Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Sci Rep ; 13(1): 19529, 2023 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-37945646

RESUMEN

Multiple sclerosis (MS), a chronic neurodegenerative disease driven by damage to the protective myelin sheath, is currently incurable. Today, all clinically available treatments modulate the immune-mediated symptoms of the disease but they fail to stop neurodegeneration in many patients. Remyelination, the regenerative process of myelin repair by oligodendrocytes, which is considered a necessary step to protect demyelinated axons and stop neuronal death, is impaired in MS patients. One of the major obstacles to finding effective remyelinating drugs is the lack of biomimetic drug screening platforms that enable quantification of compounds' potential to stimulate 3D myelination in the physiologically relevant axon-like environment. To address this need, we built a unique myelination drug discovery platform, by expanding our previously developed technology, artificial axons (AAs), which enables 3D-printing of synthetic axon mimics with the geometry and mechanical properties closely resembling those of biological axons. This platform allows for high-throughput phenotypic myelination assay based on quantification of 3D wrapping of myelin membrane around axons in response to compounds. Here, we demonstrate quantification of 3D myelin wrapping by rat oligodendrocytes around the axon mimics in response to a small library of known pro-myelinating compounds. This assay shows pro-myelinating activity for all tested compounds consistent with the published in vitro and in vivo data, demonstrating predictive power of AA platform. We find that stimulation of myelin wrapping by these compounds is dose-dependent, providing a facile means to quantify the compounds' potency and efficacy in promoting myelin wrapping. Further, the ranking of relative efficacy among these compounds differs in this 3D axon-like environment as compared to a traditional oligodendrocyte 2D differentiation assay quantifying area of deposited myelin membrane. Together, we demonstrate that the artificial axons platform and associated phenotypic myelin wrapping assay afford direct evaluation of myelin wrapping by oligodendrocytes in response to soluble compounds in an axon-like environment, providing a predictive tool for the discovery of remyelinating therapies.


Asunto(s)
Esclerosis Múltiple , Enfermedades Neurodegenerativas , Humanos , Ratas , Animales , Biomimética , Axones/fisiología , Vaina de Mielina/fisiología , Oligodendroglía/fisiología , Esclerosis Múltiple/tratamiento farmacológico
2.
iScience ; 26(3): 106156, 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36852281

RESUMEN

Promoting myelination capacity of endogenous oligodendrocyte precursor cells (OPCs) is a promising therapeutic approach for CNS demyelinating disorders such as Multiple Sclerosis (MS). To aid in the discovery of myelination-promoting compounds, we generated a genome-engineered human pluripotent stem cell (hPSC) line that consists of three reporters: identification-and-purification tag, GFP, and secreted-NanoLuc, driven by the endogenous PDGFRA, PLP1, and MBP genes, respectively. Using this cell line, we established a high-throughput drug screening platform and performed a small-molecule screen, which identified at least two myelination-promoting small-molecule (Ro1138452 and SR2211) that target prostacyclin (IP) receptor and retinoic acid receptor-related orphan receptor γ (RORγ), respectively. Single-cell-transcriptomic analysis of differentiating OPCs treated with these molecules further confirmed that they promote oligodendrocyte differentiation and revealed several pathways that are potentially modulated by them. The molecules and their target pathways provide promising targets for the possible development of remyelination-based therapy for MS and other demyelinating disorders.

3.
Glia ; 69(2): 413-423, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32902916

RESUMEN

Myelination of axons in the central nervous system (CNS) is a concerted effort between many cell types, resulting in significant cross-talk and communication among cells. Adhesion G protein-coupled receptor ADGRG1 (GPR56) is expressed in all major glial cells and regulates a wide variety of physiological processes by mediating cell-cell and cell-matrix communications. Previous literature has demonstrated the requirement of ADGRG1 in oligodendrocyte precursor cells (OPCs) during developmental myelination. However, it is unknown if ADGRG1 is responsible for myelin formation in a cell-type-specific manner. To that end, here we profiled myelin status in response to deletion of Adgrg1 specifically in OPCs, microglia, astrocytes, and neurons. Interestingly, we find that knocking out Adgrg1 in OPCs significantly decreases OPC proliferation and reduced number of myelinated axons. However, deleting Adgrg1 in microglia, astrocytes, and neurons does not impact developmental myelination. These data support an autonomous functional role for Adgrg1 in OPCs related to myelination.


Asunto(s)
Sistema Nervioso Central , Animales , Ratones , Ratones Noqueados , Vaina de Mielina , Oligodendroglía , Receptores Acoplados a Proteínas G/genética
4.
EMBO J ; 39(16): e104136, 2020 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-32452062

RESUMEN

Developmental synaptic remodeling is important for the formation of precise neural circuitry, and its disruption has been linked to neurodevelopmental disorders such as autism and schizophrenia. Microglia prune synapses, but integration of this synapse pruning with overlapping and concurrent neurodevelopmental processes, remains elusive. Adhesion G protein-coupled receptor ADGRG1/GPR56 controls multiple aspects of brain development in a cell type-specific manner: In neural progenitor cells, GPR56 regulates cortical lamination, whereas in oligodendrocyte progenitor cells, GPR56 controls developmental myelination and myelin repair. Here, we show that microglial GPR56 maintains appropriate synaptic numbers in several brain regions in a time- and circuit-dependent fashion. Phosphatidylserine (PS) on presynaptic elements binds GPR56 in a domain-specific manner, and microglia-specific deletion of Gpr56 leads to increased synapses as a result of reduced microglial engulfment of PS+ presynaptic inputs. Remarkably, a particular alternatively spliced isoform of GPR56 is selectively required for microglia-mediated synaptic pruning. Our present data provide a ligand- and isoform-specific mechanism underlying microglial GPR56-mediated synapse pruning in the context of complex neurodevelopmental processes.


Asunto(s)
Empalme Alternativo , Microglía/metabolismo , Fosfatidilserinas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sinapsis/metabolismo , Animales , Ratones , Ratones Transgénicos , Microglía/citología , Fosfatidilserinas/genética , Unión Proteica , Isoformas de Proteínas , Receptores Acoplados a Proteínas G/genética , Sinapsis/genética
5.
J Biol Chem ; 294(50): 19246-19254, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31628191

RESUMEN

Adhesion G protein-coupled receptors (aGPCRs) represent a distinct family of GPCRs that regulate several developmental and physiological processes. Most aGPCRs undergo GPCR autoproteolysis-inducing domain-mediated protein cleavage, which produces a cryptic tethered agonist (termed Stachel (stinger)), and cleavage-dependent and -independent aGPCR signaling mechanisms have been described. aGPCR G1 (ADGRG1 or G protein-coupled receptor 56 (GPR56)) has pleiotropic functions in the development of multiple organ systems, which has broad implications for human diseases. To date, two natural GPR56 ligands, collagen III and tissue transglutaminase (TG2), and one small-molecule agonist, 3-α-acetoxydihydrodeoxygedunin (3-α-DOG), have been identified, in addition to a synthetic peptide, P19, that contains seven amino acids of the native Stachel sequence. However, the mechanisms by which these natural and small-molecule agonists signal through GPR56 remain unknown. Here we engineered a noncleavable receptor variant that retains signaling competence via the P19 peptide. We demonstrate that both natural and small-molecule agonists can activate only cleaved GPR56. Interestingly, TG2 required both receptor cleavage and the presence of a matrix protein, laminin, to activate GPR56, whereas collagen III and 3-α-DOG signaled without any cofactors. On the other hand, both TG2/laminin and collagen III activate the receptor by dissociating the N-terminal fragment from its C-terminal fragment, enabling activation by the Stachel sequence, whereas P19 and 3-α-DOG initiate downstream signaling without disengaging the N-terminal fragment from its C-terminal fragment. These findings deepen our understanding of how GPR56 signals via natural ligands, and a small-molecule agonist may be broadly applicable to other aGPCR family members.


Asunto(s)
Limoninas/farmacología , Receptores Acoplados a Proteínas G/agonistas , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Células HEK293 , Humanos , Ligandos , Limoninas/química , Masculino , Ratones , Ratones Noqueados , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
6.
Cancer Cell ; 36(5): 528-544.e10, 2019 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-31631026

RESUMEN

H3K27M mutations resulting in epigenetic dysfunction are frequently observed in diffuse intrinsic pontine glioma (DIPGs), an incurable pediatric cancer. We conduct a CRISPR screen revealing that knockout of KDM1A encoding lysine-specific demethylase 1 (LSD1) sensitizes DIPG cells to histone deacetylase (HDAC) inhibitors. Consistently, Corin, a bifunctional inhibitor of HDACs and LSD1, potently inhibits DIPG growth in vitro and in xenografts. Mechanistically, Corin increases H3K27me3 levels suppressed by H3K27M histones, and simultaneously increases HDAC-targeted H3K27ac and LSD1-targeted H3K4me1 at differentiation-associated genes. Corin treatment induces cell death, cell-cycle arrest, and a cellular differentiation phenotype and drives transcriptional changes correlating with increased survival time in DIPG patients. These data suggest a strategy for treating DIPG by simultaneously inhibiting LSD1 and HDACs.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Glioma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Histona Demetilasas/antagonistas & inhibidores , Animales , Antineoplásicos/uso terapéutico , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/mortalidad , Neoplasias del Tronco Encefálico/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Línea Celular Tumoral , Cromatina/metabolismo , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glioma/genética , Glioma/mortalidad , Glioma/patología , Código de Histonas/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Histona Desacetilasas/metabolismo , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histonas/metabolismo , Humanos , Ratones , Mutación , Puente/patología , RNA-Seq , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Trends Pharmacol Sci ; 40(4): 278-293, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30871735

RESUMEN

The family of adhesion G protein-coupled receptors (aGPCRs) consists of 33 members in humans. Although the majority are orphan receptors with unknown functions, many reports have demonstrated critical functions for some members of this family in organogenesis, neurodevelopment, myelination, angiogenesis, and cancer progression. Importantly, mutations in several aGPCRs have been linked to human diseases. The crystal structure of a shared protein domain, the GPCR Autoproteolysis INducing (GAIN) domain, has enabled the discovery of a common signaling mechanism - a tethered agonist - for this class of receptors. A series of recent reports has shed new light on their biological functions and disease relevance. This review focuses on these recent advances in our understanding of aGPCR biology in the nervous system and the untapped potential of aGPCRs as novel therapeutic targets for neurological disease.


Asunto(s)
Terapia Molecular Dirigida , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Receptores Acoplados a Proteínas G/metabolismo , Animales , Desarrollo de Medicamentos/métodos , Humanos , Mutación , Enfermedades del Sistema Nervioso/genética , Enfermedades del Sistema Nervioso/fisiopatología , Receptores Acoplados a Proteínas G/efectos de los fármacos , Receptores Acoplados a Proteínas G/genética , Transducción de Señal
8.
Immunity ; 50(1): 253-271.e6, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30471926

RESUMEN

Microglia, the resident immune cells of the brain, rapidly change states in response to their environment, but we lack molecular and functional signatures of different microglial populations. Here, we analyzed the RNA expression patterns of more than 76,000 individual microglia in mice during development, in old age, and after brain injury. Our analysis uncovered at least nine transcriptionally distinct microglial states, which expressed unique sets of genes and were localized in the brain using specific markers. The greatest microglial heterogeneity was found at young ages; however, several states-including chemokine-enriched inflammatory microglia-persisted throughout the lifespan or increased in the aged brain. Multiple reactive microglial subtypes were also found following demyelinating injury in mice, at least one of which was also found in human multiple sclerosis lesions. These distinct microglia signatures can be used to better understand microglia function and to identify and manipulate specific subpopulations in health and disease.


Asunto(s)
Envejecimiento/inmunología , Lesiones Encefálicas/inmunología , Encéfalo/fisiología , Microglía/fisiología , Esclerosis Múltiple/inmunología , Adaptación Fisiológica , Envejecimiento/genética , Animales , Lesiones Encefálicas/genética , Diferenciación Celular , Enfermedades Desmielinizantes , Humanos , Longevidad , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ARN , Análisis de la Célula Individual
9.
Cell Mol Life Sci ; 75(21): 4007-4019, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29855662

RESUMEN

AIMS: G-protein coupled receptor 56 (GPR56) is the most abundant islet-expressed G-protein coupled receptor, suggesting a potential role in islet function. This study evaluated islet expression of GPR56 and its endogenous ligand collagen III, and their effects on ß-cell function. METHODS: GPR56 and collagen III expression in mouse and human pancreas sections was determined by fluorescence immunohistochemistry. Effects of collagen III on ß-cell proliferation, apoptosis, intracellular calcium ([Ca2+]i) and insulin secretion were determined by cellular BrdU incorporation, caspase 3/7 activities, microfluorimetry and radioimmunoassay, respectively. The role of GPR56 in islet vascularisation and innervation was evaluated by immunohistochemical staining for CD31 and TUJ1, respectively, in pancreases from wildtype (WT) and Gpr56-/- mice, and the requirement of GPR56 for normal glucose homeostasis was determined by glucose tolerance tests in WT and Gpr56-/- mice. RESULTS: Immunostaining of mouse and human pancreases revealed that GPR56 was expressed by islet ß-cells while collagen III was confined to the peri-islet basement membrane and islet capillaries. Collagen III protected ß-cells from cytokine-induced apoptosis, triggered increases in [Ca2+]i and potentiated glucose-induced insulin secretion from WT islets but not from Gpr56-/- islets. Deletion of GPR56 did not affect glucose-induced insulin secretion in vitro and it did not impair glucose tolerance in adult mice. GPR56 was not required for normal islet vascularisation or innervation. CONCLUSION: We have demonstrated that collagen III improves islet function by increasing insulin secretion and protecting against apoptosis. Our data suggest that collagen III may be effective in optimising islet function to improve islet transplantation outcomes, and GPR56 may be a target for the treatment of type 2 diabetes.


Asunto(s)
Colágeno/genética , Diabetes Mellitus Tipo 2/genética , Receptores Acoplados a Proteínas G/genética , Animales , Apoptosis/genética , Calcio/metabolismo , Proliferación Celular/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Humanos , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patología , Islotes Pancreáticos/metabolismo , Islotes Pancreáticos/patología , Ratones , Ratones Noqueados , Páncreas/metabolismo , Páncreas/patología
10.
Elife ; 72018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29809138

RESUMEN

In the central nervous system (CNS), myelin formation and repair are regulated by oligodendrocyte (OL) lineage cells, which sense and integrate signals from their environment, including from other glial cells and the extracellular matrix (ECM). The signaling pathways that coordinate this complex communication, however, remain poorly understood. The adhesion G protein-coupled receptor ADGRG1 (also known as GPR56) is an evolutionarily conserved regulator of OL development in humans, mice, and zebrafish, although its activating ligand for OL lineage cells is unknown. Here, we report that microglia-derived transglutaminase-2 (TG2) signals to ADGRG1 on OL precursor cells (OPCs) in the presence of the ECM protein laminin and that TG2/laminin-dependent activation of ADGRG1 promotes OPC proliferation. Signaling by TG2/laminin to ADGRG1 on OPCs additionally improves remyelination in two murine models of demyelination. These findings identify a novel glia-to-glia signaling pathway that promotes myelin formation and repair, and suggest new strategies to enhance remyelination.


Asunto(s)
Enfermedades Desmielinizantes/genética , Proteínas de Unión al GTP/genética , Microglía/metabolismo , Células Precursoras de Oligodendrocitos/metabolismo , Oligodendroglía/metabolismo , Receptores Acoplados a Proteínas G/genética , Transglutaminasas/genética , Animales , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/metabolismo , Diferenciación Celular , Linaje de la Célula/genética , Cerebelo/citología , Cerebelo/metabolismo , Enfermedades Desmielinizantes/metabolismo , Enfermedades Desmielinizantes/patología , Femenino , Proteínas de Unión al GTP/deficiencia , Regulación del Desarrollo de la Expresión Génica , Humanos , Laminina/genética , Laminina/metabolismo , Masculino , Ratones , Ratones Noqueados , Microglía/citología , Neurogénesis/genética , Células Precursoras de Oligodendrocitos/citología , Oligodendroglía/citología , Prosencéfalo/citología , Prosencéfalo/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Remielinización/genética , Transducción de Señal , Transglutaminasas/deficiencia
11.
Neuron ; 85(4): 755-69, 2015 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-25695270

RESUMEN

Myelin ensheathes axons to allow rapid propagation of action potentials and proper nervous system function. In the peripheral nervous system, Schwann cells (SCs) radially sort axons into a 1:1 relationship before wrapping an axonal segment to form myelin. SC myelination requires the adhesion G protein-coupled receptor GPR126, which undergoes autoproteolytic cleavage into an N-terminal fragment (NTF) and a seven-transmembrane-containing C-terminal fragment (CTF). Here we show that GPR126 has domain-specific functions in SC development whereby the NTF is necessary and sufficient for axon sorting, whereas the CTF promotes wrapping through cAMP elevation. These biphasic roles of GPR126 are governed by interactions with Laminin-211, which we define as a novel ligand for GPR126 that modulates receptor signaling via a tethered agonist. Our work suggests a model in which Laminin-211 mediates GPR126-induced cAMP levels to control early and late stages of SC development.


Asunto(s)
Laminina/metabolismo , Vaina de Mielina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células de Schwann/metabolismo , Animales , Animales Modificados Genéticamente , Animales Recién Nacidos , Células COS , Células Cultivadas , Chlorocebus aethiops , Embrión de Mamíferos , Embrión no Mamífero , Ganglios Espinales/citología , Humanos , Técnicas In Vitro , Laminina/genética , Larva , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Morfolinos/farmacología , Vaina de Mielina/ultraestructura , Neuroglía/metabolismo , Neuroglía/ultraestructura , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Células de Schwann/ultraestructura , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
12.
Nat Commun ; 6: 6121, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25607655

RESUMEN

Mutations in GPR56, a member of the adhesion G protein-coupled receptor family, cause a human brain malformation called bilateral frontoparietal polymicrogyria (BFPP). Magnetic resonance imaging (MRI) of BFPP brains reveals myelination defects in addition to brain malformation. However, the cellular role of GPR56 in oligodendrocyte development remains unknown. Here, we demonstrate that loss of Gpr56 leads to hypomyelination of the central nervous system in mice. GPR56 levels are abundant throughout early stages of oligodendrocyte development, but are downregulated in myelinating oligodendrocytes. Gpr56-knockout mice manifest with decreased oligodendrocyte precursor cell (OPC) proliferation and diminished levels of active RhoA, leading to fewer mature oligodendrocytes and a reduced number of myelinated axons in the corpus callosum and optic nerves. Conditional ablation of Gpr56 in OPCs leads to a reduced number of mature oligodendrocytes as seen in constitutive knockout of Gpr56. Together, our data define GPR56 as a cell-autonomous regulator of oligodendrocyte development.


Asunto(s)
Regulación de la Expresión Génica , Oligodendroglía/citología , Receptores Acoplados a Proteínas G/metabolismo , Animales , Axones/metabolismo , Encéfalo/metabolismo , Linaje de la Célula , Proliferación Celular , Supervivencia Celular , Sistema Nervioso Central/embriología , Sistema Nervioso Central/metabolismo , Cuerpo Calloso/metabolismo , Modelos Animales de Enfermedad , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Malformaciones del Desarrollo Cortical/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Mutación , Vaina de Mielina/química , Vaina de Mielina/metabolismo , Oligodendroglía/metabolismo , Nervio Óptico/metabolismo , Transducción de Señal , Tamoxifeno/química , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
13.
Ann N Y Acad Sci ; 1333: 43-64, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25424900

RESUMEN

The class of adhesion G protein-coupled receptors (aGPCRs), with 33 human homologs, is the second largest family of GPCRs. In addition to a seven-transmembrane α-helix-a structural feature of all GPCRs-the class of aGPCRs is characterized by the presence of a large N-terminal extracellular region. In addition, all aGPCRs but one (GPR123) contain a GPCR autoproteolysis-inducing (GAIN) domain that mediates autoproteolytic cleavage at the GPCR autoproteolysis site motif to generate N- and a C-terminal fragments (NTF and CTF, respectively) during protein maturation. Subsequently, the NTF and CTF are associated noncovalently as a heterodimer at the plasma membrane. While the biological function of the GAIN domain-mediated autocleavage is not fully understood, mounting evidence suggests that the NTF and CTF possess distinct biological activities in addition to their function as a receptor unit. We discuss recent advances in understanding the biological functions, signaling mechanisms, and disease associations of the aGPCRs.


Asunto(s)
Adhesión Celular , Receptores Acoplados a Proteínas G/fisiología , Animales , Discapacidades del Desarrollo/genética , Humanos , Mutación , Neoplasias/genética , Transducción de Señal , Sinapsis/fisiología
14.
PLoS One ; 8(7): e68781, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874761

RESUMEN

Loss of function mutations in GPR56, which encodes a G protein-coupled receptor, cause a specific human brain malformation called bilateral frontoparietal polymicrogyria (BFPP). Studies from BFPP postmortem brain tissue and Gpr56 knockout mice have previously showed that GPR56 deletion leads to breaches in the pial basement membrane (BM) and neuronal ectopias during cerebral cortical development. Since α3ß1 integrin also plays a role in pial BM assembly and maintenance, we evaluated whether it functions together with GPR56 in regulating the same developmental process. We reveal that loss of α3 integrin enhances the cortical phenotype associated with Gpr56 deletion, and that neuronal overmigration through a breached pial BM occurs earlier in double knockout than in Gpr56 single knockout mice. These observations provide compelling evidence of the synergism of GPR56 and α3ß1 integrin in regulating the development of cerebral cortex.


Asunto(s)
Corteza Cerebral/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Membrana Basal/metabolismo , Membrana Basal/patología , Movimiento Celular/genética , Corteza Cerebral/embriología , Corteza Cerebral/patología , Colágeno Tipo III/metabolismo , Eliminación de Gen , Integrina alfa3beta1/genética , Integrina alfa3beta1/metabolismo , Ratones , Ratones Noqueados , Neuronas/metabolismo , Fenotipo , Unión Proteica , Receptores Acoplados a Proteínas G/genética
15.
Endocrinology ; 152(7): 2909-19, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21540284

RESUMEN

Polychlorinated biphenyls (PCB) are industrial chemicals linked to developmental deficits that may be caused in part by disrupting thyroid hormone (TH) action by either reducing serum TH or interacting directly with the TH receptor (TR). Individual PCB congeners can activate the TR in vitro when the metabolic enzyme cytochrome P4501A1 (CYP1A1) is induced, suggesting that specific PCB metabolites act as TR agonists. To test this hypothesis in vivo, we compared two combinations of PCB congeners that either activate the TR (PCB 105 and 118) or not (PCB 138 and 153) in the presence or absence of a PCB congener (PCB 126) that induces CYP1A1 in vitro. Aroclor 1254 was used as a positive control, and a group treated with propylthiouracil was included to characterize the effects of low serum TH. We monitored the effects on TH signaling in several peripheral tissues by measuring the mRNA expression of well-known TH-response genes in these tissues. Aroclor 1254 and its component PCB 105/118/126 reduced total T(4) to the same extent as that of propylthiouracil but increased the expression of some TH target genes in liver. This effect was strongly correlated with CYP1A1 expression supporting the hypothesis that metabolism is necessary. Effects were gene and tissue specific, indicating that tissue-specific metabolism is an important component of PCB disruption of TH action and that PCB metabolites interact in complex ways with the TR. These are essential mechanisms to consider when evaluating the health risks of contaminant exposures, for both PCB and other polycyclic compounds known to interact with nuclear hormone receptors.


Asunto(s)
Antitiroideos/toxicidad , Contaminantes Ambientales/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Bifenilos Policlorados/toxicidad , Efectos Tardíos de la Exposición Prenatal/metabolismo , Receptores de Hormona Tiroidea/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Animales , Antitiroideos/química , Citocromo P-450 CYP1A1/biosíntesis , Citocromo P-450 CYP1A1/genética , Contaminantes Ambientales/química , Inducción Enzimática/efectos de los fármacos , Femenino , Lactancia , Masculino , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Especificidad de Órganos , Bifenilos Policlorados/química , Bifenilos Policlorados/metabolismo , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Hormonas Tiroideas/sangre , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética
16.
Toxicol Sci ; 115(1): 22-33, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20118494

RESUMEN

Glutathione S-transferases (GSTs) play an essential role in the elimination of xenobiotic-derived electrophilic metabolites and also catalyze certain steps in the conversion of endogenous molecules. Their expression is controlled by different transcription factors, such as the antioxidant-activated Nrf2 or the constitutive androstane receptor. Here, we show that the Wnt/beta-catenin pathway is also involved in the transcriptional regulation of GSTs: GSTm2, GSTm3, and GSTm6 are overexpressed in mouse hepatomas with activating Ctnnb1 (encoding beta-catenin) mutations and in transgenic hepatocytes expressing activated beta-catenin. Inversely, GSTm expression is reduced in mice with hepatocyte-specific knock out of Ctnnb1. Activation of beta-catenin-dependent signaling stimulates GSTm expression in vitro. Activation of beta-catenin in mouse hepatoma cells activates GSTm3 promoter-driven reporter activity, independently of beta-catenin/T-cell factor sites, via a retinoid X receptor-binding site. By contrast, GSTm expression is inhibited upon Ras activation in mouse liver tumors and transgenic hepatocytes. Recent studies by different groups have shown that beta-catenin-dependent signaling is involved in the transcriptional control of "perivenous" expression of various cytochrome P450s in mouse liver, whereas Ras signaling was hypothesized to antagonize the perivenous hepatocyte phenotype. In synopsis with our present results, it now appears that the Wnt/beta-catenin pathway functions as a master regulator of the expression of both phase I and phase II drug-metabolizing enzymes in perivenous hepatocytes from mouse liver.


Asunto(s)
Glutatión Transferasa/metabolismo , Hepatocitos/enzimología , Hígado/enzimología , Proteína Wnt1/metabolismo , beta Catenina/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Regulación Enzimológica de la Expresión Génica , Glutatión Transferasa/genética , Hepatocitos/patología , Inactivación Metabólica/genética , Isoenzimas , Hígado/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Mutación Puntual , Transducción de Señal/fisiología , Proteína Wnt1/genética , beta Catenina/genética
17.
Environ Health Perspect ; 115(11): 1623-30, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18007995

RESUMEN

BACKGROUND: Polychlorinated biphenyls (PCBs) may interfere with thyroid hormone (TH) signaling by reducing TH levels in blood, by exerting direct effects on TH receptors (TRs), or both. OBJECTIVE: Our objective was to identify individual PCBs that directly affect TH signaling by acting on the TR. METHODS: We administered a mixture of six PCB congeners based on their ortho substitution pattern, including PCBs 77 and 126 (non-ortho), PCBs 105 and 118 (mono-ortho), and PCBs 138 and 153 (di-ortho), to pregnant Sprague-Dawley rats from gestational days (G) 6 to 16. This mixture, or various combinations of the components, was also evaluated in a transient transfection system using GH3 cells. RESULTS: The mixture reduced serum TH levels in pregnant rats on G16 but simultaneously up-regulated the expression of malic enzyme in liver. It also functioned as a TR agonist in vitro; however, none of the individual PCB congeners comprising this mixture were active in this system. Using the aryl hydrocarbon receptor (AhR) antagonist alpha-naphthoflavone, and the cytochrome P450 (CYP)1A1 antagonist ellipticine, we show that the effect of the mixture on the thyroid hormone response element required AhR and CYP1A1. CONCLUSIONS: We propose that PCB 126 induces CYP1A1 through the AhR in GH3 cells, and that CYP1A1 activates PCB 105 and/or 118 to a form a compound that acts as a TR agonist. These data suggest that some tissues may be especially vulnerable to PCBs interfering directly with TH signaling due to their capacity to express CYP1A1 in response to coplanar PCBs (or other dioxin-like molecules) if sufficient mono-ortho PCBs are present.


Asunto(s)
Citocromo P-450 CYP1A1/biosíntesis , Bifenilos Policlorados/metabolismo , Receptores de Hormona Tiroidea/agonistas , Animales , Línea Celular , Citocromo P-450 CYP1A1/genética , Inducción Enzimática , Femenino , Malato Deshidrogenasa/genética , Malato Deshidrogenasa/metabolismo , Hipófisis/citología , Hipófisis/metabolismo , Bifenilos Policlorados/química , Embarazo , Ratas , Ratas Sprague-Dawley , Relación Estructura-Actividad , Tiroxina/sangre , Tiroxina/metabolismo , Triyodotironina/sangre , Triyodotironina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA