Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Int J Mol Sci ; 22(17)2021 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-34502484

RESUMEN

Malignant gliomas derive from brain glial cells and represent >75% of primary brain tumors. This includes anaplastic astrocytoma (grade III; AS), the most common and fatal glioblastoma multiforme (grade IV; GBM), and oligodendroglioma (ODG). We have generated patient-derived AS, GBM, and ODG cell models to study disease mechanisms and test patient-centered therapeutic strategies. We have used an aptamer-based high-throughput SOMAscan® 1.3K assay to determine the proteomic profiles of 1307 different analytes. SOMAscan® proteomes of AS and GBM self-organized into closely adjacent proteomes which were clearly distinct from ODG proteomes. GBM self-organized into four proteomic clusters of which SOMAscan® cluster 4 proteome predicted a highly inter-connected proteomic network. Several up- and down-regulated proteins relevant to glioma were successfully validated in GBM cell isolates across different SOMAscan® clusters and in corresponding GBM tissues. Slow off-rate modified aptamer proteomics is an attractive analytical tool for rapid proteomic stratification of different malignant gliomas and identified cluster-specific SOMAscan® signatures and functionalities in patient GBM cells.


Asunto(s)
Aptámeros de Nucleótidos/química , Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , Proteínas de Neoplasias/metabolismo , Proteoma/metabolismo , Proteómica , Neoplasias Encefálicas/patología , Glioma/patología , Humanos , Células Tumorales Cultivadas
2.
Nanomedicine ; 11(4): 913-25, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25680541

RESUMEN

Malignant glioma are often fatal and pose a significant therapeutic challenge. Here we have employed α-helical right handed coiled coils (RHCC) which self-assemble into tetrameric nanotubes that stably associate with platinum (Pt) (IV) compound. This Pt(IV)-RHCC complex showed superior in vitro and in vivo toxicity in human malignant glioma cells at up to 5 fold lower platinum concentrations when compared to free Pt(IV). Pt(IV)-RHCC nanotubes activated multiple cell death pathways in GB cells without affecting astrocytes in vitro or causing damage to normal mouse brain. This Pt(IV)-RHCC nanotubes may serve as a promising new therapeutic tool for low dose Pt(IV) prodrug application for highly efficient and selective treatment of human brain tumors. FROM THE CLINICAL EDITOR: The prognosis of malignant glioma remains poor despite medical advances. Platinum, one of the chemotherapeutic agents used, has significant systemic side effects. In this article, the authors employed α-helical right handed coiled coil (RHCC) protein nanotubes as a carrier for cisplatin. It was shown that the new compound achieved higher tumor kill rate but lower toxicity to normal cells and thus may hold promise to be a highly efficient treatment for the future.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Nanotubos/química , Compuestos de Platino/farmacología , Profármacos/farmacología , Animales , Antineoplásicos/química , Astrocitos/metabolismo , Astrocitos/patología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Ratones , Compuestos de Platino/química , Profármacos/química
3.
J Pathol ; 231(4): 466-79, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24014093

RESUMEN

We report a novel ligand-receptor system composed of the leucine-rich G-protein-coupled relaxin receptor, RXFP1, and the C1q-tumour necrosis factor-related protein 8 (CTRP8) in human primary brain cancer, a tumour entity devoid of the classical RXFP1 ligands, RLN1-3. In structural homology studies and computational docking experiments we delineated the N-terminal region of the globular C1q region of CTRP8 and the leucine-rich repeat units 7 and 8 of RXFP1 to mediate this new ligand-receptor interaction. CTRP8 secreted from HEK293T cells, recombinant human (rh) CTRP8, and short synthetic peptides derived from the C1q globular domain of human CTRP8 caused the activation of RXFP1 as determined by elevated intracellular cAMP levels and the induction of a marked pro-migratory phenotype in established glioblastoma (GB) cell lines and primary cells from GB patients. Employing a small competitor peptide, we were able to disrupt the CTRP8-RXFP1-induced increased GB motility. The CTRP8-RXFP1-mediated migration in GB cells involves the activation of PI3K and specific protein kinase C pathways and the increased production/secretion of the potent lysosomal protease cathepsin B (cathB), a known prognostic marker of GB. Specific inhibition of CTRP8-induced cathB activity effectively blocked the ability of primary GB to invade laminin matrices. Finally, co-immunoprecipitation studies revealed the direct interaction of human CTRP8 with RXFP1. Our results support a therapeutic approach in GB aimed at targeting multiple steps of the CTRP8-RXFP1 signalling pathway by a combined inhibitor and peptide-based strategy to block GB dissemination within the brain.


Asunto(s)
Adiponectina/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Adiponectina/farmacología , Sitios de Unión , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/patología , Catepsina B/metabolismo , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Activación Enzimática/fisiología , Glioblastoma/patología , Humanos , Invasividad Neoplásica/fisiopatología , Fosfatidilinositol 3-Quinasas/fisiología , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/fisiología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Recombinantes/farmacología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Células Tumorales Cultivadas
4.
Mol Oncol ; 12(9): 1464-1479, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29949238

RESUMEN

The C1q/TNF-related peptide 8 (CTRP8) has recently emerged as a novel ligand of the G protein-coupled receptor RXFP1 in the fatal brain tumor glioblastoma (GBM). We previously demonstrated that the CTRP8-RXFP1 ligand-receptor system promotes motility and matrix invasion of patient GBM and U87 MG cells by specific phosphorylation of PI3 kinase and protein kinase C. Here, we demonstrate a novel role for CTRP8 in protecting human GBM cells against the DNA alkylating damage of temozolomide (TMZ), the standard chemotherapy drug used to treat GBM. This DNA protective role of CTRP8 required a functional RXFP1-STAT3 signaling cascade in GBM cells. We identified N-methylpurine DNA glycosylase (MPG), a monofunctional glycosylase that initiates base excision repair pathway by generating an apurinic/apyrimidinic (AP) site, as a new CTRP8-RXFP1-STAT3 target in GBM. Upon TMZ exposure, treatment with CTRP8 reduced the formation of AP sites and double-strand DNA breaks in GBM cells. This CTRP8 effect was independent of cellular MGMT levels and was associated with decreased caspase 3/7 activity and increased survival of human GBM. CTRP8-induced RXFP1 activation caused an increase in cellular protein levels of the anti-apoptotic Bcl members and STAT3 targets Bcl-2 and Bcl-XL in human GBM. Collectively, our results demonstrate a novel multipronged and clinically relevant mechanism by which the CTRP8-RXFP1 ligand-receptor system exerts a DNA protective function against TMZ chemotherapeutic stress in GBM. This CTRP8-RXFP1-STAT3 axis is a novel determinant of TMZ responsiveness/chemoresistance and an emerging new drug target for improved treatment of human GBM.


Asunto(s)
Adiponectina/metabolismo , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Resistencia a Antineoplásicos , Glioblastoma/tratamiento farmacológico , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Factor de Transcripción STAT3/metabolismo , Temozolomida/uso terapéutico , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular Tumoral , Roturas del ADN de Doble Cadena/efectos de los fármacos , ADN Glicosilasas/genética , ADN Glicosilasas/metabolismo , Reparación del ADN , Humanos , Terapia Molecular Dirigida , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína bcl-X/metabolismo
7.
Mol Oncol ; 11(8): 1078-1098, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28500786

RESUMEN

The multikinase inhibitor and FDA-approved drug dovitinib (Dov) crosses the blood-brain barrier and was recently used as single drug application in clinical trials for GB patients with recurrent disease. The Dov-mediated molecular mechanisms in GB cells are unknown. We used GB patient cells and cell lines to show that Dov downregulated the stem cell protein Lin28 and its target high-mobility group protein A2 (HMGA2). The Dov-induced reduction in pSTAT3Tyr705 phosphorylation demonstrated that Dov negatively affects the STAT3/LIN28/Let-7/HMGA2 regulatory axis in GB cells. Consistent with the known function of LIN28 and HMGA2 in GB self-renewal, Dov reduced GB tumor sphere formation. Dov treatment also caused the downregulation of key base excision repair factors and O6 -methylguanine-DNA-methyltransferase (MGMT), which are known to have important roles in the repair of temozolomide (TMZ)-induced alkylating DNA damage. Combined Dov/TMZ treatment enhanced TMZ-induced DNA damage as quantified by nuclear γH2AX foci and comet assays, and increased GB cell apoptosis. Pretreatment of GB cells with Dov ('Dov priming') prior to TMZ treatment reduced GB cell viability independent of p53 status. Sequential treatment involving 'Dov priming' and alternating treatment cycles with TMZ and Dov substantially reduced long-term GB cell survival in MGMT+ patient GB cells. Our results may have immediate clinical implications to improve TMZ response in patients with LIN28+ /HMGA2+ GB, independent of their MGMT methylation status.


Asunto(s)
Bencimidazoles/farmacología , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Quinolonas/farmacología , Bencimidazoles/agonistas , Línea Celular Tumoral , Dacarbazina/agonistas , Dacarbazina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioblastoma/metabolismo , Glioblastoma/patología , Humanos , Proteínas de Neoplasias/biosíntesis , Quinolonas/agonistas , Temozolomida
8.
Br J Pharmacol ; 174(10): 1025-1033, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27443788

RESUMEN

We established the role of the GPCR relaxin/insulin-like family peptide receptor 1 (RXFP1 receptor) as a novel active receptor in human glioblastoma (GB), a fatal brain tumour. We identified C1q/TNF-related protein 8 (CTRP8) as a novel agonist of the RXFP1 receptor. CTRP8 enhanced the motility and matrix invasion of GB, and this involved PKC-mediated up-regulation of cathepsin B, a marker for poor prognosis in GB patients. We conclude that the absence of relaxin isoforms does not preclude the activation of the RXFP1 receptor, as the least known member of the CTRP family, CTRP8, can effectively target and activate RXFP1 receptors. LINKED ARTICLES: This article is part of a themed section on Recent Progress in the Understanding of Relaxin Family Peptides and their Receptors. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.10/issuetoc.


Asunto(s)
Adiponectina/metabolismo , Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Relaxina/metabolismo , Transducción de Señal , Adiponectina/química , Humanos , Neoplasias/patología , Conformación Proteica , Receptores Acoplados a Proteínas G/agonistas , Receptores de Péptidos/agonistas
10.
Oncol Rep ; 29(1): 149-54, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23128569

RESUMEN

We report for the first time the distribution of human INSL5 and its cognate leucine rich G-protein coupled receptor RXFP4 in the large intestine and in neuroendocrine/carcinoid tissues. Immunoreactive INSL5 was uniquely expressed by enteroendocrine cells (EECs) located within the colonic mucosa, whereas colonocytes were immunopositive for RXFP4. INSL5+ and RXFP4+ cells were also detected in human neuroendocrine/carcinoid tissues. We employed a recently described Insl5 knockout mouse model and 2 mouse models of induced colitis to address the relevance of Insl5 in EEC development and in acute inflammation of the colon. We identified INSL5 as a specific marker for synaptophysin+ EECs in the mucosa of the normal human and mouse colon. Insl5 was not essential for the development of mouse synaptophysin+ EECs. The mouse models of chemically induced colitis (dextran sulfate sodium and dinitrobenzene-sulfonic acid) failed to show changes in the numbers of Insl5+ EECs at inflammatory sites during the acute phase of colitis. In conclusion, we showed that INSL5 is a novel marker of colorectal EECs and provide first evidence for the presence of a potentially autocrine/paracrine INSL5-RXFP4 signaling system in the normal human and mouse colon and in rare human neuroendocrine tumours.


Asunto(s)
Biomarcadores de Tumor/análisis , Tumor Carcinoide/patología , Células Enteroendocrinas/patología , Insulina/fisiología , Intestino Grueso/patología , Tumores Neuroendocrinos/patología , Proteínas/fisiología , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Péptidos/metabolismo , Animales , Comunicación Autocrina , Tumor Carcinoide/metabolismo , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Células Enteroendocrinas/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Inflamación/metabolismo , Inflamación/patología , Intestino Grueso/metabolismo , Masculino , Ratones , Ratones Noqueados , Estadificación de Neoplasias , Tumores Neuroendocrinos/metabolismo , Comunicación Paracrina , Pronóstico , Células Tumorales Cultivadas
11.
Eur J Pharmacol ; 625(1-3): 23-30, 2009 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-19836372

RESUMEN

Glioblastoma multiform (GBM) is the most common malignant brain tumour in adults. Despite decades of experimentation to improve the outcome of patients with GBM this highly aggressive tumour remains fatal. Primary GBM are often characterized by the over-expression of epidermal growth factor (EGF) receptor/HER1 and/or its mutational variants, with ligand-independent, constitutively active EGF receptor vIII variant most frequently observed in GBM. EGF receptor signalling can promote tumorigenesis by increasing cell proliferation, tissue invasion, neoangiogenesis, tumour cell chemoresistance, and by inhibiting apoptosis of cancer cells. EGF receptor was the first receptor to serve as target for cancer therapy of many solid tumours. After 2 decades of intensive targeting of EGF receptor for molecular therapy, several anti-EGF receptor inhibitors are now available in the clinic. Therapeutic strategies to target EGF receptor and EGF receptor mutant forms in GBM include humanized monoclonal antibodies, tyrosine kinase inhibitors, and RNAi compounds. However, despite the fact that most EGF receptor-directed glioma therapies to date have focused on single therapeutic agents, a multi-directional approach involving targeted inhibition of multiple signalling pathways has emerged as a more robust therapeutical approach. Furthermore, the emergence of the hypothesis of "brain cancer stem cells" in the bulb of GBM identifies this population of cells with self-renewal capacity as novel obligatory targets for efficient cure of GBM. Here we summarize current findings on the clinical role of these EGF receptor inhibitory therapeutic agents in the treatment of GBM.


Asunto(s)
Antineoplásicos/farmacología , Receptores ErbB/antagonistas & inhibidores , Glioblastoma/tratamiento farmacológico , Adulto , Animales , Sistemas de Liberación de Medicamentos , Glioblastoma/fisiopatología , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología
13.
Mov Disord ; 18(10): 1169-75, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14534922

RESUMEN

Deep brain stimulation (DBS) has virtually replaced thalamotomy for the treatment of essential tremor. It is thought that the site for DBS is the same as the optimal lesion site; however, this match has not been investigated previously. We sought to determine whether the location of thalamic DBS matched the site at which thalamotomy would be performed. Eleven patients who had detailed microelectrode recording and stimulation for placement of DBS electrodes and subsequent successful tremor control were analysed. An experienced surgeon, blinded to outcome and final electrode position, selected the ideal thalamotomy site based on the reconstructed maps obtained intraoperatively. When the site of long-term clinically used DBS and theoretical thalamotomy location was calculated in three-dimensional space and compared for each of the x, y, and z axes in stereotactic space, there was no significant difference in the mediolateral location of DBS and theoretical lesion site. There was also no difference between the theoretical lesion site and the placement of the tip of the electrode; however, the active electrodes used for chronic stimulation were significantly more anterior (P = 0.005) and dorsal (P = 0.034) to the ideal thalamotomy target. This mismatch may reflect the compromise required between adverse and beneficial effects with chronic stimulation, but it also suggests different mechanisms of effect of DBS and thalamotomy.


Asunto(s)
Estimulación Eléctrica/métodos , Temblor Esencial/terapia , Psicocirugía/métodos , Tálamo/efectos de la radiación , Tálamo/cirugía , Adulto , Anciano , Anciano de 80 o más Años , Mapeo Encefálico , Estudios de Casos y Controles , Electrodos Implantados , Estudios de Evaluación como Asunto , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Examen Neurológico , Desempeño Psicomotor , Técnicas Estereotáxicas , Tálamo/anatomía & histología , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda