject(s)
Alzheimer Disease/therapy , Brain/blood supply , Brain/pathology , Cognition Disorders/therapy , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Alzheimer Disease/physiopathology , Amyloid beta-Peptides/metabolism , Animals , Cognition Disorders/physiopathology , Humans , Losartan/therapeutic use , Simvastatin/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...