Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.007
Filter
1.
Mol Brain ; 17(1): 61, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39223647

ABSTRACT

Ischemic stroke (IS) is a severe cerebrovascular disease with high disability and mortality rates, where the inflammatory response is crucial to its progression and prognosis. Efferocytosis, the prompt removal of dead cells, can reduce excessive inflammation after IS injury. While electroacupuncture (EA) has been shown to decrease inflammation post-ischemia/reperfusion (I/R), its link to efferocytosis is unclear. Our research identified ATP-binding cassette transporter A1 (Abca1) as a key regulator of the engulfment process of efferocytosis after IS by analyzing public datasets and validating findings in a mouse model, revealing its close ties to IS progression. We demonstrated that EA can reduce neuronal cell death and excessive inflammation caused by I/R. Furthermore, EA treatment increased Abca1 expression, prevented microglia activation, promoted M2 microglia polarization, and enhanced their ability to phagocytose injured neurons in I/R mice. This suggests that EA's modulation of efferocytosis could be a potential mechanism for reducing cerebral I/R injury, making regulators of efferocytosis steps a promising therapeutic target for EA benefits.


Subject(s)
ATP Binding Cassette Transporter 1 , Electroacupuncture , Inflammation , Mice, Inbred C57BL , Microglia , Phagocytosis , Reperfusion Injury , Animals , Microglia/metabolism , Microglia/pathology , Electroacupuncture/methods , ATP Binding Cassette Transporter 1/metabolism , Reperfusion Injury/pathology , Reperfusion Injury/therapy , Reperfusion Injury/metabolism , Inflammation/pathology , Male , Brain Ischemia/pathology , Brain Ischemia/metabolism , Brain Ischemia/therapy , Mice , Neurons/metabolism , Neurons/pathology , Disease Models, Animal , Efferocytosis
2.
Sci Rep ; 14(1): 21371, 2024 09 12.
Article in English | MEDLINE | ID: mdl-39266573

ABSTRACT

Atherosclerosis (AS) is a major cause of cardiovascular diseases that may lead to mortality. This study aimed to evaluate the therapeutic potential of tetrandrine in high cholesterol diet (HCD)-induced atherosclerosis, in rats, via modulation of miR-34a, as well as, Wnt5a/Ror2/ABCA1/NF-κB pathway and to compare its efficacy with atorvastatin. Induction of AS, in male rats, was done via IP administration of vitamin D3 (70 U/Kg for 3 days) together with HCD. At the end of the 9th week, rats were treated with atorvastatin at a dose of 20 mg/kg, and tetrandrine at different doses of (18.75, and 31.25 mg/kg) for 22 days. Serum inflammatory cytokines and lipid profile, liver oxidative stress parameters, and aortic tissue Wnt5a, Ror2, ABCA1, NF-κB, miR-34a levels were assessed in all experimental groups. Histopathological and Immunohistochemical assessments of aortic tissue sections were done. Results showed that tetrandrine treatment reverted the inflammatory and oxidative stress state together with reducing the serum lipids via modulating miR-34a, and Wnt5a/Ror2/ABCA1/NF-κB pathway. Moreover, it reverted the histopathological abnormalities observed in AS rats. Tetrandrine beneficial effects, in both doses, were comparable to that of atorvastatin, in most of the discussed parameters. These findings praise tetrandrine as a promising agent for management of atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter 1 , Atherosclerosis , Benzylisoquinolines , MicroRNAs , NF-kappa B , Wnt-5a Protein , Animals , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , Atherosclerosis/pathology , Wnt-5a Protein/metabolism , Rats , NF-kappa B/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Male , Benzylisoquinolines/pharmacology , Benzylisoquinolines/therapeutic use , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , Oxidative Stress/drug effects , Cholecalciferol/pharmacology , Signal Transduction/drug effects , Rats, Wistar , Diet, High-Fat/adverse effects , Cholesterol, Dietary/adverse effects
3.
Nutrients ; 16(17)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39275180

ABSTRACT

Dysfunction or loss of pancreatic ß cells can cause insulin deficiency and impaired glucose regulation, resulting in conditions like type 2 diabetes. The ATP-binding cassette transporter A1 (ABCA1) plays a key role in the reverse cholesterol transport system, and its decreased expression is associated with pancreatic ß cell lipotoxicity, resulting in abnormal insulin synthesis and secretion. Increased glutamate release can cause glucotoxicity in ß cells, though the detailed mechanisms remain unclear. This study investigated the effect of N-methyl-D-aspartic acid (NMDA) on ABCA1 expression in INS-1 cells and primary pancreatic islets to elucidate the signaling mechanisms that suppress insulin secretion. Using Western blotting, microscopy, and biochemical analyses, we found that NMDA activated the mitogen-activated protein kinase (MEK)-dependent pathway, suppressing ABCA1 protein and mRNA expression. The MEK-specific inhibitor PD98059 restored ABCA1 promoter activity, indicating the involvement of the extracellular signal-regulated kinase (MEK/ERK) pathway. Furthermore, we identified the liver X receptor (LXR) as an effector transcription factor in NMDA regulation of ABCA1 transcription. NMDA treatment increased cholesterol and triglyceride levels while decreasing insulin secretion, even under high-glucose conditions. These effects were abrogated by treatment with PD98059. This study reveals that NMDA suppresses ABCA1 expression via the MEK/ERK/LXR pathway, providing new insights into the pathological suppression of insulin secretion in pancreatic ß cells and emphasizing the importance of investigating the role of NMDA in ß cell dysfunction.


Subject(s)
ATP Binding Cassette Transporter 1 , Insulin-Secreting Cells , Liver X Receptors , MAP Kinase Signaling System , N-Methylaspartate , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/drug effects , Animals , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , N-Methylaspartate/pharmacology , Rats , Liver X Receptors/metabolism , MAP Kinase Signaling System/drug effects , Cholesterol/metabolism , Insulin/metabolism , Insulin Secretion/drug effects , Male , Extracellular Signal-Regulated MAP Kinases/metabolism , Cell Line
4.
Sci Rep ; 14(1): 18853, 2024 08 14.
Article in English | MEDLINE | ID: mdl-39143134

ABSTRACT

Eriodictyol, a flavonoid distributed in citrus fruits, has been known to exhibit anti-inflammatory activity. In this study, destabilized medial meniscus (DMM)-induced OA model was used to investigate the protective role of eriodictyol on OA. Meanwhile, we used an IL-1ß-stimulated human osteoarthritis chondrocytes model to investigate the anti-inflammatory mechanism of eriodictyol on OA. The production of nitric oxide was detected by Griess reaction. The productions of MMP1, MMP3, and PGE2 were detected by ELISA. The expression of LXRα, ABCA1, PI3K, AKT, and NF-κB were measured by western blot analysis. The results demonstrated that eriodictyol could alleviate DMM-induced OA in mice. In vitro, eriodictyol inhibited IL-1ß-induced NO, PGE2, MMP1, and MMP3 production in human osteoarthritis chondrocytes. Eriodictyol also suppressed the phosphorylation of PI3K, AKT, NF-κB p65, and IκBα induced by IL-1ß. Meanwhile, eriodictyol significantly increased the expression of LXRα and ABCA1. Furthermore, eriodictyol disrupted lipid rafts formation through reducing the cholesterol content. And cholesterol replenishment experiment showed that adding water-soluble cholesterol could reverse the anti-inflammatory effect of eriodictyol. In conclusion, the results indicated eriodictyol inhibited IL-1ß-induced inflammation in human osteoarthritis chondrocytes through suppressing lipid rafts formation, which subsequently inhibiting PI3K/AKT/NF-κB signaling pathway.


Subject(s)
Chondrocytes , Flavanones , NF-kappa B , Osteoarthritis , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Flavanones/pharmacology , Animals , Humans , Signal Transduction/drug effects , NF-kappa B/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Chondrocytes/drug effects , Chondrocytes/metabolism , Mice , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Interleukin-1beta/metabolism , Liver X Receptors/metabolism , Male , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , Disease Progression , Disease Models, Animal , Anti-Inflammatory Agents/pharmacology , Nitric Oxide/metabolism , Mice, Inbred C57BL
5.
Nutrients ; 16(15)2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39125437

ABSTRACT

In humans, α-tocopherol (α-TOC) is mainly stored in adipose tissue, where it participates in preventing damages induced by inflammation and reactive oxygen species. Factors, including genetic ones, that explain adipose tissue α-TOC concentration remain poorly understood. This study, therefore, aimed to characterize the interindividual variability of adipose tissue α-TOC concentration in healthy individuals and to identify single nucleotide polymorphisms (SNPs) associated with it. The study used a randomized cross-over design with 42 healthy adult males. α-TOC concentration was measured in fasting plasma and periumbilical adipose tissue samples, both at fast and 8 h after consumption of three standard meals. Partial least squares (PLS) regression was performed to identify SNPs associated with the interindividual variability of adipose tissue α-TOC concentration. Adipose tissue α-TOC concentration was not associated with fasting plasma concentration (Pearson's r = 0.24, 95% CI: [-0.08, 0.51]). There was a high interindividual variability of adipose tissue α-TOC concentration (CV = 61%). A PLS regression model comprising 10 SNPs in five genes (PPARG, ABCA1, BUD13, CD36, and MGLL) explained 60% (adjusted R2) of the variability of this concentration. The interindividual variability of adipose tissue α-TOC concentration in humans is due, at least partly, to SNPs in genes involved in α-TOC and triglyceride metabolism.


Subject(s)
Cross-Over Studies , Polymorphism, Single Nucleotide , Subcutaneous Fat , alpha-Tocopherol , Humans , Male , alpha-Tocopherol/blood , alpha-Tocopherol/metabolism , Adult , Subcutaneous Fat/metabolism , Young Adult , Fasting , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , CD36 Antigens/genetics , CD36 Antigens/metabolism , Healthy Volunteers
6.
Invest Ophthalmol Vis Sci ; 65(10): 29, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39167399

ABSTRACT

Purpose: Dysregulated cholesterol metabolism is critical in the pathogenesis of AMD. Cellular senescence contributes to the development of numerous age-associated diseases. In this study, we investigated the link between cholesterol burden and the cellular senescence of photoreceptors. Methods: Retinas from rod-specific ATP binding cassette subfamily A member 1 (Abca1) and G member 1 (Abcg1) (Abca1/g1-rod/-rod) knockout mice fed with a high-fat diet were analyzed for the signs of cellular senescence. Real-time quantitative PCR and immunofluorescence were used to characterize the senescence profile of the retina and cholesterol-treated photoreceptor cell line (661W). Inducible elimination of p16(Ink4a)-positive senescent cells (INK-ATTAC) mice or the administration of senolytic drugs (dasatinib and quercetin: D&Q) were used to examine the impact of senolytics on AMD-like phenotypes in Abca1/g1-rod/-rod retina. Results: Increased accumulation of senescent cells as measured by markers of cellular senescence was found in Abca1/g1-rod/-rod retina. Exogenous cholesterol also induced cellular senescence in 661W cells. Selective elimination of senescent cells in Abca1/g1-rod/-rod;INK-ATTAC mice or by administration of D&Q improved visual function, lipid accumulation in retinal pigment epithelium, and Bruch's membrane thickening. Conclusions: Cholesterol accumulation promotes cellular senescence in photoreceptors. Eliminating senescent photoreceptors improves visual function in a model of retinal neurodegeneration, and senotherapy offers a novel therapeutic avenue for further investigation.


Subject(s)
ATP Binding Cassette Transporter 1 , Cellular Senescence , Cholesterol , Disease Models, Animal , Mice, Knockout , Retinal Degeneration , Animals , Mice , Cellular Senescence/physiology , Cholesterol/metabolism , ATP Binding Cassette Transporter 1/metabolism , Retinal Degeneration/metabolism , Retinal Degeneration/pathology , Real-Time Polymerase Chain Reaction , Mice, Inbred C57BL , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/genetics , Retinal Rod Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/pathology , Retinal Rod Photoreceptor Cells/physiology
7.
Lipids Health Dis ; 23(1): 275, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39210350

ABSTRACT

Despite recent findings indicating a paradoxical association between high-density lipoprotein cholesterol (HDL-C) levels and cardiovascular disease (CVD) mortality, the impact of HDL-C on subsequent outcomes after ischemic stroke remains unclear. The study aims to investigate the relationships between HDL-C levels and post-stroke functional outcomes while examining the potential modifying influence of HDL-C-related single nucleotide polymorphisms identified through genome-wide association studies. This cohort study included 1,310 patients diagnosed with acute ischemic stroke (AIS), all of whom had their admission serum lipid profile and genotyping information. Participants were categorized into four groups based on gender and HDL-C level. Prognostic outcomes were assessed using a modified Rankin Scale (mRS) at 1, 3, and 12 months post-admission. Multivariate logistic regression and restricted cubic spline regression analysis were used to assess the associations between HDL-C levels and outcomes. The mean age of patients was 61.17 ± 12.08 years, and 69.31% were men. After adjusting confounders, patients with the highest HDL-C level group had a significantly higher risk of poor functional outcomes at 1, 3, and 12 months following stroke compared to the reference group. Restricted cubic splines depicted a nonlinear association between HDL-C levels and poor prognosis in both men and women. The ABCA1 gene rs2575876 AA genotype combined with abnormal HDL-C levels exhibited a significantly heightened risk of post-stroke adverse outcomes at 1 and 3 months compared to patients with normal HDL-C levels and GG + GA genotype. These findings suggest that the combined effects of ABCA1 genetic variants with either low or high HDL-C levels could further heighten this risk.


Subject(s)
ATP Binding Cassette Transporter 1 , Cholesterol, HDL , Ischemic Stroke , Polymorphism, Single Nucleotide , Humans , Male , Female , Middle Aged , Ischemic Stroke/genetics , Ischemic Stroke/blood , Aged , Cholesterol, HDL/blood , ATP Binding Cassette Transporter 1/genetics , Taiwan , Prognosis , Lipoproteins, HDL/blood , Lipoproteins, HDL/genetics , Risk Factors , Genotype
9.
Exp Eye Res ; 246: 110016, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39098587

ABSTRACT

Progressive Rod-Cone Degeneration (PRCD) is an integral membrane protein found in photoreceptor outer segment (OS) disc membranes and its function remains unknown. Mutations in Prcd are implicated in Retinitis pigmentosa (RP) in humans and multiple dog breeds. PRCD-deficient models exhibit decreased levels of cholesterol in the plasma. However, potential changes in the retinal cholesterol remain unexplored. In addition, impaired phagocytosis observed in these animal models points to potential deficits in the retinal pigment epithelium (RPE). Here, using a Prcd-/- murine model we investigated the alterations in the retinal cholesterol levels and impairments in the structural and functional integrity of the RPE. Lipidomic and immunohistochemical analyses show a 5-fold increase in the levels of cholesteryl esters (C.Es) and lipid deposits in the PRCD-deficient retina, respectively, indicating alterations in total retinal cholesterol. Furthermore, the RPE of Prcd-/- mice exhibit a 1.7-fold increase in the expression of lipid transporter gene ATP-binding cassette transporter A1 (Abca1). Longitudinal fundus and spectral domain optical coherence tomography (SD-OCT) examinations showed focal lesions and RPE hyperreflectivity. Strikingly, the RPE of Prcd-/- mice exhibited age-related pathological features such as lipofuscin accumulation, Bruch's membrane (BrM) deposits and drusenoid focal deposits, mirroring an Age-related Macular Degeneration (AMD)-like phenotype. We propose that the extensive lipofuscin accumulation likely impairs lysosomal function, leading to the defective phagocytosis observed in Prcd-/- mice. Our findings support the dysregulation of retinal cholesterol homeostasis in the absence of PRCD. Further, we demonstrate that progressive photoreceptor degeneration in Prcd-/- mice is accompanied by progressive structural and functional deficits in the RPE, which likely exacerbates vision loss over time.


Subject(s)
Disease Models, Animal , Retinal Pigment Epithelium , Tomography, Optical Coherence , Animals , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Mice , Lipid Metabolism , Mice, Knockout , Mice, Inbred C57BL , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Cholesterol/metabolism , Cholesterol Esters/metabolism , Cone-Rod Dystrophies/metabolism , Cone-Rod Dystrophies/genetics , Electroretinography , Bruch Membrane/metabolism , Bruch Membrane/pathology , Immunohistochemistry , Macular Degeneration/congenital
10.
Int J Mol Sci ; 25(16)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39201292

ABSTRACT

MicroRNAs (miRs) are small non-coding RNAs that regulate gene expression post-transcriptionally and are crucial in lipid metabolism. ATP-binding cassette transporter A1 (ABCA1) is essential for cholesterol efflux from cells to high-density lipoprotein (HDL). Dysregulation of miRs targeting ABCA1 can affect cholesterol homeostasis and contribute to coronary artery disease (CAD). This study aimed to investigate the expression of miRs targeting ABCA1 in human monocytes, their role in cholesterol efflux, and their relationship with CAD. We included 50 control and 50 CAD patients. RT-qPCR examined the expression of miR-33a-5p, miR-26a-5p, and miR-144-3p in monocytes. Logistic regression analysis explored the association between these miRs and CAD. HDL's cholesterol acceptance was analyzed using the J774A.1 cell line. Results showed that miR-26a-5p (p = 0.027) and ABCA1 (p = 0.003) expression levels were higher in CAD patients, while miR-33a-5p (p < 0.001) levels were lower. Downregulation of miR-33a-5p and upregulation of ABCA1 were linked to a lower CAD risk. Atorvastatin upregulated ABCA1 mRNA, and metformin downregulated miR-26a-5p in CAD patients. Decreased cholesterol efflux correlated with higher CAD risk and inversely with miRs in controls. Reduced miR-33a-5p expression and increased ABCA1 expression are associated with decreased CAD risk. miR deregulation in monocytes may influence atherosclerotic plaque formation by regulating cholesterol efflux. Atorvastatin and metformin could offer protective effects by modulating miR-33a-5p, miR-26a-5p, and ABCA1, suggesting potential therapeutic strategies for CAD prognosis and treatment.


Subject(s)
ATP Binding Cassette Transporter 1 , Coronary Artery Disease , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Coronary Artery Disease/genetics , Coronary Artery Disease/metabolism , Coronary Artery Disease/blood , Male , Female , Middle Aged , Leukocytes, Mononuclear/metabolism , Gene Expression Regulation , Aged , Cell Line , Cholesterol/metabolism , Cholesterol/blood , Monocytes/metabolism
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1869(8): 159546, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39089642

ABSTRACT

ABCA1 plays an essential role in the formation of high-density lipoprotein (HDL), and its mutations cause Tangier disease (TD), a familial HDL deficiency. In addition to the disappearance of HDL, TD patients exhibit cholesterol deposition in peripheral tissues through a mechanism poorly understood, which may contribute to the development of premature atherosclerosis. We and others previously showed that ABCA1 deficiency causes hyperactivation of the SREBP2 pathway in vitro. Here, we show using Abca1 knockout mice that ABCA1 deficiency leads to tissue-specific dysregulation of SREBP2 activity in a nutritional status-dependent manner, which may underlie the pathophysiology of TD.


Subject(s)
ATP Binding Cassette Transporter 1 , Signal Transduction , Tangier Disease , Animals , Humans , Mice , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/deficiency , Cholesterol/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, HDL/genetics , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Sterol Regulatory Element Binding Protein 2/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Tangier Disease/genetics , Tangier Disease/metabolism , Tangier Disease/pathology
12.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167476, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39181517

ABSTRACT

Hypoxic insult to the fetal brain causes loss of vulnerable premyelinating oligodendrocytes and arrested oligodendrocyte differentiation. Astrocytes influence oligodendrocyte differentiation and the astrocytic response to hypoxia could affect oligodendrocyte maturation under hypoxia. To identify pathways by which astrocytes influence oligodendroglial maturation in hypoxic injury, human fetal neural stem cell-derived astrocytes were exposed to 0.2 % oxygen for 48 hours. Transcriptomic analysis revealed the upregulation of the cholesterol-biosynthesis pathway in hypoxia-exposed astrocytes. Hypoxia-exposed primary astrocytes and astrocytic cell line (SVG) showed increased expression of hydroxy-methyl-glutaryl-CoA reductase (HMGCR), squalene epoxidase (SQLE), apolipoprotein E (apoE) and ATP-binding cassette transporter 1 (ABCA1) on qPCR and Western blot. Hypoxic SVG also showed increased cholesterol content in cells and culture supernatants and increased cell surface expression of ABCA1. Interestingly hypoxia-exposed premyelinating oligodendrocytes (Mo3.13) showed reduced cholesterol along with decreased expression of HMGCR and SQLE on qPCR and Western blot. Exogenous cholesterol increased the differentiation of Mo3.13 as measured by increased expression of myelin basic protein (MBP) on flow cytometry. Hypoxia exposure resulted in increased cholesterol transport from astrocytes to oligodendrocytes in cocultures with BODIPY-cholesterol labelled SVG and membrane-labelled Mo3.13. As exogenous cholesterol enhanced oligodendrocyte differentiation, our findings indicate that increased cholesterol synthesis by astrocytes and transport to oligodendrocytes could supplement oligodendroglial maturation in conditions of hypoxic brain injury in neonates.


Subject(s)
ATP Binding Cassette Transporter 1 , Astrocytes , Cell Differentiation , Cholesterol , Oligodendroglia , Oligodendroglia/metabolism , Oligodendroglia/pathology , Humans , Cholesterol/metabolism , Astrocytes/metabolism , Astrocytes/pathology , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , Cell Hypoxia , Myelin Sheath/metabolism , Myelin Sheath/pathology , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl CoA Reductases/genetics , Cells, Cultured , Squalene Monooxygenase/metabolism , Squalene Monooxygenase/genetics , Neural Stem Cells/metabolism , Neural Stem Cells/pathology , Cell Line
13.
Biochim Biophys Acta Mol Basis Dis ; 1870(8): 167479, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39181516

ABSTRACT

Intracellular cholesterol metabolism is regulated by the SREBP-2 and LXR signaling pathways. The effects of inflammation on these molecular mechanisms remain poorly studied, especially at the blood-brain barrier (BBB) level. Tumor necrosis factor α (TNFα) is a proinflammatory cytokine associated with BBB dysfunction. Therefore, the aim of our study was to investigate the effects of TNFα on BBB cholesterol metabolism, focusing on its underlying signaling pathways. Using a human in vitro BBB model composed of human brain-like endothelial cells (hBLECs) and brain pericytes (HBPs), we observed that TNFα increases BBB permeability by degrading the tight junction protein CLAUDIN-5 and activating stress signaling pathways in both cell types. TNFα also promotes cholesterol release and decreases cholesterol accumulation and APOE secretion. In hBLECs, the expression of SREBP-2 targets (LDLR and HMGCR) is increased, while ABCA1 expression is decreased. In HBPs, only LDLR and ABCA1 expression is increased. TNFα treatment also induces 25-hydroxycholesterol (25-HC) production, a cholesterol metabolite involved in the immune response and intracellular cholesterol metabolism. 25-HC pretreatment attenuates TNFα-induced BBB leakage and partially alleviates the effects of TNFα on ABCA1, LDLR, and HMGCR expression. Overall, our results suggest that TNFα favors cholesterol efflux via an LXR/ABCA1-independent mechanism at the BBB, while it activates the SREBP-2 pathway. Treatment with 25-HC partially reversed the effect of TNFα on the LXR/SREBP-2 pathways. Our study provides novel perspectives for better understanding cerebrovascular signaling events linked to BBB dysfunction and cholesterol metabolism in neuroinflammatory diseases.


Subject(s)
Blood-Brain Barrier , Cholesterol , Endothelial Cells , Hydroxycholesterols , Sterol Regulatory Element Binding Protein 2 , Tumor Necrosis Factor-alpha , Hydroxycholesterols/pharmacology , Hydroxycholesterols/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/drug effects , Humans , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Sterol Regulatory Element Binding Protein 2/metabolism , Sterol Regulatory Element Binding Protein 2/genetics , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Cholesterol/metabolism , Receptors, LDL/metabolism , Receptors, LDL/genetics , Signal Transduction/drug effects , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , Pericytes/metabolism , Pericytes/drug effects , Pericytes/pathology , Hydroxymethylglutaryl CoA Reductases/metabolism , Hydroxymethylglutaryl CoA Reductases/genetics , Apolipoproteins E/metabolism , Apolipoproteins E/genetics , Liver X Receptors/metabolism , Liver X Receptors/genetics , Cells, Cultured
15.
Cell ; 187(19): 5336-5356.e30, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39137777

ABSTRACT

Tumors growing in metabolically challenged environments, such as glioblastoma in the brain, are particularly reliant on crosstalk with their tumor microenvironment (TME) to satisfy their high energetic needs. To study the intricacies of this metabolic interplay, we interrogated the heterogeneity of the glioblastoma TME using single-cell and multi-omics analyses and identified metabolically rewired tumor-associated macrophage (TAM) subpopulations with pro-tumorigenic properties. These TAM subsets, termed lipid-laden macrophages (LLMs) to reflect their cholesterol accumulation, are epigenetically rewired, display immunosuppressive features, and are enriched in the aggressive mesenchymal glioblastoma subtype. Engulfment of cholesterol-rich myelin debris endows subsets of TAMs to acquire an LLM phenotype. Subsequently, LLMs directly transfer myelin-derived lipids to cancer cells in an LXR/Abca1-dependent manner, thereby fueling the heightened metabolic demands of mesenchymal glioblastoma. Our work provides an in-depth understanding of the immune-metabolic interplay during glioblastoma progression, thereby laying a framework to unveil targetable metabolic vulnerabilities in glioblastoma.


Subject(s)
Brain Neoplasms , Glioblastoma , Myelin Sheath , Tumor Microenvironment , Humans , Myelin Sheath/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioblastoma/metabolism , Glioblastoma/pathology , Animals , Mice , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Cholesterol/metabolism , Liver X Receptors/metabolism , Macrophages/metabolism , Cell Line, Tumor , ATP Binding Cassette Transporter 1/metabolism , Female , Male
16.
J Med Chem ; 67(17): 15061-15079, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39191400

ABSTRACT

Therapeutics enhancing apolipoprotein (APOE) positive function are a priority, because APOE4 is the major genetic risk factor for Alzheimer's disease (AD). The function of APOE, the key constituent of lipoprotein particles that transport cholesterol and lipids in the brain, is dependent on lipidation by ABCA1, a cell-membrane cholesterol transporter. ABCA1 transcription is regulated by liver X receptors (LXR): agonists have been shown to increase ABCA1, often accompanied by unwanted lipogenesis and elevated triglycerides (TG). Therefore, nonlipogenic ABCA1-inducers (NLAI) are needed. Two rounds of optimization of an HTS hit, derived from a phenotypic screen, gave lead compound 39 that was validated and tested in E3/4FAD mice that express human APOE3/4 and five mutant APP and PSEN1 human transgenes. Treatment with 39 increased ABCA1 expression, enhanced APOE lipidation, and reversed multiple AD phenotypes, without increasing TG. This NLAI/LXR-agonist study is the first in a human APOE-expressing model with hallmark amyloid-ß pathology.


Subject(s)
ATP Binding Cassette Transporter 1 , Alzheimer Disease , Apolipoprotein E3 , Apolipoprotein E4 , Disease Models, Animal , Mice, Transgenic , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter 1/genetics , Humans , Mice , Apolipoprotein E4/metabolism , Apolipoprotein E4/genetics , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Liver X Receptors/agonists , Liver X Receptors/metabolism , Presenilin-1/genetics , Presenilin-1/metabolism
17.
Phytomedicine ; 133: 155944, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39146879

ABSTRACT

BACKGROUND: Intrahepatic cholangiocarcinoma (ICC) remains a significant challenge in cancer therapy, especially due to its resistance to established treatments like Gemcitabine, necessitating novel therapeutic approaches. METHODS: This study utilized Gemcitabine-resistant cell lines, patient-derived organotypic tumor spheroids (PDOTs), and patient-derived xenografts (PDX) to evaluate the effects of Saikosaponin-a (SSA) on ICC cellular proliferation, migration, apoptosis, and its potential synergistic interaction with Gemcitabine. Techniques such as transcriptome sequencing, Luciferase reporter assays, and molecular docking were employed to unravel the molecular mechanisms. RESULTS: SSA exhibited antitumor effects in both in vitro and PDX models, indicating its considerable potential for ICC treatment. SSA markedly inhibited ICC progression by reducing cellular proliferation, enhancing apoptosis, and decreasing migration and invasion. Crucially, it augmented Gemcitabine's efficacy by targeting the p-AKT/BCL6/ABCA1 signaling pathway. This modulation led to the downregulation of p-AKT and suppression of BCL6 transcriptional activity, ultimately reducing ABCA1 expression and enhancing chemosensitivity to Gemcitabine. Additionally, ABCA1 was validated as a predictive biomarker for drug resistance, with a direct correlation between ABCA1 expression levels and the IC50 values of various small molecule drugs in ICC gene profiles. CONCLUSION: This study highlights the synergistic potential of SSA combined with Gemcitabine in enhancing therapeutic efficacy against ICC and identifies ABCA1 as a key biomarker for drug responsiveness. Furthermore, the introduction of the novel PDOTs microfluidic model provides enhanced insights into ICC research. This combination strategy may provide a novel approach to overcoming treatment challenges in ICC.


Subject(s)
ATP Binding Cassette Transporter 1 , Bile Duct Neoplasms , Cholangiocarcinoma , Deoxycytidine , Drug Resistance, Neoplasm , Gemcitabine , Oleanolic Acid , Proto-Oncogene Proteins c-akt , Saponins , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Oleanolic Acid/pharmacology , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , Cholangiocarcinoma/drug therapy , Humans , Cell Line, Tumor , Animals , Proto-Oncogene Proteins c-akt/metabolism , Bile Duct Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , ATP Binding Cassette Transporter 1/metabolism , Mice , Apoptosis/drug effects , Cell Proliferation/drug effects , Signal Transduction/drug effects , Drug Synergism , Xenograft Model Antitumor Assays
18.
J Hazard Mater ; 476: 135048, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-38964041

ABSTRACT

While the cardiovascular system is a primary target of organophosphorus flame retardants (OPFRs), particularly aryl-OPFRs, it is still exclusive whether the diisodecyl phenyl phosphate (DIDPP), widely used and broadly present in the environment at high concentrations, elicits atherosclerosis effects. Liver X receptors (LXRs) play a direct role in regulating the formation of atherosclerotic lesions. This study was the first to demonstrate that DIDPP acts as an LXRα ligand and functions as an LXRα antagonist with a half-maximal inhibitory concentration of 16.2 µM. We showed that treatment of an in vitro macrophage model with 1 to 10 µM of DIDPP resulted in the downregulation of direct targets of LXRα, namely ABCA1, ABCG1 and SR-B1, thereby leading to a 7.9-13.2 % reduction in cholesterol efflux. This caused dose-dependent, 24.1-43.1 % increases in the staining intensity of foam cells in the macrophage model. This atherosclerotic effect of DIDPP was proposed to be due to its antagonism of LXRα activity, as DIDPP treatment did not alter cholesterol influx. In conclusion, the findings of this study demonstrate that exposure to DIDPP may be a risk factor for atherosclerosis due to the LXRα-antagonistic activity of DIDPP and its ubiquity in the environment.


Subject(s)
Foam Cells , Liver X Receptors , Liver X Receptors/metabolism , Foam Cells/drug effects , Foam Cells/metabolism , Animals , ATP Binding Cassette Transporter 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Cholesterol/metabolism , Organophosphates/pharmacology , Organophosphates/toxicity , Mice , Humans , Flame Retardants/toxicity , Flame Retardants/pharmacology , RAW 264.7 Cells , Scavenger Receptors, Class B/metabolism
19.
Phytomedicine ; 132: 155864, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39032281

ABSTRACT

BACKGROUND: Atherosclerosis is a long-lasting inflammatory condition affecting the walls of arteries, marked by the buildup of fats, plaque formation, and vascular remodeling. Recent findings highlight the significance of cholesterol removal pathways in influencing atherosclerosis, yet the connection between cholesterol removal and regulation of macrophage inflammation remains poorly understood. RBAP could serve as an anti-inflammatory agent; however, its role in atherosclerosis and the mechanism behind it are still not well understood. PURPOSE: The objective of this research is to explore how RBAP impacts cholesterol efflux, which is a considerable element in the advancement of atherosclerosis. METHODS: An atherosclerosis mouse model was established by using an ApoE KO strain mouse on a high-fat diet (HFD) to assess the effects of RBAP, conducted either orally or through injection. Additionally, in vitro experiments were conducted where the induction of THP-1 cells was conducted for the differentiation towards macrophages, and along with mouse RAW264.7 cells, were challenged with ox-LDL to evaluate the impact of RBAP. RESULTS: In this study, RBAP was found to reduce the production and downregulate TNF-α, IL-1ß, and IL-6 levels and inhibited the activation of the TLR4/MyD88/NF-κB signaling in atherosclerosis model mice, as well as in ox-LDL-challenged THP-1 cells and mouse RAW264.7 macrophages. RBAP's effectiveness also improved the enhancement of reverse cholesterol transport (RCT) and cholesterol removal to HDL and apoA1 by increasing the activity of genes related to cholesterol removal PPARγ/LXRα/ABCA1/ABCG1, both in ApoE-/- mice and in THP-1 cells and mouse RAW264.7 macrophages. Notably, RBAP exerted similar effects on atherosclerosis model mice and macrophages to those of TAK-242, an inhibitor of the TLR4 signaling. When RBAP and TAK-242 were applied simultaneously, the improvement was not enhanced compared with either RBAP or TAK-242 treatment alone. CONCLUSION: These findings suggest that RBAP, as a TLR4 inhibitor, has anti-atherosclerotic effects by improving inflammation and promoting cholesterol effection, indicating its therapeutic potential in intervening atherosclerosis.


Subject(s)
Atherosclerosis , Cell Differentiation , Cholesterol , Foam Cells , Macrophages , Oryza , Toll-Like Receptor 4 , Animals , Atherosclerosis/drug therapy , Mice , Cholesterol/metabolism , Foam Cells/drug effects , Foam Cells/metabolism , RAW 264.7 Cells , Cell Differentiation/drug effects , Humans , Toll-Like Receptor 4/metabolism , Macrophages/drug effects , Macrophages/metabolism , Disease Models, Animal , THP-1 Cells , Male , Diet, High-Fat , ATP Binding Cassette Transporter 1/metabolism , Lipoproteins, LDL/metabolism , Mice, Inbred C57BL , Peptides/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 1/metabolism , Mice, Knockout, ApoE , NF-kappa B/metabolism , Apolipoproteins E , Anti-Inflammatory Agents/pharmacology
20.
Eur J Pharmacol ; 980: 176836, 2024 Oct 05.
Article in English | MEDLINE | ID: mdl-39032762

ABSTRACT

Dysregulated host response against infection triggers sepsis that leads to multiple organ dysfunction due to uncontrolled inflammatory responses. Despite marked progress in understanding of sepsis, numerous clinical trials for treatment of sepsis have proven daunting and a new therapeutic approach is highly needed. CE9A215 (inotodiol), a fungal secondary metabolite, has been researched for its pharmacological activities and has shown potent anti-allergic effects. In this study, we evaluated the anti-inflammatory activities of CE9A215 upon lipopolysaccharide (LPS) stimulation in vivo and in vitro for the first time. CE9A215 decreased the production of interleukin (IL)-6, tumor necrosis factor alpha (TNF-α), and IL-1ß in a concentration-dependent manner in LPS-stimulated RAW264.7 cells. Intriguingly, in human mast cell line LUVA, CE9A215 significantly lowered IL-4 and IL-10, and this effect could be beneficial for the clearance of bacterial infection. In addition, administration of CE9A215 improved the survival rate of LPS-stimulated mice and inhibited the pro-inflammatory cytokines, IL-6, TNF-α, and IL-1ß in blood. Moreover, CE9A215 enhanced the expression levels of plasma phospholipid transfer protein (PLTP), apolipoprotein E (ApoE), and ATP-binding cassette transporter (ABCA1) in LPS-stimulated RAW246.7 cells. Liver PLTP level increased significantly in the CE9A215-administered group compared with the control group, which implies that CE9A215 promotes LPS clearance and neutralization by reverse transport of LPS by increasing the expressions of PLTP, ApoE, and ABCA1. Our results highlight CE9A215's potential as a novel therapeutic option for the treatment of sepsis.


Subject(s)
ATP Binding Cassette Transporter 1 , Cytokines , Lipopolysaccharides , Sepsis , Animals , Sepsis/drug therapy , Sepsis/metabolism , Sepsis/chemically induced , Mice , Humans , RAW 264.7 Cells , ATP Binding Cassette Transporter 1/metabolism , Cytokines/metabolism , Male , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Lanosterol/analogs & derivatives , Lanosterol/pharmacology , Lanosterol/therapeutic use , Mast Cells/drug effects , Mast Cells/metabolism , Mast Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL