Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Neurosurg Focus ; 41(6): E14, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27903123

RESUMO

OBJECTIVE In this study, the authors investigated the underlying mechanisms responsible for high tumor recurrence rates of adamantinomatous craniopharyngioma (ACP) after radiotherapy and developed new targeted treatment protocols to minimize recurrence. ACPs are characterized by the activation of the receptor tyrosine kinase epidermal growth factor receptor (EGFR), known to mediate radioresistance in various tumor entities. The impact of tyrosine kinase inhibitors (TKIs) gefitinib or CUDC-101 on radiation-induced cell death and associated regulation of survivin gene expression was evaluated. METHODS The hypothesis that activated EGFR promotes radioresistance in ACP was investigated in vitro using human primary cell cultures of ACP (n = 10). The effects of radiation (12 Gy) and combined radiochemotherapy on radiosensitivity were assessed via cell death analysis using flow cytometry. Changes in target gene expression were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR). Survivin, identified in qRT-PCR to be involved in radioresistance of ACP, was manipulated by small interfering RNA (siRNA), followed by proliferation and vitality assays to further clarify its role in ACP biology. Immunohistochemically, survivin expression was assessed in patient tumors used for primary cell cultures. RESULTS In primary human ACP cultures, activation of EGFR resulted in significantly reduced cell death levels after radiotherapy. Treatment with TKIs alone and in combination with radiotherapy increased cell death response remarkably, assessed by flow cytometry. CUDC-101 was significantly more effective than gefitinib. The authors identified regulation of survivin expression after therapeutic intervention as the underlying molecular mechanism of radioresistance in ACP. EGFR activation promoting ACP cell survival and proliferation in vitro is consistent with enhanced survivin gene expression shown by qRT-PCR. TKI treatment, as well as the combination with radiotherapy, reduced survivin levels in vitro. Accordingly, ACP showed reduced cell viability and proliferation after survivin downregulation by siRNA. CONCLUSIONS These results indicate an impact of EGFR signaling on radioresistance in ACP. Inhibition of EGFR activity by means of TKI treatment acts as a radiosensitizer on ACP tumor cells, leading to increased cell death. Additionally, the results emphasize the antiapoptotic and pro-proliferative role of survivin in ACP biology and its regulation by EGFR signaling. The suppression of survivin by treatment with TKI and combined radiotherapy represents a new promising treatment strategy that will be further assessed in in vivo models of ACP.


Assuntos
Craniofaringioma/metabolismo , Proteínas Inibidoras de Apoptose/antagonistas & inibidores , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias Hipofisárias/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Tolerância a Radiação/efeitos dos fármacos , Adolescente , Adulto , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Sobrevivência Celular/efeitos da radiação , Criança , Craniofaringioma/patologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/fisiologia , Regulação para Baixo/efeitos da radiação , Feminino , Humanos , Proteínas Inibidoras de Apoptose/efeitos da radiação , Masculino , Pessoa de Meia-Idade , Neoplasias Hipofisárias/patologia , Tolerância a Radiação/fisiologia , Survivina , Células Tumorais Cultivadas
2.
Pituitary ; 17(6): 546-56, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24356780

RESUMO

INTRODUCTION: Early disease onset, clinical manifestation, histomorphology, and increased tendency to relapse distinguish the adamantinomatous craniopharyngioma (adaCP) from the more favorable papillary variant (papCP). A molecular hallmark of adaCP is the activated Wnt signaling pathway indicated by nuclear ß-catenin accumulation in a subset of tumor cells. A mouse model recently illustrated that these cells are the driving force in tumorigenesis of adaCP. This observation and the peculiar growth pattern points to the existence of a specific tumor stem cell (TSC) population in human CP. MATERIALS AND METHODS: To prove this hypothesis, the TSC markers CD133 (Prominin1) and CD44 were examined in papCP (n = 8) and adaCP (n = 25) on mRNA level using quantitative real time PCR of total tumor RNA. Furthermore, we investigated protein expression performing immunohistochemical analyses of formalin-fixed paraffin embedded tumor samples. RESULTS: PapCP revealed a homogenous CD44 expression pattern predominantly at the cell membrane, whereas CD133 labeling was hardly detectable. In adaCP, on the other hand all markers were consistently and predominantly co-expressed in nuclear ß-catenin accumulating cell clusters, which was confirmed by double immunofluorescence staining. Overall expression of CD44 was significantly decreased in adaCP versus papCP, whereas CD133 showed significantly higher protein and mRNA levels in adaCP. CONCLUSIONS: Our results indicate tumor stem cell-like characteristics of ß-catenin accumulating cell clusters in adaCP, which may represent a tumor stem cell niche and might contribute to tumor recurrence. The potential impact of these special cell groups in regard to future CP management, including postoperative follow-up and additional treatment remains to be explored.


Assuntos
Biomarcadores Tumorais , Craniofaringioma/patologia , Células-Tronco Neoplásicas/patologia , Neoplasias Hipofisárias/patologia , Via de Sinalização Wnt , Antígeno AC133 , Adolescente , Adulto , Antígenos CD/metabolismo , Criança , Pré-Escolar , Receptores ErbB/metabolismo , Éxons , Proteínas da Matriz Extracelular/metabolismo , Feminino , Glicoproteínas/metabolismo , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Masculino , Pessoa de Meia-Idade , Osteopontina/metabolismo , Peptídeos/metabolismo , Transdução de Sinais , Adulto Jovem , beta Catenina/metabolismo
3.
Nat Commun ; 8(1): 1819, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29180744

RESUMO

Senescent cells may promote tumour progression through the activation of a senescence-associated secretory phenotype (SASP), whether these cells are capable of initiating tumourigenesis in vivo is not known. Expression of oncogenic ß-catenin in Sox2+ young adult pituitary stem cells leads to formation of clusters of stem cells and induction of tumours resembling human adamantinomatous craniopharyngioma (ACP), derived from Sox2- cells in a paracrine manner. Here, we uncover the mechanisms underlying this paracrine tumourigenesis. We show that expression of oncogenic ß-catenin in Hesx1+ embryonic precursors also results in stem cell clusters and paracrine tumours. We reveal that human and mouse clusters are analogous and share a common signature of senescence and SASP. Finally, we show that mice with reduced senescence and SASP responses exhibit decreased tumour-inducing potential. Together, we provide evidence that senescence and a stem cell-associated SASP drive cell transformation and tumour initiation in vivo in an age-dependent fashion.


Assuntos
Senescência Celular/fisiologia , Craniofaringioma/metabolismo , Células-Tronco Neoplásicas/metabolismo , Neoplasias Hipofisárias/metabolismo , Compostos de Anilina/farmacologia , Animais , Compostos de Bifenilo/farmacologia , Transformação Celular Neoplásica , Criança , Craniofaringioma/patologia , Modelos Animais de Doenças , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Nitrofenóis/farmacologia , Oncogenes/fisiologia , Piperazinas/farmacologia , Hipófise/metabolismo , Hipófise/patologia , Neoplasias Hipofisárias/patologia , Proteínas Repressoras/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Sulfonamidas/farmacologia , Sequenciamento do Exoma , Adulto Jovem , beta Catenina/metabolismo
4.
Brain Pathol ; 25(1): 1-10, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24716541

RESUMO

Adamantinomatous craniopharyngiomas (adaCP) cause hypothalamic pituitary dysfunction. Elucidation of pathomechanisms underlying tumor progression is essential for the development of targeted chemotherapeutic treatment options. In order to study the mechanisms of tumor outgrowth, we implanted human primary adaCP tissue from three different surgical specimens stereotactically into the brain of immunodeficient mice (n = 20). Three months after tumor inoculation, magnetic resonance imaging and histology confirmed tumor engraftment in all 20 mice (100%) that obtained tissue transplants. The lesions invaded adjoining brain tissue with micro finger-shaped protrusions. Immunohistochemical comparison of the primary tumor and xenotransplants revealed a similar amount of proliferation (Mib-1) and cytokeratin expression pattern (KL-1). Whole tumor reconstruction using serial sections confirmed whirl-like cell clusters with nuclear ß-catenin accumulations at the tumor brain border. These whirls were surrounded by a belt of Claudin-1 expressing cells, showed an activated epidermal growth factor receptor (EGFR) and distinct CD133 as well as p21(WAF1/Cip1) positivity, indicating a tumor stem cell phenotype. Consistent with our previous in vitro studies, intracranial xenotransplants of adaCP confirmed cells with nuclear ß-catenin and activated EGFR being the driving force of tumor outgrowth. This model provides the possibility to study in vivo tumor cell migration and to test novel treatment regimens targeting this tumor stem cell niche.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Encéfalo/fisiopatologia , Craniofaringioma/fisiopatologia , Adolescente , Adulto , Animais , Encéfalo/patologia , Neoplasias Encefálicas/patologia , Claudina-1/metabolismo , Craniofaringioma/patologia , Progressão da Doença , Receptores ErbB/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Queratina-1 , Queratinas Específicas do Cabelo/metabolismo , Imageamento por Ressonância Magnética , Masculino , Camundongos Mutantes , Pessoa de Meia-Idade , Transplante de Neoplasias , Ubiquitina-Proteína Ligases/metabolismo , beta Catenina/metabolismo
5.
Neuro Oncol ; 16(2): 256-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24305709

RESUMO

BACKGROUND: Claudins are tight junction proteins expressed in epithelial tissues that play important roles in cell polarity and adhesion. Altered distribution of claudin-1(CLDN1) affects cell mobility and tumor invasiveness. Craniopharyngiomas (CPs) represent epithelial tumors of the sellar region, consisting of adamantinomatous (adaCP) and papillary (papCP) variants. Their tendency to infiltrate surrounding brain structures complicates successful surgery. Reliable markers are required to predict tumor behavior and to establish individualized treatment protocols. METHODS: We describe the distribution pattern of CLDN1 in a large cohort of 66 adaCPs, 21 papCPs, and 24 Rathke`s cleft cyst (RCC) cases using immunohistochemistry. CLDN1 mRNA levels were analyzed with qRT-PCR in 33 CP samples. The impact on the migration potential was studied in primary adaCP cell cultures (n = 11) treated with small interfering RNA (siRNA) for CLDN1. Furthermore, CLDN1 distribution patterns and expression levels were compared between invasive (n = 16) and noninvasive (n = 17) tumor groups. RESULTS: PapCPs and RCCs exhibited a distinct homogenous and membranous expression pattern, whereas CLDN1 immunoreactivity appeared weaker and more heterogeneous in adaCPs. In the latter cases, whirl-like cell clusters showed complete absence of CLDN1. mRNA analysis confirmed reduced CLDN1 levels in adaCPs versus papCPs. Interestingly, invasive tumors exhibited significantly lower CLDN1 expression compared with noninvasive counterparts regardless of CP subtype. Accordingly, siRNA treatment for CLDN1 altered tumor cell migration in vitro. CONCLUSION: CLDN1 represents a novel marker in the differential diagnosis of CP variants and RCCs. Low CLDN1 expression levels correlate with an invasive CP growth pattern and may serve as a prognostic marker.


Assuntos
Carcinoma Papilar/patologia , Claudina-1/metabolismo , Craniofaringioma/patologia , Neoplasias Hipofisárias/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Western Blotting , Carcinoma Papilar/genética , Carcinoma Papilar/metabolismo , Movimento Celular , Proliferação de Células , Criança , Pré-Escolar , Claudina-1/antagonistas & inibidores , Claudina-1/genética , Estudos de Coortes , Craniofaringioma/genética , Craniofaringioma/metabolismo , Feminino , Seguimentos , Humanos , Técnicas Imunoenzimáticas , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/metabolismo , Prognóstico , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , Adulto Jovem
6.
J Immunotoxicol ; 9(3): 301-13, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22800185

RESUMO

Chemotherapeutic agents (CT) and ionizing radiation (X-ray) induce DNA damage and primarily aim to stop the proliferation of tumor cells. However, multimodal anti-cancer therapies should finally result in tumor cell death and, best, in the induction of systemic anti-tumor immunity. Since distinct therapy-induced tumor cell death forms may create an immune activating tumor microenvironment, this study examined whether sole treatment with CT that are used in the therapy for colorectal cancer or in combination with X-ray result in colorectal tumor cell death with immunogenic potential. 5-Fluorouracil (5-FU), Oxaliplatin (Oxp), or Irinotecan (Irino) in combination with X-ray were all potent inhibitors of colorectal tumor cell colony formation. This study then examined the forms of cell death with AnnexinA5-FITC/Propidium Iodide staining. Necrosis was the prominent form of cell death induced by CT and/or X-ray. While only a combination of Irino with X-ray leads to death induction already 1 day after treatment, also the combinations of Oxp or 5-FU with X-ray and X-ray alone resulted in high necrosis rates at later time points after treatment. Inhibition of apoptosis increased the amount of necrotic tumor cells, suggesting that a programmed form of necrosis can be induced by CT + X-ray. 5-FU and Oxp alone or in combination with X-ray and Irino plus X-ray were most effective in increasing the expression of RIP, IRF-5, and p53, proteins involved in necrotic and apoptotic cell death pathways. All treatments further resulted in the release of the immune activating danger signals high-mobility group box 1 (HMGB1) and heat shock protein 70 (HSP70). The supernatants of the treated tumor cells induced maturation of dendritic cells. It is, therefore, concluded that combination of CT with X-ray is capable of inducing in vitro cell death forms of colorectal tumors with immunogenic potential.


Assuntos
Adenocarcinoma , Apoptose/imunologia , Quimiorradioterapia/métodos , Neoplasias Colorretais , Microambiente Tumoral/imunologia , Adenocarcinoma/imunologia , Adenocarcinoma/terapia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/terapia , Humanos , Proteínas de Neoplasias/imunologia , Raios X
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA