Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 366
Filter
1.
FASEB J ; 35(3): e21427, 2021 03.
Article in English | MEDLINE | ID: mdl-33629776

ABSTRACT

Porphyrins are used for cancer diagnostic and therapeutic applications, but the mechanism of how porphyrins accumulate in cancer cells remains elusive. Knowledge of how porphyrins enter cancer cells can aid the development of more accurate cancer diagnostics and therapeutics. To gain insight into porphyrin uptake mechanisms in cancer cells, we developed a flow cytometry assay to quantify cellular uptake of meso-tetra (4-carboxyphenyl) porphyrin (TCPP), a porphyrin that is currently being developed for cancer diagnostics. We found that TCPP enters cancer cells through clathrin-mediated endocytosis. The LDL receptor, previously implicated in the cellular uptake of other porphyrins, only contributes modestly to uptake. We report that TCPP instead binds strongly ( KD=42nM ) to CD320, the cellular receptor for cobalamin/transcobalamin II (Cbl/TCN2). Additionally, TCPP competes with Cbl/TCN2 for CD320 binding, suggesting that CD320 is a novel receptor for TCPP. Knockdown of CD320 inhibits TCPP uptake by up to 40% in multiple cancer cell lines, including lung, breast, and prostate cell lines, which supports our hypothesis that CD320 both binds to and transports TCPP into cancer cells. Our findings provide some novel insights into why porphyrins concentrate in cancer cells. Additionally, our study describes a novel function for the CD320 receptor which has been reported to transport only Cbl/TCN2 complexes.


Subject(s)
Neoplasms/metabolism , Porphyrins/pharmacology , Vitamin B 12/pharmacology , Biological Transport/drug effects , Endocytosis/drug effects , Endocytosis/physiology , Humans , Neoplasms/drug therapy , Porphyrins/metabolism , Receptors, LDL/drug effects , Receptors, LDL/metabolism , Vitamin B 12/metabolism
2.
Nutrients ; 14(1)2021 Dec 31.
Article in English | MEDLINE | ID: mdl-35011066

ABSTRACT

BACKGROUND: Dyslipidaemias, particularly elevated plasma low-density lipoprotein cholesterol (LDL-C) levels, are major risk factors for cardiovascular disease (CVD). Besides pharmacological approaches, a nutritional strategy for CVD prevention has gained increasing attention. Among functional foods, the hypocholesterolemic properties of soy are driven by a stimulation of LDL-receptor (LDL-R) activity. AIM: To characterize the effect of two soy peptides, namely, ß-conglycinin-derived YVVNPDNDEN and YVVNPDNNEN on the expression of proprotein convertase subtilisin/kexin type 9 (PCSK9), one of the key-regulators of the LDL-R. METHODS: PCSK9 promoter activity (luciferase assay), PCSK9 protein expression (WB) and secretion (ELISA), PCSK9 interaction with LDL-R (binding assay) and human HepG2 cells were the objects of this investigation. RESULTS: Treatment with YVVNPDNNEN peptide has led to a rise in PCSK9 gene expression (90.8%) and transcriptional activity (86.4%), and to a decrement in PCSK9 intracellular and secreted protein (-42.9%) levels. YVVNPDNNEN peptide reduced the protein expression of transcriptional factor HNF1α. Most changes driven by YVVNPDNDEN peptide were not statistically significant. Neither peptide inhibited the PCSK9-LDLR interaction. CONCLUSIONS: Although sharing a common effect on LDL-R levels through the inhibition of 3-hydroxy-3-methylglutaryl CoA reductase activity, only the YVVNPDNNEN peptide has an additional mechanism via the downregulation of PCSK9 protein levels.


Subject(s)
Antigens, Plant/chemistry , Gene Expression/drug effects , Globulins/chemistry , Peptides/pharmacology , Proprotein Convertase 9/genetics , Receptors, LDL/drug effects , Seed Storage Proteins/chemistry , Soybean Proteins/chemistry , Amino Acid Sequence , Cell Survival/drug effects , Dietary Supplements , Hep G2 Cells , Hepatocyte Nuclear Factor 1-alpha/analysis , Hepatocyte Nuclear Factor 1-alpha/genetics , Humans , Peptides/chemistry , Promoter Regions, Genetic/genetics , Proprotein Convertase 9/analysis , Proprotein Convertase 9/metabolism , Receptors, LDL/physiology
3.
ACS Chem Biol ; 15(12): 3262-3274, 2020 12 18.
Article in English | MEDLINE | ID: mdl-33270420

ABSTRACT

Alzheimer's Disease (AD) is a progressive neurodegenerative disease and the most common cause of dementia. The current treatment options for AD are limited to ameliorating cognitive decline temporarily and not reversing or preventing the progression of dementia. Hence, more effective therapeutic strategies are needed to combat this devastating disease. The low-density lipoprotein receptor has been shown to modulate the neuronal metabolism of cholesterol and apolipoprotein E, a major genetic risk factor for AD. LDLR overexpression in mice has been shown to increase amyloid-ß clearance and reduce amyloid deposition. We conducted a phenotypic screen to identify novel signaling pathways and targets that regulate LDLR expression in glial cells using an annotated compound library of approximately 29 000 compounds. The screen identified novel targets such as polo like kinase 1 (PLK1), activin receptor like kinase 5 (ALK5), and serotonin transporter (SERT). We used genetic, chemical biology and pathway analysis to confirm the target hypothesis. This work highlights that phenotypic screening is a promising strategy to identify novel mechanisms and targets for therapeutic intervention of complex neurodegenerative disorders.


Subject(s)
Receptors, LDL/drug effects , Small Molecule Libraries/pharmacology , Alzheimer Disease/pathology , Gene Knockdown Techniques , Humans , RNA, Small Interfering/genetics , Receptors, LDL/metabolism , Reproducibility of Results
4.
Nutrients ; 12(6)2020 Jun 09.
Article in English | MEDLINE | ID: mdl-32526887

ABSTRACT

This study was aimed at investigating the hypocholesterolemic effects of extra virgin olive oil (EVOO) phenols and the mechanisms behind the effect. Two phenolic extracts were prepared from EVOO of different cultivars and analyzed using the International Olive Council (IOC) official method for total phenols, a recently validated hydrolytic procedure for total hydroxytyrosol and tyrosol, and 1H-NMR analysis in order to assess their secoiridoid profiles. Both of the extracts inhibited in vitro the 3-hydroxy-3-methylglutaryl co-enzyme A reductase (HMGCoAR) activity in a dose-dependent manner. After the treatment of human hepatic HepG2 cells (25 µg/mL), they increased the low-density lipoprotein (LDL) receptor protein levels through the activation of the sterol regulatory element binding proteins (SREBP)-2 transcription factor, leading to a better ability of HepG2 cells to uptake extracellular LDL molecules with a final hypocholesterolemic effect. Moreover, both of the extracts regulated the intracellular HMGCoAR activity through the increase of its phosphorylation by the activation of AMP-activated protein kinase (AMPK)-pathways. Unlike pravastatin, they did not produce any unfavorable effect on proprotein convertase subtilisin/kexin 9 (PCSK9) protein level. Finally, the fact that extracts with different secoiridoid profiles induce practically the same biological effects suggests that the hydroxytyrosol and tyrosol derivatives may have similar roles in hypocholesterolemic activity.


Subject(s)
Anticholesteremic Agents/pharmacology , Olive Oil/chemistry , Phenols/pharmacology , Receptors, LDL/drug effects , Adenylate Kinase/metabolism , Enzyme Activation/drug effects , Hep G2 Cells , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Iridoids/analysis , Lipoproteins, LDL/metabolism , Liver/metabolism , Plant Extracts/chemistry , Receptors, LDL/metabolism , Sterol Regulatory Element Binding Protein 2/drug effects , Sterol Regulatory Element Binding Protein 2/metabolism
5.
Nutrients ; 12(5)2020 May 16.
Article in English | MEDLINE | ID: mdl-32429343

ABSTRACT

Genetic, epidemiological and pharmacological data have led to the conclusion that antagonizing or inhibiting Proprotein convertase subtilisin/kexin type 9 (PCSK9) reduces cardiovascular events. This clinical outcome is mainly related to the pivotal role of PCSK9 in controlling low-density lipoprotein (LDL) cholesterol levels. The absence of oral and affordable anti-PCSK9 medications has limited the beneficial effects of this new therapeutic option. A possible breakthrough in this field may come from the discovery of new naturally occurring PCSK9 inhibitors as a starting point for the development of oral, small molecules, to be used in combination with statins in order to increase the percentage of patients reaching their LDL-cholesterol target levels. In the present review, we have summarized the current knowledge on natural compounds or extracts that have shown an inhibitory effect on PCSK9, either in experimental or clinical settings. When available, the pharmacodynamic and pharmacokinetic profiles of the listed compounds are described.


Subject(s)
Enzyme Inhibitors/pharmacology , PCSK9 Inhibitors , Phytochemicals/pharmacology , Berberine/chemistry , Cholesterol, LDL/blood , Cholesterol, LDL/drug effects , Humans , Receptors, LDL/drug effects
6.
Pharmacol Res ; 158: 104847, 2020 08.
Article in English | MEDLINE | ID: mdl-32438039

ABSTRACT

Hypercholesterolemia has been documented to drive hormone-dependent breast cancer (BC) progression and resistance to hormonal therapy. Proprotein convertase subtilisin/kexin type-9 (PCSK9) regulates cholesterol metabolism through binding to LDL receptor (LDLR) and targeting the receptor for lysosomal degradation. Inhibition of PCSK9 is an established strategy to treat hypercholesterolemia. Pseurotin A (PS) is a unique spiro-heterocyclic γ-lactam alkaloid isolated from the fungus Aspergillus fumigatus. Preliminary studies indicated that PS lowered PCSK9 secretion in cultured HepG2 hepatocellular carcinoma cells, with an IC50 value of 1.20 µM. Docking studies suggested the ability of PS to bind at the PCSK9 narrow interface pocket that accommodates LDLR. Surface plasmon resonance (SPR) showed PS ability to inhibit the PCSK9-LDLR interaction at a concentration range of 10-150 µM. PS showed in vitro dose-dependent reduction of PCSK9, along with increased LDLR levels in hormone-dependent BT-474 and T47D breast cancer (BC) cell lines. In vivo, daily oral 10 mg/kg PS suppressed the progression of the hormone-dependent BT-474 BC cells in orthotopic nude mouse xenograft model. Immunohistochemistry (IHC) investigation of BT-474 breast tumor tissue proved the PS ability to reduce PCSK9 expression. PS also effectively suppressed BT-474 BC cells locoregional recurrence after primary tumor surgical excision. Western blot analysis showed decreased PCSK9 expression in liver tissues of PS-treated mice compared to vehicle-treated control group. PS treatment significantly reduced PCSK9 expression and normalized LDLR levels in collected primary and recurrent breast tumors at the study end. PS-treated mice showed reduced plasma cholesterol and 17ß-estradiol levels. Inhibition of tumor recurrence was associated with significant reductions in plasma level of the human BC recurrence marker CA 15-3 in treated mice at the study end. Histopathological examination of various PS-treated mice organs indicated lack of metastatic tumor cells and any pathological changes. The results of this study provide the first evidence for the suppression of the hormone-dependent breast tumor progression and recurrence by targeting the PCSK9-LDLR axis. PS is a novel first-in-class PCSK9-targeting lead appropriate for the use to control hormone-dependent BC progression and recurrence.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/drug therapy , Proprotein Convertase 9/metabolism , Pyrrolidinones/pharmacology , Receptors, LDL/drug effects , Animals , Antibiotics, Antineoplastic/therapeutic use , Breast Neoplasms/pathology , Cell Line, Tumor , Cholesterol/blood , Disease Progression , Dose-Response Relationship, Drug , Estradiol/blood , Female , Hep G2 Cells , Humans , Liver/drug effects , Liver/metabolism , Mice , Mice, Nude , Molecular Structure , Proprotein Convertase 9/drug effects , Pyrrolidinones/therapeutic use , Xenograft Model Antitumor Assays
7.
Life Sci ; 239: 117013, 2019 Dec 15.
Article in English | MEDLINE | ID: mdl-31678287

ABSTRACT

AIMS: Liver cancer is one of the leading causes of cancer mortality worldwide. Inspired by the biological structure and function of low-density lipoprotein (LDL), in this study, an ApopB-100 based targeted lipid nanoparticles was synthesized to improve the therapeutic efficacy in liver cancer treatment. MAIN METHODS: The biological composition of ApopB is similar to LDL which can effectively increase the targeting efficiency of nanoparticles in LDL receptor (LDLR)-overexpressed liver tumors. KEYFINDINGS: We have demonstrated that the co-administration of sorafenib (SRF) and Dihydroartemisinin (DHA) could exhibit synergistic anticancer effect in HepG2 liver cancer cells. DHA produced excessive cellular reactive oxygen species (ROS) and induced greater apoptosis of cancer cells. LDL-based SRF/DHA-loaded lipid nanoparticles (LD-SDN) showed remarkable decrease in the cell viability compared to that of either of single drug treated cancer cells. Combination of SRF+DHA resulted in predominant SubG1 proportion of cells. LD-SDN exhibited the highest SubG1 (%) of cells compared to that of any of the individual drugs. Most importantly, robust antitumor response and delayed tumor growth was observed for LD-SDN treated xenograft tumor model. Ki67 proliferation index of LD-SDN (22.1 ± 5.6%) is significantly lesser compared to that of either control (86.2 ± 6.9%) or SRF (75.4 ± 4.89%) or DHA (69.4 ± 6.9%). SIGNIFICANCES: These data provide strong evidence that LDL-mimetic lipid nanoformulations could be utilized as a biocompatible and tumor targeted platform for the delivery of multiple anticancer drugs in cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lipids/pharmacology , Liver Neoplasms/drug therapy , Nanoparticles , Receptors, LDL/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Artemisinins/administration & dosage , Cell Survival/drug effects , Drug Delivery Systems , Hep G2 Cells , Humans , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Reactive Oxygen Species/metabolism , Sorafenib/administration & dosage
8.
Arterioscler Thromb Vasc Biol ; 39(10): 2145-2156, 2019 10.
Article in English | MEDLINE | ID: mdl-31390883

ABSTRACT

OBJECTIVE: ApoC-III (apolipoprotein C-III) glycosylation can predict cardiovascular disease risk. Higher abundance of disialylated (apoC-III2) over monosialylated (apoC-III1) glycoforms is associated with lower plasma triglyceride levels. Yet, it remains unclear whether apoC-III glycosylation impacts TRL (triglyceride-rich lipoprotein) clearance and whether apoC-III antisense therapy (volanesorsen) affects distribution of apoC-III glycoforms. Approach and Results: To measure the abundance of human apoC-III glycoforms in plasma over time, human TRLs were injected into wild-type mice and mice lacking hepatic TRL clearance receptors, namely HSPGs (heparan sulfate proteoglycans) or both LDLR (low-density lipoprotein receptor) and LRP1 (LDLR-related protein 1). ApoC-III was more rapidly cleared in the absence of HSPG (t1/2=25.4 minutes) than in wild-type animals (t1/2=55.1 minutes). In contrast, deficiency of LDLR and LRP1 (t1/2=56.1 minutes) did not affect clearance of apoC-III. After injection, a significant increase in the relative abundance of apoC-III2 was observed in HSPG-deficient mice, whereas the opposite was observed in mice lacking LDLR and LRP1. In patients, abundance of plasma apoC-III glycoforms was assessed after placebo or volanesorsen administration. Volanesorsen treatment correlated with a statistically significant 1.4-fold increase in the relative abundance of apoC-III2 and a 15% decrease in that of apoC-III1. The decrease in relative apoC-III1 abundance was strongly correlated with decreased plasma triglyceride levels in patients. CONCLUSIONS: Our results indicate that HSPGs preferentially clear apoC-III2. In contrast, apoC-III1 is more effectively cleared by LDLR/LRP1. Clinically, the increase in the apoC-III2/apoC-III1 ratio on antisense lowering of apoC-III might reflect faster clearance of apoC-III1 because this metabolic shift associates with improved triglyceride levels.


Subject(s)
Apolipoprotein C-III/blood , Hypertriglyceridemia/drug therapy , Lipoproteins, HDL3/metabolism , Oligonucleotides/administration & dosage , Receptors, LDL/metabolism , Animals , Apolipoprotein C-III/drug effects , Disease Models, Animal , Glycosylation/drug effects , Humans , Hypertriglyceridemia/blood , Male , Mice , Molecular Targeted Therapy/methods , Receptors, LDL/drug effects , Reference Values
9.
Toxicol Appl Pharmacol ; 370: 170-177, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30914377

ABSTRACT

Proprotein convertase subtilisin/kexin type-9 (PCSK9) is most recognized serine protease for its role in cardiovascular diseases (CVD). PCSK9 regulates plasma low-density lipoprotein cholesterol (LDL-C) levels by selectively targeting hepatic LDL receptors (LDLR) for degradation, thereby serving as a potential therapeutic target for CVD. New pharmacological agents under development aim to lower the risk of CVD by inhibiting PCSK9 extracellularly, although secondary effects of this approach are not yet studied. Here we review the history of PCSK9 and rationale behind developing inhibitors for CVD. Importantly, we summarized the studies investigating the role and impact of modulated PCSK9 levels in inflammation, specifically in sepsis, rheumatoid arthritis and other chronic inflammatory conditions. Furthermore, we summarized studies that investigated the interactions of PCSK9 with pro-inflammatory pathways, such as scavenger receptor CD36 and thrombospondin 1 (TSP-1) in inflammatory diseases. This review highlights the conflicting role that PCSK9 plays in different inflammatory disease states and postulates that any unwanted effects of PCSK9 inhibition in early clinical testing should critically be examined.


Subject(s)
Inflammation/physiopathology , Proprotein Convertase 9/physiology , Antibodies, Monoclonal/pharmacology , Arthritis, Rheumatoid/physiopathology , CD36 Antigens , Cardiovascular Diseases/physiopathology , Cholesterol, LDL/blood , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation , Humans , Liver/chemistry , PCSK9 Inhibitors , Proprotein Convertase 9/genetics , RNA, Messenger/analysis , RNA, Messenger/drug effects , Receptors, LDL/drug effects , Receptors, LDL/physiology , Sepsis/physiopathology , Thrombospondin 1
10.
Eur J Nutr ; 58(3): 1283-1297, 2019 Apr.
Article in English | MEDLINE | ID: mdl-29516226

ABSTRACT

PURPOSE: Sugar-sweetened beverage intake is a risk factor for insulin resistance, dyslipidemia, fatty liver, and steatohepatitis (NASH). Sub-chronic supplementation of liquid fructose, but not glucose, in female rats increases liver and plasma triglycerides without inflammation. We hypothesized that chronic supplementation of fructose would cause NASH and liver insulin resistance. METHODS: We supplemented female Sprague-Dawley rats with water or either fructose or glucose 10% w/v solutions under isocaloric conditions for 7 months. At the end, plasma analytes, insulin, and adiponectin were determined, as well as liver triglyceride content and the expression of key genes controlling inflammation, fatty acid synthesis and oxidation, endoplasmic reticulum stress, and plasma VLDL clearance, by biochemical and histological methods. RESULTS: Although sugar-supplemented rats increased their energy intake by 50-60%, we found no manifestation of liver steatosis, fibrosis or necrosis, unchanged plasma or tissue markers of inflammation or fibrosis, and reduced liver expression of gluconeogenic enzymes, despite both sugars increased fatty acid synthesis, mTORC1, and IRE1 activity, while decreasing fatty acid oxidation and PPARα activity. Only fructose-supplemented rats were hypertriglyceridemic, showing a reduced expression of VLDL receptor and lipoprotein lipase in skeletal muscle and vWAT. Glucose-supplemented rats showed increased adiponectinemia, which would explain the different metabolic outcomes of the two sugars. CONCLUSIONS: Chronic liquid simple sugar supplementation, as the sole risk factor, is not enough for female rats to develop NASH and increased liver gluconeogenesis. Nevertheless, under isocaloric conditions, only fructose induced hypertriglyceridemia, thus confirming that also the type of nutrient matters in the development of metabolic diseases.


Subject(s)
Fatty Liver , Fructose/administration & dosage , Fructose/adverse effects , Hypertriglyceridemia/chemically induced , Inflammation , Receptors, LDL/drug effects , Animals , Disease Models, Animal , Female , Insulin Resistance , Liver/drug effects , Liver/metabolism , Rats , Rats, Sprague-Dawley , Triglycerides/metabolism
11.
Stroke ; 49(12): 3020-3029, 2018 12.
Article in English | MEDLINE | ID: mdl-30571407

ABSTRACT

Background and Purpose- Heme and iron are considered to be key factors responsible for secondary insults after intracerebral hemorrhage (ICH). Our previous study showed that LRP1 (low-density lipoprotein receptor-related protein-1)-Hx (hemopexin) facilitates removal of heme. The TLR7 (Toll-like receptor 7)-BTK (Bruton tyrosine kinase)-CRT (calreticulin) pathway regulates the expression of LRP1-Hx. This study is designed to clarify whether TLR7 activation facilitates heme scavenging and to establish the potential role of the BTK-CRT-LRP1-Hx signaling pathway in the pathophysiology of ICH. Methods- ICH was induced by stereotactic, intrastriatal injection of type VII collagenase. Mice received TLR7 agonist (imiquimod) via intraperitoneal injection after ICH induction. TLR7 inhibitor (ODN2088), BTK inhibitor (LFM-A13), and CRT agonist (thapsigargin) were given in different groups to further evaluate the underlying pathway. Mice were randomly divided into sham, ICH+vehicle (normal saline), ICH+Imiquimod (2.5, 5, and 10 µg/g), ICH+ODN2088, ICH+LFM-A13, ICH+thapsigargin, and ICH+ODN2088+thapsigargin. Imiquimod was administered twice daily starting at 6 hours after ICH; ODN2088 was administered by intracerebroventricular injection at 30 minutes, and LFM-A13 or thapsigargin was administered by intraperitoneal injection at 3 hours after ICH induction. Neurological scores, cognitive abilities, as well as brain edema, blood-brain barrier permeability, hemoglobin level, brain expression of TLR7/BTK/CRT/LRP1/Hx were analyzed. Results- Low dosage imiquimod significantly attenuated hematoma volume, brain edema, BBB permeability, and neurological deficits after ICH. Imiquimod also increased protein expressions of TLR7, BTK, CRT, LRP1, and Hx; ODN2088 reduced TLR7, BTK, CRT, LRP1, and Hx expressions. Conclusions- TLR7 plays an important role in heme scavenging after ICH by modulating the BTK-CRT-LRP1-Hx pathway. TLR7 may offer protective effects by promoting heme resolution and reduction of brain edema after ICH.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/metabolism , Brain/metabolism , Calreticulin/metabolism , Cerebral Hemorrhage/metabolism , Heme/metabolism , Hemopexin/metabolism , Membrane Glycoproteins/metabolism , Receptors, LDL/metabolism , Toll-Like Receptor 7/metabolism , Tumor Suppressor Proteins/metabolism , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/drug effects , Amides/pharmacology , Animals , Blood-Brain Barrier/drug effects , Brain/drug effects , Brain Edema/metabolism , Calreticulin/agonists , Calreticulin/drug effects , Enzyme Inhibitors/pharmacology , Hemopexin/drug effects , Imiquimod/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1 , Membrane Glycoproteins/agonists , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/drug effects , Mice , Nitriles/pharmacology , Oligodeoxyribonucleotides/pharmacology , Receptors, LDL/drug effects , Signal Transduction , Thapsigargin/pharmacology , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 7/drug effects , Tumor Suppressor Proteins/drug effects
12.
Nat Rev Endocrinol ; 15(1): 52-62, 2018 12.
Article in English | MEDLINE | ID: mdl-30367179

ABSTRACT

Clinical trials have unequivocally shown that inhibition of proprotein convertase subtilisin/kexin type 9 (PCSK9) efficaciously and safely prevents cardiovascular events by lowering levels of LDL cholesterol. PCSK9 in the circulation is derived mainly from the liver, but the protein is also expressed in the pancreas, the kidney, the intestine and the central nervous system. Although PCSK9 modulates cholesterol metabolism by regulating LDL receptor expression in the liver, in vitro and in vivo studies have suggested that PCSK9 is involved in various other physiological processes. Although therapeutic PCSK9 inhibition could theoretically have undesired effects by interfering with these non-cholesterol-related processes, studies of individuals with genetically determined reduced PCSK9 function and clinical trials of PCSK9 inhibitors have not revealed clinically meaningful adverse consequences of almost completely eradicating PCSK9 from the circulation. The clinical implications of PCSK9 functions beyond lipid metabolism in terms of wanted or unwanted effects of therapeutic PCSK9 inhibition therefore appear to be limited. The objective of this Review is to describe the physiological role of PCSK9 beyond the LDL receptor to provide a rational basis for monitoring the effects of PCSK9 inhibition as these drugs gain traction in the clinic.


Subject(s)
Cholesterol, LDL/metabolism , Lipid Metabolism/drug effects , Proprotein Convertase 9/drug effects , Proprotein Convertase 9/metabolism , Receptors, LDL/drug effects , Biomarkers/metabolism , Cardiovascular Diseases/prevention & control , Cholesterol, LDL/drug effects , Female , Humans , Hypercholesterolemia/drug therapy , Hypercholesterolemia/prevention & control , Male , Receptors, LDL/metabolism , Sensitivity and Specificity
13.
Toxicol Lett ; 296: 173-183, 2018 Oct 15.
Article in English | MEDLINE | ID: mdl-29908845

ABSTRACT

Lead exposure has been evidenced as a risk factor for Alzheimer's disease (AD), mainly affecting the ageing. However, the early manifestation and mechanisms of AD-like pathology induced by lead exposure remains to be elucidated. Considering the fact that impaired cholesterol metabolism is associated with many neurodegenerative disorders including AD, in this study we focused on the role of cholesterol metabolism in lead induced premature AD-like pathology. We treated weaning rats with lead at different concentrations for 4 weeks. We found that developmental lead exposure increased amyloid-beta (Aß) accumulation and amyloid plaque deposition in the cortex and hippocampus. Lead exposure increased amyloid precursor protein (APP) expression and activated the sterol regulatory element binding protein 2 (SREBP2)-beta secretase (BACE1) pathway. In addition, we found that lead exposure decreased cholesterol levels by upregulating the expression of liver X receptor-a (LXR-a) and ATP-binding cassette transporter protein family member A1 (ABCA1) and decreasing the expression of 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CR) and low density lipoprotein receptor (LDL-R) in young rat brain tissues. Taken together, our data demonstrated that developmental lead exposure induced early manifestation of AD-like pathology and disturbed cholesterol metabolism in young rat brains.


Subject(s)
Alzheimer Disease/chemically induced , Alzheimer Disease/pathology , Brain/pathology , Cholesterol/metabolism , Lead Poisoning, Nervous System/pathology , Lead/toxicity , Amyloid Precursor Protein Secretases/drug effects , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/drug effects , Brain/drug effects , Brain/metabolism , Brain Chemistry/drug effects , Lead/blood , Male , Organ Size/drug effects , Plaque, Amyloid/chemically induced , Plaque, Amyloid/pathology , Rats , Rats, Sprague-Dawley , Receptors, LDL/drug effects , Receptors, LDL/metabolism , Signal Transduction/drug effects , Sterol Regulatory Element Binding Proteins/drug effects
14.
Drugs ; 78(8): 799-808, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29737499

ABSTRACT

Hypercholesterolaemia is frequently observed in patients with cardiovascular diseases (CVD) and is associated with increased mortality. Statin treatment has been the standard of care for reducing low-density lipoprotein cholesterol (LDL-C) to improve cardiovascular outcomes. However, statins have limited effects in some patients and may be discontinued due to adverse effects resulting in LDL-C above target levels. The proprotein convertase subtilisin kexin type 9 (PCSK9) is a pivotal regulator in the LDL-C metabolism by degrading the LDL-C receptor on hepatocytes. Inhibition of PCSK9 by monoclonal antibodies (mAb) significantly lowers LDL-C levels and is considered to reduce the likelihood of adverse cardiac events. However, such treatment regimens are not cost-effective, and require frequent administrations at high doses that may be associated with side effects and poor drug adherence. Furthermore, it has been shown that these PCSK9 medicines may trigger the formation of antidrug antibodies followed by a significant attenuation of the LDL-C-lowering effect. Active vaccination inducing high-affinity antibodies against PCSK9 with less frequent administration intervals may be a novel promising therapeutic approach to overcome the drawback of passive immunization with PCSK9 mAb. However there is a paucity of available clinical safety and efficacy data. This article discusses challenges in the development of PCSK9 vaccines and their potential therapeutic benefits by reviewing clinical studies that evaluated the safety and efficacy of PCSK9 mAb.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Hypercholesterolemia/therapy , Hypolipidemic Agents/therapeutic use , Molecular Targeted Therapy/methods , Proprotein Convertase 9/immunology , Vaccines/therapeutic use , Cholesterol, LDL/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Immunization, Passive , Receptors, LDL/drug effects , Signal Transduction , Treatment Outcome
15.
Cardiovasc Diabetol ; 17(1): 48, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29618348

ABSTRACT

BACKGROUND: Proprotein convertase subtilisin/kexin type 9 (PCSK9), a major regulator of cholesterol homeostasis, is associated with glucose metabolism. Liraglutide, a glucagon-like peptide-1 receptor agonist, can increase insulin secretion in a glucose-dependent manner and lower blood glucose. We aimed to investigate the relationship between liraglutide and PCSK9. METHODS: At the cellular level, the expressions of PCSK9 and hepatocyte nuclear factor 1 alpha (HNF1α) protein in HepG2 cells stimulated by liraglutide was examined using Western blot. Seven-week old db/db mice and wild type (WT) mice were administered either liraglutide (200 µg/kg) or equivoluminal saline subcutaneously, twice daily for 7 weeks. Fasting glucose level, food intake and body weight were measured every week. After the 7-week treatment, the blood was collected for lipid and PCSK9 levels detection and the liver was removed from the mice for oil red O staining, immunohistochemical analysis, immunofluorescence test and Western bolt. RESULTS: Firstly, liraglutide suppressed both PCSK9 and HNF1α expression in HepG2 cells in a time and concentration dependent manner. Secondly, liraglutide induced weight loss in WT and db/db mice, decreased serum PCSK9, glucose and lipid levels and improved hepatic accumulation in db/db but not WT mice. Thirdly, liraglutide reduced both hepatic PCSK9 and low-density lipoprotein receptor (LDLR) expression with a decrease in HNF1α in db/db mice but not in WT mice. CONCLUSIONS: Liraglutide suppressed PCSK9 expression through HNF1α-dependent mechanism in HepG2 cells and db/db mice, and decreased LDLR possibly via PCSK9-independent pathways in db/db mice.


Subject(s)
Diabetes Mellitus/drug therapy , Hepatocyte Nuclear Factor 1-alpha/metabolism , Hepatocytes/drug effects , Hypoglycemic Agents/pharmacology , Incretins/pharmacology , Liraglutide/pharmacology , Proprotein Convertase 9/metabolism , Receptors, LDL/drug effects , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus/blood , Diabetes Mellitus/enzymology , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation , Hep G2 Cells , Hepatocytes/enzymology , Humans , Lipids/blood , Male , Mice , Receptors, LDL/metabolism , Signal Transduction/drug effects , Time Factors
16.
Brain Behav Immun ; 70: 36-47, 2018 05.
Article in English | MEDLINE | ID: mdl-29545118

ABSTRACT

Epidemiological evidence suggests that people with bipolar disorder prescribed lithium exhibit a lower risk of Alzheimer's disease (AD) relative to those prescribed other mood-stabilizing medicines. Lithium chloride (LiCl) reduces brain ß-amyloid (Aß) levels, and the brain clearance of Aß is reduced in AD. Therefore, the purpose of this study was to assess whether the cognitive benefits of LiCl are associated with enhanced brain clearance of exogenously-administered Aß. The brain clearance of intracerebroventricularly (icv) administered 125I-Aß42 was assessed in male Swiss outbred mice administered daily oral NaCl or LiCl (300 mg/kg for 21 days). LiCl exhibited a 31% increase in the brain clearance of 125I-Aß42 over 10 min, which was associated with a 1.6-fold increase in brain microvascular expression of the blood-brain barrier efflux transporter low density lipoprotein receptor-related protein 1 (LRP1) and increased cerebrospinal fluid (CSF) bulk-flow. 8-month-old female wild type (WT) and APP/PS1 mice were also administered daily NaCl or LiCl for 21 days, which was followed by cognitive assessment by novel object recognition and water maze, and measurement of soluble Aß42, plaque-associated Aß42, and brain efflux of 125I-Aß42. LiCl treatment restored the long-term spatial memory deficit observed in APP/PS1 mice as assessed by the water maze (back to similar levels of escape latency as WT mice), but the short-term memory deficit remained unaffected by LiCl treatment. While LiCl did not affect plaque-associated Aß42, soluble Aß42 levels were reduced by 49.9% in APP/PS1 mice receiving LiCl. The brain clearance of 125I-Aß42 decreased by 27.8% in APP/PS1 mice, relative to WT mice, however, LiCl treatment restored brain 125I-Aß42 clearance in APP/PS1 mice to a rate similar to that observed in WT mice. These findings suggest that the cognitive benefits and brain Aß42 lowering effects of LiCl are associated with enhanced brain clearance of Aß42, possibly via brain microvascular LRP1 upregulation and increased CSF bulk-flow, identifying a novel mechanism of protection by LiCl for the treatment of AD.


Subject(s)
Amyloid beta-Peptides/drug effects , Cognition/drug effects , Lithium Chloride/therapeutic use , Alzheimer Disease , Amyloid beta-Protein Precursor , Animals , Blood-Brain Barrier/drug effects , Brain , Disease Models, Animal , Lithium Chloride/pharmacology , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Memory/drug effects , Mice , Mice, Transgenic , Plaque, Amyloid , Presenilin-1 , Receptors, LDL/drug effects , Receptors, LDL/physiology , Tumor Suppressor Proteins/drug effects , Tumor Suppressor Proteins/physiology
17.
Mol Nutr Food Res ; 62(1)2018 01.
Article in English | MEDLINE | ID: mdl-28799714

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a major health issue throughout the world. However, no validated treatments for NAFLD are currently available. In-depth studies have demonstrated the efficacy of (-)-epigallocatechin-3-gallate (EGCG), a main bioactive chemical extracted from green tea, in treating NAFLD. EGCG exhibits multi-pronged preventive and therapeutic activities, including promoting lipid and glucose metabolism, anti-lipid peroxidation and anti-inflammation activities, anti-fibrosis, and anti-NAFLD related tumor, thus contributing to the mitigation of NAFLD occurrence and progression. The objectives of this paper are to review and discuss the currently known targets, signaling pathways and roles of EGCG that interfere with NAFLD pathogenesis, then providing additional experimental evidence and the foundation for the further studies and clinical applications of EGCG in the prevention and treatment of NAFLD.


Subject(s)
Catechin/analogs & derivatives , Non-alcoholic Fatty Liver Disease/drug therapy , AMP-Activated Protein Kinases/metabolism , Animals , Antioxidants/pharmacology , Autophagy/drug effects , Body Weight/drug effects , Catechin/pharmacology , Catechin/therapeutic use , Humans , Lipid Metabolism/drug effects , Non-alcoholic Fatty Liver Disease/etiology , Receptors, LDL/drug effects
18.
Eur J Nutr ; 57(1): 405-415, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28718016

ABSTRACT

PURPOSES: We previously showed that polyphenol-rich blackcurrant extract (BCE) showed a hypocholesterolemic effect in mice fed a high fat diet. As direct cholesterol removal from the body via the intestine has been recently appreciated, we investigated the effect of BCE on the modulation of genes involved in intestinal cholesterol transport using Caco-2 cells as an in vitro model. METHODS: Caco-2 cells were treated with BCE to determine its effects on mRNA and protein expression of genes important for intestinal cholesterol transport, low-density lipoprotein (LDL) uptake, cellular cholesterol content, and cholesterol transport from basolateral to apical membrane of Caco-2 cell monolayers. Cells were also treated with anthocyanin-rich or -poor fraction of BCE to determine the role of anthocyanin on BCE effects. RESULTS: BCE significantly increased protein levels of LDL receptor (LDLR) without altering its mRNA, which consequently increased LDL uptake into Caco-2 cells. This post-transcriptional induction of LDLR by BCE was markedly attenuated in the presence of rapamycin, an inhibitor of mechanistic target of rapamycin complex 1 (mTORC1). In addition, BCE altered genes involved in cholesterol transport in the enterocytes, including apical and basolateral cholesterol transporters, in such a way that could enhance cholesterol flux from the basolateral to apical side of the enterocytes. Indeed, BCE significantly increased the flux of LDL-derived cholesterol from the basolateral to the apical chamber of Caco-2 monolayer. LDLR protein levels were markedly increased by anthocyanin-rich fraction, but not by anthocyanin-free fraction. CONCLUSION: mTORC1-dependent post-transcriptional induction of LDLR by BCE anthocyanins drove the transport of LDL-derived cholesterol to the apical side of the enterocytes. This may represent a potential mechanism for the hypocholesterolemic effect of BCE.


Subject(s)
Anthocyanins/pharmacology , Cholesterol/metabolism , Fruit/chemistry , Plant Extracts/pharmacology , Receptors, LDL/genetics , Ribes , Biological Transport/drug effects , Biological Transport/genetics , Caco-2 Cells , Cholesterol, LDL/metabolism , Enterocytes/metabolism , Gene Expression/drug effects , Humans , Mechanistic Target of Rapamycin Complex 1/antagonists & inhibitors , Mechanistic Target of Rapamycin Complex 1/physiology , RNA, Messenger/analysis , Receptors, LDL/analysis , Receptors, LDL/drug effects , Sirolimus/pharmacology , Transcription, Genetic/drug effects
19.
Biosci Biotechnol Biochem ; 82(1): 57-64, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29191087

ABSTRACT

Alzheimer's disease (AD) is the most common cause of dementia among elderly population. Deranged ß-amyloid (Aß) trafficking across the blood-brain barrier is known to be a critical element in the pathogenesis of AD. In the vascular endothelial cells of hippocampus, Aß transport is mainly mediated by low-density lipoprotein-associated protein 1 (LRP1) and the receptor for advanced glycation end (RAGE) products; therefore, LRP1 and RAGE endothelial cells are potential therapeutic targets for AD. In this study, we explored the effects of Formononetin (FMN) on learning and memory improvement in APP/PS1 mice and the related mechanisms. We found that FMN significantly improved learning and memory ability by suppressing Aß production from APP processing, RAGE-dependent inflammatory signaling and promoted LRP1-dependent cerebral Aß clearance pathway. Moreover, FMN treatment alleviated ultrastructural changes in hippocampal vascular endothelial cells. In conclusion, we believe that FMN may be an efficacious and promising treatment for AD.


Subject(s)
Alzheimer Disease/drug therapy , Drug Delivery Systems , Isoflavones/therapeutic use , Neuroprotective Agents , Animals , Humans , Low Density Lipoprotein Receptor-Related Protein-1 , Mice , Neuroprotective Agents/therapeutic use , Receptor for Advanced Glycation End Products/drug effects , Receptors, LDL/drug effects , Tumor Suppressor Proteins/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL