Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
EBioMedicine ; 103: 105093, 2024 May.
Article in English | MEDLINE | ID: mdl-38569318

ABSTRACT

BACKGROUND: Human restricted genes contribute to human specific traits in the immune system. CHRFAM7A, a uniquely human fusion gene, is a negative regulator of the α7 nicotinic acetylcholine receptor (α7 nAChR), the highest Ca2+ conductor of the ACh receptors implicated in innate immunity. Understanding the mechanism of how CHRFAM7A affects the immune system remains unexplored. METHODS: Two model systems are used, human induced pluripotent stem cells (iPSC) and human primary monocytes, to characterize α7 nAChR function, Ca2+ dynamics and decoders to elucidate the pathway from receptor to phenotype. FINDINGS: CHRFAM7A/α7 nAChR is identified as a hypomorphic receptor with mitigated Ca2+ influx and prolonged channel closed state. This shifts the Ca2+ reservoir from the extracellular space to the endoplasmic reticulum (ER) leading to Ca2+ dynamic changes. Ca2+ decoder small GTPase Rac1 is then activated, reorganizing the actin cytoskeleton. Observed actin mediated phenotypes include cellular adhesion, motility, phagocytosis and tissue mechanosensation. INTERPRETATION: CHRFAM7A introduces an additional, human specific, layer to Ca2+ regulation leading to an innate immune gain of function. Through the actin cytoskeleton it drives adaptation to the mechanical properties of the tissue environment leading to an ability to invade previously immune restricted niches. Human genetic diversity predicts profound translational significance as its understanding builds the foundation for successful treatments for infectious diseases, sepsis, and cancer metastasis. FUNDING: This work is supported in part by the Community Foundation for Greater Buffalo (Kinga Szigeti) and in part by NIH grant R01HL163168 (Yongho Bae).


Subject(s)
Actin Cytoskeleton , Calcium Signaling , Induced Pluripotent Stem Cells , alpha7 Nicotinic Acetylcholine Receptor , Humans , Actin Cytoskeleton/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , alpha7 Nicotinic Acetylcholine Receptor/genetics , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Calcium/metabolism , Monocytes/metabolism , Immunity, Innate , rac1 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/genetics , Phagocytosis
2.
Methods Mol Biol ; 2742: 105-122, 2024.
Article in English | MEDLINE | ID: mdl-38165619

ABSTRACT

The combination of advanced mass spectrometry and enrichment-based sample preparation methods has enhanced analytical capabilities in clinical proteomics. In this chapter, we describe a method of proteome analysis to identify Borrelia-derived peptides in urine that includes a sample affinity enrichment method coupled with liquid chromatography tandem mass spectrometry analysis and a bioinformatic peptide authentication algorithm.


Subject(s)
Borrelia , Humans , Tandem Mass Spectrometry/methods , Peptides/chemistry , Chromatography, Liquid/methods , Proteome/analysis
3.
Int J Mol Sci ; 21(24)2020 Dec 18.
Article in English | MEDLINE | ID: mdl-33352944

ABSTRACT

Neuroinflammation in Alzheimer's disease (AD) has been the focus for identifying targetable pathways for drug development. The role of amyloid beta (Aß), a prototype of damage-associated molecular patterns (DAMPs), has been implicated in triggering an inflammatory response. As alpha7 nicotinic acetylcholine receptor (α7 nAChR) binds Aß with high affinity, α7 nAChR may play a role in Aß-induced neuroinflammation. The conundrum of how α7 nAChR as the mediator of the cholinergic anti-inflammatory response may trigger an inflammatory response has not been resolved. CHRFAM7A, the uniquely human fusion gene between ULK4 and CHRNA7, is a negative regulator of α7 nAChR ionotropic function. To provide the human context, isogenic induced pluripotent stem cell (iPSC) lines were developed from CHRFAM7A null and carrier individuals by genome-editing the null line using TALENs to knock-in CHRFAM7A. In iPSC-derived microglia-like cells, CHRFAM7A mitigated Aß uptake through the α7 nAChR. Despite the lower Aß uptake, the presence of CHRFAM7A was associated with an innate immune response that was characterized by NF-κB activation and NF-κB target transcription (TNFA, IL6, and IL1B). LPS, a prototype PAMP, induced a heightened immune response in CHRFAM7A carriers. CHRFAM7A modified the dynamics of NF-κB translocation by prolonging its nuclear presence. CHRFAM7A modified the α7 nAChR metabotropic function, resulting in a human-specific innate immune response. This iPSC model provided an opportunity to elucidate the mechanism and establish high throughput screens.


Subject(s)
Alarmins/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/metabolism , Induced Pluripotent Stem Cells/cytology , Microglia/immunology , Microglia/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Alzheimer Disease/pathology , Cell Movement , Disease Susceptibility , Gene Expression , Humans , Immunity, Innate , Immunologic Surveillance , Microglia/cytology , NF-kappa B/metabolism , Signal Transduction , alpha7 Nicotinic Acetylcholine Receptor/genetics , alpha7 Nicotinic Acetylcholine Receptor/metabolism
4.
EBioMedicine ; 59: 102892, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32818803

ABSTRACT

BACKGROUND: Cholinergic neuronal loss is one of the hallmarks of AD related neurodegeneration; however, preclinical promise of α7 nAChR drugs failed to translate into humans. CHRFAM7A, a uniquely human fusion gene, is a negative regulator of α7 nAChR and was unaccounted for in preclinical models. METHODS: Molecular methods: Function of CHRFAM7A alleles was studied in vitro in two disease relevant phenotypic readouts: electrophysiology and Aß uptake. Genome edited human induced pluripotent stem cells (iPSC) were used as a model system with the human context. Double blind pharmacogenetic study: We performed double-blind pharmacogenetic analysis on the effect of AChEI therapy based on CHRFAM7A carrier status in two paradigms: response to drug initiation and DMT effect. Mini Mental Status Examination (MMSE) was used as outcome measure. Change in MMSE score from baseline was compared by 2-tailed T-test. Longitudinal analysis of clinical outcome (MMSE) was performed using a fitted general linear model, based on an assumed autoregressive covariance structure. Model independent variables included age, sex, and medication regimen at the time of the first utilized outcome measure (AChEI alone or AChEI plus memantine), APOE4 carrier status (0, 1 or 2 alleles as categorical variables) and CHRFAM7A genotype. FINDINGS: The direct and inverted alleles have distinct phenotypes. Functional CHRFAM7A allele classifies the population as 25% non-carriers and 75% carriers. Induced pluripotent stem cell (iPSC) models α7 nAChR mediated Aß neurotoxicity. Pharmacological readout translates into both first exposure (p = 0.037) and disease modifying effect (p = 0.0048) in two double blind pharmacogenetic studies. INTERPRETATION: CHRFAM7A accounts for the translational gap in cholinergic strategies in AD. Clinical trials not accounting for this uniquely human genetic factor may have rejected drug candidates that would benefit 25% of AD. Reanalyses of the completed trials using this pharmacogenetic paradigm may identify effective therapy.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/therapy , Cholinergic Neurons/metabolism , Recombinant Fusion Proteins , alpha7 Nicotinic Acetylcholine Receptor/genetics , Alleles , Alzheimer Disease/diagnosis , Alzheimer Disease/etiology , Amyloid beta-Peptides/metabolism , Biomarkers , Cell Line , Cholinergic Antagonists/pharmacology , Cholinergic Antagonists/therapeutic use , Drug Evaluation, Preclinical , Fluorescent Antibody Technique , Gene Dosage , Gene Frequency , Genotype , Humans , Induced Pluripotent Stem Cells/metabolism , Phenotype , Synaptic Transmission , Translational Research, Biomedical , Treatment Outcome , alpha7 Nicotinic Acetylcholine Receptor/metabolism
5.
Transl Psychiatry ; 9(1): 59, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30710073

ABSTRACT

The α7 nicotinic acetylcholine receptor (α7nAChR) has been a promising target for diseases affecting cognition and higher cortical functions; however, the effect observed in animal models failed to translate into human clinical trials identifying a translational gap. CHRFAM7A is a human-specific fusion gene with properties that enable incorporation into the α7nAChR and, being human specific, CHRFAM7A effect was not accounted for in preclinical studies. We hypothesized that CHRFAM7A may account for this translational gap and understanding its function may offer novel insights when exploring α7nAChR as a drug target. CHRFAM7A is present in different copy number variations (CNV) in the human genome with high frequency. To study the functional consequences of the presence of the CHRFAM7A, two induced pluripotent stem cell (iPSC) lines (0 copy and 1 copy direct) were developed. The 0 copy line was rescued with CHRFAM7A transfection to control for genetic heterogeneity. As readouts for genotype-phenotype correlation, α7nAChR synaptic transmission and amyloid beta 1-42 (Aß1-42) uptake were tested. Synaptic transmission in the presence of CHRFAM7A demonstrated that PNU-modulated desensitization of α7nAChR currents increased as a function of CHRFAM7A dosage. CHRFAM7A mitigated the dose response of Aß1-42 uptake suggesting a protective effect beyond physiological concentrations. Furthermore, in the presence of CHRFAM7A Aß1-42 uptake activated neuronal interleukin 1ß (IL-1ß) and tumor necrosis factor α (TNF-α) without activating the canonical inflammasome pathway. Lead optimization may identify more potent molecules when the screen has a model harboring CHRFAM7A. Incorporating pharmacogenetics into clinical trials may enhance signals in efficacy measures.


Subject(s)
Induced Pluripotent Stem Cells/metabolism , Neurons/metabolism , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Amyloid beta-Peptides/administration & dosage , Amyloid beta-Peptides/metabolism , Cell Differentiation , Cells, Cultured , Gene Expression , HEK293 Cells , Humans , Inflammation/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/metabolism , Synaptic Transmission
6.
Sci Rep ; 7: 42760, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28230160

ABSTRACT

The human pyruvate dehydrogenase complex (PDC) comprises four multidomain components, E1, E3, E2 and an E3-binding protein (E3BP), the latter two forming the core as E2·E3BP sub-complex. Pyruvate flux through PDC is regulated via phosphorylation (inactivation) at E1 by four PDC kinases (PDKs), and reactivation by two PDC phosphatases. Up-regulation of PDK isoform gene expression is reported in several forms of cancer, while PDKs may be further activated by PDC by binding to the E2·E3BP core. Hence, the PDK: E2·E3BP interaction provides new therapeutic targets. We carried out both functional kinetic and thermodynamic studies to demonstrate significant differences in the activation of PDK isoforms by binding to the E2·E3BP core: (i) PDK2 needs no activation by E2·E3BP for efficient functioning, while PDK4 was the least effective of the four isoforms, and could not be activated by E2·E3BP. Hence, development of inhibitors to the interaction of PDK2 and PDK4 with E2·E3BP is not promising; (ii) Design of inhibitors to interfere with interaction of E2·E3BP with PDK1 and PDK3 is promising. PDK3 needs E2·E3BP core for activation, an activation best achieved by synergistic combination of E2-derived catalytic domain and tridomain.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Complex/metabolism , Catalytic Domain , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Thermodynamics
7.
J Enzyme Inhib Med Chem ; 31(sup4): 170-175, 2016.
Article in English | MEDLINE | ID: mdl-27435185

ABSTRACT

The last decade has witnessed the reawakening of cancer metabolism as a therapeutic target. In particular, inhibition of pyruvate dehydrogenase kinase (PDK) holds remarkable promise. Dichloroacetic acid (DCA), currently undergoing clinical trials, is a unique PDK inhibitor in which it binds to the allosteric pyruvate site of the enzyme. However, the safety of DCA as a drug is compromised by its neurotoxicity, whereas its usefulness as an investigative tool is limited by the high concentrations required to exert observable effects in cell culture. Herein, we report the identification - by making use of saturation-transfer difference NMR spectroscopy, enzymatic assays and computational methods - of furoate and thenoate derivatives as allosteric pyruvate-site-binding PDK2 inhibitors. This work substantiates the pyruvate regulatory pocket as a druggable target.


Subject(s)
Furans/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyruvic Acid/metabolism , Thiophenes/pharmacology , Allosteric Regulation/drug effects , Allosteric Site/drug effects , Dose-Response Relationship, Drug , Furans/chemical synthesis , Furans/chemistry , Humans , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/chemistry , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry
8.
Am J Physiol Endocrinol Metab ; 311(1): E117-27, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27166281

ABSTRACT

During the absorptive state, the liver stores excess glucose as glycogen and synthesizes fatty acids for triglyceride synthesis for export as very low density lipoproteins. For de novo synthesis of fatty acids from glucose, the mitochondrial pyruvate dehydrogenase complex (PDC) is the gatekeeper for the generation of acetyl-CoA from glucose-derived pyruvate. Here, we tested the hypothesis that limiting the supply of PDC-generated acetyl-CoA from glucose would have an impact on expression of key genes in the lipogenic pathway. In the present study, although the postnatal growth of liver-specific PDC-deficient (L-PDCKO) male mice was largely unaltered, the mice developed hyperinsulinemia with lower blood glucose levels in the fed state. Serum and liver lipid triglyceride and cholesterol levels remained unaltered in L-PDCKO mice. Expression of several key genes (ACL, ACC1) in the lipogenic pathway and their upstream regulators (LXR, SREBP1, ChREBP) as well as several genes in glucose metabolism (Pklr, G6pd2, Pck1) and fatty acid oxidation (FAT, Cpt1a) was downregulated in livers from L-PDCKO mice. Interestingly, there was concomitant upregulation of lipogenic genes in adipose tissue from L-PDCKO mice. Although, the total hepatic acetyl-CoA content remained unaltered in L-PDCKO mice, modified acetylation profiles of proteins in the nuclear compartment suggested an important role for PDC-generated acetyl-CoA in gene expression in de novo fatty acid synthesis in the liver. This finding has important implications for the regulation of hepatic lipid synthesis in pathological states.


Subject(s)
Acetyl Coenzyme A/metabolism , Adipose Tissue/metabolism , Gene Expression Regulation , Lipogenesis/genetics , Liver/metabolism , Mitochondria/metabolism , Pyruvate Dehydrogenase Complex/genetics , RNA, Messenger/metabolism , Animals , Blotting, Western , Cholesterol/metabolism , Fatty Acids/metabolism , Glucose/metabolism , Male , Mice , Mice, Knockout , Oxidation-Reduction , Real-Time Polymerase Chain Reaction , Subcellular Fractions , Triglycerides/metabolism
9.
Sci Signal ; 8(402): ra115, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26554819

ABSTRACT

Protein arginine methylation regulates diverse functions of eukaryotic cells, including gene expression, the DNA damage response, and circadian rhythms. We showed that arginine residues within the third intracellular loop of the human D2 dopamine receptor, which are conserved in the DOP-3 receptor in the nematode Caenorhabditis elegans, were methylated by protein arginine methyltransferase 5 (PRMT5). By mutating these arginine residues, we further showed that their methylation enhanced the D2 receptor-mediated inhibition of cyclic adenosine monophosphate (cAMP) signaling in cultured human embryonic kidney (HEK) 293T cells. Analysis of prmt-5-deficient worms indicated that methylation promoted the dopamine-mediated modulation of chemosensory and locomotory behaviors in C. elegans through the DOP-3 receptor. In addition to delineating a previously uncharacterized means of regulating GPCR (heterotrimeric guanine nucleotide-binding protein-coupled receptor) signaling, these findings may lead to the development of a new class of pharmacological therapies that modulate GPCR signaling by changing the methylation status of these key proteins.


Subject(s)
Protein-Arginine N-Methyltransferases/metabolism , Receptors, Dopamine D2/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Animals, Genetically Modified , Arginine/chemistry , Caenorhabditis elegans/drug effects , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Computational Biology , Conserved Sequence , Dopamine/metabolism , Dopamine/pharmacology , HEK293 Cells , Humans , Locomotion/drug effects , Locomotion/genetics , Locomotion/physiology , Methylation , Molecular Sequence Data , Octanols/pharmacology , Odorants , Protein-Arginine N-Methyltransferases/deficiency , Protein-Arginine N-Methyltransferases/genetics , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/genetics , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sequence Homology, Amino Acid , Signal Transduction
10.
J Cell Physiol ; 230(12): 2875-80, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25966815

ABSTRACT

A universal signaling module has been described which utilizes the nuclear form of Fibroblast growth Factor Receptor 1 (FGFR1) in a central role directing the post-mitotic development of neural cells through coordinated gene expression. In this review, we discuss in detail the current knowledge of FGFR1 nuclear interaction partners in three scenarios: (i) Engagement of FGFR1 in neuronal stem cells and regulation of neuronal differentiation; (ii) interaction with the orphan receptor Nurr1 in development of mesencephalic dopaminergic neurons; (iii) modulation of nuclear FGFR1 interactions downstream of nerve growth factor (NGF) signaling. These coalitions demonstrate the versatility of non-canonical, nuclear tyrosine kinase signaling in diverse cellular differentiation programs of neurons.


Subject(s)
Nervous System/metabolism , Neural Stem Cells/metabolism , Receptor Cross-Talk , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Dopaminergic Neurons/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Humans , Nerve Growth Factor/metabolism , Nervous System/cytology , Neurogenesis , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Signal Transduction
11.
PLoS One ; 10(4): e0123380, 2015.
Article in English | MEDLINE | ID: mdl-25923916

ABSTRACT

Genetic studies have placed the Fgfr1 gene at the top of major ontogenic pathways that enable gastrulation, tissue development and organogenesis. Using genome-wide sequencing and loss and gain of function experiments the present investigation reveals a mechanism that underlies global and direct gene regulation by the nuclear form of FGFR1, ensuring that pluripotent Embryonic Stem Cells differentiate into Neuronal Cells in response to Retinoic Acid. Nuclear FGFR1, both alone and with its partner nuclear receptors RXR and Nur77, targets thousands of active genes and controls the expression of pluripotency, homeobox, neuronal and mesodermal genes. Nuclear FGFR1 targets genes in developmental pathways represented by Wnt/ß-catenin, CREB, BMP, the cell cycle and cancer-related TP53 pathway, neuroectodermal and mesodermal programing networks, axonal growth and synaptic plasticity pathways. Nuclear FGFR1 targets the consensus sequences of transcription factors known to engage CREB-binding protein, a common coregulator of transcription and established binding partner of nuclear FGFR1. This investigation reveals the role of nuclear FGFR1 as a global genomic programmer of cell, neural and muscle development.


Subject(s)
Cell Nucleus/metabolism , Genome , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Base Sequence , Binding Sites , Cell Differentiation , Cell Line , Chromatin/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Mice , Multigene Family , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Sequence Analysis, RNA , Transcription Factors/genetics , Transcription Factors/metabolism , Tretinoin/pharmacology
12.
Biochemistry ; 54(1): 69-82, 2015 Jan 13.
Article in English | MEDLINE | ID: mdl-25436986

ABSTRACT

The human pyruvate dehydrogenase complex (PDC) comprises three principal catalytic components for its mission: E1, E2, and E3. The core of the complex is a strong subcomplex between E2 and an E3-binding protein (E3BP). The PDC is subject to regulation at E1 by serine phosphorylation by four kinases (PDK1-4), an inactivation reversed by the action of two phosphatases (PDP1 and -2). We report H/D exchange mass spectrometric (HDX-MS) and nuclear magnetic resonance (NMR) studies in the first attempt to define the interaction loci between PDK1 and PDK2 with the intact E2·E3BP core and their C-terminally truncated proteins. While the three lipoyl domains (L1 and L2 on E2 and L3 on E3BP) lend themselves to NMR studies and determination of interaction maps with PDK1 and PDK2 at the individual residue level, HDX-MS allowed studies of interaction loci on both partners in the complexes, PDKs, and other regions of the E2·E3BP core, as well, at the peptide level. HDX-MS suggested that the intact E2·E3BP core enhances the binding specificity of L2 for PDK2 over PDK1, while NMR studies detected lipoyl domain residues unique to interaction with PDK1 and PDK2. The E2·E3BP core induced more changes on PDKs than any C-terminally truncated protein, with clear evidence of greater plasticity of PDK1 than of PDK2. The effect of L1L2S paralleled HDX-MS results obtained with the intact E2·E3BP core; hence, L1L2S is an excellent candidate with which to define interaction loci with these two PDKs. Surprisingly, L3S' induced moderate interaction with both PDKs according to both methods.


Subject(s)
Deuterium Exchange Measurement/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Humans , Mass Spectrometry/methods , Molecular Sequence Data , Protein Binding/physiology , Protein Serine-Threonine Kinases/genetics , Protein Structure, Secondary , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Rats
13.
Stem Cells Transl Med ; 2(10): 776-88, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24014683

ABSTRACT

Reactivation of endogenous neurogenesis in the adult brain or spinal cord holds the key for treatment of central nervous system injuries and neurodegenerative disorders, which are major health care issues for the world's aging population. We have previously shown that activation of developmental integrative nuclear fibroblast growth factor receptor 1 (FGFR1) signaling (INFS), via gene transfection, reactivates neurogenesis in the adult brain by promoting neuronal differentiation of brain neural stem/progenitor cells (NS/PCs). In the present study, we report that targeting the α7 nicotinic acetylcholine receptors (α7nAChRs) with a specific TC-7020 agonist led to a robust accumulation of endogenous FGFR1 in the cell nucleus. Nuclear FGFR1 accumulation was accompanied by an inhibition of proliferation of NS/PCs in the subventricular zone (SVZ) and by the generation of new neurons. Neuronal differentiation was observed in different regions of the adult mouse brain, including (a) ßIII-Tubulin-expressing cortical neurons, (b) calretinin-expressing hippocampal neurons, and (c) cells in substantia nigra expressing the predopaminergic Nurr1+ phenotype. Furthermore, we showed that in vitro stimulation of neural stem/progenitor cells with α7nAChR agonist directly activated INFS and neuronal-like differentiation. TC-7020 stimulation of the ßIII-Tubulin gene was accompanied by increased binding of FGFR1, CREB binding protein, and RNA polymerase II to a Nur77 targeted promoter region. TC-7020 augmented Nur77-dependent activation of nerve growth factor inducible-B protein responsive element, indicating that α7nAChR upregulation of ßIII-Tubulin involves neurogenic FGFR1-Nur signaling. The reactivation of INFS and neurogenesis in adult brain by the α7nAChR agonist may offer a new strategy to treat brain injuries, neurodegenerative diseases, and neurodevelopmental diseases.


Subject(s)
Brain/physiology , Neurogenesis/physiology , Nicotinic Agonists/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Brain/drug effects , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Nicotinic Agonists/metabolism , Quinuclidines/pharmacology , Real-Time Polymerase Chain Reaction , Receptors, Nicotinic/metabolism , Signal Transduction/drug effects , Thiophenes/pharmacology , alpha7 Nicotinic Acetylcholine Receptor
14.
Biochem Pharmacol ; 86(8): 1099-104, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23933384

ABSTRACT

Reactivation of neurogenesis by endogenous Neural Stem/Progenitor Cells (NS/PC) in the adult brain or spinal cord holds the key for treatment of CNS injuries as well as neurodegenerative disorders, which are major healthcare issues for the world's aging population. Recent studies show that targeting the α7 nicotinic acetylcholine receptors (α7nAChR) with a specific TC-7020 agonist inhibits proliferation and stimulates neuronal differentiation of NS/PC in subventricular zone (SVZ) in the adult mouse brain. TC-7020-induced neuronogenesis is observed in different brain regions, including: (1) ßIII Tubulin-expressing cortical neurons, (2) calretinin expressing hippocampal neurons and (3) cells in substantia nigra (SN) expressing predopaminergic Nurr1+phenotype. Reactivation of developmental integrative nuclear FGFR1 signaling (INFS), via gene transfection reinstates neurogenesis in the adult brain by promoting neuronal differentiation of brain NS/PC. TC-7020 neuronogenic effect is associated with a robust accumulation of endogenous FGFR1 in the nuclei of differentiating cells. Furthermore, direct in vitro stimulation of neural stem/progenitor cells with α7nAChR agonist activates INFS and neuronal-like differentiation and activation of neuronal genes. The α7nAChR upregulation of early neuronal ßIII-Tubulin gene involves neurogenic FGFR1-Nur signaling and direct FGFR1 interaction with the gene promoter. The reactivation of developmental INFS and neurogenesis in adult brain by the α7nAChR agonist may offer new strategy to treat brain injuries, neurodegenerative and neurodevelopmental diseases.


Subject(s)
Neurogenesis/drug effects , Nicotinic Agonists/pharmacology , Quinuclidines/pharmacology , Thiophenes/pharmacology , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Animals , Neurogenesis/physiology , Neurons/cytology , Neurons/physiology
15.
PLoS One ; 8(7): e68931, 2013.
Article in English | MEDLINE | ID: mdl-23874817

ABSTRACT

Nerve growth factor (NGF) is the founding member of the polypeptide neurotrophin family responsible for neuronal differentiation. To determine whether the effects of NGF rely upon novel Integrative Nuclear FGF Receptor-1 (FGFR1) Signaling (INFS) we utilized the PC12 clonal cell line, a long-standing benchmark model of sympathetic neuronal differentiation. We demonstrate that NGF increases expression of the fgfr1 gene and promotes trafficking of FGFR1 protein from cytoplasm to nucleus by inhibiting FGFR1 nuclear export. Nuclear-targeted dominant negative FGFR1 antagonizes NGF-induced neurite outgrowth, doublecortin (dcx) expression and activation of the tyrosine hydroxylase (th) gene promoter, while active constitutive nuclear FGFR1 mimics the effects of NGF. NGF increases the expression of dcx, th, ßIII tubulin, nurr1 and nur77, fgfr1and fibroblast growth factor-2 (fgf-2) genes, while enhancing binding of FGFR1and Nur77/Nurr1 to those genes. NGF activates transcription from isolated NurRE and NBRE motifs. Nuclear FGFR1 transduces NGF activation of the Nur dimer and raises basal activity of the Nur monomer. Cooperation of nuclear FGFR1 with Nur77/Nurr1 in NGF signaling expands the integrative functions of INFS to include NGF, the first discovered pluripotent neurotrophic factor.


Subject(s)
Cell Differentiation/drug effects , Cell Nucleus/metabolism , Nerve Growth Factor/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Transcriptional Activation/drug effects , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Animals , Cell Nucleus/drug effects , Cells, Cultured , Doublecortin Protein , Humans , Nerve Growth Factor/physiology , Neurites/drug effects , Neurites/physiology , PC12 Cells , Protein Transport , Rats , Signal Transduction/drug effects , Signal Transduction/physiology
16.
J Cell Biochem ; 113(9): 2920-36, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22539306

ABSTRACT

FGF Receptor-1 (FGFR1), a membrane-targeted protein, is also involved in independent direct nuclear signaling. We show that nuclear accumulation of FGFR1 is a common response to retinoic acid (RA) in pluripotent embryonic stem cells (ESC) and neural progenitors and is both necessary and sufficient for neuronal-like differentiation and accompanying neuritic outgrowth. Dominant negative nuclear FGFR1, which lacks the tyrosine kinase domain, prevents RA-induced differentiation while full-length nuclear FGFR1 elicits differentiation in the absence of RA. Immunoprecipitation and GST assays demonstrate that FGFR1 interacts with RXR, RAR and their Nur77 and Nurr1 partners. Conditions that promote these interactions decrease the mobility of nuclear FGFR1 and RXR in live cells. RXR and FGFR1 co-associate with 5'-Fluorouridine-labeled transcription sites and with RA Responsive Elements (RARE). RA activation of neuronal (tyrosine hydroxylase) and neurogenic (fgf-2 and fgfr1) genes is accompanied by increased FGFR1, Nur, and histone H3.3 binding to their regulatory sequences. Reporter-gene assays show synergistic activations of RARE, NBRE, and NurRE by FGFR1, RAR/RXR, and Nurs. As shown for mESC differentiation, FGFR1 mediates gene activation by RA and augments transcription in the absence of RA. Cooperation of FGFR1 with RXR/RAR and Nurs at targeted genomic sequences offers a new mechanism in developmental gene regulation.


Subject(s)
Embryonic Stem Cells/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, Retinoic Acid/metabolism , Blotting, Western , Cells, Cultured , Chromatin Immunoprecipitation , Embryonic Stem Cells/cytology , Fluorescence Recovery After Photobleaching , Humans , Immunohistochemistry , Immunoprecipitation , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptors, Retinoic Acid/genetics
17.
PLoS One ; 7(4): e32294, 2012.
Article in English | MEDLINE | ID: mdl-22496730

ABSTRACT

Fungal species exhibit diverse behaviors when presented with extracellular challenges. Pathogenic fungi can undergo cell differentiation and biofilm formation in response to fluctuating nutrient levels, and these responses are required for virulence. In the model fungal eukaryote Saccharomyces cerevisiae, nutrient limitation induces filamentous growth and biofilm/mat formation. Both responses require the same signal transduction (MAPK) pathway and the same cell adhesion molecule (Flo11) but have been studied under different conditions. We found that filamentous growth and mat formation are aspects of a related response that is regulated by the MAPK pathway. Cells in yeast-form mats differentiated into pseudohyphae in response to nutrient limitation. The MAPK pathway regulated mat expansion (in the plane of the XY-axis) and substrate invasion (downward in the plane of the Z-axis), which optimized the mat's response to extracellular nutrient levels. The MAPK pathway also regulated an upward growth pattern (in the plane of the Z-axis) in response to nutrient limitation and changes in surface rigidity. Upward growth allowed for another level of mat responsiveness and resembled a type of colonial chemorepulsion. Together our results show that signaling pathways play critical roles in regulating social behaviors in which fungal cells participate. Signaling pathways may regulate similar processes in pathogens, whose highly nuanced responses are required for virulence.


Subject(s)
Biofilms , MAP Kinase Signaling System , Quorum Sensing/physiology , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae/metabolism , Adaptation, Biological , Cell Adhesion , Gene Expression Regulation, Fungal , Saccharomyces cerevisiae/genetics , Signal Transduction
18.
Curr Biol ; 20(15): 1389-95, 2010 Aug 10.
Article in English | MEDLINE | ID: mdl-20619652

ABSTRACT

Cell adhesion is a key feature in the regulation of many biological processes. In the budding yeast Saccharomyces cerevisiae, Flo11p is the major adhesion molecule that controls filamentous growth [1-3] and the expansion of interconnected cells in mats or biofilms [4]. We show here that Flo11p is shed from cells. Flo11p shedding attenuated adherence and contributed to the overall balance in adherence properties that was optimal for filamentous growth and mat formation. Shed Flo11p comprised an essential component of a fluid layer surrounding yeast mats that may be functionally analogous to the mucus secretions of higher eukaryotes. Genome-wide secretion profiling of Flo11p identified new regulatory proteins, including the furin protease Kex2p, which was required for cleavage and maturation of the Flo11p protein. Secreted mucin-like proteins may play unexpected roles in the adherence properties and virulence of microbial pathogens.


Subject(s)
Biofilms/growth & development , Membrane Glycoproteins/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/physiology , Cell Adhesion , Protein Processing, Post-Translational
19.
PLoS Genet ; 6(3): e1000883, 2010 Mar 19.
Article in English | MEDLINE | ID: mdl-20333241

ABSTRACT

An important emerging question in the area of signal transduction is how information from different pathways becomes integrated into a highly coordinated response. In budding yeast, multiple pathways regulate filamentous growth, a complex differentiation response that occurs under specific environmental conditions. To identify new aspects of filamentous growth regulation, we used a novel screening approach (called secretion profiling) that measures release of the extracellular domain of Msb2p, the signaling mucin which functions at the head of the filamentous growth (FG) MAPK pathway. Secretion profiling of complementary genomic collections showed that many of the pathways that regulate filamentous growth (RAS, RIM101, OPI1, and RTG) were also required for FG pathway activation. This regulation sensitized the FG pathway to multiple stimuli and synchronized it to the global signaling network. Several of the regulators were required for MSB2 expression, which identifies the MSB2 promoter as a target "hub" where multiple signals converge. Accessibility to the MSB2 promoter was further regulated by the histone deacetylase (HDAC) Rpd3p(L), which positively regulated FG pathway activity and filamentous growth. Our findings provide the first glimpse of a global regulatory hierarchy among the pathways that control filamentous growth. Systems-level integration of signaling circuitry is likely to coordinate other regulatory networks that control complex behaviors.


Subject(s)
MAP Kinase Signaling System , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/enzymology , Cyclic AMP/metabolism , Gene Expression Regulation, Fungal , MAP Kinase Signaling System/genetics , Promoter Regions, Genetic/genetics , Protein Binding , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
20.
Eukaryot Cell ; 8(8): 1118-33, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19502582

ABSTRACT

Many fungal species including pathogens exhibit filamentous growth (FG) as a means of foraging for nutrients. Genetic screens were performed to identify genes required for FG in the budding yeast Saccharomyces cerevisiae. Genes encoding proteins with established functions in transcriptional activation (MCM1, MATalpha2, PHD1, MSN2, SIR4, and HMS2), cell wall integrity (MPT5, WSC2, and MID2), and cell polarity (BUD5) were identified as potential regulators of FG. The transcription factors MCM1 and MATalpha2 induced invasive growth by promoting diploid-specific bipolar budding in haploid cells. Components of the cell wall integrity pathway including the cell surface proteins Slg1p/Wsc1p, Wsc2p, Mid2p, and the mitogen-activated protein kinase (MAPK) Slt2p/Mpk1p contributed to multiple aspects of the FG response including cell elongation, cell-cell adherence, and agar invasion. Mid2p and Wsc2p stimulated the FG MAPK pathway through the signaling mucin Msb2p and components of the MAPK cascade. The FG pathway contributed to cell wall integrity in parallel with the cell wall integrity pathway and in opposition with the high osmolarity glycerol response pathway. Mass spectrometry approaches identified components of the filamentous cell wall including the mucin-like proteins Msb2p, Flo11p, and subtelomeric (silenced) mucin Flo10p. Secretion of Msb2p, which occurs as part of the maturation of the protein, was inhibited by the ss-1,3-glucan layer of the cell wall, which highlights a new regulatory aspect to cell wall remodeling in this organism. Disruption of ss-1,3-glucan linkages induced mucin shedding and resulted in defects in cell-cell adhesion and invasion of cells into the agar matrix.


Subject(s)
Cell Wall/metabolism , Mitogen-Activated Protein Kinases/metabolism , Saccharomyces cerevisiae/growth & development , Signal Transduction , Cell Wall/genetics , Gene Expression Regulation, Fungal , Mitogen-Activated Protein Kinases/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...