Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 30(7): 1367-1381, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38270582

ABSTRACT

PURPOSE: Paracrine activation of pro-fibrotic hedgehog (HH) signaling in pancreatic ductal adenocarcinoma (PDAC) results in stromal amplification that compromises tumor drug delivery, efficacy, and patient survival. Interdiction of HH-mediated tumor-stroma crosstalk with smoothened (SMO) inhibitors (SHHi) "primes" PDAC patient-derived xenograft (PDX) tumors for increased drug delivery by transiently increasing vascular patency/permeability, and thereby macromolecule delivery. However, patient tumor isolates vary in their responsiveness, and responders show co-induction of epithelial-mesenchymal transition (EMT). We aimed to identify the signal derangements responsible for EMT induction and reverse them and devise approaches to stratify SHHi-responsive tumors noninvasively based on clinically-quantifiable parameters. EXPERIMENTAL DESIGN: Animals underwent diffusion-weighted magnetic resonance (DW-MR) imaging for measurement of intratumor diffusivity. In parallel, tissue-level deposition of nanoparticle probes was quantified as a marker of vascular permeability/perfusion. Transcriptomic and bioinformatic analysis was employed to investigate SHHi-induced gene reprogramming and identify key "nodes" responsible for EMT induction. RESULTS: Multiple patient tumor isolates responded to short-term SHH inhibitor exposure with increased vascular patency and permeability, with proportionate increases in tumor diffusivity. Nonresponding PDXs did not. SHHi-treated tumors showed elevated FGF drive and distinctly higher nuclear localization of fibroblast growth factor receptor (FGFR1) in EMT-polarized tumor cells. Pan-FGFR inhibitor NVP-BGJ398 (Infigratinib) reversed the SHHi-induced EMT marker expression and nuclear FGFR1 accumulation without compromising the enhanced permeability effect. CONCLUSIONS: This dual-hit strategy of SMO and FGFR inhibition provides a clinically-translatable approach to compromise the profound impermeability of PDAC tumors. Furthermore, clinical deployment of DW-MR imaging could fulfill the essential clinical-translational requirement for patient stratification.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Animals , Heterografts , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Signal Transduction , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Disease Models, Animal , Cell Line, Tumor
2.
Sci Rep ; 10(1): 20987, 2020 12 02.
Article in English | MEDLINE | ID: mdl-33268815

ABSTRACT

Homeostatic control of neuronal excitability by modulation of synaptic inhibition (I) and excitation (E) of the principal neurons is important during brain maturation. The fundamental features of in-utero brain development, including local synaptic E-I ratio and bioenergetics, can be modeled by cerebral organoids (CO) that have exhibited highly regular nested oscillatory network events. Therefore, we evaluated a 'Phase Zero' clinical study platform combining broadband Vis/near-infrared(NIR) spectroscopy and electrophysiology with studying E-I ratio based on the spectral exponent of local field potentials and bioenergetics based on the activity of mitochondrial Cytochrome-C Oxidase (CCO). We found a significant effect of the age of the healthy controls iPSC CO from 23 days to 3 months on the CCO activity (chi-square (2, N = 10) = 20, p = 4.5400e-05), and spectral exponent between 30-50 Hz (chi-square (2, N = 16) = 13.88, p = 0.001). Also, a significant effect of drugs, choline (CHO), idebenone (IDB), R-alpha-lipoic acid plus acetyl-L-carnitine (LCLA), was found on the CCO activity (chi-square (3, N = 10) = 25.44, p = 1.2492e-05), spectral exponent between 1 and 20 Hz (chi-square (3, N = 16) = 43.5, p = 1.9273e-09) and 30-50 Hz (chi-square (3, N = 16) = 23.47, p = 3.2148e-05) in 34 days old CO from schizophrenia (SCZ) patients iPSC. We present the feasibility of a multimodal approach, combining electrophysiology and broadband Vis-NIR spectroscopy, to monitor neurodevelopment in brain organoid models that can complement traditional drug design approaches to test clinically meaningful hypotheses.


Subject(s)
Brain/growth & development , Organoids/growth & development , Acetylcarnitine/pharmacology , Brain/cytology , Brain/drug effects , Brain/physiology , Case-Control Studies , Cell Line , Choline/pharmacology , Electron Transport Complex IV/metabolism , Electrophysiology , Female , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Male , Mitochondria/metabolism , Organoids/drug effects , Organoids/physiology , Proof of Concept Study , Schizophrenia/metabolism , Spectroscopy, Near-Infrared , Thioctic Acid/pharmacology , Ubiquinone/analogs & derivatives , Ubiquinone/pharmacology
3.
Front Cell Neurosci ; 14: 233, 2020.
Article in English | MEDLINE | ID: mdl-33005129

ABSTRACT

Schizophrenia (SZ) is a neurodevelopmental genetic disorder in which maternal immune activation (MIA) and increased tumor necrosis factor-α (TNF-α) may contribute. Previous studies using iPSC-derived cerebral organoids and neuronal cells demonstrated developmental malformation and transcriptional dysregulations, including TNF receptors and their signaling genes, common to SZ patients with diverse genetic backgrounds. In the present study, we examined the significance of the common TNF receptor dysregulations by transiently exposing cerebral organoids from embryonic stem cells (ESC) and from representative control and SZ patient iPSCs to TNF. In control iPSC organoids, TNF produced malformations qualitatively similar in, but generally less pronounced than, the malformations of the SZ iPSC-derived organoids. TNF and SZ alone disrupted subcortical rosettes and dispersed proliferating Ki67+ neural progenitor cells (NPC) from the organoid ventricular zone (VZ) into the cortical zone (CZ). In the CZ, the absence of large ramified pan-Neu+ neurons coincided with loss of myelinated neurites despite increased cortical accumulation of O4+ oligodendrocytes. The number of calretinin+ interneurons increased; however, they lacked the preferential parallel orientation to the organoid surface. SZ and SZ+TNF affected fine cortical and subcortical organoid structure by replacing cells with extracellular matrix (ECM)-like fibers The SZ condition increased developmental vulnerability to TNF, leading to more pronounced changes in NPC, pan-Neu+ neurons, and interneurons. Both SZ- and TNF-induced malformations were associated with the loss of nuclear (n)FGFR1 form in the CZ and its upregulation in deep IZ regions, while in earlier studies blocking nFGFR1 reproduced cortical malformations observed in SZ. Computational analysis of ChiPseq and RNAseq datasets shows that nFGFR1 directly targets neurogenic, oligodendrogenic, cell migration, and ECM genes, and that the FGFR1-targeted TNF receptor and signaling genes are overexpressed in SZ NPC. Through these changes, the developing brain with the inherited SZ genome dysregulation may suffer increased vulnerability to TNF and thus, MIA.

4.
Acta Neurobiol Exp (Wars) ; 80(3): 305-321, 2020.
Article in English | MEDLINE | ID: mdl-32990288

ABSTRACT

In humans, pyruvate dehydrogenase complex (PDC) deficiency impairs brain energy metabolism by reducing the availability of the functional acetyl­CoA pool. This "hypometabolic defect" results in congenital lactic acidosis and abnormalities of brain morphology and function, ranging from mild ataxia to profound psychomotor retardation. Our previous study showed reduction in total cell number and dendritic arbors in the cerebellar Purkinje cells in systemic PDC­deficient mice. Phenylbutyrate has been shown to increase PDC activity in cultured fibroblasts from PDC­deficient patients. Hence, we investigated the effects of postnatal (days 2­35) phenylbutyrate administration on the cerebellar Purkinje cell population in PDC­deficient female mice. Histological analyses of different regions of cerebellar cortex from the brain­specific PDC­deficient saline­injected mice revealed statistically significant reduction in the Purkinje cell density and increased cell size of the individual Purkinje cell soma compared to control PDC­normal, saline­injected group. Administration of phenylbutyrate to control mice did not cause significant changes in the Purkinje cell density and cell size in the studied regions. In contrast, administration of phenylbutyrate variably lessened the ill effects of PDC deficiency on Purkinje cell populations in different areas of the cerebellum. Our results lend further support for the possible use of phenylbutyrate as a potential treatment for PDC deficiency.


Subject(s)
Brain/drug effects , Neurons/drug effects , Phenylbutyrates/pharmacology , Purkinje Cells/drug effects , Animals , Cerebellar Cortex/drug effects , Cerebellum/drug effects , Disease Models, Animal , Mice, Transgenic , Phenylbutyrates/metabolism , Purkinje Cells/cytology
5.
Int J Mol Sci ; 22(1)2020 Dec 31.
Article in English | MEDLINE | ID: mdl-33396256

ABSTRACT

During the development of mouse embryonic stem cells (ESC) to neuronal committed cells (NCC), coordinated changes in the expression of 2851 genes take place, mediated by the nuclear form of FGFR1. In this paper, widespread differences are demonstrated in the ESC and NCC inter- and intra-chromosomal interactions, chromatin looping, the formation of CTCF- and nFGFR1-linked Topologically Associating Domains (TADs) on a genome-wide scale and in exemplary HoxA-D loci. The analysis centered on HoxA cluster shows that blocking FGFR1 disrupts the loop formation. FGFR1 binding and genome locales are predictive of the genome interactions; likewise, chromatin interactions along with nFGFR1 binding are predictive of the genome function and correlate with genome regulatory attributes and gene expression. This study advances a topologically integrated genome archipelago model that undergoes structural transformations through the formation of nFGFR1-associated TADs. The makeover of the TAD islands serves to recruit distinct ontogenic programs during the development of the ESC to NCC.


Subject(s)
CCCTC-Binding Factor/metabolism , Cell Nucleus/genetics , Chromatin/metabolism , Embryonic Stem Cells/cytology , Genome , Neurogenesis , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , CCCTC-Binding Factor/genetics , Cell Differentiation , Chromatin/genetics , Chromosomes/genetics , Embryonic Stem Cells/metabolism , Mice , Molecular Conformation , Receptor, Fibroblast Growth Factor, Type 1/genetics
6.
IEEE Trans Neural Syst Rehabil Eng ; 27(2): 108-117, 2019 02.
Article in English | MEDLINE | ID: mdl-30624220

ABSTRACT

Miniaturization of implantable devices is an important challenge for future brain-computer interface applications, and in particular for achieving precise neuron stimulation. For stimulation that utilizes light, i.e., optogenetics, the light propagation behavior and interaction at the nanoscale with elements within the neuron is an important factor that needs to be considered when designing the device. This paper analyzes the effect of light behavior for a single neuron stimulation and focuses on the impact from different cell shapes. Based on the Mie scattering theory, the paper analyzes how the shape of the soma and the nucleus contributes to the focusing effect resulting in an intensity increase, which ensures that neurons can assist in transferring light through the tissue toward the target cells. At the same time, this intensity increase can in turn also stimulate neighboring cells leading to interference within the neural circuits. This paper also analyzes the ideal placements of the device with respect to the angle and position within the cortex that can enable axonal biophoton communications, which can contain light within the cell to avoid the interference.


Subject(s)
Brain-Computer Interfaces , Nanotechnology , Neurons/physiology , Neurons/radiation effects , Optogenetics/methods , Photic Stimulation , Algorithms , Axons/radiation effects , Cell Shape/radiation effects , Cerebral Cortex/cytology , Cerebral Cortex/radiation effects , Humans , Light , Neural Stem Cells/radiation effects , Neural Stem Cells/ultrastructure , Neurons/ultrastructure , Scattering, Radiation
7.
Results Probl Cell Differ ; 66: 137-162, 2018.
Article in English | MEDLINE | ID: mdl-30209658

ABSTRACT

Schizophrenia is a neurodevelopmental disorder characterized by complex aberrations in the structure, wiring, and chemistry of multiple neuronal systems. The abnormal developmental trajectory of the brain is established during gestation, long before clinical manifestation of the disease. Over 200 genes and even greater numbers of single nucleotide polymorphisms and copy number variations have been linked with schizophrenia. How does altered function of such a variety of genes lead to schizophrenia? We propose that the protein products of these altered genes converge on a common neurodevelopmental pathway responsible for the development of brain neural circuit and neurotransmitter systems. The results of a multichanneled investigation using induced pluripotent stem cell (iPSCs)- and embryonic stem cell (ESCs)-derived neuronal committed cells (NCCs) indicate an early (preneuronal) developmental-genomic etiology of schizophrenia and that the dysregulated developmental gene networks are common to genetically unrelated cases of schizophrenia. The results support a "watershed" mechanism in which mutations within diverse signaling pathways affect the common pan-ontogenic mechanism, integrative nuclear (n)FGFR1 signaling (INFS). Dysregulation of INFS in schizophrenia NCCs deconstructs coordinated gene networks and leads to formation of new networks by the dysregulated genes. This genome deprograming affects critical gene programs and pathways for neural development and functions. Studies show that the genomic deprograming reflect an altered nFGFR1-genome interactions and deregulation of miRNA genes by nFGFR1. In addition, changes in chromatin topology imposed by nFGFR1 may play a role in coordinate gene dysregulation in schizophrenia.


Subject(s)
Gene Expression Regulation , Genome, Human/genetics , Induced Pluripotent Stem Cells/metabolism , Schizophrenia/genetics , Schizophrenia/pathology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/pathology , Humans , Induced Pluripotent Stem Cells/pathology , Mutation
8.
J Cell Physiol ; 231(6): 1199-218, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26729628

ABSTRACT

Genetic experiments have positioned the fgfr1 gene at the top of the gene hierarchy that governs gastrulation, as well as the subsequent development of the major body axes, nervous system, muscles, and bones, by affecting downstream genes that control the cell cycle, pluripotency, and differentiation, as well as microRNAs. Studies show that this regulation is executed by a single protein, the nuclear isoform of FGFR1 (nFGFR1), which integrates signals from development-initiating factors, such as retinoic acid (RA), and operates at the interface of genomic and epigenomic information. nFGFR1 cooperates with a multitude of transcriptional factors (TFs), and targets thousands of genes encoding for mRNAs, as well as miRNAs in top ontogenic networks. nFGFR1 binds to the promoters of ancient proto-oncogenes and tumor suppressor genes, in addition to binding to metazoan morphogens that delineate body axes, and construct the nervous system, as well as mesodermal and endodermal tissues. The discovery of pan-ontogenic gene programming by integrative nuclear FGFR1 signaling (INFS) impacts our understanding of ontogeny, as well as developmental pathologies, and holds new promise for reconstructive medicine, and cancer therapy.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation, Developmental , Pluripotent Stem Cells/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , Animals , Binding Sites , Gene Regulatory Networks , Genetic Predisposition to Disease , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Promoter Regions, Genetic , Protein Interaction Maps , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Transcription, Genetic
9.
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
10.
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
11.
Behav Pharmacol ; 26(1-2): 217-26, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25485645

ABSTRACT

Individuals with schizophrenia smoke at high frequency relative to the general population. Despite the harmful effects of cigarette smoking, smoking among schizophrenic patients improves cognitive impairments not addressed or worsened by common neuroleptics. Varenicline, a nonselective neuronal nicotinic receptor (NNR) agonist and full agonist of 5-HT3A receptors, helps reduce smoking among schizophrenic patients. To determine whether varenicline also improves a cognitive symptom of schizophrenia, namely, impaired sensory gating, a transgenic mouse with schizophrenia, th-fgfr1(tk-), was used. Varenicline dose-dependently increased prepulse inhibition (PPI) of the startle response, a measure of sensory gating, in th-fgfr1(tk-) mice and normalized PPI deficits relative to nontransgenic controls. With the highest dose (10 mg/kg), however, there was a robust elevation of PPI and startle response, as well as reduced exploratory behavior in the open field and elevated plus maze. Pretreatment with the nonspecific NNR antagonist mecamylamine attenuated the exaggerated PPI response and, similar to the 5-HT3A receptor antagonist ondansetron, it prevented the reduction in exploratory behavior. Collectively, these results indicate that varenicline at low-to-moderate doses may be beneficial against impaired sensory gating in schizophrenia; however, higher doses may induce anxiogenic effects, which can be prevented with antagonists of NNRs or 5-HT3A receptors.


Subject(s)
Benzazepines/pharmacology , Exploratory Behavior/drug effects , Nicotinic Agonists/pharmacology , Quinoxalines/pharmacology , Sensory Gating/drug effects , Animals , Benzazepines/administration & dosage , Cognition Disorders/drug therapy , Cognition Disorders/etiology , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Nicotinic Agonists/administration & dosage , Quinoxalines/administration & dosage , Receptors, Serotonin, 5-HT3/drug effects , Receptors, Serotonin, 5-HT3/metabolism , Reflex, Startle/drug effects , Schizophrenia/drug therapy , Schizophrenia/physiopathology , Serotonin 5-HT3 Receptor Agonists/pharmacology , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Varenicline
12.
Metab Brain Dis ; 28(4): 721-5, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24043569

ABSTRACT

Fetal development in an obese maternal intrauterine environment has been shown to predispose the offspring for a number of metabolic disorders in later life. The observation that a large percentage of women of child-bearing age in the US are overweight/obese during pregnancy is therefore a source of concern. A high fat (HF) diet-induced obesity in female rats has been used as a model for maternal obesity. The objective of this study was to determine cellular development in brains of term fetuses of obese rats fed a HF diet from the time of weaning. Fetal brains were dissected out on gestational day 21 and processed for immunohistochemical analysis in the hypothalamic as well as extra-hypothalamic regions. The major observation of this study is that fetal development in the obese HF female rat induced several alterations in the HF fetal brain. Marked increases were observed in orexigenic signaling and a significant decrease was observed for anorexigenic signaling in the vicinity of the 3rd ventricle in HF brains. Additionally, our results indicated diminished migration and maturation of stem-like cells in the 3rd ventricular region as well as in the brain cortex. The results from the present study indicate developmental alterations in the hypothalamic and extra-hypothalamic regions in the HF fetal brain suggestive of a predisposition for the development of obesity and possibly neurodevelopmental abnormalities in the offspring.


Subject(s)
Brain/embryology , Diet, High-Fat/adverse effects , Nervous System Diseases/etiology , Obesity/metabolism , Prenatal Exposure Delayed Effects/etiology , Animals , Female , Nervous System Diseases/metabolism , Nervous System Diseases/physiopathology , Obesity/etiology , Obesity/physiopathology , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/physiopathology , Rats , Rats, Sprague-Dawley
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.
Pharmacol Biochem Behav ; 111: 1-10, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23872135

ABSTRACT

The degeneration of the nigrostriatal dopamine (DA) system underlies the motor deficits in Parkinson's disease (PD). In recent years, epidemiological reports that smokers have lower incidences of PD have brought attention to the nicotinic acetylcholine system as a potential target for novel therapeutics. Nicotine, an agonist of neuronal nicotinic receptors (NNRs), modulates functions relevant to PD via stimulation of dopaminergic transmission in the nigrostriatal pathway, particularly via activation of α6ß2* and α4ß2* NNRs. Recently, reduced support of DA neurons by neurotrophic growth factors has been described in PD. Fibroblast growth factor (FGF) is critical for the development and protection of adult DA neurons. In FGF-2 knockout mice and the related th-fgfr1(tk-) mouse model there is heightened sensitivity to DA neuronal oxidative neurotoxin 6-hydroxydopamine (6-OHDA). In the present study, FGF-deficient transgenic mice th-fgfr1(tk-) were used to analyze the effects of novel full (TC-8831) and partial (TC-8581) agonists of ß2-containing nicotinic receptors on impaired motor behavior following unilateral 6-OHDA lesions. The lesions generated spontaneous (drug-naïve) turning asymmetries that correlated exponentially with the depletion of DA biomarkers in the lesioned striata. These mice also exhibited a reduced capacity to remain on the accelerating rotarod. Oral administration of TC-8831, an NNR agonist with high specificity for ß2 subunits and a full agonist at producing DA release from striatal synaptosomes, attenuated unidirectional turning and improved motor coordination. In contrast, partial ß2 NNR agonist TC-8581 had no effect on behaviors in this model. This study demonstrates the potential of NNR targeting-compounds to facilitate motor function in PD.


Subject(s)
Azabicyclo Compounds/pharmacology , Cyclopropanes/pharmacology , Disease Models, Animal , Motor Activity/drug effects , Neurons/drug effects , Nicotinic Agonists/pharmacology , Parkinson Disease/physiopathology , Pyridines/pharmacology , Receptors, Nicotinic/physiology , Animals , Behavior, Animal , Cell Line , Dopamine/metabolism , Mice , Mice, Knockout , Neurons/metabolism
16.
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
17.
Nutr Neurosci ; 16(3): 96-103, 2013 May.
Article in English | MEDLINE | ID: mdl-23006985

ABSTRACT

OBJECTIVES: Female rat neonates reared on a high carbohydrate (HC) milk formula developed chronic hyperinsulinemia and adult-onset obesity (HC phenotype). Furthermore, we have shown that fetal development in the HC intrauterine environment (maternal obesity complicated with hyperinsulinemia, hyperleptinemia, and increased levels of proinflammatory markers) resulted in increased levels of serum insulin and leptin in term HC fetuses and the spontaneous transfer of the HC phenotype to the adult offspring. The objectives of this study are to identify changes in global gene expression pattern and cellular development in term HC fetal brains in response to growth in the adverse intrauterine environment of the obese HC female rat. METHODS: GeneChip analysis was performed on total RNA obtained from fetal brains for global gene expression studies and immunohistochemical analysis was performed on fetal brain slices for investigation of cellular development in term HC fetal brains. RESULTS: Gene expression profiling identified changes in several clusters of genes that could contribute to the transfer of the maternal phenotype (chronic hyperinsulinemia and adult-onset obesity) to the HC offspring. Immunohistochemical analysis indicated diminished proliferation and neuronal maturation of stem-like cells lining the third ventricle, hypothalamic region, and the cerebral cortex in HC fetal brains. DISCUSSION: These results suggest that maternal obesity during pregnancy could alter the developmental program of specific fetal brain cell-networks. These defects could underlie pathologies such as metabolic syndrome and possibly some neurological disorders in the offspring at a later age.


Subject(s)
Dietary Carbohydrates/adverse effects , Gene Expression , Hypothalamus/embryology , Obesity/pathology , Animals , Cell Proliferation , Dietary Carbohydrates/administration & dosage , Female , Fetal Development , Gene Expression Profiling , Hyperinsulinism/pathology , Hypothalamus/cytology , Hypothalamus/pathology , Insulin/blood , Leptin/blood , Male , Phenotype , Pregnancy , Rats
18.
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
19.
J Biol Chem ; 287(24): 19827-40, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22514272

ABSTRACT

Experiments in mice deficient for Nurr1 or expressing the dominant-negative FGF receptor (FGFR) identified orphan nuclear receptor Nurr1 and FGFR1 as essential factors in development of mesencephalic dopaminergic (mDA) neurons. FGFR1 affects brain cell development by two distinct mechanisms. Activation of cell surface FGFR1 by secreted FGFs stimulates proliferation of neural progenitor cells, whereas direct integrative nuclear FGFR1 signaling (INFS) is associated with an exit from the cell cycle and neuronal differentiation. Both Nurr1 and INFS activate expression of neuronal genes, such as tyrosine hydroxylase (TH), which is the rate-limiting enzyme in dopamine synthesis. Here, we show that nuclear FGFR1 and Nurr1 are expressed in the nuclei of developing TH-positive cells in the embryonic ventral midbrain. Both nuclear receptors were effectively co-immunoprecipitated from the ventral midbrain of FGF-2-deficient embryonic mice, which previously showed an increase of mDA neurons and enhanced nuclear FGFR1 accumulation. Immunoprecipitation and co-localization experiments showed the presence of Nurr1 and FGFR1 in common nuclear protein complexes. Fluorescence recovery after photobleaching and chromatin immunoprecipitation experiments demonstrated the Nurr1-mediated shift of nuclear FGFR1-EGFP mobility toward a transcriptionally active population and that both Nurr1 and FGFR1 bind to a common region in the TH gene promoter. Furthermore, nuclear FGFR1 or its 23-kDa FGF-2 ligand (FGF-2(23)) enhances Nurr1-dependent activation of the TH gene promoter. Transcriptional cooperation of FGFR1 with Nurr1 was confirmed on isolated Nurr1-binding elements. The proposed INFS/Nurr1 nuclear partnership provides a novel mechanism for TH gene regulation in mDA neurons and a potential therapeutic target in neurodevelopmental and neurodegenerative disorders.


Subject(s)
Cell Nucleus/metabolism , Dopaminergic Neurons/metabolism , Mesencephalon/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction/physiology , Animals , Cell Line , Cell Nucleus/genetics , Dopaminergic Neurons/cytology , Mesencephalon/cytology , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Response Elements/physiology , Transcription, Genetic/physiology , Tyrosine 3-Monooxygenase/biosynthesis
20.
Integr Biol (Camb) ; 1(5-6): 394-403, 2009 Jun.
Article in English | MEDLINE | ID: mdl-20023746

ABSTRACT

Neurogenesis, the process of differentiation of neuronal stem/progenitor cells (NS/PC) into mature neurons, holds the key to the treatment of various neurodegenerative disorders, which are a major health issue for the world's aging population. We report that targeting the novel integrative nuclear FGF Receptor 1 signaling (INFS) pathway enhances the latent potential of NS/PCs to undergo neuronal differentiation, thus promoting neurogenesis in the adult brain. Employing organically modified silica (ORMOSIL)-DNA nanoplexes to efficiently transfect recombinant nuclear forms of FGFR1 and its FGF-2 ligand into the brain subventricular zone, we find that INFS stimulates the NS/PC to withdraw from the cell cycle, differentiate into doublecortin expressing migratory neuroblasts and neurons that migrate to the olfactory bulb, subcortical brain regions and in the brain cortex. Thus, nanoparticle-mediated non-viral gene transfer may be used to induce selective differentiation of NS/PCs, providing a potentially significant impact on the treatment of a broad range of neurological disorders.


Subject(s)
Brain/cytology , Brain/physiology , Nanoparticles/chemistry , Neurogenesis/physiology , Neurons/cytology , Neurons/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Transfection/methods , Animals , Cells, Cultured , Drug Carriers/chemistry , Female , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...