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










Publication year range
1.
Sci Rep ; 10(1): 8670, 2020 05 26.
Article in English | MEDLINE | ID: mdl-32457323

ABSTRACT

Tumor necrosis factor-α (TNF-α) is a pro-inflammatory cytokine, involved in Alzheimer's disease pathogenesis. Anti-TNF-α therapeutic approaches currently used in autoimmune diseases have been proposed as a therapeutic strategy in AD. We have previously examined the role of TNF-α and anti-TNF-α drugs in AD, using 5XFAD mice, and we have found a significant role for peripheral TNF-α in brain inflammation. Here we investigated the role of mouse TNF-α on the AD-like phenotype of 5XFAD mice using a knock-in mouse with deletion of the 3'UTR of the endogenous TNF-α (TNFΔARE/+) that develops rheumatoid arthritis and Crohn's disease. 5XFAD/TNFΔARE/+ mice showed significantly decreased amyloid deposition. Interestingly, microglia but not astrocytes were activated in 5XFAD/ TNFΔARE/+ brains. This microglial activation was associated with increased infiltrating peripheral leukocytes and perivascular macrophages and synaptic degeneration. APP levels and APP processing enzymes involved in Aß production remained unchanged, suggesting that the reduced amyloid burden can be attributed to the increased microglial and perivascular macrophage activation caused by TNF-α. Peripheral TNF-α levels were increased while brain TNF-α remained the same. These data provide further evidence for peripheral TNF-α as a mediator of inflammation between the periphery and the brain.


Subject(s)
3' Untranslated Regions/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Plaque, Amyloid/pathology , Tumor Necrosis Factor-alpha/genetics , Alzheimer Disease/genetics , Animals , Arthritis, Rheumatoid/genetics , Brain/pathology , Crohn Disease/genetics , Disease Models, Animal , Female , Gene Knock-In Techniques , Macrophage Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Curr Alzheimer Res ; 16(6): 559-574, 2019.
Article in English | MEDLINE | ID: mdl-30907316

ABSTRACT

After years of failed therapeutic attempts targeting beta-amyloid (Aß) in AD, there is now increasing evidence suggesting that inflammation holds a pivotal role in AD pathogenesis and immune pathways can possibly comprise primary therapeutic targets. Inflammation is a key characteristic of numerous diseases including neurodegenerative disorders and thus not surprisingly suppression of inflammation frequently constitutes a major therapeutic strategy for a wide spectrum of disorders. Several brain-resident and peripherally-derived immune populations and inflammatory mediators are involved in AD pathophysiology, with microglia comprising central cellular player in the disease process. Systemic inflammation, mostly in the form of infections, has long been observed to induce behavioral alterations and cognitive dysfunction, suggesting for a close interaction of the peripheral immune system with the brain. Systemic inflammation can result in neuroinflammation, mainly exhibited as microglial activation, production of inflammatory molecules, as well as recruitment of peripheral immune cells in the brain, thus shaping a cerebral inflammatory milieu that may seriously impact neuronal function. Increasing clinical and experimental studies have provided significant evidence that acute (e.g. infections) or chronic (e.g. autoimmune diseases like rheumatoid arthritis) systemic inflammatory conditions may be associated with increased AD risk and accelerate AD progression. Here we review the current literature that links systemic with CNS inflammation and the implications of this interaction for AD in the context of acute and chronic systemic pathologies as acute infection and rheumatoid arthritis. Elucidating the mechanisms that govern the crosstalk between the peripheral and the local brain immune system may provide the ground for new therapeutic approaches that target the immune-brain interface and shed light on the understanding of AD.


Subject(s)
Alzheimer Disease/immunology , Alzheimer Disease/physiopathology , Inflammation/immunology , Inflammation/physiopathology , Neuroimmunomodulation/physiology , Animals , Humans
3.
J Alzheimers Dis ; 60(1): 165-181, 2017.
Article in English | MEDLINE | ID: mdl-28826177

ABSTRACT

Increasing evidence suggests that neuroinflammation comprises a major characteristic of Alzheimer's disease (AD). Tumor necrosis factor-α (TNF-α) is a pleiotropic pro-inflammatory cytokine implicated in neurodegenerative diseases including AD, and has been proposed as a potent therapeutic target for AD. Although a number of studies focusing on pharmacological or genetic manipulation of TNF-α and its receptors in AD mice have provided significant knowledge regarding the role of TNF-α signaling pathway in the pathogenesis of AD, the consequences of TNF-α genetic deletion have not been thoroughly examined. Here, we focused on the effect of TNF-α deficiency on the amyloid phenotype of 5XFAD mice. Our analysis revealed that amyloid deposition, amyloid-ß (Aß) levels, and AßPP-carboxyterminal fragments are significantly reduced in the brains of 5XFAD/TNF-α-/- mice compared to the 5XFAD/TNF-α+/+. We found decreased protein levels of ß- and α-secretases in the 5XFAD/TNF-α-/- brains, suggesting for an effect of TNF-α on AßPP processing and Aß generation. We also show for the first time that TNF-α affects PS1in vivo, as 5XFAD mice lacking TNF-α expression display reduced PS1-carboxyterminal fragments implying for diminished PS1 activity. Moreover, TNF-α deficiency decreases microglial and astrocytic activation and significantly restricts the phagocytic activity of macrophages against Aß, supporting for reduced responsiveness of phagocytes toward Aß. Overall, our results reveal that TNF-α genetic deletion in 5XFAD mice attenuates amyloid plaque formation by lowering Aß generation through the reduction of functionally active PS1 and ß-secretase rather than promoting Aß clearance by phagocytic cells. Our data further suggest TNF-α inhibition as a therapeutic approach for AD.


Subject(s)
Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Gene Expression Regulation/genetics , Neuroglia/metabolism , Tumor Necrosis Factor-alpha/genetics , ADAM10 Protein/metabolism , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/genetics , Amyloid Precursor Protein Secretases/metabolism , Animals , Aspartic Acid Endopeptidases/genetics , Aspartic Acid Endopeptidases/metabolism , Brain/pathology , CD11b Antigen/metabolism , Calcium-Binding Proteins/metabolism , Cells, Cultured , Disease Models, Animal , Glial Fibrillary Acidic Protein/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism , Tumor Necrosis Factor-alpha/metabolism
4.
J Neurosci ; 37(20): 5155-5171, 2017 05 17.
Article in English | MEDLINE | ID: mdl-28442538

ABSTRACT

Increasing evidence has suggested that systemic inflammation along with local brain inflammation can play a significant role in Alzheimer's disease (AD) pathogenesis. Identifying key molecules that regulate the crosstalk between the immune and the CNS can provide potential therapeutic targets. TNF-α is a proinflammatory cytokine implicated in the pathogenesis of systemic inflammatory and neurodegenerative diseases, such as rheumatoid arthritis (RA) and AD. Recent studies have reported that anti-TNF-α therapy or RA itself can modulate AD pathology, although the underlying mechanism is unclear. To investigate the role of peripheral TNF-α as a mediator of RA in the pathogenesis of AD, we generated double-transgenic 5XFAD/Tg197 AD/TNF mice that develop amyloid deposits and inflammatory arthritis induced by human TNF-α (huTNF-α) expression. We found that 5XFAD/Tg197 mice display decreased amyloid deposition, compromised neuronal integrity, and robust brain inflammation characterized by extensive gliosis and elevated blood-derived immune cell populations, including phagocytic macrophages and microglia. To evaluate the contribution of peripheral huTNF-α in the observed brain phenotype, we treated 5XFAD/Tg197 mice systemically with infliximab, an anti-huTNF-α antibody that does not penetrate the blood-brain barrier and prevents arthritis. Peripheral inhibition of huTNF-α increases amyloid deposition, rescues neuronal impairment, and suppresses gliosis and recruitment of blood-derived immune cells, without affecting brain huTNF-α levels. Our data report, for the first time, a distinctive role for peripheral TNF-α in the modulation of the amyloid phenotype in mice by regulating blood-derived and local brain inflammatory cell populations involved in ß-amyloid clearance.SIGNIFICANCE STATEMENT Mounting evidence supports the active involvement of systemic inflammation, in addition to local brain inflammation, in Alzheimer's disease (AD) progression. TNF-α is a pluripotent cytokine that has been independently involved in the pathogenesis of systemic inflammatory rheumatoid arthritis (RA) and AD. Here we first demonstrate that manipulation of peripheral TNF-α in the context of arthritis modulates the amyloid phenotype by regulating immune cell trafficking in the mouse brain. Our study suggests that additionally to its local actions in the AD brain, TNF-α can also indirectly modulate amyloid pathology as a regulator of peripheral inflammation. Our findings may have significant implications in the treatment of RA patients with anti-TNF-α drugs and in the potential use of TNF-targeted therapies for AD.


Subject(s)
Alzheimer Disease/immunology , Amyloidogenic Proteins/immunology , Arthritis, Rheumatoid/immunology , Brain/immunology , Macrophages/immunology , Neuroglia/immunology , Tumor Necrosis Factor-alpha/immunology , Alzheimer Disease/complications , Alzheimer Disease/pathology , Animals , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/pathology , Brain/pathology , Cytokines/immunology , Female , Immunologic Factors/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuroglia/pathology
5.
J Cell Sci ; 129(20): 3705-3720, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27566162

ABSTRACT

The association of desmin with the α-crystallin Β-chain (αΒ-crystallin; encoded by CRYAB), and the fact that mutations in either one of them leads to heart failure in humans and mice, suggests a potential compensatory interplay between the two in cardioprotection. To address this hypothesis, we investigated the consequences of αΒ-crystallin overexpression in the desmin-deficient (Des-/-) mouse model, which possesses a combination of the pathologies found in most cardiomyopathies, with mitochondrial defects as a hallmark. We demonstrated that cardiac-specific αΒ-crystallin overexpression ameliorates all these defects and improves cardiac function to almost wild-type levels. Protection by αΒ-crystallin overexpression is linked to maintenance of proper mitochondrial protein levels, inhibition of abnormal mitochondrial permeability transition pore activation and maintenance of mitochondrial membrane potential (Δψm). Furthermore, we found that both desmin and αΒ-crystallin are localized at sarcoplasmic reticulum (SR)-mitochondria-associated membranes (MAMs), where they interact with VDAC, Mic60 - the core component of mitochondrial contact site and cristae organizing system (MICOS) complex - and ATP synthase, suggesting that these associations could be crucial in mitoprotection at different levels.


Subject(s)
Desmin/metabolism , Homeostasis , Mitochondria/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , alpha-Crystallin B Chain/metabolism , Animals , Antioxidants/pharmacology , Cell Survival/drug effects , Desmin/deficiency , Heart Function Tests , Homeostasis/drug effects , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Mitochondrial Permeability Transition Pore , Mitochondrial Proteins/metabolism , Myocardium/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Physical Conditioning, Animal , Sarcoplasmic Reticulum/drug effects , Sarcoplasmic Reticulum/metabolism , Stress, Physiological/drug effects , Swimming , Voltage-Dependent Anion Channels/metabolism
6.
Mech Ageing Dev ; 160: 34-40, 2016 12.
Article in English | MEDLINE | ID: mdl-27443148

ABSTRACT

Human life expectancy has increased dramatically in the last century and as a result also the prevalence of a variety of age-related diseases and syndromes. One such syndrome is frailty, which is defined as a combination of organ dysfunctions leading to increased vulnerability to adverse health outcomes. In humans, frailty is associated with various biomarkers of ageing and predicts relevant outcomes such as responses to therapies and progression of health status and mortality. Moreover, it is relatively easy to assess. To foster translation of mechanistic understanding of the ageing process and, importantly, of interventions that may extend healthy lifespan, frailty scales have been reverse translated into mice in recent years. We will review these approaches with a view to identify what is known and what is not known at present about their validity, reproducibility and reliability with a focus on the potential for further improvement.


Subject(s)
Aging , Frailty , Aging/genetics , Aging/metabolism , Aging/pathology , Animals , Disease Models, Animal , Frailty/genetics , Frailty/metabolism , Frailty/pathology , Humans , Mice
7.
Atherosclerosis ; 243(1): 77-85, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26363436

ABSTRACT

OBJECTIVE: Mutations in human apolipoprotein A-I (apoA-I) are associated with low high-density lipoprotein (HDL) cholesterol levels and pathological conditions such as premature atherosclerosis and amyloidosis. In this study we functionally characterized two natural human apoA-I mutations, L141RPisa and L159RFIN, in vivo. METHODS: We generated transgenic mice expressing either wild-type (WT) or the two mutant forms of human apoA-I on a mouse apoA-I(-/-) background and analyzed for abnormalities in their lipid and lipoprotein profiles. HDL structure and functionality, as well as atherosclerosis development following a 14-week high-fat diet were assessed in these mice. RESULTS: The expression of either apoA-I mutant was associated with markedly reduced serum apoA-I (<10% of WT apoA-I), total and HDL-cholesterol levels (∼20% and ∼7% of WT apoA-I, respectively) and the formation of few small size HDL particles with preß2 and α3, α4 electrophoretic mobility. HDL particles containing either of the two apoA-I mutants exhibited attenuated anti-oxidative properties as indicated by their inability to prevent low-density lipoprotein oxidation, and by decreased activities of paraoxonase-1 and platelet-activating factor acetylhydrolase. However, the apoA-I(L141R)Pisa or apoA-I(L159R)FIN-containing HDL particles demonstrated increased capacity to promote ATP-Binding Cassette Transporter A1-mediated cholesterol efflux from macrophages. Expression of apoA-I(L141R)Pisa or apoA-I(L159R)FIN mutations in mice was associated with increased diet-induced atherosclerosis compared to either WT apoA-I transgenic or apoA-I(-/-) mice. CONCLUSIONS: These findings suggest that natural apoA-I mutations L141RPisa and L159RFIN affect the biogenesis and the functionality of HDL in vivo and predispose to diet-induced atherosclerosis in the absence of any other genetic defect.


Subject(s)
Apolipoprotein A-I/genetics , Atherosclerosis/blood , Lipoproteins, HDL/blood , Lipoproteins, LDL/genetics , Mutation , ATP Binding Cassette Transporter 1/metabolism , Animal Feed , Animals , Antioxidants/chemistry , Aryldialkylphosphatase/metabolism , Electrophoresis, Gel, Two-Dimensional , Humans , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Platelet Activating Factor/metabolism
8.
Aging Cell ; 12(6): 1032-40, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23826707

ABSTRACT

Production of Aß by γ-secretase is a key event in Alzheimer's disease (AD). The γ-secretase complex consists of presenilin (PS) 1 or 2, nicastrin (ncstn), Pen-2, and Aph-1 and cleaves type I transmembrane proteins, including the amyloid precursor protein (APP). Although ncstn is widely accepted as an essential component of the complex required for γ-secretase activity, recent in vitro studies have suggested that ncstn is dispensable for APP processing and Aß production. The focus of this study was to answer this controversy and evaluate the role of ncstn in Aß generation and the development of the amyloid-related phenotype in the mouse brain. To eliminate ncstn expression in the mouse brain, we used a ncstn conditional knockout mouse that we mated with an established AD transgenic mouse model (5XFAD) and a neuronal Cre-expressing transgenic mouse (CamKIIα-iCre), to generate AD mice (5XFAD/CamKIIα-iCre/ncstn(f/f) mice) where ncstn was conditionally inactivated in the brain. 5XFAD/CamKIIα-iCre/ncstn(f/f) mice at 10 week of age developed a neurodegenerative phenotype with a significant reduction in Aß production and formation of Aß aggregates and the absence of amyloid plaques. Inactivation of nctsn resulted in substantial accumulation of APP-CTFs and altered PS1 expression. These results reveal a key role for ncstn in modulating Aß production and amyloid plaque formation in vivo and suggest ncstn as a target in AD therapeutics.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/metabolism , Amyloid/metabolism , Membrane Glycoproteins/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Benzothiazoles , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Disease Models, Animal , Gliosis/metabolism , Gliosis/pathology , Humans , Integrases/metabolism , Mice , Mice, Transgenic , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Neurons/metabolism , Neurons/pathology , Phenotype , Presenilin-1/metabolism , Thiazoles/metabolism
9.
Epilepsia ; 53 Suppl 6: 45-52, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23134495

ABSTRACT

Proper function of the neurovasculature is required for optimal brain function and preventing neuroinflammation and neurodegeneration. Within this review, we discuss alterations of the function of the blood-brain barrier in neurologic disorders such as multiple sclerosis, epilepsy, and Alzheimer's disease and address potential underlying mechanisms.


Subject(s)
Blood-Brain Barrier/physiopathology , Brain/pathology , Brain/physiopathology , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/physiopathology , Animals , Humans
10.
J Neurochem ; 121(2): 239-51, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22192143

ABSTRACT

Evidence accumulating during the past few years points to a significant role of matrix metalloproteinase 9 (MMP9) enzymatic activity in synaptic plasticity and cognitive processes. We have previously demonstrated that MMP9 is involved in receptor-mediated α-secretase-like cleavage of APP in vitro, resulting in increased secretion of sAPPα, the soluble N-terminal product of the non-amyloidogenic pathway known to be involved in neuronal plasticity and memory formation. To study the in vivo role of MMP9, we have generated transgenic mice over-expressing MMP9 in the brain. Herein, we demonstrate that MMP9 transgenic animals display enhanced performance in the non-spatial novel object recognition and the spatial water-maze task and that their enhanced performance was accompanied by increased dendritic spine density in the hippocampus and cortex following behavioural testing. Consistent with the above observations, the electrophysiological analysis revealed prolonged maintenance of long-term synaptic potentiation in hippocampal slices from MMP9 transgenic mice. Moreover, elevated sAPPα levels in the hippocampus and cortex of MPP9 transgenic animals were also observed. Overall, our results extend previous findings on the physiological role of MMP9 in neuronal plasticity and furthermore reveal that, APP may be one of the physiological proteolytic targets of MMP9 in vivo.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/physiology , Neuronal Plasticity/genetics , Neuronal Plasticity/physiology , Peptide Fragments/metabolism , Animals , Blotting, Western , Brain/enzymology , Brain/physiology , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Cognition/physiology , DNA/genetics , Dendritic Spines/physiology , Electrophysiological Phenomena , Exploratory Behavior/physiology , Female , Fluorescent Antibody Technique , Hippocampus/cytology , Hippocampus/physiology , Humans , Long-Term Potentiation/genetics , Long-Term Potentiation/physiology , Male , Maze Learning/physiology , Mice , Mice, Transgenic , Psychomotor Performance/physiology , Real-Time Polymerase Chain Reaction , Receptor, Platelet-Derived Growth Factor beta/genetics , Recognition, Psychology/physiology
11.
PLoS One ; 6(7): e21880, 2011.
Article in English | MEDLINE | ID: mdl-21755005

ABSTRACT

BACKGROUND: Apolipoprotein E (ApoE), a cholesterol carrier associated with atherosclerosis, is a major risk factor for Alzheimer's disease (AD). The low-density lipoprotein receptor (LDLR) regulates ApoE levels in the periphery and in the central nervous system. LDLR has been identified on astrocytes and a number of studies show that it modulates amyloid deposition in AD transgenic mice. However these findings are controversial on whether LDLR deletion is beneficial or detrimental on the AD-like phenotype of the transgenic mice. METHODOLOGY/PRINCIPAL FINDINGS: To investigate the role of LDLR in the development of the amyloid related phenotype we used an APP/PS1 transgenic mouse (5XFAD) that develops an AD-like pathology with amyloid plaques, astrocytosis and microgliosis. We found that 4 months old 5XFAD transgenic mice on the LDLR deficient background (LDLR-/-) have increased amyloid plaque deposition. This increase is associated with a significant decrease in astrocytosis and microgliosis in the 5XFAD/LDLR-/- mice. To further elucidate the role of LDLR in relation with ApoE we have generated 5XFAD transgenic mice on the ApoE deficient (ApoE-/-) or the ApoE/LDLR double deficient background (ApoE-/-/LDLR -/-). We have found that ApoE deletion in the 4 months old 5XFAD/ApoE-/- mice decreases amyloid plaque formation as expected, but has no effect on astrocytosis or microgliosis. By comparison 5XFAD/ApoE-/-LDLR -/- double deficient mice of the same age have increased amyloid deposition with decreased astrocytosis and microgliosis. CONCLUSIONS: Our analysis shows that LDL deficiency regulates astrocytosis and microgliosis in an AD mouse model. This effect is independent of ApoE, as both 5XFAD/LDLR -/- and 5XFAD/ApoE-/- LDLR -/- mice show reduction in inflammatory response and increase in amyloid deposition compared to control mice. These results demonstrate that LDLR regulates glial response in this mouse model independently of ApoE and modifies amyloid deposition.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Receptors, LDL/deficiency , Alzheimer Disease/complications , Amyloid beta-Peptides/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Astrocytes/metabolism , Astrocytes/pathology , Benzothiazoles , Disease Models, Animal , Hippocampus/metabolism , Hippocampus/pathology , Humans , Mice , Mice, Transgenic , Microglia/metabolism , Microglia/pathology , Plaque, Amyloid/complications , Plaque, Amyloid/pathology , Protein Processing, Post-Translational , Receptors, LDL/metabolism , Thiazoles/metabolism
12.
Atherosclerosis ; 214(2): 448-55, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21130455

ABSTRACT

Secretion of 27-hydroxycholesterol (27OHC) from macrophages is considered as an alternative to HDL-mediated reverse transport of excess cholesterol. We investigated 27OHC-concentrations in plasma of humans and mice with monogenic disorders of HDL metabolism. As compared to family controls mutations in the genes for apolipoprotein A-I, ATP binding cassette transporter (ABC) A1 and lecithin:cholesterol acylstransferase (LCAT) were associated with reduced concentrations of both HDL-cholesterol and HDL-27OHC whereas mutations in the genes for cholesterylester transfer protein (CETP), scavenger receptor type BI and hepatic lipase were associated with elevated HDL concentrations of either sterol. Compared to family controls and relative to the concentrations of total 27OHC and cholesterol, lower 27OHC-ester but normal cholesterylester levels were found in HDL of heterozygous LCAT mutation carriers and nonHDL of heterozygous CETP mutation carriers. In family controls, LCAT activity and CETP mass were more strongly correlated with 27OHC-ester than cholesterylester concentrations in HDL and nonHDL, respectively. These findings suggest that the formation and transfer of 27OHC-esters are more sensitive to reduced activities of LCAT and CETP, respectively, than the formation and transfer of cholesterylesters. 27OHC plasma levels were also decreased in apoA-I-, ABCA1- or LCAT-knockout mice but increased in SR-BI-knockout mice. Transplantation of ABCA1- and/or ABCG1-deficient bone marrow into LDL receptor deficient mice decreased plasma levels of 27OHC. In conclusion, mutations or absence of HDL genes lead to distinct alterations in the quantity, esterification or lipoprotein distribution of 27OHC. These findings argue against the earlier suggestion that 27OHC-metabolism in plasma occurs independently of HDL.


Subject(s)
Cholesterol, HDL/blood , Hydroxycholesterols/blood , Lipid Metabolism, Inborn Errors/blood , Mutation , ATP Binding Cassette Transporter 1 , ATP Binding Cassette Transporter, Subfamily G, Member 1 , ATP-Binding Cassette Transporters/genetics , Adolescent , Adult , Aged , Animals , Apolipoprotein A-I/genetics , Biomarkers/blood , Case-Control Studies , Cholesterol Ester Transfer Proteins/genetics , Female , Genotype , Humans , Lipase/genetics , Lipid Metabolism, Inborn Errors/genetics , Lipoproteins/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Middle Aged , Phenotype , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Scavenger Receptors, Class B/genetics , Young Adult
13.
Proc Natl Acad Sci U S A ; 107(48): 20816-21, 2010 Nov 30.
Article in English | MEDLINE | ID: mdl-21076037

ABSTRACT

Scavenger receptor class B type I (SR-BI) is a high-density lipoprotein receptor that regulates cholesterol efflux from the peripheral tissues to the liver. SR-BI has been identified on astrocytes and vascular smooth muscle cells in Alzheimer's disease brain and has been shown to mediate adhesion of microglia to fibrillar amyloid-ß (Aß). Here we report that SR-BI mediates perivascular macrophage response and regulates Aß-related pathology and cerebral amyloid angiopathy in an Alzheimer's mouse model. Reduction or deletion of SR-BI gene in heterozygous or homozygous deficient mice (SR-BI(+/-), (-/-)) resulted in a significant increase in perivascular macrophages in the brain. SR-BI deletion had no effect on apolipoprotein E or apolipoprotein AI levels in the mouse brain. Our analysis revealed increased levels of SR-BI expression in the brains of human amyloid precursor protein (Swedish, Indiana) transgenic mice (J20 line). To evaluate the role of SR-BI in Alzheimer's disease pathogenesis, we inactivated one SR-BI allele in J20 transgenic mice. SR-BI reduction in J20/SR-BI(+/-) mice enhanced fibrillar amyloid deposition and cerebral amyloid angiopathy and also exacerbated learning and memory deficits compared with J20 littermates. Immunohistochemical analysis revealed localization of SR-BI on perivascular macrophages in tight association with Aß deposits. Our data suggest that SR-BI reduction impairs the response of perivascular macrophages to Aß and enhances the Aß-related phenotype and cerebral amyloid angiopathy in J20 mice. These results reveal that SR-BI, a scavenger receptor primarily involved in high-density lipoprotein cholesterol transport, plays an essential role in Alzheimer's disease and cerebral amyloid angiopathy.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid/metabolism , Blood Vessels/metabolism , Brain/blood supply , Macrophages/metabolism , Scavenger Receptors, Class B/metabolism , Amyloid beta-Protein Precursor/metabolism , Animals , Apolipoprotein A-I/metabolism , Apolipoproteins E/metabolism , Astrocytes/metabolism , Behavior, Animal , Blood Vessels/pathology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Humans , Mice , Microglia/metabolism , Oxidative Stress , Phagocytosis , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Protein Processing, Post-Translational , Protein Transport
14.
Endocrinology ; 150(10): 4521-30, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19628574

ABSTRACT

A low high-density lipoprotein (HDL) plasma concentration and the abundance of small dense low-density lipoproteins (LDL) are risk factors for developing type 2 diabetes. We therefore investigated whether HDL and LDL play a role in the regulation of pancreatic islet cell apoptosis, proliferation, and secretory function. Isolated mouse and human islets were exposed to plasma lipoproteins of healthy human donors. In murine and human beta-cells, LDL decreased both proliferation and maximal glucose-stimulated insulin secretion. The comparative analysis of beta-cells from wild-type and LDL receptor-deficient mice revealed that the inhibitory effect of LDL on insulin secretion but not proliferation requires the LDL receptor. HDL was found to modulate the survival of both human and murine islets by decreasing basal as well as IL-1beta and glucose-induced apoptosis. IL-1beta-induced beta-cell apoptosis was also inhibited in the presence of either the delipidated protein or the deproteinated lipid moieties of HDL, apolipoprotein A1 (the main protein component of HDL), or sphingosine-1-phosphate (a bioactive sphingolipid mostly carried by HDL). In murine beta-cells, the protective effect of HDL against IL-1beta-induced apoptosis was also observed in the absence of the HDL receptor scavenger receptor class B type 1. Our data show that both LDL and HDL affect function or survival of beta-cells and raise the question whether dyslipidemia contributes to beta-cell failure and hence the manifestation and progression of type 2 diabetes mellitus.


Subject(s)
Apoptosis , Cell Proliferation , Insulin-Secreting Cells/physiology , Insulin/metabolism , Lipoproteins, HDL/physiology , Lipoproteins, LDL/physiology , Animals , Apolipoprotein A-I/metabolism , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Survival , Cells, Cultured , Female , Glucose/metabolism , Humans , Insulin Secretion , Interleukin-1beta/metabolism , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nitric Oxide Synthase Type II/metabolism , Receptors, LDL/metabolism , Scavenger Receptors, Class B/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , fas Receptor/metabolism
15.
Biochemistry ; 43(17): 5084-93, 2004 May 04.
Article in English | MEDLINE | ID: mdl-15109267

ABSTRACT

We have generated and studied the pattern of expression of transgenic mouse lines carrying the human apoA-I and apoCIII gene cluster mutated at different sites. In two lines, we have either mutated the hormone-response element (HRE) of element G of the apoCIII enhancer or the C/EBP binding site of the proximal apoA-I promoter. In a third line, we have mutated the two HREs of the apoA-I promoter and the HRE of the apoCIII enhancer. Mutations in the HRE of element G reduced the hepatic and intestinal expressions of the reporter chloramphenicol acetyltransferase (CAT) gene (which substituted the apoCIII gene) to 4 and 13% of the wild-type (WT) control, whereas the hepatic and intestinal expressions of the apoA-I gene were reduced to 92 and 25% of the WT control, respectively. A mutation in the C/EBP site increased the hepatic and intestinal expressions of the apoA-I gene approximately 1.25- and 1.6-fold, respectively, and did not affect the expression of the CAT gene. The mutation in the three HNF-4 binding sites of the apoA-I promoter/apoCIII enhancer nearly abolished the expression of apoA-I and the reporter CAT gene in all tissues. These findings establish the importance of the HREs for the hepatic and intestinal expressions of the apoA-I and apoCIII genes and suggest that C/EBP does not play a central role in the expression of the apoA-I gene.


Subject(s)
Apolipoprotein A-I/genetics , Apolipoproteins C/genetics , Enhancer Elements, Genetic , Intestinal Mucosa/metabolism , Liver/metabolism , Promoter Regions, Genetic , Response Elements , Animals , Apolipoprotein C-III , Base Sequence , Binding Sites , Cell Line , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multigene Family , Mutation , Transcription, Genetic , Transcriptional Activation
16.
Biochemistry ; 41(30): 9293-301, 2002 Jul 30.
Article in English | MEDLINE | ID: mdl-12135350

ABSTRACT

Apolipoprotein E (apoE) isoforms are key determinants of susceptibility to late-onset Alzheimer's disease (AD). The epsilon 4 and epsilon 2 alleles have been associated with increased and decreased risk for AD, respectively. We have generated and characterized transgenic mice in which the human apoE2 gene is expressed under the control of the platelet-derived growth factor B-chain (PDGF-B) promoter, or the transferrin (TF) promoter. S1 nuclease analysis and immunoblotting showed that the PDGF-B apoE2 mice express apoE2 exclusively in the brain whereas the TF apoE2 mice express apoE2 in the liver and in the brain. In the TF apoE2 mouse line, apoE2 is also detected in the plasma. The PDGF-B apoE2 and the TF apoE2 transgenic mice were bred back to apoE(-)(/)(-) background. Immunohistochemical analysis showed that the PDGF apoE2 x apoE(-)(/)(-) and the TF apoE2 x apoE(-)(/)(-) mice express human apoE2 within the neocortex in hippocampal neurons and glial cells, respectively. ApoE(-)(/)(-) mice have been shown to develop age-dependent loss of synaptophysin. Immunoblotting of mouse brain extracts and immunohistochemical analysis of brain sections showed that apoE expression in both apoE2 x apoE(-)(/)(-) transgenic lines was associated with significant recovery of brain synaptophysin levels as compared to the levels of apoE(-)(/)(-) littermates of the same age. These apoE2-expressing mice, when bred back on amyloid precursor protein (APP) transgenic mice or other mouse lines featuring alterations in lipoprotein metabolism, may provide new mouse models for elucidating the role of apoE2 in lipid homeostasis in the brain and in the pathogenesis of AD.


Subject(s)
Apolipoproteins E/genetics , Neuroglia/metabolism , Neurons/metabolism , Animals , Apolipoprotein E2 , Base Sequence , Blotting, Western , Brain/metabolism , DNA Primers , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Platelet-Derived Growth Factor/genetics , Promoter Regions, Genetic , Synaptophysin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...