Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
Comput Biol Med ; 147: 105796, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35809408

RESUMEN

Obesity is an abnormal fat accumulation disorder in the metabolic syndrome constellation, and a risk factor for diabetes, cardiovascular disorders, non-alcoholic fatty liver disease (NAFLD), and cancer. Nuclear receptors (Peroxisome proliferator-activated receptor, PPAR) are implicated in metabolic syndrome and NAFLD, and have potential for therapeutic targeting. Nuclear receptors are ligand-dependent transcription factors that have diverse roles in metabolism, including regulating genes involved in lipid and glucose metabolism, modulating inflammatory genes, and are crucial for maintaining metabolic flexibility. PPAR activates adipose triglyceride lipase, which then releases fatty acids as ligands for PPAR, indicating the interdependency of nuclear receptors and lipases. Here, molecular docking was performed with selected phytochemical ligands that can bind with PPAR-α/γ (PDB ID: 2ZNN and 2ATH, respectively) using Glide module of Schrodinger software followed by molecular dynamics simulation study using Desmond module, and ADMET analysis. Interestingly, orlistat which is a well-known lipase and fatty acid synthase inhibitor also demonstrated favorable binding affinity with both PPAR-α/γ (-10.96 kcal/mol against PPARα and -10.26 kcal/mol against PPARγ). The highest docking scores were however shown by the flavonoids - rutin (-14.88 kcal/mol against PPARα and -13.64 kcal/mol against PPARγ), and its aglycone, quercetin (-10.08 kcal/mol in PPARα and -9.89 kcal/mol in PPARγ). The other phytochemicals (genistein, esculin, daidzin, naringenin, daidzein, dihydroxy coumarin, hydroquinone) showed lower binding affinity as dual agonists. The anti-obesity effects were experimentally validated in cultured adipocytes, which revealed better lipid inhibition by rutin and quercetin than orlistat (quercetin > rutin > orlistat) pointing to their strong potential in anti-obesity treatment.


Asunto(s)
Fármacos Antiobesidad , Síndrome Metabólico , Enfermedad del Hígado Graso no Alcohólico , Fármacos Antiobesidad/farmacología , Fármacos Antiobesidad/uso terapéutico , Humanos , Ligandos , Lípidos , Simulación del Acoplamiento Molecular , Obesidad/tratamiento farmacológico , Orlistat/farmacología , PPAR alfa/química , PPAR alfa/metabolismo , PPAR gamma/química , PPAR gamma/metabolismo , Fitoquímicos/farmacología , Quercetina , Rutina/farmacología
2.
J Med Chem ; 65(3): 1961-1978, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35089724

RESUMEN

Metabolic diseases are increasing at staggering rates globally. The peroxisome proliferator-activated receptors (PPARα/γ/δ) are fatty acid sensors that help mitigate imbalances between energy uptake and utilization. Herein, we report compounds derived from phenolic lipids present in cashew nut shell liquid (CNSL), an abundant waste byproduct, in an effort to create effective, accessible, and sustainable drugs. Derivatives of anacardic acid and cardanol were tested for PPAR activity in HEK293 cell co-transfection assays, primary hepatocytes, and 3T3-L1 adipocytes. In vivo studies using PPAR-expressing zebrafish embryos identified CNSL derivatives with varying tissue-specific activities. LDT409 (23) is an analogue of cardanol with partial agonist activity for PPARα and PPARγ. Pharmacokinetic profiling showed that 23 is orally bioavailable with a half-life of 4 h in mice. CNSL derivatives represent a sustainable source of selective PPAR modulators with balanced intermediate affinities (EC50 ∼ 100 nM to 10 µM) that provide distinct and favorable gene activation profiles for the treatment of diabetes and obesity.


Asunto(s)
Ácidos Anacárdicos/farmacología , Anacardium/química , Nueces/química , PPAR alfa/agonistas , PPAR delta/agonistas , PPAR gamma/agonistas , Células 3T3-L1 , Ácidos Anacárdicos/síntesis química , Ácidos Anacárdicos/metabolismo , Ácidos Anacárdicos/farmacocinética , Animales , Diseño de Fármacos , Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , PPAR alfa/química , PPAR delta/química , PPAR gamma/química , Dominios Proteicos , Pez Cebra
3.
Yakugaku Zasshi ; 141(11): 1267-1274, 2021.
Artículo en Japonés | MEDLINE | ID: mdl-34719550

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptor-type transcription factors that consist of three subtypes (α, γ, and ß/δ) with distinct physiological functions and ligand recognition. PPARs regulate energy metabolism and therefore become therapeutic targets for various metabolic diseases. While PPARα agonists are used as anti-dyslipidemia drugs and PPARγ agonists as anti-type 2 diabetes drugs, PPAR dual/pan agonists (that acts on two or three subtypes) are expected to treat non-alcoholic steatohepatitis (NASH), pulmonary fibrosis, etc. Structural analyses of PPAR-ligand-binding domain (LBD)-ligand co-crystals using X-ray crystallography have been done mainly on PPARγ, in which ligand-free apocrystals were prepared; however, the information on PPARα-LBD and PPARδ-LBD is limited. Recently, we succeeded to obtain 34 novel co-crystal structures of PPARα-LBD and various PPARα ligands (including fibrates) using various co-crystallization techniques. This procedure is applicable to preparation of PPARδ-LBD co-crystals, and contributes to molecular design of new PPAR targeted drugs based on all three PPAR-LBD structures.


Asunto(s)
Cristalografía por Rayos X/métodos , Ligandos , PPAR alfa/química , PPAR alfa/metabolismo , Metabolismo Energético , Hipoglucemiantes , Hipolipemiantes , Enfermedades Metabólicas/metabolismo , PPAR alfa/agonistas , PPAR alfa/fisiología , Unión Proteica , Dominios Proteicos
4.
Eur J Med Chem ; 223: 113665, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34192642

RESUMEN

New types of antidiabetic agents are continually needed with diabetes becoming the epidemic in the world. Indole alkaloids play an important role in natural products owing to their variable structures and versatile biological activities like anticonvulsant, anti-inflammatory, antidiabetic, antimicrobial, and anticancer activities, which are a promising source of novel antidiabetic drugs discovery. The synthesized indole derivatives possess similar properties to natural indole alkaloids. In the last two decades, more and more indole derivatives have been designed and synthesized for searching their bioactivities. This present review describes comprehensive structures of indole compounds with the potential antidiabetic activity including natural indole alkaloids and the synthetic indole derivatives based on the structure classification, summarizes their approaches isolated from natural sources or by synthetic methods, and discusses the antidiabetic effects and the mechanisms of action. Furthermore, this review also provides briefly synthetic procedures of some important indole derivatives.


Asunto(s)
Hipoglucemiantes/química , Indoles/química , Carbolinas/química , Carbolinas/metabolismo , Carbolinas/uso terapéutico , Diabetes Mellitus/tratamiento farmacológico , Humanos , Hipoglucemiantes/síntesis química , Hipoglucemiantes/metabolismo , Hipoglucemiantes/uso terapéutico , Indoles/metabolismo , Indoles/uso terapéutico , PPAR alfa/química , PPAR alfa/metabolismo , PPAR gamma/química , PPAR gamma/metabolismo , Terpenos/química , Terpenos/metabolismo , Terpenos/uso terapéutico
5.
Cells ; 10(3)2021 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-33799988

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are a family of nuclear receptors including PPARα, PPARγ, and PPARß/δ, acting as transcription factors to regulate the expression of a plethora of target genes involved in metabolism, immune reaction, cell differentiation, and a variety of other cellular changes and adaptive responses. PPARs are activated by a large number of both endogenous and exogenous lipid molecules, including phyto- and endo-cannabinoids, as well as endocannabinoid-like compounds. In this view, they can be considered an extension of the endocannabinoid system. Besides being directly activated by cannabinoids, PPARs are also indirectly modulated by receptors and enzymes regulating the activity and metabolism of endocannabinoids, and, vice versa, the expression of these receptors and enzymes may be regulated by PPARs. In this review, we provide an overview of the crosstalk between cannabinoids and PPARs, and the importance of their reciprocal regulation and modulation by common ligands, including those belonging to the extended endocannabinoid system (or "endocannabinoidome") in the control of major physiological and pathophysiological functions.


Asunto(s)
Endocannabinoides/metabolismo , PPAR alfa/genética , PPAR delta/genética , PPAR gamma/genética , PPAR-beta/genética , Receptores de Cannabinoides/genética , Animales , Regulación de la Expresión Génica , Humanos , Ligandos , Metabolismo de los Lípidos/genética , Ratones , Ratones Noqueados , Modelos Moleculares , PPAR alfa/química , PPAR alfa/metabolismo , PPAR delta/química , PPAR delta/metabolismo , PPAR gamma/química , PPAR gamma/metabolismo , PPAR-beta/química , PPAR-beta/metabolismo , Receptores de Cannabinoides/química , Receptores de Cannabinoides/metabolismo , Transducción de Señal , Transcripción Genética
6.
STAR Protoc ; 2(1): 100364, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33718889

RESUMEN

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptor-type transcription factors with three subtypes (α, δ, and γ) that regulate cell differentiation and metabolism. Co-crystals of human PPARα-ligand-binding domain (LBD)-PPARα ligand for X-ray crystallography have been difficult to obtain. Recombinant human PPARα-LBD proteins contain intrinsic fatty acids (iFAs of Escherichia coli origin) and may be unstable without ligands during crystallization. To circumvent these limitations, we have successfully applied various crystallization techniques, including co-crystallization, cross-seeding, soaking, delipidation, and coactivator peptide supplementation. For complete details on the use and execution of this protocol, please refer to Kamata et al. (2020).


Asunto(s)
Cristalografía por Rayos X/métodos , PPAR alfa/análisis , Proteínas/aislamiento & purificación , Sitios de Unión/efectos de los fármacos , Cristalización/métodos , Humanos , Ligandos , Modelos Moleculares , PPAR alfa/química , Unión Proteica/efectos de los fármacos , Dominios Proteicos , Factores de Transcripción/química
7.
EBioMedicine ; 62: 103130, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33279456

RESUMEN

BACKGROUND: The pathophysiology of schizophrenia, a major psychiatric disorder, remains elusive. In this study, the role of peroxisome proliferator-activated receptor (PPAR)/retinoid X receptor (RXR) families, belonging to the ligand-activated nuclear receptor superfamily, in schizophrenia, was analyzed. METHODS: The PPAR/RXR family genes were screened by exploiting molecular inversion probe (MIP)-based targeted next-generation sequencing (NGS) using the samples of 1,200 Japanese patients with schizophrenia. The results were compared with the whole-genome sequencing databases of the Japanese cohort (ToMMo) and the gnomAD. To reveal the relationship between PPAR/RXR dysfunction and schizophrenia, Ppara KO mice and fenofibrate (a clinically used PPARα agonist)-administered mice were assessed by performing behavioral, histological, and RNA-seq analyses. FINDINGS: Our findings indicate that c.209-2delA, His117Gln, Arg141Cys, and Arg226Trp of the PPARA gene are risk variants for schizophrenia. The c.209-2delA variant generated a premature termination codon. The three missense variants significantly decreased the activity of PPARα as a transcription factor in vitro. The Ppara KO mice exhibited schizophrenia-relevant phenotypes, including behavioral deficits and impaired synaptogenesis in the cerebral cortex. Oral administration of fenofibrate alleviated spine pathology induced by phencyclidine, an N-methyl-d-aspartate (NMDA) receptor antagonist. Furthermore, pre-treatment with fenofibrate suppressed the sensitivity of mice to another NMDA receptor antagonist, MK-801. RNA-seq analysis revealed that PPARα regulates the expression of synaptogenesis signaling pathway-related genes. INTERPRETATION: The findings of this study indicate that the mechanisms underlying schizophrenia pathogenesis involve PPARα-regulated transcriptional machinery and modulation of synapse physiology. Hence, PPARα can serve as a novel therapeutic target for schizophrenia.


Asunto(s)
Biomarcadores , PPAR alfa/metabolismo , Esquizofrenia/metabolismo , Adulto , Anciano , Empalme Alternativo , Secuencia de Aminoácidos , Animales , Antipsicóticos/farmacología , Antipsicóticos/uso terapéutico , Línea Celular , Susceptibilidad a Enfermedades , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Modelos Moleculares , Mutación , PPAR alfa/antagonistas & inhibidores , PPAR alfa/química , PPAR alfa/genética , Conformación Proteica , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/etiología , Relación Estructura-Actividad
8.
PLoS One ; 15(6): e0235362, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32584895

RESUMEN

OBJECTIVE: Cardiovascular disease is a leading cause of death worldwide. Obesity-related metabolic disorders including dyslipidemia cause impaired collateralization under ischemic conditions, thereby resulting in exacerbated cardiovascular dysfunction. Pemafibrate is a novel selective PPARα modulator, which has been reported to improve atherogenic dyslipidemia, in particular, hypertriglyceridemia and low HDL-cholesterol. Here, we investigated whether pemafibrate modulates the revascularization process in a mouse model of hindlimb ischemia. METHODS AND RESULTS: Male wild-type (WT) mice were randomly assigned to two groups, normal diet or pemafibrate admixture diet from the ages of 6 weeks. After 4 weeks, mice were subjected to unilateral hindlimb surgery to remove the left femoral artery and vein. Pemafibrate treatment enhanced blood flow recovery and capillary formation in ischemic limbs of mice, which was accompanied by enhanced phosphorylation of endothelial nitric oxide synthase (eNOS). Treatment of cultured endothelial cells with pemafibrate resulted in increased network formation and migratory activity, which were blocked by pretreatment with the NOS inhibitor NG-nitro-L-arginine methyl ester (L-NAME). Pemafibrate treatment also increased plasma levels of the PPARα-regulated gene, fibroblast growth factor (FGF) 21 in WT mice. Systemic administration of adenoviral vectors expressing FGF21 (Ad-FGF21) to WT mice enhanced blood flow recovery, capillary density and eNOS phosphorylation in ischemic limbs. Treatment of cultured endothelial cells with FGF21 protein led to increases in endothelial cell network formation and migration, which were canceled by pretreatment with L-NAME. Furthermore, administration of pemafibrate or Ad-FGF21 had no effects on blood flow in ischemic limbs in eNOS-deficient mice. CONCLUSION: These data suggest that pemafibrate can promote revascularization in response to ischemia, at least in part, through direct and FGF21-mediated modulation of endothelial cell function. Thus, pemafibrate could be a potentially beneficial drug for ischemic vascular disease.


Asunto(s)
Benzoxazoles/farmacología , Butiratos/farmacología , Isquemia/patología , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Miembro Posterior/irrigación sanguínea , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Ratones , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , PPAR alfa/química , PPAR alfa/metabolismo , Fosforilación/efectos de los fármacos
9.
Sci Rep ; 10(1): 7623, 2020 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-32376995

RESUMEN

Small-molecule agonism of peroxisome proliferator-activated receptor α (PPARα), a ligand-activated transcriptional factor involved in regulating fatty acid metabolism, is an important approach for treating dyslipidemia. Here, we determined the structures of the ligand-binding domain (LBD) of PPARα in complex with 1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid derivatives, which were recently identified as PPARα-selective activators with markedly different structures from those of the well-known PPARα agonists fibrates. The crystal structures of the complexes showed that they form a canonical hydrogen-bond network involving helix 12 in the LBD, which is thought to be essential for PPARα activation, as also observed for fibrates. However, the phenyl side chain of the compounds occupies a small cavity between Ile272 and Ile354, which is rarely accessed by fibrates. This unique feature may be essential for subtype selectivity and combine with the well-characterized binding mode of fibrates to improve activity. These findings demonstrate the advantage of using 1H-pyrazolo-[3,4-b]pyridine as a skeleton of PPARα agonists and provide insight into the design of molecules for treating dyslipidemia.


Asunto(s)
PPAR alfa/metabolismo , Pirazoles/química , Piridinas/química , Piridinas/farmacología , Humanos , Ligandos , Simulación del Acoplamiento Molecular , PPAR alfa/química , Dominios Proteicos , Piridinas/metabolismo
10.
Fish Shellfish Immunol ; 102: 257-266, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32315742

RESUMEN

Peroxisome proliferative-activated receptor α (PPARα) belongs to the superfamily of nuclear receptors (NR). Studies have demonstrated that PPARα functions in energy metabolism, hepatic function, immune response, cell cycle, and apoptosis. In teleost fish, few studies have investigated the role of PPARα in the immune response. In this study, the grouper PPARα gene (EcPPARα) was investigated for its role in viral infection. The open reading frame of EcPPARα encoded a protein of 469 amino acids and contained an N-terminal domain (NTD), a DNA-binding domain (DBD), a hinge region, and a C-terminal ligand-binding domain (LBD). Phylogenetic analysis revealed that EcPPARα was most closely related to homologous genes in Sander lucioperca and Perca flavescens. Upon challenge with SGIV (Singapore grouper iridovirus) and RGNNV (Red-spotted grouper nervous necrosis virus), EcPPARα expression levels were significantly upregulated in different tissues. Subcellular localization analysis showed that the EcPPARα protein localized throughout the cytoplasm and nucleus with diffuse intracellular expression patterns, which is consistent with the localization pattern of mammalian PPARs. Based on morphological observation of cytopathic effect (CPEs), viral gene expression mRNAs, and virus titer assays, the results presented here showed that an overexpression of EcPPARα promoted SGIV production in grouper spleen cells. Overexpression of EcPPARα significantly inhibited the expression of several cytokines, including interferon-related genes (IFN-γ, ISG15, MXI, MXII, MAVS and MDA5), inflammatory cytokines (IL-1ß, IL-6, IL-8, TNF-α) and Toll like receptor adaptors (TRAF6 and MyD88). Luciferase activity of IFN-α, IFN-γ, ISRE and NF-κB promoters was also significantly decreased in EcPPARα overexpression cells. Due to these detected interferon-related genes and inflammatory cytokines play important antiviral effect against SGIV in grouper, we speculated that the promotion effect of EcPPARα on SGIV replication may be caused by down-regulation of interferon and inflammatory response. In addition, through apoptotic body observation, capspase-3 activity detection, and flow cytometry analysis, it was found that overexpression of EcPPARα promoted SGIV-induced apoptosis in fathead minnow (FHM) cells. These data may increase an understanding of the role of PPARα in fish antiviral immune responses and apoptosis.


Asunto(s)
Lubina/genética , Lubina/inmunología , Enfermedades de los Peces/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , PPAR alfa/genética , PPAR alfa/inmunología , Secuencia de Aminoácidos , Animales , Infecciones por Virus ADN/inmunología , Infecciones por Virus ADN/veterinaria , Proteínas de Peces/química , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Perfilación de la Expresión Génica/veterinaria , Nodaviridae/fisiología , PPAR alfa/química , Filogenia , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/veterinaria , Ranavirus/fisiología , Alineación de Secuencia/veterinaria
11.
Int J Mol Sci ; 21(1)2020 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-31935812

RESUMEN

The selective PPARα modulator (SPPARMα) is expected to medicate dyslipidemia with minimizing adverse effects. Recently, pemafibrate was screened from the ligand library as an SPPARMα bearing strong potency. Several clinical pieces of evidence have proved the usefulness of pemafibrate as a medication; however, how pemafibrate works as a SPPARMα at the molecular level is not fully known. In this study, we investigate the molecular mechanism behind its novel SPPARMα character through a combination of approaches of X-ray crystallography, isothermal titration calorimetry (ITC), and fragment molecular orbital (FMO) analysis. ITC measurements have indicated that pemafibrate binds more strongly to PPARα than to PPARγ. The crystal structure of PPARα-ligand binding domain (LBD)/pemafibrate/steroid receptor coactivator-1 peptide (SRC1) determined at 3.2 Å resolution indicates that pemafibrate binds to the ligand binding pocket (LBP) of PPARα in a Y-shaped form. The structure also reveals that the conformation of the phenoxyalkyl group in pemafibrate is flexible in the absence of SRC1 coactivator peptide bound to PPARα; this gives a freedom for the phenoxyalkyl group to adopt structural changes induced by the binding of coactivators. FMO calculations have indicated that the accumulation of hydrophobic interactions provided by the residues at the LBP improve the interaction between pemafibrate and PPARα compared with the interaction between fenofibrate and PPARα.


Asunto(s)
Benzoxazoles/farmacología , Butiratos/farmacología , Simulación del Acoplamiento Molecular , PPAR alfa/química , Benzoxazoles/química , Sitios de Unión , Butiratos/química , Cristalografía por Rayos X , Humanos , Simulación de Dinámica Molecular , PPAR alfa/metabolismo , Unión Proteica
12.
J Biomol Struct Dyn ; 38(2): 511-523, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-30767625

RESUMEN

Diabetes is a foremost health problem globally susceptible to increased mortality and morbidity. The present therapies in the antidiabetic class have sound adverse effects and thus, emphasis on the further need to develop effective medication therapy. Peroxisome proliferator-activated receptor alpha-gamma dual approach represents an interesting target for developing novel anti-diabetic drug along with potential anti-hyperlipidimic activity. In the current study, the peroxisome proliferator-activated receptor alpha-gamma agonistic hits were screened by hierarchical virtual screening of drug like compounds followed by molecular dynamics simulation and knowledge-based structure-activity relation analysis. The key amino acid residues of binding pockets of both target proteins were acknowledged as essential and were found to be associated in the key interactions with the most potential dual hit. This dual targeted approach of structure based computational technique was undertaken to identify prevalent promising hits for both targets with binding energy and absorption distribution metabolism excretion prediction supported the analysis of their pharmacokinetic potential. In addition, stability analysis using molecular dynamics simulation of the target protein complexes was performed with the most promising dual targeted hit found in this study. Further, comparative analysis of binding site of both targets was done for the development of knowledge-based structure-activity relationship, which may useful for successful designing of dual agonistic candidates. AbbreviationsADMEabsorption distribution metabolism excretionHTVShighthroughput virtual screeningMDmolecular dynamicsMMGBSAmolecular mechanics generalized bonn solvation accessiblePDBprotein data bankPPARperoxisome proliferator-activated receptorRMSDRoot mean square deviationRMSFRoot mean square fluctuationSARstructural activity relationshipSPsimple precisionT2DMTypeII diabetes mellitusXPExtra precisionCommunicated by Ramaswamy H. Sarma.


Asunto(s)
Enfermedades Metabólicas/tratamiento farmacológico , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , PPAR alfa/agonistas , PPAR gamma/agonistas , Evaluación Preclínica de Medicamentos , Ligandos , PPAR alfa/química , PPAR gamma/química , Reproducibilidad de los Resultados , Relación Estructura-Actividad
13.
Int J Mol Sci ; 20(22)2019 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-31766193

RESUMEN

Pemafibrate is the first clinically-available selective peroxisome proliferator-activated receptor α modulator (SPPARMα) that has been shown to effectively improve hypertriglyceridemia and low high-density lipoprotein cholesterol (HDL-C) levels. Global gene expression analysis reveals that the activation of PPARα by pemafibrate induces fatty acid (FA) uptake, binding, and mitochondrial or peroxisomal oxidation as well as ketogenesis in mouse liver. Pemafibrate most profoundly induces HMGCS2 and PDK4, which regulate the rate-limiting step of ketogenesis and glucose oxidation, respectively, compared to other fatty acid metabolic genes in human hepatocytes. This suggests that PPARα plays a crucial role in nutrient flux in the human liver. Additionally, pemafibrate induces clinically favorable genes, such as ABCA1, FGF21, and VLDLR. Furthermore, pemafibrate shows anti-inflammatory effects in vascular endothelial cells. Pemafibrate is predicted to exhibit beneficial effects in patients with atherogenic dyslipidemia and diabetic microvascular complications.


Asunto(s)
Benzoxazoles/farmacología , Butiratos/farmacología , PPAR alfa/metabolismo , Transcriptoma/efectos de los fármacos , Animales , Ácidos Grasos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/metabolismo , Humanos , Modelos Moleculares , Oxidación-Reducción/efectos de los fármacos , PPAR alfa/agonistas , PPAR alfa/química
14.
Mol Metab ; 29: 55-64, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31668392

RESUMEN

OBJECTIVE: Fatty acids are important for biological function; however, in excess, they can cause metabolic dysregulation. Methods to image and detect fatty acids in real time are lacking. Therefore, the current study examined the dynamics of fatty acid trafficking and signaling utilizing novel fluorescent and luminescent approaches. METHODS: We generated fluorescent and luminescent-based genetically-encoded sensors based upon the ligand-dependent interaction between PPARα and SRC-1 to image and detect cellular dynamics of fatty acid trafficking. RESULTS: The use of a fluorescent sensor demonstrates that fatty acids traffic rapidly from lipid droplets to the nucleus. Both major lipases ATGL and HSL contribute to fatty acid signaling from lipid droplet to nucleus, however, their dynamics differ. Furthermore, direct activation of lipolysis, independent of receptor-mediated signaling is sufficient to promote lipid droplet to nuclear trafficking of fatty acids. A luminescent-based sensor that reports intracellular fatty acid levels is amenable to high-throughput analysis. CONCLUSIONS: Fatty acids traffic from lipid droplets to the nucleus within minutes of stimulated lipolysis. Genetically-encoded fluorescent and luminescent based sensors can be used to probe the dynamics of fatty acid trafficking and signaling.


Asunto(s)
Ácidos Grasos/metabolismo , Colorantes Fluorescentes/química , Microscopía Fluorescente , Transporte Biológico , Núcleo Celular/metabolismo , Colorimetría , Ácidos Grasos/análisis , Genes Reporteros , Células HEK293 , Humanos , Ligandos , Gotas Lipídicas/metabolismo , Lipólisis , Coactivador 1 de Receptor Nuclear/genética , Coactivador 1 de Receptor Nuclear/metabolismo , PPAR alfa/química , PPAR alfa/genética , PPAR alfa/metabolismo , Transducción de Señal , Esterol Esterasa/metabolismo
15.
J Alzheimers Dis ; 71(2): 389-397, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31424405

RESUMEN

Mounting evidence has identified that impaired amyloid-ß (Aß) clearance might contribute to Alzheimer's disease (AD) pathology. The lysosome-autophagy network plays an important role in protein homeostasis and cell health by removing abnormal protein aggregates via intracellular degradation. Therefore, stimulation of cellular degradative machinery for efficient removal of Aß has emerged as a growing field in AD research. However, mechanisms controlling such pathways and drugs to promote such mechanisms are poorly understood. Aspirin is a widely used drug throughout the world and recent studies have identified a new function of this drug. At low doses, aspirin stimulates lysosomal biogenesis and autophagy to clear amyloid plaques in an animal model of AD. This review delineates such functions of aspirin and analyzes underlying mechanisms that involve peroxisome proliferator-activated receptor alpha (PPARα)-mediated transcription of transcription factor EB (TFEB), the master regulator of lysosomal biogenesis.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Aspirina/metabolismo , PPAR alfa/metabolismo , Placa Amiloide/tratamiento farmacológico , Placa Amiloide/metabolismo , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Aspirina/uso terapéutico , Humanos , PPAR alfa/química , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , Estructura Secundaria de Proteína
16.
Ecotoxicol Environ Saf ; 181: 164-171, 2019 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-31185430

RESUMEN

Short-chain chlorinated paraffins (SCCPs) are frequently detected in environmental matrices and human tissues. It was hypothesized that SCCPs might interact with the peroxisome proliferator-activated receptor α (PPARα). In the present study, an in vitro, dual-luciferase reporter gene assay and in silico molecular docking analysis were employed together to study the interactions between SCCPs congeners and PPARα. Expressions of genes downstream in pathways activated by PPARα in liver of rats exposed to 1, 10, or 100 mg/kg bm/d of C10-13-CPs (56.5% Cl) for 28 days were examined to confirm activation potencies of SCCPs toward PPARα signaling. Effects of exposure to C10-13-CPs (56.5% Cl) on fatty acid metabolism in rat liver were also explored via a pseudo-targeted metabolomics strategy. Our results showed that C10-13-CPs (56.5% Cl) caused a dose-dependent greater expression of luciferase activity of rat PPARα. Molecular docking modeling revealed that SCCPs had a strong capacity to bind with PPARα only through hydrophobic interactions and the binding affinity was dependent on the degree of chlorination in SCCPs congeners. In livers of male rats, exposure to 100 mg/kg bm/d of C10-13-CPs (56.5% Cl) resulted in up-regulated expressions of 11 genes that are downstream in the PPARα-activated pathway and regulate catabolism of fatty acid. Consistently, accelerated fatty acid oxidation was observed mainly characterized by lesser concentrations of ∑fatty acids in livers of rats. Overall, these results demonstrated, for the first time, that SCCPs could activate rat PPARα signaling and thereby disrupt metabolism of fatty acid in livers of male rats.


Asunto(s)
Ácidos Grasos/metabolismo , Hígado/efectos de los fármacos , PPAR alfa/metabolismo , Parafina/toxicidad , Animales , Expresión Génica/efectos de los fármacos , Genes Reporteros , Halogenación , Hígado/metabolismo , Luciferasas/genética , Masculino , Simulación del Acoplamiento Molecular , PPAR alfa/química , Parafina/química , Ratas , Transducción de Señal , Regulación hacia Arriba
17.
Sci Rep ; 9(1): 6918, 2019 05 06.
Artículo en Inglés | MEDLINE | ID: mdl-31061404

RESUMEN

Peroxisome proliferator-activated receptor alfa (PPARA/NR1C1) is a ligand activated nuclear receptor that is a key regulator of lipid metabolism in tissues with high fatty acid catabolism such as the liver. Here, we cloned PPARA from polar bear liver tissue and studied in vitro transactivation of polar bear and human PPARA by environmental contaminants using a luciferase reporter assay. Six hinge and ligand-binding domain amino acids have been substituted in polar bear PPARA compared to human PPARA. Perfluorocarboxylic acids (PFCA) and perfluorosulfonic acids induced the transcriptional activity of both human and polar bear PPARA. The most abundant PFCA in polar bear tissue, perfluorononanoate, increased polar bear PPARA-mediated luciferase activity to a level comparable to that of the potent PPARA agonist WY-14643 (~8-fold, 25 µM). Several brominated flame retardants were weak agonists of human and polar bear PPARA. While single exposures to polychlorinated biphenyls did not, or only slightly, increase the transcriptional activity of PPARA, a technical mixture of PCBs (Aroclor 1254) strongly induced the transcriptional activity of human (~8-fold) and polar bear PPARA (~22-fold). Polar bear PPARA was both quantitatively and qualitatively more susceptible than human PPARA to transactivation by less lipophilic compounds.


Asunto(s)
Contaminantes Ambientales/toxicidad , PPAR alfa/genética , Transcripción Genética/efectos de los fármacos , Ursidae , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Evolución Molecular , Humanos , Modelos Moleculares , PPAR alfa/química , Bifenilos Policlorados/toxicidad , Conformación Proteica , Alineación de Secuencia , Especificidad de la Especie
18.
ChemMedChem ; 14(14): 1343-1348, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31141287

RESUMEN

Automated computational analogue design and scoring can speed up hit-to-lead optimization and appears particularly promising in selective optimization of side-activities (SOSA) where possible analogue diversity is confined. Probing this concept, we employed the cysteinyl leukotriene receptor 1 (CysLT1 R) antagonist cinalukast as lead for which we discovered peroxisome proliferator-activated receptor α (PPARα) modulatory activity. We automatically generated a virtual library of close analogues and classified these roughly 8000 compounds for PPARα agonism and CysLT1 R antagonism using automated affinity scoring and machine learning. A computationally preferred analogue for SOSA was synthesized, and in vitro characterization indeed revealed a marked activity shift toward enhanced PPARα activation and diminished CysLT1 R antagonism. Thereby, this prospective application study highlights the potential of automating SOSA.


Asunto(s)
PPAR alfa/agonistas , Bibliotecas de Moléculas Pequeñas/química , Sitios de Unión , Humanos , Antagonistas de Leucotrieno/química , Ligandos , Simulación del Acoplamiento Molecular , PPAR alfa/química , PPAR alfa/metabolismo , Prueba de Estudio Conceptual , Receptores de Leucotrienos/química , Bibliotecas de Moléculas Pequeñas/metabolismo , Tiazoles/química
19.
J Food Drug Anal ; 27(1): 295-304, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30648583

RESUMEN

Peroxisome proliferator-activated receptor α (PPARα) is a nuclear hormone receptor that transcriptionally regulates lipid metabolism and inflammation; therefore, PPARα agonists are promising agents to treat dyslipidemia and metabolic disorders. PPARα full agonists, such as fibrates, are effective anti-hypertriglyceride agents, but their use is limited by adverse side effects. Hence, the aim of this study was to identify small molecules that can activate PPARα while minimizing the adverse effects. Antrodia cinnamomea, a rare medical mushroom, has been used widely in Asian countries for the treatment of various diseases, including liver diseases. Antcin B, H and K (antcins) and ergostatrien-3ß-ol (EK100) are bioactive compounds isolated from A. cinnamomea with anti-inflammatory actions. Antcins, ergostane-type triterpenoids, contain the polar head with carboxylate group and the sterol-based body. Here, we showed at the first time that sterol-based compounds, antcins, but not EK100, activate PPARα in a cell-based transactivation study. The in silico docking studies presented several significant molecular interactions of antcins, including Tyr314, and His440 in the ligand-binding domain of PPARα, and these interactions are required for helix 12 (H12) stabilization. We propose that PPARα activation activity of antcins is related to their binding mode which requires conventional H12 stabilization, and that antcins can be developed as safe selective PPARα modulators.


Asunto(s)
Antrodia/química , Colestenos/química , Colestenonas/química , Ergosterol/análogos & derivados , PPAR alfa/agonistas , Extractos Vegetales/química , Triterpenos/química , Ergosterol/química , Humanos , Simulación del Acoplamiento Molecular , PPAR alfa/química , PPAR alfa/metabolismo
20.
Biochim Biophys Acta Gen Subj ; 1863(3): 586-597, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30611848

RESUMEN

BACKGROUND: The nuclear Peroxisome Proliferator Activated Receptors (PPARs) are ligand-activated transcription factors playing a fundamental role in energy homeostasis and metabolism. Consequently, functional impairment or dysregulation of these receptors lead to a variety of metabolic diseases. While some phytocannabinoids (pCBs) are known to activate PPARγ, no data have been reported so far on their possible activity at PPARα. METHODS: The putative binding modes of pCBs into PPARα/γ Ligand Binding Domains were found and assessed by molecular docking and molecular dynamics. Luciferase assays validated in silico predictions whereas the biological effects of such PPARα/γ ligands were assessed in HepG2 and 3T3L1 cell cultures. RESULTS: The in silico study identified cannabigerolic acid (CBGA), cannabidiolic acid (CBDA) and cannabigerol (CBG) from C. sativa as PPARα/γ dual agonists, suggesting their binding modes toward PPARα/γ isoforms and predicting their activity as full or partial agonists. These predictions were confirmed by luciferase functional assays. The resulting effects on downstream gene transcription in adipocytes and hepatocytes were also observed, establishing their actions as functional dual agonists. CONCLUSIONS: Our work broadens the activity spectrum of CBDA, CBGA and CBG by providing evidence that these pCBs act as dual PPARα/γ agonists with the ability to modulate the lipid metabolism. GENERAL SIGNIFICANCE: Dual PPARα/γ agonists have emerged as an attractive alternative to selective PPAR agonists to treat metabolic disorders. We identified some pCBs as dual PPARα/γ agonists, potentially useful for the treatment of dyslipidemia and type 2 diabetes mellitus.


Asunto(s)
Cannabinoides/análisis , Cannabinoides/aislamiento & purificación , PPAR alfa/agonistas , PPAR gamma/agonistas , Fitoquímicos , Células 3T3-L1 , Animales , Células COS , Cannabinoides/química , Cannabinoides/farmacología , Chlorocebus aethiops , Biología Computacional , Regulación de la Expresión Génica/efectos de los fármacos , Células Hep G2 , Humanos , Ratones , Modelos Moleculares , Simulación del Acoplamiento Molecular , PPAR alfa/química , PPAR alfa/metabolismo , PPAR gamma/química , PPAR gamma/metabolismo , Fitoquímicos/análisis , Fitoquímicos/química , Fitoquímicos/aislamiento & purificación , Fitoquímicos/farmacología , Unión Proteica , Elementos de Respuesta/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...