Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 196
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Mol Cell Biochem ; 468(1-2): 21-34, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32146648

RESUMO

Neuroblastoma (NB) is the common pediatric tumor of the sympathetic nervous system characterized by poor prognosis. Owing to the challenges such as high tumor heterogeneity, multidrug resistance, minimal residual disease, etc., there is an immediate need for exploring new therapeutic strategies and effective treatments for NB. Herein, in the current study, we explored the unexplored response of NB cells to the second-generation histone deacetylase inhibitor (HDACi) JNJ-26481585(JNJ) and the lysosomotropic agent, Chloroquine (CQ) alone and upon JNJ/CQ treatment as a plausible therapeutic. We identify that while JNJ alone induced autophagy in NB cells, JNJ/CQ treatment decreased the viability and proliferation of NB cells in vitro by switching from autophagy to apoptosis. Further we found that autophagy inhibition by CQ pre-treatment led to the generation of ROS and a decrease in the mitochondrial membrane potential (MMP) that subsequently caused caspase-3-mediated apoptotic cell death in NB cells. Corroborating the above observations, we found that the ROS scavenger N-acetylcysteine (NAC) countered caspase-3 activity and the cells were rescued from apoptosis. Finally, these observations establish that JNJ/CQ treatment resulted in cell death in NB cells by triggering the formation of ROS and disruption of MMP, suggesting that modulation of JNJ-induced autophagy by CQ represents a promising new therapeutic approach in NB.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Cloroquina/farmacologia , Ácidos Hidroxâmicos/farmacologia , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Neoplasias do Sistema Nervoso Periférico/tratamento farmacológico , Acetilcisteína/farmacologia , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sinergismo Farmacológico , Sequestradores de Radicais Livres/farmacologia , Inibidores de Histona Desacetilases/farmacologia , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neoplasias do Sistema Nervoso Periférico/genética , Neoplasias do Sistema Nervoso Periférico/metabolismo , Espécies Reativas de Oxigênio/metabolismo
2.
Pharmacol Res ; 131: 164-176, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29466695

RESUMO

Neuroblastoma is the most common extracranical tumor of childhood and the most deadly tumor of infancy. It is characterized by early age onset and high frequencies of metastatic disease but also the capacity to spontaneously regress. Despite intensive therapy, the survival for patients with high-risk neuroblastoma and those with recurrent or relapsed disease is low. Hence, there is an urgent need to develop new therapies for these patient groups. The molecular pathogenesis based on high-throughput omics technologies of neuroblastoma is beginning to be resolved which have given the opportunity to develop personalized therapies for high-risk patients. Here we discuss the potential of developing targeted therapies against aberrantly expressed molecules detected in sub-populations of neuroblastoma patients and how these selected targets can be drugged in order to overcome treatment resistance, improve survival and quality of life for these patients and also the possibilities to transfer preclinical research into clinical testing.


Assuntos
Antineoplásicos/uso terapêutico , Terapia de Alvo Molecular/métodos , Neuroblastoma/tratamento farmacológico , Neoplasias do Sistema Nervoso Periférico/tratamento farmacológico , Antineoplásicos/farmacologia , Criança , Descoberta de Drogas/métodos , Resistencia a Medicamentos Antineoplásicos , Drogas em Investigação/farmacologia , Drogas em Investigação/uso terapêutico , Humanos , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Qualidade de Vida , Sistema Nervoso Simpático/efeitos dos fármacos , Sistema Nervoso Simpático/metabolismo , Sistema Nervoso Simpático/patologia
3.
Proc Natl Acad Sci U S A ; 111(19): E2008-17, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24778213

RESUMO

The ability of cancer cells to invade along nerves is associated with aggressive disease and diminished patient survival rates. Perineural invasion (PNI) may be mediated by nerve secretion of glial cell line-derived neurotrophic factor (GDNF) attracting cancer cell migration through activation of cell surface Ret proto-oncogene (RET) receptors. GDNF family receptor (GFR)α1 acts as coreceptor with RET, with both required for response to GDNF. We demonstrate that GFRα1 released by nerves enhances PNI, even in the absence of cancer cell GFRα1 expression. Cancer cell migration toward GDNF, RET phosphorylation, and MAPK pathway activity are increased with exposure to soluble GFRα1 in a dose-dependent fashion. Dorsal root ganglia (DRG) release soluble GFRα1, which potentiates RET activation and cancer cell migration. In vitro DRG coculture assays of PNI show diminished PNI with DRG from GFRα1(+/-) mice compared with GFRα1(+/+) mice. An in vivo murine model of PNI demonstrates that cancer cells lacking GFRα1 maintain an ability to invade nerves and impair nerve function, whereas those lacking RET lose this ability. A tissue microarray of human pancreatic ductal adenocarcinomas demonstrates wide variance of cancer cell GFRα1 expression, suggesting an alternate source of GFRα1 in PNI. These findings collectively demonstrate that GFRα1 released by nerves enhances PNI through GDNF-RET signaling and that GFRα1 expression by cancer cells enhances but is not required for PNI. These results advance a mechanistic understanding of PNI and implicate the nerve itself as a key facilitator of this adverse cancer cell behavior.


Assuntos
Adenocarcinoma/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas c-ret/metabolismo , Células 3T3 , Adenocarcinoma/patologia , Animais , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Técnicas de Cocultura , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Nus , Invasividade Neoplásica , Tecido Nervoso/metabolismo , Tecido Nervoso/patologia , Neoplasias Pancreáticas/patologia , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Proto-Oncogene Mas , RNA Interferente Pequeno/genética , Neuropatia Ciática/metabolismo , Neuropatia Ciática/patologia , Solubilidade
4.
Lab Invest ; 96(12): 1268-1278, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27775688

RESUMO

NF-κB has an essential role in the initiation and progression of pancreatic cancer and specifically mediates the induction of epithelial-mesenchymal transition and invasiveness. In this study, we demonstrate the importance of activated NF-κB signaling in EMT induction, lymphovascular metastasis, and neural invasion. Modulation of NF-κB activity was accomplished through the specific NF-κB inhibitor (BAY 11-7085), triptolide, and Minnelide treatment, as well as overexpression of IKBα repressor and IKK activator plasmids. In the classical lymphovascular metastatic cascade, inhibition of NF-κB decreased the expression of several EMT transcription factors (SNAI1, SNAI2, and ZEB1) and mesenchymal markers (VIM and CDH2) and decreased in vitro invasion, which was rescued by IKK activation. This was further demonstrated in vivo via BAY 11-7085 treatment in a orthotopic model of pancreatic cancer. In vivo NF-κB inhibition decreased tumor volume; decreased tumor EMT gene expression, while restoring cell-cell junctions; and decreasing overall metastasis. Furthermore, we demonstrate the importance of active NF-κB signaling in neural invasion. Triptolide treatment inhibits Nerve Growth Factor (NGF) mediated, neural-tumor co-culture in vitro invasion, and dorsal root ganglia (DRG) neural outgrowth through a disruption in tumor-neural cross talk. In vivo, Minnelide treatment decreased neurotrophin expression, nerve density, and sciatic nerve invasion. Taken together, this study demonstrates the importance of NF-κB signaling in the progression of pancreatic cancer through the modulation of EMT induction, lymphovascular invasion, and neural invasion.


Assuntos
Transição Epitelial-Mesenquimal , NF-kappa B/metabolismo , Pâncreas/metabolismo , Neoplasias Pancreáticas/metabolismo , Nervos Periféricos/metabolismo , Neoplasias do Sistema Nervoso Periférico/secundário , Transdução de Sinais , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular , Linhagem Celular Tumoral , Técnicas de Cocultura , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Gânglios Espinais/citologia , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Gânglios Espinais/patologia , Humanos , Metástase Linfática/patologia , Metástase Linfática/prevenção & controle , Camundongos , Camundongos Nus , Inibidor de NF-kappaB alfa/genética , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/antagonistas & inibidores , Invasividade Neoplásica/patologia , Transplante de Neoplasias , Pâncreas/efeitos dos fármacos , Pâncreas/patologia , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Nervos Periféricos/citologia , Nervos Periféricos/efeitos dos fármacos , Nervos Periféricos/patologia , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Neoplasias do Sistema Nervoso Periférico/prevenção & controle , Proteínas Recombinantes/metabolismo , Nervo Isquiático/citologia , Nervo Isquiático/efeitos dos fármacos , Nervo Isquiático/metabolismo , Nervo Isquiático/patologia , Transdução de Sinais/efeitos dos fármacos
5.
Histochem Cell Biol ; 146(4): 467-77, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27278446

RESUMO

Schwann cells generate myelin sheaths around the axons of the peripheral nervous system, thus facilitating efficient nerve impulse propagation. Two main tumor types can arise from peripheral nerves, schwannomas and neurofibromas, which are sometimes difficult to distinguish and may require the use of diagnostic biomarkers. Here, we characterize a new marker for Schwann cells and its potential use as a diagnostic marker for schwannomas. Immunohistochemistry for Glu-tubulin, a posttranslational modification of α-tubulin, was performed in mouse and human tissues. This technique labels Schwann cells but not oligodendrocytes. All peripheral nerves were immunoreactive for this antibody, including large nerve trunks, thin myelinated nerves, as well as the myenteric and submucous plexus of the digestive tract. In the mouse brain, many neurons were immunoreactive for Glu-tubulin but oligodendrocytes were negative. During embryo development, immunoreactive nerves were already found at E10. In Schwann cells, the staining is restricted to the myelin sheaths and is not present in the perinuclear cytoplasm or the Ranvier nodes. Primary cultures of fibroblasts and Schwann cells were established from mouse sciatic nerves, and Western blot analysis showed that Glu-tubulin immunoreactivity was found in the Schwann cells but not in the fibroblasts. Clinical specimens of schwannomas (n = 20) and neurofibromas (n = 20) were stained with anti-Glu-tubulin antibodies. Schwannomas presented a strong staining in all tumor cells, whereas neurofibromas had a light speckled staining pattern, easily distinguishable from the one found in schwannomas. In conclusion, Glu-tubulin can be used as a marker of Schwann cells and can help in diagnosing peripheral nerve tumors.


Assuntos
Neurilemoma/metabolismo , Neurofibroma/metabolismo , Neoplasias do Sistema Nervoso Periférico/metabolismo , Células de Schwann/metabolismo , Tubulina (Proteína)/metabolismo , Animais , Biomarcadores/análise , Biomarcadores/metabolismo , Células Cultivadas , Diagnóstico Diferencial , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neurilemoma/diagnóstico , Neurofibroma/diagnóstico , Neoplasias do Sistema Nervoso Periférico/diagnóstico , Células de Schwann/patologia , Tubulina (Proteína)/análise
7.
Eur Radiol ; 24(2): 405-12, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24097302

RESUMO

OBJECTIVES: To evaluate the usefulness of normalising intra-tumour tracer accumulation on (18)F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) to reference tissue uptake for characterisation of peripheral nerve sheath tumours (PNSTs) in neurofibromatosis type 1 (NF1) compared with the established maximum standardised uptake value (SUVmax) cut-off of >3.5. METHODS: Forty-nine patients underwent FDG PET/CT. Intra-tumour tracer uptake (SUVmax) was normalised to three different reference tissues (tumour-to-liver, tumour-to-muscle and tumour-to-fat ratios). Receiver operating characteristic (ROC) analyses were used out to assess the diagnostic performance. Histopathology and follow-up served as the reference standard. RESULTS: Intra-tumour tracer uptake correlated significantly with liver uptake (rs= 0.58, P = 0.016). On ROC analysis, the optimum threshold for tumour-to-liver ratio was >2.6 (AUC = 0.9735). Both the SUVmax cut-off value of >3.5 and a tumour-to-liver ratio >2.6 provided a sensitivity of 100 %, but specificity was significantly higher for the latter (90.3% vs 79.8%; P = 0.013). CONCLUSIONS: In patients with NF1, quantitative (18)F-FDG PET imaging may identify malignant change in neurofibromas with high accuracy. Specificity could be significantly increased by using the tumour-to-liver ratio. The authors recommend further evaluation of a tumour-to-liver ratio cut-off value of >2.6 for diagnostic intervention planning. KEY POINTS: • (18)F-FDG PET/CT is used for detecting malignancy in PNSTs in NF1 patients • An SUV max cut-off value may give false-positive results for benign plexiform neurofibromas • Specificity can be significantly increased using a tumour-to-liver ratio.


Assuntos
Fluordesoxiglucose F18 , Fígado/metabolismo , Neoplasias de Bainha Neural/diagnóstico , Neurofibromatose 1/diagnóstico , Neoplasias do Sistema Nervoso Periférico/diagnóstico , Tomografia por Emissão de Pósitrons/métodos , Tomografia Computadorizada por Raios X/métodos , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Diagnóstico Diferencial , Feminino , Fluordesoxiglucose F18/farmacocinética , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/metabolismo , Estadiamento de Neoplasias , Neoplasias de Bainha Neural/complicações , Neoplasias de Bainha Neural/metabolismo , Neurofibromatose 1/complicações , Neurofibromatose 1/metabolismo , Neoplasias do Sistema Nervoso Periférico/complicações , Neoplasias do Sistema Nervoso Periférico/metabolismo , Curva ROC , Compostos Radiofarmacêuticos/farmacocinética , Adulto Jovem
8.
Ann Diagn Pathol ; 18(2): 95-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24461704

RESUMO

Benign peripheral nerve sheath tumors (PNSTs) showing more than one line of differentiation (hybrid PNSTs) have been increasingly recognized, mainly due to awareness of their existence and as a consequence of increased use of immunohistochemisty during the last decade. Two recent studies suggested overrepresentation of hybrid tumors among benign PNSTs in patients with neurofibromatosis type 1 (NF-1). This study was performed to assess the presence of perineurial cells in microscopic (early) neurofibromatous lesions and normal-looking peripheral nerves in specimens from 5 patients with NF-1 using markers of perineurial cell differentiation (epithelial membrane antigen, claudin-1, and glucose transporter 1). In 2 patients, multiple normal looking nerve fibers as well as hypertrophied nerves and microscopic tumor nodules showed variable intraneural perineurial cell proliferations that frequently occupied the whole nerve fascicle resulting in multiple microscopic reticular perineurioma-like nodules (microscopic hybrid neurofibromatosis/perineuriomatosis). None of the cases showed the onion skin pattern of intraneural perineurioma. However, other nerve fibers within the same specimens showed normal compact rim of perineurium without any detectable intraneural perineurial cells. Both patients had concurrent multiple larger PNSTs (plexiform neurofibromas, hybrid neurofibroma/perineurioma and lesions with features intermediate between the 2 types). One specimen harboring high-grade malignant PNST and 2 specimens with large solitary neurofibromas displayed no intraneural perineurial cells. These observations suggest that intraneural perineurial proliferations are part of the early lesions in the setting of constitutional NF-1 inactivation and support the concept of pure and hybrid perineuriomatous lesions as novel member of the spectrum of PNSTs in NF-1.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias de Bainha Neural/patologia , Neurofibroma/patologia , Neurofibromatose 1/patologia , Neoplasias do Sistema Nervoso Periférico/patologia , Adulto , Proliferação de Células , Claudina-1/metabolismo , Diagnóstico Diferencial , Feminino , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Humanos , Pessoa de Meia-Idade , Mucina-1/metabolismo , Neoplasias de Bainha Neural/metabolismo , Neoplasias de Bainha Neural/cirurgia , Neurofibroma/metabolismo , Neurofibroma/cirurgia , Neurofibromatose 1/metabolismo , Neurofibromatose 1/cirurgia , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/cirurgia
9.
Mol Cancer ; 12(1): 70, 2013 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-23835063

RESUMO

BACKGROUND: Neuroblastoma (NB) tumours are commonly divided into three cytogenetic subgroups. However, by unsupervised principal components analysis of gene expression profiles we recently identified four distinct subgroups, r1-r4. In the current study we characterized these different subgroups in more detail, with a specific focus on the fourth divergent tumour subgroup (r4). METHODS: Expression microarray data from four international studies corresponding to 148 neuroblastic tumour cases were subject to division into four expression subgroups using a previously described 6-gene signature. Differentially expressed genes between groups were identified using Significance Analysis of Microarray (SAM). Next, gene expression network modelling was performed to map signalling pathways and cellular processes representing each subgroup. Findings were validated at the protein level by immunohistochemistry and immunoblot analyses. RESULTS: We identified several significantly up-regulated genes in the r4 subgroup of which the tyrosine kinase receptor ERBB3 was most prominent (fold change: 132-240). By gene set enrichment analysis (GSEA) the constructed gene network of ERBB3 (n = 38 network partners) was significantly enriched in the r4 subgroup in all four independent data sets. ERBB3 was also positively correlated to the ErbB family members EGFR and ERBB2 in all data sets, and a concurrent overexpression was seen in the r4 subgroup. Further studies of histopathology categories using a fifth data set of 110 neuroblastic tumours, showed a striking similarity between the expression profile of r4 to ganglioneuroblastoma (GNB) and ganglioneuroma (GN) tumours. In contrast, the NB histopathological subtype was dominated by mitotic regulating genes, characterizing unfavourable NB subgroups in particular. The high ErbB3 expression in GN tumour types was verified at the protein level, and showed mainly expression in the mature ganglion cells. CONCLUSIONS: Conclusively, this study demonstrates the importance of performing unsupervised clustering and subtype discovery of data sets prior to analyses to avoid a mixture of tumour subtypes, which may otherwise give distorted results and lead to incorrect conclusions. The current study identifies ERBB3 as a clear-cut marker of a GNB/GN-like expression profile, and we suggest a 7-gene expression signature (including ERBB3) as a complement to histopathology analysis of neuroblastic tumours. Further studies of ErbB3 and other ErbB family members and their role in neuroblastic differentiation and pathogenesis are warranted.


Assuntos
Biomarcadores Tumorais/metabolismo , Ganglioneuroblastoma/metabolismo , Ganglioneuroma/metabolismo , Neoplasias do Sistema Nervoso Periférico/metabolismo , Receptor ErbB-3/metabolismo , Biomarcadores Tumorais/genética , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Redes Reguladoras de Genes , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Receptor ErbB-3/genética , Transcriptoma , Regulação para Cima
10.
Acta Chir Belg ; 113(4): 298-300, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24224442

RESUMO

Ganglioneuromas are rare, benign, well-differentiated, slowgrowing tumors of the sympathetic nervous system, composed of large, mature neurons in a stroma composed of Schwann cells. Ganglioneuromas are derived from the neural crest cells and can arise anywhere from the base of the skull to the pelvis. The pre-sacral area is a very rare location for ganglioneuromas to develop. We describe the case of a 31 year old woman, who was incidentally found to have an abnormal pre-sacral mass. The following work-up, revealed the mass to be growing on imagery (computed tomography and magnetic resonance imagery) and fluorine-18 fluorodeoxiglucose avid. The mass was removed by assisted laparoscopy and was found to be a benign ganglioneuroma. This is the first described case of fluorine-18 fluorodeoxiglucose avid, pre-sacral, benign ganglioneuroma.


Assuntos
Fluordesoxiglucose F18 , Ganglioneuroma/diagnóstico , Imageamento por Ressonância Magnética/métodos , Neoplasias do Sistema Nervoso Periférico/diagnóstico , Tomografia por Emissão de Pósitrons/métodos , Adulto , Diagnóstico Diferencial , Feminino , Fluordesoxiglucose F18/farmacocinética , Ganglioneuroma/metabolismo , Humanos , Laparoscopia , Plexo Lombossacral , Estadiamento de Neoplasias , Neoplasias do Sistema Nervoso Periférico/metabolismo , Compostos Radiofarmacêuticos/farmacocinética
11.
Zhonghua Bing Li Xue Za Zhi ; 42(5): 305-10, 2013 May.
Artigo em Zh | MEDLINE | ID: mdl-24004586

RESUMO

OBJECTIVE: To study the clinicopathologic characteristics of peripheral neuroblastic tumors and to evaluate the prognostic significance of these features. METHODS: The clinical and pathologic findings were retrospectively reviewed in 121 cases of peripheral neuroblastic tumor. The clinical outcomes of patients were evaluated. The three-year event-free survival rate was analyzed, with respect to age of patients, Evan's staging, International Neuroblastoma Pathology Classification and mitosis-karyorrhexis index. RESULTS: The median age at diagnosis was 2.7 years; and 96 cases (79.3%) occurred in patients younger than 5 years old. The number of cases in Evan's staging I, II, III, IV and IVs was 24, 39, 24, 29 and 5, respectively. There were 82 cases of neuroblastoma (NB) (including 2 cases of undifferentiated NB, 52 cases of poorly differentiated NB and 28 cases of differentiating NB), 9 cases of ganglioneuroblastoma, intermixed type (GNBi), 19 cases of ganglioneuroma, maturing type (GN) and 11 cases of ganglioneuroblastoma, nodular type (GNBn). Forty-nine cases were in the favorable histology subgroup and 72 cases in the unfavorable histology subgroup. The overall three-year event-free survival rate of the 121 cases was 73.0% ± 4.3%. The three-year event-free survival rates were associated with age (P = 0.002), Evan's staging (P = 0.000), histologic category (P = 0.000), mitosis-karyorrhexis index (P = 0.043), prognostic subgroup (P = 0.000). CONCLUSIONS: Most of the peripheral neuroblastic tumors occur in the children younger than 5 years old. It is composed of NB, GNBi, GN and GNBn. The three-year event-free survival rate is approximately 70%. Significant prognostic parameters include age of patients, Evan's staging, International Neuroblastoma Pathology Classification and mitosis-karyorrhexis index.


Assuntos
Neuroblastoma/patologia , Neoplasias do Sistema Nervoso Periférico/patologia , Fatores Etários , Antígenos Nucleares/metabolismo , Criança , Pré-Escolar , Intervalo Livre de Doença , Feminino , Ganglioneuroblastoma/metabolismo , Ganglioneuroblastoma/patologia , Ganglioneuroblastoma/cirurgia , Ganglioneuroma/metabolismo , Ganglioneuroma/patologia , Ganglioneuroma/cirurgia , Humanos , Lactente , Recém-Nascido , Masculino , Estadiamento de Neoplasias , Proteínas do Tecido Nervoso/metabolismo , Nestina/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/cirurgia , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/cirurgia , Fosfopiruvato Hidratase/metabolismo , Estudos Retrospectivos , Proteínas S100/metabolismo
12.
Histopathology ; 61(6): 1006-16, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22804730

RESUMO

AIMS: More than 50% of neuroblastomas (NBs) present with haematogenous and/or lymphatic metastasis; however, little is known about the clinicopathological significance in NBs of the key lymphangiogenesis growth factors vascular endothelial growth factor (VEGF)-C and VEGF-D and the receptor VEGFR-3. METHODS AND RESULTS: Ninety-three NBs and nine ganglioneuromas (GNs) were immunostained for VEGF-C, VEGF-D and VEGFR-3. VEGF-C and VEGF-D were present in 76% and 82% of the NBs, respectively. There was no significant difference in VEGF-C expression between NBs and GNs. VEGF-D expression was significantly higher in NBs compared with GNs and in MYCN-amplified NBs. VEGFR-3 tumoral cell expression (VEGFR-3c), present in 48% of the NBs, was significantly higher in NBs from children ≥ 18 months at presentation and those belonging to a high-risk group. VEGFR-3 lymphovascular density was increased significantly in NBs compared with GNs and in NBs associated with adverse clinicopathological and biological factors. Lymphovascular invasion, assessed in VEGFR-3-stained vessels, was present in ∼50% of NBs. Cox regression analyses demonstrated that VEGFR-3c expression was associated with a significantly shorter event-free survival and that its effect was independent of the important pathological variable, mitosis-karyorrhexis index. CONCLUSIONS: VEGF-D and VEGFR-3 up-regulation support tumour progression in NB and VEGFR-3c may provide a useful prognostic marker in NBs.


Assuntos
Biomarcadores Tumorais/metabolismo , Neuroblastoma/metabolismo , Neoplasias do Sistema Nervoso Periférico/metabolismo , Fator C de Crescimento do Endotélio Vascular/metabolismo , Fator D de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Adolescente , Criança , Pré-Escolar , Progressão da Doença , Ganglioneuroma/diagnóstico , Ganglioneuroma/metabolismo , Ganglioneuroma/mortalidade , Regulação Neoplásica da Expressão Gênica , Humanos , Lactente , Recém-Nascido , Estimativa de Kaplan-Meier , Linfangiogênese/fisiologia , Neuroblastoma/diagnóstico , Neuroblastoma/mortalidade , Neoplasias do Sistema Nervoso Periférico/diagnóstico , Neoplasias do Sistema Nervoso Periférico/mortalidade , Prognóstico , Estudos Retrospectivos , Regulação para Cima , Fator C de Crescimento do Endotélio Vascular/genética , Fator D de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética
13.
Neurochem Res ; 37(6): 1364-71, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22476983

RESUMO

Some 20 years ago c-Fos was identified as a member of the AP-1 family of inducible transcription factors (Angel and Karin in Biochim Biophys Acta 1072:129-157, 1991). More recently, an additional activity was described for this protein: it associates to the endoplasmic reticulum and activates the biosynthesis of phospholipids (Bussolino et al. in FASEB J 15:556-558, 2001), (Gil et al. in Mol Biol Cell 15:1881-1894, 2004), the quantitatively most important components of cellular membranes. This latter activity of c-Fos determines the rate of membrane genesis and consequently of growth in differentiating PC12 cells (Gil et al. in Mol Biol Cell 15:1881-1894, 2004). In addition, it has been shown that c-Fos is over-expressed both in PNS and CNS tumors (Silvestre et al. in PLoS One 5(3):e9544, 2010). Herein, it is shown that c-Fos-activated phospholipid synthesis is required to support membrane genesis during the exacerbated growth characteristic of brain tumor cells. Specifically blocking c-Fos-activated phospholipid synthesis significantly reduces proliferation of tumor cells in culture. Blocking c-Fos expression also prevents tumor progression in mice intra-cranially xeno-grafted human brain tumor cells. In NPcis mice, an animal model of the human disease Neurofibromatosis Type I (Cichowski and Jacks in Cell 104:593-604, 2001), animals spontaneously develop tumors of the PNS and the CNS, provided they express c-Fos (Silvestre et al. in PLoS One 5(3):e9544, 2010). Treatment of PNS tumors with an antisense oligonucleotide that specifically blocks c-Fos expression also blocks tumor growth in vivo. These results disclose cytoplasmic c-Fos as a new target for effectively controlling brain tumor growth.


Assuntos
Proliferação de Células/efeitos dos fármacos , Neoplasias do Sistema Nervoso Central/patologia , Neoplasias do Sistema Nervoso Periférico/patologia , Fosfolipídeos/biossíntese , Proteínas Proto-Oncogênicas c-fos/metabolismo , Animais , Linhagem Celular Tumoral , Sistema Nervoso Central/metabolismo , Neoplasias do Sistema Nervoso Central/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Camundongos , Oligonucleotídeos Antissenso/metabolismo , Células PC12 , Neoplasias do Sistema Nervoso Periférico/metabolismo , Ratos
14.
Mod Pathol ; 24(3): 343-54, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21297585

RESUMO

Neurothekeomas and dermal nerve sheath myxomas have previously been considered related cutaneous neoplasms of peripheral nerve sheath origin based on light microscopic similarities. However, recent immunohistochemical and ultrastructural data indicate nerve sheath myxomas exhibit true nerve sheath differentiation, whereas no such compelling evidence exists for neurothekeomas. Although neurothekeomas lack a specific immunohistochemical profile, similar antigen expression and histopathologic patterns suggest neurothekeomas may be categorized as fibrohistiocytic tumors. To date, no known molecular studies have examined the histogenetic relationship of these tumors. We report the first microarray-based gene expression profile study of these entities on formalin-fixed paraffin-embedded tissues. Cases of dermal schwannomas, dermal nerve sheath myxomas, myxoid/mixed/cellular neurothekeomas, and cellular fibrous histiocytomas diagnosed in the past 3 years were identified in our database. Archival formalin-fixed paraffin-embedded tissue from 28 patients was selected for microarray analysis (seven schwannomas, five nerve sheath myxomas, nine myxoid/mixed/cellular neurothekeomas and seven cellular fibrous histiocytomas). Following tumor RNA isolation, amplification and labeling using commercially available kits, labeled targets were hybridized to the Affymetrix GeneChip Human Genome U133 Plus 2.0 Array (Santa Clara, CA, USA). Acquisition of array images and data analyses was performed using appropriate software. Hierarchical clustering and principal component analysis demonstrated discrete groups, which correlated with histopathologically identified diagnoses. Dermal nerve sheath myxomas demonstrate very similar molecular genetic signatures to dermal schwannomas, whereas neurothekeomas of all subtypes more closely resemble cellular fibrous histiocytomas. We are the first to report distinct gene expression profiles for nerve sheath myxomas and neurothekeomas, which further substantiates the argument that these are separate entities. Our molecular data confirms that dermal nerve sheath myxomas are of peripheral nerve sheath origin, and suggests that neurothekeomas may actually be a variant of fibrous histiocytomas.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neurotecoma/genética , Neoplasias Cutâneas/genética , Adolescente , Adulto , Biomarcadores Tumorais/metabolismo , Feminino , Perfilação da Expressão Gênica , Histiocitoma Fibroso Maligno/genética , Histiocitoma Fibroso Maligno/metabolismo , Histiocitoma Fibroso Maligno/patologia , Humanos , Masculino , Análise em Microsséries , Neurilemoma/genética , Neurilemoma/metabolismo , Neurilemoma/patologia , Neurotecoma/metabolismo , Neurotecoma/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Neoplasias do Sistema Nervoso Periférico/genética , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Pele/inervação , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Adulto Jovem
15.
Biochem Pharmacol ; 188: 114547, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33838132

RESUMO

Perineural invasion (PNI) is characterized by an encounter between the cancer cells and neuronal fibers and holds an extremely poor prognosis for malignant tumors. The exact molecular mechanism behind PNI yet remains to be explored. However, it is worth-noting that an involvement of the neuroactive molecules plays a major part in this process. A complex signaling network comprising the interplay between immunological cascades and neurogenic molecules such as tumor-derived neurotrophins, neuromodulators, and growth factors constitutes an active microenvironment for PNI associated with malignancy. The present review aims at discussing the following points in relation to PNI: a) Communication between PNI and neuroplasticity mechanisms can explain the pathophysiology of poor, short and long-term outcomes in cancer patients; b) Neuroactive molecules can significantly alter the neurons and cancer cells so as to sustain PNI progression; c) Finally, careful manipulation of neurogenic pathways and/or their crosstalk with the immunological molecules implicated in PNI could provide a potential breakthrough in cancer therapeutics.


Assuntos
Fatores de Crescimento Neural/metabolismo , Neurotransmissores/metabolismo , Nervos Periféricos/metabolismo , Nervos Periféricos/patologia , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Animais , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Fatores de Crescimento Neural/genética , Neurotransmissores/genética , Neoplasias do Sistema Nervoso Periférico/genética , Microambiente Tumoral/fisiologia
16.
Int J Cancer ; 126(2): 563-71, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19634141

RESUMO

Neurofibromatosis type 1 (NF1) patients are prone to the development of malignant tumors, the most common being Malignant Peripheral Nerve Sheath Tumor (MPNST). NF1-MPNST patients have an overall poor survival due to systemic metastasis. Currently, the management of MPNSTs includes surgery and radiation; however, conventional chemotherapy is not very effective, underscoring the need for effective biologically-targeted therapies. Recently, the NF1 gene product, neurofibromin, was shown to negatively regulate the phosphoinositide-3-kinase (PI3K)/Protein Kinase-B (Akt)/mammalian Target Of Rapamycin (mTOR) pathway, with loss of neurofibromin expression in established human MPNST cell lines associated with high levels of mTOR activity. We developed and characterized a human NF1-MPNST explant grown subcutaneously in NOD-SCID mice, to evaluate the effect of the mTOR inhibitor rapamycin. We demonstrate that rapamycin significantly inhibited human NF1-MPNST mTOR pathway activation and explant growth in vivo at doses as low as 1.0 mg/kg/day, without systemic toxicities. While rapamycin was effective at reducing NF1-MPNST proliferation and angiogenesis, with decreased CyclinD1 and VEGF respectively, there was no increase in tumor apoptosis. Rapamycin effectively decreased activation of S6 downstream of mTOR, but there was accompanied increased Akt activation. This study demonstrates the therapeutic potential and limitations of rapamycin in NF1-associated, and likely sporadic, MPNSTs.


Assuntos
Neurofibromatose 1/tratamento farmacológico , Neoplasias do Sistema Nervoso Periférico/tratamento farmacológico , Sirolimo/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Ciclina D1/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neurofibromatose 1/metabolismo , Neurofibromatose 1/patologia , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR , Carga Tumoral/efeitos dos fármacos , Fator A de Crescimento do Endotélio Vascular/metabolismo , Adulto Jovem
17.
World J Surg ; 34(5): 1039-45, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20119692

RESUMO

BACKGROUND: Angiogenesis and lymphangiogenesis are important processes in the progression of malignant tumors. Previous studies have shown that nerve growth factor-beta (NGF-beta) can promote the initiation and progression of many tumors. In addition, vascular endothelial growth factor-C (VEGF-C) has become recognized as the most important lymphangiogenic factor. In the present study, the expression of NGF-beta in human hilar cholangiocarcinoma and its relationship with lymphangiogenesis, lymph node metastasis, nerve infiltration, and VEGF-C expression was investigated. METHODS: Nerve growth factor-beta and VEGF-C expression were investigated by immunohistochemistry in samples from 28 cases of hilar cholangiocarcinoma. The lymphatic vessel density (LVD) in the tumor tissue that indicated lymphangiogenesis were calculated by immunostaining with the lymphendothelial-specific antibody D2-40. The relationship between NGF-beta expression and VEGF-C expression, lymphangiogenesis, lymph node metastasis, and nerve infiltration was evaluated. RESULTS: The overexpression of NGF-beta and VEGF-C occurred in 57.1% (16/28) and 46.4% (13/28) of tumor samples, respectively. Nerve growth factor-beta overexpression was highly correlated with VEGF-C overexpression (P = 0.005), LVD (P < 0.001), lymph node metastasis (P = 0.021), nerve infiltration (P = 0.019), and tumor stage (P = 0.040). Furthermore, VEGF-C overexpression was highly correlated with LVD (P < 0.001) and lymph node metastasis (P < 0.001). However, there was no statistic significance in the relation between NGF-beta expression and sex (P = 0.185), age (P = 0.387), maximal tumor size (P = 0.736), Bismuth classification (P = 0.627) as well as histological grade (P = 0.203). CONCLUSIONS: Nerve growth factor-beta might promote lymph node metastasis and nerve infiltration in human hilar cholangiocarcinoma.


Assuntos
Neoplasias dos Ductos Biliares/patologia , Ductos Biliares Intra-Hepáticos , Colangiocarcinoma/patologia , Linfonodos/patologia , Fator de Crescimento Neural/biossíntese , Neoplasias do Sistema Nervoso Periférico/patologia , Adulto , Idoso de 80 Anos ou mais , Neoplasias dos Ductos Biliares/metabolismo , Colangiocarcinoma/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Neoplasias do Sistema Nervoso Periférico/metabolismo , Fator C de Crescimento do Endotélio Vascular/biossíntese
18.
Genetika ; 46(3): 314-20, 2010 Mar.
Artigo em Russo | MEDLINE | ID: mdl-20391775

RESUMO

The protein Merlin is involved in the regulation of cell proliferation and differentiation in the eyes and wings of Drosophila and is a homolog of the human protein encoded by the Neurofibromatosis 2 (NF2) gene whose mutations cause auricular nerve tumors. Recent studies show that Merlin and Expanded cooperatively regulate the recycling of membrane receptors, such as the epidermal growth factor receptor (EGFR). By performing a search for potential genetic interactions between Merlin (Mer) and the genes important for vesicular trafficking, we found that ectopic expression in the wing pouch of the clathrin adapter protein Lap involved in clathrin-mediated receptor endocytosis resulted in the formation of extra vein materials. On the one hand, coexpression of wild-type Merlin and lap in the wing pouch restored normal venation, while overexpression of a dominant-negative mutant Mer(DBB) together with lap enhanced ectopic vein formation. Using various constructs with Merlin truncated copies, we showed the C-terminal portion of the Merlin protein to be responsible for the Merlin-lap genetic interaction. Furthermore, we showed that the Merlin and Lap proteins colocalized at the cortex of the wing imaginal disc cells.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Endocitose/fisiologia , Neurofibromina 2/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Proliferação de Células , Clatrina/genética , Clatrina/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Neoplasias da Orelha/genética , Neoplasias da Orelha/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Olho/embriologia , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neurofibromina 2/genética , Neoplasias do Sistema Nervoso Periférico/genética , Neoplasias do Sistema Nervoso Periférico/metabolismo , Receptores de Peptídeos de Invertebrados/genética , Receptores de Peptídeos de Invertebrados/metabolismo , Asas de Animais/embriologia
19.
Arkh Patol ; 72(4): 52-5, 2010.
Artigo em Russo | MEDLINE | ID: mdl-21086641

RESUMO

The authors report a rare a case of gliosarcoma metastases in a 28-year-old male patient. The cauda equina roots were involved after brain tumor 16 months ago, which, on microscopic study, had a biphasic pattern and heterogeneous staining in the reaction with antibody to GFAP and vimentin; the tumor cells did not express EMA, EA, and desmin. Gliosarcoma was diagnosed, by taking into account morphological and immunohistochemical data. Tumor tissue of the cauda equine roots had the same immunophenotype as the brain tumor with a predominance of glial component, which permitted the source of metastases to be ascertained.


Assuntos
Neoplasias Encefálicas , Cauda Equina/patologia , Gliossarcoma , Neoplasias do Sistema Nervoso Periférico , Adulto , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Cauda Equina/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Gliossarcoma/metabolismo , Gliossarcoma/patologia , Humanos , Masculino , Metástase Neoplásica , Proteínas de Neoplasias/metabolismo , Neoplasias do Sistema Nervoso Periférico/metabolismo , Neoplasias do Sistema Nervoso Periférico/patologia , Neoplasias do Sistema Nervoso Periférico/secundário , Vimentina/metabolismo
20.
J Neuroimmunol ; 207(1-2): 39-44, 2009 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-19174309

RESUMO

Dehydroepiandrosterone (DHEA) is an abundant adrenal steroid in serum of humans, and has been reported to have anti-inflammatory, anti-proliferative, and certain immune-regulating properties. Experimental autoimmune neuritis (EAN) is a Th1 cell-mediated animal model of Guillain-Barré syndrome (GBS) in humans. In the present study, DHEA was administered subcutaneously to Lewis rats immunized with bovine peripheral myelin (BPM) in Freund's complete adjuvant. Rats treated with DHEA displayed significant delay in onset, decreased inflammatory cell infiltration in the PNS. Benefit was associated with significant decreases in numbers of IFN-gamma and TNF-alpha expressing cells in the PNS, BPM-stimulated T cell proliferation and IFN-gamma, TNF-alpha-secretion in the spleen cells. Only 2 mg DHEA-treated EAN rats decreased peak clinical score. No significant difference of supernatant IL-10 was found among the treatment and control groups. These results suggest that DHEA can ameliorate the severity of EAN by suppressing the proliferation of autoreactive T cell and expression of pro-inflammatory cytokines.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Desidroepiandrosterona/administração & dosagem , Neurite Autoimune Experimental/tratamento farmacológico , Análise de Variância , Animais , Bovinos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Ensaio de Imunoadsorção Enzimática/métodos , Feminino , Adjuvante de Freund , Interferon-alfa/metabolismo , Interferon gama/metabolismo , Bainha de Mielina/imunologia , Neurite Autoimune Experimental/etiologia , Neurite Autoimune Experimental/imunologia , Neurite Autoimune Experimental/patologia , Neoplasias do Sistema Nervoso Periférico/metabolismo , Ratos , Ratos Endogâmicos Lew , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA