Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Arch Pharm Res ; 45(5): 340-351, 2022 May.
Article in English | MEDLINE | ID: mdl-35608792

ABSTRACT

Adipose browning has recently been reported to be a novel therapeutic strategy for obesity. Because the retinoic acid receptor (RAR) is a potential target involved in browning, adapalene (AD), an anti-acne agent with RAR agonism, was examined in detail for its effects on adipose browning and the underlying mechanisms in vitro and in vivo. AD upregulated the expression of adipose browning-related markers in a concentration-dependent manner, promoted mitochondrial biogenesis, increased oxygen consumption rates, and lowered lipid droplet sizes in differentiated 3T3/L1 white adipocytes. Among the three retinoic acid receptors (RARα, RARß, and RARγ), knockdown of the gene encoding RARß mitigated AD-induced adipose browning. Similarly, LE135 (a selective RARß antagonist) attenuated AD action, suggesting that AD promotes adipose browning through RARß. Sequential phosphorylation of p38 mitogen-activated protein kinase (MAPK) and activating transcription factor 2 (ATF2) was critical for AD-induced adipose browning, based on the observations that either SB203580 (a p38 MAPK inhibitor) or ATF2 siRNA reduced the effects of AD. In vivo browning effects of AD were confirmed in C57BL/6J mice and high-fat diet-induced obese (DIO) mice after oral administration of AD either acutely or chronically. This study identifies new actions of AD as an adipose browning agent and demonstrates that RARß activation followed by increased phosphorylation of p38 MAPK and ATF2 appears to be a key mechanism of AD action.


Subject(s)
Activating Transcription Factor 2 , Adapalene , Adipose Tissue, White , Lipid Regulating Agents , Receptors, Retinoic Acid , p38 Mitogen-Activated Protein Kinases , 3T3-L1 Cells , Activating Transcription Factor 2/metabolism , Adapalene/administration & dosage , Adapalene/pharmacology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Administration, Oral , Animals , Lipid Regulating Agents/administration & dosage , Lipid Regulating Agents/pharmacology , Mice , Mice, Inbred C57BL , Phosphorylation , Receptors, Retinoic Acid/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
2.
Eur J Pharmacol ; 926: 175040, 2022 Jul 05.
Article in English | MEDLINE | ID: mdl-35598846

ABSTRACT

Extensive studies have shown that the increasing brown adipose tissue (BAT) mass/activity possesses a strong ability to prevent obesity and its related complications. The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signal pathway is known to play a role in adipocyte differentiation and development. However, its impact on thermogenic properties of mature brown adipocytes has not yet been clarified. Nifuroxazide (NFX), a potent inhibitor of STAT3, has received widespread attention due to its alternative anti-tumor and anti-inflammatory effects. Herein, we report that NFX induces lipolysis with subsequent downregulation of ACCα and FAS, while ATGL and pHSL levels are elevated in mature brown adipocytes. Furthermore, NFX treatment promotes the mitochondrial respiration of mature brown adipocytes, as evidenced by increased expression of thermogenic transcriptional factors and mitochondrial content. In addition, it also alleviates the IL-6 and TNFα inhibition on brown thermogenic programming via suppressing the STAT3/NF-κB/IL-6 signaling pathway. In general, these findings suggest that the blockade of the JAK/STAT3 pathway by NFX has a pro-thermogenic effect on mature brown adipocytes which opens new perspectives for NFX repurposing and potential therapeutic route to counteract obesity and related metabolic disorders.


Subject(s)
Adipocytes, Brown , Hydroxybenzoates , Lipid Regulating Agents , Mitochondria , Nitrofurans , STAT3 Transcription Factor , Uncoupling Protein 1 , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Humans , Hydroxybenzoates/pharmacology , Interleukin-6/metabolism , Lipid Regulating Agents/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Nitrofurans/pharmacology , Obesity/metabolism , Obesity/prevention & control , Obesity/therapy , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Thermogenesis/drug effects , Uncoupling Protein 1/biosynthesis , Uncoupling Protein 1/metabolism
3.
J Am Coll Cardiol ; 78(18): 1831-1843, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34711342

ABSTRACT

Detecting familial hypercholesterolemia (FH) early and "normalizing" low-density lipoprotein (LDL) cholesterol values are the 2 pillars for effective cardiovascular disease prevention in FH. Combining lipid-lowering therapies targeting synergistic/complementary metabolic pathways makes this feasible, even among severe phenotypes. For LDL receptor-dependent treatments, PCSK9 remains the main target for adjunctive therapy to statins and ezetimibe through a variety of approaches. These include protein inhibition (adnectins), inhibition of translation at mRNA level (antisense oligonucleotides or small interfering RNA), and creation of loss-of-function mutations through base-pair editing. For patients with little LDL receptor function, LDL receptor-independent treatment targeting ANGPTL3 through monoclonal therapies are now available, or in the future, antisense/small interfering RNA-based approaches offer alternative approaches. Finally, first-in-human studies are ongoing, testing adenovirus-mediated gene therapy transducing healthy LDLR DNA in patients with HoFH. Further development of the CRISPR cas technology, which has shown promising results in vivo on introducing PCSK9 loss-of-function mutations, will move a single-dose, curative treatment for FH closer.


Subject(s)
Cardiovascular Diseases , Hyperlipoproteinemia Type II , Lipid Regulating Agents , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Cholesterol, LDL/blood , Drug Development , Early Diagnosis , Humans , Hyperlipoproteinemia Type II/genetics , Hyperlipoproteinemia Type II/metabolism , Hyperlipoproteinemia Type II/therapy , Lipid Regulating Agents/classification , Lipid Regulating Agents/pharmacology , Therapies, Investigational
4.
J Am Coll Cardiol ; 78(16): 1635-1654, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34649702

ABSTRACT

Coronavirus disease-2019 (COVID-19) is associated with systemic inflammation, endothelial activation, and multiorgan manifestations. Lipid-modulating agents may be useful in treating patients with COVID-19. These agents may inhibit viral entry by lipid raft disruption or ameliorate the inflammatory response and endothelial activation. In addition, dyslipidemia with lower high-density lipoprotein cholesterol and higher triglyceride levels portend worse outcomes in patients with COVID-19. Upon a systematic search, 40 randomized controlled trials (RCTs) with lipid-modulating agents were identified, including 17 statin trials, 14 omega-3 fatty acids RCTs, 3 fibrate RCTs, 5 niacin RCTs, and 1 dalcetrapib RCT for the management or prevention of COVID-19. From these 40 RCTs, only 2 have reported preliminary results, and most others are ongoing. This paper summarizes the ongoing or completed RCTs of lipid-modulating agents in COVID-19 and the implications of these trials for patient management.


Subject(s)
COVID-19 Drug Treatment , COVID-19/prevention & control , Fatty Acids, Omega-3/therapeutic use , Fibric Acids/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Niacin/therapeutic use , Amides/pharmacology , Amides/therapeutic use , Esters/pharmacology , Esters/therapeutic use , Fatty Acids, Omega-3/pharmacology , Fibric Acids/pharmacology , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipid Regulating Agents/pharmacology , Lipid Regulating Agents/therapeutic use , Niacin/pharmacology , Randomized Controlled Trials as Topic , Sulfhydryl Compounds/pharmacology , Sulfhydryl Compounds/therapeutic use
5.
PLoS Pathog ; 17(7): e1009734, 2021 07.
Article in English | MEDLINE | ID: mdl-34310651

ABSTRACT

Animal African Trypanosomiasis (AAT) is a debilitating livestock disease prevalent across sub-Saharan Africa, a main cause of which is the protozoan parasite Trypanosoma congolense. In comparison to the well-studied T. brucei, there is a major paucity of knowledge regarding the biology of T. congolense. Here, we use a combination of omics technologies and novel genetic tools to characterise core metabolism in T. congolense mammalian-infective bloodstream-form parasites, and test whether metabolic differences compared to T. brucei impact upon sensitivity to metabolic inhibition. Like the bloodstream stage of T. brucei, glycolysis plays a major part in T. congolense energy metabolism. However, the rate of glucose uptake is significantly lower in bloodstream stage T. congolense, with cells remaining viable when cultured in concentrations as low as 2 mM. Instead of pyruvate, the primary glycolytic endpoints are succinate, malate and acetate. Transcriptomics analysis showed higher levels of transcripts associated with the mitochondrial pyruvate dehydrogenase complex, acetate generation, and the glycosomal succinate shunt in T. congolense, compared to T. brucei. Stable-isotope labelling of glucose enabled the comparison of carbon usage between T. brucei and T. congolense, highlighting differences in nucleotide and saturated fatty acid metabolism. To validate the metabolic similarities and differences, both species were treated with metabolic inhibitors, confirming that electron transport chain activity is not essential in T. congolense. However, the parasite exhibits increased sensitivity to inhibition of mitochondrial pyruvate import, compared to T. brucei. Strikingly, T. congolense exhibited significant resistance to inhibitors of fatty acid synthesis, including a 780-fold higher EC50 for the lipase and fatty acid synthase inhibitor Orlistat, compared to T. brucei. These data highlight that bloodstream form T. congolense diverges from T. brucei in key areas of metabolism, with several features that are intermediate between bloodstream- and insect-stage T. brucei. These results have implications for drug development, mechanisms of drug resistance and host-pathogen interactions.


Subject(s)
Trypanosoma brucei brucei/metabolism , Trypanosoma congolense/metabolism , Animals , Lipid Regulating Agents/pharmacology , Mice , Trypanosoma brucei brucei/drug effects , Trypanosoma congolense/drug effects , Trypanosomiasis, African
6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1866(10): 158981, 2021 10.
Article in English | MEDLINE | ID: mdl-34119681

ABSTRACT

Lipid droplets (LDs) perform several important functions like inflammatory responses, membrane trafficking, acts as secondary messengers, etc. rather than simply working as an energy reservoir. LDs have been implicated as a controlling factor in the progression of atherosclerosis followed by foam cell formation that derives from macrophages during the differentiation process. However, the role of LDs in monocyte differentiation or its further immunological function is still an area that mandates in-depth investigation. We report that LD dynamics is important for differentiation of monocytes and is absolutely required for sustained and prolonged functional activity of differentiated macrophages. In THP-1 cell line model system, we elucidated that increase in total LD content in monocyte by external lipid supplements, can induce monocyte differentiation independent of classical stimuli, PMA. Differential expression of PLIN2 and ATGL during the event, together with abrogation of de novo lipogenesis further confirmed the fact. Besides, an increase in LD content by free fatty acid supplement was able to exert a synergistic effect with PMA on differentiation and phagocytic activity compared to when they are used alone. Additionally, we have shown Rab5a to play a vital role in LDs biosynthesis/maturation in monocytes and thereby directly affecting differentiation of monocytes into macrophages via AKT pathway. Thus our study reveals the multi-faceted function of LDs during the process of monocyte to macrophage differentiation and thereby helping to maintain the functional activity.


Subject(s)
Lipid Droplets/metabolism , Lipid Metabolism/immunology , Macrophages/immunology , Monocytes/immunology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Endocytosis/drug effects , Endocytosis/immunology , Humans , Intravital Microscopy , Lipid Droplets/drug effects , Lipid Metabolism/drug effects , Lipid Regulating Agents/pharmacology , Macrophages/cytology , Macrophages/metabolism , Monocytes/cytology , Monocytes/metabolism , THP-1 Cells , rab5 GTP-Binding Proteins/metabolism
7.
Postgrad Med ; 133(6): 651-664, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33900135

ABSTRACT

Treatment with icosapent ethyl 4 g/day, a highly purified and stable ethyl ester of eicosapentaenoic acid (EPA), demonstrated a significant reduction in atherosclerotic cardiovascular disease (ASCVD) events and death in REDUCE-IT. However, analyses of REDUCE-IT and meta-analyses have suggested that this clinical benefit is greater than can be achieved by triglyceride reduction alone. EPA therefore may have additional pleiotropic effects, including anti-inflammatory and anti-aggregatory mechanisms. EPA competes with arachidonic acid for cyclooxygenase and lipoxygenase, producing anti-inflammatory and anti-aggregatory metabolites rather than the more deleterious metabolites associated with arachidonic acid. Changing the EPA:arachidonic acid ratio may shift metabolic status from pro-inflammatory/pro-aggregatory to anti-inflammatory/anti-aggregatory. EPA also has antioxidant effects and increases synthesis of nitric oxide. Incorporation of EPA into phospholipid bilayers influences membrane structure and may help to prevent cardiac arrhythmias. Clinically, this may translate into improved vascular health, including regression of atherosclerotic plaque. Overall, EPA has a range of pleiotropic effects that contribute to a reduction in ASCVD.


Subject(s)
Atherosclerosis , Eicosapentaenoic Acid/analogs & derivatives , Plaque, Atherosclerotic , Anti-Inflammatory Agents/pharmacology , Atherosclerosis/immunology , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Eicosapentaenoic Acid/pharmacology , Humans , Lipid Regulating Agents/pharmacology , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/metabolism , Platelet Aggregation Inhibitors/pharmacology , Treatment Outcome
8.
Open Heart ; 8(1)2021 04.
Article in English | MEDLINE | ID: mdl-33888593

ABSTRACT

Icosapent ethyl (Vascepa) is a purified preparation of the omega-3 fatty acid eicosapentaenoic acid, which is marketed by Amarin Pharma based in Ireland. The product was initially approved by the US Food and Drug Administration for the use of a high dose (4 g/day) in the treatment of hypertriglyceridaemia. On the basis of the results of the REDUCE-IT (Reduction of Cardiovascular Events with Icosapent Ethyl Intervention Trial), the agency later granted a label extension to include the additional indication of a reduction in risk of cardiovascular events in persons with serum triglyceride levels of 150 mg/dL or greater and established cardiovascular disease or diabetes. Data supporting the efficacy of omega-3 fatty acids in the prevention of cardiovascular disease have been inconsistent and controversial. The story of the development of icosapent ethyl has been fraught with challenges, including the invalidation of six core patents on the product, and recently, the completion of a new clinical trial, STRENGTH (Long-Term Outcomes Study to Assess STatin Residual Risk Reduction With EpaNova in HiGh CV Risk PatienTs With Hypertriglyceridemia), that directly contradicts REDUCE-IT and calls into question whether icosapent ethyl is actually effective in the secondary prevention of cardiovascular events. This article traces the course of the development of this fascinating product and discusses its complex medical, regulatory and legal history, which is still continuing to unfold.


Subject(s)
Biomedical Research/legislation & jurisprudence , Cardiovascular Diseases/prevention & control , Eicosapentaenoic Acid/analogs & derivatives , Secondary Prevention/methods , Eicosapentaenoic Acid/pharmacology , Humans , Lipid Regulating Agents/pharmacology
9.
Am J Med ; 134(9): 1085-1090, 2021 09.
Article in English | MEDLINE | ID: mdl-33864765

ABSTRACT

Elevated triglyceride (TG) levels have been linked to residual atherosclerotic cardiovascular risk in patients with controlled low-density lipoprotein cholesterol. However, outcome trials testing TG-lowering agents have failed to demonstrate cardiovascular risk reduction in statin-treated subjects. One such example is the recent STRENGTH trial, which tested mixed omega fatty acids (n3-FAs, 4 g/d) in high-risk patients with elevated TGs. Similar to trials using fibrates and niacin, the STRENGTH trial failed despite effective TG lowering. Results from these studies have contributed to skepticism about the use of TG-lowering therapy for cardiovascular risk. However, new mechanistic insights are provided by the REDUCE-IT trial that used icosapent ethyl (IPE), a purified formulation of the n3-FA eicosapentaenoic acid. In high-risk patients, IPE reduced a composite of cardiovascular events (25%, P < .001) in a manner not predicted by TG lowering. Benefits with IPE appear linked to broad pleiotropic actions associated with on-treatment eicosapentaenoic acid levels. These studies indicate that although TGs are a potential biomarker of cardiovascular risk, there is no evidence that TG lowering itself is an effective strategy for reducing such risk.


Subject(s)
Cardiovascular Diseases , Eicosapentaenoic Acid/analogs & derivatives , Hypertriglyceridemia/drug therapy , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/prevention & control , Clinical Trials as Topic , Eicosapentaenoic Acid/pharmacology , Heart Disease Risk Factors , Humans , Lipid Regulating Agents/pharmacology , Risk Adjustment/methods
10.
J Atheroscler Thromb ; 28(7): 665-678, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-33867421

ABSTRACT

Familial hypercholesterolemia (FH) is an inherited disorder with retarded clearance of plasma LDL caused by mutations of the genes involved in the LDL receptor-mediated pathway and most of them exhibit autosomal dominant inheritance. Homozygotes of FH (HoFH) may have plasma LDL-C levels, which are at least twice as high as those of heterozygous FH (HeFH) and therefore four times higher than normal levels. Prevalence of HoFH had been estimated as 1 in 1,000,000 before but more recent genetic analysis surveys predict 1 in 170,000 to 300,000. Since LDL receptor activity is severely impaired, HoFH patients do not or very poorly respond to medications to enhance activity, such as statins, and have a poorer prognosis compared to HeFH. HoFH should therefore be clinically distinguished from HeFH. Thorough family studies and genetic analysis are recommended for their accurate diagnosis.Fatal cardiovascular complications could develop even in the first decade of life for HoFH, so aggressive lipid-lowering therapy should be initiated as early as possible. Direct removal of plasma LDL by lipoprotein apheresis has been the principal measure for these patients. However, this treatment alone may not achieve stable LDL-C target levels and combination with drugs should be considered. The lipid-lowering effects of statins and PCSK9 inhibitors substantially vary depending on the remaining LDL receptor activity of individual patients. On the other hand, the action an MTP inhibitor is independent of LDL receptor activity, and it is effective in most HoFH cases.This review summarizes the key clinical issues of HoFH as well as insurance coverage available under the Japanese public healthcare system.


Subject(s)
Blood Component Removal/methods , Early Medical Intervention , Homozygous Familial Hypercholesterolemia , LDL-Receptor Related Proteins/genetics , Lipid Regulating Agents , Cholesterol, LDL/blood , Early Medical Intervention/methods , Early Medical Intervention/organization & administration , Heart Disease Risk Factors , Homozygous Familial Hypercholesterolemia/diagnosis , Homozygous Familial Hypercholesterolemia/drug therapy , Homozygous Familial Hypercholesterolemia/epidemiology , Homozygous Familial Hypercholesterolemia/genetics , Humans , Insurance Coverage , Japan/epidemiology , Lipid Regulating Agents/classification , Lipid Regulating Agents/pharmacology , Prognosis
11.
Heart ; 107(17): 1369-1375, 2021 09.
Article in English | MEDLINE | ID: mdl-33795379

ABSTRACT

Elevated levels of low-density lipoprotein cholesterol (LDL-C) are associated with increased risk of coronary heart disease and stroke. Guidelines for the management of dyslipidaemia from the European Society of Cardiology (ESC) and the European Atherosclerosis Society (EAS) were updated in late 2019 in light of recent intervention trials involving the use of innovative lipid-lowering agents in combination with statins. The new guidelines advocate achieving very low LDL-C levels in individuals at highest risk, within the paradigm of 'lower is better'. With the advent of combination therapy using ezetimibe and/or proprotein convertase subtilisin/kexin type 9 inhibitors in addition to statins, the routine attainment of extremely low LDL-C levels in the clinic has become a reality. Moreover, clinical trials in this setting have shown that, over the 5-7 years of treatment experience to date, profound LDL-C lowering leads to further reduction in cardiovascular events compared with more moderate lipid lowering, with no associated safety concerns. These reassuring findings are bolstered by genetic studies showing lifelong very low LDL-C levels (<1.4 mmol/L; <55 mg/dL) are associated with lower cardiovascular risk than in the general population, with no known detrimental health effects. Nevertheless, long-term safety studies are required to consolidate the present evidence base. This review summarises key data supporting the ESC/EAS recommendation to reduce markedly LDL-C levels, with aggressive goals for LDL-C in patients at highest risk, and provides expert opinion on its significance for clinical practice.


Subject(s)
Cardiovascular Diseases , Cholesterol, LDL/blood , Dyslipidemias , Lipid Regulating Agents/pharmacology , Cardiovascular Diseases/blood , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Heart Disease Risk Factors , Humans , Risk Adjustment
13.
Curr Atheroscler Rep ; 22(12): 72, 2020 10 03.
Article in English | MEDLINE | ID: mdl-33009957

ABSTRACT

PURPOSE OF REVIEW: The review highlights selected studies related to cardiovascular disease (CVD) prevention that were presented at the 2020 European Society of Cardiology (ESC) Congress-The Digital Experience. RECENT FINDINGS: The studies reviewed include clinical trials on novel RNA interference-based lipid-lowering therapies AKCEA-APOCIII-LRx and vupanorsen (AKCEA-ANGPTL3-LRx); the EVAPORATE trial assessing the effects of icosapent ethyl on coronary plaque volume progression; the LoDoCo2 trial evaluating the efficacy of low-dose colchicine in cardiovascular disease risk reduction among patients with chronic coronary artery disease; as well as the EMPEROR-Reduced trial evaluating cardiovascular and renal outcomes with empagliflozin in patients with heart failure and reduced ejection fraction. In addition, we review the BPLTTC analysis on blood pressure treatment across blood pressure levels and CVD status and discuss findings from the BRACE CORONA study that examined continuing versus suspending angiotensin-converting enzyme inhibitor or angiotensin receptor blockers in patients on these antihypertensive medications who were hospitalized with COVID-19 infection. The studies presented at the 2020 digital ESC Congress highlight the continuing advancements in the field of CVD prevention.


Subject(s)
Betacoronavirus/physiology , Cardiology , Cardiovascular Agents/pharmacology , Cardiovascular Diseases , Coronavirus Infections , Lipid Regulating Agents/pharmacology , Pandemics , Pneumonia, Viral , Benzhydryl Compounds/pharmacology , COVID-19 , Cardiology/methods , Cardiology/trends , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/prevention & control , Clinical Trials as Topic , Congresses as Topic , Coronavirus Infections/epidemiology , Coronavirus Infections/therapy , Eicosapentaenoic Acid/analogs & derivatives , Eicosapentaenoic Acid/pharmacology , Europe , Glucosides/pharmacology , Humans , Oligonucleotides/pharmacology , Pneumonia, Viral/epidemiology , Pneumonia, Viral/therapy , SARS-CoV-2 , Societies, Medical , Telecommunications
14.
Microvasc Res ; 132: 104054, 2020 11.
Article in English | MEDLINE | ID: mdl-32768464

ABSTRACT

Ischemic stroke is a leading cause of mortality worldwide that occurs following the reduction or interruption of blood brain supply, characterized by a cascade of early events as oxidative stress and ensuing neuro-inflammation, energy failure and the burst of intracellular Ca++ resulting in activation of phospholipases and large increase in FFA including arachidonic acid, ultimately leading to nervous cell death. Grape Seed Flour (GSF) is a complex polyphenolic mixture harboring antioxidant, anti-inflammatory and neuroprotective properties. Orlistat (Xenical ™,Xe) is a gastro-intestinal lipase inhibitor and an anti-obesity agent. In an earlier study we reported the higher efficiency in neuroprotection against HFD-induced brain lipotoxicity when combining the two drugs (GSF + Xe). As a result repurposing Xe as an adjunct to GSF therapy against stroke appeared relevant and worthy of investigation. I/R insult disrupted the blood brain barrier (BBB) as assessed by EB dye extravasation, increased water and Na+ within the brain. Ultrastructurally I/R altered the brain blood capillaries at the vicinity of hippocampus dentate gyrus area as assessed by transmission and scanning electron microscopy. I/R altered lipid metabolism as revealed by LDL/HDL ratio, lipase activity, and FFA profiles. Moreover, I/R induced neuro-inflammation as assessed by down-regulation of anti-inflammatory CD 56 and up-regulation of pro-inflammatory CD 68 antigen. Importantly almost all I/R-induced disturbances were retrieved partially upon Xe or GSF on their own, and optimally when combining the two drugs. Xe per se is protective against I/R injury and the best neuroprotection was obtained when associating low dosage Xe with high dosage GSF, enabling neuroprevention and cell survival within hippocampus dentate gyrus area as revealed by increased staining of Ki 67 proliferation biomarker.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Blood-Brain Barrier/drug effects , Brain Edema/prevention & control , Capillary Permeability/drug effects , Grape Seed Extract/pharmacology , Lipid Metabolism/drug effects , Lipid Regulating Agents/pharmacology , Neuroprotective Agents/pharmacology , Orlistat/pharmacology , Reperfusion Injury/prevention & control , Stroke/prevention & control , Animals , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/ultrastructure , Brain Edema/metabolism , Brain Edema/pathology , Disease Models, Animal , Inflammation Mediators/metabolism , Male , Rats, Wistar , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Stroke/metabolism , Stroke/pathology
15.
J Cell Mol Med ; 24(16): 9267-9279, 2020 08.
Article in English | MEDLINE | ID: mdl-32639107

ABSTRACT

The ω-3 fatty acids exert as an antioxidant via the G protein-coupled receptor 120 (GPR120). Icosapent ethyl, a purified eicosapentaenoic acid, showed a marked reduction in sudden cardiac death. Connexin43 is sensitive to redox status. We assessed whether icosapent ethyl attenuates fatal arrhythmias after myocardial infarction, a status of high oxidative stress, through increased connexin43 expression and whether the GPR120 signalling is involved in the protection. Male Wistar rats after ligating coronary artery were assigned to either vehicle or icosapent ethyl for 4 weeks. The postinfarction period was associated with increased oxidative-nitrosative stress. In concert, myocardial connexin43 levels revealed a significant decrease in vehicle-treated infarcted rats compared with sham. These changes of oxidative-nitrosative stress and connexin43 levels were blunted after icosapent ethyl administration. Provocative arrhythmias in the infarcted rats treated with icosapent ethyl were significantly improved than vehicle. Icosapent ethyl significantly increased GPR120 compared to vehicle after infarction. The effects of icosapent ethyl on superoxide and connexin43 were similar to GPR120 agonist GW9508. Besides, the effects of icosapent ethyl on oxidative-nitrosative stress and connexin43 phosphorylation were abolished by administering AH-7614, an inhibitor of GPR120. SIN-1 abolished the Cx43 phosphorylation of icosapent ethyl without affecting GPR120 levels. Taken together, chronic use of icosapent ethyl after infarction is associated with up-regulation of connexin43 phosphorylation through a GPR120-dependent antioxidant pathway and thus plays a beneficial effect on arrhythmogenic response to programmed electrical stimulation.


Subject(s)
Connexin 43/metabolism , Eicosapentaenoic Acid/analogs & derivatives , Lipid Regulating Agents/pharmacology , Myocardial Infarction/complications , Receptors, G-Protein-Coupled/metabolism , Tachycardia, Ventricular/drug therapy , Animals , Connexin 43/genetics , Eicosapentaenoic Acid/pharmacology , Male , Phosphorylation , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Tachycardia, Ventricular/etiology , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/pathology
16.
Theranostics ; 10(15): 6561-6580, 2020.
Article in English | MEDLINE | ID: mdl-32550890

ABSTRACT

Patients with cervical cancer (CCa) with lymph node metastasis (LNM) have an extremely poor prognosis. Elucidation of the molecular mechanisms underlying LNM may provide clinical therapeutic strategies for CCa. Upregulation of fatty acid-binding protein 5 (FABP5) expression in CCa tumours was demonstrated to positively correlate with LNM. However, the precise role and mechanisms of FABP5 in the LNM of CCa remain unknown. Methods: The diagnostic value of FABP5 as a predictor of LNM in CCa was evaluated in CCa tumour samples. The functional role of FABP5 and its upstream and downstream regulatory factors were investigated by gain-of-function and loss-of-function assays in vitro and in vivo. A mouse model of LNM was used to determine the effect of FABP5 on LNM and the therapeutic value of FABP5 targeting. Results: We demonstrated that FABP5 was markedly upregulated in CCa with LNM and correlated with poor prognosis. FABP5 protein was an independent predictor of LNM in a multivariate logistic analysis. Furthermore, FABP5 promoted epithelial-mesenchymal transition, lymphangiogenesis, and LNM by reprogramming fatty acid (FA) metabolism. Mechanistically, FABP5 promoted lipolysis and FA synthesis, which led to an increase in intracellular fatty acids (FAs) that activated NF-κB signalling, thus inducing LNM. Importantly, administration of orlistat, which attenuates FA metabolism reprogramming, inhibited FABP5-induced LNM in CCa. The pro-metastatic effect of FABP5 was reduced by miR-144-3p. Moreover, miR-144-3p was significantly downregulated and FABP5 was upregulated in CCa in a hypoxic microenvironment. Conclusion: Our findings highlight a FA metabolism-dependent mechanism of FABP5-induced LNM. Moreover, the expression and biological function of FABP5 can be regulated by miR-144-3p in hypoxia. Our study identifies FABP5 as a potential diagnostic biomarker and therapeutic target for LNM in CCa.


Subject(s)
Cell Movement , Cellular Reprogramming , Epithelial-Mesenchymal Transition , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Uterine Cervical Neoplasms/pathology , Adult , Aged , Animals , Cell Line, Tumor , Cell Proliferation , Fatty Acid-Binding Proteins/genetics , Female , Humans , Lipid Regulating Agents/pharmacology , Lymphatic Metastasis , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Orlistat/pharmacology , Prognosis , Signal Transduction , Tumor Microenvironment , Up-Regulation , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Xenograft Model Antitumor Assays
17.
Gastroenterology ; 159(3): 999-1014.e9, 2020 09.
Article in English | MEDLINE | ID: mdl-32450149

ABSTRACT

BACKGROUND & AIMS: Development of nonalcoholic steatohepatitis (NASH) is associated with reductions in hepatic microRNA122 (MIR122); the RAR related orphan receptor A (RORA) promotes expression of MIR122. Increasing expression of RORA in livers of mice increases expression of MIR122 and reduces lipotoxicity. We investigated the effects of a RORA agonist in mouse models of NASH. METHODS: We screened a chemical library to identify agonists of RORA and tested their effects on a human hepatocellular carcinoma cell line (Huh7). C57BL/6 mice were fed a chow or high-fat diet (HFD) for 4 weeks to induce fatty liver. Mice were given hydrodynamic tail vein injections of a MIR122 antagonist (antagomiR-122) or a control antagomiR once each week for 3 weeks while still on the HFD or chow diet, or intraperitoneal injections of the RORA agonist RS-2982 or vehicle, twice each week for 3 weeks. Livers, gonad white adipose, and skeletal muscle were collected and analyzed by reverse-transcription polymerase chain reaction, histology, and immunohistochemistry. A separate group of mice were fed an atherogenic diet, with or without injections of RS-2982 for 3 weeks; livers were analyzed by immunohistochemistry, and plasma was analyzed for levels of aminotransferases. We analyzed data from liver tissues from patients with NASH included in the RNA-sequencing databases GSE33814 and GSE89632. RESULTS: Injection of mice with antagomiR-122 significantly reduced levels of MIR122 in plasma, liver, and white adipose tissue; in mice on an HFD, antagomiR-122 injections increased fat droplets and total triglyceride content in liver and reduced ß-oxidation and energy expenditure, resulting in significantly more weight gain than in mice given the control microRNA. We identified RS-2982 as an agonist of RORA and found it to increase expression of MIR122 promoter activity in Huh7 cells. In mice fed an HFD or atherogenic diet, injections of RS-2982 increased hepatic levels of MIR122 precursors and reduced hepatic synthesis of triglycerides by reducing expression of biosynthesis enzymes. In these mice, RS-2982 significantly reduced hepatic lipotoxicity, reduced liver fibrosis, increased insulin resistance, and reduced body weight compared with mice injected with vehicle. Patients who underwent cardiovascular surgery had increased levels of plasma MIR122 compared to its levels before surgery; increased expression of plasma MIR122 was associated with increased levels of plasma free fatty acids and levels of RORA. CONCLUSIONS: We identified the compound RS-2982 as an agonist of RORA that increases expression of MIR122 in cell lines and livers of mice. Mice fed an HFD or atherogenic diet given injections of RS-2982 had reduced hepatic lipotoxicity, liver fibrosis, and body weight compared with mice given the vehicle. Agonists of RORA might be developed for treatment of NASH.


Subject(s)
Lipid Regulating Agents/pharmacology , MicroRNAs/genetics , Non-alcoholic Fatty Liver Disease/drug therapy , Nuclear Receptor Subfamily 1, Group F, Member 1/agonists , Obesity/drug therapy , Animals , Antagomirs/administration & dosage , Benzamides/pharmacology , Benzamides/therapeutic use , Body Weight , Cell Line, Tumor , Datasets as Topic , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Acids, Nonesterified/blood , Fatty Acids, Nonesterified/metabolism , Humans , Insulin Resistance , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Lipid Regulating Agents/therapeutic use , Liver/drug effects , Liver/pathology , Male , Mice , MicroRNAs/antagonists & inhibitors , MicroRNAs/blood , Mutation , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Promoter Regions, Genetic/drug effects , Up-Regulation/drug effects
18.
J Atheroscler Thromb ; 27(2): 119-131, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-31748470

ABSTRACT

A number of effective drugs have been developed through animal experiments, contributing to the health of many patients. In particular, the WHHL rabbit family (WHHL rabbits and its advanced strains (coronary atherosclerosis-prone WHHL-CA rabbits and myocardial infarction-prone WHHLMI rabbits) developed at Kobe University (Kobe, Japan) contributed greatly in the development of cholesterol-lowering agents. The WHHL rabbit family is animal models for human familial hypercholesterolemia, coronary atherosclerosis, and coronary heart disease. At the end of breeding of the WHHL rabbit family, this review summarizes the contribution of the WHHL rabbit family to the development of lipid-lowering agents and anti-atherosclerosis agents. Studies using the WHHL rabbit family demonstrated, for the first time in the world, that lowering serum cholesterol levels or preventing LDL oxidation can suppress the progression and destabilization of coronary lesions. In addition, the WHHL rabbit family contributed to the development of various compounds that exhibit lipid-lowering and anti-atherosclerotic effects and has also been used in studies of gene therapeutics. Furthermore, this review also discusses the causes of the increased discrepancy in drug development between the results of animal experiments and clinical studies, which became a problem in recent years, and addresses the importance of the selection of appropriate animal models used in studies in addition to an appropriate study design.


Subject(s)
Coronary Artery Disease , Disease Models, Animal , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Hyperlipoproteinemia Type II/drug therapy , Myocardial Infarction , Rabbits , Animals , Coronary Artery Disease/drug therapy , Coronary Artery Disease/metabolism , Drug Development/methods , History, 20th Century , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/history , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipid Regulating Agents/history , Lipid Regulating Agents/pharmacology , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism
19.
PLoS One ; 14(6): e0218459, 2019.
Article in English | MEDLINE | ID: mdl-31233523

ABSTRACT

Interruption of bile acid recirculation through inhibition of the apical sodium-dependent bile acid transporter (ASBT) is a promising strategy to alleviate hepatic cholesterol accumulation in non-alcoholic steatohepatitis (NASH), and improve the metabolic aspects of the disease. Potential disease-attenuating effects of the ASBT inhibitor volixibat (5, 15, and 30 mg/kg) were investigated in high-fat diet (HFD)-fed Ldlr-/-.Leiden mice over 24 weeks. Plasma and fecal bile acid levels, plasma insulin, lipids, and liver enzymes were monitored. Final analyses included liver histology, intrahepatic lipids, mesenteric white adipose tissue mass, and liver gene profiling. Consistent with its mechanism of action, volixibat significantly increased the total amount of bile acid in feces. At the highest dose, volixibat significantly attenuated the HFD-induced increase in hepatocyte hypertrophy, hepatic triglyceride and cholesteryl ester levels, and mesenteric white adipose tissue deposition. Non-alcoholic fatty liver disease activity score (NAS) was significantly lower in volixibat-treated mice than in the HFD controls. Gene profiling showed that volixibat reversed the inhibitory effect of the HFD on metabolic master regulators, including peroxisome proliferator-activated receptor-γ coactivator-1ß, insulin receptor, and sterol regulatory element-binding transcription factor 2. Volixibat may have beneficial effects on physiological and metabolic aspects of NASH pathophysiology.


Subject(s)
Benzothiepins/pharmacology , Energy Metabolism/drug effects , Glycosides/pharmacology , Lipid Regulating Agents/pharmacology , Non-alcoholic Fatty Liver Disease/metabolism , Organic Anion Transporters, Sodium-Dependent/antagonists & inhibitors , Symporters/antagonists & inhibitors , Adipose Tissue, Beige/metabolism , Animals , Bile Acids and Salts/metabolism , Biomarkers , Disease Models, Animal , Lipid Metabolism/drug effects , Male , Mice , Mice, Knockout , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Risk Factors
20.
Clin Pharmacol Drug Dev ; 7(7): 773-780, 2018 09.
Article in English | MEDLINE | ID: mdl-29659193

ABSTRACT

We sought to establish the bioequivalence of 2 weight-loss aids: orlistat 27-mg chewable tablet and orlistat 60-mg capsule, measured pharmacodynamically as percentage fecal fat excretion. Two open-label, single-center, randomized, 3-period, 3-treatment crossover studies were conducted in adults with body mass index 25-33 kg/m2 . For each 9-day treatment period, subjects received orlistat 27-mg chewable tablet, 60-mg capsule, or 120-mg capsules (2 60-mg capsules) 3 times daily; a 2-day washout separated treatments. Primary bioequivalence analyses were based on 2 1-sided tests of the 90% CI of the ratio of geometric means using log-transformed data (study 1) and by the dose-scale method to calculate bias-corrected and accelerated 90% CI of relative bioavailability (f) using nontransformed data (study 2). Bioequivalence was established if 90% CIs fell within 0.80-1.25. In total, 48 and 144 subjects were randomized in study 1 and study 2, respectively. Bioequivalence between the formulations was established in both studies: study 1 ratio of geometric means of percentage fecal fat excretion was 0.96 (2 1-sided tests, 90% CI 0.87-1.06); study 2-point estimate of f was 1.09 (bias-corrected and accelerated 90% CI 0.98-1.22). Tolerability of the 27-mg tablet was consistent with the 60-mg capsule; mild gastrointestinal effects were most common.


Subject(s)
Feces/chemistry , Lipid Regulating Agents/administration & dosage , Lipid Regulating Agents/pharmacokinetics , Orlistat/administration & dosage , Orlistat/pharmacokinetics , Administration, Oral , Adult , Biological Availability , Capsules , Cross-Over Studies , Female , Healthy Volunteers , Humans , Lipid Regulating Agents/adverse effects , Lipid Regulating Agents/pharmacology , Lipids/analysis , Male , Middle Aged , Orlistat/adverse effects , Orlistat/pharmacology , Tablets , Therapeutic Equivalency , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL