Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Radiol Case Rep ; 19(8): 3561-3568, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38933655

RESUMEN

For maxillary gingival carcinomas, especially those in the molar region, surgical resection is often performed beyond the maxillary tuberosity. Bleeding from the posterior superior alveolar or maxillary artery into the pterygoid process is difficult to stop during partial maxillary resection. Advances in catheterization and materials have enabled the embolization of various vessels. In this report, we describe two cases of maxillary gingival cancer in which preoperative endovascular arterial embolization prevented bleeding due to unexpected vascular injury, allowing for a safe surgery with minimal blood loss. This technique effectively avoids emergency hemostasis for unexpected bleeding when resecting gingival cancers in the maxillary molar region.

2.
Am J Case Rep ; 24: e938904, 2023 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-36967573

RESUMEN

BACKGROUND Osteomas are benign tumors characterized by proliferation of dense or trabecular bone. Most osteomas of the head and neck occur in the mandible, they rarely occur in the maxillary sinus, and free osteomas are extremely rare. While usually detected incidentally on plain radiographs, symptoms appear when the osteoma obstructs the sinus orifice or invades the adjacent orbit or intracranial structures. Herein, we report a case of a patient with an isolated laminar osteoma arising in the maxillary sinus. CASE REPORT A 52-year-old man presented to our hospital with a radiopaque mass in the right maxillary sinus. An oval mass of size 2.7×2.3 cm was observed in the right maxillary sinus on computed tomography, and no sinusitis was present. Under general anesthesia, the mass was removed orally via the modified Caldwell-Luc method. Histopathologic examination revealed layered compact bony tissue covered by the sinus membrane. The mass was free from the inferior wall of the right maxillary sinus, and a part of the mass was covered by the sinus membrane. The postoperative course was uneventful, and no recurrence was noted after 5 years. CONCLUSIONS We experienced a case of an extremely rare osteoma in the maxillary sinus. The osteoma was solitary and free within the maxillary sinus. The treatment performed was surgical excision using the modified Caldwell-Luc method. The cause of the free osteoma was thought to be odontogenic maxillary sinusitis caused by apical periodontitis of the nearby tooth.


Asunto(s)
Osteoma , Neoplasias de los Senos Paranasales , Masculino , Humanos , Persona de Mediana Edad , Seno Maxilar/diagnóstico por imagen , Seno Maxilar/patología , Tomografía Computarizada por Rayos X , Radiografía , Osteoma/diagnóstico por imagen , Osteoma/cirugía , Anestesia General
3.
Cureus ; 15(12): e50228, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38192955

RESUMEN

Bone cysts involving the maxillary sinus are frequently observed, and controlling bleeding from the maxillary or posterior superior alveolar arteries is extremely difficult when the surgical site extends into the palatine fossa or the wing socket behind the maxillary sinus. In this report, we describe a case wherein preoperative endovascular arterial embolization prevented bleeding owing to an unexpected vascular injury that occurred during the removal of a dentigerous cyst from the maxillary sinus. This resulted in a safe operation with less intraoperative bleeding. Although this approach carries the risk of complications, such as paralysis, around the affected area, the likelihood of such complications is low. This approach is useful for performing a safe surgery with minimal blood loss because it avoids the need for emergency hemostasis for major intraoperative hemorrhage.

4.
Front Cell Dev Biol ; 10: 1061216, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36531939

RESUMEN

The expression profiles of exosomal microRNAs (miRNAs) are regulated by the microenvironment, and appropriate priming with mesenchymal stem cells (MSCs) is one of the strategies to enhance the paracrine potency of MSCs. Our previous work demonstrated that exosomes from tumor necrosis factor (TNF)-α-primed human gingiva-derived MSCs (GMSCs) could be a therapeutic tool against periodontitis, and that TNFα-inducible exosomal miR-1260b is essential for the inhibition of alveolar bone loss. However, the precise molecular mechanism underlying miR-1260b-mediated inhibition of osteoclastogenesis is not yet fully understood. Here, we found that the activating transcription factor (ATF)-6ß, a novel miR-1260b-targeting gene, is critical for the regulation of osteoclastogenesis under endoplasmic reticulum (ER) stress. An experimental periodontal mouse model demonstrated that induction of ER stress was accompanied by enhanced ATF6ß expression, and local administration of miR-1260b and ATF6ß siRNA using polyethylenimine nanoparticles (PEI-NPs) significantly suppressed the periodontal bone resorption. In periodontal ligament (PDL) cells, the ER stress inducer, tunicamycin, enhanced the expression of the receptor activator of NF-κB ligand (RANKL), while miR-1260b-mediated downregulation of ATF6ß caused RANKL inhibition. Furthermore, the secretome from miR-1260b/ATF6ß-axis-activated PDL cells inhibited osteoclastogenesis in human CD14+ peripheral blood-derived monocytes. These results indicate that the miR-1260b/ATF6ß axis mediates the regulation of ER stress, which may be used as a novel therapeutic strategy to treat periodontal disease.

5.
Sci Rep ; 12(1): 13344, 2022 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-35922474

RESUMEN

Immunoregulatory properties of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) are promising. Gingival tissue-derived MSCs (GMSCs) have unique immunoregulatory capacity and secrete large amounts of EVs. Recent findings suggest that priming MSCs with inflammatory stimuli is an effective strategy for cell-free therapy. However, the precise mechanism by which the contents of EVs are customized has not been fully elucidated. Here, we show that EVs derived from GMSCs primed with a combination of two pro-inflammatory cytokines, tumor necrosis factor-α (TNF-α) and interferon-α (IFN-α), synergistically promote anti-inflammatory M2 macrophage polarization by increasing the expression of cluster of differentiation 73 (CD73) and CD5 molecule-like (CD5L). Expression of CD73 by TNF-α/IFN-α stimulation was transcriptionally upregulated by the activation of mammalian target of rapamycin signaling and nuclear translocation of hypoxia-inducible factor 1α in GMSCs. TNF-α/IFN-α treatment also significantly increased the expression of CD5L mRNA via the transcription factor DNA-binding protein inhibitor ID3 and liver X receptor. Interestingly, exosomal CD5L is a prerequisite for the synergistic effect of EVs-mediated M2 macrophage polarization. These results indicate that combined pre-licensing with TNF-α and IFN-α in GMSCs is ideal for enhancing the anti-inflammatory function of EVs, which contributes to the establishment of a therapeutic tool.


Asunto(s)
Vesículas Extracelulares , Factor de Necrosis Tumoral alfa , Vesículas Extracelulares/metabolismo , Interferón-alfa/metabolismo , Interferón-alfa/farmacología , Activación de Macrófagos , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
6.
Am J Case Rep ; 22: e929674, 2021 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-33617517

RESUMEN

BACKGROUND Neurofibromas are benign tumors of neurological origin caused by the proliferation of Schwann cells and fibroblasts; they often occur in the skin and nerves as a symptom of von Recklinghausen disease. Solitary neurofibromas are also known to occur on their own, but solitary development in the hard palate is extremely rare and difficult to distinguish from schwannomas. The neural origin of solitary neurofibromas is also difficult to determine intraoperatively, and there have been no reports that clearly identify the neural origin of neurofibromas in the hard palate. CASE REPORT We report a case of a solitary neurofibroma originating in the hard palate in a 24-year-old woman. She presented to our department with a 1.2×0.8-cm dome-shaped left palate mass. After identification of the nerve at the source, the tumor was resected under general anesthesia. Histopathology was positive for S-100 and CD34 immunostaining, as well as for Alcian blue. Eventually, the mass was diagnosed as a neurofibroma. CONCLUSIONS Solitary neurofibromas originating in the hard palate are difficult to differentiate from other neoplastic lesions, especially schwannomas, based on clinical findings alone. Therefore, it is important to perform a biopsy and immunostaining of the biopsied specimens for S-100 and CD34. In neurofibromas, tumor cells are loose and delicate, often with wavy or serpentine nuclei, and S-100 protein-positive cells are sparser than in schwannomas. An overall pathological diagnosis should be made with regard to CD34, taking into account that schwannomas are CD34-negative and neurofibromas are CD34-positive.


Asunto(s)
Neurilemoma , Neurofibroma , Neurofibromatosis 1 , Adulto , Biopsia , Femenino , Humanos , Neurofibroma/diagnóstico , Neurofibroma/cirugía , Paladar Duro , Adulto Joven
7.
Acta Biomater ; 122: 306-324, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33359765

RESUMEN

Mesenchymal stem cell (MSC)-derived exosome plays a central role in the cell-free therapeutics involving MSCs and the contents can be customized under disease-associated microenvironments. However, optimal MSC-preconditioning to enhance its therapeutic potential is largely unknown. Here, we show that preconditioning of gingival tissue-derived MSCs (GMSCs) with tumor necrosis factor-alpha (TNF-α) is ideal for the treatment of periodontitis. TNF-α stimulation not only increased the amount of exosome secreted from GMSCs, but also enhanced the exosomal expression of CD73, thereby inducing anti-inflammatory M2 macrophage polarization. The effect of GMSC-derived exosomes on inflammatory bone loss were examined by ligature-induced periodontitis model in mice. Local injection of GMSC-derived exosomes significantly reduced periodontal bone resorption and the number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts, and these effects were further enhanced by preconditioning of GMSCs with TNF-α. Thus, GMSC-derived exosomes also exhibited anti-osteoclastogenic activity. Receptor activator of NF-κB ligand (RANKL) expression was regulated by Wnt5a in periodontal ligament cells (PDLCs), and exosomal miR-1260b was found to target Wnt5a-mediated RANKL pathway and inhibit its osteoclastogenic activity. These results indicate that significant ability of the TNF-α-preconditioned GMSC-derived exosomes to regulate inflammation and osteoclastogenesis paves the way for establishment of a therapeutic approach for periodontitis.


Asunto(s)
Pérdida de Hueso Alveolar , Exosomas , Animales , Encía , Humanos , Macrófagos , Ratones , Osteoclastos , Factor de Necrosis Tumoral alfa
8.
Front Immunol ; 11: 709, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32373130

RESUMEN

Enamel matrix derivatives (EMDs)-based periodontal tissue regenerative therapy is known to promote healing with minimal inflammatory response after periodontal surgery, i. e., it promotes wound healing with reduced pain and swelling. It has also been reported that macrophages stimulated with amelogenin, a major component of EMD, produce various anti-inflammatory cytokines and growth factors. We previously found that stimulation of monocytes with murine recombinant M180 (rM180) amelogenin suppresses major histocompatibility complex class II (MHC II) gene expression using microarray analysis. However, the detailed molecular mechanisms for this process remain unclear. In the present study, we demonstrated that rM180 amelogenin selectively downmodulates the interferon gamma (IFNγ)-induced cell surface expression of MHC II molecules in macrophages and this mechanism mediated by rM180 appeared to be widely conserved across species. Furthermore, rM180 accumulated in the nucleus of macrophages at 15 min after stimulation and inhibited the protein expression of class II transactivator (CIITA) which controls the transcription of MHC II by IFNγ. In addition, reduced MHC II expression on macrophages pretreated with rM180 impaired the expression of T cell activation markers CD25 and CD69, T cell proliferation ability, and IL-2 production by allogenic CD4+ T lymphocytes in mixed lymphocyte reaction assay. The chromatin immunoprecipitation assay showed that IFNγ stimulation increased the acetylation of histone H3 lysine 27, which is important for conversion to euchromatin, as well as the trimethylation of histone H3 lysine 4 levels in the CIITA promoter IV (p-IV) region, but both were suppressed in the group stimulated with IFNγ after rM180 treatment. In conclusion, the present study shows that amelogenin suppresses MHC II expression by altering chromatin structure and inhibiting CIITA p-IV transcription activity, and attenuates subsequent T cell activation. Clinically observed acceleration of wound healing after periodontal surgery by amelogenin may be partially mediated by the mechanism elucidated in this study. In addition, the use of recombinant amelogenin is safe because it is biologically derived protein. Therefore, amelogenin may also be used in future as an immunosuppressant with minimal side effects for organ transplantation or MHC II-linked autoimmune diseases such as type I diabetes, multiple sclerosis, and rheumatoid arthritis, among others.


Asunto(s)
Amelogenina/farmacología , Regulación hacia Abajo/efectos de los fármacos , Eucromatina/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Interferón gamma/metabolismo , Macrófagos/inmunología , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/efectos de los fármacos , Transactivadores/genética , Amelogenina/genética , Animales , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células RAW 264.7 , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Células THP-1
9.
Arch Oral Biol ; 83: 241-251, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28822800

RESUMEN

OBJECTIVES: Amelogenin, the major component of the enamel matrix derivative (EMD), has been suggested as a bioactive candidate for periodontal regeneration. Apart from producing a regenerative effect on periodontal tissues, amelogenin has also been reported to have an anti-inflammatory effect. However, the precise molecular mechanisms underlying these effects remain unclear. In the present study, we examined the immunomodulatory effects of amelogenin on macrophages. DESIGN: Human phorbol 12-myristate 13-acetate (PMA)-differentiated U937 macrophages and CD14+ peripheral blood-derived monocytes (PBMC)-derived macrophages were stimulated with recombinant amelogenin (rM180). After performing a detailed microarray analysis, the effects of rM180 on macrophage phenotype and signal transduction pathways were evaluated by real-time polymerase chain reaction, enzyme-linked immunosorbent assay, confocal microscopy and flow cytometry. RESULTS: The microarray analysis demonstrated that rM180 increased the expression of anti-inflammatory genes in lipopolysaccharide (LPS)-challenged macrophages after 24h, while it temporarily up-regulated inflammatory responses at 4h. rM180 significantly enhanced the expression of M2 macrophage markers (CD163 and CD206). rM180-induced M2 macrophage polarisation was associated with morphological changes as well as vascular endothelial growth factor (VEGF) production. rM180 enhanced prostaglandin E2 (PGE2) expression, and the activation of the cAMP/cAMP-responsive element binding (CREB) signaling pathway was involved in amelogenin-induced M2 macrophage polarisation. Blocking of PGE2 signaling by indomethacin specifically abrogated rM180 with or without LPS-induced M2 shift in PBMC-derived macrophages. CONCLUSION: Amelogenin could reprogram macrophages into the anti-inflammatory M2 phenotype. It could therefore contribute to the early resolution of inflammation in periodontal lesions and provide a suitable environment for remodeling-periodontal tissues.


Asunto(s)
Amelogenina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Dinoprostona/fisiología , Macrófagos/efectos de los fármacos , Transducción de Señal/fisiología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Lipopolisacáridos , Análisis por Micromatrices , Microscopía Confocal , Fenotipo , Reacción en Cadena de la Polimerasa , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular/metabolismo
10.
J Cell Biochem ; 116(4): 628-39, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25399781

RESUMEN

Sprouty was identified as an inhibitor of the fibroblast growth factor (FGF) receptor, and Sprouty2 (Spry2) functions as a negative regulator of receptor tyrosine kinase signaling. In this study, we investigated how inhibition of Spry2 affects osteoblasts and gingival epithelial cells in periodontal tissue regeneration in vitro. Transduction of a dominant-negative mutant of Spry2 (Y55A-Spry2) enhanced basic fibroblast growth factor (bFGF)- and epidermal growth factor (EGF)-induced ERK activation in MC3T3-E1 osteoblastic cells. In contrast, it decreased their activation in GE1 cells. Consistent with these observations, Y55A-Spry2 increased osteoblast proliferation with bFGF and EGF stimulation, whereas the proliferation of Y55A-Spry2-introduced GE1 cells was decreased via the ubiquitination and degradation of EGF receptors (EGFRs). In addition, Y55A-Spry2 caused upregulation of Runx2 expression and downregulation of Twist, a negative regulator of Runx2, with treatment of bFGF and EGF, resulting in enhanced osteoblastogenesis accompanied by alkaline phosphatase activation and osteocalcin expression in MC3T3-E1 cells. These data suggest that suppression of Spry2 expression induces proliferation and differentiation of osteoblastic cells after the addition of a bFGF and EGF cocktail but inhibits proliferation in gingival epithelial cells. These in vitro experiments may provide a molecular basis for novel therapeutic approaches in periodontal tissue regeneration. Taken together, our study proposes that combined application of an inhibitor for tyrosine 55 of Spry2, bFGF, and EGF may effectively allow alveolar bone growth and block the ingrowth of gingival epithelial cells toward bony defects, biologically mimicking a barrier effect in guided tissue regeneration, with in vivo investigation in the future.


Asunto(s)
Células Epiteliales/fisiología , Encía/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Mutación , Osteoblastos/fisiología , Animales , Diferenciación Celular , Proliferación Celular , Factor de Crecimiento Epidérmico/farmacología , Factor 2 de Crecimiento de Fibroblastos/farmacología , Expresión Génica , Ratones , Proteínas Serina-Treonina Quinasas
11.
PLoS One ; 8(10): e78129, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24167599

RESUMEN

Emdogain (enamel matrix derivative, EMD) is well recognized in periodontology. It is used in periodontal surgery to regenerate cementum, periodontal ligament, and alveolar bone. However, the precise molecular mechanisms underlying periodontal regeneration are still unclear. In this study, we investigated the proteins bound to amelogenin, which are suggested to play a pivotal role in promoting periodontal tissue regeneration. To identify new molecules that interact with amelogenin and are involved in osteoblast activation, we employed coupling affinity chromatography with proteomic analysis in fractionated SaOS-2 osteoblastic cell lysate. In SaOS-2 cells, many of the amelogenin-interacting proteins in the cytoplasm were mainly cytoskeletal proteins and several chaperone molecules of heat shock protein 70 (HSP70) family. On the other hand, the proteomic profiles of amelogenin-interacting proteins in the membrane fraction of the cell extracts were quite different from those of the cytosolic-fraction. They were mainly endoplasmic reticulum (ER)-associated proteins, with lesser quantities of mitochondrial proteins and nucleoprotein. Among the identified amelogenin-interacting proteins, we validated the biological interaction of amelogenin with glucose-regulated protein 78 (Grp78/Bip), which was identified in both cytosolic and membrane-enriched fractions. Confocal co-localization experiment strongly suggested that Grp78/Bip could be an amelogenin receptor candidate. Further biological evaluations were examined by Grp78/Bip knockdown analysis with and without amelogenin. Within the limits of the present study, the interaction of amelogenin with Grp78/Bip contributed to cell proliferation, rather than correlate with the osteogenic differentiation in SaOS-2 cells. Although the biological significance of other interactions are not yet explored, these findings suggest that the differential effects of amelogenin-derived osteoblast activation could be of potential clinical significance for understanding the cellular and molecular bases of amelogenin-induced periodontal tissue regeneration.


Asunto(s)
Amelogenina/metabolismo , Diferenciación Celular/fisiología , Membrana Celular/metabolismo , Citosol/metabolismo , Proteínas de Choque Térmico/metabolismo , Osteoblastos/metabolismo , Proteómica , Amelogenina/genética , Animales , Línea Celular , Membrana Celular/genética , Chaperón BiP del Retículo Endoplásmico , Técnicas de Silenciamiento del Gen , Proteínas de Choque Térmico/genética , Humanos , Ratones , Osteoblastos/citología , Periodoncio/citología , Periodoncio/metabolismo , Regeneración/fisiología
12.
PLoS One ; 8(7): e67906, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874464

RESUMEN

Bisphosphonate-related osteonecrosis of the jaw (BRONJ) is a serious side effect of nitrogen-containing bisphosphonate (NBP) use. Many studies have shown that BRONJ is limited to the jawbone and does not occur in the other bones. We hypothesized that BRONJ is related to local bacterial iections and involves the innate immune system. To examine the relationship between BRONJ and innate immunity, we examined the effects of NBPs on macrophages, one of the important cell types in innate immunity. The expression of toll-like receptor-4 (TLR4) in cells after pretreatment with zoledronic acid (ZOL) did not considerably differ from that in untreated control cells. However, cytokine levels and nitric oxide (NO) production increased after pretreatment with ZOL. Furthermore, ZOL induced NF-κB activation by enhancing IκB-α degradation. Lipopolysaccharide (LPS)-induced apoptosis also increased after pretreatment with ZOL. This effect was mediated by a reduction of suppressor of cytokine signaling-1 (SOCS1), which is a negative regulator of myeloid differentiation primary response gene 88 (MyD 88)-dependent signaling. These results suggest that ZOL induced excessive innate immune response and proinflammatory cytokine production and that these processes may be involved in the bone destruction observed in BRONJ.


Asunto(s)
Difosfonatos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Imidazoles/farmacología , Lipopolisacáridos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Línea Celular , Citocinas/genética , Citocinas/metabolismo , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Activación de Macrófagos/efectos de los fármacos , Activación de Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Ratones , Factor 88 de Diferenciación Mieloide/metabolismo , FN-kappa B/metabolismo , Óxido Nítrico/biosíntesis , Proteína 1 Supresora de la Señalización de Citocinas , Receptor Toll-Like 4/metabolismo , Ácido Zoledrónico
13.
Genes Cells ; 10(9): 887-95, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16115197

RESUMEN

Caveolin-1 (Cav-1) has been suggested to function as a negative regulator of mitogen-stimulated proliferation and the Ras-p42/44 ERK (MAP kinase) pathway in a variety of cell types. However, the molecular basis of this suppression has not been clarified. Spred/Sprouty family proteins are also negative regulators of the ERK pathway by interacting with Raf-1. The Spred/Sprouty family proteins contain a cysteine-rich (CR) domain at the C-terminus, which is thought to be palmitoylated like Cav-1 and necessary for membrane anchoring. In this study, we demonstrated that Spred-1 localized in cholesterol-rich membrane raft/caveola fractions and interacted with Cav-1. To clarify the biological effect of Cav-1/Spred-1 interaction, we used hematopoietic cells that lacked expression of caveolins but expressed Spred-1. Forced expression of Cav-1 suppressed SCF- and IL-3-induced proliferation and ERK activation. Furthermore, forced expression of exogenous Spred-1 in Cav-1-expressing cells further suppressed proliferation and ERK activation. These data suggest that Spred-1 inhibits ERK activation in collaboration with Cav-1.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Proteínas Represoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Caveolas/metabolismo , Células Cultivadas , Activación Enzimática , Inmunoprecipitación , Microscopía Electrónica , Estructura Terciaria de Proteína , Proteínas Represoras/farmacología , Transducción de Señal , Fracciones Subcelulares , Factores de Tiempo , Transfección
14.
Nat Neurosci ; 8(7): 855-7, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15937482

RESUMEN

We report here that loss of the Sprouty2 gene (also known as Spry2) in mice resulted in enteric nerve hyperplasia, which led to esophageal achalasia and intestinal pseudo-obstruction. Glial cell line-derived neurotrophic factor (GDNF) induced hyperactivation of ERK and Akt in enteric nerve cells. Anti-GDNF antibody administration corrected nerve hyperplasia in Sprouty2-deficient mice. We show Sprouty2 to be a negative regulator of GDNF for the neonatal development or survival of enteric nerve cells.


Asunto(s)
Sistema Nervioso Entérico/patología , Acalasia del Esófago/genética , Eliminación de Gen , Factores de Crecimiento Nervioso/metabolismo , Neuronas/patología , Proteínas/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Anticuerpos/farmacología , Sistema Nervioso Entérico/metabolismo , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial , Hiperplasia , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Ratones , Ratones Noqueados , Factores de Crecimiento Nervioso/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt
15.
J Biol Chem ; 279(50): 52543-51, 2004 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-15465815

RESUMEN

Sprouty/Spred family proteins have been identified as negative regulators of growth factor-induced ERK/mitogen-activated protein (MAP) kinase activation. However, it has not been clarified whether these proteins regulate cytokine-induced ERK activity. We found that Spred-1 is highly expressed in interleukin-3 (IL-3)-dependent hematopoietic cell lines and bone marrow-derived mast cells. To investigate the roles of Spred-1 in hematopoiesis, we expressed wild-type Spred-1 and a dominant negative form of Spred-1, DeltaC-Spred, in IL-3- and stem cell factor (SCF)-dependent cell lines as well as hematopoietic progenitor cells from mouse bone marrow by retrovirus gene transfer. In IL-3-dependent Ba/F3 cells expressing c-kit, forced expression of Spred-1 resulted in a reduced proliferation rate and ERK activation in response to not only SCF but also IL-3. In contrast, DeltaC-Spred augmented IL-3-induced cell proliferation and ERK activation. Wild-type Spred-1 inhibited colony formation of bone marrow cells in the presence of cytokines, whereas DeltaC-Spred-1 expression enhanced colony formation. Augmentation of ERK activation and proliferation in response to IL-3 was also observed in Spred-1-deficient bone marrow-derived mast cells. These data suggest that Spred-1 negatively regulates hematopoiesis by suppressing not only SCF-induced but also IL-3-induced ERK activation.


Asunto(s)
Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interleucina-3/metabolismo , Proteínas Represoras/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Línea Celular , Ensayo de Unidades Formadoras de Colonias , Activación Enzimática , Expresión Génica , Hematopoyesis , Humanos , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Represoras/genética , Factor de Células Madre/metabolismo
17.
Nat Cell Biol ; 5(5): 427-32, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12717443

RESUMEN

The signalling cascade including Raf, mitogen-activated protein kinase (MAPK) kinase and extracellular-signal-regulated kinase (ERK) is important in many facets of cellular regulation. Raf is activated through both Ras-dependent and Ras-independent mechanisms, but the regulatory mechanisms of Raf activation remain unclear. Two families of membrane-bound molecules, Sprouty and Sprouty-related EVH1-domain-containing protein (Spred) have been identified and characterized as negative regulators of growth-factor-induced ERK activation. But the molecular functions of mammalian Sproutys have not been clarified. Here we show that mammalian Sprouty4 suppresses vascular epithelial growth factor (VEGF)-induced, Ras-independent activation of Raf1 but does not affect epidermal growth factor (EGF)-induced, Ras-dependent activation of Raf1. Sprouty4 binds to Raf1 through its carboxy-terminal cysteine-rich domain, and this binding is necessary for the inhibitory activity of Sprouty4. In addition, Sprouty4 mutants of the amino-terminal region containing the conserved tyrosine residue, which is necessary for suppressing fibroblast growth factor signalling, still inhibit the VEGF-induced ERK pathway. Our results show that receptor tyrosine kinases use distinct pathways for Raf and ERK activation and that Sprouty4 differentially regulates these pathways.


Asunto(s)
Membrana Celular/metabolismo , Células Eucariotas/metabolismo , Sustancias de Crecimiento/metabolismo , Sistema de Señalización de MAP Quinasas/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Proto-Oncogénicas c-raf/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Animales , Membrana Celular/efectos de los fármacos , Factores de Crecimiento Endotelial/metabolismo , Factores de Crecimiento Endotelial/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Células Eucariotas/citología , Células Eucariotas/efectos de los fármacos , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Péptidos y Proteínas de Señalización Intercelular/farmacología , Péptidos y Proteínas de Señalización Intracelular , Linfocinas/metabolismo , Linfocinas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Mutación/genética , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Estructura Terciaria de Proteína/efectos de los fármacos , Estructura Terciaria de Proteína/genética , Proteínas Proto-Oncogénicas c-raf/genética , Proteínas Tirosina Quinasas Receptoras/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Proteínas ras/genética , Proteínas ras/metabolismo
18.
Biochem Biophys Res Commun ; 302(4): 767-72, 2003 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-12646235

RESUMEN

We have reported on Spred-1 and Spred-2, which inhibit MAP kinase activation by interacting with c-kit and ras/raf. Here, we report the cloning of a third member in this family, Spred-3. Spred-3 is expressed exclusively in the brain and its gene locates in chromosome 19q13.13 in human. Like Spred-1 and -2, Spred-3 contains an EVH1 domain in the N-terminus and a Sprouty-related cysteine-rich region (SPR domain) in the C-terminus that is necessary for membrane localization. However, Spred-3 does not possess a functional c-kit binding domain (KBD), since the critical amino acid Arg residue in this region was replaced with Gly in Spred-3. Although Spred-3 suppressed growth factor-induced MAP kinase (Erk) activation, inhibitory activity of Spred-3 was lower than that of Spred-1 or Spred-2. By the analysis of chimeric molecules between Spred-3 and Spred-1, we found that the SPR domain, rather than KBD, is responsible for efficient Erk suppression. The finding of Spred-3 revealed the presence of a novel family of regulators for the Ras/MAP kinase pathway, each member of which may have different specificities for extracellular signals.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Cromosomas Humanos Par 19 , Clonación Molecular , Activación Enzimática , Humanos , Ratones , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas Represoras/química , Alineación de Secuencia , Distribución Tisular , Tirosina/metabolismo
19.
Artículo en Inglés | MEDLINE | ID: mdl-12424446

RESUMEN

OBJECTIVE: This study evaluated the effects of marsupialization on odontogenic keratocysts (OKCs) and its role in conjunction with enucleation and curettage. STUDY DESIGN: Twenty-eight primary OKCs, treated by marsupialization before enucleation and curettage, were examined in this study. The effect of marsupialization and recurrence data after a follow-up period of at least 3 years were evaluated. The changes of growth characteristics during marsupialization were analyzed by means of histopathologic and immunohistochemical studies with monoclonal anti-Ki-67 antibody. RESULTS: The effect of marsupialization was evaluated as extremely effective (64.3%), moderately effective (32.1%), and poorly effective (3.6%). In 5 lesions, the cysts disappeared macroscopically and further surgery was not done. Recurrence was observed in 6 lesions (21.4%), and there was no significant difference in recurrence rates between the lesions treated with or without marsupialization. There appeared to be a predilection for recurrence in the lesions in the mandibular ramus region and also for radiographically multilocular lesions. Microscopic examination showed substantial changes from a parakeratinized or orthokeratinized epithelium into a hyperplastic, stratified, nonkeratinizing squamous epithelium after marsupialization in many cases. There was no significant difference in labeling index between premarsupialization (20.2% +/- 12.0%) and postmarsupialization (15.3% +/- 10.3%). CONCLUSION: Marsupialization was found to be effective as a preliminary treatment for large OKCs. This procedure does not appear to affect the recurrence tendency of OKCs, and the probable changes in growth characteristics become rather less aggressive.


Asunto(s)
Enfermedades Mandibulares/cirugía , Quistes Odontogénicos/cirugía , Procedimientos Quirúrgicos Orales/métodos , Adolescente , Adulto , Anciano , Niño , Descompresión Quirúrgica/métodos , Femenino , Estudios de Seguimiento , Humanos , Inmunohistoquímica , Antígeno Ki-67/análisis , Masculino , Enfermedades Mandibulares/fisiopatología , Enfermedades Maxilares/fisiopatología , Enfermedades Maxilares/cirugía , Persona de Mediana Edad , Quistes Odontogénicos/fisiopatología , Recurrencia , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA