Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 303(6): F873-85, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22759395

RESUMEN

Factors that initiate cellular damage and trigger the inflammatory response cascade and renal injury are not completely understood after renal ischemia-reperfusion injury (IRI). High-mobility group box-1 protein (HMGB1) is a damage-associated molecular pattern molecule that binds to chromatin, but upon signaling undergoes nuclear-cytoplasmic translocation and release from cells. Immunohistochemical and Western blot analysis identified HMGB1 nuclear-cytoplasmic translocation and release from renal cells (particularly vascular and tubular cells) into the venous circulation after IRI. Time course analysis indicated HMGB1 release into the venous circulation progressively increased parallel to increased renal ischemic duration. Ethyl pyruvate (EP) treatment blocked H(2)O(2) (oxidative stress)-induced HMGB1 release from human umbilical vein endothelial cells in vitro, and in vivo resulted in nuclear retention and significant blunting of HMGB1 release into the circulation after IRI. EP treatment before IRI improved short-term serum creatinine and albuminuria, proinflammatory cyto-/chemokine release, and long-term albuminuria and fibrosis. The renoprotective effect of EP was abolished when exogenous HMGB1 was injected, suggesting EP's therapeutic efficacy is mediated by blocking HMGB1 translocation and release. To determine the independent effects of circulating HMGB1 after injury, exogenous HMGB1 was administered to healthy animals at pathophysiological dose. HMGB1 administration induced a rapid surge in systemic circulating cyto-/chemokines (including TNF-α, eotaxin, G-CSF, IFN-γ, IL-10, IL-1α, IL-6, IP-10, and KC) and led to mobilization of bone marrow CD34+Flk1+ cells into the circulation. Our results indicate that increased ischemic duration causes progressively enhanced HMGB1 release into the circulation triggering damage/repair signaling, an effect inhibited by EP because of its ability to block HMGB1 nuclear-cytoplasmic translocation.


Asunto(s)
Proteínas del Grupo de Alta Movilidad/metabolismo , Riñón/irrigación sanguínea , Daño por Reperfusión/metabolismo , Proteínas Represoras/metabolismo , Animales , Células de la Médula Ósea/fisiología , Creatinina/sangre , Citocinas/sangre , Proteínas del Grupo de Alta Movilidad/sangre , Proteínas del Grupo de Alta Movilidad/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Peróxido de Hidrógeno/metabolismo , Masculino , Ratones , Estrés Oxidativo/efectos de los fármacos , Piruvatos/farmacología , Proteínas Represoras/sangre , Proteínas Represoras/farmacología
2.
Dis Markers ; 2022: 5045873, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35845134

RESUMEN

Background: Primary ovarian insufficiency (POI) is a female disease characterized by ovarian function loss under 40 years old. Transplantation of exosomes is an encouraging regenerative medicine method that has the potential for restoring ovarian functions post-POI with high efficiency. Therefore, we investigate the therapeutic efficacy and potential mechanisms of human umbilical cord mesenchymal stem cell- (UCMSC-) derived exosomes on ovarian dysfunction post-POI. Methods: The model of POI was established by intraperitoneal injection with 5 mg/kg cisplatin. The mouse ovarian function was detected by measuring the levels of anti-Mullerian hormone, follicle-stimulating hormone, and estradiol and detecting the morphological changes. For in vitro experiments, the characterization and identification of UCMSCs and UCMSC-derived exosomes were done by observation of morphologies and flow cytometry. To exclude the interference effect of nonspecific precipitation substances, UCMSCs were treated with RNase A or RNase A in combination with Triton X-100. Granulosa cell (GC) identification was performed using immunofluorescence. GC proliferation and viability were assessed using 5-ethynyl-2'-deoxyuridine (EdU) assays and Cell Counting Kit-8 (CCK-8), and GC apoptosis was calculated by flow cytometry. Gene expression and protein levels were evaluated using reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blotting. The binding relationship between miR-29a and HMG-box transcription factor (HBP1) was verified by luciferase reporter assays. Results: In vitro, the human UCMSC-derived exosomes carrying miR-29a upregulation promoted the proliferation of GCs and suppressed their apoptosis. In vivo, miR-29a upregulation reserved the mature follicles and restored the ovarian functions. miR-29a targeted HBP1 and negatively regulated its expression. HBP1 upregulation rescued the miR-29a upregulation-induced inhibition in GC apoptosis and inactivated the Wnt/ß-catenin pathway. Conclusion: The exosomal miR-29a derived from human UCMSCs improves the ovarian function by targeting HBP1 and activating the Wnt/ß-catenin pathway.


Asunto(s)
Exosomas , Células Madre Mesenquimatosas , MicroARNs , Insuficiencia Ovárica Primaria , Adulto , Animales , Apoptosis , Exosomas/metabolismo , Femenino , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas del Grupo de Alta Movilidad/farmacología , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Insuficiencia Ovárica Primaria/genética , Insuficiencia Ovárica Primaria/metabolismo , Insuficiencia Ovárica Primaria/terapia , Proteínas Represoras/metabolismo , Ribonucleasa Pancreática , Factores de Transcripción/metabolismo , Cordón Umbilical/metabolismo , beta Catenina
3.
Eur Cell Mater ; 21: 355-63, 2011 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-21484705

RESUMEN

In previous research, it was shown that recombinant HMGA2 protein enhances the proliferation of porcine chondrocytes grown in vitro, opening up promising applications of this embryonic architectural transcription factor for tissue engineering, such as in cartilage repair. In this paper, we describe the development and analyses of two synthetic fragments comprising the functional AT-hook motifs of the HMGA2 protein, as well as the nuclear transport domain. They can be synthesised up to large scales, while eliminating some of the problems of recombinant protein production, including unwanted modification or contamination by the expression hosts, or of gene therapy approaches such as uncontrolled viral integration and transgene expression even after therapy. Application of one of these peptides onto porcine hyaline cartilage chondrocytes, grown in in vitro monolayer cell culture, showed a growth-promoting effect similar to that of the wild type HMGA2 protein. Furthermore, it also promoted cell growth of adult adipose tissue derived stem cells. Due to its proliferation inducing function and vast availability, this peptide is thus suitable for further application and investigation in various fields such as tissue engineering and stem cell research.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Condrocitos/efectos de los fármacos , Proteínas del Grupo de Alta Movilidad/farmacología , Fragmentos de Péptidos/farmacología , Células Madre/efectos de los fármacos , Tejido Adiposo/citología , Secuencia de Aminoácidos , Análisis de Varianza , Animales , Células Cultivadas , Condrocitos/citología , Perros , Relación Dosis-Respuesta a Droga , Proteínas del Grupo de Alta Movilidad/química , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Microscopía Fluorescente , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Células Madre/citología , Porcinos , Factores de Tiempo
4.
J Exp Med ; 192(4): 565-70, 2000 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-10952726

RESUMEN

Lipopolysaccharide (LPS) is lethal to animals because it activates cytokine release, causing septic shock and tissue injury. Early proinflammatory cytokines (e.g., tumor necrosis factor [TNF] and interleukin [IL]-1) released within the first few hours of endotoxemia stimulate mediator cascades that persist for days and can lead to death. High mobility group 1 protein (HMG-1), a ubiquitous DNA-binding protein, was recently identified as a "late" mediator of endotoxin lethality. Anti-HMG-1 antibodies neutralized the delayed increase in serum HMG-1, and protected against endotoxin lethality, even when passive immunization was delayed until after the early cytokine response. Here we examined whether HMG-1 might stimulate cytokine synthesis in human peripheral blood mononuclear cell cultures. Addition of purified recombinant HMG-1 to human monocyte cultures significantly stimulated the release of TNF, IL-1alpha, IL-1beta, IL-1RA, IL-6, IL-8, macrophage inflammatory protein (MIP)-1alpha, and MIP-1beta; but not IL-10 or IL-12. HMG-1 concentrations that activated monocytes were within the pathological range previously observed in endotoxemic animals, and in serum obtained from septic patients. HMG-1 failed to stimulate cytokine release in lymphocytes, indicating that cellular stimulation was specific. Cytokine release after HMG-1 stimulation was delayed and biphasic compared with LPS stimulation. Computer-assisted image analysis demonstrated that peak intensity of HMG-1-induced cellular TNF staining was comparable to that observed after maximal stimulation with LPS. Administration of HMG-1 to Balb/c mice significantly increased serum TNF levels in vivo. Together, these results indicate that, like other cytokine mediators of endotoxin lethality (e.g., TNF and IL-1), extracellular HMG-1 is a regulator of monocyte proinflammatory cytokine synthesis.


Asunto(s)
Proteínas Portadoras/farmacología , Citocinas/biosíntesis , Proteínas del Grupo de Alta Movilidad/farmacología , Monocitos/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Proteínas Portadoras/genética , Células Cultivadas , Quimiocina CCL3 , Quimiocina CCL4 , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Proteína HMGB1 , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Interleucina-1/metabolismo , Interleucina-10/metabolismo , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Lipopolisacáridos/farmacología , Proteínas Inflamatorias de Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Proteínas Recombinantes/farmacología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
5.
Dev Cell ; 9(5): 593-603, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16256735

RESUMEN

Sox9 and the closely related factor Sox10 are essential for the formation of neural crest precursor cells, and play divergent roles in the process by which these cells are subsequently directed to form specific derivatives. These group E Sox factors have also been implicated in the development of the vertebrate inner ear. Despite their importance, however, the mechanisms that allow SoxE proteins to regulate such a diverse range of cell types have remained poorly understood. Here we demonstrate that during vertebrate development, the activities of individual SoxE factors are well conserved and are regulated by SUMOylation. We show that SoxE mutants that cannot be SUMOylated, or that mimic constitutive SUMOylation, are each able to mediate a subset of the diverse activities characteristic of wild-type SoxE proteins. These findings provide important mechanistic insight into how the activity of widely deployed developmental regulatory proteins can be directed to specific developmental events.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Oído Interno/embriología , Proteínas del Grupo de Alta Movilidad/metabolismo , Cresta Neural/embriología , Factores de Transcripción/metabolismo , Xenopus/embriología , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/farmacología , Oído Interno/citología , Oído Interno/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/farmacología , Melanocitos/citología , Melanocitos/metabolismo , Cresta Neural/citología , Cresta Neural/metabolismo , Neuroglía/citología , Neuroglía/metabolismo , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Factor de Transcripción SOX9 , Factores de Transcripción SOXE , Proteína SUMO-1/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/farmacología , Técnicas del Sistema de Dos Híbridos , Xenopus/genética
6.
Int J Biochem Cell Biol ; 40(8): 1536-42, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18191612

RESUMEN

The high mobility group box (HMGB) 1 protein is a very abundant and conserved protein that is implicated in many key cellular events but its functions within the nucleus remain elusive. The role of this protein in replication of closed circular DNA containing a eukaryotic origin of replication has been studied in vitro by using native and recombinant HMGB1 as well as various modified HMGB1 preparations such as truncated protein, lacking its C-terminal tail, in vivo acetylated protein, and recombinant HMGB1 phosphorylated in vitro by protein kinase C (PKC). Native HMGB1 extracted from tumour cells inhibits replication and this effect is reduced upon acetylation and completely abolished upon removal of the acidic C-terminal tail. Recombinant HMGB1, however, fails to inhibit replication but it acquires such a property following in vitro phosphorylation by PKC.


Asunto(s)
Replicación del ADN/efectos de los fármacos , Proteínas del Grupo de Alta Movilidad/farmacología , Proteínas Represoras/farmacología , Acetilación , Animales , Afidicolina/farmacología , ADN Circular/metabolismo , Proteína HMGB1 , Humanos , Fosforilación , Proteína Quinasa C/metabolismo , Ratas , Proteínas Recombinantes/farmacología , Células Tumorales Cultivadas
7.
J Neurosci ; 26(24): 6413-21, 2006 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-16775128

RESUMEN

Cerebral ischemic injury proceeds with excitotoxicity-induced acute neuronal death in the ischemic core and with delayed damage processes in the penumbra. However, knowledge concerning the direct mediators that connect these two processes is limited. Here, we demonstrate that high-mobility group box 1 (HMGB1), a nonhistone DNA-binding protein, is massively released into the extracellular space immediately after ischemic insult and that it subsequently induces neuroinflammation in the postischemic brain. Short hairpin (sh)RNA-mediated HMGB1 downregulation in the postischemic brain suppressed infarct size, microglia activation, and proinflammatory marker induction, indicating that HMGB1 plays a crucial role in the inflammatory process. The proinflammatory cytokine-like function of extracellular HMGB1 was further verified in primary cortical cultures and microglial cultures. HMGB1 was found to accumulate in NMDA-treated primary cortical culture media, and supernatants collected from these cultures were found to trigger microglia activation, the hallmark of brain inflammation. Moreover, treatment with recombinant HMGB1 also induced microglial activation, but HMGB1-depleted supernatant produced by anti-HMGB1 antibody treatment or by HMGB1 shRNA expression did not, thus demonstrating the essential role of HMGB1 in microglial activation. Together, these results indicate that HMGB1 functions as a novel proinflammatory cytokine-like factor that connects excitotoxicity-induced acute damage processes and delayed inflammatory processes in the postischemic brain.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Inflamación/metabolismo , Inflamación/patología , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Animales , Northern Blotting/métodos , Western Blotting/métodos , Encéfalo/patología , Muerte Celular/fisiología , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Embrión de Mamíferos , Inhibidores Enzimáticos/toxicidad , Agonistas de Aminoácidos Excitadores/toxicidad , Expresión Génica/efectos de los fármacos , Expresión Génica/fisiología , Proteína HMGB1 , Proteínas del Grupo de Alta Movilidad/farmacología , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Inflamación/etiología , Masculino , Ratones , Microglía/metabolismo , Microglía/fisiología , N-Metilaspartato/toxicidad , Neuronas/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Estaurosporina/toxicidad , Factores de Tiempo , Transfección/métodos
8.
J Thromb Haemost ; 5(1): 109-16, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17239166

RESUMEN

BACKGROUND: Sepsis is a life-threatening disorder resulting from systemic inflammatory and coagulatory responses to infection. High-mobility group box 1 protein (HMGB1), an abundant intranuclear protein, was recently identified as a potent lethal mediator of sepsis. However, the precise mechanisms by which HMGB1 exerts its lethal effects in sepsis have yet to be confirmed. We recently reported that plasma HMGB1 levels correlated with disseminated intravascular coagulation (DIC) score, indicating that HMGB1 might play an important role in the pathogenesis of DIC. OBJECTIVES: To investigate the mechanisms responsible for the lethal effects of HMGB1, and more specifically, to explore the effects of HMGB1 on the coagulation system. METHODS: Rats were exposed to thrombin with or without HMGB1, and a survival analysis, pathologic analyses and blood tests were conducted. The effects of HMGB1 on the coagulation cascade, anticoagulant pathways and surface expression of procoagulant or anticoagulant molecules were examined in vitro. RESULTS: Compared to thrombin alone, combined administration of thrombin and HMGB1 resulted in excessive fibrin deposition in glomeruli, prolonged plasma clotting times, and increased mortality. In vitro, HMGB1 did not affect clotting times, but inhibited the anticoagulant protein C pathway mediated by the thrombin-thrombomodulin complex, and stimulated tissue factor expression on monocytes. CONCLUSIONS: These findings demonstrate the procoagulant role of HMGB1 in vivo and in vitro. During sepsis, massive accumulation of HMGB1 in the systemic circulation would promote the development of DIC.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Coagulantes/farmacología , Coagulación Intravascular Diseminada/sangre , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas Represoras/metabolismo , Trombosis/sangre , Animales , Pruebas de Coagulación Sanguínea , Células Cultivadas , Coagulantes/toxicidad , Citocinas/sangre , Modelos Animales de Enfermedad , Coagulación Intravascular Diseminada/inducido químicamente , Coagulación Intravascular Diseminada/metabolismo , Coagulación Intravascular Diseminada/patología , Activación Enzimática/efectos de los fármacos , Fibrina/metabolismo , Proteína HMGB1 , Hemólisis/efectos de los fármacos , Proteínas del Grupo de Alta Movilidad/farmacología , Proteínas del Grupo de Alta Movilidad/toxicidad , Humanos , Inflamación/sangre , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Proteína C/metabolismo , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/farmacología , Proteínas Represoras/toxicidad , Trombina , Tromboplastina/metabolismo , Trombosis/inducido químicamente , Trombosis/metabolismo , Trombosis/patología
9.
Biochim Biophys Acta ; 866(4): 233-41, 1986 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-3964709

RESUMEN

The endogenous DNA methylase in nuclei isolated from growing mouse cells preferentially methylates DNA in micrococcal nuclease-resistant regions probably as a result of the location in these regions of the preponderance of hemimethylated sites. Added mouse ascites cell DNA methylase catalyses the methylation of exposed, nuclease-sensitive DNA in chromatin from growing or non-growing mouse or insect cells. The poor acceptor ability of nuclease-resistant regions in this situation is due to the presence of histone proteins which block de novo methylation. Transcriptionally active regions of chromatin are selectively methylated in vitro by either endogenous or added DNA methylase.


Asunto(s)
Cromatina/metabolismo , Animales , Células Cultivadas , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasas/análisis , Proteínas del Grupo de Alta Movilidad/farmacología , Metilación , Ratones , Nucleoproteínas/farmacología , Transcripción Genética
10.
Biochim Biophys Acta ; 1395(2): 228-36, 1998 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-9473681

RESUMEN

Up-stream stimulatory factor (USF)1 is a human transcription factor which binds specifically to the E-box in the Ad MLP located at - 58 from the start site. The nature of USF binding on a Ad MLP DNA fragment was investigated in the presence of DTT and also in the presence of purified HMG-1 using electrophoretic mobility shift assay. We show that the binding capacity of USF for the E-box increases significantly with increasing DTT concentrations. At the higher DTT levels, a second USF-DNA complex is formed in which there is co-occupation of both the E-box and the initiator sequence. The stability of the second complex is largely refractory to an excess of unlabeled oligonucleotide which contains the initiator sequence. These findings indicate a cooperative binding interaction between USF ligands bound simultaneously at the E-box and the Inr sequence. Two models are proposed which are consistent with these data. Furthermore, experiments indicate that the presence of HMG-1, a nuclear protein known to influence transcriptional activity, increases USF binding activity at the E-box by as much as 100%. These findings indicate that both reducing conditions and HMG-1 may act as modulators of USF-regulated transcription.


Asunto(s)
Adenoviridae/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ditiotreitol/farmacología , Proteínas del Grupo de Alta Movilidad/metabolismo , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas del Grupo de Alta Movilidad/farmacología , Humanos , Oxidación-Reducción , Proteínas Recombinantes de Fusión/metabolismo , Factores Estimuladores hacia 5' , Proteínas Virales
11.
Biochim Biophys Acta ; 1354(3): 279-90, 1997 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-9427537

RESUMEN

We have determined the effect of HMG-1 bound to cisplatin-damaged DNA on the activities of calf helicase E. DNase I protection analysis demonstrated HMG-1 bound a cisplatin-damaged 24 base oligonucleotide annealed to M13mp18. Exonuclease digestion experiments revealed that greater than 90% of the DNA substrates contained a single site specific cisplatin adduct and, maximally, 65% of the substrates were bound by HMG-1. Helicase E catalyzed displacement of the cisplatin-damaged DNA oligonucleotide was inhibited by HMG-1 in a concentration-dependent manner. Time course experiments revealed a decreased rate of displacement in reactions containing HMG-1. The maximum inhibition observed was 55% and taking into account that only 65% of the substrates had HMG-1 bound, approximately 85% inhibition was observed on platinated DNA substrates containing HMG-1. Inhibition of helicase activity was proportional to the amount of substrate bound by HMG-1 based on the displacement and exonuclease assays at varying HMG-1 concentrations. The ability of helicase E to displace an undamaged DNA oligonucleotide from a cisplatin-damaged DNA template was also inhibited by HMG-1. Interestingly, HMG-1 had no effect on the rate of DNA-dependent ATP hydrolysis catalyzed by helicase E on the same DNA substrate. The inhibition of helicase activity by HMG-1 binding cisplatin-damaged DNA further supports a role for HMG-1 inhibiting DNA repair which may contribute to cellular sensitivity to cisplatin.


Asunto(s)
Adenosina Trifosfatasas/antagonistas & inhibidores , Proteínas Portadoras/farmacología , Cisplatino/toxicidad , Daño del ADN/efectos de los fármacos , ADN Helicasas/antagonistas & inhibidores , ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/farmacología , Animales , Proteínas Portadoras/metabolismo , Catálisis , Bovinos , Cisplatino/metabolismo , Desoxirribonucleasa I , Proteína HMGB1 , Proteínas del Grupo de Alta Movilidad/metabolismo , Cinética , Especificidad por Sustrato
12.
Mol Endocrinol ; 16(3): 529-40, 2002 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11875113

RESUMEN

Steroidogenic factor 1 (SF-1) is an essential factor in endocrine proliferation and gene expression. Despite the fact that SF-1 expression is restricted to specialized cells within the endocrine system, the only identified regulatory factors of SF-1 are the ubiquitously expressed E-box proteins (upstream stimulatory factors 1 and 2). Sequence examination of the SF-1 proximal promoter revealed a conserved site of AACAAAG (Sox-BS1), which matches exactly the defined consensus Sox protein binding element. Among the approximately 20 known members of the Sox gene family, we focused on Sox3, Sox8, and Sox9, based on their coexpression with SF-1 in the embryonic testis. Indeed, all three of these Sox proteins were capable of binding the proximal Sox-BS1 within the SF-1 promoter (-110 to -104), albeit with differing affinities. Of the three Sox proteins, Sox9 exhibited high-affinity binding to the Sox-BS1 element and consistently activated SF-1 promoter-reporter constructs. Mutating the Sox-BS1 attenuated SF-1 promoter activity in both embryonic and postnatal Sertoli cells, as well as in the adrenocortical cell line, Y1. Our findings, taken together with the overlapping expression profiles of Sox9 and SF-1, and the similar intersex phenotypes associated with both SOX9 and SF-1 human mutations, suggest that Sox9 up-regulates SF-1 and accounts partially for the sexually dimorphic expression pattern of SF-1 observed during male gonadal differentiation.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas del Grupo de Alta Movilidad/farmacología , Factores de Transcripción/genética , Factores de Transcripción/farmacología , Corteza Suprarrenal/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular , Secuencia de Consenso , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Fushi Tarazu , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas de Homeodominio , Humanos , Masculino , Ratones , Mutagénesis Sitio-Dirigida , Regiones Promotoras Genéticas , Ratas , Ratas Sprague-Dawley , Receptores Citoplasmáticos y Nucleares , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción SOX9 , Factores de Transcripción SOXB1 , Factores de Transcripción SOXE , Análisis de Secuencia de ADN , Células de Sertoli/metabolismo , Caracteres Sexuales , Factor Esteroidogénico 1 , Testículo/embriología , Testículo/metabolismo , Factores de Transcripción/metabolismo , Transfección
13.
Endocrinology ; 142(9): 3987-95, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11517178

RESUMEN

Phex is an endopetidase that regulates systemic phosphate homeostasis. We investigated Phex gene transcription by cloning and performing functional analysis of the 2736 bp of the 5' flanking region of the mouse Phex gene containing its promoter. We identified a transcription start site, a consensus TATA-box, and multiple potential cis-acting regulator elements. To determine whether the promoter directs cell-type restricted Phex expression, we transfected full-length and 5'-deleted Phex luciferase reporter constructs into various cell lines. Phex-expressing C5.18 chondrocytes displayed the highest activity of the transfected Phex promoter constructs compared with non-Phex-expressing COS-7 cells, whereas promoter activity was intermediate in ROS 17/2.8 osteoblasts and maturation stage-dependent in MC3T3-E1 osteoblasts. Analysis of sequential 5'-deletion mutants of the Phex promoter in ROS 17/2.8 cells revealed bimodal activity, suggesting that both positive and negative cis-acting regions may be present. The chondrogenic factor SOX9 markedly stimulated Phex promoter activity, whereas Cbfa1, PTH, and 1,25(OH)(2)D(3) had no effect. Our findings are consistent with the predominant expression of Phex in bone and cartilage. Additional studies will be needed to confirm the regulatory regions in the Phex promoter that function in a cell-restricted manner.


Asunto(s)
Clonación Molecular , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/fisiología , Proteínas/genética , Regiones no Traducidas 5'/genética , Secuencia de Aminoácidos/genética , Animales , Secuencia de Bases/genética , Línea Celular , Senescencia Celular/fisiología , Fibroblastos/metabolismo , Proteínas del Grupo de Alta Movilidad/farmacología , Ratones , Datos de Secuencia Molecular , Osteoblastos/metabolismo , Endopeptidasa Neutra Reguladora de Fosfato PHEX , Regiones Promotoras Genéticas/efectos de los fármacos , ARN Mensajero/metabolismo , Factor de Transcripción SOX9 , Factores de Transcripción/farmacología , Transcripción Genética/fisiología , Regulación hacia Arriba
14.
FEBS Lett ; 327(2): 141-4, 1993 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-8335103

RESUMEN

The high mobility group (HMG) proteins represent a class of chromosomal non-histone proteins with an assumed influence on transcription. In this context, the effect of the maize HMGa protein on reporter gene expression was examined. Transient co-transformation experiments in maize protoplasts with plasmid constructs directing the synthesis of the maize HMGa protein and with a luciferase reporter plasmid demonstrated a stimulatory effect of the HMGa protein on the reporter gene expression. Additional experiments with HMGa deletion constructs indicated that the HMG-Box DNA-binding motif is important for the observed effect, while the acidic carboxy-terminal domain of the HMGa protein appears to be dispensable.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/farmacología , Proteínas de Plantas , Protoplastos , Zea mays/genética , Luciferasas/genética , Plásmidos , Transformación Genética
15.
FEBS Lett ; 433(1-2): 174-8, 1998 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-9738956

RESUMEN

NF-Y is a conserved sequence-specific transcription factor binding to CCAAT boxes. The chromatin-associated HMGI proteins influence promoter activities through positive and negative effects on binding of transcription factors. It was previously shown that HMGI(Y) synergizes the binding of NF-Y to the alpha2-collagen CCAAT box [Currie, R.A. (1997) J. Biol Chem. 272, 30880-30888]. Using recombinant proteins, we confirm that at low concentrations of NF-Y, HMGI(Y) acts synergistically on the alpha2-collagen CCAAT and we extend this observation to HMGI and HMGI-C. However, enhancement of DNA binding to gamma-globin, alpha-globin and MHC class II Ea CCAAT boxes was not observed. At high concentrations, HMGI proteins inhibit binding to alpha2-collagen and to gamma-globin, but not to high affinity Ea or a-globin CCAAT. In none of our experiments did we see a ternary complex between NF-Y, HMGI(Y) and DNA. In protein competition experiments, NF-Y affinity was at least two orders of magnitude higher, even in the context of the suboptimal gamma-globin CCAAT. Our data prove that HMGI proteins have complex positive and negative effects on NF binding to some, but not to all CCAAT boxes, suggesting that this phenomenon is dictated by the sequences flanking the pentanucleotide rather than direct protein-protein interactions.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/farmacología , Secuencias Reguladoras de Ácidos Nucleicos , Animales , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT , Colágeno/genética , Globinas/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Ratones
16.
J Neuroimmunol ; 148(1-2): 63-73, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14975587

RESUMEN

Glatiramer acetate (GLAT) is a mixture of basic polypeptides that have been shown to suppress experimental autoimmune encephalomyelitis (EAE). As Copaxone, GLAT is approved for the treatment of relapsing-remitting multiple sclerosis (MS). Different immunomechanisms have been suggested to contribute to the beneficial effects of GLAT which rely on blockade of MHC class II molecules or cross-recognition with myelin basic protein (MBP). Because GLAT could also inhibit experimental autoimmunity not related to myelin proteins, we searched for additional, less-restricted immunomodulatory actions of GLAT. Using freshly isolated resident peritoneal macrophages from naive Lewis rats, it is shown that GLAT profoundly modulates cytokine secretion of the cells. In unseparated macrophages (MPhi) and MPhi of low density, GLAT enhanced constitutive and LPS-induced production of interleukin 10 (IL-10) while LPS-induced synthesis of tumor necrosis factor-alpha (TNF-alpha) was dose-dependently suppressed by GLAT. Although both basic proteins GLAT and MBP facilitated adherence of MPhi, MBP had opposite effects on cytokine production suggesting unique properties of GLAT. In contrast to MPhi, peritoneal mast cells produced only little amounts of cytokines. The inductive effect of GLAT on IL-10 production by antigen-presenting cells was also observed in bone marrow-derived rat dendritic cells (DCs) which, unlike MPhi, were not suppressed in their production of TNF-alpha. Induction of IL-10 in different antigen-presenting cells is a new immunomodulatory mechanism of GLAT. In part, it goes along with the inhibition of TNF-alpha and may be a common basis for the known beneficial effects of GLAT on various cellular autoimmune responses including MS.


Asunto(s)
Antígenos CD , Antígenos de Neoplasias , Antígenos de Superficie , Proteínas Aviares , Proteínas Sanguíneas , Células Dendríticas/efectos de los fármacos , Interleucina-10/metabolismo , Macrófagos Peritoneales/efectos de los fármacos , Péptidos/farmacología , Animales , Antígenos de Diferenciación/metabolismo , Basigina , Células Cultivadas , Células Dendríticas/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas , Ensayo de Inmunoadsorción Enzimática/métodos , Citometría de Flujo/métodos , Acetato de Glatiramer , Proteínas del Grupo de Alta Movilidad/farmacología , Inmunosupresores/farmacología , Lipopolisacáridos/farmacología , Macrófagos Peritoneales/metabolismo , Mastocitos/efectos de los fármacos , Glicoproteínas de Membrana/metabolismo , Proteína Básica de Mielina/metabolismo , Ratas , Ratas Endogámicas Lew , Factor de Necrosis Tumoral alfa/metabolismo
17.
Mol Cell Endocrinol ; 136(1): 67-78, 1997 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-9510069

RESUMEN

The rat is frequently used as a model to study the role of progesterone (P) in regulating FSH secretion and synthesis. The ability of P to modulate rat FSH-beta mRNA levels suggests the presence of a functional hormone response element. We have found three PRE-like sequences upstream of the transcription start site in the rat FSH-beta gene. These sequences are herein referred to as PRE-like sequence #1, #2 and #3 with #1 being most distal from the start site. The current studies determined whether these PRE-like sequences bound P receptor (PR) and were functional in regulating the induction of expression by P. Electrophoretic mobility shift assays (EMSA) demonstrated that a single 289 base pair (bp) DNA fragment encompassing all three PRE-like sequences specifically bound PR. Further, PR bound with high affinity to double-stranded oligonucleotides representing individual PRE-like sequences #1, #2 and, with lower affinity to a double-stranded oligonucleotide representing PRE-like sequence, #3. We have cloned a 361 bp sequence from the promoter region of the rat FSH-beta gene encompassing all three PRE-like sequences into a luciferase reporter vector (pGL3-promoter) yielding pFSHbeta361-luc+ which when transiently transfected into primary rat pituitary cell cultures, conferred P-responsiveness to a heterologous promoter. P-responsiveness was dependent upon the presence of PR and was blocked by the PR antagonist RU-486. These data strongly suggest the presence of functional PRE's in the rat FSH-beta gene promoter.


Asunto(s)
Hormona Folículo Estimulante/genética , Regulación de la Expresión Génica/genética , Progesterona/farmacología , Regiones Promotoras Genéticas/genética , Receptores de Progesterona/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Células Cultivadas , Clonación Molecular , ADN/metabolismo , Femenino , Hormona Folículo Estimulante de Subunidad beta , Proteínas del Grupo de Alta Movilidad/farmacología , Humanos , Mifepristona/farmacología , Datos de Secuencia Molecular , Adenohipófisis/citología , Progesterona/antagonistas & inhibidores , Congéneres de la Progesterona/farmacología , Promegestona/farmacología , Ratas , Proteínas Recombinantes de Fusión , Transfección
18.
Life Sci ; 66(18): 1725-31, 2000 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-10809169

RESUMEN

High mobility group (HMG) I-type chromosomal phosphoproteins HMG I/Y and HMG I-C were investigated following morphine treatment of C6BU-1 glioma. Cells were labelled with [32P]-orthophosphoric acid. Electrophoretic profiles and autoradiograms of the control cells revealed the presence of HMG I and HMG I-C proteins. HMG Y was not detected. Northern blot analysis showed a single HMG I/Y transcript. Treatment with morphine lowered the [32P]-incorporation in HMG I and HMGI-C proteins and the level of the HMG I/Y transcript. However, it did not change the protein ratios on the Coomassie stained gels. These results suggest that morphine may trigger independent reaction pathways affecting either transcription regulation and/or postsynthetic phosphorylation of the preexisting HMG I-type proteins. In addition, opposing changes in the postsynthetic phosphorylation of HMG 14 and histones H1AB were also noticed.


Asunto(s)
Analgésicos Opioides/farmacología , Neoplasias Encefálicas/metabolismo , Cromosomas/metabolismo , Glioma/metabolismo , Proteínas del Grupo de Alta Movilidad/farmacología , Morfina/farmacología , Factores de Transcripción/farmacología , Animales , Autorradiografía , Northern Blotting , Cromosomas/efectos de los fármacos , Electroforesis en Gel de Poliacrilamida , Proteína HMGA1a , Ratas , Colorantes de Rosanilina
20.
Tissue Eng Part A ; 15(3): 473-7, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18721076

RESUMEN

The low capability of self-repair in hyaline cartilage tissue and chondrocytes de-differentiating when grown in vitro (e.g., for tissue engineering approaches) limits articular cartilage repair. It has been shown that the embryonic architectural transcription factors of the high-mobility-group-A (HMGA) protein family affect the regulation of cell differentiation by influencing the state of cell chromatin and are involved in hyaline cartilage development by affecting the expression of chondrocyte-specific marker genes. Thus, the control of cartilage cell proliferation and differentiation by HMGA proteins promises to be an important aspect in cartilage tissue repair. To elucidate the effects on the proliferative activity of hyaline chondrocytes, HMGA proteins were recombinantly expressed, highly purified, and applied to porcine hyaline cartilage cells growing in in vitro monolayer cell culture. Direct application of HMGA1a, HMGA1b, and HMGA2 proteins onto porcine chondrocytes was shown to have a highly significant influence on cell proliferation. Greater proliferation of chondrocytes was achieved than in the untreated control group, indicating a promising approach to enhancing cartilage tissue repair.


Asunto(s)
Condrocitos/citología , Condrocitos/efectos de los fármacos , Proteínas del Grupo de Alta Movilidad/farmacología , Animales , Cartílago/citología , Cartílago/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Condrocitos/metabolismo , Humanos , Sus scrofa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA