35 results on '"Stachowiak MK"'
Search Results
2. Systems Genome: Coordinated Gene Activity Networks, Recurring Coordination Modules, and Genome Homeostasis in Developing Neurons.
- Author
-
Dhiman S, Manoj N, Liput M, Sangwan A, Diehl J, Balcerak A, Sudhakar S, Augustyniak J, Jornet JM, Bae Y, Stachowiak EK, Dutta A, and Stachowiak MK
- Subjects
- Animals, Humans, Cell Differentiation genetics, Genome, Neurogenesis genetics, Receptor, Fibroblast Growth Factor, Type 1 genetics, Receptor, Fibroblast Growth Factor, Type 1 metabolism, Gene Regulatory Networks, Homeostasis genetics, Neurons metabolism
- Abstract
As human progenitor cells differentiate into neurons, the activities of many genes change; these changes are maintained within a narrow range, referred to as genome homeostasis. This process, which alters the synchronization of the entire expressed genome, is distorted in neurodevelopmental diseases such as schizophrenia. The coordinated gene activity networks formed by altering sets of genes comprise recurring coordination modules, governed by the entropy-controlling action of nuclear FGFR1, known to be associated with DNA topology. These modules can be modeled as energy-transferring circuits, revealing that genome homeostasis is maintained by reducing oscillations (noise) in gene activity while allowing gene activity changes to be transmitted across networks; this occurs more readily in neuronal committed cells than in neural progenitors. These findings advance a model of an "entangled" global genome acting as a flexible, coordinated homeostatic system that responds to developmental signals, is governed by nuclear FGFR1, and is reprogrammed in disease.
- Published
- 2024
- Full Text
- View/download PDF
3. Global Genome Conformational Programming during Neuronal Development Is Associated with CTCF and Nuclear FGFR1-The Genome Archipelago Model.
- Author
-
Decker B, Liput M, Abdellatif H, Yergeau D, Bae Y, Jornet JM, Stachowiak EK, and Stachowiak MK
- Subjects
- 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, CCCTC-Binding Factor metabolism, Cell Nucleus genetics, Chromatin metabolism, Embryonic Stem Cells cytology, Genome, Neurogenesis, Receptor, Fibroblast Growth Factor, Type 1 metabolism
- 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.
- Published
- 2020
- Full Text
- View/download PDF
4. A proof of concept 'phase zero' study of neurodevelopment using brain organoid models with Vis/near-infrared spectroscopy and electrophysiology.
- Author
-
Dutta A, Karanth SS, Bhattacharya M, Liput M, Augustyniak J, Cheung M, Stachowiak EK, and Stachowiak MK
- Subjects
- 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, Brain growth & development, Organoids growth & development
- 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.
- Published
- 2020
- Full Text
- View/download PDF
5. Immune Factor, TNFα, Disrupts Human Brain Organoid Development Similar to Schizophrenia-Schizophrenia Increases Developmental Vulnerability to TNFα.
- Author
-
Benson CA, Powell HR, Liput M, Dinham S, Freedman DA, Ignatowski TA, Stachowiak EK, and Stachowiak MK
- 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., (Copyright © 2020 Benson, Powell, Liput, Dinham, Freedman, Ignatowski, Stachowiak and Stachowiak.)- Published
- 2020
- Full Text
- View/download PDF
6. Bioenergy Crisis in Coronavirus Diseases?
- Author
-
Dutta A, Das A, Kondziella D, and Stachowiak MK
- Abstract
Coronavirus disease (COVID-19) has been declared as a pandemic by the World Health Organization (WHO).[...]., Competing Interests: The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results
- Published
- 2020
- Full Text
- View/download PDF
7. Phenylbutyrate administration reduces changes in the cerebellar Purkinje cells population in PDC‑deficient mice.
- Author
-
Klejbor I, Mahmood S, Melka N, Ebertowska A, Morys J, Stachowiak EK, Stachowiak MK, and Patel MS
- Subjects
- Animals, Cerebellar Cortex drug effects, Cerebellum drug effects, Disease Models, Animal, Mice, Transgenic, Phenylbutyrates metabolism, Purkinje Cells cytology, Brain drug effects, Neurons drug effects, Phenylbutyrates pharmacology, Purkinje Cells drug effects
- Abstract
In humans, pyruvate dehydrogenase complex (PDC) deficiency impairs brain energy metabolism by reducing the availability of the functional acetyl‑CoA 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 PDC‑deficient mice. Phenylbutyrate has been shown to increase PDC activity in cultured fibroblasts from PDC‑deficient patients. Hence, we investigated the effects of postnatal (days 2‑35) phenylbutyrate administration on the cerebellar Purkinje cell population in PDC‑deficient female mice. Histological analyses of different regions of cerebellar cortex from the brain‑specific PDC‑deficient saline‑injected mice revealed statistically significant reduction in the Purkinje cell density and increased cell size of the individual Purkinje cell soma compared to control PDC‑normal, saline‑injected 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.
- Published
- 2020
8. Analysis of Light Propagation on Physiological Properties of Neurons for Nanoscale Optogenetics.
- Author
-
Wirdatmadja S, Johari P, Desai A, Bae Y, Stachowiak EK, Stachowiak MK, Jornet JM, and Balasubramaniam S
- Subjects
- 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, Brain-Computer Interfaces, Nanotechnology, Neurons physiology, Neurons radiation effects, Optogenetics methods, Photic Stimulation
- 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.
- Published
- 2019
- Full Text
- View/download PDF
9. Cerebral organoids reveal early cortical maldevelopment in schizophrenia-computational anatomy and genomics, role of FGFR1.
- Author
-
Stachowiak EK, Benson CA, Narla ST, Dimitri A, Chuye LEB, Dhiman S, Harikrishnan K, Elahi S, Freedman D, Brennand KJ, Sarder P, and Stachowiak MK
- Subjects
- Calbindin 2 metabolism, Cerebral Cortex metabolism, Embryonic Stem Cells pathology, Humans, Interneurons metabolism, Interneurons pathology, Receptor, Fibroblast Growth Factor, Type 1 genetics, Reelin Protein, Schizophrenia genetics, Schizophrenia metabolism, Cerebral Cortex pathology, Receptor, Fibroblast Growth Factor, Type 1 metabolism, Schizophrenia pathology
- Abstract
Studies of induced pluripotent stem cells (iPSCs) from schizophrenia patients and control individuals revealed that the disorder is programmed at the preneuronal stage, involves a common dysregulated mRNA transcriptome, and identified Integrative Nuclear FGFR1 Signaling a common dysregulated mechanism. We used human embryonic stem cell (hESC) and iPSC-derived cerebral organoids from four controls and three schizophrenia patients to model the first trimester of in utero brain development. The schizophrenia organoids revealed an abnormal scattering of proliferating Ki67+ neural progenitor cells (NPCs) from the ventricular zone (VZ), throughout the intermediate (IZ) and cortical (CZ) zones. TBR1 pioneer neurons and reelin, which guides cortico-petal migration, were restricted from the schizophrenia cortex. The maturing neurons were abundantly developed in the subcortical regions, but were depleted from the schizophrenia cortex. The decreased intracortical connectivity was denoted by changes in the orientation and morphology of calretinin interneurons. In schizophrenia organoids, nuclear (n)FGFR1 was abundantly expressed by developing subcortical cells, but was depleted from the neuronal committed cells (NCCs) of the CZ. Transfection of dominant negative and constitutively active nFGFR1 caused widespread disruption of the neuro-ontogenic gene networks in hESC-derived NPCs and NCCs. The fgfr1 gene was the most prominent FGFR gene expressed in NPCs and NCCs, and blocking with PD173074 reproduced both the loss of nFGFR1 and cortical neuronal maturation in hESC cerebral organoids. We report for the first time, progression of the cortical malformation in schizophrenia and link it to altered FGFR1 signaling. Targeting INFS may offer a preventive treatment of schizophrenia.
- Published
- 2017
- Full Text
- View/download PDF
10. Global Developmental Gene Programing Involves a Nuclear Form of Fibroblast Growth Factor Receptor-1 (FGFR1).
- Author
-
Terranova C, Narla ST, Lee YW, Bard J, Parikh A, Stachowiak EK, Tzanakakis ES, Buck MJ, Birkaya B, and Stachowiak MK
- Subjects
- 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, Cell Nucleus metabolism, Genome, Receptor, Fibroblast Growth Factor, Type 1 metabolism
- 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/β-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.
- Published
- 2015
- Full Text
- View/download PDF
11. Regulation of neuronal differentiation by proteins associated with nuclear bodies.
- Author
-
Förthmann B, van Bergeijk J, Lee YW, Lübben V, Schill Y, Brinkmann H, Ratzka A, Stachowiak MK, Hebert M, Grothe C, and Claus P
- Subjects
- Animals, Cells, Cultured, Fibroblast Growth Factor 2 metabolism, Humans, Neurites physiology, Nuclear Proteins metabolism, PC12 Cells, Protein Binding, Rats, SMN Complex Proteins metabolism, Cell Differentiation genetics, Cell Nucleus metabolism, Coiled Bodies metabolism, Neurogenesis genetics, Neurons physiology, SMN Complex Proteins physiology
- Abstract
Nuclear bodies are large sub-nuclear structures composed of RNA and protein molecules. The Survival of Motor Neuron (SMN) protein localizes to Cajal bodies (CBs) and nuclear gems. Diminished cellular concentration of SMN is associated with the neurodegenerative disease Spinal Muscular Atrophy (SMA). How nuclear body architecture and its structural components influence neuronal differentiation remains elusive. In this study, we analyzed the effects of SMN and two of its interaction partners in cellular models of neuronal differentiation. The nuclear 23 kDa isoform of Fibroblast Growth Factor - 2 (FGF-2(23)) is one of these interacting proteins - and was previously observed to influence nuclear bodies by destabilizing nuclear gems and mobilizing SMN from Cajal bodies (CBs). Here we demonstrate that FGF-2(23) blocks SMN-promoted neurite outgrowth, and also show that SMN disrupts FGF-2(23)-dependent transcription. Our results indicate that FGF-2(23) and SMN form an inactive complex that interferes with neuronal differentiation by mutually antagonizing nuclear functions. Coilin is another nuclear SMN binding partner and a marker protein for Cajal bodies (CBs). In addition, coilin is essential for CB function in maturation of small nuclear ribonucleoprotein particles (snRNPs). The role of coilin outside of Cajal bodies and its putative impacts in tissue differentiation are poorly defined. The present study shows that protein levels of nucleoplasmic coilin outside of CBs decrease during neuronal differentiation. Overexpression of coilin has an inhibitory effect on neurite outgrowth. Furthermore, we find that nucleoplasmic coilin inhibits neurite outgrowth independent of SMN binding revealing a new function for coilin in neuronal differentiation.
- Published
- 2013
- Full Text
- View/download PDF
12. Activation of developmental nuclear fibroblast growth factor receptor 1 signaling and neurogenesis in adult brain by α7 nicotinic receptor agonist.
- Author
-
Narla ST, Klejbor I, Birkaya B, Lee YW, Morys J, Stachowiak EK, Prokop D, Bencherif M, and Stachowiak MK
- Subjects
- 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, Brain physiology, Neurogenesis physiology, Nicotinic Agonists pharmacology, Receptor, Fibroblast Growth Factor, Type 1 metabolism, Signal Transduction physiology
- 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) βIII-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 βIII-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 βIII-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.
- Published
- 2013
- Full Text
- View/download PDF
13. NGF-induced cell differentiation and gene activation is mediated by integrative nuclear FGFR1 signaling (INFS).
- Author
-
Lee YW, Stachowiak EK, Birkaya B, Terranova C, Capacchietti M, Claus P, Aletta JM, and Stachowiak MK
- Subjects
- 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, Cell Differentiation drug effects, Cell Nucleus metabolism, Nerve Growth Factor pharmacology, Receptor, Fibroblast Growth Factor, Type 1 physiology, Transcriptional Activation drug effects
- 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, βIII 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.
- Published
- 2013
- Full Text
- View/download PDF
14. Immobile survival of motoneuron (SMN) protein stored in Cajal bodies can be mobilized by protein interactions.
- Author
-
Förthmann B, Brinkmann H, Ratzka A, Stachowiak MK, Grothe C, and Claus P
- Subjects
- Fibroblast Growth Factor 2 metabolism, Fluorescence Recovery After Photobleaching, HEK293 Cells, Humans, Muscular Atrophy metabolism, Muscular Atrophy pathology, Nuclear Proteins metabolism, Protein Binding, Protein Interaction Maps, Protein Isoforms metabolism, Ribonucleoprotein, U4-U6 Small Nuclear metabolism, Coiled Bodies metabolism, SMN Complex Proteins metabolism
- Abstract
Reduced levels of survival of motoneuron (SMN) protein lead to spinal muscular atrophy, but it is still unknown how SMN protects motoneurons in the spinal cord against degeneration. In the nucleus, SMN is associated with two types of nuclear bodies denoted as gems and Cajal bodies (CBs). The 23 kDa isoform of fibroblast growth factor-2 (FGF-2(23)) is a nuclear protein that binds to SMN and destabilizes the SMN-Gemin2 complex. In the present study, we show that FGF-2(23) depletes SMN from CBs without affecting their general structure. FRAP analysis of SMN-EGFP in CBs demonstrated that the majority of SMN in CBs remained mobile and allowed quantification of fast, slow and immobile nuclear SMN populations. The potential for SMN release was confirmed by in vivo photoconversion of SMN-Dendra2, indicating that CBs concentrate immobile SMN that could have a specialized function in CBs. FGF-2(23) accelerated SMN release from CBs, accompanied by a conversion of immobile SMN into a mobile population. Furthermore, FGF-2(23) caused snRNP accumulation in CBs. We propose a model in which Cajal bodies store immobile SMN that can be mobilized by its nuclear interaction partner FGF-2(23), leading to U4 snRNP accumulation in CBs, indicating a role for immobile SMN in tri-snRNP assembly.
- Published
- 2013
- Full Text
- View/download PDF
15. Cerebral Developmental Abnormalities in a Mouse with Systemic Pyruvate Dehydrogenase Deficiency.
- Author
-
Pliss L, Hausknecht KA, Stachowiak MK, Dlugos CA, Richards JB, and Patel MS
- Subjects
- Animals, Brain metabolism, Brain pathology, Brain Diseases etiology, Carbohydrate Metabolism, Female, Lipogenesis physiology, Male, Mice, Mice, Knockout, Pyruvate Dehydrogenase Complex Deficiency Disease physiopathology, Brain abnormalities, Brain Diseases pathology, Disease Models, Animal, Pyruvate Dehydrogenase Complex physiology, Pyruvate Dehydrogenase Complex Deficiency Disease complications
- Abstract
Unlabelled: Pyruvate dehydrogenase (PDH) complex (PDC) deficiency is an inborn error of pyruvate metabolism causing a variety of neurologic manifestations. Systematic analyses of development of affected brain structures and the cellular processes responsible for their impairment have not been performed due to the lack of an animal model for PDC deficiency., Methods: In the present study we investigated a murine model of systemic PDC deficiency by interrupting the X-linked Pdha1 gene encoding the α subunit of PDH to study its role on brain development and behavioral studies., Results: Male embryos died prenatally but heterozygous females were born. PDC activity was reduced in the brain and other tissues in female progeny compared to age-matched control females. Immunohistochemical analysis of several brain regions showed that approximately 40% of cells were PDH(-). The oxidation of glucose to CO2 and incorporation of glucose-carbon into fatty acids were reduced in brain slices from 15 day-old PDC-deficient females. Histological analyses showed alterations in several structures in white and gray matters in 35 day-old PDC-deficient females. Reduction in total cell number and reduced dendritic arbors in Purkinje neurons were observed in PDC-deficient females. Furthermore, cell proliferation, migration and differentiation into neurons by newly generated cells were reduced in the affected females during pre- and postnatal periods. PDC-deficient mice had normal locomotor activity in a novel environment but displayed decreased startle responses to loud noises and there was evidence of abnormal pre-pulse inhibition of the startle reflex., Conclusions: The results show that a reduction in glucose metabolism resulting in deficit in energy production and fatty acid biosynthesis impairs cellular differentiation and brain development in PDC-deficient mice.
- Published
- 2013
- Full Text
- View/download PDF
16. Cooperation of nuclear fibroblast growth factor receptor 1 and Nurr1 offers new interactive mechanism in postmitotic development of mesencephalic dopaminergic neurons.
- Author
-
Baron O, Förthmann B, Lee YW, Terranova C, Ratzka A, Stachowiak EK, Grothe C, Claus P, and Stachowiak MK
- Subjects
- 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, 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
- 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.
- Published
- 2012
- Full Text
- View/download PDF
17. Fibroblast growth factor receptor-1 (FGFR1) nuclear dynamics reveal a novel mechanism in transcription control.
- Author
-
Dunham-Ems SM, Lee YW, Stachowiak EK, Pudavar H, Claus P, Prasad PN, and Stachowiak MK
- Subjects
- Animals, CREB-Binding Protein metabolism, Cell Compartmentation, Cell Line, Fibroblast Growth Factor 2 metabolism, Fluorescence Recovery After Photobleaching, Green Fluorescent Proteins metabolism, Humans, Mice, Models, Biological, Molecular Weight, Nuclear Matrix metabolism, Protein Binding, Protein Structure, Tertiary, Protein Transport, Receptor, Fibroblast Growth Factor, Type 1 chemistry, Recombinant Fusion Proteins metabolism, Ribosomal Protein S6 Kinases, 90-kDa metabolism, Transcriptional Activation genetics, Transfection, Cell Nucleus metabolism, Receptor, Fibroblast Growth Factor, Type 1 metabolism, Transcription, Genetic
- Abstract
Nuclear FGFR1 acts as a developmental gene regulator in cooperation with FGF-2, RSK1, and CREB-binding protein (CBP). FRAP analysis revealed three nuclear FGFR1 populations: i) a fast mobile, ii) a slower mobile population reflecting chromatin-bound FGFR1, and iii) an immobile FGFR1 population associated with the nuclear matrix. Factors (cAMP, CBP) that induce FGFR1-mediated gene activation shifted FGFR1 from the nuclear matrix (immobile) to chromatin (slow) and reduced the movement rate of the chromatin-bound population. Transcription inhibitors accelerated FGFR1 movement; the content of the chromatin-bound slow FGFR1 decreased, whereas the fast population increased. The transcriptional activation appears to involve conversion of the immobile matrix-bound and the fast nuclear FGFR1 into a slow chromatin-binding population through FGFR1's interaction with CBP, RSK1, and the high-molecular-weight form of FGF-2. Our findings support a general mechanism in which gene activation is governed by protein movement and collisions with other proteins and nuclear structures.
- Published
- 2009
- Full Text
- View/download PDF
18. Factors controlling fibroblast growth factor receptor-1's cytoplasmic trafficking and its regulation as revealed by FRAP analysis.
- Author
-
Dunham-Ems SM, Pudavar HE, Myers JM, Maher PA, Prasad PN, and Stachowiak MK
- Subjects
- Animals, Brefeldin A pharmacology, Cattle, Cell Nucleus chemistry, Cell Nucleus metabolism, Cells, Cultured, Fluorescence Recovery After Photobleaching, Golgi Apparatus metabolism, Green Fluorescent Proteins analysis, Green Fluorescent Proteins genetics, Humans, Models, Biological, Protein Transport drug effects, Receptor, Fibroblast Growth Factor, Type 1 analysis, Receptor, Fibroblast Growth Factor, Type 1 genetics, Recombinant Fusion Proteins analysis, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Ribosomal Protein S6 Kinases, 90-kDa analysis, Cytoplasm metabolism, Protein Biosynthesis, Receptor, Fibroblast Growth Factor, Type 1 metabolism, Ribosomal Protein S6 Kinases, 90-kDa metabolism
- Abstract
Biochemical and microscopic studies have indicated that FGFR1 is a transmembrane and soluble protein present in the cytosol and nucleus. How FGFR1 enters the cytosol and subsequently the nucleus to control cell development and associated gene activities has become a compelling question. Analyses of protein synthesis, cytoplasmic subcompartmental distribution and movement of FGFR1-EGFP and FGFR1 mutants showed that FGFR1 exists as three separate populations (a) a newly synthesized, highly mobile, nonglycosylated, cytosolic receptor that is depleted by brefeldin A and resides outside the ER-Golgi lumen, (b) a slowly diffusing membrane receptor population, and (c) an immobile membrane pool increased by brefeldin A. RSK1 increases the highly mobile cytosolic FGFR1 population and its overall diffusion rate leading to increased FGFR1 nuclear accumulation, which coaccumulates with RSK1. A model is proposed in which newly synthesized FGFR1 can enter the (a) "nuclear pathway," where the nonglycosylated receptor is extruded from the pre-Golgi producing highly mobile cytosolic receptor molecules that rapidly accumulate in the nucleus or (b) "membrane pathway," in which FGFR1 is processed through the Golgi, where its movement is spatially restricted to trans-Golgi membranes with limited lateral mobility. Entrance into the nuclear pathway is favored by FGFR1's interaction with kinase active RSK1.
- Published
- 2006
- Full Text
- View/download PDF
19. In vivo gene transfer to the brain cortex using a single injection of HSV-1 vector into the medial septum.
- Author
-
Tabbaa S, Corso T, Jenkins L, Tran R, Bloom D, and Stachowiak MK
- Subjects
- Animals, Genes, Reporter, Humans, Microinjections, Random Allocation, Rats, Rats, Inbred F344, beta-Galactosidase genetics, beta-Galactosidase metabolism, Gene Transfer Techniques, Genetic Vectors, Herpesvirus 1, Human genetics, Herpesvirus 1, Human metabolism, Septal Nuclei anatomy & histology, Septal Nuclei physiology
- Abstract
This study shows that an ICP4-replication-deficient herpes simplex virus containing the Moloney murine leukaemia virus LTR fused with the coding sequence for the beta-galactosidase gene can be used as a very effective vector for delivering the beta-galactosidase reporter gene into the rat brain septum. F344 rats received bilateral stereotaxic injections into the nucleus of the diagonal band and into the medial septum. The X-gal stain was used to detect the activity of the expressed beta-galactosidase enzyme. The delivered reporter gene was expressed successfully not only in the neuronal cells of the injected areas but also in cells that project to the injection area such as cortex cells about 6 mm away from the injection sites. Expression was visible at 1, 3 and 9 weeks following injection. We conclude that this vector can effectively deliver genes into different regions of the mature mammalian brain and also to areas distant from the injection site.
- Published
- 2005
20. Organically modified silica nanoparticles: a nonviral vector for in vivo gene delivery and expression in the brain.
- Author
-
Bharali DJ, Klejbor I, Stachowiak EK, Dutta P, Roy I, Kaur N, Bergey EJ, Prasad PN, and Stachowiak MK
- Subjects
- Animals, Brain cytology, Bromodeoxyuridine metabolism, Cell Proliferation, Green Fluorescent Proteins genetics, Mice, Microscopy, Confocal, Receptor, Fibroblast Growth Factor, Type 1 metabolism, Silicon Dioxide, Stem Cells physiology, Brain metabolism, DNA metabolism, Gene Expression, Genetic Therapy methods, Genetic Vectors therapeutic use, Nanostructures, Transfection methods
- Abstract
This article reports on the application of organically modified silica (ORMOSIL) nanoparticles as a nonviral vector for efficient in vivo gene delivery. Highly monodispersed, stable aqueous suspension of nanoparticles, surface-functionalized with amino groups for binding of DNA, were prepared and characterized. Stereotaxic injections of nanoparticles, complexed with plasmid DNA encoding for EGFP, into the mouse ventral midbrain and into lateral ventricle, allowed us to fluorescently visualize the extensive transfection of neuronal-like cells in substantia nigra and areas surrounding the lateral ventricle. No ORMOSIL-based toxicity was observed 4 weeks after transfection. The efficiency of transfection equaled or exceeded that obtained in studies using a viral vector. An in vivo optical imaging technique (a fiber-based confocal fluorescent imaging system) provided an effective means to show the retention of viability of the transfected cells. The ORMOSIL-mediated transfections also were used to manipulate the biology of the neural stem/progenitor cells in vivo. Transfection of a plasmid expressing the nucleus-targeting fibroblast growth factor receptor type 1 resulted in significant inhibition of the in vivo incorporation of bromodeoxyuridine into the DNA of the cells in the subventricular zone and the adjacent rostral migratory stream. This in vivo approach shows that the nuclear receptor can control the proliferation of the stem/progenitor cells in this region of the brain. The results of this nanomedicine approach using ORMOSIL nanoparticles as a nonviral gene delivery platform have a promising future direction for effective therapeutic manipulation of the neural stem/progenitor cells as well as in vivo targeted brain therapy.
- Published
- 2005
- Full Text
- View/download PDF
21. Control of CREB-binding protein signaling by nuclear fibroblast growth factor receptor-1: a novel mechanism of gene regulation.
- Author
-
Fang X, Stachowiak EK, Dunham-Ems SM, Klejbor I, and Stachowiak MK
- Subjects
- Amino Acid Substitution, CREB-Binding Protein, Cell Differentiation physiology, Cells, Cultured, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells physiology, Humans, Mutagenesis, Site-Directed, Promoter Regions, Genetic, RNA Polymerase II metabolism, Receptor Protein-Tyrosine Kinases genetics, Receptor, Fibroblast Growth Factor, Type 1, Receptors, Fibroblast Growth Factor genetics, Signal Transduction, Transcription, Genetic, Transcriptional Activation, Transfection, Cell Nucleus physiology, Gene Expression Regulation, Nuclear Proteins physiology, Receptor Protein-Tyrosine Kinases physiology, Receptors, Fibroblast Growth Factor physiology, Trans-Activators physiology
- Abstract
In integrative nuclear fibroblast growth factor receptor-1 (FGFR1) signaling a newly synthesized FGFR1 translocates to the nucleus to stimulate cell differentiation and associated gene activities. The present study shows that FGFR1 accumulates and interacts with the transcriptional co-activator CREB-binding protein (CBP) in nuclear speckle domains in the developing brain and in neural progenitor-like cells in vitro, which accompanies differentiation and postmitotic growth. Cell differentiation and gene activation by nuclear FGFR1 do not require tyrosine kinase activity. Instead, FGFR1 stimulates transcription in cooperation with CBP by increasing recruitment of RNA polymerase II and histone acetylation at the active gene promoter. FGFR1 is a multifactorial protein whose N terminus interacts with CBP and C terminus with ribosomal S6 kinase 1 (RSK1). Nuclear FGFR1 augments CBP-mediated transcription by 1) releasing the CBP C-terminal domain from RSK1 inhibition and 2) activating the CBP N-terminal domain. The interaction of FGFR1 with CBP and RSK1 allows activation of gene transcription and may play a role in cell differentiation.
- Published
- 2005
- Full Text
- View/download PDF
22. Voltage-sensitive and ligand-gated channels in differentiating neural stem-like cells derived from the nonhematopoietic fraction of human umbilical cord blood.
- Author
-
Sun W, Buzanska L, Domanska-Janik K, Salvi RJ, and Stachowiak MK
- Subjects
- Antigens, CD34 biosynthesis, Cell Differentiation, Cell Line, Electrophysiology, Excitatory Amino Acid Antagonists metabolism, Fetal Blood cytology, Glycine chemistry, Humans, Immunohistochemistry, Ions, Kainic Acid metabolism, Leukocyte Common Antigens biosynthesis, Ligands, N-Methylaspartate metabolism, Neurotransmitter Agents metabolism, Oligonucleotide Array Sequence Analysis, Patch-Clamp Techniques, Phenotype, Potassium chemistry, Receptors, GABA metabolism, Receptors, Glutamate metabolism, Sodium chemistry, Stem Cells metabolism, Umbilical Veins metabolism, Fetal Blood metabolism, Neurons metabolism, Stem Cells cytology
- Abstract
Fetal cells with the characteristics of neural stem cells (NSCs) can be derived from the nonhematopoietic fraction of human umbilical cord blood (HUCB), expanded as a nonimmortalized cell line (HUCB-NSC), and further differentiated into neuron-like cells (HUCB-NSCD); however, the functional and neuronal properties of these cells are poorly understood. To address this issue, we used whole-cell patch-clamp recordings, gene microarrays, and immunocytochemistry to identify voltage-gated channels and ligand-gated receptors on HUCB-NSCs and HUCB-NSCDs. Gene microarray analysis identified genes for voltage-dependent potassium and sodium channels and the neurotransmitter receptors acetylcholine (ACh), gamma-aminobutyric acid (GABA), glutamate, glycine, 5-hydroxytryptamine (5-HT), and dopamine (DA). Several of these genes (GABA-A, glycine and glutamate receptors, voltage-gated potassium channels, and voltage-gated sodium type XII alpha channels) were not expressed in the HUCB mono-nuclear fraction (HUCB-MC), which served as a starting cell population for HUCB-NSC. HUCB-NSCD acquired neuronal phenotypes and displayed an inward rectifying potassium current (Kir) and an outward rectifying potassium current (I(K+)). Kir was present on most HUCB-NSCs and HUCB-NSCDs, whereas I(K+) was present only on HUCB-NSCDs. Many HUCB-NSCDs were immunopositive for glutamate, glycine, nicotinic ACh, DA, 5-HT, and GABA receptors. Kainic acid (KA), a non-N-methyl-D-asparate (NMDA) glutamate-receptor agonist, induced an inward current in some HUCB-NSCDs. KA, glycine, DA, ACh, GABA, and 5-HT partially blocked Kir through their respective receptors. These results suggest that HUCB-NSCs differentiate toward neuron-like cells, with functional voltage- and ligand-gated channels identified in other neuronal systems.
- Published
- 2005
- Full Text
- View/download PDF
23. Assessment of viral and non-viral gene transfer into adult rat brains using HSV-1, calcium phosphate and PEI-based methods.
- Author
-
Corso TD, Torres G, Goulah C, Roy I, Gambino AS, Nayda J, Buckley T, Stachowiak EK, Bergey EJ, Pudavar H, Dutta P, Bloom DC, Bowers WJ, and Stachowiak MK
- Subjects
- Animals, Animals, Newborn, Brain metabolism, Brain pathology, Brain virology, Cells, Cultured, Cricetinae, Mice, NIH 3T3 Cells, Nanotechnology, Rats, Receptor, Fibroblast Growth Factor, Type 1, Substantia Nigra virology, Transduction, Genetic methods, Calcium Phosphates chemistry, Genetic Vectors, Herpesvirus 1, Human genetics, Parkinson Disease etiology, Polyethyleneimine chemistry, Receptor Protein-Tyrosine Kinases chemistry, Receptor Protein-Tyrosine Kinases genetics, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Fibroblast Growth Factor chemistry, Receptors, Fibroblast Growth Factor genetics, Receptors, Fibroblast Growth Factor metabolism, Substantia Nigra metabolism
- Abstract
CNS gene transfer could provide new approaches to the modelling of neurodegenerative diseases and devising potential therapies. One such disorder is Parkinson's disease (PD), in which dysfunction of several different metabolic processes has been implicated. Here we review the literature on gene transfer systems based on herpes simplex virus type 1 (HSV-1) and non-viral polyethyleneimine (PEI) and calcium phosphate nanoparticle methods. We also assess the usefulness of various CNS gene delivery methods and present some of our own data to exemplify such usefulness. Our data result from vectors stereotaxically introduced to the substantia nigra (SN) of adult rats and evaluated 1 week and/or 1 month post injection using histochemical methods to assess recombinant ss-galactosidase enzyme activity. Gene transfer using PEI or calcium phosphate-mediated transfections was observed for both methods and PEI was comparable to that of HSV-1 amplicon. Our data show that the amplicon delivery was markedly increased when packaged with a helper virus and was similar to the expression profile achieved with a full-size replication-defective HSV-1 recombinant (8117/43). We also examine whether PEI or HSV-1 amplicon-mediated gene transfer could facilitate assessment of the biological effects induced by a dominant negative FGF receptor-1 mutant to model the reduced FGF signalling thought to occur in Parkinson's disease.
- Published
- 2005
24. 90-kDa ribosomal S6 kinase is a direct target for the nuclear fibroblast growth factor receptor 1 (FGFR1): role in FGFR1 signaling.
- Author
-
Hu Y, Fang X, Dunham SM, Prada C, Stachowiak EK, and Stachowiak MK
- Subjects
- Animals, Brain metabolism, Cells, Cultured, Cyclic AMP metabolism, Cyclic AMP Response Element-Binding Protein metabolism, Humans, Neurons cytology, Neurons metabolism, Protein Binding, Protein Structure, Tertiary, Rats, Rats, Sprague-Dawley, Receptor Protein-Tyrosine Kinases genetics, Receptor, Fibroblast Growth Factor, Type 1, Receptors, Fibroblast Growth Factor genetics, Ribosomal Protein S6 Kinases, 90-kDa genetics, Subcellular Fractions metabolism, Two-Hybrid System Techniques, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Fibroblast Growth Factor metabolism, Ribosomal Protein S6 Kinases, 90-kDa metabolism
- Abstract
Fibroblast growth factor receptor 1 (FGFR1) is a transmembrane protein capable of transducing stimulation by secreted FGFs. In addition, newly synthesized FGFR1 enters the nucleus in response to cellular stimulation and during development. Nuclear FGFR1 can transactivate CRE (cAMP responsive element), activate CRE-binding protein (CREB)-binding protein (CBP) and gene activities causing cellular growth and differentiation. Here, a yeast two-hybrid assay was performed to identify FGFR1-binding proteins and the mechanism of nuclear FGFR1 action. Ten FGFR1-binding proteins were identified. Among the proteins detected with the intracellular FGFR1 domain was a 90-kDa ribosomal S6 kinase (RSK1), a regulator of CREB, CBP, and histone phosphorylation. FGFR1 bound to the N-terminal region of RSK1. The FGFR1-RSK1 interaction was confirmed by co-immunoprecipitation and colocalization in the nucleus and cytoplasm of mammalian cells. Predominantly nuclear FGFR1-RSK1 interaction was observed in the rat brain during neurogenesis and in cAMP-stimulated cultured neural cells. In TE671 cells, transfected FGFR1 colocalized and coimmunoprecipitated, almost exclusively, with nuclear RSK1. Nuclear RSK1 kinase activity and RSK1 activation of CREB were enhanced by transfected FGFR1. In contrast, kinase-deleted FGFR1 (TK-), which did not bind to RSK1 failed to stimulate nuclear RSK1 activity or RSK1 activation of CREB. Kinase inactive FGFR1 (K514A) bound effectively to nuclear RSK1, but it failed to stimulate RSK1. Thus, active FGFR1 kinase regulates the functions of nuclear RSK1. The interaction of nuclear FGFR1 with pluripotent RSK1 offers a new mechanism through which FGFR1 may control fundamental cellular processes.
- Published
- 2004
- Full Text
- View/download PDF
25. Novel nuclear signaling pathway mediates activation of fibroblast growth factor-2 gene by type 1 and type 2 angiotensin II receptors.
- Author
-
Peng H, Moffett J, Myers J, Fang X, Stachowiak EK, Maher P, Kratz E, Hines J, Fluharty SJ, Mizukoshi E, Bloom DC, and Stachowiak MK
- Subjects
- Adrenal Medulla cytology, Adrenal Medulla metabolism, Angiotensin II analogs & derivatives, Angiotensin II pharmacology, Animals, Binding Sites, Cattle, Cell Nucleus genetics, Cells, Cultured, DNA Footprinting, Deoxyribonuclease I metabolism, Fibroblast Growth Factor 2 metabolism, Gene Expression Regulation drug effects, Imidazoles pharmacology, Promoter Regions, Genetic, Pyridines pharmacology, Receptor, Angiotensin, Type 1, Receptor, Angiotensin, Type 2, Recombinant Proteins genetics, Recombinant Proteins metabolism, Response Elements physiology, Trans-Activators metabolism, Tyrosine metabolism, Cell Nucleus metabolism, Fibroblast Growth Factor 2 genetics, Receptors, Angiotensin metabolism, Signal Transduction
- Abstract
In bovine adrenal medullary cells synergistically acting type 1 and type 2 angiotensin II (AII) receptors activate the fibroblast growth factor-2 (FGF-2) gene through a unique AII-responsive promoter element. Both the type 1 and type 2 AII receptors and the downstream cyclic adenosine 1',3'-monophosphate- and protein kinase C-dependent signaling pathways activate the FGF-2 promoter through a novel signal-transducing mechanism. This mechanism, which we have named integrative nuclear FGF receptor-1 signaling, involves the nuclear translocation of FGF receptor-1 and its subsequent transactivation of the AII-responsive element in the FGF-2 promoter.
- Published
- 2001
- Full Text
- View/download PDF
26. Polyethyleneimine-mediated transfection of cultured postmitotic neurons from rat sympathetic ganglia and adult human retina.
- Author
-
Horbinski C, Stachowiak MK, Higgins D, and Finnegan SG
- Subjects
- Animals, Animals, Newborn, Cells, Cultured, DNA metabolism, Dose-Response Relationship, Drug, Ganglia, Sympathetic cytology, Gene Expression, Humans, Luminescent Proteins biosynthesis, Luminescent Proteins genetics, Mitosis, Neurons cytology, Neurons drug effects, Plasmids genetics, Polyethyleneimine pharmacology, Rats, Retina cytology, beta-Galactosidase biosynthesis, beta-Galactosidase genetics, Ganglia, Sympathetic metabolism, Neurons metabolism, Polyethyleneimine metabolism, Retina metabolism, Transfection methods
- Abstract
Background: Chemical methods of transfection that have proven successful with cell lines often do not work with primary cultures of neurons. Recent data, however, suggest that linear polymers of the cation polyethyleneimine (PEI) can facilitate the uptake of nucleic acids by neurons. Consequently, we examined the ability of a commercial PEI preparation to allow the introduction of foreign genes into postmitotic mammalian neurons. Sympathetic neurons were obtained from perinatal rat pups and maintained for 5 days in vitro in the absence of nonneuronal cells. Cultures were then transfected with varying amounts of a plasmid encoding either E. coli beta-galactosidase or enhanced green fluorescence protein (EGFP) using PEI., Results: Optimal transfection efficiency was observed with 1 microg/ml of plasmid DNA and 5 microg/ml PEI. Expression of beta-galactosidase was both rapid and stable, beginning within 6 hours and lasting for at least 21 days. A maximum yield was obtained within 72 hours with approximately 9% of the neurons expressing beta-galactosidase, as assessed by both histochemistry and antibody staining. Cotransfection of two plasmids encoding reporter genes was achieved. Postmitotic neurons from adult human retinal cultures also demonstrated an ability to take up and express foreign DNA using PEI as a vector., Conclusions: These data suggest that PEI is a useful agent for the stable expression of plasmid-encoded genes in neuronal cultures.
- Published
- 2001
- Full Text
- View/download PDF
27. Gene transfer into the central nervous system using herpes simplex virus-1 vectors.
- Author
-
Tabbaa S, Goulah C, Tran RK, Lis A, Korody R, Stachowski B, Horowitz JM, Torres G, Stachowiak EK, Bloom DC, and Stachowiak MK
- Subjects
- Animals, Brain cytology, Genetic Therapy methods, Genetic Vectors, Male, Rats, Rats, Inbred F344, Stereotaxic Techniques, beta-Galactosidase analysis, Brain metabolism, Gene Transfer Techniques, Herpesvirus 1, Human, beta-Galactosidase genetics
- Abstract
Manipulation of gene expression in developing or in mature central nervous systems (CNS) holds a promise for the resolution of many compelling neurobiological questions, including the feasibility of gene therapy to treat diseases of the brain. In this context, a number of viral vectors have been used in recent years to introduce and express genes into the CNS. This article discusses a gene transfer system based on the Herpes Simplex Virus-1 (HSV-1). We describe here the use of non-replicating, non-toxic HSV-1 vector, 8117/43, in a series of studies carried in our joint program. This vector proves further the utility of HSV-1 as a delivery vehicle to a number of distinct sites within the CNS.
- Published
- 2000
28. Transcriptional regulation of fibroblast growth factor-2 expression in human astrocytes: implications for cell plasticity.
- Author
-
Moffett J, Kratz E, Myers J, Stachowiak EK, Florkiewicz RZ, and Stachowiak MK
- Subjects
- Astrocytes cytology, Astrocytes physiology, Base Sequence, Binding Sites, Brain cytology, Brain metabolism, Cell Nucleus metabolism, Cells, Cultured, Colforsin pharmacology, DNA-Binding Proteins metabolism, Epidermal Growth Factor pharmacology, Fibroblast Growth Factor 2 biosynthesis, Fibroblast Growth Factor 2 pharmacology, Humans, Interleukin-1 pharmacology, Luciferases biosynthesis, Mitosis, Molecular Sequence Data, Oligodeoxyribonucleotides pharmacology, Oligonucleotides, Antisense pharmacology, Polymerase Chain Reaction, Recombinant Fusion Proteins biosynthesis, Tetradecanoylphorbol Acetate pharmacology, Transfection, Astrocytes metabolism, Fibroblast Growth Factor 2 genetics, Gene Expression Regulation drug effects, Neuronal Plasticity physiology, Promoter Regions, Genetic, Transcription, Genetic drug effects
- Abstract
Induction of the fibroblast growth factor-2 (FGF-2) gene and the consequent accumulation of FGF-2 in the nucleus are operative events in mitotic activation and hypertrophy of human astrocytes. In the brain, these events are associated with cellular degeneration and may reflect release of the FGF-2 gene from cell contact inhibition. We used cultures of human astrocytes to examine whether expression of FGF-2 is also controlled by soluble growth factors. Treatment of subconfluent astrocytes with interleukin-1beta, epidermal or platelet-derived growth factors, 18-kDa FGF-2, or serum or direct stimulation of protein kinase C (PKC) with phorbol 12-myristate 13-acetate or adenylate cyclase with forskolin increased the levels of 18-, 22-, and 24-kDa FGF-2 isoforms and FGF-2 mRNA. Transfection of FGF-2 promoter-luciferase constructs identified a unique -555/-513 bp growth factor-responsive element (GFRE) that confers high basal promoter activity and activation by growth factors to a downstream promoter region. It also identified a separate region (-624/-556 bp) essential for PKC and cAMP stimulation. DNA-protein binding assays indicated that novel cis-acting elements and trans-acting factors mediate activation of the FGF-2 gene. Southwestern analysis identified 40-, 50-, 60-, and 100-kDa GFRE-binding proteins and 165-, 112-, and 90-kDa proteins that interacted with the PKC/cAMP-responsive region. The GFRE and the element essential for PKC and cAMP stimulation overlap with the region that mediates cell contact inhibition of the FGF-2 promoter. The results show a two-stage regulation of the FGF-2 gene: 1) an initial induction by reduced cell contact, and 2) further activation by growth factors or the PKC-signaling pathway. The hierarchic regulation of the FGF-2 gene promoter by cell density and growth factors or PKC reflects a two-stage activation of protein binding to the GFRE and to the PKC/cAMP-responsive region, respectively.
- Published
- 1998
- Full Text
- View/download PDF
29. Cruciform-extruding regulatory element controls cell-specific activity of the tyrosine hydroxylase gene promoter.
- Author
-
Kim EL, Peng H, Esparza FM, Maltchenko SZ, and Stachowiak MK
- Subjects
- Animals, Base Sequence, Cattle, Cell Line, Cells, Cultured, DNA, Superhelical drug effects, DNA, Superhelical metabolism, Gene Expression Regulation, Enzymologic drug effects, Humans, Luciferases biosynthesis, Molecular Sequence Data, Mutagenesis, Site-Directed, Neuroblastoma, Organ Specificity, Osmium Tetroxide pharmacology, Recombinant Fusion Proteins biosynthesis, Recombinant Proteins biosynthesis, Sequence Deletion, Transfection, Tumor Cells, Cultured, Adrenal Medulla enzymology, Chromaffin Cells enzymology, DNA, Superhelical chemistry, Nucleic Acid Conformation, Promoter Regions, Genetic, Regulatory Sequences, Nucleic Acid, Tyrosine 3-Monooxygenase biosynthesis, Tyrosine 3-Monooxygenase genetics
- Abstract
Tyrosine hydroxylase (TH) is expressed specifically in catecholaminergic cells. We have identified a novel regulatory sequence in the upstream region of the bovine TH gene promoter formed by a dyad symmetry element (DSE1;-352/-307 bp). DSE1 supports TH promoter activity in TH-expressing bovine adrenal medulla chromaffin (BAMC) cells and inhibits promoter activity in non-expressing TE671 cells. DNase I footprinting of relaxed TH promoter DNA showed weak binding of nuclear BAMC cell proteins to a short sequence in the right DSE1 arm. In BAMC cells, deletion of the right arm markedly reduced the expression of luciferase from the TH promoter. However, deletion of the left DSE1 arm or its reversed orientation (RevL) also inactivated the TH promoter. In supercoiled TH promoter, DSE1 assumes a cruciform-like conformation i.e., it binds cruciform-specific 2D3 antibody, and S1 nuclease-cleavage and OsO4-modification assays have identified an imperfect cruciform extruded by the DSE1. DNase I footprinting of supercoiled plasmid showed that cruciformed DSE1 is targeted by nuclear proteins more efficiently than the linear duplex isomer and that the protected site encompasses the left arm and center of DSE1. Our results suggest that the disruption of intrastrand base-pairing preventing cruciform formation and protein binding to DSE1 is responsible for its inactivation in DSE1 mutants. DSE1 cruciform may act as a target site for activator (BAMC cells) and repressor (TE671) proteins. Its extrusion emerges as a novel mechanism that controls cell-specific promoter activity.
- Published
- 1998
- Full Text
- View/download PDF
30. Nuclear accumulation of fibroblast growth factor receptors in human glial cells--association with cell proliferation.
- Author
-
Stachowiak EK, Maher PA, Tucholski J, Mordechai E, Joy A, Moffett J, Coons S, and Stachowiak MK
- Subjects
- Astrocytes chemistry, Astrocytes metabolism, Blotting, Western, Cell Division physiology, Cell Nucleus chemistry, Cells, Cultured, Fibroblast Growth Factor 2 metabolism, Glioma chemistry, Humans, Immunohistochemistry, Iodine Radioisotopes, Mitogens physiology, Receptor, Fibroblast Growth Factor, Type 1, Receptors, Cytoplasmic and Nuclear chemistry, Receptors, Cytoplasmic and Nuclear metabolism, Receptors, Fibroblast Growth Factor chemistry, Receptors, Fibroblast Growth Factor immunology, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Subcellular Fractions, Transfection, Cell Nucleus metabolism, Glioma metabolism, Neuroglia cytology, Neuroglia metabolism, Receptor Protein-Tyrosine Kinases, Receptors, Fibroblast Growth Factor metabolism
- Abstract
In this study we describe the presence of high affinity FGF-2 binding sites in the nuclei of U251MG glioma cells (K(d)=7 pM). Immunoprecipitation of total cell extracts with FGF receptor (FGFR) 1-4 antibodies showed that U251MG glioma cells express only FGFR1. [125I]FGF-2 cross linking to nuclear extracts followed by FGFR1 immunoprecipitation showed that FGFR1 may account for the nuclear FGF-2 binding sites. Western blot analysis demonstrated the presence of 103, 118 kDa and small amounts of 145 kDa FGFR1 isoforms in the nuclei of glioma cells. All isoforms contain both the C- and N-terminal domains. Nuclear FGFR1 retains kinase activity. Immunocytochemistry using confocal microscopy showed specific FGFR1 immunoreactivity within the nuclear interior. In continuously proliferating glioma cells, nuclear FGFR1 is constitutively expressed, independent of cell density. In contrast, in nontransformed human astrocytes, nuclear FGFR1 levels fluctuate with the proliferative state of the cell. In quiescent, confluent astrocytes nuclear FGFR1 protein was depleted. An accumulation of nuclear FGFR1 was observed following the transition to a subconfluent, proliferating state. Transfection of a pcDNA3.1-FGFR1 expression vector into glioma cells that do not express FGFR1 resulted in the nuclear accumulation of FGFR1, increased cell proliferation, and stimulated transition from the G0/G1 to the S-phase of the cell cycle. The increased proliferative rate was resistant to inhibition by the cell-impermeable FGF binding antagonist, myoinositol hexakis [dihydrogen phosphate]. Our results suggest that the constitutive nuclear presence of FGFR1 contributes to the increased proliferation of glioma cells while the transient nuclear accumulation of FGFR1 in normal astrocytes may play a role in the transition to a reactive state.
- Published
- 1997
- Full Text
- View/download PDF
31. Nuclear accumulation of FGF-2 is associated with proliferation of human astrocytes and glioma cells.
- Author
-
Joy A, Moffett J, Neary K, Mordechai E, Stachowiak EK, Coons S, Rankin-Shapiro J, Florkiewicz RZ, and Stachowiak MK
- Subjects
- Astrocytes metabolism, Cell Communication, Cell Count, Cell Division, Cells, Cultured, Female, Glioblastoma metabolism, Humans, Male, Neuroglia cytology, Neuroglia metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptor, Fibroblast Growth Factor, Type 2, Receptors, Fibroblast Growth Factor metabolism, Astrocytes cytology, Cell Nucleus metabolism, Fibroblast Growth Factor 2 metabolism, Glioblastoma pathology
- Abstract
FGF-2 has been implicated in the neoplastic transformation of glioma cells and in the transition of normal quiescent astrocytes to a proliferating, reactive state. In the present study we have observed that in human glial cells, levels and subcellular localization of FGF-2 are different in quiescent and proliferating cells. FGF-2 was detected in the cytoplasm of non-reactive astrocytes in human brain sections. In contrast FGF-2 was located within the cytoplasm and nuclei of reactive astrocytes in gliotic brain tissue and in neoplastic cells of glioma tumors. In vitro, FGF-2 was found predominantly in the nucleus of subconfluent proliferating astrocytes, but was detected only in the cytoplasm of density arrested quiescent astrocytes. Our results suggest that reduced cell contact stimulates nuclear accumulation of FGF-2, accompanying mitotic activation of reactive human astrocytes. FGF-2 was constitutively localized to the nucleus of continuously proliferating glioma cells independent of cell density. A role for intracellular FGF-2 was further suggested by the observation that glioma cells that are not stimulated to proliferate by extracellular FGF-2 proliferated faster when transfected with FGF-2 expressing vectors. This increased proliferation correlated with nuclear accumulation of FGF-2. Cell proliferation was attenuated by 5'-deoxy-5'-methylthioadenosine, a FGF-2 receptor tyrosine kinase inhibitor that acts within the cell, but was unaffected by myo-inositol hexakis [dihydrogen phosphate] that disrupts FGF-2 binding to plasma membrane receptors. Our results indicate that FGF-2 serves as a nuclear regulator of proliferation in astrocytic cells. In glioma cells, the constitutive presence of FGF-2 in the nucleus may promote proliferation that is insensitive to cell contact inhibition.
- Published
- 1997
- Full Text
- View/download PDF
32. Nuclear accumulation of fibroblast growth factor receptors is regulated by multiple signals in adrenal medullary cells.
- Author
-
Stachowiak MK, Maher PA, Joy A, Mordechai E, and Stachowiak EK
- Subjects
- Adrenal Medulla cytology, Animals, Cattle, Cell Nucleus metabolism, Cells, Cultured, Kinetics, Microscopy, Immunoelectron, Protein-Tyrosine Kinases metabolism, Signal Transduction, Subcellular Fractions metabolism, Adrenal Medulla metabolism, Receptors, Fibroblast Growth Factor metabolism
- Abstract
In an effort to determine the localization of fibroblast growth factor (FGF) receptors (FGFR) that could mediate the intracellular action of FGF-2, we discovered the presence of high-affinity. FGF-2 binding sites in the nuclei of bovine adrenal medullary cells (BAMC). Western blot analysis demonstrated the presence of 103-, 118-, and 145-kDa forms of FGFR1 in nuclei isolated from BAMC. 125I-FGF-2 cross-linking to nuclear extracts followed by FGFR1 immunoprecipitation showed that FGFR1 can account for the nuclear FGF-2 binding sites. Nuclear FGFR1 has kinase activity and undergoes autophosphorylation. Immunocytochemistry with the use of confocal and electron microscopes demonstrated the presence of FGFR1 within the nuclear interior. Nuclear subfractionation followed by Western blot or immunoelectron microscopic analysis showed that the nuclear FGFR1 is contained in the nuclear matrix and the nucleoplasm. Agents that induce translocation of endogenous FGF-2 to the nucleus (forskolin, carbachol, or angiotensin II) increased the intranuclear accumulation of FGFR1. This accumulation was accompanied by an overall increase in FGF-2-inducible tyrosine kinase activity. Our findings suggest a novel mode for growth factor action whereby growth factor receptors translocate to the nucleus in parallel with their ligand and act as direct mediators of nuclear responses to cell stimulation.
- Published
- 1996
- Full Text
- View/download PDF
33. Promoter regions involved in density-dependent regulation of basic fibroblast growth factor gene expression in human astrocytic cells.
- Author
-
Moffett J, Kratz E, Florkiewicz R, and Stachowiak MK
- Subjects
- Cells, Cultured, Child, DNA-Binding Proteins metabolism, Gene Expression Regulation, Neoplastic, Glioma pathology, Histones genetics, Humans, Male, Nuclear Proteins metabolism, RNA, Messenger genetics, Transcriptional Activation, Astrocytes physiology, Cell Count, Fibroblast Growth Factor 2 genetics, Promoter Regions, Genetic
- Abstract
Expression of mitogenic basic fibroblast growth factor (bFGF) in the central nervous system is inhibited by direct cell contact and is implicated in reactive and neoplastic transformation of astrocytes. The molecular mechanisms controlling expression of bFGF were examined in cultures of human astrocytes. Cell-density-dependent depletion of bFGF mRNA levels parallels changes in bFGF gene protein. Regulation of transcription of a bFGF luciferase reporter gene containing an upstream region (bp -1800 to +314) of the bFGF gene promoter mimicks the density-dependent regulation of the endogenous bFGF gene in transfected astrocytes. Deletion analysis has identified a fragment (bp -650 to -513) and sequences further downstream (bp -274 to +314) as the regions required for the regulation of bFGF gene activity by cell density. Unlike in astrocytes, changing the cell density of glioma cell cultures does not affect the levels of bFGF protein and mRNA. bFGF luciferase constructs were expressed at the same level in high- or low-density cultures of glioma cells, indicating altered regulation of the bFGF gene promoter. Electrophoretic mobility shift assays showed binding of nuclear proteins to a fragment of bFGF gene promoter from bp -650 to -453. This binding was abolished by a deletion of the upstream cell-density-responsive region (bp -650 to -512). Binding was observed with nuclear extracts from subconfluent astrocytes but was reduced in extracts from confluent astrocytes. Our results indicate that induction of bFGF in astrocytes upon reduction of cell density is mediated transcriptionally by positive trans-acting factors interacting with bFGF promoter. In contrast, nuclear proteins from glioma cells bind to the promoter region from bp -650 to -453 independent of cell density. Thus, the constitutive binding of trans-acting factor(s) to the region of the bFGF promoter from bp -650 to -453 may be responsible for the continuous expression of bFGF that leads to the uncontrolled growth of glioma cells.
- Published
- 1996
- Full Text
- View/download PDF
34. Regulation of bFGF gene expression and subcellular distribution of bFGF protein in adrenal medullary cells.
- Author
-
Stachowiak MK, Moffett J, Joy A, Puchacz E, Florkiewicz R, and Stachowiak EK
- Subjects
- Adrenal Medulla cytology, Animals, Base Sequence, Cattle, Cell Nucleus chemistry, Cells, Cultured, Colforsin pharmacology, Cyclic AMP pharmacology, Cytoplasm chemistry, Fibroblast Growth Factor 2 biosynthesis, Humans, Models, Biological, Molecular Sequence Data, Promoter Regions, Genetic genetics, Protein Kinase C pharmacology, Receptors, Angiotensin physiology, Receptors, Cholinergic physiology, Signal Transduction physiology, Tetradecanoylphorbol Acetate pharmacology, Transcriptional Activation, Adrenal Medulla metabolism, Fibroblast Growth Factor 2 analysis, Fibroblast Growth Factor 2 genetics, Gene Expression Regulation drug effects
- Abstract
Basic fibroblast growth factor (bFGF), a potent mitogenic/neurotrophic factor, controls the development and plasticity of many types of neural cells. In adrenal chromaffin cells, the appearance of bFGF protein coincided with the establishment of functional innervation, suggesting induction by trans-synaptic signals. In cultured bovine adrenal medullary cells Western blot analysis revealed 18-, 23-, and 24-kD bFGF isoforms in the cytosolic and nuclear fractions. Stimulation of acetylcholine nicotinic receptors or hormonal angiotensin II receptors or the direct stimulation of adenylate cyclase with forskolin or protein kinase C (PKC) with PMA increased the content of all bFGF isoforms. Increases in the levels of intracellular bFGF did not result in detectable presence of bFGF proteins in culture medium. Instead, bFGF proteins accumulated in the cytoplasm or the nucleus depending on whether PKC or cAMP pathways were activated. The long-term nuclear forskolin-induced accumulation of bFGF was prevented by cycloheximide or by antisense bFGF oligonucleotide and was also accompanied by an increase in bFGF mRNA. We used luciferase reporter plasmids containing the human bFGF promoter to show that the induction of bFGF resulted from transcriptional activation of the bFGF gene and was mediated by regulatory sequences located upstream from its transcription start site. Stimulation of bFGF gene expression by forskolin and PMA was synergistic and was mediated through different promoter regions. The results suggest that stimulation by cAMP and PKC is mediated through novel cis elements. The regulation of bFGF protein content also involves posttranscriptional mechanisms since changes in the levels of individual bFGF isoforms were different depending on whether cells were treated with carbachol or angiotensin II, forskolin, or PMA. The present study indicates that bFGF is an intracrine cytoplasmic-nuclear factor, whose expression is regulated by trans-synaptic and hormonal stimuli and which may act as a direct mediator of genomic responses to afferent stimulation.
- Published
- 1994
- Full Text
- View/download PDF
35. Short and long term regulation of catecholamine biosynthetic enzymes by angiotensin in cultured adrenal medullary cells. Molecular mechanisms and nature of second messenger systems.
- Author
-
Stachowiak MK, Jiang HK, Poisner AM, Tuominen RK, and Hong JS
- Subjects
- Adrenal Medulla drug effects, Angiotensin II pharmacology, Animals, Calcimycin pharmacology, Calcium physiology, Calcium Channels physiology, Calmodulin antagonists & inhibitors, Calmodulin physiology, Cattle, Cells, Cultured, Colforsin pharmacology, Enzyme Activation drug effects, Gene Expression drug effects, Kinetics, Membrane Potentials drug effects, Phenylethanolamine N-Methyltransferase genetics, Protein Kinase C metabolism, RNA, Messenger biosynthesis, Saralasin pharmacology, Tyrosine 3-Monooxygenase genetics, Veratridine pharmacology, Adrenal Medulla enzymology, Angiotensin II analogs & derivatives, Catecholamines biosynthesis, Phenylethanolamine N-Methyltransferase metabolism, Second Messenger Systems, Tyrosine 3-Monooxygenase metabolism
- Abstract
The purpose of this study was to examine the effects of angiotensin on the enzyme activities and gene expression of two catecholamine synthesizing enzymes, tyrosine hydroxylase (TH) and phenylethanolamine N-methyltransferase (PNMT), in bovine adrenal medullary (AM) cells. Short term (15 min) incubation of cultured AM cells with 2 nM [Sar1]angiotensin II (s1-AII) did not increase basal secretion of catecholamines; however, longer incubations (3, 24, or 72 h) produced 4-10-fold increases. To determine whether angiotensin affects synthesis of catecholamines, the activities of TH and PNMT were examined. Incubation with s1-AII (15-30 min) decreased the Km of TH for its biopterine cofactor [6R)-5,6,7,8-tetrahydro-1-biopterin dihydrochloride (BH4] without affecting the Vmax, suggesting activation of TH. After long term incubation (72 h) the Km value was identical to that of control, while increases in the apparent Vmax were observed. PNMT activity was unaffected during a 30-min treatment with s1-AII; however, 2-fold increases occurred after a 48-72-h incubation. s1-AII (24 h) increased the relative abundance of TH and PNMT mRNAs, suggesting that the long term increase in enzyme activities reflected increased expression of TH and PNMT genes. Maximal increases were observed at 2 nM s1-AII and the changes were antagonized by saralasin. Induction of TH mRNA by s1-AII was additive to the effects of veratridine or forskolin indicating that effects of angiotensin were not due to membrane depolarization or increased cyclic AMP levels. Incubation with Ca2+ ionophore A23187 increased TH and PNMT mRNA levels in AM cells raising the possibility that the increase in cellular [Ca2+] could mediate effects of angiotensin. Angiotensin-induced increases in TH and PNMT mRNA were inhibited by nifedipine indicating involvement of voltage-dependent Ca2+ channels. In addition, the increases in TH, but not PNMT mRNA, were antagonized by dantrolene, which inhibits mobilization of Ca2+ from intracellular stores. Calmodulin involvement was suggested by the inhibition of s1-AII induced changes in mRNA with 1 microM calmidazolium.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1990
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.