Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Proc Natl Acad Sci U S A ; 111(6): 2325-30, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24449858

ABSTRACT

Small-fiber neuropathy (SFN) is a disorder of peripheral nerves commonly found in patients with diabetes mellitus, HIV infection, and those receiving chemotherapy. The complexity of disease etiology has led to a scarcity of effective treatments. Using two models of progressive SFN, we show that overexpression of glial cell line-derived neurotrophic factor (GDNF) in skin keratinocytes or topical application of XIB4035, a reported nonpeptidyl agonist of GDNF receptor α1 (GFRα1), are effective treatments for SFN. We also demonstrate that XIB4035 is not a GFRα1 agonist, but rather it enhances GFRα family receptor signaling in conjunction with ligand stimulation. Taken together, our results indicate that topical application of GFRα/RET receptor signaling modulators may be a unique therapy for SFN, and we have identified XIB4035 as a candidate therapeutic agent.


Subject(s)
Erythromelalgia/drug therapy , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Quinolines/therapeutic use , Signal Transduction , Administration, Topical , Animals , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Ligands , Mice , Mice, Transgenic , Quinolines/administration & dosage , Small Molecule Libraries
2.
Proc Natl Acad Sci U S A ; 107(39): 17005-10, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20837532

ABSTRACT

Recent studies indicate that molecules released by glia can induce synapse formation. However, what induces glia to produce such signals, their identity, and their in vivo relevance remain poorly understood. Here we demonstrate that supporting cells of the vestibular organ--cells that have many characteristics of glia--promote synapse formation only when induced by neuron-derived signals. Furthermore, we identify BDNF as the synaptogenic signal produced by these nonneuronal cells. Mice in which erbB signaling has been eliminated in supporting cells have vestibular dysfunction caused by failure of synapse formation between hair cells and sensory neurons. This phenotype correlates with reduced BDNF expression in supporting cells and is rescued by reexpression of BDNF in these cells. Furthermore, knockdown of BDNF expression in supporting cells postnatally phenocopies the loss of erbB signaling. These results indicate that vestibular supporting cells contribute in vivo to vestibular synapse formation and that this is mediated by reciprocal signals between sensory neurons and supporting cells involving erbB receptors and BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Epithelium/physiology , ErbB Receptors/metabolism , Neuroglia/physiology , Synapses/physiology , Vestibule, Labyrinth/physiology , Animals , ErbB Receptors/genetics , Glial Fibrillary Acidic Protein , Hair Cells, Vestibular/metabolism , Hair Cells, Vestibular/physiology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Receptor, ErbB-4 , Vestibule, Labyrinth/cytology
3.
J Med Chem ; 65(6): 4600-4615, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35293760

ABSTRACT

Inhibition of the S-adenosyl methionine (SAM)-producing metabolic enzyme, methionine adenosyltransferase 2A (MAT2A), has received significant interest in the field of medicinal chemistry due to its implication as a synthetic lethal target in cancers with the deletion of the methylthioadenosine phosphorylase (MTAP) gene. Here, we report the identification of novel MAT2A inhibitors with distinct in vivo properties that may enhance their utility in treating patients. Following a high-throughput screening, we successfully applied the structure-based design lessons from our first-in-class MAT2A inhibitor, AG-270, to rapidly redesign and optimize our initial hit into two new lead compounds: a brain-penetrant compound, AGI-41998, and a potent, but limited brain-penetrant compound, AGI-43192. We hope that the identification and first disclosure of brain-penetrant MAT2A inhibitors will create new opportunities to explore the potential therapeutic effects of SAM modulation in the central nervous system (CNS).


Subject(s)
Methionine Adenosyltransferase , Neoplasms , Brain/metabolism , Drug Design , Humans , Neoplasms/drug therapy , S-Adenosylmethionine/metabolism
4.
J Med Chem ; 64(8): 4430-4449, 2021 04 22.
Article in English | MEDLINE | ID: mdl-33829783

ABSTRACT

The metabolic enzyme methionine adenosyltransferase 2A (MAT2A) was recently implicated as a synthetic lethal target in cancers with deletion of the methylthioadenosine phosphorylase (MTAP) gene, which is adjacent to the CDKN2A tumor suppressor and codeleted with CDKN2A in approximately 15% of all cancers. Previous attempts to target MAT2A with small-molecule inhibitors identified cellular adaptations that blunted their efficacy. Here, we report the discovery of highly potent, selective, orally bioavailable MAT2A inhibitors that overcome these challenges. Fragment screening followed by iterative structure-guided design enabled >10 000-fold improvement in potency of a family of allosteric MAT2A inhibitors that are substrate noncompetitive and inhibit release of the product, S-adenosyl methionine (SAM), from the enzyme's active site. We demonstrate that potent MAT2A inhibitors substantially reduce SAM levels in cancer cells and selectively block proliferation of MTAP-null cells both in tissue culture and xenograft tumors. These data supported progressing AG-270 into current clinical studies (ClinicalTrials.gov NCT03435250).


Subject(s)
Enzyme Inhibitors/chemistry , Methionine Adenosyltransferase/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/genetics , Binding Sites , Crystallography, X-Ray , Drug Design , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/therapeutic use , Homozygote , Humans , Methionine Adenosyltransferase/metabolism , Molecular Dynamics Simulation , Neoplasms/drug therapy , Purine-Nucleoside Phosphorylase/metabolism , S-Adenosylmethionine/metabolism , Structure-Activity Relationship
5.
Cancer Cell ; 39(2): 209-224.e11, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33450196

ABSTRACT

The methylthioadenosine phosphorylase (MTAP) gene is located adjacent to the cyclin-dependent kinase inhibitor 2A (CDKN2A) tumor-suppressor gene and is co-deleted with CDKN2A in approximately 15% of all cancers. This co-deletion leads to aggressive tumors with poor prognosis that lack effective, molecularly targeted therapies. The metabolic enzyme methionine adenosyltransferase 2α (MAT2A) was identified as a synthetic lethal target in MTAP-deleted cancers. We report the characterization of potent MAT2A inhibitors that substantially reduce levels of S-adenosylmethionine (SAM) and demonstrate antiproliferative activity in MTAP-deleted cancer cells and tumors. Using RNA sequencing and proteomics, we demonstrate that MAT2A inhibition is mechanistically linked to reduced protein arginine methyltransferase 5 (PRMT5) activity and splicing perturbations. We further show that DNA damage and mitotic defects ensue upon MAT2A inhibition in HCT116 MTAP-/- cells, providing a rationale for combining the MAT2A clinical candidate AG-270 with antimitotic taxanes.


Subject(s)
DNA Damage/drug effects , Enzyme Inhibitors/pharmacology , Methionine Adenosyltransferase/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , Purine-Nucleoside Phosphorylase/genetics , RNA Splicing/drug effects , RNA, Messenger/genetics , Animals , Cell Line , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16 , DNA Damage/genetics , Gene Deletion , HCT116 Cells , HEK293 Cells , Humans , Methionine Adenosyltransferase/genetics , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasms/drug therapy , Neoplasms/genetics , RNA Splicing/genetics , S-Adenosylmethionine/metabolism
6.
ACS Med Chem Lett ; 11(10): 1899-1904, 2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33062171

ABSTRACT

Hepatocellular carcinoma (HCC) accounts for a majority of primary liver cancer and is one of the most common forms of cancer worldwide. Aberrant signaling of the FGF19-FGFR4 pathway leads to HCC in mice and is hypothesized to be a driver in FGF19 amplified HCC in humans. Multiple small molecule inhibitors have been pursued as targeted therapies for HCC in recent years, including several selective FGFR4 inhibitors that are currently being evaluated in clinical trials. Herein, we report a novel series of highly selective, covalent 2-amino-6,8-dimethyl-pyrido[2,3-d]pyrimidin-7(8H)-ones that potently and selectively inhibit FGFR4 signaling through covalent modification of Cys552, which was confirmed by X-ray crystallography. Correlative target occupancy and pFGFR4 inhibition were observed in vivo, as well as tumor regression in preclinical models of orthotopic and sorafenib-resistant HCC.

7.
Nat Commun ; 10(1): 96, 2019 01 09.
Article in English | MEDLINE | ID: mdl-30626880

ABSTRACT

Aberrant metabolism of cancer cells is well appreciated, but the identification of cancer subsets with specific metabolic vulnerabilities remains challenging. We conducted a chemical biology screen and identified a subset of neuroendocrine tumors displaying a striking pattern of sensitivity to inhibition of the cholesterol biosynthetic pathway enzyme squalene epoxidase (SQLE). Using a variety of orthogonal approaches, we demonstrate that sensitivity to SQLE inhibition results not from cholesterol biosynthesis pathway inhibition, but rather surprisingly from the specific and toxic accumulation of the SQLE substrate, squalene. These findings highlight SQLE as a potential therapeutic target in a subset of neuroendocrine tumors, particularly small cell lung cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Delivery Systems , Drug Screening Assays, Antitumor , Squalene Monooxygenase/antagonists & inhibitors , Squalene Monooxygenase/metabolism , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cholesterol/biosynthesis , Gene Deletion , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans
8.
Cell Chem Biol ; 25(6): 705-717.e11, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29628435

ABSTRACT

Activating KRAS mutations are major oncogenic drivers in multiple tumor types. Synthetic lethal screens have previously been used to identify targets critical for the survival of KRAS mutant cells, but their application to drug discovery has proven challenging, possibly due in part to a failure of monolayer cultures to model tumor biology. Here, we report the results of a high-throughput synthetic lethal screen for small molecules that selectively inhibit the growth of KRAS mutant cell lines in soft agar. Chemoproteomic profiling identifies the target of the most KRAS-selective chemical series as dihydroorotate dehydrogenase (DHODH). DHODH inhibition is shown to perturb multiple metabolic pathways. In vivo preclinical studies demonstrate strong antitumor activity upon DHODH inhibition in a pancreatic tumor xenograft model.


Subject(s)
Oxidoreductases Acting on CH-CH Group Donors/metabolism , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrimidines/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Dihydroorotate Dehydrogenase , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Female , Humans , Mice , Mice, SCID , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Pyrimidines/chemistry , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Tumor Cells, Cultured
9.
J Comp Neurol ; 501(1): 30-7, 2007 Mar 01.
Article in English | MEDLINE | ID: mdl-17206617

ABSTRACT

Recent studies indicate that neurotrophin 3 (NT3) may be important for the maintenance and function of the adult inner ear, but the pattern of postnatal NT3 expression in this organ has not been characterized. We used a reporter mouse in which cells expressing NT3 also express beta-galactosidase, allowing for their histochemical visualization, to determine the pattern of NT3 expression in cochlear and vestibular organs. We analyzed animals from birth (P0) to adult (P135). At P0, NT3 was strongly expressed in supporting cells and hair cells of all vestibular and cochlear sense organs, Reissner's membrane, saccular membrane, and the dark cells adjacent to canal organs. With increasing age, staining disappeared in most cell types but remained relatively high in inner hair cells (IHCs) and to a lesser extent in IHC supporting cells. In the cochlea, by P0 there is a longitudinal gradient (apex > base) that persists into adulthood. In vestibular maculae, staining gradients are: striolar > extrastriolar regions and supporting cells > hair cells. By P135, cochlear staining is restricted to IHCs and their supporting cells, with stronger expression in the apex than the base. By the same age, in the vestibular organs, NT3 expression is weak and restricted to saccular and utricular supporting cells. These results suggest that NT3 might play a long-term role in the maintenance and functioning of the adult auditory and vestibular systems and that supporting cells are the main source of this factor in the adult.


Subject(s)
Aging/metabolism , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Ear, Inner/metabolism , Neurotrophin 3/metabolism , Animals , Cochlea/metabolism , Hair Cells, Auditory, Inner/metabolism , Immunohistochemistry , Lac Operon , Mice , Mice, Transgenic , Saccule and Utricle/metabolism , Staining and Labeling , Tissue Distribution , beta-Galactosidase/metabolism
10.
PLoS One ; 12(9): e0185092, 2017.
Article in English | MEDLINE | ID: mdl-28950000

ABSTRACT

Tumor cells display fundamental changes in metabolism and nutrient uptake in order to utilize additional nutrient sources to meet their enhanced bioenergetic requirements. Glutamine (Gln) is one such nutrient that is rapidly taken up by tumor cells to fulfill this increased metabolic demand. A vital step in the catabolism of glutamine is its conversion to glutamate by the mitochondrial enzyme glutaminase (GLS). This study has identified GLS a potential therapeutic target in breast cancer, specifically in the basal subtype that exhibits a deregulated glutaminolysis pathway. Using inducible shRNA mediated gene knockdown, we discovered that loss of GLS function in triple-negative breast cancer (TNBC) cell lines with a deregulated glutaminolysis pathway led to profound tumor growth inhibition in vitro and in vivo. GLS knockdown had no effect on growth and metabolite levels in non-TNBC cell lines. We rescued the anti-tumor effect of GLS knockdown using shRNA resistant cDNAs encoding both GLS isoforms and by addition of an α-ketoglutarate (αKG) analog thus confirming the critical role of GLS in TNBC. Pharmacological inhibition of GLS with the small molecule inhibitor CB-839 reduced cell growth and led to a decrease in mammalian target of rapamycin (mTOR) activity and an increase in the stress response pathway driven by activating transcription factor 4 (ATF4). Finally, we found that GLS inhibition synergizes with mTOR inhibition, which introduces the possibility of a novel therapeutic strategy for TNBC. Our study revealed that GLS is essential for the survival of TNBC with a deregulated glutaminolysis pathway. The synergistic activity of GLS and mTOR inhibitors in TNBC cell lines suggests therapeutic potential of this combination for the treatment of vulnerable subpopulations of TNBC.


Subject(s)
Glutaminase/metabolism , Glutamine/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , Triple Negative Breast Neoplasms/enzymology , Cell Line, Tumor , Female , Humans , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
11.
Cell Rep ; 15(3): 574-587, 2016 Apr 19.
Article in English | MEDLINE | ID: mdl-27068473

ABSTRACT

Homozygous deletions of p16/CDKN2A are prevalent in cancer, and these mutations commonly involve co-deletion of adjacent genes, including methylthioadenosine phosphorylase (MTAP). Here, we used shRNA screening and identified the metabolic enzyme, methionine adenosyltransferase II alpha (MAT2A), and the arginine methyltransferase, PRMT5, as vulnerable enzymes in cells with MTAP deletion. Metabolomic and biochemical studies revealed a mechanistic basis for this synthetic lethality. The MTAP substrate methylthioadenosine (MTA) accumulates upon MTAP loss. Biochemical profiling of a methyltransferase enzyme panel revealed that MTA is a potent and selective inhibitor of PRMT5. MTAP-deleted cells have reduced PRMT5 methylation activity and increased sensitivity to PRMT5 depletion. MAT2A produces the PRMT5 substrate S-adenosylmethionine (SAM), and MAT2A depletion reduces growth and PRMT5 methylation activity selectively in MTAP-deleted cells. Furthermore, this vulnerability extends to PRMT5 co-complex proteins such as RIOK1. Thus, the unique biochemical features of PRMT5 create an axis of targets vulnerable in CDKN2A/MTAP-deleted cancers.


Subject(s)
Adenosine/analogs & derivatives , Antigens, Neoplasm/metabolism , Gene Deletion , Methionine Adenosyltransferase/metabolism , Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Purine-Nucleoside Phosphorylase/metabolism , Signal Transduction , Thionucleosides/metabolism , Adenosine/metabolism , Genomics , HCT116 Cells , Humans , Multiprotein Complexes/metabolism , Neoplasms/metabolism , Purine-Nucleoside Phosphorylase/deficiency , RNA, Small Interfering/metabolism
12.
Cell Rep ; 17(3): 876-890, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27732861

ABSTRACT

Although aberrant metabolism in tumors has been well described, the identification of cancer subsets with particular metabolic vulnerabilities has remained challenging. Here, we conducted an siRNA screen focusing on enzymes involved in the tricarboxylic acid (TCA) cycle and uncovered a striking range of cancer cell dependencies on OGDH, the E1 subunit of the alpha-ketoglutarate dehydrogenase complex. Using an integrative metabolomics approach, we identified differential aspartate utilization, via the malate-aspartate shuttle, as a predictor of whether OGDH is required for proliferation in 3D culture assays and for the growth of xenograft tumors. These findings highlight an anaplerotic role of aspartate and, more broadly, suggest that differential nutrient utilization patterns can identify subsets of cancers with distinct metabolic dependencies for potential pharmacological intervention.


Subject(s)
Aspartic Acid/metabolism , Ketoglutarate Dehydrogenase Complex/metabolism , Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Respiration/drug effects , Citric Acid Cycle/drug effects , Enzyme Inhibitors/pharmacology , Gene Knockdown Techniques , Humans , RNA, Small Interfering/metabolism
13.
Cancer Res ; 73(19): 6024-35, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23928993

ABSTRACT

HER2/HER3 dimerization resulting from overexpression of HER2 or neuregulin (NRG1) in cancer leads to HER3-mediated oncogenic activation of phosphoinositide 3-kinase (PI3K) signaling. Although ligand-blocking HER3 antibodies inhibit NRG1-driven tumor growth, they are ineffective against HER2-driven tumor growth because HER2 activates HER3 in a ligand-independent manner. In this study, we describe a novel HER3 monoclonal antibody (LJM716) that can neutralize multiple modes of HER3 activation, making it a superior candidate for clinical translation as a therapeutic candidate. LJM716 was a potent inhibitor of HER3/AKT phosphorylation and proliferation in HER2-amplified and NRG1-expressing cancer cells, and it displayed single-agent efficacy in tumor xenograft models. Combining LJM716 with agents that target HER2 or EGFR produced synergistic antitumor activity in vitro and in vivo. In particular, combining LJM716 with trastuzumab produced a more potent inhibition of signaling and cell proliferation than trastuzumab/pertuzumab combinations with similar activity in vivo. To elucidate its mechanism of action, we solved the structure of LJM716 bound to HER3, finding that LJM716 bound to an epitope, within domains 2 and 4, that traps HER3 in an inactive conformation. Taken together, our findings establish that LJM716 possesses a novel mechanism of action that, in combination with HER2- or EGFR-targeted agents, may leverage their clinical efficacy in ErbB-driven cancers.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/pathology , Neuregulin-1/metabolism , Protein Conformation/drug effects , Receptor, ErbB-3/antagonists & inhibitors , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Female , Humans , Immunoblotting , Immunoprecipitation , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Phosphorylation/drug effects , Protein Multimerization/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/immunology , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/immunology , Receptor, ErbB-3/metabolism , Signal Transduction , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
14.
J Assoc Res Otolaryngol ; 11(1): 19-26, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19820996

ABSTRACT

Recent studies indicate that supporting cells play important roles in inner ear development, function, and regeneration after injury, but the molecular mechanisms underlying these processes remain poorly understood. Inducible cell-specific gene recombination in supporting cells could be a powerful tool to study the roles of specific molecules in these cells. Here we tested the feasibility, effectiveness, and cell specificity of inducible Cre-mediated gene recombination in the postnatal inner ear using mice that express an inducible form of Cre (CreER(T)) under the transcriptional control of the proteolipid protein (PLP) promoter. We assessed the pattern of tamoxifen-induced gene recombination in the inner ear using the ROSA26-LacZ reporter line, in which the beta-galactosidase gene is expressed only after Cre-mediated excision of a loxP-flanked stop cassette. Recombination was detected in cochlear inner phalangeal cells, supporting cells surrounding hair cells in vestibular maculae and cristae. Recombination also occurred in Schwann cells. We also found that this CreER(T) line can be used to increase and decrease the levels of expression of a trophic factor, brain-derived neurotrophic factor, specifically in supporting cells. These results show that PLP/CreER(T) mice are a powerful tool to dissect gene function in inner ear supporting cells.


Subject(s)
Ear, Inner , Gene Expression Regulation, Developmental , Neuroglia/physiology , Recombination, Genetic/physiology , Schwann Cells/physiology , Animals , Antineoplastic Agents, Hormonal , Brain-Derived Neurotrophic Factor/genetics , Ear, Inner/cytology , Ear, Inner/embryology , Ear, Inner/physiology , Genes, Reporter , Hair Cells, Vestibular/physiology , Integrases/genetics , Lac Operon , Mice , Mice, Transgenic , Myelin Proteolipid Protein/genetics , Promoter Regions, Genetic/genetics , Tamoxifen , Transgenes/genetics
15.
Nat Med ; 16(12): 1429-33, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21076395

ABSTRACT

Aberrant activation of the Hedgehog (Hh) pathway can drive tumorigenesis. To investigate the mechanism by which glioma-associated oncogene family zinc finger-1 (GLI1), a crucial effector of Hh signaling, regulates Hh pathway activation, we searched for GLI1-interacting proteins. We report that the chromatin remodeling protein SNF5 (encoded by SMARCB1, hereafter called SNF5), which is inactivated in human malignant rhabdoid tumors (MRTs), interacts with GLI1. We show that Snf5 localizes to Gli1-regulated promoters and that loss of Snf5 leads to activation of the Hh-Gli pathway. Conversely, re-expression of SNF5 in MRT cells represses GLI1. Consistent with this, we show the presence of a Hh-Gli-activated gene expression profile in primary MRTs and show that GLI1 drives the growth of SNF5-deficient MRT cells in vitro and in vivo. Therefore, our studies reveal that SNF5 is a key mediator of Hh signaling and that aberrant activation of GLI1 is a previously undescribed targetable mechanism contributing to the growth of MRT cells.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics , Rhabdoid Tumor/genetics , Signal Transduction/genetics , Transcription Factors/metabolism , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , DNA Primers/genetics , DNA-Binding Proteins/genetics , Gene Expression Profiling , Humans , Immunoblotting , In Situ Hybridization , Mass Spectrometry , Mice , Microarray Analysis , SMARCB1 Protein , Transcription Factors/genetics , Zinc Finger Protein GLI1
16.
J Neurosci Res ; 85(10): 2080-6, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17492793

ABSTRACT

Oligodendrocytes (OLs), the myelinating cells of the central nervous system, have specialized morphologies that subserve their function. Numerous qualitative studies suggest that OLs in different brain regions can differ in their morphological characteristics, including number of branches and internodes, internode length, etc. However, progress in identifying and characterizing the diverse types of OLs and their distribution in the brain has been made difficult by several technical constraints. Here we report a new strategy to analyze OL morphology with a high degree of quantitative power and throughput. We used confocal microscopy and three-dimensional cell tracing software to study OLs in the frontal cortex of mice expressing enhanced green fluorescent protein (eGFP) under the control of the proteolipid protein (Plp) gene promoter. Three-dimensional reconstructions were then used to analyze and quantify cell morphology, including total process length, total process surface area, total internode length, number of primary processes, number of branch points, and number of internodes. In addition, these reconstructions were subjected to Sholl analysis, which allows for the quantitative measure of OL arbor complexity. By using this approach, we identified and characterized a previously undescribed population of small OLs with a compact but complex morphology that includes numerous branching processes and a large number of short internodes. Our data suggest that other populations of OLs remain to be identified and characterized and that the tools we have developed could help in the process of characterizing them.


Subject(s)
Frontal Lobe/cytology , Image Processing, Computer-Assisted , Imaging, Three-Dimensional , Microscopy, Confocal , Oligodendroglia/ultrastructure , Animals , Dendrites/ultrastructure , Fluorescent Dyes , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Oligodendroglia/metabolism
17.
Proc Natl Acad Sci U S A ; 104(19): 8131-6, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17483467

ABSTRACT

Several psychiatric disorders are associated with white matter defects, suggesting that oligodendrocyte (OL) abnormalities underlie some aspects of these diseases. Neuregulin 1 (NRG1) and its receptor, erbB4, are genetically linked with susceptibility to schizophrenia and bipolar disorder. In vitro studies suggest that NRG1-erbB signaling is important for OL development. To test whether erbB signaling contributes to psychiatric disorders by regulating the structure or function of OLs, we analyzed transgenic mice in which erbB signaling is blocked in OLs in vivo. Here we show that loss of erbB signaling leads to changes in OL number and morphology, reduced myelin thickness, and slower conduction velocity in CNS axons. Furthermore, these transgenic mice have increased levels of dopamine receptors and transporters and behavioral alterations consistent with neuropsychiatric disorders. These results indicate that defects in white matter can cause alterations in dopaminergic function and behavior relevant to neuropsychiatric disorders.


Subject(s)
Dopamine/physiology , ErbB Receptors/physiology , Mental Disorders/etiology , Myelin Sheath/physiology , Oligodendroglia/physiology , Signal Transduction/physiology , Amphetamine/pharmacology , Animals , Anxiety/etiology , Cyclic Nucleotide Phosphodiesterases, Type 1 , Mice , Mice, Transgenic , Motor Activity , Nerve Tissue Proteins/physiology , Neural Conduction , Neuregulin-1 , Phosphoric Diester Hydrolases/genetics , Promoter Regions, Genetic , Receptor, ErbB-4 , Social Behavior
18.
Glia ; 54(6): 578-90, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16921523

ABSTRACT

Fibroblast growth factor 2 (FGF2) inhibits oligodendrocyte progenitor cell (OPC) differentiation during development and limits remyelination following chronic demyelination. The current study examines the mechanism underlying this effect of FGF2 expression on OPC differentiation. Retroviral lineage tracing demonstrates a direct in vivo effect of FGF receptor (FGFR) signaling on OPC differentiation. Retrovirus expressing a dominant negative FGFR construct (FGFRdn) and green fluorescent protein (GFP) was injected into the dorsal columns of postnatal day 7 (P7) mice followed by perfusion at P28. Among the GFP-labeled cells, FGFRdn retrovirus generated a higher proportion of oligodendrocytes than did control infections. This result from FGFRdn expression in OPCs was similar to the result obtained in our previous study using control retrovirus in FGF2 null mice. Further, in vitro retroviral siRNA expression distinguishes the function of specific FGFR isoforms in OPC responses to FGF2. FGF2 inhibition of OPC differentiation was effectively blocked by siRNA targeted to FGFR1, but not FGFR2 or FGFR3. We propose a model of direct FGF2 activation of FGFR1 leading to inhibition of OPC differentiation. This signaling pathway may be an important regulator of oligodendrocyte generation during myelination in development and may perturb OPC generation of remyelinating oligodendrocytes in demyelinating disease.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Fibroblast Growth Factor 2/pharmacology , Oligodendroglia/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cells, Cultured , Fibroblast Growth Factor 2/metabolism , Genetic Vectors/genetics , Green Fluorescent Proteins , Growth Inhibitors/metabolism , Growth Inhibitors/pharmacology , Male , Mice , Mice, Inbred C57BL , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/metabolism , Nerve Regeneration/physiology , Oligodendroglia/drug effects , RNA, Small Interfering/genetics , Rats , Receptor, Fibroblast Growth Factor, Type 1/drug effects , Recombinant Fusion Proteins/genetics , Retroviridae/genetics , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/drug effects , Transfection/methods
19.
Cell ; 127(1): 185-97, 2006 Oct 06.
Article in English | MEDLINE | ID: mdl-17018285

ABSTRACT

Embryonic multipotent neural precursors are exposed to extracellular signals instructing them to adopt different fates, neuronal or glial. However, the mechanisms by which precursors integrate these signals to make timely fate choices remained undefined. Here we show that direct nuclear signaling by a receptor tyrosine kinase inhibits the responses of precursors to astrocyte differentiation factors while maintaining their neurogenic potential. Upon neuregulin-induced activation and presenilin-dependent cleavage of ErbB4, the receptor's intracellular domain forms a complex with TAB2 and the corepressor N-CoR. This complex undergoes nuclear translocation and binds promoters of astrocytic genes, repressing their expression. Consistent with this observation, astrogenesis occurs precociously in ErbB4 knockout mice. Our studies define how presenilin-dependent nuclear signaling by a receptor tyrosine kinase directly regulates gene transcription and cell fate. This pathway could be of importance for neural stem cell biology and for understanding the pathogenesis of Alzheimer's disease.


Subject(s)
Astrocytes/physiology , Brain , Cell Nucleus/metabolism , ErbB Receptors/metabolism , Presenilins/metabolism , Signal Transduction/physiology , Active Transport, Cell Nucleus/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Astrocytes/cytology , Brain/cytology , Brain/embryology , Cell Differentiation/physiology , Cell Line , ErbB Receptors/genetics , Gene Expression Regulation, Developmental , Humans , Mice , Mice, Knockout , Multiprotein Complexes , Neuregulin-1/metabolism , Neurons/cytology , Neurons/physiology , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nuclear Receptor Co-Repressor 1 , Presenilins/genetics , Promoter Regions, Genetic , Protein Structure, Tertiary , Rats , Receptor, ErbB-4 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Time Factors , Two-Hybrid System Techniques
20.
Glia ; 49(4): 542-54, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15578654

ABSTRACT

Analysis of fibroblast growth factor 2 null (FGF2-/-) and wild-type (FGF2+/+) mice was used to interpret the potential in vivo role of endogenous FGF2 on oligodendrocyte lineage cell (OLC) responses during oligodendrogenesis and myelination. In wild-type mouse spinal cord, FGF2 levels increased approximately threefold between the first and second postnatal weeks, a period corresponding with the peak of oligodendrogenesis. Absence of this developmental FGF2 elevation in FGF2-/- mice eliminated the transient overproduction of oligodendrocytes that is known to occur at the peak of oligodendrogenesis in wild-type mice. Absence of FGF2 did not affect oligodendrocyte progenitor (OP) density or proliferation, based on BrdU incorporation, and also did not alter survival, based on TUNEL analysis. To examine OLC differentiation in vivo, retrovirus encoding-enhanced green fluorescent protein (GFP) was injected into the spinal cord to heritably label endogenous cycling cells in the white matter at postnatal day 7 and then identify the generated cells at postnatal day 28. Phenotypes of cells expressing GFP were identified by morphology and immunolabeling, using CC1 for oligodendrocytes and NG2 combined with platelet-derived growth factor alpha receptor for OPs. Within the population of GFP-labeled cells, the proportion of oligodendrocytes was higher in FGF2-/- mice, indicating that endogenous FGF2 inhibited OLC differentiation in wild-type mice. Furthermore, in FGF2-/- mice fewer cells appeared to be generated from an initial retrovirus-labeled cell, consistent with more frequent differentiation into post-mitotic oligodendrocytes. This in vivo analysis demonstrates that the predominant role of endogenous FGF2 on OLCs in development is inhibition of differentiation.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Oligodendroglia/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptors, Fibroblast Growth Factor/genetics , Spinal Cord/growth & development , Spinal Cord/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Antigens/metabolism , Autophagy-Related Proteins , Biomarkers , Bromodeoxyuridine , Cell Line , Cell Proliferation , Cell Survival/physiology , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mice , Mice, Knockout , Myelin Sheath/metabolism , Oligodendroglia/cytology , Proteoglycans/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 2 , Receptors, Fibroblast Growth Factor/metabolism , Retroviridae/genetics , Spinal Cord/cytology , Stem Cells/cytology , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL