Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38947019

RESUMEN

Background . Interactions among tumor, immune, and vascular niches play major roles in driving glioblastoma (GBM) malignancy and treatment responses. The composition, heterogeneity, and localization of extracellular core matrix proteins (CMPs) that mediate such interactions, however, are not well understood. Methods . Here, through computational genomics and proteomics approaches, we analyzed the functional and clinical relevance of CMP expression in GBM at bulk, single cell, and spatial anatomical resolution. Results . We identified genes encoding CMPs whose expression levels categorize GBM tumors into CMP expression-high (M-H) and CMP expression-low (M-L) groups. CMP enrichment is associated with worse patient survival, specific driver oncogenic alterations, mesenchymal state, infiltration of pro-tumor immune cells, and immune checkpoint gene expression. Anatomical and single-cell transcriptome analyses indicate that matrisome gene expression is enriched in vascular and leading edge/infiltrative niches that are known to harbor glioma stem cells driving GBM progression. Finally, we identified a 17-gene CMP expression signature, termed Matrisome 17 (M17) signature that further refines the prognostic value of CMP genes. The M17 signature is a significantly stronger prognostic factor compared to MGMT promoter methylation status as well as canonical subtypes, and importantly, potentially predicts responses to PD1 blockade. Conclusion . The matrisome gene expression signature provides a robust stratification of GBM patients by survival and potential biomarkers of functionally relevant GBM niches that can mediate mesenchymal-immune cross talk. Patient stratification based on matrisome profiles can contribute to selection and optimization of treatment strategies.

2.
Res Sq ; 2023 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-37790408

RESUMEN

Interactions among tumor, immune and vascular niches play major roles in driving glioblastoma (GBM) malignancy and treatment responses. The composition, heterogeneity, and localization of extracellular core matrix proteins (CMPs) that mediate such interactions, however, are not well understood. Here, we characterize functional and clinical relevance of genes encoding CMPs in GBM at bulk, single cell, and spatial anatomical resolution. We identify a "matrix code" for genes encoding CMPs whose expression levels categorize GBM tumors into matrisome-high and matrisome-low groups that correlate with worse and better patient survival, respectively. The matrisome enrichment is associated with specific driver oncogenic alterations, mesenchymal state, infiltration of pro-tumor immune cells and immune checkpoint gene expression. Anatomical and single cell transcriptome analyses indicate that matrisome gene expression is enriched in vascular and leading edge/infiltrative anatomic structures that are known to harbor glioma stem cells driving GBM progression. Finally, we identified a 17-gene matrisome signature that retains and further refines the prognostic value of genes encoding CMPs and, importantly, potentially predicts responses to PD1 blockade in clinical trials for GBM. The matrisome gene expression profiles provide potential biomarkers of functionally relevant GBM niches that contribute to mesenchymal-immune cross talk and patient stratification which could be applied to optimize treatment responses.

3.
bioRxiv ; 2023 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-37333072

RESUMEN

Interactions among tumor, immune and vascular niches play major roles in driving glioblastoma (GBM) malignancy and treatment responses. The composition, heterogeneity, and localization of extracellular core matrix proteins (CMPs) that mediate such interactions, however, are not well understood. Here, we characterize functional and clinical relevance of genes encoding CMPs in GBM at bulk, single cell, and spatial anatomical resolution. We identify a "matrix code" for genes encoding CMPs whose expression levels categorize GBM tumors into matrisome-high and matrisome-low groups that correlate with worse and better survival, respectively, of patients. The matrisome enrichment is associated with specific driver oncogenic alterations, mesenchymal state, infiltration of pro-tumor immune cells and immune checkpoint gene expression. Anatomical and single cell transcriptome analyses indicate that matrisome gene expression is enriched in vascular and leading edge/infiltrative anatomic structures that are known to harbor glioma stem cells driving GBM progression. Finally, we identified a 17-gene matrisome signature that retains and further refines the prognostic value of genes encoding CMPs and, importantly, potentially predicts responses to PD1 blockade in clinical trials for GBM. The matrisome gene expression profiles may provide biomarkers of functionally relevant GBM niches that contribute to mesenchymal-immune cross talk and patient stratification to optimize treatment responses.

4.
Biomedicines ; 11(3)2023 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-36979714

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) is the most aggressive molecular subtype of breast cancer, and current treatments are only partially effective in disease control. More effective combination approaches are needed to improve the survival of TNBC patients. Eribulin mesylate, a non-taxane microtubule dynamics inhibitor, is approved by the U.S. Food and Drug Administration to treat metastatic breast cancer after at least two previous chemotherapeutic regimens. However, eribulin as a single agent has limited therapeutic efficacy against TNBC. METHODS: High-throughput kinome library RNAi screening, Ingenuity Pathway Analysis, and STRING analysis were performed to identify target kinases for combination with eribulin. The identified combinations were validated using in vivo and ex vivo proliferation assays. RESULTS: We identified 135 potential kinase targets whose inhibition enhanced the antiproliferation effect of eribulin in TNBC cells, with the PI3K/Akt/mTOR and the MAPK/JNK pathways emerging as the top candidates. Indeed, copanlisib (pan-class I PI3K inhibitor), everolimus (mTOR inhibitor), trametinib (MEK inhibitor), and JNK-IN-8 (pan-JNK inhibitor) produced strong synergistic antiproliferative effects when combined with eribulin, and the PI3K and mTOR inhibitors had the most potent effects in vitro. CONCLUSIONS: Our data suggest a new strategy of combining eribulin with PI3K or mTOR inhibitors to treat TNBC.

5.
Nat Commun ; 13(1): 767, 2022 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-35140215

RESUMEN

A major rate-limiting step in developing more effective immunotherapies for GBM is our inadequate understanding of the cellular complexity and the molecular heterogeneity of immune infiltrates in gliomas. Here, we report an integrated analysis of 201,986 human glioma, immune, and other stromal cells at the single cell level. In doing so, we discover extensive spatial and molecular heterogeneity in immune infiltrates. We identify molecular signatures for nine distinct myeloid cell subtypes, of which five are independent prognostic indicators of glioma patient survival. Furthermore, we identify S100A4 as a regulator of immune suppressive T and myeloid cells in GBM and demonstrate that deleting S100a4 in non-cancer cells is sufficient to reprogram the immune landscape and significantly improve survival. This study provides insights into spatial, molecular, and functional heterogeneity of glioma and glioma-associated immune cells and demonstrates the utility of this dataset for discovering therapeutic targets for this poorly immunogenic cancer.


Asunto(s)
Inmunoterapia , Proteína de Unión al Calcio S100A4/aislamiento & purificación , Análisis de la Célula Individual/métodos , Animales , Neoplasias Encefálicas/inmunología , Femenino , Glioma/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Células Mieloides , Pronóstico , Proteína de Unión al Calcio S100A4/genética , Microambiente Tumoral/inmunología
6.
Cancer Res Commun ; 2(6): 402-416, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-36688010

RESUMEN

The emergence of treatment resistance significantly reduces the clinical utility of many effective targeted therapies. Although both genetic and epigenetic mechanisms of drug resistance have been reported, whether these mechanisms are stochastically selected in individual tumors or governed by a predictable underlying principle is unknown. Here, we report that the dependence of cancer stem cells (CSCs), not bulk tumor cells, on the targeted pathway determines the molecular mechanism of resistance in individual tumors. Using both spontaneous and transplantable mouse models of sonic hedgehog (SHH) medulloblastoma (MB) treated with an SHH/Smoothened inhibitor, sonidegib/LDE225, we show that genetic-based resistance occurs only in tumors that contain SHH-dependent CSCs (SD-CSCs). In contrast, SHH MBs containing SHH-dependent bulk tumor cells but SHH-independent CSCs (SI-CSCs) acquire resistance through epigenetic reprogramming. Mechanistically, elevated proteasome activity in SMOi-resistant SI-CSC MBs alters the tumor cell maturation trajectory through enhanced degradation of specific epigenetic regulators, including histone acetylation machinery components, resulting in global reductions in H3K9Ac, H3K14Ac, H3K56Ac, H4K5Ac, and H4K8Ac marks and gene expression changes. These results provide new insights into how selective pressure on distinct tumor cell populations contributes to different mechanisms of resistance to targeted therapies. This insight provides a new conceptual framework to understand responses and resistance to SMOis and other targeted therapies.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Animales , Ratones , Transducción de Señal , Proteínas Hedgehog/genética , Meduloblastoma/genética , Neoplasias Cerebelosas/tratamiento farmacológico , Células Madre Neoplásicas/metabolismo
8.
Neurotherapeutics ; 18(3): 1710-1728, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34160773

RESUMEN

CAG repeat expansion is the genetic cause of nine incurable polyglutamine (polyQ) diseases with neurodegenerative features. Silencing repeat RNA holds great therapeutic value. Here, we developed a repeat-based RNA-cleaving DNAzyme that catalyzes the destruction of expanded CAG repeat RNA of six polyQ diseases with high potency. DNAzyme preferentially cleaved the expanded allele in spinocerebellar ataxia type 1 (SCA1) cells. While cleavage was non-allele-specific for spinocerebellar ataxia type 3 (SCA3) cells, treatment of DNAzyme leads to improved cell viability without affecting mitochondrial metabolism or p62-dependent aggresome formation. DNAzyme appears to be stable in mouse brain for at least 1 month, and an intermediate dosage of DNAzyme in a SCA3 mouse model leads to a significant reduction of high molecular weight ATXN3 proteins. Our data suggest that DNAzyme is an effective RNA silencing molecule for potential treatment of multiple polyQ diseases.


Asunto(s)
ADN Catalítico/administración & dosificación , ADN Catalítico/genética , Enfermedad de Machado-Joseph/genética , Péptidos/genética , ARN/genética , Expansión de Repetición de Trinucleótido/genética , Animales , Ataxina-3/genética , Línea Celular Tumoral , Silenciador del Gen/fisiología , Células HEK293 , Humanos , Enfermedad de Machado-Joseph/terapia , Ratones , Péptidos/metabolismo , Ataxias Espinocerebelosas/genética , Ataxias Espinocerebelosas/terapia , Técnicas Estereotáxicas
9.
Front Immunol ; 11: 1191, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32625208

RESUMEN

The application of tumor immunotherapy to glioblastoma (GBM) is limited by an unprecedented degree of immune suppression due to factors that include high numbers of immune suppressive myeloid cells, the blood brain barrier, and T cell sequestration to the bone marrow. We previously identified an increase in immune suppressive myeloid-derived suppressor cells (MDSCs) in GBM patients, which correlated with poor prognosis and was dependent on macrophage migration inhibitory factor (MIF). Here we examine the MIF signaling axis in detail in murine MDSC models, GBM-educated MDSCs and human GBM. We found that the monocytic subset of MDSCs (M-MDSCs) expressed high levels of the MIF cognate receptor CD74 and was localized in the tumor microenvironment. In contrast, granulocytic MDSCs (G-MDSCs) expressed high levels of the MIF non-cognate receptor CXCR2 and showed minimal accumulation in the tumor microenvironment. Furthermore, targeting M-MDSCs with Ibudilast, a brain penetrant MIF-CD74 interaction inhibitor, reduced MDSC function and enhanced CD8 T cell activity in the tumor microenvironment. These findings demonstrate the MDSC subsets differentially express MIF receptors and may be leveraged for specific MDSC targeting.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Células Supresoras de Origen Mieloide/inmunología , Receptores Inmunológicos/inmunología , Escape del Tumor/inmunología , Animales , Humanos , Inmunoterapia/métodos , Factores Inhibidores de la Migración de Macrófagos/inmunología , Factores Inhibidores de la Migración de Macrófagos/metabolismo , Ratones , Piridinas/farmacología , Receptores Inmunológicos/metabolismo , Escape del Tumor/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
10.
Nat Commun ; 11(1): 1508, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32198351

RESUMEN

Tumour cell phagocytosis by antigen presenting cells (APCs) is critical to the generation of antitumour immunity. However, cancer cells can evade phagocytosis by upregulating anti-phagocytosis molecule CD47. Here, we show that CD47 blockade alone is inefficient in stimulating glioma cell phagocytosis. However, combining CD47 blockade with temozolomide results in a significant pro-phagocytosis effect due to the latter's ability to induce endoplasmic reticulum stress response. Increased tumour cell phagocytosis subsequently enhances antigen cross-presentation and activation of cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) in APCs, resulting in more efficient T cell priming. This bridging of innate and adaptive responses inhibits glioma growth, but also activates immune checkpoint. Sequential administration of an anti-PD1 antibody overcomes this potential adaptive resistance. Together, these findings reveal a dynamic relationship between innate and adaptive immune regulation in tumours and support further investigation of phagocytosis modulation as a strategy to enhance cancer immunotherapy responses.


Asunto(s)
Inmunidad Adaptativa , Glioblastoma/inmunología , Glioma/inmunología , Inmunidad Innata , Fagocitosis/inmunología , Animales , Presentación de Antígeno , Apoptosis , Antígeno CD47/efectos de los fármacos , Antígeno CD47/metabolismo , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Glioblastoma/patología , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Monitorización Inmunológica , Nucleotidiltransferasas/metabolismo , Linfocitos T/inmunología , Temozolomida/farmacología
11.
Nat Biomed Eng ; 4(1): 69-83, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31844155

RESUMEN

Exosomes are attractive as nucleic-acid carriers because of their favourable pharmacokinetic and immunological properties and their ability to penetrate physiological barriers that are impermeable to synthetic drug-delivery vehicles. However, inserting exogenous nucleic acids, especially large messenger RNAs, into cell-secreted exosomes leads to low yields. Here we report a cellular-nanoporation method for the production of large quantities of exosomes containing therapeutic mRNAs and targeting peptides. We transfected various source cells with plasmid DNAs and stimulated the cells with a focal and transient electrical stimulus that promotes the release of exosomes carrying transcribed mRNAs and targeting peptides. Compared with bulk electroporation and other exosome-production strategies, cellular nanoporation produced up to 50-fold more exosomes and a more than 103-fold increase in exosomal mRNA transcripts, even from cells with low basal levels of exosome secretion. In orthotopic phosphatase and tensin homologue (PTEN)-deficient glioma mouse models, mRNA-containing exosomes restored tumour-suppressor function, enhanced inhibition of tumour growth and increased survival. Cellular nanoporation may enable the use of exosomes as a universal nucleic-acid carrier for applications requiring transcriptional manipulation.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Electroporación/métodos , Exosomas/metabolismo , Glioma/tratamiento farmacológico , ARN Mensajero/uso terapéutico , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Ratones Endogámicos C57BL , MicroARNs/metabolismo , MicroARNs/uso terapéutico , Nanotecnología , ARN Mensajero/metabolismo , Transducción de Señal
12.
Mol Cell ; 70(5): 825-841.e6, 2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29861161

RESUMEN

Super-enhancers are large clusters of enhancers that activate gene expression. Broad trimethyl histone H3 lysine 4 (H3K4me3) often defines active tumor suppressor genes. However, how these epigenomic signatures are regulated for tumor suppression is little understood. Here we show that brain-specific knockout of the H3K4 methyltransferase MLL4 (a COMPASS-like enzyme, also known as KMT2D) in mice spontaneously induces medulloblastoma. Mll4 loss upregulates oncogenic Ras and Notch pathways while downregulating neuronal gene expression programs. MLL4 enhances DNMT3A-catalyzed DNA methylation and SIRT1/BCL6-mediated H4K16 deacetylation, which antagonize expression of Ras activators and Notch pathway components, respectively. Notably, Mll4 loss downregulates tumor suppressor genes (e.g., Dnmt3a and Bcl6) by diminishing broad H3K4me3 and super-enhancers and also causes widespread impairment of these epigenomic signatures during medulloblastoma genesis. These findings suggest an anti-tumor role for super-enhancers and provide a unique tumor-suppressive mechanism in which MLL4 is necessary to maintain broad H3K4me3 and super-enhancers at tumor suppressor genes.


Asunto(s)
Neoplasias Cerebelosas/genética , Metilación de ADN , Genes Supresores de Tumor , N-Metiltransferasa de Histona-Lisina/genética , Meduloblastoma/genética , Oncogenes , Procesamiento Proteico-Postraduccional , Acetilación , Animales , Proliferación Celular , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Regulación Neoplásica de la Expresión Génica , Genes ras , N-Metiltransferasa de Histona-Lisina/deficiencia , Lisina , Meduloblastoma/metabolismo , Meduloblastoma/patología , Ratones Noqueados , Proteínas Proto-Oncogénicas c-bcl-6/genética , Proteínas Proto-Oncogénicas c-bcl-6/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal , Sirtuina 1/genética , Sirtuina 1/metabolismo
13.
Cancer Res ; 77(19): 5360-5373, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28807938

RESUMEN

Glioma stem cells (GSC) and epithelial-mesenchymal transition (EMT) are strongly associated with therapy resistance and tumor recurrence, but the underlying mechanisms are incompletely understood. Here, we show that S100A4 is a novel biomarker of GSCs. S100A4+ cells in gliomas are enriched with cancer cells that have tumor-initiating and sphere-forming abilities, with the majority located in perivascular niches where GSCs are found. Selective ablation of S100A4-expressing cells was sufficient to block tumor growth in vitro and in vivo We also identified S100A4 as a critical regulator of GSC self-renewal in mouse and patient-derived glioma tumorspheres. In contrast with previous reports of S100A4 as a reporter of EMT, we discovered that S100A4 is an upstream regulator of the master EMT regulators SNAIL2 and ZEB along with other mesenchymal transition regulators in glioblastoma. Overall, our results establish S100A4 as a central node in a molecular network that controls stemness and EMT in glioblastoma, suggesting S100A4 as a candidate therapeutic target. Cancer Res; 77(19); 5360-73. ©2017 AACR.


Asunto(s)
Biomarcadores/metabolismo , Neoplasias Encefálicas/patología , Transición Epitelial-Mesenquimal , Glioblastoma/patología , Células Madre Neoplásicas/patología , Proteína de Unión al Calcio S100A4/metabolismo , Animales , Apoptosis , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Ratones , Células Madre Neoplásicas/metabolismo , Proteína de Unión al Calcio S100A4/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Nat Nanotechnol ; 12(8): 763-769, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28459470

RESUMEN

Tumour-targeted immunotherapy offers the unique advantage of specific tumouricidal effects with reduced immune-associated toxicity. However, existing platforms suffer from low potency, inability to generate long-term immune memory and decreased activities against tumour-cell subpopulations with low targeting receptor levels. Here we adopted a modular design approach that uses colloidal nanoparticles as substrates to create a multivalent bi-specific nanobioconjugate engager (mBiNE) to promote selective, immune-mediated eradication of cancer cells. By simultaneously targeting the human epidermal growth factor receptor 2 (HER2) expressed by cancer cells and pro-phagocytosis signalling mediated by calreticulin, the mBiNE stimulated HER2-targeted phagocytosis and produced durable antitumour immune responses against HER2-expressing tumours. Interestingly, although the initial immune activation mediated by the mBiNE was receptor dependent, the subsequent antitumour immunity also generated protective effects against tumour-cell populations that lacked the HER2 receptor. Thus, the mBiNE represents a new targeted, nanomaterial-immunotherapy platform to stimulate innate and adaptive immunity and promote a universal antitumour response.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Inmunoterapia/métodos , Nanoconjugados/química , Neoplasias/terapia , Receptor ErbB-2/inmunología , Inmunidad Adaptativa , Animales , Coloides , Humanos , Inmunidad Innata , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/patología , Células THP-1
15.
J Neurooncol ; 133(1): 47-57, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28432589

RESUMEN

Despite multimodal treatment that includes surgery, radiation and chemotherapy, virtually all glioblastomas (GBM) recur, indicating that these interventions are insufficient to eradicate all malignant cells. To identify potential new therapeutic targets in GBMs, we examined the expression and function of proteins that are associated with therapy resistance and cancer cell survival. We measured the expression of eight such proteins in 50 GBM samples by immunohistochemistry and analyzed patient survival. We report that GBM patients with high expression of ABCG2 (also called BCRP) or XIAP at the protein level had worse survival than those with low expression. The adjusted hazard ratio for ABCG2 was 2.35 and for XIAP was 2.65. Since glioma stem cells (GSCs) have been shown to be more resistant than bulk tumor cells to anti-cancer therapies and to express high levels of these proteins, we also sought to determine if ABCG2 and XIAP have functional roles in GSCs. We used small molecule inhibitors to treat patient-derived GBM tumorspheres in vitro and observed that inhibitors of ABCG2, Ko143 and fumitremorgin, significantly reduced self-renewal. These results suggest that ABCG2 and XIAP proteins may be useful indicators of patient survival and that inhibition of ABCG2 may be a promising therapeutic strategy in GBMs.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Proteínas de Neoplasias/metabolismo , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/radioterapia , Células Cultivadas , Dacarbazina/análogos & derivados , Dacarbazina/uso terapéutico , Dicetopiperazinas/farmacología , Femenino , Estudios de Seguimiento , Glioblastoma/tratamiento farmacológico , Glioblastoma/mortalidad , Glioblastoma/radioterapia , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Indoles/farmacología , Masculino , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Persona de Mediana Edad , Proteínas de Neoplasias/antagonistas & inhibidores , Trasplante de Neoplasias , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Temozolomida
16.
Exp Mol Med ; 49(4): e317, 2017 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-28408749

RESUMEN

Gliosarcoma (GS) is a rare variant (2%) of glioblastoma (GBM) that poses clinical genomic challenges because of its poor prognosis and limited genomic information. To gain a comprehensive view of the genomic alterations in GS and to understand the molecular etiology of GS, we applied whole-exome sequencing analyses for 28 GS cases (6 blood-matched fresh-frozen tissues for the discovery set, 22 formalin-fixed paraffin-embedded tissues for the validation set) and copy-number variation microarrays for 5 blood-matched fresh-frozen tissues. TP53 mutations were more prevalent in the GS cases (20/28, 70%) compared to the GBM cases (29/90, 32%), and the GS patients with TP53 mutations showed a significantly shorter survival (multivariate Cox analysis, hazard ratio=23.9, 95% confidence interval, 2.87-199.63, P=0.003). A pathway analysis showed recurrent alterations in MAPK signaling (EGFR, RASGRF2 and TP53), phosphatidylinositol/calcium signaling (CACNA1s, PLCs and ITPRs) and focal adhesion/tight junction (PTEN and PAK3) pathways. Genomic profiling of the matched recurrent GS cases detected the occurrence of TP53 mutations in two recurrent GS cases, which suggests that TP53 mutations play a role in treatment resistance. Functionally, we found that TP53 mutations are associated with the epithelial-mesenchymal transition (EMT) process of sarcomatous components of GS. We provide the first comprehensive genome-wide genetic alternation profiling of GS, which suggests novel prognostic subgroups in GS patients based on their TP53 mutation status and provides new insight in the pathogenesis and targeted treatment of GS.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Transición Epitelial-Mesenquimal/genética , Gliosarcoma/genética , Mutación , Proteína p53 Supresora de Tumor/genética , Neoplasias Encefálicas/patología , Señalización del Calcio , Línea Celular Tumoral , Femenino , Gliosarcoma/patología , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Persona de Mediana Edad , Uniones Estrechas/genética , Uniones Estrechas/metabolismo
18.
Oncotarget ; 7(44): 72322-72342, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27608848

RESUMEN

In the last several years, our appreciation of intra-tumoral heterogeneity has greatly increased due to accumulating evidence for the co-existence of genetically and epigenetically divergent cancer cells residing in different microenvironments within a tumor. Herein, we review recent literature discussing intra-tumoral heterogeneity in the context of therapy resistance mechanisms at the genetic, epigenetic and microenvironmental levels. We illustrate the influence of tumor microenvironment on therapy resistance and epigenetic states of cancer cells by highlighting the role of cancer stem cells in therapy resistance. We also summarize different strategies that have been employed to address various resistance mechanisms at genetic, epigenetic, and microenvironmental levels in preclinical and clinical studies. We propose that future personalized cancer therapy design needs to incorporate dynamic and comprehensive analyses of tumor heterogeneity landscape and multi-dimensional mechanisms of therapy resistance.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Heterogeneidad Genética , Terapia Molecular Dirigida/tendencias , Neoplasias/genética , Microambiente Tumoral/genética , Antineoplásicos/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Epigénesis Genética , Humanos , Terapia Molecular Dirigida/métodos , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Células Madre Neoplásicas/patología , Transducción de Señal/genética
19.
Nat Commun ; 6: 6548, 2015 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-25813983

RESUMEN

Basal-like breast cancer (BLBC) is a heterogeneous disease with poor prognosis; however, its cellular origins and aetiology are poorly understood. In this study, we show that inhibitor of differentiation 4 (ID4) is a key regulator of mammary stem cell self-renewal and marks a subset of BLBC with a putative mammary basal cell of origin. Using an ID4GFP knock-in reporter mouse and single-cell transcriptomics, we show that ID4 marks a stem cell-enriched subset of the mammary basal cell population. ID4 maintains the mammary stem cell pool by suppressing key factors required for luminal differentiation. Furthermore, ID4 is specifically expressed by a subset of human BLBC that possess a very poor prognosis and a transcriptional signature similar to a mammary stem cell. These studies identify ID4 as a mammary stem cell regulator, deconvolute the heterogeneity of BLBC and link a subset of mammary stem cells to the aetiology of BLBC.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Inhibidoras de la Diferenciación/genética , Glándulas Mamarias Animales/citología , ARN Mensajero/metabolismo , Células Madre/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Técnicas de Sustitución del Gen , Humanos , Proteínas Inhibidoras de la Diferenciación/metabolismo , Glándulas Mamarias Animales/metabolismo , Ratones , Trasplante de Neoplasias , Fenotipo , Reacción en Cadena en Tiempo Real de la Polimerasa
20.
Front Oncol ; 4: 323, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25452936

RESUMEN

Prom1/CD133 has been identified in colorectal, hepatocellular, and pancreatic cancer as a cancer stem cell marker and has been used as such to predict colon cancer recurrence in humans. Its potential molecular function as well as its role as a marker of intestinal regeneration is still not fully known. We evaluated the role of Prom1 in intestinal regeneration in inflammatory bowel disease (IBD), determined the function of Prom1, and characterized the effect of a lack of Prom1 on intestinal tumor formation in animal models. Our results suggest that Apc mutations lead to an increase in Prom1 expressing cells in the intestinal crypt stem cell compartment and in early intestinal adenomas. Also, Prom1 knockout mice are more susceptible to intestinal tumor formation. We conclude that Prom1 likely plays a role in regulating intestinal homeostasis and that these results clearly illustrate the role of Prom1 in intestinal regeneration. We further conclude that Prom1 may provide a novel therapeutic target for patients with gastrointestinal conditions such as IBD, short bowel syndrome, and colorectal cancer.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...