Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 270
Filtrer
1.
Open Biol ; 14(6): 230349, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38862017

RÉSUMÉ

Coronavirus disease 2019 (COVID-19) was initially considered a primarily respiratory disease but is now known to affect other organs including the heart and brain. A major route by which COVID-19 impacts different organs is via the vascular system. We studied the impact of apolipoprotein E (APOE) genotype and inflammation on vascular infectivity by pseudo-typed severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) viruses in mouse and human cultured endothelial cells and pericytes. Possessing the APOE4 allele or having existing systemic inflammation is known to enhance the severity of COVID-19. Using targeted replacement human APOE3 and APOE4 mice and inflammation induced by bacterial lipopolysaccharide (LPS), we investigated infection by SARS-CoV-2. Here, we show that infectivity was higher in murine cerebrovascular pericytes compared to endothelial cells and higher in cultures expressing APOE4. Furthermore, increasing the inflammatory state of the cells by prior incubation with LPS increased infectivity into human and mouse pericytes and human endothelial cells. Our findings provide insights into the mechanisms underlying severe COVID-19 infection, highlighting how risk factors such as APOE4 genotype and prior inflammation may exacerbate disease severity by augmenting the virus's ability to infect vascular cells.


Sujet(s)
COVID-19 , Cellules endothéliales , Péricytes , SARS-CoV-2 , Péricytes/virologie , Péricytes/métabolisme , Péricytes/anatomopathologie , Humains , Animaux , SARS-CoV-2/physiologie , SARS-CoV-2/pathogénicité , COVID-19/virologie , COVID-19/anatomopathologie , Souris , Cellules endothéliales/virologie , Cellules endothéliales/métabolisme , Cellules endothéliales/anatomopathologie , Facteurs de risque , Lipopolysaccharides/pharmacologie , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Inflammation/virologie , Inflammation/anatomopathologie
2.
Front Endocrinol (Lausanne) ; 15: 1374825, 2024.
Article de Anglais | MEDLINE | ID: mdl-38742194

RÉSUMÉ

Increasing evidence suggests that female individuals have a higher Alzheimer's disease (AD) risk associated with post-menopausal loss of circulating estradiol (E2). However, clinical data are conflicting on whether E2 lowers AD risk. One potential contributing factor is APOE. The greatest genetic risk factor for AD is APOE4, a factor that is pronounced in female individuals post-menopause. Clinical data suggests that APOE impacts the response of AD patients to E2 replacement therapy. However, whether APOE4 prevents, is neutral, or promotes any positive effects of E2 is unclear. Therefore, our goal was to determine whether APOE modulates the impact of E2 on behavior and AD pathology in vivo. To that end, mice that express human APOE3 (E3FAD) or APOE4 (E4FAD) and overproduce Aß42 were ovariectomized at either 4 months (early) or 8 months (late) and treated with vehicle or E2 for 4 months. In E3FAD mice, we found that E2 mitigated the detrimental effect of ovariectomy on memory, with no effect on Aß in the early paradigm and only improved learning in the late paradigm. Although E2 lowered Aß in E4FAD mice in the early paradigm, there was no impact on learning or memory, possibly due to higher Aß pathology compared to E3FAD mice. In the late paradigm, there was no effect on learning/memory and Aß pathology in E4FAD mice. Collectively, these data support the idea that, in the presence of Aß pathology, APOE impacts the response to E2 supplementation post-menopause.


Sujet(s)
Apolipoprotéine E3 , Apolipoprotéine E4 , Oestradiol , Ovariectomie , Animaux , Femelle , Humains , Souris , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Peptides bêta-amyloïdes/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Comportement animal/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Oestradiol/pharmacologie , Souris transgéniques
3.
J Neuroimmune Pharmacol ; 19(1): 22, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38771543

RÉSUMÉ

SARS-CoV-2 spike proteins have been shown to cross the blood-brain barrier (BBB) in mice and affect the integrity of human BBB cell models. However, the effects of SARS-CoV-2 spike proteins in relation to sporadic, late onset, Alzheimer's disease (AD) risk have not been extensively investigated. Here we characterized the individual and combined effects of SARS-CoV-2 spike protein subunits S1 RBD, S1 and S2 on BBB cell types (induced brain endothelial-like cells (iBECs) and astrocytes (iAstrocytes)) generated from induced pluripotent stem cells (iPSCs) harboring low (APOE3 carrier) or high (APOE4 carrier) relative Alzheimer's risk. We found that treatment with spike proteins did not alter iBEC integrity, although they induced the expression of several inflammatory cytokines. iAstrocytes exhibited a robust inflammatory response to SARS-CoV-2 spike protein treatment, with differences found in the levels of cytokine secretion between spike protein-treated APOE3 and APOE4 iAstrocytes. Finally, we tested the effects of potentially anti-inflammatory drugs during SARS-CoV-2 spike protein exposure in iAstrocytes, and discovered different responses between spike protein treated APOE4 iAstrocytes and APOE3 iAstrocytes, specifically in relation to IL-6, IL-8 and CCL2 secretion. Overall, our results indicate that APOE3 and APOE4 iAstrocytes respond differently to anti-inflammatory drug treatment during SARS-CoV-2 spike protein exposure with potential implications to therapeutic responses.


Sujet(s)
Apolipoprotéine E3 , Apolipoprotéine E4 , Astrocytes , Barrière hémato-encéphalique , Cytokines , Glycoprotéine de spicule des coronavirus , Barrière hémato-encéphalique/métabolisme , Humains , Cytokines/métabolisme , Glycoprotéine de spicule des coronavirus/métabolisme , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Astrocytes/métabolisme , Astrocytes/virologie , Astrocytes/effets des médicaments et des substances chimiques , Apolipoprotéine E3/métabolisme , Cellules souches pluripotentes induites/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , SARS-CoV-2 , COVID-19/métabolisme , COVID-19/immunologie , Cellules cultivées
4.
Traffic ; 25(5): e12937, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38777335

RÉSUMÉ

The polymorphic APOE gene is the greatest genetic determinant of sporadic Alzheimer's disease risk: the APOE4 allele increases risk, while the APOE2 allele is neuroprotective compared with the risk-neutral APOE3 allele. The neuronal endosomal system is inherently vulnerable during aging, and APOE4 exacerbates this vulnerability by driving an enlargement of early endosomes and reducing exosome release in the brain of humans and mice. We hypothesized that the protective effects of APOE2 are, in part, mediated through the endosomal pathway. Messenger RNA analyses showed that APOE2 leads to an enrichment of endosomal pathways in the brain when compared with both APOE3 and APOE4. Moreover, we show age-dependent alterations in the recruitment of key endosomal regulatory proteins to vesicle compartments when comparing APOE2 to APOE3. In contrast to the early endosome enlargement previously shown in Alzheimer's disease and APOE4 models, we detected similar morphology and abundance of early endosomes and retromer-associated vesicles within cortical neurons of aged APOE2 targeted-replacement mice compared with APOE3. Additionally, we observed increased brain extracellular levels of endosome-derived exosomes in APOE2 compared with APOE3 mice during aging, consistent with enhanced endosomal cargo clearance by exosomes to the extracellular space. Our findings thus demonstrate that APOE2 enhances an endosomal clearance pathway, which has been shown to be impaired by APOE4 and which may be protective due to APOE2 expression during brain aging.


Sujet(s)
Vieillissement , Apolipoprotéine E2 , Encéphale , Endosomes , Exosomes , Animaux , Humains , Souris , Vieillissement/métabolisme , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/génétique , Apolipoprotéine E2/métabolisme , Apolipoprotéine E2/génétique , Apolipoprotéine E3/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E4/métabolisme , Apolipoprotéine E4/génétique , Encéphale/métabolisme , Endosomes/métabolisme , Exosomes/métabolisme , Souris de lignée C57BL , Neurones/métabolisme
5.
Arterioscler Thromb Vasc Biol ; 44(6): 1346-1364, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38660806

RÉSUMÉ

BACKGROUND: Atherosclerosis is the major underlying pathology of cardiovascular disease and is driven by dyslipidemia and inflammation. Inhibition of the immunoproteasome, a proteasome variant that is predominantly expressed by immune cells and plays an important role in antigen presentation, has been shown to have immunosuppressive effects. METHODS: We assessed the effect of ONX-0914, an inhibitor of the immunoproteasomal catalytic subunits LMP7 (proteasome subunit ß5i/large multifunctional peptidase 7) and LMP2 (proteasome subunit ß1i/large multifunctional peptidase 2), on atherosclerosis and metabolism in LDLr-/- and APOE*3-Leiden.CETP mice. RESULTS: ONX-0914 treatment significantly reduced atherosclerosis, reduced dendritic cell and macrophage levels and their activation, as well as the levels of antigen-experienced T cells during early plaque formation, and Th1 cells in advanced atherosclerosis in young and aged mice in various immune compartments. Additionally, ONX-0914 treatment led to a strong reduction in white adipose tissue mass and adipocyte progenitors, which coincided with neutrophil and macrophage accumulation in white adipose tissue. ONX-0914 reduced intestinal triglyceride uptake and gastric emptying, likely contributing to the reduction in white adipose tissue mass, as ONX-0914 did not increase energy expenditure or reduce total food intake. Concomitant with the reduction in white adipose tissue mass upon ONX-0914 treatment, we observed improvements in markers of metabolic syndrome, including lowered plasma triglyceride levels, insulin levels, and fasting blood glucose. CONCLUSIONS: We propose that immunoproteasomal inhibition reduces 3 major causes underlying cardiovascular disease, dyslipidemia, metabolic syndrome, and inflammation and is a new target in drug development for atherosclerosis treatment.


Sujet(s)
Tissu adipeux blanc , Athérosclérose , Modèles animaux de maladie humaine , Syndrome métabolique X , Souris de lignée C57BL , Proteasome endopeptidase complex , Récepteurs aux lipoprotéines LDL , Animaux , Athérosclérose/anatomopathologie , Athérosclérose/prévention et contrôle , Athérosclérose/traitement médicamenteux , Athérosclérose/immunologie , Athérosclérose/génétique , Athérosclérose/métabolisme , Syndrome métabolique X/traitement médicamenteux , Syndrome métabolique X/immunologie , Tissu adipeux blanc/métabolisme , Tissu adipeux blanc/effets des médicaments et des substances chimiques , Tissu adipeux blanc/anatomopathologie , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs aux lipoprotéines LDL/déficit , Proteasome endopeptidase complex/métabolisme , Mâle , Inhibiteurs du protéasome/pharmacologie , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Maladies de l'aorte/prévention et contrôle , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/enzymologie , Maladies de l'aorte/immunologie , Maladies de l'aorte/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/immunologie , Plaque d'athérosclérose , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Souris invalidées pour les gènes ApoE , Souris , Métabolisme énergétique/effets des médicaments et des substances chimiques , Oligopeptides
6.
JCI Insight ; 9(8)2024 04 22.
Article de Anglais | MEDLINE | ID: mdl-38646937

RÉSUMÉ

Sepsis is a leading cause of mortality worldwide, and pneumonia is the most common cause of sepsis in humans. Low levels of high-density lipoprotein cholesterol (HDL-C) levels are associated with an increased risk of death from sepsis, and increasing levels of HDL-C by inhibition of cholesteryl ester transfer protein (CETP) decreases mortality from intraabdominal polymicrobial sepsis in APOE*3-Leiden.CETP mice. Here, we show that treatment with the CETP inhibitor (CETPi) anacetrapib reduced mortality from Streptococcus pneumoniae-induced sepsis in APOE*3-Leiden.CETP and APOA1.CETP mice. Mechanistically, CETP inhibition reduced the host proinflammatory response via attenuation of proinflammatory cytokine transcription and release. This effect was dependent on the presence of HDL, leading to attenuation of immune-mediated organ damage. In addition, CETP inhibition promoted monocyte activation in the blood prior to the onset of sepsis, resulting in accelerated macrophage recruitment to the lung and liver. In vitro experiments demonstrated that CETP inhibition significantly promoted the activation of proinflammatory signaling in peripheral blood mononuclear cells and THP1 cells in the absence of HDL; this may represent a mechanism responsible for improved bacterial clearance during sepsis. These findings provide evidence that CETP inhibition represents a potential approach to reduce mortality from pneumosepsis.


Sujet(s)
Protéines de transfert des esters de cholestérol , Monocytes , Streptococcus pneumoniae , Animaux , Femelle , Humains , Souris , Apolipoprotéine E3/métabolisme , Protéines de transfert des esters de cholestérol/antagonistes et inhibiteurs , Protéines de transfert des esters de cholestérol/métabolisme , Cholestérol HDL/sang , Cholestérol HDL/métabolisme , Modèles animaux de maladie humaine , Macrophages/immunologie , Macrophages/métabolisme , Monocytes/immunologie , Monocytes/métabolisme , Pneumonie à pneumocoques/immunologie , Pneumonie à pneumocoques/mortalité , Pneumonie à pneumocoques/métabolisme , Pneumonie à pneumocoques/microbiologie , Sepsie/immunologie , Sepsie/mortalité , Sepsie/microbiologie , Sepsie/métabolisme , Streptococcus pneumoniae/immunologie , Cellules THP-1
7.
FEBS Lett ; 598(8): 902-914, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38529702

RÉSUMÉ

Apolipoprotein E (apoE) is a regulator of lipid metabolism, cholesterol transport, and the clearance and aggregation of amyloid ß in the brain. The three human apoE isoforms apoE2, apoE3, and apoE4 only differ in one or two residues. Nevertheless, the functions highly depend on the isoform types and lipidated states. Here, we generated novel anti-apoE monoclonal antibodies (mAbs) and obtained an apoE4-selective mAb whose epitope is within residues 110-117. ELISA and bio-layer interferometry measurements demonstrated that the dissociation constants of mAbs are within the nanomolar range. Using the generated antibodies, we successfully constructed sandwich ELISA systems, which can detect all apoE isoforms or selectively detect apoE4. These results suggest the usability of the generated anti-apoE mAbs for selective detection of apoE isoforms.


Sujet(s)
Anticorps monoclonaux , Apolipoprotéines E , Isoformes de protéines , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/composition chimique , Humains , Isoformes de protéines/immunologie , Apolipoprotéines E/métabolisme , Apolipoprotéines E/génétique , Apolipoprotéines E/composition chimique , Apolipoprotéines E/immunologie , Animaux , Épitopes/immunologie , Épitopes/composition chimique , Test ELISA/méthodes , Souris , Apolipoprotéine E4/génétique , Apolipoprotéine E4/immunologie , Apolipoprotéine E4/métabolisme , Souris de lignée BALB C , Apolipoprotéine E3/immunologie , Apolipoprotéine E3/génétique , Apolipoprotéine E3/composition chimique , Apolipoprotéine E3/métabolisme
8.
Mol Neurodegener ; 19(1): 24, 2024 Mar 11.
Article de Anglais | MEDLINE | ID: mdl-38468308

RÉSUMÉ

Microglia are highly dynamic cells that play a critical role in tissue homeostasis through the surveillance of brain parenchyma and response to cues associated with damage. Aging and APOE4 genotype are the strongest risk factors for Alzheimer's disease (AD), but how they affect microglial dynamics remains unclear. Using ex vivo confocal microscopy, we analyzed microglial dynamic behaviors in the entorhinal cortex (EC) and hippocampus CA1 of 6-, 12-, and 21-month-old mice APOE3 or APOE4 knock-in mice expressing GFP under the CX3CR1 promoter. To study microglia surveillance, we imaged microglia baseline motility for 20 min and measured the extension and retraction of processes. We found that APOE4 microglia exhibited significantly less brain surveillance (27%) compared to APOE3 microglia in 6-month-old mice; aging exacerbated this deficit. To measure microglia response to damage, we imaged process motility in response to ATP, an injury-associated signal, for 30 min. We found APOE4 microglia extended their processes significantly slower (0.9 µm/min, p < 0.005) than APOE3 microglia (1.1 µm/min) in 6-month-old animals. APOE-associated alterations in microglia motility were observed in 12- and 21-month-old animals, and this effect was exacerbated with aging in APOE4 microglia. We measured protein and mRNA levels of P2RY12, a core microglial receptor required for process movement in response to damage. We found that APOE4 microglia express significantly less P2RY12 receptors compared to APOE3 microglia despite no changes in P2RY12 transcripts. To examine if the effect of APOE4 on the microglial response to ATP also applied to amyloid ß (Aß), we infused locally Hi-Lyte Fluor 555-labeled Aß in acute brain slices of 6-month-old mice and imaged microglia movement for 2 h. APOE4 microglia showed a significantly slower (p < 0.0001) process movement toward the Aß, and less Aß coverage at early time points after Aß injection. To test whether P2RY12 is involved in process movement in response to Aß, we treated acute brain slices with a P2RY12 antagonist before Aß injection; microglial processes no longer migrated towards Aß. These results provide mechanistic insights into the impact of APOE4 genotype and aging in dynamic microglial behaviors prior to gross Aß pathology and could help explain how APOE4 brains are more susceptible to AD pathogenesis.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Animaux , Souris , Adénosine triphosphate/métabolisme , Adénosine triphosphate/pharmacologie , Maladie d'Alzheimer/métabolisme , Peptides bêta-amyloïdes/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Encéphale/métabolisme , Génotype , Souris transgéniques , Microglie/métabolisme
9.
J Alzheimers Dis ; 97(2): 753-775, 2024.
Article de Anglais | MEDLINE | ID: mdl-38217595

RÉSUMÉ

BACKGROUND: Apolipoprotein E4 (APOE4) is the most prevalent genetic risk factor of Alzheimer's disease. Several studies suggest that APOE4 binding to its receptors is associated with their internalization and accumulation in intracellular compartments. Importantly, this phenomenon also occurs with other, non-ApoE receptors. Based on these observations, we hypothesized that APOE4 pathological effects are mediated by impairment in the life cycle of distinct receptors (APOER2, LRP1, IR, VEGFR). OBJECTIVE: To examine the effects of APOE genotype on receptors protein levels and compartmentalization. METHODS: Primary mouse neurons were prepared from APOE3 or APOE4 targeted replacement mice, or APOE-KO mice. Specific receptors protein levels were evaluated in these neurons, utilizing immunofluorescent staining. Additionally, surface membrane protein levels of those receptors were assessed by cell surface biotinylation assay and ELISA. Receptors' colocalization with intracellular compartments was assessed by double staining and confocal microscopy, followed by colocalization analysis. Finally, LRP1 or APOER2 were knocked-down with CRISPR/Cas9 system to examine their role in mediating APOE4 effects on the receptors. RESULTS: Our results revealed lower receptors' levels in APOE4, specifically on the membrane surface. Additionally, APOE4 affects the compartmentation of these receptors in two patterns: the first was observed with LRP1 and was associated with decreased receptor levels in numerous intracellular compartments. The second was obtained with the other receptors and was associated with their accumulation in early endosomes and their decrease in the late endosomes. CONCLUSIONS: These results provide a unifying mechanism, in which APOE4 drives the down regulation of various receptors, which plays important roles in distinct APOE4 related pathological processes.


Sujet(s)
Maladie d'Alzheimer , Souris , Animaux , Maladie d'Alzheimer/anatomopathologie , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Souris transgéniques , Apolipoprotéines E , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme
10.
FEBS Lett ; 598(3): 347-362, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38279679

RÉSUMÉ

The low-density lipoprotein (LDL) receptor-related protein (LRP)1 participates in the metabolism of apolipoprotein (apo) E-containing lipoproteins (apoE-LP). We investigated the effects of modifications of cysteine (Cys)-thiol of apoE on LRP1-mediated metabolism. Among the three isoforms, apoE2-LP exhibited the lowest affinity for LRP1 but was significantly catabolized, whereas apoE4-LP was sufficiently bound to LRP1 but showed the lowest catabolic capability. The reduction enhanced the binding and suppressed the catabolism of apoE3-LP, but had no effect on apoE2-LP. The formation of disulfide-linked complexes with apoAII suppressed binding, but enhanced the catabolism of apoE2-LP. Redox modifications of apoE-Cys-thiol may modulate the LRP1-mediated metabolism of apoE2- or apoE3-LP, but not apoE4-LP. The failure of this function may be involved in the pathophysiology of dyslipidemia.


Sujet(s)
Apolipoprotéines E , Thiols , Apolipoprotéine E2/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Apolipoprotéines E/métabolisme , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Triglycéride/métabolisme , Isoformes de protéines/génétique , Isoformes de protéines/métabolisme , Protéines de transport
11.
Neuron ; 112(7): 1100-1109.e5, 2024 Apr 03.
Article de Anglais | MEDLINE | ID: mdl-38266643

RÉSUMÉ

The Apolipoprotein E gene (APOE) is of great interest due to its role as a risk factor for late-onset Alzheimer's disease. ApoE is secreted by astrocytes in the central nervous system in high-density lipoprotein (HDL)-like lipoproteins. Structural models of lipidated ApoE of high resolution could aid in a mechanistic understanding of how ApoE functions in health and disease. Using monoclonal Fab and F(ab')2 fragments, we characterize the structure of lipidated ApoE on astrocyte-secreted lipoproteins. Our results provide support for the "double-belt" model of ApoE in nascent discoidal HDL-like lipoproteins, where two ApoE proteins wrap around the nanodisc in an antiparallel conformation. We further show that lipidated, recombinant ApoE accurately models astrocyte-secreted ApoE lipoproteins. Cryogenic electron microscopy of recombinant lipidated ApoE further supports ApoE adopting antiparallel dimers in nascent discoidal lipoproteins.


Sujet(s)
Apolipoprotéines E , Astrocytes , Lipoprotéines , Astrocytes/métabolisme , Apolipoprotéines E/génétique , Lipoprotéines HDL/composition chimique , Lipoprotéines HDL/métabolisme , Système nerveux central/métabolisme , Apolipoprotéine E4/métabolisme , Apolipoprotéine E3/métabolisme
12.
Cell ; 187(2): 428-445.e20, 2024 01 18.
Article de Anglais | MEDLINE | ID: mdl-38086389

RÉSUMÉ

A recent case report described an individual who was a homozygous carrier of the APOE3 Christchurch (APOE3ch) mutation and resistant to autosomal dominant Alzheimer's Disease (AD) caused by a PSEN1-E280A mutation. Whether APOE3ch contributed to the protective effect remains unclear. We generated a humanized APOE3ch knock-in mouse and crossed it to an amyloid-ß (Aß) plaque-depositing model. We injected AD-tau brain extract to investigate tau seeding and spreading in the presence or absence of amyloid. Similar to the case report, APOE3ch expression resulted in peripheral dyslipidemia and a marked reduction in plaque-associated tau pathology. Additionally, we observed decreased amyloid response and enhanced microglial response around plaques. We also demonstrate increased myeloid cell phagocytosis and degradation of tau aggregates linked to weaker APOE3ch binding to heparin sulfate proteoglycans. APOE3ch influences the microglial response to Aß plaques, which suppresses Aß-induced tau seeding and spreading. The results reveal new possibilities to target Aß-induced tauopathy.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Apolipoprotéine E3 , Protéines tau , Animaux , Humains , Souris , Maladie d'Alzheimer/anatomopathologie , Peptides bêta-amyloïdes/métabolisme , Protéines amyloïdogènes/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Encéphale/métabolisme , Modèles animaux de maladie humaine , Souris transgéniques , Microglie/métabolisme , Plaque amyloïde/métabolisme , Protéines tau/génétique , Protéines tau/métabolisme , Présentations de cas cliniques comme sujet
13.
Alzheimers Dement ; 20(2): 819-836, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-37791598

RÉSUMÉ

INTRODUCTION: We discovered that the APOE3 Christchurch (APOE3Ch) variant may provide resistance to Alzheimer's disease (AD). This resistance may be due to reduced pathological interactions between ApoE3Ch and heparan sulfate proteoglycans (HSPGs). METHODS: We developed and characterized the binding, structure, and preclinical efficacy of novel antibodies targeting human ApoE-HSPG interactions. RESULTS: We found that one of these antibodies, called 7C11, preferentially bound ApoE4, a major risk factor for sporadic AD, and disrupts heparin-ApoE4 interactions. We also determined the crystal structure of a Fab fragment of 7C11 and used computer modeling to predict how it would bind to ApoE. When we tested 7C11 in mouse models, we found that it reduced recombinant ApoE-induced tau pathology in the retina of MAPT*P301S mice and curbed pTau S396 phosphorylation in brains of systemically treated APOE4 knock-in mice. Targeting ApoE-HSPG interactions using 7C11 antibody may be a promising approach to developing new therapies for AD.


Sujet(s)
Maladie d'Alzheimer , Apolipoprotéine E4 , Souris , Humains , Animaux , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Protéoglycanes à sulfate d'héparane/métabolisme , Phosphorylation , Apolipoprotéines E/métabolisme , Maladie d'Alzheimer/anatomopathologie , Facteurs immunologiques , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme
14.
Int J Biol Macromol ; 254(Pt 2): 127799, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37923037

RÉSUMÉ

A significant portion of brain-tumor patients suffer from 'brain-tumor-related epilepsy (BTE)' which results in depression, anxiety and hampered quality of life. Conventional anti-epileptic drugs indicate negative interaction with other drugs augmenting the poor outcome of overall therapy. Levetiracetam (LVM) has evidenced effectiveness for BTE but its hydrophilicity restricts the passage into blood-brain barrier. The majority of lipid nanoparticles fails to load hydrophilic drug sufficiently. Therefore, lipid-drug conjugates (LDC) were synthesized using stearic acid via amide bond formation confirmed by FTIR and NMR. The nanoparticles of synthesized LDC were prepared by solvent injection method followed by functionalization with Apolipoprotein E3 (ApoE3@LDC-NP). The nanoparticles were characterized by DSC, XRD, particle size (131.6 ± 1.24 nm), zeta potential (-15.6 ± 0.09 mV), and for storage stability. In-vitro release study indicated initial burst release of 20 ± 0.63 % followed by sustained release up to 30 h (66 ± 1.40 %) for ApoE3@LDC-NP. The cell-line study on HEK293 indicated no significant cytotoxic effect and greater cell uptake through U87MG cell line. The pharmacokinetic and bio-distribution study indicated 2.5-fold greater brain-targeting of ApoE3@LDC-NP as compared to LVM solution. It proved safe in the haemolysis study and exhibited the absence of tissue necrosis. Thus, ApoE3@LDC-NP might be a promising approach for effective brain-targeting of LVM for improved clinical response in BTE.


Sujet(s)
Tumeurs du cerveau , Nanoparticules , Humains , Apolipoprotéine E3/métabolisme , Lévétiracétam/pharmacologie , Lévétiracétam/métabolisme , Lévétiracétam/usage thérapeutique , Cellules HEK293 , Qualité de vie , Encéphale/métabolisme , Liposomes/métabolisme , Vecteurs de médicaments/composition chimique , Nanoparticules/composition chimique , Tumeurs du cerveau/traitement médicamenteux , Lignée cellulaire tumorale , Taille de particule , Systèmes de délivrance de médicaments
15.
Exp Neurol ; 371: 114609, 2024 01.
Article de Anglais | MEDLINE | ID: mdl-37944881

RÉSUMÉ

Many cancer patients experience serious cognitive problems related to their treatment, which can greatly affect their quality of life. The molecular mechanisms of this cancer chemotherapy-induced cognitive impairment (CICI) are unknown, thus slowing the development of preventative approaches. We hypothesized that cancer chemotherapies could induce cellular senescence in the brain, creating a pro-inflammatory environment and damaging normal brain communication. We tested this hypothesis using the common chemotherapeutic agent doxorubicin in two independent mouse models. In the first model, we used mice that express tdTomato under the pdkn2a (p16) promoter; p16 is a regulator of cellular senescence, and its upregulation is denoted by the presence of fluorescently tagged cells. Two weeks after exposure to three doses of 5 mg/kg doxorubicin, the number of tdTomato positive cells were increased nearly three-fold in both the cerebral cortex and the hippocampus. tdTomato staining co-localized with neurons, microglia, oligodendrocyte precursor cells, and endothelial cells, but not astrocytes. In the second model, we used APOE knock-in mice, since the APOE4 allele is a risk factor for CICI in humans and mouse models. We isolated RNA from the cerebral cortex of APOE3 and APOE4 mice from one to 21 days after a single dose of 10 mg/kg doxorubicin. Using NanoString analysis of over 700 genes related to neuroinflammation and RT-qPCR analysis of cerebral cortex transcripts, we found two-fold induction of four senescence-related genes at three weeks in the APOE4 mice compared to the APOE3 control mice: p21(cdkn1a), p16, Gadd45a, and Egr1. We conclude that doxorubicin promotes cellular senescence pathways in the brain, supporting the hypothesis that drugs to eliminate senescent cells could be useful in preventing CICI.


Sujet(s)
Maladie d'Alzheimer , Tumeurs , Humains , Souris , Animaux , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Souris transgéniques , Cellules endothéliales/métabolisme , Qualité de vie , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Encéphale/métabolisme , Doxorubicine/toxicité , Génotype , Maladie d'Alzheimer/métabolisme
16.
AAPS PharmSciTech ; 24(8): 223, 2023 Nov 10.
Article de Anglais | MEDLINE | ID: mdl-37945928

RÉSUMÉ

Rivastigmine hydrogen tartrate (RHT) is an acetylcholinesterase (AChE) inhibitor used in the management of Alzheimer's disease (AD). RHT is a BCS class-I drug that undergoes significant first-pass metabolism. Permeating a hydrophilic drug through the brain remains a major challenge in brain delivery. In this study, the RHT was incorporated inside the hydrophilic core of liposomes (LPS) and then coated with the ApoE3. ApoE3-coated RHT-loaded liposomes (ApoE3-RHT-LPS) were fabricated through the thin film hydration method using DSPE-PEG. The coating of LPS with ApoE3 enhances brain uptake and improves Aß clearance. The results obtained from the physicochemical characterization demonstrated that ApoE3-RHT-LPS shows a particle size of 128.6 ± 2.16 nm and a zeta potential of 16.6 ± 1.19. The % entrapment efficiency and % drug loading were found to be 75% and 17.84%, respectively. The data obtained from TEM and SEM studies revealed that the particle size of the LPS was less than 200 nm. An in vitro AChE assay was performed, and the results demonstrated the AChE inhibitory potential of ApoE3-RHT-LPS. Through the intravenous route, an in vivo pharmacokinetic study of formulation displayed improved brain uptake of RHT by ~ 1.3-fold than pure RHT due to ApoE3 coating. In vivo, biodistribution studies in vital organs suggested that the biodistribution of RHT to the liver was significantly reduced (p < 0.001), signifying an increase in the drug's half-life and blood circulation time. All research findings suggested that ApoE3 coating and LPS strategy are proven effective for improving the brain uptake of RHT designed for the management of AD.


Sujet(s)
Maladie d'Alzheimer , Liposomes , Humains , Rivastigmine , Liposomes/composition chimique , Apolipoprotéine E3/métabolisme , Apolipoprotéine E3/pharmacologie , Acetylcholinesterase/métabolisme , Acetylcholinesterase/pharmacologie , Acetylcholinesterase/usage thérapeutique , Distribution tissulaire , Lipopolysaccharides , Encéphale/métabolisme , Anticholinestérasiques , Maladie d'Alzheimer/traitement médicamenteux , Maladie d'Alzheimer/métabolisme , Taille de particule
17.
Cell Rep ; 42(10): 113183, 2023 10 31.
Article de Anglais | MEDLINE | ID: mdl-37777962

RÉSUMÉ

Recent developments in genome sequencing have expanded the knowledge of genetic factors associated with late-onset Alzheimer's disease (AD). Among them, genetic variant ε4 of the APOE gene (APOE4) confers the greatest disease risk. Dysregulated glucose metabolism is an early pathological feature of AD. Using isogenic ApoE3 and ApoE4 astrocytes derived from human induced pluripotent stem cells, we find that ApoE4 increases glycolytic activity but impairs mitochondrial respiration in astrocytes. Ultrastructural and autophagy flux analyses show that ApoE4-induced cholesterol accumulation impairs lysosome-dependent removal of damaged mitochondria. Acute treatment with cholesterol-depleting agents restores autophagic activity, mitochondrial dynamics, and associated proteomes, and extended treatment rescues mitochondrial respiration in ApoE4 astrocytes. Taken together, our study provides a direct link between ApoE4-induced lysosomal cholesterol accumulation and abnormal oxidative phosphorylation.


Sujet(s)
Maladie d'Alzheimer , Cellules souches pluripotentes induites , Humains , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Astrocytes/métabolisme , Phosphorylation oxydative , Cellules cultivées , Cellules souches pluripotentes induites/métabolisme , Apolipoprotéine E3/métabolisme , Cholestérol/métabolisme , Maladie d'Alzheimer/métabolisme , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme
18.
Nat Immunol ; 24(11): 1854-1866, 2023 Nov.
Article de Anglais | MEDLINE | ID: mdl-37857825

RÉSUMÉ

Microglial involvement in Alzheimer's disease (AD) pathology has emerged as a risk-determining pathogenic event. While apolipoprotein E (APOE) is known to modify AD risk, it remains unclear how microglial apoE impacts brain cognition and AD pathology. Here, using conditional mouse models expressing apoE isoforms in microglia and central nervous system-associated macrophages (CAMs), we demonstrate a cell-autonomous effect of apoE3-mediated microglial activation and function, which are negated by apoE4. Expression of apoE3 in microglia/CAMs improves cognitive function, increases microglia surrounding amyloid plaque and reduces amyloid pathology and associated toxicity, whereas apoE4 expression either compromises or has no effects on these outcomes by impairing lipid metabolism. Single-cell transcriptomic profiling reveals increased antigen presentation and interferon pathways upon apoE3 expression. In contrast, apoE4 expression downregulates complement and lysosomal pathways, and promotes stress-related responses. Moreover, in the presence of mouse endogenous apoE, microglial apoE4 exacerbates amyloid pathology. Finally, we observed a reduction in Lgals3-positive responsive microglia surrounding amyloid plaque and an increased accumulation of lipid droplets in APOE4 human brains and induced pluripotent stem cell-derived microglia. Our findings establish critical isoform-dependent effects of microglia/CAM-expressed apoE in brain function and the development of amyloid pathology, providing new insight into how apoE4 vastly increases AD risk.


Sujet(s)
Maladie d'Alzheimer , Souris , Animaux , Humains , Maladie d'Alzheimer/génétique , Maladie d'Alzheimer/anatomopathologie , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme , Microglie/métabolisme , Apolipoprotéine E3/génétique , Apolipoprotéine E3/métabolisme , Plaque amyloïde/métabolisme , Plaque amyloïde/anatomopathologie , Apolipoprotéines E/génétique , Apolipoprotéines E/métabolisme , Encéphale , Homéostasie , Souris transgéniques
19.
J Lipid Res ; 64(9): 100425, 2023 09.
Article de Anglais | MEDLINE | ID: mdl-37579971

RÉSUMÉ

Plasma levels of HDL cholesterol are inversely associated with CVD progression. It is becoming increasingly clear that HDL plays important roles in immunity that go beyond its traditionally understood roles in lipid transport. We previously reported that HDL interaction with regulatory T cells (Treg) protected them from apoptosis, which could be a mechanism underlying the broad anti-inflammatory effect of HDL. Herein, we extend our work to show that HDL interacts mainly with memory Treg, particularly with the highly suppressive effector memory Treg, by limiting caspase-dependent apoptosis in an Akt-dependent manner. Reconstitution experiments identified the protein component of HDL as the primary driver of the effect, though the most abundant HDL protein, apolipoprotein A-I (APOA1), was inactive. In contrast, APOE-depleted HDL failed to rescue effector memory Treg, suggesting the critical role of APOE proteins. HDL particles reconstituted with APOE, and synthetic phospholipids blunted Treg apoptosis at physiological concentrations. The APOE3 and APOE4 isoforms were the most efficient. Similar results were obtained when lipid-free recombinant APOEs were tested. Binding experiments showed that lipid-free APOE3 bound to memory Treg but not to naive Treg. Overall, our results show that APOE interaction with Treg results in blunted caspase-dependent apoptosis and increased survival. As dysregulation of HDL-APOE levels has been reported in CVD and obesity, our data bring new insight on how this defect may contribute to these diseases.


Sujet(s)
Maladies cardiovasculaires , Lymphocytes T régulateurs , Humains , Lymphocytes T régulateurs/métabolisme , Apolipoprotéine E3/métabolisme , Apolipoprotéines E/métabolisme , Apolipoprotéine E4/génétique , Apolipoprotéine E4/métabolisme
20.
Arterioscler Thromb Vasc Biol ; 43(10): 1952-1966, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37650329

RÉSUMÉ

BACKGROUND: Specialized brain endothelial cells and human APOE3 are independently important for neurovascular function, yet whether APOE3 expression by endothelial cells contributes to brain function is currently unknown. In the present study, we determined whether the loss of endothelial cell APOE3 impacts brain vascular and neural function. METHODS: We developed APOE3fl/fl/Cdh5(PAC)-CreERT2+/- (APOE3Cre+/-) and APOE3fl/fl/Cdh5(PAC)-CreERT2-/- (APOE3Cre-/-, control) mice and induced endothelial cell APOE3 knockdown with tamoxifen at ≈4 to 5 weeks of age. Neurovascular and neuronal function were evaluated by biochemistry, immunohistochemistry, behavioral testing, and electrophysiology at 9 months of age. RESULTS: We found that the loss of endothelial APOE3 expression was sufficient to cause neurovascular dysfunction including higher permeability and lower vessel coverage in tandem with deficits in spatial memory and fear memory extinction and a disruption of cortical excitatory/inhibitory balance. CONCLUSIONS: Our data collectively support the novel concept that endothelial APOE3 plays a critical role in the regulation of the neurovasculature, neural circuit function, and behavior.


Sujet(s)
Encéphale , Cellules endothéliales , Souris , Humains , Animaux , Apolipoprotéine E3/métabolisme , Cellules endothéliales/métabolisme , Encéphale/métabolisme , Apolipoprotéine E4
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...