Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Cell Mol Life Sci ; 80(2): 42, 2023 Jan 16.
Article in English | MEDLINE | ID: mdl-36645496

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) play vital roles in normal brain functions (i.e., learning, memory, and neuronal development) and various neuropathological conditions, such as epilepsy, autism, Parkinson's disease, Alzheimer's disease, and traumatic brain injury. Endogenous neuroactive steroids such as 24(S)-hydroxycholesterol (24(S)-HC) have been shown to influence NMDAR activity, and positive allosteric modulators (PAMs) derived from 24(S)-hydroxycholesterol scaffold can also enhance NMDAR function. This study describes the structural determinants and mechanism of action for 24(S)-hydroxycholesterol and two novel synthetic analogs (SGE-550 and SGE-301) on NMDAR function. We also show that these agents can mitigate the altered function caused by a set of loss-of-function missense variants in NMDAR GluN subunit-encoding GRIN genes associated with neurological and neuropsychiatric disorders. We anticipate that the evaluation of novel neuroactive steroid NMDAR PAMs may catalyze the development of new treatment strategies for GRIN-related neuropsychiatric conditions.


Subject(s)
Alzheimer Disease , Nervous System Diseases , Neurosteroids , Humans , Receptors, N-Methyl-D-Aspartate/metabolism , Neurosteroids/pharmacology , Neurosteroids/therapeutic use , Hydroxycholesterols/pharmacology , Hydroxycholesterols/therapeutic use , Nervous System Diseases/drug therapy , Nervous System Diseases/genetics , Alzheimer Disease/drug therapy , Steroids/pharmacology , Allosteric Regulation/physiology
2.
mBio ; 12(6): e0290721, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34781692

ABSTRACT

Oncogenic gammaherpesviruses express viral products during latent and lytic infection that block the innate immune response. Previously, we found that Kaposi's sarcoma herpesvirus (KSHV/human herpesvirus-8) viral microRNAs (miRNAs) downregulate cholesterol biogenesis, and we hypothesized that this prevents the production of 25-hydroxycholesterol (25HC), a cholesterol derivative. 25HC blocks KSHV de novo infection of primary endothelial cells at a postentry step and decreases viral gene expression of LANA (latency-associated nuclear antigen) and RTA. Herein we expanded on this observation by determining transcriptomic changes associated with 25HC treatment of primary endothelial cells using RNA sequencing (RNA-Seq). We found that 25HC treatment inhibited KSHV gene expression and induced interferon-stimulated genes (ISGs) and several inflammatory cytokines (interleukin 8 [IL-8], IL-1α). Some 25HC-induced genes were partially responsible for the broadly antiviral effect of 25HC against several viruses. Additionally, we found that 25HC inhibited infection of primary B cells by a related oncogenic virus, Epstein-Barr virus (EBV/human herpesvirus-4) by suppressing key viral genes such as LMP-1 and inducing apoptosis. RNA-Seq analysis revealed that IL-1 and IL-8 pathways were induced by 25HC in both primary endothelial cells and B cells. We also found that the gene encoding cholesterol 25-hydroxylase (CH25H), which converts cholesterol to 25HC, can be induced by type I interferon (IFN) in human B cell-enriched peripheral blood mononuclear cells (PBMCs). We propose a model wherein viral miRNAs target the cholesterol pathway to prevent 25HC production and subsequent induction of antiviral ISGs. Together, these results answer some important questions about a widely acting antiviral (25HC), with implications for multiple viral and bacterial infections. IMPORTANCE A cholesterol derivative, 25-hydroxycholesterol (25HC), has been demonstrated to inhibit infections from widely different bacteria and viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, its mechanism of activity is still not fully understood. In this work, we look at gene expression changes in the host and virus after 25HC treatment to find clues about its antiviral activity. We likewise demonstrate that 25HC is also antiviral against EBV, a common cancer-causing virus. We compared our results with previous data from antiviral screening assays and found the same pathways resulting in antiviral activity. Together, these results bring us closer to understanding how a modified form of cholesterol works against several viruses.


Subject(s)
Cytokines/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/drug effects , Herpesvirus 8, Human/drug effects , Hydroxycholesterols/pharmacology , Hydroxycholesterols/therapeutic use , Inflammation/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/virology , Cells, Cultured , Cytokines/genetics , Endothelial Cells/drug effects , Endothelial Cells/immunology , Endothelial Cells/virology , Epstein-Barr Virus Infections/drug therapy , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/genetics , Humans , Hydroxycholesterols/immunology , Sequence Analysis, RNA , Virus Latency , Virus Replication
3.
Int J Mol Sci ; 22(21)2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34769299

ABSTRACT

The COVID-19 pandemic caused by SARS-CoV-2 infection poses a serious threat to global public health and the economy. The enzymatic product of cholesterol 25-hydroxylase (CH25H), 25-Hydroxycholesterol (25-HC), was reported to have potent anti-SARS-CoV-2 activity. Here, we found that the combination of 25-HC with EK1 peptide, a pan-coronavirus (CoV) fusion inhibitor, showed a synergistic antiviral activity. We then used the method of 25-HC modification to design and synthesize a series of 25-HC-modified peptides and found that a 25-HC-modified EK1 peptide (EK1P4HC) was highly effective against infections caused by SARS-CoV-2, its variants of concern (VOCs), and other human CoVs, such as HCoV-OC43 and HCoV-229E. EK1P4HC could protect newborn mice from lethal HCoV-OC43 infection, suggesting that conjugation of 25-HC with a peptide-based viral inhibitor was a feasible and universal strategy to improve its antiviral activity.


Subject(s)
Antiviral Agents/pharmacology , Hydroxycholesterols/chemistry , Lipopeptides/chemistry , SARS-CoV-2/drug effects , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Body Weight/drug effects , COVID-19/virology , Coronavirus 229E, Human/drug effects , Coronavirus 229E, Human/pathogenicity , Coronavirus Infections/drug therapy , Coronavirus Infections/mortality , Coronavirus Infections/virology , Coronavirus OC43, Human/drug effects , Coronavirus OC43, Human/pathogenicity , Disease Models, Animal , Drug Synergism , Humans , Hydroxycholesterols/pharmacology , Hydroxycholesterols/therapeutic use , Lipopeptides/pharmacology , Lipopeptides/therapeutic use , Mice , Mice, Inbred BALB C , Polyethylene Glycols/chemistry , SARS-CoV-2/isolation & purification , SARS-CoV-2/physiology , Survival Rate , Virus Internalization/drug effects , COVID-19 Drug Treatment
4.
Cells ; 10(11)2021 11 05.
Article in English | MEDLINE | ID: mdl-34831255

ABSTRACT

Acetaminophen (APAP) overdose is one of the most frequent causes of acute liver failure (ALF). N-acetylcysteine (NAC) is currently being used as part of the standard care in the clinic but its usage has been limited in severe cases, in which liver transplantation becomes the only treatment option. Therefore, there still is a need for a specific and effective therapy for APAP induced ALF. In the current study, we have demonstrated that treatment with 25-Hydroxycholesterol 3-Sulfate (25HC3S) not only significantly reduced mortality but also decreased the plasma levels of liver injury markers, including LDH, AST, and ALT, in APAP overdosed mouse models. 25HC3S also decreased the expression of those genes involved in cell apoptosis, stabilized mitochondrial polarization, and significantly decreased the levels of oxidants, malondialdehyde (MDA), and reactive oxygen species (ROS). Whole genome bisulfite sequencing analysis showed that 25HC3S increased demethylation of 5mCpG in key promoter regions and thereby increased the expression of those genes involved in MAPK-ERK and PI3K-Akt signaling pathways. We concluded that 25HC3S may alleviate APAP induced liver injury via up-regulating the master signaling pathways and maintaining mitochondrial membrane polarization. The results suggest that 25HC3S treatment facilitates the recovery and significantly decreases the mortality of APAP induced acute liver injury and has a synergistic effect with NAC in propylene glycol (PG) for the injury.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Cholesterol Esters/therapeutic use , Hydroxycholesterols/therapeutic use , Mitochondria/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/physiopathology , Cholesterol Esters/pharmacology , CpG Islands/genetics , DNA Demethylation , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Hydroxycholesterols/pharmacology , Liver/drug effects , Liver/injuries , Liver/metabolism , Liver/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred C57BL , Mitochondria/drug effects , Models, Biological , Organ Specificity/drug effects , Oxidants/metabolism
5.
Brain ; 143(9): 2709-2720, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32830245

ABSTRACT

Anti-N-methyl-d-aspartate receptor (NMDAR) encephalitis is an immune-mediated disease characterized by a complex neuropsychiatric syndrome in association with an antibody-mediated decrease of NMDAR. About 85% of patients respond to immunotherapy (and removal of an associated tumour if it applies), but it often takes several months or more than 1 year for patients to recover. There are no complementary treatments, beyond immunotherapy, to accelerate this recovery. Previous studies showed that SGE-301, a synthetic analogue of 24(S)-hydroxycholesterol, which is a potent and selective positive allosteric modulator of NMDAR, reverted the memory deficit caused by phencyclidine (a non-competitive antagonist of NMDAR), and prevented the NMDAR dysfunction caused by patients' NMDAR antibodies in cultured neurons. An advantage of SGE-301 is that it is optimized for systemic delivery such that plasma and brain exposures are sufficient to modulate NMDAR activity. Here, we used SGE-301 to confirm that in cultured neurons it prevented the antibody-mediated reduction of receptors, and then we applied it to a previously reported mouse model of passive cerebroventricular transfer of patient's CSF antibodies. Four groups were established: mice receiving continuous (14-day) infusion of patients' or controls' CSF, treated with daily subcutaneous administration of SGE-301 or vehicle (no drug). The effects on memory were examined with the novel object location test at different time points, and the effects on synaptic levels of NMDAR (assessed with confocal microscopy) and plasticity (long-term potentiation) were examined in the hippocampus on Day 18, which in this model corresponds to the last day of maximal clinical and synaptic alterations. As expected, mice infused with patient's CSF antibodies, but not those infused with controls' CSF, and treated with vehicle developed severe memory deficit without locomotor alteration, accompanied by a decrease of NMDAR clusters and impairment of long-term potentiation. All antibody-mediated pathogenic effects (memory, synaptic NMDAR, long-term potentiation) were prevented in the animals treated with SGE-301, despite this compound not antagonizing antibody binding. Additional investigations on the potential mechanisms related to these SGE-301 effects showed that (i) in cultured neurons SGE-301 prolonged the decay time of NMDAR-dependent spontaneous excitatory postsynaptic currents suggesting a prolonged open time of the channel; and (ii) it significantly decreased, without fully preventing, the internalization of antibody-bound receptors suggesting that additional, yet unclear mechanisms, contribute in keeping unchanged the surface NMDAR density. Overall, these findings suggest that SGE-301, or similar NMDAR modulators, could potentially serve as complementary treatment for anti-NMDAR encephalitis and deserve future investigations.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis/metabolism , Anti-N-Methyl-D-Aspartate Receptor Encephalitis/therapy , Autoantibodies/administration & dosage , Autoantibodies/cerebrospinal fluid , Receptors, N-Methyl-D-Aspartate/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Animals , Cells, Cultured , HEK293 Cells , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Hydroxycholesterols/chemistry , Hydroxycholesterols/pharmacology , Hydroxycholesterols/therapeutic use , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques
6.
J Pharmacol Exp Ther ; 374(1): 142-150, 2020 07.
Article in English | MEDLINE | ID: mdl-32341017

ABSTRACT

Nonalcoholic fatty liver disease is a chronic inflammatory liver disease. It is associated with obesity and type 2 diabetes. Oxycholesterols are metabolites of cholesterol, and several of them can act on the G protein-coupled receptor, G protein-coupled receptor 183 (GPR183)/Epstein-Barr virus-induced gene 2. We found expression of GPR183 in human hepatoma cell lines and in vivo induction of GPR183 expression in mouse livers after high-fat diet feeding. Therefore, the role of oxycholesterols and GPR183 in hepatocytes was studied using a model of hepatic steatosis induced by liver X receptor (LXR) activation. LXR activation by T0901317 resulted in fat accumulation in Hep3B human hepatoma cells. This lipid accumulation was inhibited by 7α,25-dihydroxycholesterol, the most potent agonist of GPR183. The protective effects of 7α,25-dihydroxycholesterol were suppressed by a specific GPR183 antagonist, NIBR189 [(2E)-3-(4-Bromophenyl)-1-[4-4-methoxybenzoyl)-1-piperazinyl]-2-propene-1-one]. T0901317 treatment induced expression of the major transcription factor for lipogenesis, sterol regulatory element-binding protein 1c (SREBP-1c). 7α,25-Dihydroxycholesterol inhibited the induction of SREBP-1c proteins in a GPR183-dependent manner. Using inhibitors specific for intracellular signaling molecules, 7α,25-dihydroxycholesterol-induced suppression of hepatocellular steatosis was shown to be mediated through Gi/o proteins, p38 mitogen-activated protein kinases, phosphoinositide 3-kinase, and AMP-activated protein kinase. In addition, the inhibitory effect of 7α,25-dihydroxycholesterol was validated in HepG2 cells and primary mouse hepatocytes. Therefore, the present report suggests that 7α,25-dihydroxycholesterol-GPR183 signaling may suppress hepatocellular steatosis in the liver. SIGNIFICANCE STATEMENT: Oxycholesterols, which are metabolites of cholesterol, act on the G protein-coupled receptor, G protein-coupled receptor 183 (GPR183)/Epstein-Barr virus-induced gene 2, which is expressed in human hepatoma cell lines, and its expression is induced in vivo in mouse livers after high-fat diet feeding. Activation of GPR183 inhibits fat accumulation in primary mouse hepatocytes and HepG2 cells through Gi/o proteins, p38 mitogen-activated protein kinases, phosphoinositide 3-kinase, and AMP-activated protein kinase.


Subject(s)
Hepatocytes/drug effects , Hepatocytes/metabolism , Hydroxycholesterols/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Receptors, G-Protein-Coupled/metabolism , Adipose Tissue/drug effects , Adipose Tissue/pathology , Animals , Cell Line, Tumor , Diet, High-Fat/adverse effects , Gene Expression Regulation/drug effects , Hepatocytes/pathology , Humans , Hydroxycholesterols/therapeutic use , Liver X Receptors/metabolism , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism
8.
Anticancer Res ; 40(2): 779-788, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32014920

ABSTRACT

BACKGROUND/AIM: Oxysterol plays important physiological roles in diverse biological processes including apoptosis. However, the mechanisms underlying oxysterol-induced apoptosis remain unknown. 25-hydroxycholesterol (25-HC) is an oxysterol synthesized by cholesterol 25-hydroxylase from cholesterol during sterol metabolism. The aim of present study was to investigate 25-HC-induced apoptosis and associated signalling pathways in FaDu cells, which is originated form human head and neck squamous cell carcinoma cells. MATERIALS AND METHODS: 25-HC-induced apoptosis was investigated by cell cytotoxicity assay using MTT, cell viability assay using cell LIVE/DEAD cell viability assay, haematoxylin & eosin staining, nuclear staining, fluorescence-activated cell sorting, western blotting using specific antibodies associated with extrinsic and intrinsic apoptosis pathways, and caspase-3/-7 activity assay in FaDu cells. RESULTS: 25-HC dose-dependently decreased the viability of FaDu cells and up-regulated apoptotic events, such as alteration in morphology, and nuclear condensation. Flow cytometric analysis showed an increase in apoptotic population upon 25-HC treatment, suggesting that 25-HC induces apoptosis in FaDu cells. Moreover, 25-HC-induced apoptosis in FaDu cells was dependent on the activation of caspases by Fas antigen ligand-triggered death receptor-mediated extrinsic pathway and mitochondria-dependent intrinsic pathway via mitogen activated protein kinases. CONCLUSION: Cholesterol-derived oxysterol, 25-HC has potential anti-cancer function in FaDu cells and may have potential properties for the discovery of anti-cancer agents.


Subject(s)
Apoptosis/drug effects , Hydroxycholesterols/therapeutic use , Squamous Cell Carcinoma of Head and Neck/drug therapy , Cell Line, Tumor , Humans , Hydroxycholesterols/pharmacology , Squamous Cell Carcinoma of Head and Neck/pathology
9.
Int J Biol Sci ; 16(2): 298-308, 2020.
Article in English | MEDLINE | ID: mdl-31929757

ABSTRACT

Myocardial ischemia-reperfusion (IR) injury occurs when occlusive coronary artery restores blood supply after events such as myocardial infarction, stroke, cardiac arrest and resuscitation, and organ transplantation. However, the mechanisms involved are poorly understood, and effective pharmacological interventions are still lacking. A previous study demonstrated that 25-hydroxycholesterol (25-HC) contributed to lipid metabolism and cholesterol metabolism as an oxysterol molecule. We herein explored whether 25-hydroxycholesterol (25-HC) has cardioprotective properties against IR injury and explored its underlying mechanisms. 25-HC was administered before reperfusion procedure in IR injury model mice. We found that 25-HC significantly reduced the IR-induced infarct size and improved cardiac function, and this protective effect was associated with reduced phosphorylation of p38-MAPK and JNK1/2. Besides, 25-HC also inhibited the Bax/Bcl-2 ratio and the relative expression of cleaved caspase-3. Furthermore, 25-HC decreased the PARP activity, indicating that 25-HC ameliorates IR injury via the PARP pathway. The 25-HC group abolished cardioprotection in the presence of little PARP activity, suggesting that the PARP activity is essential for 25-HC to exert its effect during IR injury. Our primary study indicates that 25-HC ameliorated IR injury by inhibiting the PARP activity and decreasing myocardial apoptosis, which makes it a potential therapeutic drug in IR injury of the heart.


Subject(s)
Hydroxycholesterols/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cells, Cultured , Echocardiography , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Oxidative Stress/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Curr Opin Ophthalmol ; 31(1): 61-66, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31770163

ABSTRACT

PURPOSE OF REVIEW: Age-related cataract occurs when crystallin proteins in the lens partially unfold and subsequently aggregate. Physicians and traditional healers alike have been exploring pharmacologic cataract treatment for hundreds of years. Currently, surgery is the only effective treatment. However, there are an abundance of homeopathic and alternative remedies that have been suggested as treatment for cataract. This article reviews the current understanding of cataract development and discusses several homeopathic remedies purported to treat age-related cataract. Additionally, we will present an overview of evidence regarding the development of pharmacologic cataract reversal therapies. RECENT FINDINGS: Some homeopathic therapies have been shown to prevent cataract development in experimental models. More studies are required to elucidate the potential medicinal and toxic properties of the various alternative therapies. However, in recent years, scientists have begun to investigate substances that address cataract by reversing lens protein aggregation. One such compound, lanosterol, was reported to reverse cataract opacity in vitro and in animal models. Subsequently, 25-hydroxycholesterol and rosmarinic acid were identified as having similar properties. SUMMARY: Although challenges and uncertainties remain, further research has the potential to lead to the development of a nonsurgical therapeutic option for age-related cataract.


Subject(s)
Cataract/therapy , Materia Medica , Animals , Antioxidants/therapeutic use , Cinnamates/therapeutic use , Depsides/therapeutic use , Humans , Hydroxycholesterols/therapeutic use , Rosmarinic Acid
11.
Sci Rep ; 8(1): 9341, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29921877

ABSTRACT

Cataract, the leading cause of vision impairment worldwide, arises from abnormal aggregation of crystallin lens proteins. Presently, surgical removal is the only therapeutic approach. Recent findings have triggered renewed interest in development of non-surgical treatment alternatives. However, emerging treatments are yet to achieve full and consistent lens clearance. Here, the first ex vivo assay to screen for drug candidates that reduce human lenticular protein aggregation was developed. This assay allowed the identification of two leading compounds as facilitating the restoration of nearly-complete transparency of phacoemulsified cataractous preparation ex vivo. Mechanistic studies demonstrated that both compounds reduce cataract microparticle size and modify their amyloid-like features. In vivo studies confirmed that the lead compound, rosmarinic acid, delays cataract formation and reduces the severity of lens opacification in model rats. Thus, the ex vivo assay may provide an initial platform for broad screening of potential novel therapeutic agents towards pharmacological treatment of cataract.


Subject(s)
Cataract/drug therapy , Cataract/pathology , Aged , Aged, 80 and over , Animals , Cinnamates/therapeutic use , Crystallins/metabolism , Depsides/therapeutic use , Doxycycline/therapeutic use , Female , Humans , Hydroxycholesterols/therapeutic use , Male , Rats , Rats, Wistar , Rosmarinic Acid
12.
Metabolism ; 71: 83-93, 2017 06.
Article in English | MEDLINE | ID: mdl-28521882

ABSTRACT

BACKGROUND: Oxysterol sulfation plays a fundamental role in the regulation of many biological events. Its products, 25-hydroxycholesterol 3-sulfate (25HC3S) and 25-hydroxycholesterol 3, 25-disulfate (25HCDS), have been demonstrated to be potent regulators of lipid metabolism, inflammatory response, cell apoptosis, and cell survival. In the present study, we tested these products' potential to treat LPS-induced acute liver failure in a mouse model. METHODS: Acute liver failure mouse model was established by intravenous injection with LPS. The injured liver function was treated with intraperitoneal administration of 25HC, 25HC3S or 25HCDS. Serum enzymatic activities were determined in our clinic laboratory. ELISA assays were used to detect pro-inflammatory factor levels in sera. Western blot, Real-time Quantitative PCR and RT2 Profiler PCR Array analysis were used to determine levels of gene expression. RESULTS: Administration of 25HC3S/25HCDS decreased serum liver-impaired markers; suppressed secretion of pro-inflammatory factors; alleviated liver, lung, and kidney injury; and subsequently increased the survival rate in the LPS-induced mouse model. These effects resulted from the inhibition of the expression of genes involved in the pro-inflammatory response and apoptosis and the simultaneous induction of the expression of genes involved in cell survival. Compared to 25HC, 25HC3S and 25HCDS exhibited significantly stronger effects in these activities, indicating that the cholesterol metabolites play an important role in the inflammatory response, cell apoptosis, and cell survival in vivo. CONCLUSIONS: 25HC3S/25HCDS has potential to serve as novel biomedicines in the therapy of acute liver failure and acute multiple organ failure.


Subject(s)
Chemical and Drug Induced Liver Injury/mortality , Chemical and Drug Induced Liver Injury/prevention & control , Cholesterol Esters/therapeutic use , Cholesterol/metabolism , Hydroxycholesterols/therapeutic use , Lipopolysaccharides , Animals , Biomarkers/blood , Chemical and Drug Induced Liver Injury/pathology , Cytokines/blood , Gene Expression Regulation/drug effects , Liver/pathology , Liver Function Tests , Male , Mice , Mice, Inbred C57BL , Survival Rate
13.
Int J Mol Sci ; 17(1)2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26797605

ABSTRACT

Adipogenesis is the process by which precursor stem cells differentiate into lipid laden adipocytes. Adipogenesis is regulated by a complex and highly orchestrated gene expression program. In mammalian cells, the peroxisome proliferator-activated receptor γ (PPARγ), and the CCAAT/enhancer binding proteins (C/EBPs) such as C/EBPα, ß and δ are considered the key early regulators of adipogenesis, while fatty acid binding protein 4 (FABP4), adiponectin, and fatty acid synthase (FAS) are responsible for the formation of mature adipocytes. Excess accumulation of lipids in the adipose tissue leads to obesity, which is associated with cardiovascular diseases, type II diabetes and other pathologies. Thus, investigating adipose tissue development and the underlying molecular mechanisms is vital to develop therapeutic agents capable of curbing the increasing incidence of obesity and related pathologies. In this review, we address the process of adipogenic differentiation, key transcription factors and proteins involved, adipogenic regulators and potential anti-adipogenic bioactive molecules.


Subject(s)
Adipogenesis/drug effects , Genistein/therapeutic use , Hydroxycholesterols/therapeutic use , Obesity/drug therapy , Stilbenes/therapeutic use , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/pathology , Adipogenesis/genetics , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , CCAAT-Enhancer-Binding Protein-delta/genetics , CCAAT-Enhancer-Binding Protein-delta/metabolism , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation , Humans , Mice , Obesity/genetics , Obesity/metabolism , Obesity/pathology , PPAR gamma/genetics , PPAR gamma/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Resveratrol , Signal Transduction
14.
Science ; 350(6261): 674-7, 2015 Nov 06.
Article in English | MEDLINE | ID: mdl-26542570

ABSTRACT

Cataracts reduce vision in 50% of individuals over 70 years of age and are a common form of blindness worldwide. Cataracts are caused when damage to the major lens crystallin proteins causes their misfolding and aggregation into insoluble amyloids. Using a thermal stability assay, we identified a class of molecules that bind α-crystallins (cryAA and cryAB) and reversed their aggregation in vitro. The most promising compound improved lens transparency in the R49C cryAA and R120G cryAB mouse models of hereditary cataract. It also partially restored protein solubility in the lenses of aged mice in vivo and in human lenses ex vivo. These findings suggest an approach to treating cataracts by stabilizing α-crystallins.


Subject(s)
Cataract/drug therapy , Hydroxycholesterols/pharmacology , alpha-Crystallin A Chain/chemistry , alpha-Crystallin B Chain/chemistry , Amyloid/antagonists & inhibitors , Amyloid/chemistry , Animals , Calorimetry, Differential Scanning , Cataract/genetics , Disease Models, Animal , Gene Knock-In Techniques , Humans , Hydroxycholesterols/chemistry , Hydroxycholesterols/therapeutic use , Mice , Protein Conformation/drug effects , Protein Stability/drug effects , alpha-Crystallin A Chain/genetics , alpha-Crystallin B Chain/genetics
15.
Spine J ; 15(4): 733-42, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25450659

ABSTRACT

BACKGROUND CONTEXT: The nonunion rate after lumbar spinal fusion is as high as 25%. Recombinant human bone morphogenetic protein 2 (rhBMP2) has been used as a biological adjunct to promote bony fusion. However, recently there have been concerns about BMP2. Oxysterol 133 (Oxy133) has been shown to promote excellent fusion rates in rodent lumbar spine models and offers a potential alternative to rhBMP2. PURPOSE: The purpose of this study was to compare the fusion rate of rhBMP2 and Oxy133 in a randomized controlled trial using a posterolateral lumbar rabbit spinal fusion model. STUDY DESIGN: This was a randomized control animal study. METHODS: Twenty-four male adult white New Zealand rabbits (3-3.5 kg) underwent bilateral posterolateral lumbar spinal fusion at L4-L5. Rabbits were divided into four groups: control (A), 30-µg rhBMP2 (B), 20-mg Oxy133 (C), and 60-mg Oxy133 (D). At 4 weeks, fusion was evaluated by fluoroscopy, and at 8 weeks, the rabbits were sacrificed and fusion was evaluated radiographically, by manual palpation, and with microcomputed tomography. RESULTS: Fusion rates by radiographic analysis at 8 weeks were Group A, 40.0%; Group B, 91.7%; Group C, 91.7%; and Group D, 100%. Evaluation of fusion masses by manual palpation of excised spines after sacrifice showed the following fusion rates: Group A, 0%; Group B, 83.3%; Group C, 83.3%; and Group D, 90%. Microcomputed tomography scanning confirmed these findings. CONCLUSIONS: These findings in a rabbit model demonstrate that both 20- and 60-mg Oxy133 doses promote fusion that is equivalent to fusion induced by 30-µg rhBMP2 and significantly greater than the control group. The present findings confirm that Oxy133 is a promising candidate for therapeutic development as an alternative to rhBMP2 to promote spinal fusion.


Subject(s)
Bone Morphogenetic Protein 2/therapeutic use , Hydroxycholesterols/therapeutic use , Lumbar Vertebrae/surgery , Spinal Fusion/methods , Transforming Growth Factor beta/therapeutic use , Animals , Lumbar Vertebrae/diagnostic imaging , Male , Models, Animal , Rabbits , Radiography , Recombinant Proteins/therapeutic use , Treatment Outcome
16.
Behav Brain Res ; 278: 356-9, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25453744

ABSTRACT

In spite of the fact that cholesterol does not pass the blood-brain barrier, treatment of mice with dietary cholesterol causes significant effects on a number of genes in the brain and in addition a memory impairment. We have suggested that these effects are mediated by 27-hydroxycholesterol, which is able to pass the blood-brain barrier. To test this hypothesis we utilized Cyp27-/- mice lacking 27-hydroxycholesterol. The negative effect on memory observed after treatment of wildtype mice with dietary cholesterol was not observed in these mice. The cholesterol diet reduced the levels of the "memory protein" Arc (Activity Regulated Cytoskeleton associated protein) in the hippocampus of the wildtype mice but not in the hippocampus of the Cyp27-/- mice. The results are consistent with 27-hydroxycholesterol as the mediator of the negative effects of cholesterol on cognition.


Subject(s)
Cholesterol/adverse effects , Cognition Disorders/etiology , Cognition Disorders/genetics , Hydroxycholesterols/therapeutic use , AIDS-Related Complex/genetics , AIDS-Related Complex/metabolism , Animals , Body Weight/drug effects , Body Weight/genetics , Cholestanetriol 26-Monooxygenase/deficiency , Cholestanetriol 26-Monooxygenase/genetics , Cholic Acid/administration & dosage , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/metabolism , Spatial Learning/drug effects
17.
Biochem Biophys Res Commun ; 447(1): 152-7, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24704452

ABSTRACT

Cholesterosis is a disease of cholesterol metabolism characterized by the presence of excessive lipid droplets in the cytoplasm. These lipid droplets are mainly composed of cholesterol esters derived from free cholesterol. The removal of excess cholesterol from gallbladder epithelial cells (GBECs) is very important for the maintenance of intracellular cholesterol homeostasis and the preservation of gallbladder function. Several lines of evidence have indicated that the activation of either peroxisome proliferator-activated receptor gamma (PPARγ) or liver X receptor α (LXRα) relates to cholesterol efflux. While pioglitazone can regulate the activation of PPARγ, 22(R)-hydroxycholesterol can activate LXRα and is a metabolic intermediate in the biosynthesis of steroid hormones. However, the effect of 22(R)-hydroxycholesterol in combination with pioglitazone on cholesterosis of the gallbladder is unclear. GBECs were treated with pioglitazone, 22(R)-hydroxycholesterol or PPARγ siRNA followed by Western blot analysis for ATP-binding cassette transporter A1 (ABCA1), PPARγ and LXRα. Cholesterol efflux to apoA-I was determined, and Oil Red O staining was performed to monitor variations in lipid levels in treated GBECs. Our data showed that 22(R)-hydroxycholesterol can modestly up-regulate LXRα while simultaneously increasing ABCA1 by 56%. The combination of 22(R)-hydroxycholesterol and pioglitazone resulted in a 3.64-fold increase in ABCA1 expression and a high rate of cholesterol efflux. Oil Red O staining showed an obvious reduction in the lipid droplets associated with cholesterosis in GBECs. In conclusion, the present findings indicate that the anti-lipid deposition action of 22(R)-hydroxycholesterol combined with pioglitazone involves the activation of the PPARγ-LXRα-ABCA1 pathway, increased ABCA1 expression and the efflux of cholesterol from GBECs. Thus, 22(R)-hydroxycholesterol synergistically combined with pioglitazone to produce a remarkable effect on lipid deposition in cholesterosis GBECs.


Subject(s)
ATP Binding Cassette Transporter 1/physiology , Cholesterol Esters/metabolism , Gallbladder Diseases/drug therapy , Hydroxycholesterols/therapeutic use , Hypercholesterolemia/drug therapy , Orphan Nuclear Receptors/physiology , PPAR gamma/physiology , Thiazolidinediones/therapeutic use , Cells, Cultured , Drug Synergism , Epithelial Cells/metabolism , Gallbladder/cytology , Humans , Liver X Receptors , Pioglitazone
18.
Lipids Health Dis ; 10: 44, 2011 Mar 09.
Article in English | MEDLINE | ID: mdl-21388551

ABSTRACT

Oxysterols are oxidation products of cholesterol that are generated by enzymatic reactions mediated by cytochrome P450 family enzymes or by non-enzymatic reactions involving reactive oxygen and nitrogen species. Oxysterols play various regulatory roles in normal cellular processes such as cholesterol homeostasis by acting as intermediates in cholesterol catabolism. Pathological effects of oxysterols have also been described, and various reports have implicated oxysterols in several disease states, including atherosclerosis, neurological disease, and cancer. Numerous studies show that oxysterols are associated with various types of cancer, including cancers of the colon, lung, skin, breast and bile ducts. The molecular mechanisms whereby oxysterols contribute to the initiation and progression of cancer are an area of active investigation. This review focuses on the current state of knowledge regarding the role of oxysterols in carcinogenesis. Mutagenicity of oxysterols has been described in both nuclear and mitochondrial DNA. Certain oxysterols such as cholesterol-epoxide and cholestanetriol have been shown to be mutagenic and genotoxic. Oxysterols possess pro-oxidative and pro-inflammatory properties that can contribute to carcinogenesis. Oxysterols can induce the production of inflammatory cytokines such as interleukin-8 and interleukin-1ß. Certain oxysterols are also involved in the induction of cyclo-oxygenase-2 expression. Inflammatory effects can also be mediated through the activation of liver-X-receptor, a nuclear receptor for oxysterols. Thus, several distinct molecular mechanisms have been described showing that oxysterols contribute to the initiation and progression of cancers arising in various organ systems.


Subject(s)
Carcinogens/pharmacology , Cholesterol/metabolism , Hydroxycholesterols/pharmacology , Neoplasms/chemically induced , Animals , Bile Duct Neoplasms/chemically induced , Carcinogens/toxicity , Cholangiocarcinoma/chemically induced , Colonic Neoplasms/chemically induced , Humans , Hydroxycholesterols/adverse effects , Hydroxycholesterols/therapeutic use , Lung Neoplasms/chemically induced , Lung Neoplasms/drug therapy , Mutagens/pharmacology , Oxidation-Reduction
19.
Endocrinology ; 149(8): 3778-88, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18450964

ABSTRACT

Prostate cancer is the most commonly diagnosed and the second leading cause of cancer death in men. The androgens-androgen receptor signaling plays an important role in normal prostate development, as well as in prostatic diseases, such as benign hyperplasia and prostate cancer. Accordingly, androgen ablation has been the most effective endocrine therapy for hormone-dependent prostate cancer. Here, we report a novel nuclear receptor-mediated mechanism of androgen deprivation. Genetic or pharmacological activation of the liver X receptor (LXR) in vivo lowered androgenic activity by inducing the hydroxysteroid sulfotransferase 2A1, an enzyme essential for the metabolic deactivation of androgens. Activation of LXR also inhibited the expression of steroid sulfatase in the prostate, which may have helped to prevent the local conversion of sulfonated androgens back to active metabolites. Interestingly, LXR also induced the expression of selected testicular androgen synthesizing enzymes. At the physiological level, activation of LXR in mice inhibited androgen-dependent prostate regeneration in castrated mice. Treatment with LXR agonists inhibited androgen-dependent proliferation of prostate cancer cells in a LXR- and sulfotransferase 2A1-dependent manner. In summary, we have revealed a novel function of LXR in androgen homeostasis, an endocrine role distinct to the previously known sterol sensor function of this receptor. LXR may represent a novel therapeutic target for androgen deprivation, and may aid in the treatment and prevention of hormone-dependent prostate cancer.


Subject(s)
Androgens/biosynthesis , Benzoates/therapeutic use , Benzylamines/therapeutic use , DNA-Binding Proteins/agonists , Hydroxycholesterols/therapeutic use , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Receptors, Cytoplasmic and Nuclear/agonists , Androgens/pharmacology , Animals , Benzoates/pharmacology , Benzylamines/pharmacology , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/physiology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Enzymologic/drug effects , Humans , Hydroxycholesterols/pharmacology , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Hormone-Dependent/metabolism , Orphan Nuclear Receptors , Prostate/drug effects , Prostate/physiology , Prostatic Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/physiology , Regeneration/drug effects , Sulfotransferases/genetics , Testosterone/blood
20.
Neuroscience ; 138(4): 1097-103, 2006.
Article in English | MEDLINE | ID: mdl-16442740

ABSTRACT

7beta-Hydroxycholesterol has been previously demonstrated to inhibit astrocytosis in injured cortex or spinal cord of rats. In this study, we explored the inhibitory effects of the liposome containing 7beta-hydroxycholesterol on the reactive astrocytosis caused by the injection of iron into the hippocampus of rats and furthermore evaluated the involvement of reactive astrocytosis in iron-induced epilepsy. Injection of ferric chloride solution unilaterally into the hippocampus of rats induced spontaneous spiking activity ipsilaterally then developed into bilateral hippocampi and generalized convulsive seizures within the first week post-operation, and spontaneous epileptiform activity and generalized seizures lasted as long as 2 weeks post-operation, whereas none of the rats injected with sodium chloride solution unilaterally into the hippocampus developed generalized seizures. With immunohistochemistry and Western blot analyses, apparent reactive astrocytosis in bilateral hippocampi was detected using antibody against glial fibrillary acidic protein 14 days after the injection of ferric chloride solution, but no significant differences were found in the amount of synaptophysin protein, a presynaptic vesicle protein, as compared with the rats injected with sodium chloride solution. Infusion of liposome suspension containing 7beta-hydroxycholesterol into the same site immediately after the injection of ferric chloride solution reduced the extent of the reactive astrocytosis by 50%-55% of the amount of glial fibrillary acidic protein in the hippocampi of both hemispheres, and non-significantly elevated the amount of synaptophysin protein in both sides of hippocampus. However, these effects did not significantly modify the seizure latency and the incidence of generalized seizures in the rats. These findings demonstrate the effects of 7beta-hydroxycholesterol on the inhibition of reactive astrocytosis caused by iron deposition in the hippocampus of rats, and suggest that the reactive astrocytosis may not play a causal role in the development of iron-induced seizures.


Subject(s)
Epilepsy/drug therapy , Gliosis/drug therapy , Hippocampus/drug effects , Hydroxycholesterols/pharmacology , Iron Metabolism Disorders/drug therapy , Iron/antagonists & inhibitors , Action Potentials/drug effects , Action Potentials/physiology , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Brain Injuries/complications , Brain Injuries/metabolism , Brain Injuries/physiopathology , Chlorides , Convulsants/antagonists & inhibitors , Convulsants/toxicity , Disease Models, Animal , Down-Regulation/drug effects , Down-Regulation/physiology , Epilepsy/chemically induced , Epilepsy/physiopathology , Epilepsy, Temporal Lobe/chemically induced , Epilepsy, Temporal Lobe/drug therapy , Epilepsy, Temporal Lobe/physiopathology , Ferric Compounds/antagonists & inhibitors , Ferric Compounds/toxicity , Glial Fibrillary Acidic Protein/metabolism , Gliosis/chemically induced , Gliosis/physiopathology , Hippocampus/metabolism , Hippocampus/physiopathology , Hydroxycholesterols/therapeutic use , Iron/metabolism , Iron/toxicity , Iron Metabolism Disorders/metabolism , Iron Metabolism Disorders/physiopathology , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Rats , Reaction Time/drug effects , Reaction Time/physiology , Synaptophysin/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...