Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Endocrine ; 51(3): 524-33, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25982150

RESUMO

First-line treatment of patients with growth hormone secreting adenomas is surgical resection. Disease control can be obtained by surgery (one or multiple steps), in case followed by medical treatment or adjuvant radiation therapy (radiosurgery or radiotherapy). The impact of pre-surgical treatment with somatostatin analogs (SSAs) on surgical outcome is still controversial. The aim of this study is to retrospectively evaluate the impact of SSA pre-treatment on biochemical outcome and post-surgical hypopituitarism in a consecutive surgical series from a single referral centre, with data covering 17 years' experience and to investigate the possible predictive value of early postoperative insulin-like factor 1 (IGF-I) on long-term biochemical control. Data from 68 acromegalic patients were revised. Endocrinological long-term follow-up (minimum 6 months) was available for 57 patients. Eighty-eight percent of patients received a single-step surgical treatment (single surgery, with or without adjuvant medical therapy). The remaining 12% underwent a multi-step strategy: redo-surgery (three macroadenomas) and/or radiation (four macro- and two microadenomas). Pre-surgical SSA treatment was performed in 77.9% and resulted in a significant lowering of basal IGF-I values (p = 0.0001). Early post-surgical IGF-I was significantly lower in patients biochemically controlled with single surgery alone (p = 0.016) and after overall treatment strategies (p = 0.005). Normalization of GH and IGF-I was obtained in 56.1%, and normalization of either one of them in 27.8% of patients. No major surgery-related complications occurred. Post-treatment hypopituitarism occurred in 11.9% and was lower in SSA pre-treated patients. Our results well compare with other recently published series. Very early post-surgical IGF-I improvement might be a useful predictor for biochemical disease control. Moreover, our results suggest that pre-surgical treatment with somatostatin analogs seems to prevent hypopituitarism.


Assuntos
Acromegalia/tratamento farmacológico , Acromegalia/cirurgia , Antagonistas de Hormônios/uso terapêutico , Somatostatina/análogos & derivados , Somatostatina/uso terapêutico , Adolescente , Adulto , Idoso , Criança , Terapia Combinada , Feminino , Seguimentos , Hormônio do Crescimento/sangue , Adenoma Hipofisário Secretor de Hormônio do Crescimento/tratamento farmacológico , Adenoma Hipofisário Secretor de Hormônio do Crescimento/cirurgia , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Pessoa de Meia-Idade , Neoplasias Hipofisárias/tratamento farmacológico , Neoplasias Hipofisárias/cirurgia , Reoperação/estatística & dados numéricos , Estudos Retrospectivos , Resultado do Tratamento , Adulto Jovem
2.
Transl Res ; 160(5): 355-62, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22735029

RESUMO

Meningioma is one of the most common intracranial tumors and is graded according to the World Health Organization (WHO) classification system. Although these tumors are often surgically curable, a malignant behavior also may occur in meningiomas with benign histologic profiles (WHO I). Thus, it is mandatory to identify biomolecular parameters useful to improve the classification of these tumors. HOXA genes belong to the HOX gene family that encodes homeodomain-containing transcription factors known to be key regulators of embryonic development, involved in cell growth and differentiation and in the development of the central nervous system. Moreover, altered HOXA gene methylation and expression have prognostic value in many tumors. The purpose of this study was to determine whether the level of HOXA3, 7, 9, and 10 methylation in meningioma could be a biomarker linked to the pathologic characteristics of the tumor. We found that methylation levels of HOXA7, 9, and 10 in 131 meningioma samples were significantly higher in WHO II/III tumors compared with WHO I tumors. Moreover, in newly diagnosed WHO I meningiomas, HOXA7, 9, and 10 methylation was significantly lower than in WHO I samples derived from recurring tumors, and multiple meningiomas presented significantly higher HOXA 10 methylation with respect to solitary meningiomas. This study demonstrates that HOXA7, 9, and 10 are methylation targets in meningioma, associated with histopathology and clinical aggressiveness parameters. Our findings suggest the possibility of detecting the malignancy potential of meningioma by assessing the HOXA methylation level and identifying patients at higher risk who could benefit from closer follow-up or postoperative adjuvant treatments.


Assuntos
Proteínas de Homeodomínio/genética , Neoplasias Meníngeas/genética , Meningioma/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores , Metilação de DNA , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Pessoa de Meia-Idade , Adulto Jovem
3.
J Cancer Res Clin Oncol ; 138(1): 35-47, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21947269

RESUMO

PURPOSE: The purpose of this study was to determine whether specific HOXA epigenetic signatures could differentiate glioma with distinct biological, pathological, and clinical characteristics. METHODS: We evaluated HOXA3, 7, 9, and 10 methylation in 63 glioma samples by MassARRAY and pyrosequencing. RESULTS: We demonstrated the direct statistical correlation between the level of methylation of all HOXA genes examined and WHO grading. Moreover, in glioblastoma patients, higher level of HOXA9 and HOXA10 methylation significantly correlated with increased survival probability (HOXA9-HR: 0.36, P = 0.007; HOXA10-HR: 0.46, P = 0.045; combined HOXA9 and 10-HR 0.28, P = 0.004). CONCLUSIONS: This study identifies HOXA3, 7, 9, and 10 as methylation targets mainly in high-grade glioma and hypermethylation of the HOXA9 and 10 as prognostic factor in glioblastoma patients. Our data indicate that these epigenetic changes may be biomarkers of clinically different subgroups of glioma patients that could eventually benefit from personalized therapeutic strategies.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Metilação de DNA , Glioma/genética , Glioma/patologia , Proteínas de Homeodomínio/genética , Neoplasias Encefálicas/metabolismo , Cromossomos Humanos Par 7 , Análise por Conglomerados , Amplificação de Genes , Glioma/metabolismo , Proteínas Homeobox A10 , Proteínas de Homeodomínio/biossíntese , Humanos , Gradação de Tumores , Taxa de Sobrevida
4.
Pediatr Blood Cancer ; 57(2): 240-6, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21671360

RESUMO

BACKGROUND: Children treated with cranial radiotherapy (CRT) for leukemia are at risk of developing central nervous system injuries. Magnetic resonance imaging (MRI) represents the examination method of choice for evaluating radiation-induced brain complications. The purpose of this report is to describe the spectrum of MRI abnormalities detected in a group of survivors of leukemia treated with cranial irradiation. PROCEDURES: In this cross-sectional, single center study, 56 patients (median age at follow-up 19 years) receiving CRT as cranial prophylaxis (CP) included in the leukemia protocol (total dose 1,800-2,400 cGy) and/or in the total body irradiation regimen (990-1,200 cGy) before hematopoietic stem cell transplant, were evaluated by MRI after a median interval of 11 years (range 2-27) following CRT. RESULTS: Fifty-nine MRI abnormalities (32 cavernomas, nine focal areas of gliosis, seven dystrophic mineralizations, five cerebral atrophies, four pituitary atrophies, one diffuse radiation leukoencephalopathy, and one meningioma) were found in 43 patients. The longest interval between CRT and MRI and oldest age at follow-up represented the two risk factors that were statistically associated with MRI lesions (P = 0.032 and 0.033, respectively). Cerebral cavernomas (CC) were the most frequent MRI abnormalities (57%). All patients with CC were asymptomatic at diagnosis and during follow-up, except one who had aspecific neurological manifestations and micro hemorrhages. CONCLUSIONS: These results confirm that total doses and modalities of fractionation dose of CRT were not significantly associated with MRI abnormalities. Moreover, in our experience none of the patients developed neurological symptoms related to MRI abnormalities, and furthermore, the CC remained substantially stable during follow-up.


Assuntos
Doenças do Sistema Nervoso Central/epidemiologia , Doenças do Sistema Nervoso Central/patologia , Irradiação Craniana/efeitos adversos , Leucemia/radioterapia , Imageamento por Ressonância Magnética , Adolescente , Adulto , Doenças do Sistema Nervoso Central/etiologia , Neoplasias do Sistema Nervoso Central/epidemiologia , Neoplasias do Sistema Nervoso Central/etiologia , Neoplasias do Sistema Nervoso Central/patologia , Criança , Pré-Escolar , Estudos Transversais , Relação Dose-Resposta à Radiação , Feminino , Seguimentos , Hemangioma Cavernoso do Sistema Nervoso Central/epidemiologia , Hemangioma Cavernoso do Sistema Nervoso Central/etiologia , Hemangioma Cavernoso do Sistema Nervoso Central/patologia , Humanos , Lactente , Itália/epidemiologia , Masculino , Neoplasias Induzidas por Radiação/epidemiologia , Neoplasias Induzidas por Radiação/etiologia , Neoplasias Induzidas por Radiação/patologia , Fatores de Risco , Sobreviventes
5.
J Neuroimmunol ; 234(1-2): 115-23, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21316111

RESUMO

CXCR4 and CXCR7 chemokine receptors, and their ligands CXCL11 and CXCL12, have been often involved in tumor cell proliferation and survival. We report the expression pattern of these ligand/receptor pairs in 22 human meningiomas. High CXCR7 and CXCL12 expression was associated with high-proliferative tumors. CXCR7 levels were correlated to the content of both ligands, suggesting a possible autocrine regulation. CXCR4 and CXCL12 were homogeneously expressed within tumor cells, while CXCR7 was mainly detected in tumor endothelial cells and CXCL11 in pericytes. Our results highlight the preferential CXCR7 and CXCL12 expression within more aggressive tumors and the possible role of CXCR7 in meningioma vascularization.


Assuntos
Vasos Sanguíneos/metabolismo , Neoplasias Meníngeas/patologia , Meningioma/patologia , Receptores CXCR/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Antígenos CD34/metabolismo , Vasos Sanguíneos/patologia , Dura-Máter/metabolismo , Células Endoteliais/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Indóis , Antígeno Ki-67/metabolismo , Masculino , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Microvasos , Pessoa de Meia-Idade , RNA Mensageiro/metabolismo , Receptores CXCR/classificação , Receptores CXCR/genética , Estatística como Assunto
6.
Mol Cancer Res ; 7(3): 383-92, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19276180

RESUMO

It has been reported that cancer stem cells may contribute to glioma radioresistance through preferential activation of the DNA damage checkpoint response and an increase in DNA repair capacity. We have examined DNA repair in five stem and nonstem glioma cell lines. The population doubling time was significantly increased in stem compared with nonstem cells, and enhanced activation of Chk1 and Chk2 kinases was observed in untreated CD133(+) compared with CD133(-) cells. Neither DNA base excision or single-strand break repair nor resolution of pH2AX nuclear foci were increased in CD133(+) compared with CD133(-) cells. We conclude that glioma stem cells display elongated cell cycle and enhanced basal activation of checkpoint proteins that might contribute to their radioresistance, whereas enhanced DNA repair is not a common feature of these cells.


Assuntos
Neoplasias Encefálicas/genética , Reparo do DNA , Glioblastoma/genética , Células-Tronco Neoplásicas/fisiologia , Antígeno AC133 , Animais , Antígenos CD/biossíntese , Antígenos CD/genética , Apoptose/fisiologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Quinase 1 do Ponto de Checagem , Quinase do Ponto de Checagem 2 , Dano ao DNA , Ativação Enzimática , Glioblastoma/metabolismo , Glioblastoma/patologia , Glicoproteínas/biossíntese , Glicoproteínas/genética , Humanos , Cariotipagem , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Peptídeos/genética , Proteínas Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo
7.
J Immunol ; 182(6): 3530-9, 2009 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-19265131

RESUMO

In this study, cancer cells were isolated from tumor specimens of nine glioblastoma patients. Glioblastoma cells, cultured under suitable culture conditions, displayed markers typical of neural stem cells, were capable of partial multilineage differentiation in vitro, and gave origin to infiltrating tumors when orthotopically injected in NOD/SCID mice. These cells, although resistant to freshly isolated NK cells, were highly susceptible to lysis mediated by both allogeneic and autologous IL-2 (or IL-15)-activated NK cells. Indeed, all stem cell-cultured glioblastoma cells analyzed did not express protective amounts of HLA class I molecules, while expressing various ligands of activating NK receptors that triggered optimal NK cell cytotoxicity. Importantly, glioblastoma stem cells expressed high levels of PVR and Nectin-2, the ligands of DNAM-1-activating NK receptor.


Assuntos
Citotoxicidade Imunológica , Glioblastoma/imunologia , Glioblastoma/patologia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/patologia , Células-Tronco Neoplásicas/imunologia , Células-Tronco Neoplásicas/patologia , Animais , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Glioblastoma/metabolismo , Humanos , Imunidade Inata , Células Matadoras Naturais/metabolismo , Ligantes , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Linfócitos do Interstício Tumoral/transplante , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Células-Tronco Neoplásicas/metabolismo , Receptores de Células Matadoras Naturais/biossíntese , Receptores de Células Matadoras Naturais/genética , Receptores de Células Matadoras Naturais/fisiologia , Células Tumorais Cultivadas
8.
J Biol Chem ; 284(11): 7138-48, 2009 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-19147502

RESUMO

Because a subpopulation of cancer stem cells (tumor-initiating cells, TICs) is believed to be responsible for the development, progression, and recurrence of many tumors, we evaluated the in vitro sensitivity of human glioma TICs to epidermal growth factor receptor (EGFR) kinase inhibitors (erlotinib and gefitinib) and possible molecular determinants for their effects. Cells isolated from seven glioblastomas (GBM 1-7) and grown using neural stem cell permissive conditions were characterized for in vivo tumorigenicity, expression of tumor stem cell markers (CD133, nestin), and multilineage differentiation properties, confirming that these cultures are enriched in TICs. TIC cultures were challenged with increasing concentrations of erlotinib and gefitinib, and their survival was evaluated after 1-4 days. In most cases, a time- and concentration-dependent cell death was observed, although GBM 2 was completely insensitive to both drugs, and GBM 7 was responsive only to the highest concentrations tested. Using a radioligand binding assay, we show that all GBM TICs express EGFR. Erlotinib and gefitinib inhibited EGFR and ERK1/2 phosphorylation/activation in all GBMs, irrespective of the antiproliferative response observed. However, under basal conditions GBM 2 showed a high Akt phosphorylation that was completely insensitive to both drugs, whereas GBM 7 was completely insensitive to gefitinib, and Akt inactivation occurred only for the highest erlotinib concentration tested, showing a precise relationship with the antiproliferative effects of the drug. Interestingly, in GBM 2, phosphatase and tensin homolog expression was significantly down-regulated, possibly accounting for the insensitivity to the drugs. In conclusion, glioma TICs are responsive to anti-EGFR drugs, but phosphatase and tensin homolog expression and Akt inhibition seem to be necessary for such effect.


Assuntos
Receptores ErbB/metabolismo , Glioma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Idoso , Animais , Receptores ErbB/antagonistas & inibidores , Cloridrato de Erlotinib , Feminino , Gefitinibe , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioma/tratamento farmacológico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas dos Microfilamentos/biossíntese , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tensinas , Fatores de Tempo , Células Tumorais Cultivadas
9.
Clin Cancer Res ; 14(16): 5022-32, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18698020

RESUMO

PURPOSE: Hypothalamic or locally produced growth factors and cytokines control pituitary development, functioning, and cell division. We evaluated the expression of the chemokine stromal cell-derived factor 1 (SDF1) and its receptor CXCR4 in human pituitary adenomas and normal pituitary tissues and their role in cell proliferation. EXPERIMENTAL DESIGN: The expression of SDF1 and CXCR4 in 65 human pituitary adenomas and 4 human normal pituitaries was determined by reverse transcription-PCR, immunohistochemistry, and confocal immunofluorescence. The proliferative effect of SDF1 was evaluated in eight fibroblast-free human pituitary adenoma cell cultures. RESULTS: CXCR4 mRNA was expressed in 92% of growth hormone (GH)-secreting pituitary adenomas (GHoma) and 81% of nonfunctioning pituitary adenomas (NFPA), whereas SDF1 was identified in 63% and 78% of GHomas and NFPAs, respectively. Immunostaining for CXCR4 and SDF1 showed a strong homogenous labeling in all tumoral cells in both GHomas and NFPAs. In normal tissues, CXCR4 and SDF1 were expressed only in a subset of anterior pituitary cells, with a lower expression of SDF1 compared with its cognate receptor. CXCR4 and SDF1 were not confined to a specific cell population in the anterior pituitary but colocalized with discrete subpopulations of GH-, prolactin-, and adrenocorticorticotropic hormone-secreting cells. Conversely, most of the SDF1-containing cells expressed CXCR4. In six of eight pituitary adenoma primary cultures, SDF1 induced a statistically significant increase in DNA synthesis that was prevented by the treatment with the CXCR4 antagonist AMD3100 or somatostatin. CONCLUSIONS: CXCR4 and SDF1 are overexpressed in human pituitary adenomas and CXCR4 activation may contribute to pituitary cell proliferation and, possibly, to adenoma development in humans.


Assuntos
Quimiocina CXCL12/biossíntese , Neoplasias Hipofisárias/metabolismo , Receptores CXCR4/biossíntese , Proliferação de Células , Imunofluorescência , Humanos , Imuno-Histoquímica , Hibridização In Situ , Microscopia Confocal , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
10.
Endocr Relat Cancer ; 15(2): 583-96, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18509006

RESUMO

Dopamine D2 and somatostatin receptors (sstrs) were reported to affect non-functioning pituitary adenoma (NFPA) proliferation in vitro. However, the reported results differ according to the experimental conditions used. We established an experimental protocol allowing reproducible evaluation of NFPA cell proliferation in vitro, to test and compare the antiproliferative effects of dopamine and somatostatin analogs (alone or in combination) with the activity of the dopamine-somatostatin chimeric molecule BIM-23A760. The protocol was utilized by four independent laboratories, studying 38 fibroblast-deprived NFPA cell cultures. Cells were characterized for GH, POMC, sstr1-sstr5, total dopamine D2 receptor (D2R) (in all cases), and D2 receptor long and short isoforms (in 15 out of 38 cases) mRNA expression and for alpha-subunit, LH, and FSH release. D2R, sstr3, and sstr2 mRNAs were consistently observed, with the dominant expression of D2R (2.9+/-2.6 copy/copy beta-glucuronidase; mean+/-s.e.m.), when compared with sstr3 and sstr2 (0.6+/-1.0 and 0.3+/-0.6 respectively). BIM-23A760, a molecule with high affinity for D2R and sstr2, significantly inhibited [3H]thymidine incorporation in 23 out of 38 (60%) NFPA cultures (EC50=1.2 pM and Emax=-33.6+/-3.7%). BIM-23A760 effects were similar to those induced by the selective D2R agonist cabergoline that showed a statistically significant inhibition in 18 out of 27 tumors (compared with a significant inhibition obtained in 17 out of 27 tumors using BIM-23A760, in the same subgroup of adenomas analyzed), while octreotide was effective in 13 out of 27 cases. In conclusion, superimposable data generated in four independent laboratories using a standardized protocol demonstrate that, in vitro, chimeric dopamine/sstr agonists are effective in inhibiting cell proliferation in two-thirds of NFPAs.


Assuntos
Adenoma/tratamento farmacológico , Adenoma/patologia , Dopamina/análogos & derivados , Neoplasias Hipofisárias/tratamento farmacológico , Neoplasias Hipofisárias/patologia , Somatostatina/análogos & derivados , Adulto , Idoso , Antineoplásicos Hormonais/farmacologia , Cabergolina , Divisão Celular/efeitos dos fármacos , Dopamina/farmacologia , Antagonistas de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Ergolinas/farmacologia , Feminino , Fibroblastos/citologia , Humanos , Masculino , Pessoa de Meia-Idade , Octreotida/farmacologia , RNA Mensageiro/metabolismo , Receptores de Dopamina D2/genética , Receptores de Somatostatina/genética , Somatostatina/farmacologia , Sulpirida/farmacologia , Timidina/metabolismo , Trítio , Células Tumorais Cultivadas
11.
Pituitary ; 11(1): 93-102, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17458701

RESUMO

Herein we report a rare case of a pituitary metastasis from a neuroendocrine tumour mimicking an adenoma. Moreover, starting from this unusual case, the relevant literature concerning the diagnosis and management of patients with metastasis at pituitary level is reviewed. A 69-year-old woman was admitted to our Unit for severe headache, diplopia, and critical visual field impairment. MRI showed a large pituitary mass compressing the optic chiasm and infiltrating the cavernous sinus. Trans-sphenoidal biopsy revealed a pituitary metastasis from a neuroendocrine tumour, in line with the multiple liver lesions that were already considered metastases from an ileal primary neuroendocrine tumour. In vitro receptor characterisation of both pituitary and liver tissues by immunohistochemistry showed a heterogeneous somatostatin receptor subtype pattern, with a predominant expression of sst(2) within the pituitary lesion. However, the liver metastasis receptor profile was completely different from the pituitary. Octreotide LAR was administered first, followed by receptor radiometabolic therapy with radiolabelled somatostatin analogues ((90)Y-DOTATOC and (177)Lu-DOTATATE). After 16 months, MRI showed a significant shrinkage of the sellar mass. Moreover, disappearance of diplopia and visual defects, together with a considerable improvement in quality of life were gradually recorded. To our knowledge, this is the first case of combined treatment using "cold" and radiolabelled octreotide in a pituitary metastasis from a neuroendocrine tumour.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Neoplasias do Íleo/patologia , Tumores Neuroendócrinos/terapia , Octreotida/análogos & derivados , Octreotida/uso terapêutico , Compostos Organometálicos/uso terapêutico , Neoplasias Hipofisárias/terapia , Compostos Radiofarmacêuticos/uso terapêutico , Adenoma/diagnóstico , Adulto , Idoso , Biópsia , Preparações de Ação Retardada , Diagnóstico Diferencial , Diplopia/etiologia , Diplopia/terapia , Feminino , Cefaleia/etiologia , Cefaleia/terapia , Humanos , Imuno-Histoquímica , Neoplasias Hepáticas/secundário , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/complicações , Tumores Neuroendócrinos/secundário , Neoplasias Hipofisárias/complicações , Neoplasias Hipofisárias/secundário , Qualidade de Vida , Cintilografia , Fatores de Tempo , Resultado do Tratamento
12.
Neurosurgery ; 61(2): E418-9; discussion E419, 2007 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-17762725

RESUMO

OBJECTIVE: Idiopathic myelofibrosis is a clonal stem cell disorder that leads to ineffective erythropoiesis accompanied by reactive myelofibrosis (bone marrow fibrosis). As a consequence, extramedullary hematopoiesis characteristically develops. The central nervous system is rarely affected; the spinal canal and the cranial meninges are generally the preferred locations. Extramedullary hematopoiesis within central nervous system primary tumors have already been reported but, to our knowledge, never before in a patient with evidence of idiopathic myelofibrosis. CLINICAL PRESENTATION: A patient experiencing generalized idiopathic myelofibrosis developed a hemorrhagic intracranial meningioma containing islets of extramedullary myeloid metaplasia. INTERVENTION: The tumor was radically removed through a right frontal craniotomy. After surgery, the patient recovered completely and was discharged with a normal neurological status. After 6 years, the patient is in excellent condition with no sign of recurrence on magnetic resonance imaging scans. CONCLUSION: The reasons for this uncommon association are uncertain, but we hypothesize that myeloid islets may be involved in the origin of the tumor as well as in its acute hemorrhagic onset. Moreover, we suggest that in the presence of proven idiopathic myelofibrosis intracranial myeloid metaplasia should be ruled out by appropriate neuroimaging and considered as a potential diagnosis in the presence of brain lesions.


Assuntos
Neoplasias Encefálicas/patologia , Hematopoese Extramedular , Neoplasias Meníngeas/patologia , Meningioma/patologia , Mielofibrose Primária/patologia , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/cirurgia , Craniotomia , Humanos , Imageamento por Ressonância Magnética , Masculino , Neoplasias Meníngeas/diagnóstico por imagem , Neoplasias Meníngeas/cirurgia , Meningioma/diagnóstico por imagem , Meningioma/cirurgia , Pessoa de Meia-Idade , Tomografia Computadorizada por Raios X
14.
J Clin Endocrinol Metab ; 92(5): 1592-9, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17311860

RESUMO

CONTEXT: Criteria to define the response to somatostatin (SS) analogs (SSA) in acromegaly are based on biochemical control of the disease. However, the mechanisms of action of SSAs in inhibiting tumor growth and hormonal secretion are only partially understood, and the two effects may occur independently. OBJECTIVE: The objective of the study was to investigate the dissociation between antiproliferative and antisecretive effects of SSA in an octreotide-resistant patient displaying dramatic tumor shrinkage during primary therapy with octreotide LAR. DESIGN AND SETTING: We characterized somatostatin and dopamine D(2) receptor expression by immunohistochemistry and real-time RT-PCR. The effects of different receptor-selective, bispecific analogs, and chimeric somatostatin/dopamine compounds on GH secretion and cell proliferation in primary cell cultures of the tumor were assessed. RESULTS: The expression of SS receptor subtypes (sst)(5) and D(2) receptor was higher, compared with the other receptor subtypes. GH inhibition by SS-14 and the two chimeric somatostatin/dopamine compounds was scant but greater than subtype-selective and sst(2)/sst(5) bispecific agonists. Conversely, cell growth was potently inhibited by all test substances. However, SS-14, sst(2)/sst(5) bispecific agonist, and chimeric molecules were more potent than the other compounds. CONCLUSIONS: The significant antiproliferative effect of octreotide seems to be related to the higher expression of sst(5) and the negligible antihormonal effect to the lower expression of sst(2). However, activation of multiple receptors by new analogs may produce better control of tumor cell activities. The dissociation between antisecretive and antiproliferative effects observed in vivo and in vitro confirms that SSAs may induce tumor shrinkage despite the lack of effect on GH secretion.


Assuntos
Acromegalia/tratamento farmacológico , Adenoma Acidófilo/tratamento farmacológico , Octreotida/uso terapêutico , Neoplasias Hipofisárias/tratamento farmacológico , Acromegalia/metabolismo , Acromegalia/patologia , Adenoma Acidófilo/metabolismo , Adenoma Acidófilo/patologia , Adulto , Cabergolina , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Preparações de Ação Retardada , Ergolinas/uso terapêutico , Hormônio do Crescimento Humano/sangue , Humanos , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/metabolismo , Imageamento por Ressonância Magnética , Masculino , Microscopia Eletrônica , Octreotida/administração & dosagem , Neoplasias Hipofisárias/metabolismo , Neoplasias Hipofisárias/patologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Somatostatina/genética , Timidina/metabolismo
15.
J Clin Neurosci ; 14(4): 355-8, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17240151

RESUMO

Forty-four evaluable patients with intracranial meningiomas were assessed for the expression of the cell-cycle regulator cyclin D1 and of proteins involved in proliferation and apoptosis such as PCNA, MIB-1, p53 and bcl-2. Analyses were carried out by western blot and immunohistochemistry after immediate processing of fresh tumor specimens. By western blot, expression of cyclin D1 significantly correlated with p53 (p=0.02) and with proliferative activity, as assessed by PCNA expression (p=0.0009). By immunohistochemistry, a significant relationship between cyclin D1 and the proliferation marker MIB-1 was confirmed (p=0.05), whereas significance with bcl-2 expression was not found (p=0.01). Moreover, although the association with tumor grade appeared of borderline statistical significance (p=0.07), all the grade II/III meningiomas showed increased expression of cyclin D1 and high proliferative activity. In conclusion, data from this preliminary study seem to suggest a potential value of the combined expression of cyclin D1 and proliferation indicators in defining subgroups of meningiomas with a more aggressive biological behavior.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/metabolismo , Ciclina D1/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Meningioma/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Proliferação de Células , Feminino , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Masculino , Pessoa de Meia-Idade , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Índice de Gravidade de Doença , Proteína Supressora de Tumor p53/metabolismo
16.
Neuro Oncol ; 9(1): 3-11, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17108064

RESUMO

Chemokines participate in cellular processes associated with tumor proliferation, migration, and angiogenesis. We previously demonstrated that stromal cell-derived factor 1 (SDF1) exerts a mitogenic activity in glioblastomas through the activation of its receptor CXCR4. Here we studied the expression of this chemokine in human meningiomas and its possible role in cell proliferation. Reverse transcriptase-PCR analysis for CXCR4 and SDF1 was performed on 55 human meningiomas (47 WHO grade I, 5 WHO II, and 3 WHO III). Immunolabeling for CXCR4 and SDF1 was performed on paraffin-embedded sections of these tumors. [(3)H]Thymidine uptake and Western blot analyses were performed on primary meningeal cell cultures of tumors to evaluate the proliferative activity of human SDF1alpha (hSDF1alpha) in vitro and the involvement of extracellular signal-regulated kinase 1/2 (ERK1/2) activation in this process. CXCR4 mRNA was expressed by 78% of the tumor specimens and SDF1 mRNA by 53%. CXCR4 and SDF1 were often detected in the same tumor tissues and colocalized with epithelial membrane antigen immunostaining. In 9 of 12 primary cultures from meningiomas, hSDF1alpha induced significant cell proliferation that was strongly reduced by the mitogen-activated protein kinase kinase inhibitor PD98059, involving ERK1/2 activation in the proliferative signal of hSDF1alpha. In fact, CXCR4 stimulation led to ERK1/2 phosphorylation/activation. In addition, the hSDF1alpha-induced cell proliferation was significantly correlated with the MIB1 staining index in the corresponding surgical specimen. In conclusion, we found that human meningiomas express CXCR4 and SDF1 and that hSDF1alpha induces proliferation in primary meningioma cell cultures through the activation of ERK1/2.


Assuntos
Proliferação de Células , Quimiocinas CXC/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Receptores CXCR4/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Western Blotting , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patologia , Quimiocina CXCL12 , Quimiocinas CXC/genética , Feminino , Humanos , Técnicas Imunoenzimáticas , Técnicas In Vitro , Masculino , Neoplasias Meníngeas/genética , Meningioma/genética , Meningioma/patologia , Pessoa de Meia-Idade , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Receptores CXCR4/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Células Estromais/metabolismo , Células Tumorais Cultivadas
17.
J Pediatr Endocrinol Metab ; 19 Suppl 1: 381-8, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16700314

RESUMO

The transsphenoidal approach has specific indications in the management of craniopharyngiomas. Usually, it is best reserved for patients with preferably cystic extra-arachnoid-infradiaphragmatic tumors with small suprasellar extension. Moreover, it is definitely less traumatic than transcranial approaches and it has been proven to be feasible also in paediatric patients. When possible, radical removal of these tumours must be the goal of surgery, but this attitude, which reduces but not eliminates the risk of relapse, has to be counterbalanced by heavy morbidity and even mortality, especially in children. In this view, many neurosurgeons favour a more 'conservative' approach with subtotal removal followed by radiotherapy whose dramatic efficacy on craniopharyngiomas is well known. With these premises, a transsphenoidal approach is realistically applicable to a greater number of large cystic craniopharyngiomas if the aim is not radical removal, but is to drain them into the sphenoid sinus to relieve mass effect symptoms (cystosphenoidostomy), and delay radiotherapy and its detrimental effects on visual and pituitary function, especially in younger patients, to a more suitable time after surgery.


Assuntos
Craniofaringioma/cirurgia , Procedimentos Neurocirúrgicos , Neoplasias Hipofisárias/cirurgia , Osso Esfenoide , Atividades Cotidianas , Adolescente , Criança , Feminino , Humanos , Masculino , Procedimentos Neurocirúrgicos/efeitos adversos , Procedimentos Neurocirúrgicos/mortalidade , Seio Esfenoidal/fisiologia , Resultado do Tratamento
18.
Neurochem Int ; 49(5): 423-32, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16621164

RESUMO

Chemokines have been involved in cellular processes associated to malignant transformation such as proliferation, migration and angiogenesis. The expression of five CXC chemokine receptors and their main ligands was analysed by RT-PCR in 31 human astrocytic neoplasms. The mRNAs for all the receptors analysed were identified in a high percentage of tumours, while their ligands showed lower expression. CXCR4 and SDF1 were the most frequently mRNA identified (29/31 and 13/31 of the gliomas studied, respectively). Thus, we further analysed the cell localization of CXCR4 and SDF1 in immunohistochemistry experiments. We show a marked co-localization of CXCR4 and SDF1 in tumour cells, mainly evident in psudolpalisade and microcystic degeneration areas and in the vascular endothelium. In addition, hSDF1alpha induced a significant increase of DNA synthesis in primary human glioblastoma cell cultures and chemotaxis in a glioblastoma cell line. These results provide evidence of the expression of multiple CXC chemokines and their receptors in brain tumours and that in particular CXCR4 and SDF1 sustain proliferation and migration of glioma cells to promote malignant progression.


Assuntos
Neoplasias Encefálicas/metabolismo , Movimento Celular/fisiologia , Proliferação de Células , Quimiocinas CXC/fisiologia , Glioma/metabolismo , Receptores de Quimiocinas/metabolismo , Sequência de Bases , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Quimiocinas CXC/metabolismo , Primers do DNA , Glioma/patologia , Humanos , Imuno-Histoquímica , Ligantes , Receptores de Quimiocinas/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Mol Pharmacol ; 69(2): 539-46, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16258074

RESUMO

We used GH4C1 cells as a model to study the effects of the chemokine stromal cell-derived factor 1 (SDF1) in pituitary functions. In these cells, SDF1alpha induced proliferation and growth hormone secretion, suggesting a possible regulatory role for this chemokine at pituitary level. We evaluated the intracellular signaling involved in these effects: SDF1alpha increased cytosolic [Ca(2+)] and activated Pyk2, extracellular signal-regulated kinases 1 and 2 (ERK1/2), and large-conductance Ca(2+)-activated K(+) channels (BK(Ca)) channels. To correlate these intracellular effectors with the proliferative and secretory effects, we inhibited their activity using BAPTA-AM (Ca(2+) chelator), 2'-amino-3'-methoxyflavone (PD98059; a mitogen-activated protein kinase kinase inhibitor), salicylate (Pyk2 inhibitor), and tetraethyl ammonium (K(+) channel blocker). All of these compounds reverted SDF1alpha-induced proliferation, suggesting the involvement of multiple intracellular pathways. Conversely, only BAPTA-AM reverted growth hormone secretion. To identify a possible cross-talk and a molecular ordering among these pathways, we tested these antagonists on SDF1alpha-dependent activation of ERK1/2, Pyk2, and BK(Ca) channels. From these experiments, we observed that the inhibition of [Ca(2+)](i) increase or BK(Ca) channel activity did not affect ERK1/2 activation by SDF1alpha; Pyk2 activation was purely Ca(2+)-dependent, not involving ERK1/2 or BK(Ca) channels; and BK(Ca) channel activity was antagonized by Pyk2 but not by ERK1/2 inhibitors. These data suggest that an SDF1alpha-dependent increase of [Ca(2+)](i) activates Pyk2, which in turn regulates BK(Ca) channel activity. Conversely, ERK1/2 activation is an independent phenomenon. In conclusion, we demonstrate that SDF1alpha causes both proliferation and growth hormone release from pituitary adenoma cells, suggesting that the activation of CXCR4 may represent a novel regulatory mechanism for growth hormone secretion and pituitary cell proliferation, which may contribute to pituitary adenoma development.


Assuntos
Adenoma/metabolismo , Quimiocinas CXC/farmacologia , Hormônio do Crescimento/metabolismo , Hipófise/efeitos dos fármacos , Neoplasias Hipofisárias/metabolismo , Receptores CXCR4/agonistas , Animais , Cálcio/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células , Quimiocina CXCL12 , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Hipófise/metabolismo , Hipófise/fisiologia , Ratos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Ann N Y Acad Sci ; 1090: 332-43, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17384278

RESUMO

Recent evidence indicates that cancer cells express chemokine (CK) receptors and that their signaling is crucial for tumor proliferation, migration, and angiogenesis. The profiles of expression of CXC CK receptors (CXCR1-5) and their main ligands (growth-related oncogene, GRO1-2-3/CXCL1-2-3; interleukin 8, IL-8/CXCL8; monokine-induced gamma-interferon MIG/CXCL9; gamma-interferon-inducible-protein-10, IP-10/CXCL10; stromal cell-derived factor-1, SDF1/CXCL12; B-cell activating CK-1, BCA-1/CXCL13) were analyzed by reverse transcription polymerase chain reaction (RT-PCR) in surgical samples of human meningiomas. All the five receptors displayed high percentages of positive cases: 92% CXCR1, 89% CXCR2, 83% CXCR3, 78% CXCR4, and 94% CXCR5. Conversely, their ligands showed a lower pattern of expression: 40% IL-8, 42% GRO1-3, 42% IP-10, 28% MIG, 53% SDF1, and 3% BCA-1. SDF1/CXCR4 interaction plays a pivotal role in cancer proliferation. Thus, the signaling mechanisms activated by the exclusive binding between SDF1 and CXCR4 was investigated in 12 primary cultures from meningioma tissues. CXCR4 was functionally coupled as demonstrated by the significant increase of DNA synthesis in meningioma cells in response to SDF1, measured by [3H]-thymidine uptake. In three primary cultures, the SDF1-dependent mitogenic activity was associated with a marked phosphorylation of extracellular signal-regulated kinase (ERK1/2) as evaluated by Western blots. PD98059 (a MEK inhibitor) significantly reduced ERK1/2 activation, thus linking the SDF1/CXCR4 pathway to meningioma cell proliferation via ERK1/2 signal transduction. We demonstrate, for the first time in human meningiomas, the simultaneous expression of CXCR1-5 and their CKs and the mitogenic activity of SDF1/CXCR4, suggesting a pivotal role of these receptor-ligand pairs in meningeal tumors.


Assuntos
Proliferação de Células , Quimiocinas CXC/metabolismo , Meningioma/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptores CXCR4/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Quimiocina CXCL12 , Primers do DNA , Ativação Enzimática , Feminino , Humanos , Masculino , Meningioma/enzimologia , Meningioma/patologia , Pessoa de Meia-Idade , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA