Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nat Chem Biol ; 20(3): 365-372, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37828400

ABSTRACT

Stimulator of interferon genes (STING) is a dimeric transmembrane adapter protein that plays a key role in the human innate immune response to infection and has been therapeutically exploited for its antitumor activity. The activation of STING requires its high-order oligomerization, which could be induced by binding of the endogenous ligand, cGAMP, to the cytosolic ligand-binding domain. Here we report the discovery through functional screens of a class of compounds, named NVS-STGs, that activate human STING. Our cryo-EM structures show that NVS-STG2 induces the high-order oligomerization of human STING by binding to a pocket between the transmembrane domains of the neighboring STING dimers, effectively acting as a molecular glue. Our functional assays showed that NVS-STG2 could elicit potent STING-mediated immune responses in cells and antitumor activities in animal models.


Subject(s)
Adaptor Proteins, Signal Transducing , Membrane Proteins , Animals , Humans , Adaptor Proteins, Signal Transducing/metabolism , Biological Assay , Cytosol , Immunity, Innate , Ligands , Membrane Proteins/metabolism
2.
Nature ; 475(7357): 524-7, 2011 Jul 27.
Article in English | MEDLINE | ID: mdl-21796212

ABSTRACT

Epstein-Barr virus-induced gene 2 (EBI2, also known as GPR183) is a G-protein-coupled receptor that is required for humoral immune responses; polymorphisms in the receptor have been associated with inflammatory autoimmune diseases. The natural ligand for EBI2 has been unknown. Here we describe the identification of 7α,25-dihydroxycholesterol (also called 7α,25-OHC or 5-cholesten-3ß,7α,25-triol) as a potent and selective agonist of EBI2. Functional activation of human EBI2 by 7α,25-OHC and closely related oxysterols was verified by monitoring second messenger readouts and saturable, high-affinity radioligand binding. Furthermore, we find that 7α,25-OHC and closely related oxysterols act as chemoattractants for immune cells expressing EBI2 by directing cell migration in vitro and in vivo. A critical enzyme required for the generation of 7α,25-OHC is cholesterol 25-hydroxylase (CH25H). Similar to EBI2 receptor knockout mice, mice deficient in CH25H fail to position activated B cells within the spleen to the outer follicle and mount a reduced plasma cell response after an immune challenge. This demonstrates that CH25H generates EBI2 biological activity in vivo and indicates that the EBI2-oxysterol signalling pathway has an important role in the adaptive immune response.


Subject(s)
Hydroxycholesterols/pharmacology , Receptors, Cell Surface/immunology , Animals , Antibody Formation/immunology , B-Lymphocytes , Cell Line , Cell Movement/drug effects , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Hydroxycholesterols/chemistry , Liver/chemistry , Mice , Mice, Knockout , Receptors, G-Protein-Coupled , Sheep , T-Lymphocytes/immunology
3.
J Am Chem Soc ; 135(5): 1669-72, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23330637

ABSTRACT

The identification of factors that promote ß cell proliferation could ultimately move type 1 diabetes treatment away from insulin injection therapy and toward a cure. We have performed high-throughput, cell-based screens using rodent ß cell lines to identify molecules that induce proliferation of ß cells. Herein we report the discovery and characterization of WS6, a novel small molecule that promotes ß cell proliferation in rodent and human primary islets. In the RIP-DTA mouse model of ß cell ablation, WS6 normalized blood glucose and induced concomitant increases in ß cell proliferation and ß cell number. Affinity pulldown and kinase profiling studies implicate Erb3 binding protein-1 and the IκB kinase pathway in the mechanism of action of WS6.


Subject(s)
High-Throughput Screening Assays , Islets of Langerhans/drug effects , Urea/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Islets of Langerhans/cytology , Mice , Molecular Structure , Molecular Weight , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
4.
Proc Natl Acad Sci U S A ; 107(38): 16542-7, 2010 Sep 21.
Article in English | MEDLINE | ID: mdl-20823227

ABSTRACT

Adult neurogenesis occurs in mammals and provides a mechanism for continuous neural plasticity in the brain. However, little is known about the molecular mechanisms regulating hippocampal neural progenitor cells (NPCs) and whether their fate can be pharmacologically modulated to improve neural plasticity and regeneration. Here, we report the characterization of a small molecule (KHS101) that selectively induces a neuronal differentiation phenotype. Mechanism of action studies revealed a link of KHS101 to cell cycle exit and specific binding to the TACC3 protein, whose knockdown in NPCs recapitulates the KHS101-induced phenotype. Upon systemic administration, KHS101 distributed to the brain and resulted in a significant increase in neuronal differentiation in vivo. Our findings indicate that KHS101 accelerates neuronal differentiation by interaction with TACC3 and may provide a basis for pharmacological intervention directed at endogenous NPCs.


Subject(s)
Cell Differentiation/drug effects , Neurogenesis/drug effects , Neurons/cytology , Neurons/drug effects , Thiazoles/pharmacology , Adult Stem Cells/cytology , Adult Stem Cells/drug effects , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Hippocampus/cytology , Male , Neurons/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley , Thiazoles/chemistry , Thiazoles/pharmacokinetics
5.
Proc Natl Acad Sci U S A ; 107(8): 3552-7, 2010 Feb 23.
Article in English | MEDLINE | ID: mdl-20133595

ABSTRACT

Approximately 3,500 mammalian genes are predicted to be secreted or single-pass transmembrane proteins. The function of the majority of these genes is still unknown, and a number of the encoded proteins might find use as new therapeutic agents themselves or as targets for small molecule or antibody drug development. To analyze the physiological activities of the extracellular proteome, we developed a large-scale, high-throughput protein expression, purification, and screening platform. For this study, the complete human extracellular proteome was analyzed and prioritized based on genome-wide disease association studies to select 529 initial target genes. These genes were cloned into three expression vectors as native sequences and as N-terminal and C-terminal Fc fusions to create an initial collection of 806 purified secreted proteins. To determine its utility, this library was screened in an OCT4-based cellular assay to identify regulators of human embryonic stem-cell self-renewal. We found that the pigment epithelium-derived factor can promote long-term pluripotent growth of human embryonic stem cells without bFGF or TGFbeta/Activin/Nodal ligand supplementation. Our results further indicate that activation of the pigment epithelium-derived factor receptor-Erk1/2 signaling pathway by the pigment epithelium-derived factor is sufficient to maintain the self-renewal of pluripotent human embryonic stem cells. These experiments illustrate the potential for discovering novel biological functions by directly screening protein diversity in cell-based phenotypic or reporter assays.


Subject(s)
Embryonic Stem Cells/metabolism , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Proteome/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Genome-Wide Association Study , High-Throughput Screening Assays , Humans , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Octamer Transcription Factor-3/genetics , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/drug effects , Proteome/genetics , Receptors, Neuropeptide/genetics , Receptors, Neuropeptide/metabolism , Signal Transduction
6.
Proc Natl Acad Sci U S A ; 106(22): 8912-7, 2009 Jun 02.
Article in English | MEDLINE | ID: mdl-19447925

ABSTRACT

Ectopic expression of defined transcription factors can reprogram somatic cells to induced pluripotent stem (iPS) cells, but the utility of iPS cells is hampered by the use of viral delivery systems. Small molecules offer an alternative to replace virally transduced transcription factors with chemical signaling cues responsible for reprogramming. In this report we describe a small-molecule screening platform applied to identify compounds that functionally replace the reprogramming factor Klf4. A series of small-molecule scaffolds were identified that activate Nanog expression in mouse fibroblasts transduced with a subset of reprogramming factors lacking Klf4. Application of one such molecule, kenpaullone, in lieu of Klf4 gave rise to iPS cells that are indistinguishable from murine embryonic stem cells. This experimental platform can be used to screen large chemical libraries in search of novel compounds to replace the reprogramming factors that induce pluripotency. Ultimately, such compounds may provide mechanistic insight into the reprogramming process.


Subject(s)
Benzazepines/pharmacology , Cell Differentiation , Epigenesis, Genetic/drug effects , Fibroblasts/drug effects , Indoles/pharmacology , Pluripotent Stem Cells/cytology , Small Molecule Libraries/pharmacology , Animals , Fibroblasts/cytology , Genes, Reporter , Homeodomain Proteins/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Luciferases/genetics , Mice , Nanog Homeobox Protein
7.
Cell Metab ; 3(5): 367-78, 2006 May.
Article in English | MEDLINE | ID: mdl-16679294

ABSTRACT

Insulin resistance is a primary defect in type 2 diabetes characterized by impaired peripheral glucose uptake and insufficient suppression of hepatic glucose output. Insulin signaling inhibits liver glucose production by inducing nuclear exclusion of the gluconeogenic transcription factor FOXO1 in an Akt-dependent manner. Through the concomitant application of genome-scale functional screening and quantitative image analysis, we have identified PTP-MEG2 as a modulator of insulin-dependent FOXO1 subcellular localization. Ectopic expression of PTP-MEG2 in cells inhibited insulin-induced phosphorylation of the insulin receptor, while RNAi-mediated reduction of PTP-MEG2 transcript levels enhanced insulin action. Additionally, adenoviral-mediated depletion of PTP-MEG2 in livers of diabetic (db/db) mice resulted in insulin sensitization and normalization of hyperglycemia. These data implicate PTP-MEG2 as a mediator of blood glucose homeostasis through antagonism of insulin signaling, and suggest that modulation of PTP-MEG2 activity may be an effective strategy in the treatment of type 2 diabetes.


Subject(s)
Insulin/metabolism , Liver/metabolism , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Animals , Blood Glucose/metabolism , Cell Line, Tumor , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/enzymology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/metabolism , Green Fluorescent Proteins/genetics , Hepatocytes/drug effects , Hepatocytes/enzymology , Humans , Insulin Resistance , Liver/drug effects , Liver/enzymology , Male , Mice , Promoter Regions, Genetic , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases, Non-Receptor , RNA Interference , Rats , Rats, Sprague-Dawley , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Recombinant Fusion Proteins/metabolism , Transfection
8.
Bioorg Med Chem ; 17(3): 977-80, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-18329275

ABSTRACT

A small molecule inhibitor of alpha4 integrin-dependent cell migration was identified through a cell-based screen of small molecule libraries. Biochemical and cellular experiments suggest that this molecule functions by interacting with gamma-parvin. This molecule should serve as a useful tool to study alpha4 integrin signaling and may lead to new therapeutics for the treatment of autoimmune diseases.


Subject(s)
Aniline Compounds/pharmacology , Cell Movement/drug effects , Integrin alpha4/metabolism , Tubercidin/analogs & derivatives , Actinin/antagonists & inhibitors , Actinin/metabolism , Adenosine Kinase/antagonists & inhibitors , Adenosine Kinase/metabolism , Aniline Compounds/chemical synthesis , Aniline Compounds/chemistry , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Humans , Integrin alpha4/drug effects , Jurkat Cells , RNA Interference , Signal Transduction , Small Molecule Libraries , Tubercidin/chemical synthesis , Tubercidin/chemistry , Tubercidin/pharmacology
9.
Cell Rep ; 25(11): 3074-3085.e5, 2018 12 11.
Article in English | MEDLINE | ID: mdl-30540940

ABSTRACT

Intratumoral (IT) STING activation results in tumor regression in preclinical models, yet factors dictating the balance between innate and adaptive anti-tumor immunity are unclear. Here, clinical candidate STING agonist ADU-S100 (S100) is used in an IT dosing regimen optimized for adaptive immunity to uncover requirements for a T cell-driven response compatible with checkpoint inhibitors (CPIs). In contrast to high-dose tumor ablative regimens that result in systemic S100 distribution, low-dose immunogenic regimens induce local activation of tumor-specific CD8+ effector T cells that are responsible for durable anti-tumor immunity and can be enhanced with CPIs. Both hematopoietic cell STING expression and signaling through IFNAR are required for tumor-specific T cell activation, and in the context of optimized T cell responses, TNFα is dispensable for tumor control. In a poorly immunogenic model, S100 combined with CPIs generates a survival benefit and durable protection. These results provide fundamental mechanistic insights into STING-induced anti-tumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunity , Membrane Proteins/metabolism , Neoplasms/immunology , Animals , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Cytokines/metabolism , Dose-Response Relationship, Immunologic , Drug Resistance, Neoplasm , Hematopoiesis , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/pathology , Programmed Cell Death 1 Receptor/metabolism , S100 Proteins/administration & dosage , S100 Proteins/immunology
12.
Chem Biol ; 11(2): 247-59, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15123286

ABSTRACT

Hymenialdisine (HMD) is a sponge-derived natural product kinase inhibitor with nanomolar activity against CDKs, Mek1, GSK3beta, and CK1 and micromolar activity against Chk1. In order to explore the broader application of the pyrrolo[2,3-c]azepine skeleton of HMD as a general kinase inhibitory scaffold, we searched for additional protein targets using affinity chromatography in conjunction with the synthesis of diverse HMD analogs and profiled HMD against a panel of 60 recombinant enzymes. This effort has led to nanomolar to micromolar inhibitors of 11 new targets including p90RSK, KDR, c-Kit, Fes, MAPK1, PAK2, PDK1, PKCtheta, PKD2, Rsk1, and SGK. The synthesis of HMD analogs has resulted in the identification of compounds with enhanced and/or dramatically altered selectivities relative to HMD (28n) and in molecules with antiproliferative activities 30-fold higher than HMD (28p).


Subject(s)
Azepines/chemistry , Enzyme Inhibitors/chemistry , Phosphotransferases/antagonists & inhibitors , Pyrroles/chemistry , Amino Acid Sequence , Azepines/chemical synthesis , Azepines/pharmacology , Chromatography, Affinity , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Molecular Sequence Data , Phosphotransferases/metabolism , Pyrroles/chemical synthesis , Pyrroles/pharmacology , Structure-Activity Relationship
14.
ACS Chem Biol ; 8(7): 1407-11, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23621521

ABSTRACT

Retinal pigment epithelial (RPE) cells form a monolayer adjacent to the retina and play a critical role in the visual light cycle. Degeneration of RPE cells results in retinal disorders such as age-related macular degeneration. Cell transplant strategies have potential therapeutic value for such disorders; however, risks associated with an inadequate supply of donor cells limit their therapeutic success. The identification of factors that proliferate RPE cells ex vivo could provide a renewable source of cells for transplantation. Here, we report that a small molecule (WS3) can reversibly proliferate primary RPE cells isolated from fetal and adult human donors. Following withdrawal of WS3, RPE cells differentiate into a functional monolayer, as exhibited by their expression of mature RPE genes and phagocytosis of photoreceptor outer segments. Furthermore, chemically expanded RPE cells preserve vision when transplanted into dystrophic Royal College of Surgeons (RCS) rats, a well-established model of retinal degeneration.


Subject(s)
Biotin/analogs & derivatives , Epithelial Cells/cytology , Epithelial Cells/drug effects , Phenylurea Compounds/pharmacology , Pyrimidines/pharmacology , Retina/cytology , Small Molecule Libraries/pharmacology , Animals , Biotin/chemistry , Biotin/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Fetal Stem Cells , Fluorescent Antibody Technique , Humans , Molecular Structure , Phenylurea Compounds/chemistry , Pyrimidines/chemistry , Rats , Retina/drug effects , Retinal Degeneration/drug therapy
15.
Science ; 336(6082): 717-21, 2012 May 11.
Article in English | MEDLINE | ID: mdl-22491093

ABSTRACT

Osteoarthritis (OA) is a degenerative joint disease that involves the destruction of articular cartilage and eventually leads to disability. Molecules that promote the selective differentiation of multipotent mesenchymal stem cells (MSCs) into chondrocytes may stimulate the repair of damaged cartilage. Using an image-based high-throughput screen, we identified the small molecule kartogenin, which promotes chondrocyte differentiation (median effective concentration = 100 nM), shows chondroprotective effects in vitro, and is efficacious in two OA animal models. Kartogenin binds filamin A, disrupts its interaction with the transcription factor core-binding factor ß subunit (CBFß), and induces chondrogenesis by regulating the CBFß-RUNX1 transcriptional program. This work provides new insights into the control of chondrogenesis that may ultimately lead to a stem cell-based therapy for osteoarthritis.


Subject(s)
Anilides/pharmacology , Cartilage, Articular/cytology , Chondrocytes/drug effects , Chondrogenesis , Mesenchymal Stem Cells/drug effects , Osteoarthritis/drug therapy , Phthalic Acids/pharmacology , Anilides/administration & dosage , Anilides/chemistry , Anilides/therapeutic use , Animals , Cattle , Cell Nucleus/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrocytes/physiology , Contractile Proteins/metabolism , Core Binding Factor Alpha 2 Subunit/metabolism , Core Binding Factor beta Subunit/metabolism , Disease Models, Animal , Filamins , High-Throughput Screening Assays , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Microfilament Proteins/metabolism , Osteoarthritis/pathology , Osteoarthritis/physiopathology , Phthalic Acids/administration & dosage , Phthalic Acids/chemistry , Phthalic Acids/therapeutic use , Regeneration , Small Molecule Libraries , Structure-Activity Relationship
16.
PLoS One ; 7(2): e31226, 2012.
Article in English | MEDLINE | ID: mdl-22359577

ABSTRACT

Tumor progenitor cells represent a population of drug-resistant cells that can survive conventional chemotherapy and lead to tumor relapse. However, little is known of the role of tumor progenitors in prostate cancer metastasis. The studies reported herein show that the CXCR4/CXCL12 axis, a key regulator of tumor dissemination, plays a role in the maintenance of prostate cancer stem-like cells. The CXCL4/CXCR12 pathway is activated in the CD44(+)/CD133(+) prostate progenitor population and affects differentiation potential, cell adhesion, clonal growth and tumorigenicity. Furthermore, prostate tumor xenograft studies in mice showed that a combination of the CXCR4 receptor antagonist AMD3100, which targets prostate cancer stem-like cells, and the conventional chemotherapeutic drug Taxotere, which targets the bulk tumor, is significantly more effective in eradicating tumors as compared to monotherapy.


Subject(s)
Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/metabolism , Receptors, CXCR4/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols , Benzylamines , Cell Adhesion , Cell Proliferation , Chemokine CXCL12/metabolism , Cyclams , Docetaxel , Heterocyclic Compounds/pharmacology , Humans , Male , Mice , Neoplasm Metastasis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Taxoids/pharmacology
17.
ACS Chem Biol ; 6(11): 1223-31, 2011 Nov 18.
Article in English | MEDLINE | ID: mdl-21936526

ABSTRACT

Here we report the design and evaluation of a bifunctional, small molecule switch that induces a targeted immune response against tumors in vivo. A high affinity ligand for prostate specific membrane antigen (PSMA) was conjugated to a hapten that binds dinitrophenyl (DNP)-specific antibodies. When introduced into hu-PBL-NOD/SCID mice previously immunized with a KLH-DNP immunogen, this conjugate induced a targeted antibody-dependent cellular cytotoxicity (ADCC) response to PSMA-expressing tumor cells in a mouse xenograft model. The ability to create a small molecule inducible antibody response against self-antigens using endogenous non-autoreactive antibodies may provide advantages over the autologous immune response generated by conventional vaccines in certain therapeutic settings.


Subject(s)
2,4-Dinitrophenol/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Cancer Vaccines/chemistry , Cancer Vaccines/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , 2,4-Dinitrophenol/chemistry , Animals , Antibodies, Neoplasm/immunology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, Neoplasm/immunology , Antigens, Surface/metabolism , Autoantigens/immunology , Cancer Vaccines/therapeutic use , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Glutamate Carboxypeptidase II/metabolism , Humans , Ligands , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Prostatic Neoplasms/pathology
18.
Clin Cancer Res ; 16(23): 5692-702, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-21138868

ABSTRACT

PURPOSE: The cancer stem cell hypothesis predicts that standard prostate cancer monotherapy eliminates bulk tumor cells but not a tumor-initiating cell population, eventually leading to relapse. Many studies have sought to determine the underlying differences between bulk tumor and cancer stem cells. EXPERIMENTAL DESIGN: Our previous data suggest that the PTEN/PI3K/AKT pathway is critical for the in vitro maintenance of CD133(+)/CD44(+) prostate cancer progenitors and, consequently, that targeting PI3K signaling may be beneficial in treatment of prostate cancer. RESULTS: Here, we show that inhibition of PI3K activity by the dual PI3K/mTOR inhibitor NVP-BEZ235 leads to a decrease in the population of CD133(+)/CD44(+) prostate cancer progenitor cells in vivo. Moreover, the combination of the PI3K/mTOR modulator NVP-BEZ235, which eliminates prostate cancer progenitor populations, and the chemotherapeutic drug Taxotere, which targets the bulk tumor, is significantly more effective in eradicating tumors in a prostate cancer xenograft model than monotherapy. CONCLUSION: This combination treatment ultimately leads to the expansion of cancer progenitors with a PTEN E91D mutation, suggesting that the analysis of PTEN mutations could predict therapeutic response to the dual therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma/drug therapy , Neoplastic Stem Cells/drug effects , Prostatic Neoplasms/drug therapy , AC133 Antigen , Animals , Antigens, CD/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Drug Delivery Systems/methods , Glycoproteins/metabolism , Humans , Hyaluronan Receptors/metabolism , Imidazoles/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , PTEN Phosphohydrolase/genetics , Peptides/metabolism , Prostatic Neoplasms/pathology , Quinolines/administration & dosage , Xenograft Model Antitumor Assays
19.
Cell Stem Cell ; 4(5): 416-26, 2009 May 08.
Article in English | MEDLINE | ID: mdl-19427291

ABSTRACT

Embryonic stem cells (ESCs) are an attractive source of cells for disease modeling in vitro and may eventually provide access to cells/tissues for the treatment of many degenerative diseases. However, applications of ESC-derived cell types are largely hindered by the lack of highly efficient methods for lineage-specific differentiation. Using a high-content screen, we have identified a small molecule, named stauprimide, that increases the efficiency of the directed differentiation of mouse and human ESCs in synergy with defined extracellular signaling cues. Affinity-based methods revealed that stauprimide interacts with NME2 and inhibits its nuclear localization. This, in turn, leads to downregulation of c-Myc, a key regulator of the pluripotent state. Thus, our findings identify a chemical tool that primes ESCs for efficient differentiation through a mechanism that affects c-Myc expression, and this study points to an important role for NME2 in ESC self-renewal.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Enzyme Inhibitors/pharmacology , Staurosporine/analogs & derivatives , Animals , Cell Differentiation/drug effects , Chromatin Immunoprecipitation , Down-Regulation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endoderm/metabolism , Genes, myc , Humans , Mice , NM23 Nucleoside Diphosphate Kinases/antagonists & inhibitors , NM23 Nucleoside Diphosphate Kinases/genetics , NM23 Nucleoside Diphosphate Kinases/metabolism , Staurosporine/pharmacology
20.
ACS Chem Biol ; 2(3): 171-5, 2007 Mar 20.
Article in English | MEDLINE | ID: mdl-17348628

ABSTRACT

An essential function of the human epidermis is the maintenance of a protective barrier against the environment. As a consequence, keratinocytes, which make up this layer of the skin, undergo an elaborate process of self-renewal, terminal differentiation, and cell death. Misregulation of these processes can lead to several human diseases, including psoriasis and basal cell and squamous cell carcinomas. To identify novel regulators of keratinocyte differentiation, a cell-based screen of small-molecule libraries was carried out for molecules that induce terminal differentiation of normal human epidermal keratinocytes. One class of molecules was identified, the 2-(3,4,5-trimethoxyphenylamino)-pyrrolo[2,3-d]pyrimidines, which were shown to induce differentiation of epidermal progenitor cells to terminally differentiated keratinocytes. These molecules serve as useful mechanistic probes of the cellular differentiation programs that regulate the formation and homeostasis of the epidermis and may lead to novel therapeutic approaches for the treatment of skin hyperproliferative disorders.


Subject(s)
Cell Differentiation/drug effects , Epidermal Cells , Keratinocytes/cytology , Cell Division/drug effects , Epidermis/drug effects , Humans , Keratinocytes/drug effects , Pyrimidines/pharmacology , Pyrroles/pharmacology , Skin Diseases/drug therapy , Skin Diseases/pathology , Stem Cells/cytology , Stem Cells/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL