Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Microb Cell Fact ; 11: 78, 2012 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-22694812

RESUMEN

BACKGROUND: Recent successes in the determination of G-protein coupled receptor (GPCR) structures have relied on the ability of receptor variants to overcome difficulties in expression and purification. Therefore, the quick screening of functionally expressed stable receptor variants is vital. RESULTS: We developed a platform using Saccharomyces cerevisiae for the rapid construction and evaluation of functional GPCR variants for structural studies. This platform enables us to perform a screening cycle from construction to evaluation of variants within 6-7 days. We firstly confirmed the functional expression of 25 full-length class A GPCRs in this platform. Then, in order to improve the expression level and stability, we generated and evaluated the variants of the four GPCRs (hADRB2, hCHRM2, hHRH1 and hNTSR1). These stabilized receptor variants improved both functional activity and monodispersity. Finally, the expression level of the stabilized hHRH1 in Pichia pastoris was improved up to 65 pmol/mg from negligible expression of the functional full-length receptor in S. cerevisiae at first screening. The stabilized hHRH1 was able to be purified for use in crystallization trials. CONCLUSIONS: We demonstrated that the S. cerevisiae system should serve as an easy-to-handle and rapid platform for the construction and evaluation of GPCR variants. This platform can be a powerful prescreening method to identify a suitable GPCR variant for crystallography.


Asunto(s)
Receptores Acoplados a Proteínas G/metabolismo , Saccharomyces cerevisiae/metabolismo , Cristalización , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Pichia/metabolismo , Plásmidos/genética , Plásmidos/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Histamínicos H1/genética , Receptores Histamínicos H1/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
2.
Nature ; 482(7384): 237-40, 2012 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-22286059

RESUMEN

G-protein-coupled receptors are the largest class of cell-surface receptors, and these membrane proteins exist in equilibrium between inactive and active states. Conformational changes induced by extracellular ligands binding to G-protein-coupled receptors result in a cellular response through the activation of G proteins. The A(2A) adenosine receptor (A(2A)AR) is responsible for regulating blood flow to the cardiac muscle and is important in the regulation of glutamate and dopamine release in the brain. Here we report the raising of a mouse monoclonal antibody against human A(2A)AR that prevents agonist but not antagonist binding to the extracellular ligand-binding pocket, and describe the structure of A(2A)AR in complex with the antibody Fab fragment (Fab2838). This structure reveals that Fab2838 recognizes the intracellular surface of A(2A)AR and that its complementarity-determining region, CDR-H3, penetrates into the receptor. CDR-H3 is located in a similar position to the G-protein carboxy-terminal fragment in the active opsin structure and to CDR-3 of the nanobody in the active ß(2)-adrenergic receptor structure, but locks A(2A)AR in an inactive conformation. These results suggest a new strategy to modulate the activity of G-protein-coupled receptors.


Asunto(s)
Regulación Alostérica/efectos de los fármacos , Anticuerpos Monoclonales/farmacología , Agonismo Inverso de Drogas , Receptor de Adenosina A2A/metabolismo , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Receptores Acoplados a Proteínas G/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Regiones Determinantes de Complementariedad/inmunología , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/farmacología , Ligandos , Ratones , Modelos Moleculares , Opsinas/inmunología , Pichia , Conformación Proteica/efectos de los fármacos , Receptor de Adenosina A2A/química , Receptor de Adenosina A2A/inmunología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/química
3.
Nature ; 482(7386): 547-51, 2012 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-22278061

RESUMEN

The parasympathetic branch of the autonomic nervous system regulates the activity of multiple organ systems. Muscarinic receptors are G-protein-coupled receptors that mediate the response to acetylcholine released from parasympathetic nerves. Their role in the unconscious regulation of organ and central nervous system function makes them potential therapeutic targets for a broad spectrum of diseases. The M2 muscarinic acetylcholine receptor (M2 receptor) is essential for the physiological control of cardiovascular function through activation of G-protein-coupled inwardly rectifying potassium channels, and is of particular interest because of its extensive pharmacological characterization with both orthosteric and allosteric ligands. Here we report the structure of the antagonist-bound human M2 receptor, the first human acetylcholine receptor to be characterized structurally, to our knowledge. The antagonist 3-quinuclidinyl-benzilate binds in the middle of a long aqueous channel extending approximately two-thirds through the membrane. The orthosteric binding pocket is formed by amino acids that are identical in all five muscarinic receptor subtypes, and shares structural homology with other functionally unrelated acetylcholine binding proteins from different species. A layer of tyrosine residues forms an aromatic cap restricting dissociation of the bound ligand. A binding site for allosteric ligands has been mapped to residues at the entrance to the binding pocket near this aromatic cap. The structure of the M2 receptor provides insights into the challenges of developing subtype-selective ligands for muscarinic receptors and their propensity for allosteric regulation.


Asunto(s)
Antagonistas Colinérgicos/química , Antagonistas Colinérgicos/farmacología , Quinuclidinil Bencilato/análogos & derivados , Quinuclidinil Bencilato/química , Quinuclidinil Bencilato/farmacología , Receptor Muscarínico M2/antagonistas & inhibidores , Receptor Muscarínico M2/química , Acetilcolina/análogos & derivados , Acetilcolina/química , Acetilcolina/metabolismo , Acetilcolinesterasa/química , Acetilcolinesterasa/metabolismo , Regulación Alostérica , Sitios de Unión , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Antagonistas Colinérgicos/metabolismo , Cristalografía por Rayos X , Evolución Molecular , Humanos , Ligandos , Modelos Moleculares , Conformación Proteica , Quinuclidinil Bencilato/metabolismo , Receptor Muscarínico M2/genética , Receptor Muscarínico M2/metabolismo , Tirosina/química , Tirosina/metabolismo
4.
Microb Cell Fact ; 10: 24, 2011 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-21513509

RESUMEN

BACKGROUND: Various protein expression systems, such as Escherichia coli (E. coli), Saccharomyces cerevisiae (S. cerevisiae), Pichia pastoris (P. pastoris), insect cells and mammalian cell lines, have been developed for the synthesis of G protein-coupled receptors (GPCRs) for structural studies. Recently, the crystal structures of four recombinant human GPCRs, namely ß2 adrenergic receptor, adenosine A2a receptor, CXCR4 and dopamine D3 receptor, were successfully determined using an insect cell expression system. GPCRs expressed in insect cells are believed to undergo mammalian-like posttranscriptional modifications and have similar functional properties than in mammals. Crystal structures of GPCRs have not yet been solved using yeast expression systems. In the present study, P. pastoris and insect cell expression systems for the human muscarinic acetylcholine receptor M2 subtype (CHRM2) were developed and the quantity and quality of CHRM2 synthesized by both expression systems were compared for the application in structural studies. RESULTS: The ideal conditions for the expression of CHRM2 in P. pastoris were 60 hr at 20°C in a buffer of pH 7.0. The specific activity of the expressed CHRM2 was 28.9 pmol/mg of membrane protein as determined by binding assays using [3H]-quinuclidinyl benzilate (QNB). Although the specific activity of the protein produced by P. pastoris was lower than that of Sf9 insect cells, CHRM2 yield in P. pastoris was 2-fold higher than in Sf9 insect cells because P. pastoris was cultured at high cell density. The dissociation constant (Kd) for QNB in P. pastoris was 101.14 ± 15.07 pM, which was similar to that in Sf9 insect cells (86.23 ± 8.57 pM). There were no differences in the binding affinity of CHRM2 for QNB between P. pastoris and Sf9 insect cells. CONCLUSION: Compared to insect cells, P. pastoris is easier to handle, can be grown at lower cost, and can be expressed quicker at a large scale. Yeast, P. pastoris, and insect cells are all effective expression systems for GPCRs. The results of the present study strongly suggested that protein expression in P. pastoris can be applied to the structural and biochemical studies of GPCRs.


Asunto(s)
Expresión Génica , Técnicas Genéticas , Pichia/genética , Receptor Muscarínico M2/genética , Receptor Muscarínico M2/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Humanos , Datos de Secuencia Molecular , Pichia/metabolismo , Procesamiento Proteico-Postraduccional , Spodoptera
5.
Protein Expr Purif ; 79(1): 81-7, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21515379

RESUMEN

Anion exchangers are membrane proteins that have been identified in a wide variety of species, where they transport Cl(-) and HCO3(-)across the cell membrane. In this study, we cloned an anion-exchange protein from the genome of the basidiomycete Phanerochaete chrysosporium (PcAEP). PcAEP is a 618-amino acid protein that is homologous to the human anion exchanger (AE1) with 22.9% identity and 40.3% similarity. PcAEP was overexpressed by introducing the PcAEP gene into the genome of Pichia pastoris. As a result, PcAEP localized in the membrane of P. pastoris and was solubilized successfully by n-dodecyl-ß-D-maltoside. His-tagged PcAEP was purified as a single band on SDS-PAGE using immobilized metal affinity chromatography and gel filtration chromatography. Purified PcAEP was found to bind to SITS, an inhibitor of the AE family, suggesting that the purified protein is folded properly. PcAEP expressed and purified using the present system could be useful for biological and structural studies of the anion exchange family of proteins.


Asunto(s)
Antiportadores/genética , Clonación Molecular , Proteínas Fúngicas/genética , Phanerochaete/genética , Pichia/genética , Secuencia de Aminoácidos , Proteína 1 de Intercambio de Anión de Eritrocito/genética , Antiportadores/análisis , Antiportadores/aislamiento & purificación , Membrana Celular/ultraestructura , ADN Complementario/genética , Electroforesis en Gel de Poliacrilamida , Proteínas Fúngicas/análisis , Proteínas Fúngicas/aislamiento & purificación , Humanos , Datos de Secuencia Molecular , Alineación de Secuencia , Solubilidad , Regulación hacia Arriba
6.
Biotechnol Lett ; 33(1): 103-7, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20936326

RESUMEN

PURPOSE OF WORK: Soluble protein expression is an important first step during various types of protein studies. Here, we present the screening strategy of secretable mutant. The strategy aimed to identify those cysteine residues that provoke protein misfolding in the heterologous expression system. Intentional mutagenesis studies should consider the size of the library and the time required for expression screening. Here, we proposed a cysteine-to-serine shuffling mutation strategy (CS shuffling) using a Saccharomyces cerevisiae expression system. This strategy of site-directed shuffling mutagenesis of cysteine-to-serine residues aims to identify the cysteine residues that cause protein misfolding in heterologous expression. In the case of a nonglycosylated mutant of the taste-modifying protein miraculin (MCL), which was used here as a model protein, 25% of all constructs obtained from CS shuffling expressed MCL mutant, and serine mutations were found at Cys47 or Cys92, which are involved in the formation of the disulfide bond. This indicates that these residues had the potential to provoke protein misfolding via incorrect disulfide bonding. The CS shuffling can be performed using a small library and within one week, and is an effective screening strategy of soluble protein expression.


Asunto(s)
Cisteína/genética , Glicoproteínas/metabolismo , Mutagénesis Sitio-Dirigida/métodos , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Serina/genética , Secuencia de Aminoácidos , Vectores Genéticos , Glicoproteínas/genética , Datos de Secuencia Molecular , Pliegue de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Edulcorantes/metabolismo
7.
Biochim Biophys Acta ; 1800(9): 986-92, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20542090

RESUMEN

BACKGROUND: Miraculin (MCL) is a taste-modifying protein that converts sourness into sweetness. The molecular mechanism underlying the taste-modifying action of MCL is unknown. METHODS: Here, a yeast expression system for MCL was constructed to accelerate analysis of its structure-function relationships. The Saccharomyces cerevisiae expression system has advantages as a high-throughput analysis system, but compared to other hosts it is characterized by a relatively low level of recombinant protein expression. To alleviate this weakness, in this study we optimized the codon usage and signal-sequence as the first step. Recombinant MCL (rMCL) was expressed and purified, and the sensory taste was analyzed. RESULTS: As a result, a 2 mg/l yield of rMCL was successfully obtained. Although sensory taste evaluation showed that rMCL was flat in taste under all the pH conditions employed, taste-modifying activity similar to that of native MCL was recovered after deglycosylation. Mutagenetic analysis revealed that the N-glycan attached to Asn42 was bulky in rMCL. CONCLUSIONS: The high-mannose-type N-glycan attached in yeast blocks the taste-modifying activity of rMCL. GENERAL SIGNIFICANCE: The bulky N-glycan attached to Asn42 may cause steric hindrance in the interaction between active residues and the sweet taste receptor hT1R2/hT1R3.


Asunto(s)
Glicoproteínas , Polisacáridos , Receptores Acoplados a Proteínas G/agonistas , Proteínas Recombinantes , Gusto/efectos de los fármacos , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Glicoproteínas/farmacología , Humanos , Polisacáridos/biosíntesis , Polisacáridos/genética , Polisacáridos/farmacología , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Saccharomyces cerevisiae/genética , Gusto/fisiología
8.
Biochem Biophys Res Commun ; 382(4): 704-10, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19306845

RESUMEN

Human TAS2 receptors (hTAS2Rs) perceive bitter tastants, but few studies have explored the structure-function relationships of these receptors. In this paper, we report our trials on the large-scale preparations of hTAS2Rs for structural analysis. Twenty-five hTAS2Rs were expressed using a GFP-fusion yeast system in which the constructs and the culture conditions (e.g., the signal sequence, incubation time and temperature after induction) were optimized by measuring GFP fluorescence. After optimization, five hTAS2Rs (hTAS2R7, hTAS2R8, hTAS2R16, hTAS2R41, and hTAS2R48) were expressed at levels greater than 1mg protein/L of culture, which is a preferable level for purification and crystallization. Among these five bitter taste receptors, hTAS2R41 exhibited the highest detergent solubilization efficiency of 87.1% in n-dodecyl-beta-d-maltopyranoside (DDM)/cholesteryl hemisuccinate (CHS). Fluorescence size-exclusion chromatography showed that hTAS2R41 exhibited monodispersity in DDM/CHS without aggregates, suggesting that hTAS2R41 is a good target for future crystallization trials.


Asunto(s)
Ingeniería Genética/métodos , Receptores Acoplados a Proteínas G/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Cromatografía en Gel , Clonación Molecular , Fluorescencia , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Humanos , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Saccharomyces cerevisiae/genética , Solubilidad , Percepción del Gusto
9.
Biochem Biophys Res Commun ; 380(2): 271-6, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19167344

RESUMEN

N-linked glycosylation is the most common post-translational modification of G-protein-coupled receptors (GPCRs) and is correlated to the localization and function of the receptors depending on each receptor. However, heterogeneity of glycosylation can interfere with protein crystallization. The removal of N-linked glycosylation from membrane proteins improves the ability to crystallize these proteins. We screened 25 non-glycosylated GPCRs for functional receptor production in the methylotrophic yeast Pichia pastoris using specific ligand-receptor binding assays. We found that five clones were expressed at greater than 10 pmol/mg, 9 clones at 1-10 pmol/mg and 11 clones at less than 1 pmol/mg of membrane protein. Further optimization of culture parameters including culture scale, induction time, pH and temperature enabled us to achieve expression of a functional human muscarinic acetylcholine receptor subtype 2 (CHRM2) with a B(max) value of 51.2 pmol/mg of membrane protein. Approximately 1.9 mg of the human CHRM2 was produced from a 1-L culture.


Asunto(s)
Pichia , Receptor Muscarínico M2/biosíntesis , Receptores Acoplados a Proteínas G/biosíntesis , Proteínas Recombinantes/biosíntesis , Glicosilación , Humanos , Biosíntesis de Proteínas , Receptor Muscarínico M2/química , Receptores Acoplados a Proteínas G/química , Proteínas Recombinantes/química
10.
Biochem Biophys Res Commun ; 371(4): 841-5, 2008 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-18474222

RESUMEN

Crystallization of eukaryotic membrane proteins is a challenging, iterative process. The protein of interest is often modified in an attempt to improve crystallization and diffraction results. To accelerate this process, we took advantage of a GFP-fusion yeast expression system that uses PCR to direct homologous recombination and gene cloning. We explored the possibility of employing more than one PCR fragment to introduce various mutations in a single step, and found that when up to five PCR fragments were co-transformed into yeast, the recombination frequency was maintained as the number of fragments was increased. All transformants expressed the model membrane protein, while the resulting plasmid from each clone contained the designed mutations only. Thus, we have demonstrated a technique allowing the expression of mutant membrane proteins within 5 days, combining a GFP-fusion expression system and yeast homologous recombination.


Asunto(s)
Clonación Molecular/métodos , Proteínas de la Membrana/biosíntesis , Proteínas Recombinantes/biosíntesis , Saccharomyces cerevisiae/genética , Animales , Proteínas Fluorescentes Verdes/genética , Proteínas de la Membrana/genética , Ratones , Mutación , Reacción en Cadena de la Polimerasa , Biosíntesis de Proteínas , Proteínas Recombinantes/genética , Recombinación Genética , Canales Catiónicos TRPM/biosíntesis , Canales Catiónicos TRPM/genética , Transformación Genética
11.
PLoS One ; 3(2): e1666, 2008 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-18301744

RESUMEN

BACKGROUND: We demonstrated that mouse embryonic stem (ES) cells-derived vascular endothelial growth factor receptor-2 (VEGF-R2) positive cells could differentiate into both endothelial cells (EC) and mural cells (MC), and termed them as vascular progenitor cells (VPC). Recently, we have established a method to expand monkey and human ES cells-derived VPC with the proper differentiation stage in a large quantity. Here we investigated the therapeutic potential of human VPC-derived EC and MC for vascular regeneration. METHODS AND RESULTS: After the expansion of human VPC-derived vascular cells, we transplanted these cells to nude mice with hindlimb ischemia. The blood flow recovery and capillary density in ischemic hindlimbs were significantly improved in human VPC-derived EC-transplanted mice, compared to human peripheral and umbilical cord blood-derived endothelial progenitor cells (pEPC and uEPC) transplanted mice. The combined transplantation of human VPC-derived EC and MC synergistically improved blood flow of ischemic hindlimbs remarkably, compared to the single cell transplantations. Transplanted VPC-derived vascular cells were effectively incorporated into host circulating vessels as EC and MC to maintain long-term vascular integrity. CONCLUSIONS: Our findings suggest that the combined transplantation of human ES cells-derived EC and MC can be used as a new promising strategy for therapeutic vascular regeneration in patients with tissue ischemia.


Asunto(s)
Isquemia/terapia , Neovascularización Fisiológica , Trasplante de Células Madre/métodos , Animales , Diferenciación Celular , Células Madre Embrionarias/citología , Células Endoteliales/trasplante , Miembro Posterior , Humanos , Ratones , Ratones Desnudos , Músculo Liso Vascular/citología , Miocitos del Músculo Liso/citología , Flujo Sanguíneo Regional , Trasplante Heterólogo
12.
Arterioscler Thromb Vasc Biol ; 27(10): 2127-34, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17872458

RESUMEN

OBJECTIVE: We demonstrated previously that mouse embryonic stem (ES) cell-derived vascular endothelial growth factor receptor-2 (VEGF-R2)-positive cells can differentiate into both vascular endothelial cells and mural cells. This time, we investigated kinetics of differentiation of human ES cells to vascular cells and examined their potential as a source for vascular regeneration. METHODS AND RESULTS: Unlike mouse ES cells, undifferentiated human ES cells already expressed VEGF-R2, but after differentiation, a VEGF-R2-positive but tumor rejection antigen 1-60 (TRA1-60)-negative population emerged. These VEGF-R2-positive but tumor rejection antigen 1-60-negative cells were also positive for platelet-derived growth factor receptor alpha and beta chains and could be effectively differentiated into both VE-cadherin+ endothelial cell and alpha-smooth muscle actin+ mural cell. VE-cadherin+ cells, which were also CD34+ and VEGF-R2+ and thought to be endothelial cells in the early differentiation stage, could be expanded while maintaining their maturity. Their transplantation to the hindlimb ischemia model of immunodeficient mice contributed to the construction of new blood vessels and improved blood flow. CONCLUSIONS: We could identify the differentiation process from human ES cells to vascular cell components and demonstrate that expansion and transplantation of vascular cells at the appropriate differentiation stage may constitute a novel strategy for vascular regenerative medicine.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Células Madre Embrionarias/metabolismo , Células Endoteliales/metabolismo , Miocitos del Músculo Liso/metabolismo , Regeneración , Actinas/metabolismo , Proteínas Angiogénicas/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos de Neoplasias/metabolismo , Cadherinas/metabolismo , Línea Celular , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Células Madre Embrionarias/inmunología , Células Endoteliales/inmunología , Células Endoteliales/trasplante , Miembro Posterior/irrigación sanguínea , Humanos , Isquemia/metabolismo , Isquemia/fisiopatología , Isquemia/cirugía , Cinética , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Miocitos del Músculo Liso/inmunología , Miocitos del Músculo Liso/trasplante , Neovascularización Fisiológica , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Flujo Sanguíneo Regional , Trasplante de Células Madre , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 26(9): 1977-84, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16809546

RESUMEN

OBJECTIVE: The acquisition of arterial or venous identity is highlighted in vascular development. Previously, we have reported an embryonic stem (ES) cell differentiation system that exhibits early vascular development using vascular endothelial growth factor (VEGF) receptor-2 (VEGFR2)-positive cells as common vascular progenitors. In this study, we constructively induced differentiation of arterial and venous endothelial cells (ECs) in vitro to elucidate molecular mechanisms of arterial-venous specification. METHODS AND RESULTS: ECs were induced from VEGFR2+ progenitor cells with various conditions. VEGF was essential to induce ECs. Addition of 8bromo-cAMP or adrenomedullin (AM), an endogenous ligand-elevating cAMP, enhanced VEGF-induced EC differentiation. Whereas VEGF alone mainly induced venous ECs, 8bromo-cAMP (or AM) with VEGF supported substantial induction of arterial ECs. Stimulation of cAMP pathway induced Notch signal activation in ECs. The arterializing effect of VEGF and cAMP was abolished in recombination recognition sequence binding protein at the Jkappa site deficient ES cells lacking Notch signal activation or in ES cells treated with gamma-secretase inhibitor. Nevertheless, forced Notch activation by the constitutively active Notch1 alone did not induce arterial ECs. CONCLUSIONS: Adrenomedullin/cAMP is a novel signaling pathway to activate Notch signaling in differentiating ECs. Coordinated signaling of VEGF, Notch, and cAMP is required to induce arterial ECs from vascular progenitors.


Asunto(s)
Arterias/citología , Diferenciación Celular/fisiología , AMP Cíclico/metabolismo , Células Endoteliales/citología , Péptidos/metabolismo , Receptores Notch/metabolismo , Células Madre/citología , 8-Bromo Monofosfato de Adenosina Cíclica/farmacología , Adrenomedulina , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Sinergismo Farmacológico , Ratones , Péptidos/farmacología , Transducción de Señal/fisiología , Factor A de Crecimiento Endotelial Vascular/farmacología , Venas/citología
14.
Endocrinology ; 147(4): 1642-53, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16384868

RESUMEN

Adrenomedullin (AM) is a vasodilating hormone secreted mainly from vascular wall, and its expression is markedly enhanced after stroke. We have revealed that AM promotes not only vasodilation but also vascular regeneration. In this study, we focused on the roles of AM in the ischemic brain and examined its therapeutic potential. We developed novel AM-transgenic (AM-Tg) mice that overproduce AM in the liver and performed middle cerebral artery occlusion for 20 min (20m-MCAO) to examine the effects of AM on degenerative or regenerative processes in ischemic brain. The infarct area and gliosis after 20m-MCAO was reduced in AM-Tg mice in association with suppression of leukocyte infiltration, oxidative stress, and apoptosis in the ischemic core. In addition, vascular regeneration and subsequent neurogenesis were enhanced in AM-Tg mice, preceded by increase in mobilization of CD34(+) mononuclear cells, which can differentiate into endothelial cells. The vasculo-neuro-regenerative actions observed in AM-Tg mice in combination with neuroprotection resulted in improved recovery of motor function. Brain edema was also significantly reduced in AM-Tg mice via suppression of vascular permeability. In vitro, AM exerted direct antiapoptotic and neurogenic actions on neuronal cells. Exogenous administration of AM in mice after 20m-MCAO also reduced the infarct area, and promoted vascular regeneration and functional recovery. In summary, this study suggests the neuroprotective and vasculo-neuro-regenerative roles of AM and provides basis for a new strategy to rescue ischemic brain through its multiple hormonal actions.


Asunto(s)
Isquemia Encefálica/terapia , Regeneración Nerviosa , Fármacos Neuroprotectores , Péptidos/fisiología , Adrenomedulina , Animales , Infarto Cerebral/terapia , Cuerpo Estriado/irrigación sanguínea , Miembro Posterior/irrigación sanguínea , Humanos , Ratones , Ratones Transgénicos , Neovascularización Fisiológica , Péptidos/genética , Especies Reactivas de Oxígeno
15.
J Clin Invest ; 114(6): 784-94, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15372102

RESUMEN

Production of thromboxane (TX) A2 and PG I2/prostacyclin (PGI2) is increased in patients with atherosclerosis. However, their roles in atherogenesis have not been critically defined. To examine this issue, we cross-bred atherosclerosis-prone apoE-deficient mice with mice deficient in either the TXA receptor (TP) or the PGI receptor (IP). Although they showed levels of serum cholesterol and triglyceride similar to those of apoE-deficient mice, apoE-/-TP-/- mice exhibited a significant delay in atherogenesis, and apoE-/-IP-/- mice exhibited a significant acceleration in atherogenesis compared with mice deficient in apoE alone. The plaques in apoE-/-IP-/- mice showed partial endothelial disruption and exhibited enhanced expression of ICAM-1 and decreased expression of platelet endothelial cell adhesion molecule 1 (PECAM-1) in the overlying endothelial cells compared with those of apoE-/-TP-/- mice. Platelet activation with thrombin ex vivo revealed higher and lower sensitivity for surface P-selectin expression in platelets of apoE-/-IP-/- and apoE-/-TP-/- mice, respectively, than in those of apoE-/- mice. Intravital microscopy of the common carotid artery revealed a significantly greater number of leukocytes rolling on the vessel walls in apoE-/-IP-/- mice than in either apoE-/-TP-/- or apoE-/- mice. We conclude that TXA2 promotes and PGI2 prevents the initiation and progression of atherogenesis through control of platelet activation and leukocyte-endothelial cell interaction.


Asunto(s)
Apolipoproteínas E/deficiencia , Arteriosclerosis/patología , Epoprostenol/metabolismo , Tromboxano A2/metabolismo , Animales , Apolipoproteínas E/genética , Arteriosclerosis/sangre , Arteriosclerosis/inmunología , Arteriosclerosis/fisiopatología , Molécula 1 de Adhesión Intercelular/genética , Macrófagos/citología , Macrófagos/patología , Ratones , Ratones Noqueados , Agregación Plaquetaria , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Receptores de Epoprostenol/deficiencia , Receptores de Epoprostenol/genética , Receptores de Tromboxanos/deficiencia , Receptores de Tromboxanos/genética
16.
FEBS Lett ; 544(1-3): 86-92, 2003 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-12782295

RESUMEN

We previously reported that adrenomedullin (AM), a vasodilating hormone secreted from blood vessels, promotes proliferation and migration of human umbilical vein endothelial cells (HUVECs). In this study, we examined the ability of AM to promote vascular regeneration. AM increased the phosphorylation of Akt in HUVECs and the effect was inhibited by the AM antagonists and the inhibitors for protein kinase A (PKA) or phosphatidylinositol 3-kinase (PI3K). AM promoted re-endothelialization in vitro of wounded monolayer of HUVECs and neo-vascularization in vivo in murine gel plugs. These effects were also inhibited by the AM antagonists and the inhibitors for PKA or PI3K. The findings suggest that AM plays significant roles in vascular regeneration, associated with PKA- and PI3K-dependent activation of Akt in endothelial cells, and possesses therapeutic potential for vascular injury and tissue ischemia.


Asunto(s)
Endotelio Vascular/citología , Neovascularización Fisiológica , Péptidos/metabolismo , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas/metabolismo , Adrenomedulina , Animales , Materiales Biocompatibles/farmacología , Western Blotting , División Celular , Movimiento Celular , Células Cultivadas , Colágeno/farmacología , AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Endotelio/fisiología , Activación Enzimática , Humanos , Laminina/farmacología , Ratones , Péptidos/química , Péptidos/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Unión Proteica , Proteoglicanos/farmacología , Proteínas Proto-Oncogénicas c-akt , Regeneración , Factores de Tiempo , Cicatrización de Heridas
17.
Circulation ; 107(16): 2085-8, 2003 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-12707232

RESUMEN

BACKGROUND: We demonstrated that vascular endothelial growth factor receptor 2 (VEGF-R2)-positive cells derived from mouse embryonic stem (ES) cells can differentiate into both endothelial cells and mural cells to suffice as vascular progenitor cells (VPCs). Here we examined whether VPCs occur in primate ES cells and investigated the differences in VPC differentiation kinetics between primate and mouse ES cells. METHODS AND RESULTS: In contrast to mouse ES cells, undifferentiated monkey ES cells expressed VEGF-R2. By culturing these undifferentiated ES cells for 4 days on OP9 feeder layer, VEGF-R2 expression disappeared, and then reappeared after 8 days of differentiation. We then isolated these VEGF-R2-positive and vascular endothelial cadherin (VEcadherin)-negative cells by flow cytometry sorting. Additional 5-day reculture of these VEGF-R2+ VEcadherin- cells on OP9 feeder layer resulted in the appearance of platelet endothelial cell adhesion molecule-1 (PECAM1)-positive, VEcadherin-positive, endothelial nitric oxide synthase (eNOS)-positive endothelial cells. On a collagen IV-coated dish in the presence of serum, these cells differentiated into smooth muscle actin (SMA)-positive and calponin-positive mural cells (pericytes or vascular smooth muscle cells). Addition of 50 ng/mL VEGF to the culture on a collagen IV-coated dish resulted in the appearance of PECAM1+ cells surrounded by SMA+ cells. In addition, these differentiated VEGF-R2+ cells can form tube-like structures in a 3-dimensional culture. CONCLUSIONS: Our findings indicate that differentiation kinetics of VPCs derived from primate and mouse ES cells were different. Differentiated VEGF-R2+ VEcadherin- cells can act as VPCs in primates. To seek the clinical potential of VPCs for vascular regeneration, investigations of primate ES cells are indispensable.


Asunto(s)
Embrión de Mamíferos/citología , Endotelio Vascular/citología , Músculo Liso Vascular/citología , Células Madre/fisiología , Animales , Diferenciación Celular , Línea Celular , Cinética , Macaca fascicularis , Ratones , Especificidad de la Especie , Células Madre/química , Receptor 2 de Factores de Crecimiento Endotelial Vascular/análisis
18.
Proc Natl Acad Sci U S A ; 100(6): 3404-9, 2003 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-12621153

RESUMEN

Natriuretic peptides (NPs), which consist of atrial, brain, and C-type natriuretic peptides (ANP, BNP, and CNP, respectively), are characterized as cardiac or vascular hormones that elicit their biological effects by activation of the cGMPcGMP-dependent protein kinase (cGK) pathway. We recently reported that adenoviral gene transfer of CNP into rabbit blood vessels not only suppressed neointimal formation but also accelerated reendothelialization, a required step for endothelium-dependent vasorelaxation and antithrombogenicity. Accordingly, we investigated the therapeutic potential of the NPscGMPcGK pathway for vascular regeneration. In transgenic (Tg) mice that overexpress BNP in response to hindlimb ischemia, neovascularization with appropriate mural cell coating was accelerated without edema or bleeding, and impaired angiogenesis by the suppression of nitric oxide production was effectively rescued. Furthermore, in BNP-Tg mice, inflammatory cell infiltration in ischemic tissue and vascular superoxide production were suppressed compared with control mice. Ischemia-induced angiogenesis was also significantly potentiated in cGK type I Tg mice, but attenuated in cGK type I knockout mice. NPs significantly stimulated capillary network formation of cultured endothelial cells by cGK stimulation and subsequent Erk12 activation. Furthermore, gene transfer of CNP into ischemic muscles effectively accelerated angiogenesis. These findings reveal an action of the NPscGMPcGK pathway to exert multiple vasculoprotective and regenerative actions in the absence of apparent adverse effects, and therefore suggest that NPs as the endogenous cardiovascular hormone can be used as a strategy of therapeutic angiogenesis in patients with tissue ischemia.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Vasos Sanguíneos/fisiología , Proteínas Quinasas Dependientes de GMP Cíclico/fisiología , GMP Cíclico/fisiología , Péptido Natriurético Encefálico/fisiología , Regeneración/efectos de los fármacos , Regeneración/fisiología , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/fisiología , Factor Natriurético Atrial/uso terapéutico , Células Cultivadas , Expresión Génica , Técnicas de Transferencia de Gen , Humanos , Inflamación/etiología , Inflamación/patología , Isquemia/terapia , Ratones , Ratones Noqueados , Ratones Transgénicos , Péptido Natriurético Encefálico/genética , Péptido Natriurético Encefálico/uso terapéutico , Péptido Natriurético Tipo-C/genética , Péptido Natriurético Tipo-C/fisiología , Péptido Natriurético Tipo-C/uso terapéutico , Neovascularización Fisiológica
19.
Blood ; 101(7): 2675-8, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12480710

RESUMEN

We demonstrated that Flk-1(+) cells derived from mouse embryonic stem (ES) cells can differentiate into both endothelial cells (ECs) and mural cells (MCs) to suffice as vascular progenitor cells (VPCs). In the present study, we investigated the importance of the stage of ES cell differentiation on effective participation in adult neovascularization. We obtained Flk-1(+) LacZ-expressing undifferentiated VPCs. Additional culture of these VPCs with vascular endothelial growth factor (VEGF) resulted in a mixture of ECs and MCs (differentiated VPCs). We injected VPCs subcutaneously into tumor-bearing mice. Five days after the injection, whereas undifferentiated VPCs were often detected as nonvascular cells, differentiated VPCs were more specifically incorporated into developing vasculature mainly as ECs. VPC-derived MCs were also detected in vascular walls. Furthermore, transplantation of differentiated VPCs augmented tumor blood flow in nude mice. These results indicate that a specific vascular contribution in adult neovascularization can be achieved by selective transplantation of ES cell-derived VPCs in appropriate differentiation stages, which should be the basis for vascular regeneration schemes.


Asunto(s)
Endotelio Vascular/citología , Neovascularización Fisiológica , Trasplante de Células Madre , Células Madre/citología , Animales , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular , Embrión de Mamíferos , Ratones , Ratones Desnudos , Neoplasias Experimentales/irrigación sanguínea , Neovascularización Patológica , Ratas , Células Madre Totipotentes/citología , Células Tumorales Cultivadas , Receptor 2 de Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...