Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
3.
Mol Neurodegener ; 18(1): 7, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36707892

ABSTRACT

BACKGROUND: PICALM is one of the most significant susceptibility factors for Alzheimer's disease (AD). In humans and mice, PICALM is highly expressed in brain endothelium. PICALM endothelial levels are reduced in AD brains. PICALM controls several steps in Aß transcytosis across the blood-brain barrier (BBB). Its loss from brain endothelium in mice diminishes Aß clearance at the BBB, which worsens Aß pathology, but is reversible by endothelial PICALM re-expression. Thus, increasing PICALM at the BBB holds potential to slow down development of Aß pathology. METHODS: To identify a drug that could increase PICALM expression, we screened a library of 2007 FDA-approved drugs in HEK293t cells expressing luciferase driven by a human PICALM promoter, followed by a secondary mRNA screen in human Eahy926 endothelial cell line. In vivo studies with the lead hit were carried out in Picalm-deficient (Picalm+/-) mice, Picalm+/-; 5XFAD mice and Picalmlox/lox; Cdh5-Cre; 5XFAD mice with endothelial-specific Picalm knockout. We studied PICALM expression at the BBB, Aß pathology and clearance from brain to blood, cerebral blood flow (CBF) responses, BBB integrity and behavior. RESULTS: Our screen identified anti-malaria drug artesunate as the lead hit. Artesunate elevated PICALM mRNA and protein levels in Eahy926 endothelial cells and in vivo in brain capillaries of Picalm+/- mice by 2-3-fold. Artesunate treatment (32 mg/kg/day for 2 months) of 3-month old Picalm+/-; 5XFAD mice compared to vehicle increased brain capillary PICALM levels by 2-fold, and reduced Aß42 and Aß40 levels and Aß and thioflavin S-load in the cortex and hippocampus, and vascular Aß load by 34-51%. Artesunate also increased circulating Aß42 and Aß40 levels by 2-fold confirming accelerated Aß clearance from brain to blood. Consistent with reduced Aß pathology, treatment of Picalm+/-; 5XFAD mice with artesunate improved CBF responses, BBB integrity and behavior on novel object location and recognition, burrowing and nesting. Endothelial-specific knockout of PICALM abolished all beneficial effects of artesunate in 5XFAD mice indicating that endothelial PICALM is required for its therapeutic effects. CONCLUSIONS: Artesunate increases PICALM levels and Aß clearance at the BBB which prevents development of Aß pathology and functional deficits in mice and holds potential for translation to human AD.


Subject(s)
Alzheimer Disease , Antimalarials , Monomeric Clathrin Assembly Proteins , Animals , Mice , Humans , Infant , Blood-Brain Barrier/metabolism , Artesunate/pharmacology , Artesunate/metabolism , Artesunate/therapeutic use , Antimalarials/pharmacology , Antimalarials/metabolism , Antimalarials/therapeutic use , Endothelial Cells/metabolism , HEK293 Cells , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/metabolism , Mice, Transgenic , Monomeric Clathrin Assembly Proteins/metabolism , Monomeric Clathrin Assembly Proteins/pharmacology
4.
Front Cell Neurosci ; 16: 1078919, 2022.
Article in English | MEDLINE | ID: mdl-36523817

ABSTRACT

[This corrects the article DOI: 10.3389/fncel.2020.00027.].

5.
J Exp Med ; 219(1)2022 01 03.
Article in English | MEDLINE | ID: mdl-34846535

ABSTRACT

Subcortical white matter (WM) stroke accounts for 25% of all strokes and is the second leading cause of dementia. Despite such clinical importance, we still do not have an effective treatment for ischemic WM stroke, and the mechanisms of WM postischemic neuroprotection remain elusive. 3K3A-activated protein C (APC) is a signaling-selective analogue of endogenous blood protease APC that is currently in development as a neuroprotectant for ischemic stroke patients. Here, we show that 3K3A-APC protects WM tracts and oligodendrocytes from ischemic injury in the corpus callosum in middle-aged mice by activating protease-activated receptor 1 (PAR1) and PAR3. We show that PAR1 and PAR3 were also required for 3K3A-APC's suppression of post-WM stroke microglia and astrocyte responses and overall improvement in neuropathologic and functional outcomes. Our data provide new insights into the neuroprotective APC pathway in the WM and illustrate 3K3A-APC's potential for treating WM stroke in humans, possibly including multiple WM strokes that result in vascular dementia.


Subject(s)
Corpus Callosum/metabolism , Ischemia/metabolism , Oligodendroglia/metabolism , Protein C/metabolism , White Matter/metabolism , Animals , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Chenodeoxycholic Acid/analogs & derivatives , Chenodeoxycholic Acid/pharmacology , Corpus Callosum/drug effects , Disease Models, Animal , Enzyme Activation/drug effects , Fibrinolytic Agents/metabolism , Fibrinolytic Agents/pharmacology , Humans , Ischemia/physiopathology , Ischemia/prevention & control , Male , Mice, Inbred C57BL , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Protein C/pharmacology , Receptor, PAR-1/metabolism , Receptors, Thrombin/metabolism , Stroke/metabolism , Stroke/prevention & control
6.
Nat Aging ; 1(6): 506-520, 2021 06.
Article in English | MEDLINE | ID: mdl-35291561

ABSTRACT

Apolipoprotein E4 (APOE4), the main susceptibility gene for Alzheimer's disease (AD), leads to vascular dysfunction, amyloid-ß pathology, neurodegeneration and dementia. How these different pathologies contribute to advanced-stage AD remains unclear. Using aged APOE knock-in mice crossed with 5xFAD mice, we show that, compared to APOE3, APOE4 accelerates blood-brain barrier (BBB) breakdown, loss of cerebral blood flow, neuronal loss and behavioral deficits independently of amyloid-ß. BBB breakdown was associated with activation of the cyclophilin A-matrix metalloproteinase-9 BBB-degrading pathway in pericytes. Suppression of this pathway improved BBB integrity and prevented further neuronal loss and behavioral deficits in APOE4;5FAD mice while having no effect on amyloid-ß pathology. Thus, APOE4 accelerates advanced-stage BBB breakdown and neurodegeneration in Alzheimer's mice via the cyclophilin A pathway in pericytes independently of amyloid-ß, which has implication for the pathogenesis and treatment of vascular and neurodegenerative disorder in AD.


Subject(s)
Alzheimer Disease , Neurodegenerative Diseases , Mice , Animals , Apolipoprotein E4/genetics , Alzheimer Disease/genetics , Cyclophilin A/genetics , Amyloid beta-Peptides/metabolism
8.
Front Cell Neurosci ; 14: 27, 2020.
Article in English | MEDLINE | ID: mdl-32116568

ABSTRACT

Pericytes are perivascular mural cells that enwrap brain capillaries and maintain blood-brain barrier (BBB) integrity. Most studies suggest that pericytes regulate cerebral blood flow (CBF) and oxygen delivery to activated brain structures, known as neurovascular coupling. While we have previously shown that congenital loss of pericytes leads over time to aberrant hemodynamic responses, the effects of acute global pericyte loss on neurovascular coupling have not been studied. To address this, we used our recently reported inducible pericyte-specific Cre mouse line crossed to iDTR mice carrying Cre-dependent human diphtheria toxin (DT) receptor, which upon DT treatment leads to acute pericyte ablation. As expected, DT led to rapid progressive loss of pericyte coverage of cortical capillaries up to 50% at 3 days post-DT, which correlated with approximately 50% reductions in stimulus-induced CBF responses measured with laser doppler flowmetry (LDF) and/or intrinsic optical signal (IOS) imaging. Endothelial response to acetylcholine, microvascular density, and neuronal evoked membrane potential responses remained, however, unchanged, as well as arteriolar smooth muscle cell (SMC) coverage and functional responses to adenosine, as we previously reported. Together, these data suggest that neurovascular uncoupling in this model is driven by pericyte loss, but not other vascular deficits or neuronal dysfunction. These results further support the role of pericytes in CBF regulation and may have implications for neurological conditions associated with rapid pericyte loss such as hypoperfusion and stroke, as well as conditions where the exact time course of global regional pericyte loss is less clear, such as Alzheimer's disease (AD) and other neurogenerative disorders.

9.
Neurobiol Dis ; 136: 104745, 2020 03.
Article in English | MEDLINE | ID: mdl-31931140

ABSTRACT

Food restriction has been widely associated with beneficial effects on brain aging and age-related neurodegenerative diseases such as Alzheimer's disease. However, previous studies on the effects of food restriction on aging- or pathology-related cognitive decline are controversial, emphasizing the importance of the type, onset and duration of food restriction. In the present study, we assessed the effects of preventive every-other-day (EOD) feeding regimen on neurodegenerative phenotype in 5XFAD transgenic mice, a commonly used mouse model of Alzheimer's disease. EOD feeding regimen was introduced to transgenic female mice at the age of 2 months and the effects on amyloid-ß (Aß) accumulation, gliosis, synaptic plasticity, and blood-brain barrier breakdown were analyzed in cortical tissue of 6-month-old animals. Surprisingly, significant increase of inflammation in the cortex of 5XFAD fed EOD mice was observed, reflected by the expression of microglial and astrocytic markers. This increase in reactivity and/or proliferation of glial cells was accompanied by an increase in proinflammatory cytokine TNF-α, p38 MAPK and EAAT2, and a decrease in GAD67. NMDA receptor subunit 2B, related to glutamate excitotoxicity, was increased in the cortex of 5XFAD-EOD mice indicating additional alterations in glutamatergic signaling. Furthermore, 4 months of EOD feeding regimen had led to synaptic plasticity proteins reduction and neuronal injury in 5XFAD mice. However, EOD feeding regimen did not affect Aß load and blood-brain barrier permeability in the cortex of 5XFAD mice. Our results demonstrate that EOD feeding regimen exacerbates Alzheimer's disease-like neurodegenerative and neuroinflammatory changes irrespective of Aß pathology in 5XFAD mice, suggesting that caution should be paid when using food restrictions in the prodromal phase of this neurodegenerative disease.


Subject(s)
Alzheimer Disease/metabolism , Disease Models, Animal , Fasting/adverse effects , Fasting/metabolism , Inflammation Mediators/metabolism , Neurons/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Female , Inflammation/metabolism , Inflammation/pathology , Mice , Mice, Transgenic , Neurons/pathology
10.
Nat Neurosci ; 22(7): 1089-1098, 2019 07.
Article in English | MEDLINE | ID: mdl-31235908

ABSTRACT

Pericytes are positioned between brain capillary endothelial cells, astrocytes and neurons. They degenerate in multiple neurological disorders. However, their role in the pathogenesis of these disorders remains debatable. Here we generate an inducible pericyte-specific Cre line and cross pericyte-specific Cre mice with iDTR mice carrying Cre-dependent human diphtheria toxin receptor. After pericyte ablation with diphtheria toxin, mice showed acute blood-brain barrier breakdown, severe loss of blood flow, and a rapid neuron loss that was associated with loss of pericyte-derived pleiotrophin (PTN), a neurotrophic growth factor. Intracerebroventricular PTN infusions prevented neuron loss in pericyte-ablated mice despite persistent circulatory changes. Silencing of pericyte-derived Ptn rendered neurons vulnerable to ischemic and excitotoxic injury. Our data demonstrate a rapid neurodegeneration cascade that links pericyte loss to acute circulatory collapse and loss of PTN neurotrophic support. These findings may have implications for the pathogenesis and treatment of neurological disorders that are associated with pericyte loss and/or neurovascular dysfunction.


Subject(s)
Carrier Proteins/physiology , Cytokines/physiology , Nerve Degeneration/physiopathology , Nerve Tissue Proteins/physiology , Neurons/pathology , Pericytes/physiology , Shock/physiopathology , Animals , Brain Ischemia/physiopathology , Capillaries/physiopathology , Carrier Proteins/therapeutic use , Cells, Cultured , Cerebrovascular Circulation/physiology , Cytokines/deficiency , Cytokines/therapeutic use , Endothelial Cells/cytology , Female , Genes, Reporter , Infusions, Intraventricular , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Nerve Degeneration/drug therapy , Neuroglia/metabolism , Neurons/metabolism , Neurotoxins/toxicity , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism , Shock/metabolism , Shock/pathology
11.
J Exp Med ; 216(2): 279-293, 2019 02 04.
Article in English | MEDLINE | ID: mdl-30647119

ABSTRACT

3K3A-activated protein C (APC), a cell-signaling analogue of endogenous blood serine protease APC, exerts vasculoprotective, neuroprotective, and anti-inflammatory activities in rodent models of stroke, brain injury, and neurodegenerative disorders. 3K3A-APC is currently in development as a neuroprotectant in patients with ischemic stroke. Here, we report that 3K3A-APC inhibits BACE1 amyloidogenic pathway in a mouse model of Alzheimer's disease (AD). We show that a 4-mo daily treatment of 3-mo-old 5XFAD mice with murine recombinant 3K3A-APC (100 µg/kg/d i.p.) prevents development of parenchymal and cerebrovascular amyloid-ß (Aß) deposits by 40-50%, which is mediated through NFκB-dependent transcriptional inhibition of BACE1, resulting in blockade of Aß generation in neurons overexpressing human Aß-precursor protein. Consistent with reduced Aß deposition, 3K3A-APC normalized hippocampus-dependent behavioral deficits and cerebral blood flow responses, improved cerebrovascular integrity, and diminished neuroinflammatory responses. Our data suggest that 3K3A-APC holds potential as an effective anti-Aß prevention therapy for early-stage AD.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/metabolism , Protein C/pharmacology , Recombinant Proteins/pharmacology , Signal Transduction/drug effects , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/genetics , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Disease Models, Animal , Mice , Mice, Transgenic , Signal Transduction/genetics
12.
Nat Protoc ; 13(6): 1377-1402, 2018 06.
Article in English | MEDLINE | ID: mdl-29844521

ABSTRACT

Cerebrovascular dysfunction has an important role in the pathogenesis of multiple brain disorders. Measurement of hemodynamic responses in vivo can be challenging, particularly as techniques are often not described in sufficient detail and vary between laboratories. We present a set of standardized in vivo protocols that describe high-resolution two-photon microscopy and intrinsic optical signal (IOS) imaging to evaluate capillary and arteriolar responses to a stimulus, regional hemodynamic responses, and oxygen delivery to the brain. The protocol also describes how to measure intrinsic NADH fluorescence to understand how blood O2 supply meets the metabolic demands of activated brain tissue, and to perform resting-state absolute oxygen partial pressure (pO2) measurements of brain tissue. These methods can detect cerebrovascular changes at far higher resolution than MRI techniques, although the optical nature of these techniques limits their achievable imaging depths. Each individual procedure requires 1-2 h to complete, with two to three procedures typically performed per animal at a time. These protocols are broadly applicable in studies of cerebrovascular function in healthy and diseased brain in any of the existing mouse models of neurological and vascular disorders. All these procedures can be accomplished by a competent graduate student or experienced technician, except the two-photon measurement of absolute pO2 level, which is better suited to a more experienced, postdoctoral-level researcher.


Subject(s)
Cerebrovascular Circulation/physiology , Cerebrovascular Disorders/diagnostic imaging , Cerebrovascular Disorders/pathology , Hemodynamics , Hypoxia/diagnostic imaging , Hypoxia/pathology , Intravital Microscopy/methods , Animals , Intravital Microscopy/standards , Mice
13.
Nat Med ; 24(3): 326-337, 2018 03.
Article in English | MEDLINE | ID: mdl-29400711

ABSTRACT

Diffuse white-matter disease associated with small-vessel disease and dementia is prevalent in the elderly. The biological mechanisms, however, remain elusive. Using pericyte-deficient mice, magnetic resonance imaging, viral-based tract-tracing, and behavior and tissue analysis, we found that pericyte degeneration disrupted white-matter microcirculation, resulting in an accumulation of toxic blood-derived fibrin(ogen) deposits and blood-flow reductions, which triggered a loss of myelin, axons and oligodendrocytes. This disrupted brain circuits, leading to white-matter functional deficits before neuronal loss occurs. Fibrinogen and fibrin fibrils initiated autophagy-dependent cell death in oligodendrocyte and pericyte cultures, whereas pharmacological and genetic manipulations of systemic fibrinogen levels in pericyte-deficient, but not control mice, influenced the degree of white-matter fibrin(ogen) deposition, pericyte degeneration, vascular pathology and white-matter changes. Thus, our data indicate that pericytes control white-matter structure and function, which has implications for the pathogenesis and treatment of human white-matter disease associated with small-vessel disease.


Subject(s)
Central Nervous System/physiopathology , Dementia/physiopathology , Leukoencephalopathies/physiopathology , White Matter/physiopathology , Animals , Axons/pathology , Blood Vessels/diagnostic imaging , Blood Vessels/pathology , Blood-Brain Barrier/pathology , Brain/diagnostic imaging , Brain/metabolism , Brain/physiopathology , Central Nervous System/blood supply , Central Nervous System/diagnostic imaging , Dementia/blood , Dementia/diagnostic imaging , Humans , Leukoencephalopathies/blood , Leukoencephalopathies/diagnostic imaging , Magnetic Resonance Imaging , Mice , Microcirculation , Myelin Sheath/metabolism , Pericytes/metabolism , Pericytes/pathology , White Matter/blood supply , White Matter/diagnostic imaging
14.
Nat Neurosci ; 20(3): 406-416, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28135240

ABSTRACT

Pericytes are perivascular mural cells of brain capillaries. They are positioned centrally in the neurovascular unit between endothelial cells, astrocytes and neurons. This position allows them to regulate key neurovascular functions of the brain. The role of pericytes in the regulation of cerebral blood flow (CBF) and neurovascular coupling remains, however, under debate. Using loss-of-function pericyte-deficient mice, here we show that pericyte degeneration diminishes global and individual capillary CBF responses to neuronal stimuli, resulting in neurovascular uncoupling, reduced oxygen supply to the brain and metabolic stress. Neurovascular deficits lead over time to impaired neuronal excitability and neurodegenerative changes. Thus, pericyte degeneration as seen in neurological disorders such as Alzheimer's disease may contribute to neurovascular dysfunction and neurodegeneration associated with human disease.


Subject(s)
Brain/blood supply , Cell Death/physiology , Nerve Degeneration/physiopathology , Oxygen/metabolism , Pericytes/pathology , Animals , Brain/metabolism , Capillaries/physiology , Female , Homeodomain Proteins/genetics , Male , Mice , Mice, Transgenic , Neurons/physiology , Receptor, Platelet-Derived Growth Factor beta/genetics , Stress, Physiological/physiology , Vasodilation/physiology
15.
Neurochem Int ; 96: 69-76, 2016 06.
Article in English | MEDLINE | ID: mdl-26939764

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of death and disability in humans. Subsequent pathological events occurring in the brain after TBI, referred to as secondary injury, continue to damage surrounding tissue resulting in substantial neuronal loss. Using an animal model of TBI we examined the effect of dietary restriction (DR) on the neuroapoptosis and Bcl-2 family genes as the main regulators of the intrinsic apoptotic pathway. Bcl-2, Bcl-xl and Bax mRNA and protein expression in the ipsilateral cortex of adult Wistar rats exposed to DR before TBI were studied from 2 to 28 days post injury. Our results showed that DR suppressed neuroapoptosis and promoted significant upregulation of antiapoptotic Bcl-2 and Bcl-xl mRNAs in the ipsilateral cortex following injury. Expression of the proapoptotic Bax gene increased in ad libitum (AL) fed rats but remained unchanged in rats exposed to DR. Although the expression of Bcl-2, Bcl-xl and Bax proteins was changed in a similar manner in both experimental groups, DR promoted a continuous increase in the Bcl-2:Bax protein ratio throughout the recovery period. Together with our previous finding that DR mediates inhibition of the extrinsic apoptotic pathway the present work reveals that modulation of the intrinsic pathway contributes to the beneficial effect of DR in brain injury. These findings provide new insight into the effects of DR on pro-survival signaling after injury, lending further support to its neuroprotective effect.


Subject(s)
Brain Injuries/metabolism , Caloric Restriction/trends , Cerebral Cortex/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , bcl-2-Associated X Protein/biosynthesis , bcl-X Protein/biosynthesis , Animals , Apoptosis/physiology , Brain Injuries/pathology , Brain Injuries/prevention & control , Cerebral Cortex/pathology , Male , Random Allocation , Rats , Rats, Wistar
16.
J Steroid Biochem Mol Biol ; 149: 43-52, 2015 May.
Article in English | MEDLINE | ID: mdl-25616002

ABSTRACT

Diminished glucocorticoid signaling is associated with an age-related decline in hippocampal functioning. In this study we demonstrate the effect of intermittent, every other day (EOD) feeding on the glucocorticoid hormone/glucocorticoid receptor (GR) system in the hippocampus of middle-aged (18-month-old) and aged (24-month-old) Wistar rats. In aged ad libitum-fed rats, a decrease in the level of total GR and GR phosphorylated at Ser(232) (pGR) was detected. Conversely, aged rats subjected to EOD feeding, starting from 6 months of age, showed an increase in GR and pGR levels and a higher content of hippocampal corticosterone. Furthermore, prominent nuclear staining of pGR was observed in CA1 pyramidal and DG granule neurons of aged EOD-fed rats. These changes were accompanied by increased Sgk-1 and decreased GFAP transcription, pointing to upregulated transcriptional activity of GR. EOD feeding also induced an increase in the expression of the mineralocorticoid receptor. Our results reveal that intermittent feeding restores impaired GR signaling in the hippocampus of aged animals by inducing rather than by stabilizing GR signaling during aging.


Subject(s)
Aging , Food Deprivation/physiology , Hippocampus/physiology , Receptors, Glucocorticoid/metabolism , Signal Transduction , 11-beta-Hydroxysteroid Dehydrogenase Type 1/analysis , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , Animals , Corticosterone/analysis , Corticosterone/metabolism , HSP90 Heat-Shock Proteins/analysis , HSP90 Heat-Shock Proteins/metabolism , Immediate-Early Proteins/genetics , Male , Phosphotransferases/analysis , Phosphotransferases/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, Glucocorticoid/analysis , Tacrolimus Binding Proteins/analysis , Tacrolimus Binding Proteins/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...