Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Int. j. morphol ; 42(4): 1080-1095, ago. 2024. ilus, tab
Article in English | LILACS | ID: biblio-1569275

ABSTRACT

SUMMARY: Marein is a flavonoid compound that reduces blood glucose and lipids and has a protective effect in diabetes. However, the effect and mechanism(s) of marein on renal endothelial-mesenchymal transition in diabetic kidney disease (DKD) have not been elucidated. In this study, single-cell sequencing data on DKD were analyzed using a bioinformation method, and the data underwent reduced dimension clustering. It was found that endothelial cells could be divided into five subclusters. The developmental sequence of the subclusters was 0, 1, 4, 2, and 3, of which subcluster 3 had the most interstitial phenotype.The expression of mesenchymal marker protein:Vimentin(VIM), Fibronectin(FN1), and fibroblast growth factor receptor 1 (FGFR1) increased with the conversion of subclusters. In db/db mice aged 13-14 weeks, which develop DKD complications after 8-12 weeks of age, marein reduced blood levels of glucose, creatinine, and urea nitrogen, improved structural damage in kidney tissue, and reduced collagen deposition and the expression of FN1 and VIM. Marein also up-regulated autophagy marker:Light chain 3II/I(LC3II/I) and decreased FGFR1 expression in renal tissue. In an endothelial-mesenchymal transition model, a high glucose level induced a phenotypic change in human umbilical vein endothelial cells. Marein decreased endothelial cell migration, improved endothelial cell morphology, and decreased the expression of VIM and FN1. The use of the FGFR1 inhibitor, AZD4547, and autophagy inhibitor, 3-Methyladenine(3-MA), further demonstrated the inhibitory effect of marein on high glucose-induced endothelial-mesenchymal transition by reducing FGFR1 expression and up-regulating the autophagy marker protein, LC3II/I. In conclusion, this study suggests that marein has a protective effect on renal endothelial- mesenchymal transition in DKD, which may be mediated by inducing autophagy and down-regulating FGFR1 expression.


La mareína es un compuesto flavonoide que reduce la glucosa y los lípidos en sangre y tiene un efecto protector en la diabetes. Sin embargo, no se han dilucidado el efecto y los mecanismos de la mareína sobre la transición endotelial- mesenquimatosa renal en la enfermedad renal diabética (ERD). En este estudio, los datos de secuenciación unicelular sobre DKD se analizaron utilizando un método de bioinformación y los datos se sometieron a una agrupación de dimensiones reducidas. Se descubrió que las células endoteliales podían dividirse en cinco subgrupos. La secuencia de desarrollo de los subgrupos fue 0, 1, 4, 2 y 3, de los cuales el subgrupo 3 tenía el fenotipo más intersticial. La expresión de la proteína marcadora mesenquimatosa: vimentina (VIM), fibronectina (FN1) y receptor del factor de crecimiento de fibroblastos. 1 (FGFR1) aumentó con la conversión de subgrupos. En ratones db/db de 13 a 14 semanas de edad, que desarrollan complicaciones de DKD después de las 8 a 12 semanas de edad, la mareína redujo los niveles sanguíneos de glucosa, creatinina y nitrógeno ureico, mejoró el daño estructural en el tejido renal y redujo la deposición y expresión de colágeno de FN1 y VIM. Marein también aumentó el marcador de autofagia: Cadena ligera 3II/I (LC3II/I) y disminuyó la expresión de FGFR1 en el tejido renal. En un modelo de transición endotelial-mesenquimal, un nivel alto de glucosa indujo un cambio fenotípico en las células endoteliales de la vena umbilical humana. Marein disminuyó la migración de células endoteliales, mejoró la morfología de las células endoteliales y disminuyó la expresión de VIM y FN1. El uso del inhibidor de FGFR1, AZD4547, y del inhibidor de la autofagia, 3-metiladenina (3-MA), demostró aún más el efecto inhibidor de la mareína en la transición endotelial-mesenquimal inducida por niveles altos de glucosa al reducir la expresión de FGFR1 y regular positivamente la proteína marcadora de autofagia. , LC3II/I. En conclusión, este estudio sugiere que la mareína tiene un efecto protector sobre la transición endotelial-mesenquimatosa renal en la ERC, que puede estar mediada por la inducción de autofagia y la regulación negativa de la expresión de FGFR1.


Subject(s)
Chalcones/pharmacology , Diabetic Nephropathies/drug therapy , Endothelial-Mesenchymal Transition , Autophagy , Computational Biology , Receptor, Fibroblast Growth Factor, Type 1
2.
Article in English | MEDLINE | ID: mdl-38760286

ABSTRACT

Tenosynovial giant cell tumor is a benign neoplasm arising from the synovium of joints, including the temporomandibular joint (TMJ). Despite its benign nature, these tumors may exhibit aggressive behavior. A 57-year-old woman with a swollen, hardened area in the left TMJ was referred to the university´s clinic. The diagnosis of tenosynovial giant cell tumor was made based on the presence of hyperplastic synovial lining containing mononuclear and giant cells, hemorrhagic areas, hemosiderin deposits, and calcification foci in the biopsy. A low condylectomy was performed, and histopathologic analysis of the surgical piece upheld the diagnosis. Due to histopathologic resemblance with other giant cell-rich lesions (giant cell granuloma of the jaws, brown tumor of hyperparathyroidism, and non-ossifying fibroma) for which signature mutations are known, mutational analysis of KRAS, FGFR1, and TRPV4 genes was conducted. The results revealed wild-type sequences for all the mutations tested, thereby supporting the diagnosis of tenosynovial giant cell tumor.


Subject(s)
Giant Cell Tumor of Tendon Sheath , Humans , Female , Middle Aged , Giant Cell Tumor of Tendon Sheath/pathology , Giant Cell Tumor of Tendon Sheath/genetics , Giant Cell Tumor of Tendon Sheath/surgery , Diagnosis, Differential , Biopsy , Temporomandibular Joint Disorders/pathology , Temporomandibular Joint Disorders/surgery , Temporomandibular Joint Disorders/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , DNA Mutational Analysis , Proto-Oncogene Proteins p21(ras)
3.
Rev. Esc. Enferm. USP ; Rev. Esc. Enferm. USP;58: e20230183, 2024. tab, graf
Article in English | LILACS, BDENF - Nursing | ID: biblio-1565123

ABSTRACT

ABSTRACT Objective: To observe the therapeutic effect of gentiopicroside, as the main component of Gentianaceae, on wounds in pressure injury (PI) model rats and explore its mechanism. Method: Male Sprague Dawley rats were randomly divided into control group, model group and gentiopicroside groups (50, 100 and 200 mg·kg-1·d-1 for 9 consecutive days). The mice's skeletal muscle fibroblast line NOR-10 cells were collected after being treated with gentiopicroside (0.2~5.0 M) and basic fibroblast growth factor receptor 1 (bFGFR1) inhibitor (5.0 M SU5402) for 7 days. Results: Compared to the model group, the gentiopicroside groups showed significantly increased wound healing rates, reduced inflammatory cells in the wound tissues, and significantly increased expression levels of proliferating cell nuclear antigen (PCNA) and bFGFR1, accompanied by increased proliferation of new myofibroblasts. Gentiopicroside upregulated the mRNA expression of bFGFR1 and PCNA in NOR-10 cells in a dose-dependent manner; however, SU5402 reversed the effect of gentiopicroside. Conclusion: Gentiopicroside may promote myofibroblast proliferation by upregulating the expression of bFGFR1 and PCNA and ultimately accelerating the healing of PI wounds.


RESUMO Objetivo: Observar o efeito terapêutico do gentiopicrosídeo como principal componente das Gentianáceas em feridas de lesão por pressão (LP) em modelos de ratos e explorar seu mecanismo. Métodos: Ratos Sprague Dawley machos foram divididos aleatoriamente em grupo controle, grupo modelo e grupos gentiopicrosídeo (50, 100 e 200 mg·kg-1·d-1 por 9 dias consecutivos). As células NOR-10 da linha de fibroblastos do músculo esquelético de camundongos foram coletadas após serem tratadas com gentiopicrosídeo (0,2~5,0 μM) e inibidor do receptor 1 do fator de crescimento fibroblástico básico (bFGFR1) (5.0 μM SU5402) por 7 dias. Resultados: Em comparação com o grupo modelo, os grupos gentiopicrosídeo apresentaram taxas de cicatrização de feridas significativamente maiores, menos células inflamatórias nos tecidos da ferida e níveis de expressão de antígeno nuclear de proliferação celular (PCNA) e bFGFR1 significativamente maiores, acompanhados por aumento da proliferação de novos miofibroblastos. O gentiopicrosídeo regulou positivamente a expressão de mRNA de bFGFR1 e PCNA em células NOR-10 de maneira dependente da dose, enquanto o SU5402 reverteu o efeito do gentiopicrosídeo. Conclusão: O gentiopicrosídeo pode promover a proliferação de miofibroblastos, suprarregulando a expressão de bFGFR1 e PCNA e, em última análise, acelerando a cicatrização de feridas de LP.


RESUMEN Objetivo: Observar el efecto terapéutico del gentiopicrósido como componente principal de la Gentianaceae en heridas por lesión por presión (LP) en modelos de ratas y explorar su mecanismo. Método: Se dividieron aleatoriamente ratas macho Sprague Dawley en grupo control, grupo modelo y grupos gentiopicrósido (50, 100 y 200 mg·kg-1·d-1 durante 9 días consecutivos). Se recogieron células NOR-10 de la línea de fibroblastos de músculo esquelético de ratón después de ser tratadas con gentiopicrósido (0.2~5.0 μM) y un inhibidor del receptor 1 del factor de crecimiento de fibroblastos básico (bFGFR1) (5.0 μM SU5402) durante 7 días. Resultados: En comparación con el grupo modelo, los grupos de gentiopicrósido mostraron tasas de curación de heridas significativamente más altas, menos células inflamatorias en los tejidos de la herida y niveles de expresión significativamente mayores del antígeno nuclear de proliferación celular (PCNA) y bFGFR1, acompañados de una mayor proliferación de nuevos miofibroblastos. El gentiopicrósido podría regular positivamente la expresión de ARNm de bFGFR1 y PCNA en células NOR-10 de manera dependiente de la dosis, sin embargo, SU5402 revirtió el efecto del gentiopicrósido. Conclusión: El gentiopicrósido puede promover la proliferación de miofibroblastos al aumentar la expresión de bFGFR1 y PCNA y, en última instancia, acelerar la cicatrización de las heridas de LP.


Subject(s)
Humans , Wound Healing , Pressure Ulcer , Proliferating Cell Nuclear Antigen , Gentianaceae , Receptor, Fibroblast Growth Factor, Type 1
4.
J Med Case Rep ; 17(1): 39, 2023 Jan 26.
Article in English | MEDLINE | ID: mdl-36698221

ABSTRACT

BACKGROUND: Myeloproliferative neoplasms are a group of diseases with diverse biological and clinical characteristics. As a provisional separate entity, myeloid/lymphoid neoplasms with eosinophilia and genetic rearrangement have been described, which may present an initial clinical behavior of myeloproliferation and be characterized by varied genetic rearrangements. One of these entities is associated with FGFR1 rearrangements, characterized by its low prevalence and few treatment options. CASE PRESENTATION: We present the case of a 53-year-old Mestizo male patient of Hispanic origin who initially presented weight loss and fatigue, with a complete blood count showing leukocytosis and eosinophilia, with an initial diagnosis of nonspecific myeloproliferative disorder. In a next-generation sequencing study, BCR::FGFR1 rearrangement was documented, a diagnosis of myeloid/lymphoid neoplasia with eosinophilia and BCR::FGFR1 rearrangement was made, and hydroxyurea therapy was initiated. Subsequently, transformation to cortical T-lymphoblastic leukemia/lymphoma and erythroid precursors was documented, requiring management with chemotherapy. CONCLUSIONS: Myeloid/lymphoid neoplasms with eosinophilia and genetic rearrangements constitute a group of deeply heterogeneous diseases with variable clinical and diagnostic characteristics and whose treatment is not clearly defined.


Subject(s)
Eosinophilia , Lymphoma, Non-Hodgkin , Lymphoma , Myeloproliferative Disorders , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Male , Middle Aged , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Eosinophilia/genetics , Gene Rearrangement , Lymphoma, Non-Hodgkin/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics
5.
Brain Res ; 1803: 148234, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36634900

ABSTRACT

Diabetes mellitus (DM) is a chronic metabolic disease, mainly characterized by increased blood glucose and insulin dysfunction. In response to the persistent systemic hyperglycemic state, numerous metabolic and physiological complications have already been well characterized. However, its relationship to bone fragility, cognitive deficits and increased risk of dementia still needs to be better understood. The impact of chronic hyperglycemia on bone physiology and architecture was assessed in a model of chronic hyperglycemia induced by a single intraperitoneal administration of streptozotocin (STZ; 55 mg/kg) in Wistar rats. In addition, the bone-to-brain communication was investigated by analyzing the gene expression and methylation status of genes that encode the main osteokines released by the bone [Fgf23 (fibroblast growth factor 23), Bglap (bone gamma-carboxyglutamate protein) and Lcn2 (lipocalin 2) and their receptors in both, the bone and the brain [Fgfr1 (fibroblast growth factor receptor 1), Gpr6A (G-protein coupled receptor family C group 6 member A), Gpr158 (G protein-coupled receptor 158) and Slc22a17 (Solute carrier family 22 member 17)]. It was observed that chronic hyperglycemia negatively impacted on bone biology and compromised the balance of the bone-brain endocrine axis. Ultrastructural disorganization was accompanied by global DNA hypomethylation and changes in gene expression of DNA-modifying enzymes that were accompanied by changes in the methylation status of the osteokine promoter region Bglap and Lcn2 (lipocalin 2) in the femur. Additionally, the chronic hyperglycemic state was accompanied by modulation of gene expression of the osteokines Fgf23 (fibroblast growth factor 23), Bglap (bone gamma-carboxyglutamate protein) and Lcn2 (lipocalin 2) in the different brain regions. However, transcriptional regulation mediated by DNA methylation was observed only for the osteokine receptors, Fgfr1(fibroblast growth factor receptor 1) in the striatum and Gpr158 (G protein-coupled receptor 158) in the hippocampus. This is a pioneer study demonstrating that the chronic hyperglycemic state compromises the crosstalk between bone tissue and the brain, mainly affecting the hippocampus, through transcriptional silencing of the Bglap receptor by hypermethylation of Gpr158 gene.


Subject(s)
Fibroblast Growth Factor-23 , Hyperglycemia , Receptors, G-Protein-Coupled , Animals , Rats , 1-Carboxyglutamic Acid/genetics , 1-Carboxyglutamic Acid/metabolism , Bone and Bones/metabolism , Brain/metabolism , Epigenetic Repression , Hippocampus/metabolism , Homeostasis , Hyperglycemia/metabolism , Lipocalin-2/metabolism , Osteocalcin/genetics , Osteocalcin/metabolism , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptors, G-Protein-Coupled/metabolism
6.
J Oral Pathol Med ; 52(2): 119-126, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36565263

ABSTRACT

BACKGROUND: Fibroblast growth factor receptor 1 is a potential prognostic factor for tongue squamous cell carcinoma and is associated with oral epithelial dysplasia grade in oral leukoplakia. METHODS: Thirty cases of tongue squamous cell carcinoma and 30 cases of oral leukoplakia were analyzed. Fibroblast growth factor receptor 1 and phosphorylated Akt protein expression were analyzed by immunohistochemistry and quantified using a digital algorithm. Fibroblast growth factor receptor 1 gene amplification was analyzed by fluorescent in situ hybridization in the tongue squamous cell carcinoma cases. RESULTS: Clinical appearance and dysplasia grade were correlated with oral leukoplakia malignant transformation. Oral leukoplakia cases presenting high fibroblast growth factor receptor 1 expression showed a higher risk of malignant transformation (p = 0.016, HR: 7.3, 95% CI: 1.4-37.4). Phosphorylated Akt showed faint to no expression in oral leukoplakia, which did not correlate with dysplasia grade or malignant transformation. High expression of fibroblast growth factor receptor 1 and phosohorylated Akt were associated with poor overall survival and disease-free survival in tongue squamous cell carcinoma, although only fibroblast growth factor receptor 1 expression was significantly associated with poor overall survival (p = 0.024; HR: 4.9, 95% CI: 1.2-19.9). Cases presenting double fibroblast growth factor receptor 1/phosphorylated Akt overexpression (n = 8) showed markedly impaired overall survival (p = 0.020; HR: 6.4, 95% CI: 1.3-31.1) and disease-free survival (p = 0.001, HR: 13.0, 95% CI: 3.0-55.7). Fibroblast growth factor receptor 1 amplification was observed in 16.6% of tongue squamous cell carcinoma cases, being correlated with vascular and neural invasion (p = 0.001 and 0.017, respectively), but not with fibroblast growth factor receptor 1 protein expression, overall survival, or disease-free survival. CONCLUSION: Fibroblast growth factor receptor 1 protein expression is an important prognostic factor in oral leukoplakia and tongue squamous cell carcinoma.


Subject(s)
Carcinoma, Squamous Cell , Tongue Neoplasms , Humans , Carcinoma, Squamous Cell/pathology , Tongue Neoplasms/pathology , Prognosis , Receptor, Fibroblast Growth Factor, Type 1/genetics , In Situ Hybridization, Fluorescence , Proto-Oncogene Proteins c-akt/genetics , Leukoplakia, Oral/pathology , Tongue/pathology
7.
s.l; CONETEC; 30 oct. 2022.
Non-conventional in Spanish | BRISA/RedTESA | ID: biblio-1400072

ABSTRACT

INTRODUCCIÓN: Los colangiocarcinomas abarcan todos los tumores que se originan en el epitelio de la vía biliar. Más del 90 % son adenocarcinomas y generalmente se clasifican como intrahepáticos o extrahepático. La mediana de edad en el momento del diagnóstico es de 65 años, y tasas de sobrevida relativa a los 5 años son cercanas al 10%. La resección completa con márgenes negativos es el único tratamiento potencialmente curativo para pacientes con enfermedad resecable. Sin embargo, la mayoría de los pacientes son diagnosticados con enfermedad no resecable, localmente avanzada o metastásica debido a la ausencia de síntomas hasta un momento posterior del curso de la enfermedad. Para los pacientes con estadio avanzado o irresecable y de buen estado general, el tratamiento de primera línea estándar es gemcitabina y cisplatino. En el entorno de segunda línea una vez que la enfermedad ha progresado el esquema de quimioterapia mFOLFOX (ácido folínico o leucovorina, fluorouracilo y oxaliplatino) es el régimen de tratamiento preferido. El perfil molecular de los colangiocarcinomas desempeña un papel cada vez más importante en la determinación del pronóstico y la selección de tratamientos. Recientemente se han identificado alteraciones genéticas del receptor del factor de crecimiento fibroblástico (FGFR, su sigla del inglés Fibroblast Growth Factor Receptor) en aproximadamente en un 10-16% de los colangiocarcinomas intrahepáticos y se han denominado indistintamente traslocaciones, fusiones o reordenamientos. Estas fusiones o reordenamientos rara vez se producen en los colangiocarcinomas extra-hepáticos. TECNOLOGÍA: Pemigatinib (Pemazyre®) es un inhibidor de la cinasa de FGFR1, 2 y 3 que inhibe la fosforilación y la señalización de FGFR y disminuye la viabilidad celular en las células que expresan alteraciones genéticas de FGFR, incluyendo mutaciones puntuales, amplificaciones y fusiones o reordenamientos. OBJETIVO: El objetivo del presente informe es evaluar rápidamente los parámetros de eficacia, seguridad, costos y recomendaciones disponibles acerca del empleo de pemigatinib en el tratamiento de pacientes con colangiocarcinoma. MÉTODOS: Se realizó una búsqueda bibliográfica en las principales bases de datos tales como PUBMED, LILACS, BRISA, COCHRANE, SCIELO, EMBASE, TRIPDATABASE como así también en sociedades científicas, agencias reguladoras, financiadores de salud y agencias de evaluación de tecnologías sanitarias. Se priorizó la inclusión de revisiones sistemáticas, ensayos clínicos controlados aleatorizados, evaluación de tecnología sanitaria y guías de práctica clínica de alta calidad metodológica. En PubMed se utilizó la estrategia de búsqueda que se detalla en el Anexo I. La fecha de búsqueda de información fue hasta el 24 de octubre de 2022. Para la búsqueda en Pubmed se utilizó la siguiente estrategia de búsqueda: (pemigatinib [Supplementary Concept] OR pemigatinib [tiab] OR Pemazyre [tiab] OR INCB054828 [tiab]) AND (Cholangiocarcinoma [MESH] OR cholangiocarcinoma [tiab]). RECOMENDACIONES: Las guías de la Red Nacional de Centros para el Tratamiento Integral del cáncer (NCCN, su sigla del inglés National Comprehensive Cancer Network) de los Estados Unidos, menciona al pemigatinib como una de las opciones de tratamiento para el colangiocarcinoma irresecable o metastásico con fusiones o reordenamientos de FGFR2, después de la progresión de la enfermedad a una primera línea de tratamento. La Sociedad Española de Oncología Médica (SEOM) recomienda que los pacientes candidatos al tratamiento deben enrolarse en ensayos clínicos. Añade que cuando esté disponible el fármaco en el país, se debería realizar una caracterización molecular exhaustiva del tumor en aquellos pacientes con estadios avanzado; ya que ciertas alteraciones genéticas, como las mutaciones de IDH1, las fusiones de FGFR2, mutaciones del gen BRAF, la inestabilidad de los microsatélites y los reordenamientos de NTRK, entre otros, pueden beneficiarse de terapias dirigidas específicas. La valoración del beneficio clínico según la de la Sociedad Europea de Oncología Médica (ESMO, su sigla del inglés European Society for Medical Oncology) para considerar la relevancia del beneficio clínico de forma sistematizada para este fármaco en la indicación evaluada es de 3 puntos. Según esta escala las calificaciones más altas en el ámbito no curativo son 5 y 4, que indican una magnitud sustancial del beneficio clínico. En Argentina, la Asociación de Oncología Clínica (AAOC) en sus recomendaciones actuales para el tratamiento oncológico del cáncer de vía biliar no menciona su uso dentro de las opciones terapêuticas. CONCLUSIONES: La eficacia y seguridad de pemigatinib frente a otros fármacos disponibles en la indicación evaluada no pudo ser establecida debido a que no se encontraron estudios comparativos. La evidencia que sustenta la aprobación de comercialización de pemigatinib para el tratamiento de adultos con colangiocarcinoma avanzado progresados a una primera línea de tratamiento y con alteraciones moleculares en el receptor del factor de crecimiento de fibroblastos 2, se basa en un único ensayo clínico no aleatorizado en pocos participantes. Este estudio mostró que aquellos adultos que utilizaron el tratamiento tendrían una tasa de respuesta cercanas al 37% al mediano plazo. Las agencias regulatorias relevadas han autorizado su comercialización de forma acelerada, bajo la denominación de medicamento huérfano, y sujeta a un ensayo de confirmación por parte del productor de la tecnología. Una guía de la Asociación Argentina de Oncología Clínica actualizada no lo menciona dentro de las opciones actuales de tratamiento, mientras que el resto de las recomendaciones provenientes de países de altos ingresos relevados lo mencionan como una opción terapéutica. Reino Unido brinda cobertura en la indicación evaluada sujeta a un acuerdo de comercialización con un descuento en el precio de venta. Otros países, como España y Canadá, no han autorizado su cobertura debido a que consideran que la evidencia que sustenta su utilización es insuficiente y existe incertidumbre sobre su beneficio en comparación con otras alternativas terapéuticas disponibles. No se hallaron evaluaciones económicas publicadas, aunque el costo del fármaco es muy elevado.


Subject(s)
Humans , Cholangiocarcinoma/drug therapy , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Argentina , Efficacy , Cost-Benefit Analysis
8.
Clin Transl Oncol ; 24(12): 2453-2465, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36002765

ABSTRACT

PURPOSE: To investigate the role and mechanism of TNF-inducible protein 3(TNFAIP3) in breast cancer angiogenesis induced by fibroblast growth factor receptor1 (FGFR1) activation. METHODS: The immunohistochemical assay was used to detect the expression of vascular endothelial cell marker CD31 and CD105 in mice DCIS.COM-iFGFR1 transplanted tumor (previously established by our group). The effects of TNFAIP3 knockout/knockdown breast cancer cell lines on angiogenesis, migration, and invasion of Human Umbilical Vein Endothelial Cells (HUVEC) were detected by the tubulogenesis and Trewells assay. RNA-seq analysis of TNFAIP3 downstreams differential genes after TNFAIP3 knockdown. The expression and secretion of VEGFA after FGFR1 activation in breast cancer cells were detected by qPCR, Western blot, and ELISA. RESULTS: Immunohistochemistry showed that TNFAIP3 knockout inhibited the expression of CD31 and CD105 in DCIS grafted tumors promoted by FGFR1 activation. Tubulogenesis and Trewells experiments showed that TNFAIP3 gene knockout/knockdown inhibited the angiogenesis, migration, and invasion of HUVEC cells promoted by FGFR1 activation. qPCR assay showed that VEGFA mRNA level in the TNFAIP3 knockdown cell line was significantly down-regulated (p < 0.05). qPCR, Western blot and ELISA results showed that TNFAIP3 gene knockout/knockdown could inhibit the expression and secretion of VEGFA in breast cancer cells induced by FGFR1 activation. CONCLUSION: TNFAIP3 promotes breast cancer angiogenesis induced by FGFR1 activation through the expression and secretion of VEGFA.


Subject(s)
Breast Neoplasms , Carcinoma, Intraductal, Noninfiltrating , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Female , Fibroblast Growth Factors , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Mice , Neovascularization, Pathologic , RNA, Messenger , Receptor, Fibroblast Growth Factor, Type 1 , Tumor Necrosis Factor alpha-Induced Protein 3 , Vascular Endothelial Growth Factor A/metabolism
9.
Arch Oral Biol ; 135: 105372, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35151029

ABSTRACT

OBJECTIVE: To evaluate previous nonsyndromic cleft lip with or without cleft palate (NSCL±P) associated signals in 4p16.2, 8p11.23, 12q13.13, 12q13.2 and 17q21.32 in a multiethnic Brazilian cohort. DESIGN: The single nucleotide polymorphisms (SNPs) rs34246903 in 4p16.2, rs13317 in 8p11.23 (FGFR1, fibroblast growth factor receptor 1), rs3741442 in 12q13.13, rs705704 in 12q13.2 and rs4968247 in 17q21.32 were genotyped with TaqMan allelic discrimination assays in a case-control sample including 801 NSCL±P patients [233 nonsyndromic cleft lip (NSCLO) and 568 nonsyndromic cleft lip and palate (NSCLP)] and 881 healthy controls. Multiple logistic regression analyses, considering sex and genomic ancestry as covariates, were conducted, and the p value was adjusted with Bonferroni multiple correction testing (p ≤ 0.01). RESULTS: Although several associations were identified, those that resisted the multiple correction testing involved the alleles and genotypes of rs34246903 and rs13317. The NSCLO group had a lower frequency of the minor C allele of rs34246903 compared to controls, giving an odds ratio (OR) of 0.74 [95% confidence interval (CI): 0.59-0.93, p = 0.01]. The rs34246903 CC genotype (homozygous) and the recessive model revealed significant protective associations with NSCLO, yielding ORs of 0.50 (95% CI: 0.29-0.85, p = 0.005) and 0.55 (95% CI: 0.33-0.93, p = 0.01) respectively. The presence of C variant allele of rs13317 (OR: 0.81, 95% CI: 0.69-0.96, p = 0.01) as well the TC genotype (OR: 0.77, 95% CI: 0.62-0.94, p = 0.01) and the dominant model (OR: 0.77, 95% CI: 0.63-0.94, p = 0.009) showed significant associations with reduced risk of NSCL±P. CONCLUSION: Our study is the first to support the association of rs34246903 (4p16.2) with NSCLO and rs13317 within FGFR1 with NSCL±P in the highly admixed Brazilian population. Further studies are needed to determine the functionality of those SNPs or to identify the causal markers in linkage disequilibrium with those susceptibility markers.


Subject(s)
Cleft Lip , Cleft Palate , Receptor, Fibroblast Growth Factor, Type 1/genetics , Alleles , Brazil , Case-Control Studies , Cleft Lip/genetics , Cleft Palate/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Genotype , Humans , Polymorphism, Single Nucleotide
10.
Clin Transl Oncol ; 23(4): 874-881, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32880048

ABSTRACT

PURPOSE: Endocrine therapy is a mainstay for the treatment of hormone receptor-positive breast cancer (BC); however, only a fraction of patients experience a pronounced response to antagonists of estrogen signaling. There is a need to identify predictors for efficacy of this treatment. METHODS: This study included 138 patients with newly diagnosed metastatic BC, who received upfront endocrine therapy. Archival biopsy specimens were tested for CCND1 and FGFR1 gene amplification and mRNA expression by PCR-based methods. RESULTS: CCND1 and FGFR1 amplification was detected in 24 (17.9%) and 28 (20.9%) of 134 evaluable cases, respectively; 9 carcinomas had concurrent alterations of these two genes. Presence of amplification in at least one locus was more common in tumors of higher grade (p = 0.018) and was associated with higher Ki-67 proliferation index (p = 0.036). CCND1 gene amplification was associated with shorter progression-free survival (PFS) in patients receiving aromatase inhibitors (AI) [16.0 months vs. 32.4 months, HR = 3.16 (95% CI 1.26-7.93), p = 0.014]. FGFR1 status did not significantly affect PFS of AI-treated women; however, objective response to AI was observed less frequently in FGFR1-amplified BC as compared to cases with normal FGFR1 copy number [2/15 (13.3%) vs. 22/46 (47.8%), p = 0.031]. Meanwhile, CCND1/FGFR1 gene status did not influence the outcome of tamoxifen-treated patients. CONCLUSION: Presence of CCND1 and/or FGFR1 amplification is associated with worse outcomes of AI therapy in patients with metastatic BC.


Subject(s)
Aromatase Inhibitors/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cyclin D1/genetics , Gene Amplification , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Cell Proliferation , Drug Resistance, Neoplasm , Female , Humans , Kaplan-Meier Estimate , Ki-67 Antigen/metabolism , Middle Aged , Progression-Free Survival , RNA, Messenger/metabolism , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Tamoxifen/therapeutic use , Treatment Outcome
11.
Rev. ADM ; 77(6): 287-294, nov.-dic. 2020. ilus, tab
Article in Spanish | LILACS | ID: biblio-1150870

ABSTRACT

Introducción: El carcinoma oral de células escamosas (COCE) es una neoplasia epitelial maligna que se presenta frecuentemente entre la quinta y sexta década de la vida. Su compleja patogénesis incluye el proceso de angiogénesis y la regulación del microambiente tumoral como mecanismos de progresión tumoral. Objetivo: Determinar la relación entre las variables clínicas e histológicas del COCE con la inmunoexpresión de VEGF, FGF-1, FGFR-1, TGFB-1, TGFBR-II y CD105. Material y métodos: Nueve casos de COCE; tres bien (BD), tres moderado (MD) y tres pobremente diferenciados (PD) obtenidos del Departamento de Patología y Medicina Bucal, División de Estudios de Postgrado e Investigación. Se aplicó la técnica de inmunohistoquímica por peroxidasa para identificar la expresión de VEGF, FGF-1, FGFR- 1, TGFB-1, TGFBR-II y CD105. El análisis de inmunoexpresión se realizó con el programa ImageJ. Se aplicó la prueba de Kruskal-Wallis y correlación de Spearman (p < 0.05). Resultados: La inmunoexpresión de VEGF fue mayor en los COCE PD, FGFR-1 fue positivo en los BD, mientras que FGF, TGFB-1 y TGFBR-II fueron negativos. El análisis de microdensidad vascular (MVD) indicó mayor número de vasos CD105 positivos en los carcinomas BD, seguidos de los PD y MD. Conclusión: Considerando los resultados obtenidos podemos concluir que la angiogénesis es un fenómeno constante independiente del grado de diferenciación que durante el proceso de transformación de una neoplasia requerirá la formación de vasos sanguíneos y que este proceso puede ser modulado por factores de crecimiento tales como los analizados en este trabajo (AU)


Introduction: Oral squamous cell carcinoma (OSCC) is a malignant epithelial neoplasm that frequently occurs between the fifth and sixth decade of life. Its complex pathogenesis includes the angiogenesis process and the regulation of the tumor microenvironment as mechanisms of tumor progression. Objective: To determine the relationship between the clinical and histological variables of OSCC with the immunoexpression of VEGF, FGF-1, FGFR-1, TGFB- 1, TGFBR-II and CD105. Material and methods: Nine cases of OSCC; three well (WD), three moderate (MD) and three poorly differentiated (PD) obtained from the Oral Medicine and Pathology Department, Division of Graduate Studies and Research. The peroxidase immunohistochemistry technique was performed to identify the expression of VEGF, FGF-1, FGFR-1, TGFB-1, TGFBR-II and CD105. The immunoexpression analysis was performed with the ImageJ software. The Kruskal-Wallis and Spearman correlation test were performed (p < 0.05). Results: VEGF immunoexpression was higher in PD OSCC, while FGFR-1 was predominantly positive in WD; FGF, TGFB-1 and TGFBR-II were negative. Vascular microdensity analysis (MVD) indicated a greater number of CD105 positive vessels in WD carcinomas, followed by PD and MD. Conclusion: Considering the results obtained, we can conclude that angiogenesis is a constant phenomenon independent of the degree of differentiation; that during the transformation process of a neoplasm it will require the formation of blood vessels and that this process can be modulated by growth factors such as those analyzed in this work (AU)


Subject(s)
Humans , Male , Female , Adult , Middle Aged , Aged , Carcinoma, Squamous Cell/immunology , Fibroblast Growth Factor 1 , Vascular Endothelial Growth Factor A , Blood Vessels , Immunohistochemistry , Histological Techniques , Intercellular Signaling Peptides and Proteins , Receptor, Fibroblast Growth Factor, Type 1 , Endoglin , Mexico
12.
Acta Cir Bras ; 34(12): e201901202, 2020.
Article in English | MEDLINE | ID: mdl-32049183

ABSTRACT

PURPOSE: To explore the potential role and unclear molecular mechanisms of vaccarin in wound healing. METHODS: Rats' skin excision model to study the effects of vaccarin on wound healing in vivo . Hematoxylin and eosin staining was performed to evaluate Histopathologic characteristics. Immunohistochemistry was employed to assess the effects of vaccarin in accelerating angiogenesis. Western blot was used to evaluate relative protein expressed levels. RESULTS: Vaccarin could significantly promote wound healing and endothelial cells and fibroblasts proliferation in the wound site. Immunohistochemistry and Western blot studies showed that the nodal proteins and receptor (bFGFR) related to angiogenesis signaling pathway were activated, and the microvascular density in the wound site was markedly higher than that in the control group. CONCLUSIONS: The present study was the first to demonstrate that vaccarin is able to induce angiogenesis and accelerate wound healing in vivo by increasing expressions of p-Akt, p-Erk and p-bFGFR. This process is mediated by MAPK/ERK and PI3K/AKT signaling pathways.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Caryophyllaceae/chemistry , Mitogen-Activated Protein Kinase Kinases/drug effects , Phosphatidylinositol 3-Kinases/drug effects , Plant Extracts/pharmacology , Wound Healing/drug effects , Animals , Blotting, Western , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Fibroblasts/drug effects , Immunohistochemistry , Male , Mitogen-Activated Protein Kinase Kinases/analysis , Phosphatidylinositol 3-Kinases/analysis , Plant Extracts/chemistry , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 1/analysis , Receptor, Fibroblast Growth Factor, Type 1/drug effects , Reproducibility of Results , Signal Transduction , Time Factors
13.
J Pathol ; 250(2): 126-133, 2020 02.
Article in English | MEDLINE | ID: mdl-31705763

ABSTRACT

Next-generation sequencing has revealed mutations in several bone-related lesions and was recently used to uncover the genetic basis of giant cell lesions of the jaws (GCLJ). Consistent with their benign nature, GCLJ show a low tumor mutation burden. They also harbor somatic, heterozygous, mutually exclusive mutations in TRPV4, KRAS, or FGFR1. These signature mutations occur only in a subset of lesional cells, suggesting the existence of a 'landscaping effect', with mutant cells inducing abnormal accumulation of non-mutant cells that form the tumor mass. Osteoclast-rich lesions with histological similarities to GCLJ can occur in the jaws sporadically or in association with genetically inherited syndromes. Based on recent results, the pathogenesis of a subgroup of sporadic GCLJ seems closely related to non-ossifying fibroma of long bones, with both lesions sharing MAPK pathway-activating mutations. In this review, we extrapolate from these recent findings to contextualize GCLJ genetics and we highlight the therapeutic implications of this new information. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Giant Cell Tumors/genetics , Jaw Neoplasms/genetics , Giant Cell Tumors/pathology , Giant Cell Tumors/therapy , Granuloma, Giant Cell/genetics , Granuloma, Giant Cell/pathology , Granuloma, Giant Cell/therapy , High-Throughput Nucleotide Sequencing/methods , Humans , Jaw Neoplasms/pathology , Jaw Neoplasms/therapy , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , TRPV Cation Channels/genetics
14.
Am J Case Rep ; 20: 1566-1571, 2019 Oct 25.
Article in English | MEDLINE | ID: mdl-31649234

ABSTRACT

BACKGROUND Encephalocraniocutaneous lipomatosis is a rare neurocutaneous disorder characterized by cutaneous, ocular, and central nervous system anomalies; its molecular etiology was recently identified. This report describes the surgical treatment and genetic characterization of a giant ocular lipodermoid cyst secondary to encephalocraniocutaneous lipomatosis. CASE REPORT An 11-year-old girl with past medical history of absence seizures presented with a reddish protruding mass in her right eye involving the temporal conjunctiva and the peripheral temporal cornea; eyelid closure was not possible due to mass protrusion. She also presented skin tags at the level of the external canthus and 3 alopecic areas at the level of the scalp compatible with nevus psiloliparus. No family history was reported. A dermoid cyst was suspected and excisional biopsy was performed under general anesthesia. A large conjunctival and lamellar corneoscleral resection was done, followed by a corneal tectonic graft. Molecular analysis was carried out, including PCR and Sanger sequencing on DNA obtained from the mass. After surgery, the patient achieved complete eyelid closure, reduction of ocular surface symptoms, and improved aesthetic appearance. Histological analysis confirmed a lipodermoid cyst; genetic tests confirmed a mosaic activating mutation in FGFR1 (c.1638C>A, p.Asn546Lys). The diagnosis was encephalocraniocutaneous lipomatosis. CONCLUSIONS ECCL is a rare condition; an accurate diagnosis comprising clinical and genetic aspects can facilitate the monitoring of possible complications, improve the multidisciplinary treatment, and provide valuable information for future therapy developments. In this case, the patient's quality of life improved significantly, ocular symptoms disappeared, and a good esthetic appearance was achieved.


Subject(s)
Dermoid Cyst/genetics , Dermoid Cyst/surgery , Eye Diseases/diagnosis , Eye Diseases/genetics , Eye Neoplasms/genetics , Eye Neoplasms/surgery , Lipomatosis/diagnosis , Lipomatosis/genetics , Neurocutaneous Syndromes/diagnosis , Neurocutaneous Syndromes/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Biopsy , Child , Corneal Transplantation , DNA Mutational Analysis , Dermoid Cyst/etiology , Eye Diseases/complications , Eye Neoplasms/etiology , Female , Humans , Lipomatosis/complications , Neurocutaneous Syndromes/complications , Polymerase Chain Reaction , Seizures/etiology
15.
Br J Pharmacol ; 176(23): 4462-4473, 2019 12.
Article in English | MEDLINE | ID: mdl-31351013

ABSTRACT

BACKGROUND AND PURPOSE: Pulmonary arterial hypertension (PAH) is a progressive disease associated with high morbidity and mortality, despite advances in medical therapy. We compared the effects of infigratinib (NVP-BGJ398), a new FGF receptor-1 inhibitor, with or without the PDE-5 inhibitor sildenafil, on vascular function and remodelling as well as on gene expression of signal transducers for receptors of TGF-ß (Smads-1/2/4) and transcription factor of endothelial-mesenchymal transition (Twist-1) in established experimental PAH. Types I and III pro-collagen and TGF-ß expressions in lung fibroblasts were analysed in vitro after the different treatments. EXPERIMENTAL APPROACH: PAH was induced in male Wistar rats with monocrotaline. 14 days later, treatments [sildenafil (SIL), infigratinib (INF) or their combination (SIL+INF)] were given for another 14 days. On Day 28, echocardiography and haemodynamic assays were performed, and lungs and pulmonary vessels were removed for analysis by histology, immunohistochemistry and RT-PCR. Fibroblasts prepared from PAH lungs were also analysed for TGF-ß and pro-collagen. KEY RESULTS: Only the combination of infigratinib and sildenafil significantly improved right ventricular systolic pressure and vascular remodelling parameters (right ventricular hypertrophy, smooth muscle α-actin, vessel wall thickness, and vascular collagen content). Infigratinib may act by reducing gene expression of Smads-1/4 and Twist-1 in lung tissue, as well as TGF-ß and types I and III pro-collagen in lung fibroblasts. CONCLUSIONS AND IMPLICATIONS: In this model of monocrotaline-induced PAH, the combination of the new inhibitor of FGF receptor-1, infigratinib, and sildenafil effectively improved haemodynamics and decreased vascular remodelling.


Subject(s)
Antihypertensive Agents/pharmacology , Phenylurea Compounds/pharmacology , Pulmonary Arterial Hypertension/drug therapy , Pyrimidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Sildenafil Citrate/pharmacology , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/chemistry , Injections, Intraperitoneal , Male , Monocrotaline , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/chemistry , Pulmonary Arterial Hypertension/chemically induced , Pulmonary Arterial Hypertension/metabolism , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Rats , Rats, Wistar , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Sildenafil Citrate/administration & dosage
16.
Mol Genet Genomic Med ; 7(5): e625, 2019 05.
Article in English | MEDLINE | ID: mdl-30891959

ABSTRACT

BACKGROUND: Postzygotic KRAS, HRAS, NRAS, and FGFR1 mutations result in a group of mosaic RASopathies characterized by related developmental anomalies in eye, skin, heart, and brain. These oculocutaneous disorders include oculoectodermal syndrome (OES) encephalo-cranio-cutaneous lipomatosis (ECCL), and Schimmelpenning-Feuerstein-Mims syndrome (SFMS). Here, we report the results of the clinical and molecular characterization of a novel cohort of patients with oculocutaneous mosaic RASopathies. METHODS: Two OES, two ECCL, and two SFMS patients were ascertained in the study. In addition, two subjects with unilateral isolated epibulbar dermoids were also enrolled. Molecular analysis included PCR amplification and Sanger sequencing of KRAS, HRAS, NRAS, and FGFR1 genes in DNA obtained from biopsies (skin/epibulbar dermoids), buccal mucosa, and blood leukocytes. Massive parallel sequencing was employed in two cases with low-level mosaicism. RESULTS: In DNA from biopsies, mosaicism for pathogenic variants, including KRAS p.Ala146Thr in two OES subjects, FGFR1 p.Asn546Lys and KRAS p.Ala146Val in ECCL patients, and KRAS p.Gly12Asp in both SFMS patients, was demonstrated. No mutations were shown in DNA from conjunctival lesions in two subjects with isolated epibubar dermoids. CONCLUSION: Our study allowed the expansion of the clinical spectrum of mosaic RASopathies and supports that mosaicism for recurrent mutations in KRAS and FGFR1 is a commonly involved mechanism in these rare oculocutaneous anomalies.


Subject(s)
Dermoid Cyst/genetics , Ectodermal Dysplasia/genetics , Eye Diseases/genetics , Lipomatosis/genetics , Neurocutaneous Syndromes/genetics , Nevus, Sebaceous of Jadassohn/genetics , Phenotype , Receptor, Fibroblast Growth Factor, Type 1/genetics , Dermoid Cyst/pathology , Ectodermal Dysplasia/pathology , Eye Diseases/pathology , GTP Phosphohydrolases/genetics , Humans , Lipomatosis/pathology , Membrane Proteins/genetics , Mosaicism , Neurocutaneous Syndromes/pathology , Nevus, Sebaceous of Jadassohn/pathology , Proto-Oncogene Proteins p21(ras)/genetics
17.
Histopathology ; 74(2): 311-320, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30129658

ABSTRACT

AIMS: Fibroblast growth factor (FGF)-2 and fibroblast growth factor receptor (FGFR)-1 are associated with tumour invasiveness, cell proliferation, angiogenesis, and metastasis. The aims of this study were to investigate FGF-2 expression and FGFR-1 expression in oral epithelial dysplasia (OED) and oral tongue squamous cell carcinoma (OTSCC), and their correlation with OTSCC patients' prognosis. METHODS AND RESULTS: One hundred and sixty-seven cases were retrospectively selected, including 85 surgical specimens of patients with OTSCC, 46 incisional biopsies of OTSCC, and 36 incisional biopsies of OED. Tissue sections were subjected to immunohistochemical staining for FGF-2 and FGFR-1, and digitally scored. Elevated scores of FGF-2 and FGFR-1 immunostaining were associated with high-grade OEDs. FGF-2 positivity in the stroma was associated with vascular invasion and a worse prognosis, in both overall survival (OS) and disease-free survival (DFS) analyses, in univariate and multivariate models. FGFR-1 positivity in the stroma was correlated with lymph node metastasis and distant metastasis. FGFR-1 expression in either the malignant cells or the stroma was strongly correlated with shorter OS and DFS. CONCLUSIONS: Taken together, our findings suggest that increased FGF-2 expression and increased FGFR-1 expression are associated with high-grade OEDs, and are correlated with the presence of metastasis and adverse outcomes in OTSCC patients.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Fibroblast Growth Factor 2/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Tongue Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Disease-Free Survival , Female , Humans , Lymphatic Metastasis/pathology , Male , Middle Aged , Neoplasm Invasiveness/pathology , Prognosis , Retrospective Studies , Survival Rate , Tongue Neoplasms/mortality , Tongue Neoplasms/pathology , Young Adult
18.
Acta cir. bras ; Acta cir. bras;34(12): e201901202, 2019. graf
Article in English | LILACS | ID: biblio-1054685

ABSTRACT

Abstract Purpose To explore the potential role and unclear molecular mechanisms of vaccarin in wound healing. Methods Rats' skin excision model to study the effects of vaccarin on wound healing in vivo . Hematoxylin and eosin staining was performed to evaluate Histopathologic characteristics. Immunohistochemistry was employed to assess the effects of vaccarin in accelerating angiogenesis. Western blot was used to evaluate relative protein expressed levels. Results Vaccarin could significantly promote wound healing and endothelial cells and fibroblasts proliferation in the wound site. Immunohistochemistry and Western blot studies showed that the nodal proteins and receptor (bFGFR) related to angiogenesis signaling pathway were activated, and the microvascular density in the wound site was markedly higher than that in the control group. Conclusions The present study was the first to demonstrate that vaccarin is able to induce angiogenesis and accelerate wound healing in vivo by increasing expressions of p-Akt, p-Erk and p-bFGFR. This process is mediated by MAPK/ERK and PI3K/AKT signaling pathways.


Subject(s)
Animals , Male , Wound Healing/drug effects , Plant Extracts/pharmacology , Phosphatidylinositol 3-Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/drug effects , Caryophyllaceae/chemistry , Angiogenesis Inducing Agents/pharmacology , Time Factors , Immunohistochemistry , Plant Extracts/chemistry , Signal Transduction , Blotting, Western , Reproducibility of Results , Rats, Sprague-Dawley , Phosphatidylinositol 3-Kinases/analysis , Mitogen-Activated Protein Kinase Kinases/analysis , Endothelial Cells/drug effects , Cell Proliferation/drug effects , Receptor, Fibroblast Growth Factor, Type 1/analysis , Receptor, Fibroblast Growth Factor, Type 1/drug effects , Fibroblasts/drug effects
19.
Rev. bras. pesqui. méd. biol ; Braz. j. med. biol. res;52(1): e7816, 2019. tab, graf
Article in English | LILACS | ID: biblio-974271

ABSTRACT

Fibroblast growth factor receptor 1 (FGFR1) has been reported in gastric cancer to be a prognostic factor. However, miR-497-targeted FGFR1 has not been explored in the carcinogenesis of gastric cancer. The present study intended to revalidate the prognostic significance of FGFR1 in patients with gastric cancer, and the mechanism of miR-497-regulated FGFR1 was investigated in gastric cancer cell proliferation and apoptosis. The messenger RNA (mRNA) and protein levels were assayed by RT-qPCR and western blotting, respectively. The targeted genes were predicted by a bioinformatics algorithm and confirmed by a dual luciferase reporter assay. Cell proliferation was analyzed by CCK-8 assay. Annexin V-FITC/PI staining was used to evaluate the apoptosis in AGS and SGC-7901 cells. FGFR1 was frequently up-regulated in gastric cancer tissues and associated with poor overall survival in patients with gastric cancer. Interestingly, FGFR1 loss-of-function resulted in a significant growth inhibition and apoptosis in AGS and SGC-7901 cells. In addition, we found that miR-497 was inhibited in gastric cancer tissues and cell lines, while overexpression of miR-497 could suppress proliferation and induce apoptosis in AGS and SGC-7901 cells. Importantly, bioinformatics analysis and experimental data suggested that FGFR1 was a direct target of miR-497, which could inhibit FGFR1 expression when transfected with miR-497 mimics. Furthermore, we found that overexpression of FGFR1 reversed the growth inhibition and apoptosis of miR-497 mimics in AGS and SGC-7901 cells. These findings suggested that overexpression of miR-497 inhibited proliferation and induced apoptosis in gastric cancer through the suppression of FGFR1.


Subject(s)
Humans , Stomach Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Prognosis , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Immunohistochemistry , Signal Transduction , Blotting, Western , Apoptosis , Disease Progression , Cell Line, Tumor , Cell Proliferation , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Real-Time Polymerase Chain Reaction
20.
Braz J Med Biol Res ; 52(1): e7816, 2018 Nov 23.
Article in English | MEDLINE | ID: mdl-30484492

ABSTRACT

Fibroblast growth factor receptor 1 (FGFR1) has been reported in gastric cancer to be a prognostic factor. However, miR-497-targeted FGFR1 has not been explored in the carcinogenesis of gastric cancer. The present study intended to revalidate the prognostic significance of FGFR1 in patients with gastric cancer, and the mechanism of miR-497-regulated FGFR1 was investigated in gastric cancer cell proliferation and apoptosis. The messenger RNA (mRNA) and protein levels were assayed by RT-qPCR and western blotting, respectively. The targeted genes were predicted by a bioinformatics algorithm and confirmed by a dual luciferase reporter assay. Cell proliferation was analyzed by CCK-8 assay. Annexin V-FITC/PI staining was used to evaluate the apoptosis in AGS and SGC-7901 cells. FGFR1 was frequently up-regulated in gastric cancer tissues and associated with poor overall survival in patients with gastric cancer. Interestingly, FGFR1 loss-of-function resulted in a significant growth inhibition and apoptosis in AGS and SGC-7901 cells. In addition, we found that miR-497 was inhibited in gastric cancer tissues and cell lines, while overexpression of miR-497 could suppress proliferation and induce apoptosis in AGS and SGC-7901 cells. Importantly, bioinformatics analysis and experimental data suggested that FGFR1 was a direct target of miR-497, which could inhibit FGFR1 expression when transfected with miR-497 mimics. Furthermore, we found that overexpression of FGFR1 reversed the growth inhibition and apoptosis of miR-497 mimics in AGS and SGC-7901 cells. These findings suggested that overexpression of miR-497 inhibited proliferation and induced apoptosis in gastric cancer through the suppression of FGFR1.


Subject(s)
Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Stomach Neoplasms/genetics , Apoptosis , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Disease Progression , Humans , Immunohistochemistry , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL