Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Curr Opin Cell Biol ; 85: 102279, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37984008

RESUMO

Glioblastoma is the most common and aggressive primary brain tumor, characterized by a highly complex and heterogeneous tumor immune microenvironment (TIME). In this review, we discuss the impact of tumor-intrinsic and tumor-extrinsic drivers that contribute to heterogeneity in the adult glioblastoma TIME, focusing on four main factors: genetic drivers, sex, age, and standard of care therapy. We describe recent insights into how each of these factors affects key aspects ranging from TIME composition to therapy response, with an emphasis on the cross-talk between tumor and immune cells. Deciphering these local interactions is fundamental to understanding therapy resistance and identifying novel immunomodulatory strategies.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Adulto , Humanos , Glioblastoma/genética , Glioblastoma/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Microambiente Tumoral
2.
Cancer Res ; 83(7): 1031-1047, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-36753744

RESUMO

The survival rate for patients with head and neck cancer (HNC) diagnosed with cervical lymph node (cLN) or distant metastasis is low. Genomic alterations in the HRAS oncogene are associated with advanced tumor stage and metastasis in HNC. Elucidation of the molecular mechanisms by which mutated HRAS (HRASmut) facilitates HNC metastasis could lead to improved treatment options for patients. Here, we examined metastasis driven by mutant HRAS in vitro and in vivo using HRASmut human HNC cell lines, patient-derived xenografts, and a novel HRASmut syngeneic model. Genetic and pharmacological manipulations indicated that HRASmut was sufficient to drive invasion in vitro and metastasis in vivo. Targeted proteomic analysis showed that HRASmut promoted AXL expression via suppressing the Hippo pathway and stabilizing YAP1 activity. Pharmacological blockade of HRAS signaling with the farnesyltransferase inhibitor tipifarnib activated the Hippo pathway and reduced the nuclear export of YAP1, thus suppressing YAP1-mediated AXL expression and metastasis. AXL was required for HRASmut cells to migrate and invade in vitro and to form regional cLN and lung metastases in vivo. In addition, AXL-depleted HRASmut tumors displayed reduced lymphatic and vascular angiogenesis in the primary tumor. Tipifarnib treatment also regulated AXL expression and attenuated VEGFA and VEGFC expression, thus regulating tumor-induced vascular formation and metastasis. Our results indicate that YAP1 and AXL are crucial factors for HRASmut-induced metastasis and that tipifarnib treatment can limit the metastasis of HNC tumors with HRAS mutations by enhancing YAP1 cytoplasmic sequestration and downregulating AXL expression. SIGNIFICANCE: Mutant HRAS drives metastasis of head and neck cancer by switching off the Hippo pathway to activate the YAP1-AXL axis and to stimulate lymphovascular angiogenesis.


Assuntos
Neoplasias de Cabeça e Pescoço , Proteômica , Humanos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Linhagem Celular Tumoral , Transdução de Sinais , Neoplasias de Cabeça e Pescoço/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo
3.
J Clin Invest ; 133(1)2023 01 03.
Artigo em Inglês | MEDLINE | ID: mdl-36594465

RESUMO

Glioblastoma (GBM) is the most belligerent and frequent brain tumor in adults. Research over the past two decades has provided increased knowledge of the genomic and molecular landscape of GBM and highlighted the presence of a high degree of inter- and intratumor heterogeneity within the neoplastic compartment. It is now appreciated that GBMs are composed of multiple distinct and impressionable neoplastic and non-neoplastic cell types that form the unique brain tumor microenvironment (TME). Non-neoplastic cells in the TME form reciprocal interactions with neoplastic cells to promote tumor growth and invasion, and together they influence the tumor response to standard-of-care therapies as well as emerging immunotherapies. One of the most prevalent non-neoplastic cell types in the GBM TME are myeloid cells, the most abundant of which are of hematopoietic origin, including monocytes/monocyte-derived macrophages. Less abundant, although still a notable presence, are neutrophils of hematopoietic origin and intrinsic brain-resident microglia. In this Review we focus on neutrophils and monocytes that infiltrate tumors from the blood circulation, their heterogeneity, and their interactions with neoplastic cells and other non-neoplastic cells in the TME. We conclude with an overview of challenges in targeting these cells and discuss avenues for therapeutic exploitation to improve the dismal outcomes of patients with GBM.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Humanos , Monócitos/patologia , Glioblastoma/genética , Glioblastoma/terapia , Glioblastoma/metabolismo , Neutrófilos/patologia , Macrófagos/metabolismo , Microglia/patologia , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/metabolismo , Microambiente Tumoral/genética
4.
J Nanotheranostics ; 3(4): 177-188, 2022 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-36324626

RESUMO

Background: Glioblastoma is the most lethal primary brain malignancy in adults. Standard of care treatment, consisting of temozolomide (TMZ) and adjuvant radiotherapy (RT), mostly does not prevent local recurrence. The inability of drugs to enter the brain, in particular antibody-based drugs and radiosensitizers, is a crucial limitation to effective glioblastoma therapy. Methods: Here, we developed a combined strategy using radiosensitizer gold nanoparticles coated with insulin to cross the blood-brain barrier and shuttle tumor-targeting antibodies (cetuximab) into the brain. Results: Following intravenous injection to an orthotopic glioblastoma mouse model, the nanoparticles specifically accumulated within the tumor. Combining targeted nanoparticle injection with TMZ and RT standard of care significantly inhibited tumor growth and extended survival, as compared to standard of care alone. Histological analysis of tumors showed that the combined treatment eradicated tumor cells, and decreased tumor vascularization, proliferation, and repair. Conclusions: Our findings demonstrate radiosensitizer nanoparticles that effectively deliver antibodies into the brain, target the tumor, and effectively improve standard of care treatment outcome in glioblastoma.

5.
Cancers (Basel) ; 14(20)2022 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-36291914

RESUMO

Brain tumors comprise over 100 types of masses, differing in the following: location; patient age; molecular, histological, and immunohistochemical characteristics; and prognosis and treatment. Glioma tumors originate from neuroglia, cells supporting the brain. Palladin, a structural protein widely expressed in mammalian tissues, has a pivotal role in cytoskeletal dynamics and motility in health and disease. Palladin is linked to the progression of breast, pancreatic, and renal cancers. In the central nervous system, palladin is involved in embryonic development, neuronal maturation, the cell cycle, differentiation, and apoptosis. However, the role of palladin in brain tumors is unknown. In this work, we explored palladin's role in glioma. We analyzed clinical data, along with bulk and single-cell gene expression. We then validated our results using IHC staining of tumor samples, together with qRT-PCR of glioma cell lines. We determined that wild-type palladin-4 is overexpressed in adult gliomas and is correlated with a decrease in survival. Palladin expression outperformed clinically used prognostic markers and was most prominent in glioblastoma. Finally, we showed that palladin originates from the malignant cell population. Our findings indicate that palladin expression might be linked to adult glioma progression and is associated with prognosis.

6.
Elife ; 112022 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-35642785

RESUMO

Cancer stem cells (CSCs) alone can initiate and maintain tumors, but the function of non-cancer stem cells (non-CSCs) that form the tumor bulk remains poorly understood. Proteomic analysis showed a higher abundance of the extracellular matrix small leucine-rich proteoglycan fibromodulin (FMOD) in the conditioned medium of differentiated glioma cells (DGCs), the equivalent of glioma non-CSCs, compared to that of glioma stem-like cells (GSCs). DGCs silenced for FMOD fail to cooperate with co-implanted GSCs to promote tumor growth. FMOD downregulation neither affects GSC growth and differentiation nor DGC growth and reprogramming in vitro. DGC-secreted FMOD promotes angiogenesis by activating integrin-dependent Notch signaling in endothelial cells. Furthermore, conditional silencing of FMOD in newly generated DGCs in vivo inhibits the growth of GSC-initiated tumors due to poorly developed vasculature and increases mouse survival. Collectively, these findings demonstrate that DGC-secreted FMOD promotes glioma tumor angiogenesis and growth through paracrine signaling in endothelial cells and identifies a DGC-produced protein as a potential therapeutic target in glioma.


Assuntos
Células Endoteliais , Glioma , Animais , Células Endoteliais/metabolismo , Fibromodulina/metabolismo , Glioma/patologia , Integrinas/metabolismo , Camundongos , Células-Tronco Neoplásicas/metabolismo , Neovascularização Patológica/metabolismo , Proteômica
7.
STAR Protoc ; 3(1): 101106, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35098162

RESUMO

The immune fraction of the tumor microenvironment has been proven to play a fundamental role in glioblastoma progression and therapeutic response. Here, we present a detailed magnetic-bead-enrichment-based protocol to isolate and analyze the composition of this fraction from mouse brain tumors. The protocol is optimized to achieve high yields of viable immune cells. We also detail characterization of the immune subtypes by FACS analysis. Our procedure is applicable for either lentiviral-induced tumors or transplant models in syngeneic immunocompetent mice. For complete details on the use and execution of this protocol, please refer to Magod et al. (2021).


Assuntos
Neoplasias Encefálicas/imunologia , Microambiente Tumoral , Animais , Neoplasias Encefálicas/patologia , Modelos Animais de Doenças , Citometria de Fluxo , Camundongos
8.
Cell Rep ; 36(5): 109480, 2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34348160

RESUMO

Recent multi-omics studies show different immune tumor microenvironment (TME) compositions in glioblastoma (GBM). However, temporal comprehensive knowledge of the TME from initiation of the disease remains sparse. We use Cre recombinase (Cre)-inducible lentiviral murine GBM models to compare the cellular evolution of the immune TME in tumors initiated from different oncogenic drivers. We show that neutrophils infiltrate early during tumor progression primarily in the mesenchymal GBM model. Depleting neutrophils in vivo at the onset of disease accelerates tumor growth and reduces the median overall survival time of mice. We show that, as a tumor progresses, bone marrow-derived neutrophils are skewed toward a phenotype associated with pro-tumorigenic processes. Our findings suggest that GBM can remotely regulate systemic myeloid differentiation in the bone marrow to generate neutrophils pre-committed to a tumor-supportive phenotype. This work reveals plasticity in the systemic immune host microenvironment, suggesting an additional point of intervention in GBM treatment.


Assuntos
Medula Óssea/patologia , Neoplasias Encefálicas/patologia , Carcinogênese/patologia , Glioma/patologia , Neutrófilos/patologia , Microambiente Tumoral , Animais , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Modelos Animais de Doenças , Progressão da Doença , Feminino , Glioma/irrigação sanguínea , Glioma/tratamento farmacológico , Humanos , Terapia de Imunossupressão , Integrases/metabolismo , Camundongos Endogâmicos C57BL , Mutação/genética , Estadiamento de Neoplasias , Neovascularização Patológica/patologia , Análise de Sobrevida
9.
Nat Commun ; 12(1): 3615, 2021 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-34127674

RESUMO

Glioblastoma is considered one of the most aggressive malignancies in adult and pediatric patients. Despite decades of research no curative treatment is available and it thus remains associated with a very dismal prognosis. Although recent pre-clinical and clinical studies have demonstrated the feasibility of chimeric antigen receptors (CAR) T cell immunotherapeutic approach in glioblastoma, tumor heterogeneity and antigen loss remain among one of the most important challenges to be addressed. In this study, we identify p32/gC1qR/HABP/C1qBP to be specifically expressed on the surface of glioma cells, making it a suitable tumor associated antigen for redirected CAR T cell therapy. We generate p32 CAR T cells and find them to recognize and specifically eliminate p32 expressing glioma cells and tumor derived endothelial cells in vitro and to control tumor growth in orthotopic syngeneic and xenograft mouse models. Thus, p32 CAR T cells may serve as a therapeutic option for glioblastoma patients.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Antineoplásicos/farmacologia , Glioma/imunologia , Glioma/terapia , Linfócitos T/imunologia , Idoso , Animais , Antígenos de Neoplasias/imunologia , Neoplasias Encefálicas , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Feminino , Glioblastoma/genética , Glioblastoma/patologia , Glioma/genética , Glioma/metabolismo , Humanos , Imunoterapia Adotiva , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Proteínas Mitocondriais/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Receptores de Antígenos Quiméricos/imunologia , Serina Endopeptidases/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Nat Commun ; 12(1): 1912, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33771989

RESUMO

Glioblastoma (GB) is a highly invasive type of brain cancer exhibiting poor prognosis. As such, its microenvironment plays a crucial role in its progression. Among the brain stromal cells, the microglia were shown to facilitate GB invasion and immunosuppression. However, the reciprocal mechanisms by which GB cells alter microglia/macrophages behavior are not fully understood. We propose that these mechanisms involve adhesion molecules such as the Selectins family. These proteins are involved in immune modulation and cancer immunity. We show that P-selectin mediates microglia-enhanced GB proliferation and invasion by altering microglia/macrophages activation state. We demonstrate these findings by pharmacological and molecular inhibition of P-selectin which leads to reduced tumor growth and increased survival in GB mouse models. Our work sheds light on tumor-associated microglia/macrophage function and the mechanisms by which GB cells suppress the immune system and invade the brain, paving the way to exploit P-selectin as a target for GB therapy.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , Macrófagos/metabolismo , Microglia/metabolismo , Selectina-P/genética , Animais , Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos SCID , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/genética , Neoplasias Experimentais/metabolismo , Selectina-P/antagonistas & inibidores , Selectina-P/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética
11.
Sci Adv ; 6(47)2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33208369

RESUMO

Harnessing CRISPR-Cas9 technology for cancer therapeutics has been hampered by low editing efficiency in tumors and potential toxicity of existing delivery systems. Here, we describe a safe and efficient lipid nanoparticle (LNP) for the delivery of Cas9 mRNA and sgRNAs that use a novel amino-ionizable lipid. A single intracerebral injection of CRISPR-LNPs against PLK1 (sgPLK1-cLNPs) into aggressive orthotopic glioblastoma enabled up to ~70% gene editing in vivo, which caused tumor cell apoptosis, inhibited tumor growth by 50%, and improved survival by 30%. To reach disseminated tumors, cLNPs were also engineered for antibody-targeted delivery. Intraperitoneal injections of EGFR-targeted sgPLK1-cLNPs caused their selective uptake into disseminated ovarian tumors, enabled up to ~80% gene editing in vivo, inhibited tumor growth, and increased survival by 80%. The ability to disrupt gene expression in vivo in tumors opens new avenues for cancer treatment and research and potential applications for targeted gene editing of noncancerous tissues.


Assuntos
Nanopartículas , Neoplasias , Sistemas CRISPR-Cas , Edição de Genes , Técnicas de Transferência de Genes , Lipossomos , Neoplasias/genética , Neoplasias/terapia
12.
Oncogene ; 39(46): 6990-7004, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33077835

RESUMO

Interconversion of transformed non-stem cells to cancer stem cells, termed cancer cell plasticity, contributes to intra-tumor heterogeneity and its molecular mechanisms are currently unknown. Here, we have identified Tenascin C (TNC) to be upregulated and secreted in mesenchymal glioblastoma (MES GBM) subtype with high NF-κB signaling activity. Silencing TNC decreases proliferation, migration and suppresses self-renewal of glioma stem cells. Loss of TNC in MES GBM compromises de-differentiation of transformed astrocytes and blocks the ability of glioma stem cells to differentiate into tumor derived endothelial cells (TDEC). Inhibition of NF-κB activity or TNC knockdown in tumor cells decreased their tumorigenic potential in vivo. Our results uncover a link between NF-κB activation in MES GBM and high levels of TNC in GBM extracellular matrix. We suggest that TNC plays an important role in the autocrine regulation of glioma cell plasticity and hence can be a potential molecular target for MES GBM.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Células-Tronco Mesenquimais/patologia , Células-Tronco Neoplásicas/patologia , Tenascina/metabolismo , Animais , Astrócitos/patologia , Desdiferenciação Celular , Plasticidade Celular , Transformação Celular Neoplásica/patologia , Matriz Extracelular/patologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Tenascina/genética , Microambiente Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Int J Cancer ; 146(1): 115-122, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31211411

RESUMO

Epigenetic transformations may provide early indicators for cancer and other disease. Specifically, the amount of genomic 5-hydroxymethylcytosine (5-hmC) was shown to be globally reduced in a wide range of cancers. The integration of this global biomarker into diagnostic workflows is hampered by the limitations of current 5-hmC quantification methods. Here we present and validate a fluorescence-based platform for high-throughput and cost-effective quantification of global genomic 5-hmC levels. We utilized the assay to characterize cancerous tissues based on their 5-hmC content, and observed a pronounced reduction in 5-hmC level in various cancer types. We present data for glioblastoma, colorectal cancer, multiple myeloma, chronic lymphocytic leukemia and pancreatic cancer, compared to corresponding controls. Potentially, the technique could also be used to follow response to treatment for personalized treatment selection. We present initial proof-of-concept data for treatment of familial adenomatous polyposis.


Assuntos
5-Metilcitosina/análogos & derivados , Biomarcadores Tumorais/metabolismo , Epigênese Genética , Ensaios de Triagem em Larga Escala/métodos , Neoplasias/genética , 5-Metilcitosina/metabolismo , Animais , Análise Custo-Benefício , Fluorescência , Ensaios de Triagem em Larga Escala/economia , Humanos , Camundongos , Neoplasias/classificação , Estudo de Prova de Conceito
14.
J Control Release ; 308: 109-118, 2019 08 28.
Artigo em Inglês | MEDLINE | ID: mdl-31255690

RESUMO

Tumor-selective drug conjugates can potentially improve the prognosis for patients affected by glioblastoma (GBM) - the most common and malignant type of brain cancer with no effective cure. Here we evaluated a novel tumor penetrating peptide that targets cell surface p32, LinTT1 (AKRGARSTA), as a GBM targeting ligand for systemically-administered nanoparticles. LinTT1-functionalization increased tumor homing of iron oxide nanoworms (NWs) across a panel of five GBM models ranging from infiltratively-disseminating to angiogenic phenotypes. LinTT1-NWs homed to CD31-positive tumor blood vessels, including to transdifferentiated endothelial cells, and showed co-localization with tumor macrophages and lymphatic vessels. LinTT1 functionalization also resulted in increased GBM delivery of other types of systemically-administered nanoparticles: silver nanoparticles and albumin-paclitaxel nanoparticles. Finally, LinTT1-guided proapoptotic NWs exerted strong anti-glioma activity in two models of GBM, including doubling the lifespan of the mice in an aggressive orthotopic stem cell-like GBM that recapitulates the histological hallmarks of human GBM. Our study suggests that LinTT1 targeting strategy can be used to increase GBM uptake of systemic nanoparticles for improved imaging and therapy.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Glioblastoma/tratamento farmacológico , Nanopartículas , Peptídeos/administração & dosagem , Albuminas/administração & dosagem , Albuminas/farmacocinética , Animais , Linhagem Celular Tumoral , Células Endoteliais/metabolismo , Feminino , Compostos Férricos/química , Glioblastoma/patologia , Humanos , Masculino , Nanopartículas Metálicas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Paclitaxel/administração & dosagem , Paclitaxel/farmacocinética , Peptídeos/química , Prata/química , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Elife ; 62017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28079521

RESUMO

Protein kinase A (PKA) plays critical roles in neuronal function that are mediated by different regulatory (R) subunits. Deficiency in either the RIß or the RIIß subunit results in distinct neuronal phenotypes. Although RIß contributes to synaptic plasticity, it is the least studied isoform. Using isoform-specific antibodies, we generated high-resolution large-scale immunohistochemical mosaic images of mouse brain that provided global views of several brain regions, including the hippocampus and cerebellum. The isoforms concentrate in discrete brain regions, and we were able to zoom-in to show distinct patterns of subcellular localization. RIß is enriched in dendrites and co-localizes with MAP2, whereas RIIß is concentrated in axons. Using correlated light and electron microscopy, we confirmed the mitochondrial and nuclear localization of RIß in cultured neurons. To show the functional significance of nuclear localization, we demonstrated that downregulation of RIß, but not of RIIß, decreased CREB phosphorylation. Our study reveals how PKA isoform specificity is defined by precise localization.


Assuntos
Química Encefálica , Proteínas Quinases Dependentes de AMP Cíclico/análise , Isoformas de Proteínas/análise , Animais , Axônios/química , Dendritos/química , Imuno-Histoquímica , Camundongos
16.
Oncotarget ; 7(7): 7403-14, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26859681

RESUMO

Malignant peripheral nerve sheath tumors (MPNSTs) are highly aggressive sarcomas that arise sporadically or in association with the Neurofibromatosis type 1 (NF1) cancer predisposition syndrome. In individuals with NF1, MPNSTs are hypothesized to arise from Nf1-deficient Schwann cell precursor cells following the somatic acquisition of secondary cooperating genetic mutations (e.g., p53 loss). To model this sequential genetic cooperativity, we coupled somatic lentivirus-mediated p53 knockdown in the adult right sciatic nerve with embryonic Schwann cell precursor Nf1 gene inactivation in two different Nf1 conditional knockout mouse strains. Using this approach, ~60% of mice with Periostin-Cre-mediated Nf1 gene inactivation (Periostin-Cre; Nf1(flox/flox) mice) developed tumors classified as low-grade MPNSTs following p53 knockdown (mean, 6 months). Similarly, ~70% of Nf1+/- mice with GFAP-Cre-mediated Nf1 gene inactivation (GFAP-Cre; Nf1(flox/null) mice) developed low-grade MPNSTs following p53 knockdown (mean, 3 months). In addition, wild-type and Nf1+/- mice with GFAP-Cre-mediated Nf1 loss develop MPNSTs following somatic p53 knockout with different latencies, suggesting potential influences of Nf1+/- stromal cells in MPNST pathogenesis. Collectively, this new MPNST model system permits the analysis of somatically-acquired events as well as tumor microenvironment signals that potentially cooperate with Nf1 loss in the development and progression of this deadly malignancy.


Assuntos
Células-Tronco Embrionárias/patologia , Neurilemoma/patologia , Neurofibromina 1/fisiologia , Células de Schwann/patologia , Nervo Isquiático/patologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Animais Recém-Nascidos , Western Blotting , Células Cultivadas , Células-Tronco Embrionárias/metabolismo , Técnicas Imunoenzimáticas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurilemoma/genética , Células de Schwann/metabolismo , Nervo Isquiático/metabolismo
17.
Sci Adv ; 2(1): e1501292, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26824076

RESUMO

Glioblastoma multiforme (GBM) is the most common and lethal form of intracranial tumor. We have established a lentivirus-induced mouse model of malignant gliomas, which faithfully captures the pathophysiology and molecular signature of mesenchymal human GBM. RNA-Seq analysis of these tumors revealed high nuclear factor κB (NF-κB) activation showing enrichment of known NF-κB target genes. Inhibition of NF-κB by either depletion of IκB kinase 2 (IKK2), expression of a IκBαM super repressor, or using a NEMO (NF-κB essential modifier)-binding domain (NBD) peptide in tumor-derived cell lines attenuated tumor proliferation and prolonged mouse survival. Timp1, one of the NF-κB target genes significantly up-regulated in GBM, was identified to play a role in tumor proliferation and growth. Inhibition of NF-κB activity or silencing of Timp1 resulted in slower tumor growth in both mouse and human GBM models. Our results suggest that inhibition of NF-κB activity or targeting of inducible NF-κB genes is an attractive therapeutic approach for GBM.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , NF-kappa B/genética , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Quinase I-kappa B/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Peptídeos/genética , Transdução de Sinais/genética , Regulação para Cima/genética
18.
Crit Rev Oncog ; 19(5): 327-36, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25404148

RESUMO

Glioblastoma (GBM) is the most common and malignant type of primary brain tumor. It represents one of the deadliest human cancers, with an average survival at diagnosis of about 1 year. This poor prognosis is due to therapeutic resistance and tumor recurrence after surgical removal. One of the most important hallmarks of GBM is tumor heterogeneity. Intertumor heterogeneity is mostly characterized by distinct genetic alterations that occur in individual tumors originating in the same organ and allows the classification of these tumors into different molecular subtypes. Intratumor heterogeneity-the diversity within individual tumors-has become the focus of research interest in the past few years, and tumor cell plasticity as a new source of cancer stem cells has added another level of complexity to this phenomenon. This review describes the molecular heterogeneity of GBMs at the transcriptome level and the expression profile-based classification of histopathologically indistinguishable tumors into different subtypes. In addition, the role of dedifferentiation of tumor cells into a stem cell-like state is discussed as a source of cellular heterogeneity within tumors, highlighting tumor cell plasticity as an important driver of GBM heterogeneity. Understanding tumor heterogeneity will help design better therapies against GBM and avoid tumor recurrence.


Assuntos
Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Animais , Humanos , Camundongos , Células-Tronco Neoplásicas/patologia
19.
EMBO Rep ; 15(3): 244-53, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24531722

RESUMO

Regenerative medicine aims to replace the lost or damaged cells in the human body through a new source of healthy transplanted cells or by endogenous repair. Although human embryonic stem cells were first thought to be the ideal source for cell therapy and tissue repair in humans, the discovery by Yamanaka and colleagues revolutionized the field. Almost any differentiated cell can be sent back in time to a pluripotency state by expressing the appropriate transcription factors. The process of somatic reprogramming using Yamanaka factors, many of which are oncogenes, offers a glimpse into how cancer stem cells may originate. In this review we discuss the similarities between tumor dedifferentiation and somatic cell reprogramming and how this may pose a risk to the application of this new technology in regenerative medicine.


Assuntos
Desdiferenciação Celular , Reprogramação Celular , Células-Tronco Neoplásicas/citologia , Animais , Carcinógenos/metabolismo , Linhagem da Célula , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/fisiologia
20.
Mol Ther ; 21(12): 2195-204, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23959073

RESUMO

Antiangiogenic therapy is a promising new treatment modality for cancer, but it generally produces only transient tumor regression. We have previously devised a tumor-targeted nanosystem, in which a pentapeptide, CGKRK, delivers a proapoptotic peptide into the mitochondria of tumor blood vessel endothelial cells and tumor cells. The treatment was highly effective in glioblastoma mouse models completely refractory to other antiangiogenic treatments. Here, we identify p32/gC1qR/HABP, a mitochondrial protein that is also expressed at the cell surface of activated (angiogenic) endothelial cells and tumor cells, as a receptor for the CGKRK peptide. The results demonstrate the ability of p32 to cause internalization of a payload bound to p32 into the cytoplasm. We also show that nardilysin, a protease capable of cleaving CGKRK, plays a role in the internalization of a p32-bound payload. As p32 is overexpressed and surface displayed in breast cancers, we studied the efficacy of the nanosystem in this cancer. We show highly significant treatment results in an orthotopic model of breast cancer. The specificity of cell surface p32 for tumor-associated cells, its ability to carry payloads to mitochondria, and the efficacy of the system in important types of cancer make the nanosystem a promising candidate for further development.


Assuntos
Inibidores da Angiogênese/farmacologia , Neoplasias da Mama/terapia , Glicoproteínas de Membrana/metabolismo , Proteínas Mitocondriais/metabolismo , Nanopartículas/química , Peptídeos/farmacologia , Receptores de Complemento/metabolismo , Inibidores da Angiogênese/uso terapêutico , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Neoplasias Mamárias Experimentais , Glicoproteínas de Membrana/genética , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/genética , Terapia de Alvo Molecular , Especificidade de Órgãos , Peptídeos/administração & dosagem , Peptídeos/uso terapêutico , Receptores de Complemento/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA