Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Mol Cell Neurosci ; 99: 103386, 2019 09.
Article in English | MEDLINE | ID: mdl-31202891

ABSTRACT

A subset of individuals with major depressive disorder (MDD) elects treatment with complementary and alternative medicines (CAMs), including the omega-3 fatty acids docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA). Previous studies in rodents suggest that DHA modulates neurodevelopmental processes, including adult neurogenesis and neuroplasticity, but the molecular and cellular mechanisms of DHA's potential therapeutic effect in the context of human neurobiology have not been well established. Here we sought to address this knowledge gap by investigating the effects of DHA using human iPSC-derived neural progenitor cells (NPCs) and post-mitotic neurons using pathway-selective reporter genes, multiplexed mRNA expression profiling, and a panel of metabolism-based viability assays. Finally, real-time, live-cell imaging was employed to monitor neurite outgrowth upon DHA treatment. Overall, these studies showed that DHA treatment (0-50 µM) significantly upregulated both WNT and CREB signaling pathways in human neuronal cells in a dose-dependent manner with 2- to 3-fold increases in pathway activation. Additionally, we observed that DHA treatment enhanced survival of iPSC-derived NPCs and differentiation of post-mitotic neurons with live-cell imaging, revealing increased neurite outgrowth with DHA treatment within 24 h. Taken together, this study provides evidence that DHA treatment activates critical pathways regulating neuroplasticity, which may contribute to enhanced neuronal cell viability and neuronal connectivity. The extent to which these pathways represent molecular mechanisms underlying the potential beneficial effects of omega-3 fatty acids in MDD and other brain disorders merits further investigation.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Docosahexaenoic Acids/pharmacology , Neural Stem Cells/metabolism , Wnt Signaling Pathway , Cell Line , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neuronal Outgrowth
2.
Cell ; 161(5): 999-1011, 2015 May 21.
Article in English | MEDLINE | ID: mdl-26000480

ABSTRACT

Despite all modern advances in medicine, an effective drug treatment of obesity has not been found yet. Discovery of leptin two decades ago created hopes for treatment of obesity. However, development of leptin resistance has been a big obstacle, mitigating a leptin-centric treatment of obesity. Here, by using in silico drug-screening methods, we discovered that Celastrol, a pentacyclic triterpene extracted from the roots of Tripterygium Wilfordi (thunder god vine) plant, is a powerful anti-obesity agent. Celastrol suppresses food intake, blocks reduction of energy expenditure, and leads to up to 45% weight loss in hyperleptinemic diet-induced obese (DIO) mice by increasing leptin sensitivity, but it is ineffective in leptin-deficient (ob/ob) and leptin receptor-deficient (db/db) mouse models. These results indicate that Celastrol is a leptin sensitizer and a promising agent for the pharmacological treatment of obesity.


Subject(s)
Anti-Obesity Agents/administration & dosage , Obesity/drug therapy , Animals , Anti-Obesity Agents/metabolism , Energy Metabolism , Gene Expression Profiling , Glucose/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Mice , Pentacyclic Triterpenes , Plant Extracts/administration & dosage , Tripterygium/chemistry , Triterpenes/administration & dosage
3.
Sci Transl Med ; 7(288): 288ra77, 2015 May 20.
Article in English | MEDLINE | ID: mdl-25995223

ABSTRACT

The emergence of drug resistance is a major limitation of current antimalarials. The discovery of new druggable targets and pathways including those that are critical for multiple life cycle stages of the malaria parasite is a major goal for developing next-generation antimalarial drugs. Using an integrated chemogenomics approach that combined drug resistance selection, whole-genome sequencing, and an orthogonal yeast model, we demonstrate that the cytoplasmic prolyl-tRNA (transfer RNA) synthetase (PfcPRS) of the malaria parasite Plasmodium falciparum is a biochemical and functional target of febrifugine and its synthetic derivative halofuginone. Febrifugine is the active principle of a traditional Chinese herbal remedy for malaria. We show that treatment with febrifugine derivatives activated the amino acid starvation response in both P. falciparum and a transgenic yeast strain expressing PfcPRS. We further demonstrate in the Plasmodium berghei mouse model of malaria that halofuginol, a new halofuginone analog that we developed, is active against both liver and asexual blood stages of the malaria parasite. Halofuginol, unlike halofuginone and febrifugine, is well tolerated at efficacious doses and represents a promising lead for the development of dual-stage next-generation antimalarials.


Subject(s)
Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Antimalarials/pharmacology , Enzyme Inhibitors/pharmacology , Malaria, Falciparum/drug therapy , Piperidines/pharmacology , Plasmodium falciparum/drug effects , Protozoan Proteins/antagonists & inhibitors , Quinazolines/pharmacology , Quinazolinones/pharmacology , Amino Acyl-tRNA Synthetases/metabolism , Animals , Antimalarials/chemistry , Antimalarials/toxicity , Computer-Aided Design , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Design , Drug Resistance , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/toxicity , Erythrocytes/parasitology , Liver/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Mice , Models, Molecular , Molecular Structure , Molecular Targeted Therapy , Piperidines/chemistry , Piperidines/toxicity , Plasmodium falciparum/enzymology , Protozoan Proteins/metabolism , Quinazolines/chemistry , Quinazolines/toxicity , Quinazolinones/chemistry , Quinazolinones/toxicity , Structure-Activity Relationship , Time Factors
4.
J Surg Res ; 194(2): 544-550, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25479907

ABSTRACT

BACKGROUND: Lipopolysaccharide (LPS) has a deleterious effect on several organs, including the liver, and eventually leads to endotoxic shock and death. LPS-induced hepatotoxicity is characterized by disturbed intracellular redox balance and excessive reactive oxygen species (ROS) accumulation, leading to liver injury. We have shown that treatment with suberoylanilide hydroxamic acid (SAHA), a histone deacetylase inhibitor, improves survival in a murine model of LPS-induced shock, but the protective effect of SAHA against liver damage remains unknown. The goal of this study was to investigate the mechanism underlying SAHA action in murine livers. METHOD: Male C57BL/6J mice (6-8 wk), weighing 20-25 g, were randomly divided into three groups: (A) a sham group was given isotonic sodium chloride solution (10 µL/g body weight, intraperitoneal, i.p.) with dimethyl sulfoxide (DMSO; 1 µL/g body weight, i.p.); (B) an LPS group was challenged with LPS (20 mg/kg, i.p.) dissolved in isotonic sodium chloride solution with DMSO; (C) and an LPS plus SAHA group was treated with SAHA (50 mg/kg, i.p.) dissolved in DMSO immediately after injection of LPS (20 mg/kg, i.p.). Mice were anesthetized, and their livers were harvested 6 or 24 h after injection to analyze whether SAHA affected production of ROS and activation of apoptotic proteins in the liver cells of challenged mice. RESULTS: SAHA counteracted LPS-induced production of ROS (thiobarbituric acid reactive substances and nitrite) and reversed an LPS-induced decrease in antioxidant enzyme, glutathione. SAHA also attenuated LPS-induced hepatic apoptosis. Moreover, SAHA inhibited activation of the redox-sensitive kinase, apoptosis signal-regulating kinase-1, and the mitogen-activated protein kinases, p38 and Jun N-terminal kinase. CONCLUSIONS: Our data indicate, for the first time, that SAHA is capable of alleviating LPS-induced hepatotoxicity and suggest that a blockade of the upstream events required for apoptosis signal-regulating kinase-1 action may serve as a new therapeutic option in the treatment of LPS-induced inflammatory conditions.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cyclooxygenase 2/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Lipopolysaccharides , Liver/drug effects , Liver/enzymology , MAP Kinase Kinase Kinase 5/metabolism , Male , Mice, Inbred C57BL , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Random Allocation , Vorinostat
5.
Chembiochem ; 15(13): 1920-30, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25111632

ABSTRACT

Malaria, an infectious disease caused by eukaryotic parasites of the genus Plasmodium, afflicts hundreds of millions of people every year. Both the parasite and its host utilize protein kinases to regulate essential cellular processes. Bioinformatic analyses of parasite genomes predict at least 65 protein kinases, but their biological functions and therapeutic potential are largely unknown. We profiled 1358 small-molecule kinase inhibitors to evaluate the role of both the human and the malaria kinomes in Plasmodium infection of liver cells, the parasites' obligatory but transient developmental stage that precedes the symptomatic blood stage. The screen identified several small molecules that inhibit parasite load in liver cells, some with nanomolar efficacy, and each compound was subsequently assessed for activity against blood-stage malaria. Most of the screening hits inhibited both liver- and blood-stage malaria parasites, which have dissimilar gene expression profiles and infect different host cells. Evaluation of existing kinase activity profiling data for the library members suggests that several kinases are essential to malaria parasites, including cyclin-dependent kinases (CDKs), glycogen synthase kinases, and phosphoinositide-3-kinases. CDK inhibitors were found to bind to Plasmodium protein kinase 5, but it is likely that these compounds target multiple parasite kinases. The dual-stage inhibition of the identified kinase inhibitors makes them useful chemical probes and promising starting points for antimalarial development.


Subject(s)
Genome, Protozoan/genetics , Malaria/genetics , Plasmodium/genetics , Protein Kinases/genetics , Animals , Antimalarials/chemistry , Computational Biology , Drug Evaluation, Preclinical , Humans , Liver/parasitology , Malaria/parasitology , Male , Mice , Mice, Inbred C57BL , Plasmodium/enzymology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries
6.
Nat Chem Biol ; 8(3): 311-7, 2012 Feb 12.
Article in English | MEDLINE | ID: mdl-22327401

ABSTRACT

Febrifugine, the bioactive constituent of one of the 50 fundamental herbs of traditional Chinese medicine, has been characterized for its therapeutic activity, though its molecular target has remained unknown. Febrifugine derivatives have been used to treat malaria, cancer, fibrosis and inflammatory disease. We recently demonstrated that halofuginone (HF), a widely studied derivative of febrifugine, inhibits the development of T(H)17-driven autoimmunity in a mouse model of multiple sclerosis by activating the amino acid response (AAR) pathway. Here we show that HF binds glutamyl-prolyl-tRNA synthetase (EPRS), inhibiting prolyl-tRNA synthetase activity; this inhibition is reversed by the addition of exogenous proline or EPRS. We further show that inhibition of EPRS underlies the broad bioactivities of this family of natural product derivatives. This work both explains the molecular mechanism of a promising family of therapeutics and highlights the AAR pathway as an important drug target for promoting inflammatory resolution.


Subject(s)
Amino Acyl-tRNA Synthetases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Piperidines/pharmacology , Quinazolines/pharmacology , Quinazolinones/pharmacology , Amino Acyl-tRNA Synthetases/chemistry , Amino Acyl-tRNA Synthetases/metabolism , Animals , Cell Differentiation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Mice , Mice, Inbred C57BL , Piperidines/chemistry , Quinazolines/chemistry , Quinazolinones/chemistry , Structure-Activity Relationship , Th17 Cells/drug effects , Th17 Cells/enzymology , Th17 Cells/immunology , Th17 Cells/metabolism
7.
ACS Chem Biol ; 4(7): 527-33, 2009 Jul 17.
Article in English | MEDLINE | ID: mdl-19572548

ABSTRACT

The nematode Caenorhabditis elegans is a unique whole animal model system for identifying small molecules with in vivo anti-infective properties. C. elegans can be infected with a broad range of human pathogens, including Enterococcus faecalis, an important human nosocomial pathogen. Here, we describe an automated, high-throughput screen of 37,200 compounds and natural product extracts for those that enhance survival of C. elegans infected with E. faecalis. Using a robot to dispense live, infected animals into 384-well plates and automated microscopy and image analysis, we identified 28 compounds and extracts not previously reported to have antimicrobial properties, including six structural classes that cure infected C. elegans animals but do not affect the growth of the pathogen in vitro, thus acting by a mechanism of action distinct from antibiotics currently in clinical use.


Subject(s)
Anti-Bacterial Agents/pharmacology , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/microbiology , Disease Models, Animal , Enterococcus faecalis/drug effects , Animals , Anti-Bacterial Agents/chemistry , Combinatorial Chemistry Techniques , Drug Evaluation, Preclinical , Enterococcus faecalis/growth & development , Gram-Positive Bacterial Infections/drug therapy , Humans , Molecular Structure
8.
Antimicrob Agents Chemother ; 53(6): 2564-8, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19307367

ABSTRACT

Malaria infects 500 million people annually, a number that is likely to rise as drug resistance to currently used antimalarials increases. During its intraerythrocytic stage, the causative parasite, Plasmodium falciparum, metabolizes hemoglobin and releases toxic heme, which is neutralized by a parasite-specific crystallization mechanism to form hemozoin. Evidence suggests that chloroquine, the most successful antimalarial agent in history, acts by disrupting the formation of hemozoin. Here we describe the development of a 384-well microtiter plate screen to detect small molecules that can also disrupt heme crystallization. This assay, which is based on a colorimetric assay developed by Ncokazi and Egan (K. K. Ncokazi and T. J. Egan, Anal. Biochem. 338:306-319, 2005), requires no parasites or parasite-derived reagents and no radioactive materials and is suitable for a high-throughput screening platform. The assay's reproducibility and large dynamic range are reflected by a Z factor of 0.74. A pilot screen of 16,000 small molecules belonging to diverse structural classes was conducted. The results of the target-based assay were compared with a whole-parasite viability assay of the same small molecules to identify small molecules active in both assays.


Subject(s)
Antimalarials/pharmacology , Colorimetry/methods , Drug Evaluation, Preclinical/methods , Heme/chemistry , Hemeproteins/biosynthesis , Animals , Crystallization , Dose-Response Relationship, Drug , Plasmodium falciparum/drug effects , Plasmodium falciparum/growth & development
9.
Proc Natl Acad Sci U S A ; 105(28): 9751-6, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18607002

ABSTRACT

The discovery of new small molecules and their testing in rational combination poses an ongoing problem for rare diseases, in particular, for pediatric cancers such as neuroblastoma. Despite maximal cytotoxic therapy with double autologous stem cell transplantation, outcome remains poor for children with high-stage disease. Because differentiation is aberrant in this malignancy, compounds that modulate transcription, such as histone deacetylase (HDAC) inhibitors, are of particular interest. However, as single agents, HDAC inhibitors have had limited efficacy. In the present study, we use an HDAC inhibitor as an enhancer to screen a small-molecule library for compounds inducing neuroblastoma maturation. To quantify differentiation, we use an enabling gene expression-based screening strategy. The top hit identified in the screen was all-trans-retinoic acid. Secondary assays confirmed greater neuroblastoma differentiation with the combination of an HDAC inhibitor and a retinoid versus either alone. Furthermore, effects of combination therapy were synergistic with respect to inhibition of cellular viability and induction of apoptosis. In a xenograft model of neuroblastoma, animals treated with combination therapy had the longest survival. This work suggests that testing of an HDAC inhibitor and retinoid in combination is warranted for children with neuroblastoma and demonstrates the success of a signature-based screening approach to prioritize compound combinations for testing in rare diseases.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Differentiation/drug effects , Drug Evaluation, Preclinical/methods , Histone Deacetylase Inhibitors , Neuroblastoma/drug therapy , Retinoids/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Survival , Drug Synergism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Expression Profiling , Humans , Neuroblastoma/pathology , Retinoids/therapeutic use , Small Molecule Libraries , Xenograft Model Antitumor Assays
10.
Bioorg Med Chem Lett ; 18(9): 2809-12, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18430569

ABSTRACT

Histone deacetylases (HDACs) regulate many important physiological processes and the discovery of small molecules that modulate HDAC activity has both academic and clinical relevance. HDAC inhibitors, most notably SAHA, have been pursued as cancer chemotherapeutics but may be useful in treating psychiatric disorders, malaria, and other diseases. Herein, we describe an inexpensive and robust assay, based on fluorescence polarization, for HDAC ligand discovery. The assay is well suited for high-throughput screening and enzyme kinetic studies.


Subject(s)
Biological Assay/methods , Enzyme Inhibitors/pharmacokinetics , Fluorescence Resonance Energy Transfer/methods , Fluorescent Dyes/chemistry , Histone Deacetylase Inhibitors , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Ligands , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL