Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 4758, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38902234

RESUMO

To uncover molecular changes underlying blood-brain-barrier dysfunction in Alzheimer's disease, we performed single nucleus RNA sequencing in 24 Alzheimer's disease and control brains and focused on vascular and astrocyte clusters as main cell types of blood-brain-barrier gliovascular-unit. The majority of the vascular transcriptional changes were in pericytes. Of the vascular molecular targets predicted to interact with astrocytic ligands, SMAD3, upregulated in Alzheimer's disease pericytes, has the highest number of ligands including VEGFA, downregulated in Alzheimer's disease astrocytes. We validated these findings with external datasets comprising 4,730 pericyte and 150,664 astrocyte nuclei. Blood SMAD3 levels are associated with Alzheimer's disease-related neuroimaging outcomes. We determined inverse relationships between pericytic SMAD3 and astrocytic VEGFA in human iPSC and zebrafish models. Here, we detect vast transcriptome changes in Alzheimer's disease at the gliovascular-unit, prioritize perturbed pericytic SMAD3-astrocytic VEGFA interactions, and validate these in cross-species models to provide a molecular mechanism of blood-brain-barrier disintegrity in Alzheimer's disease.


Assuntos
Doença de Alzheimer , Astrócitos , Barreira Hematoencefálica , Pericitos , Proteína Smad3 , Fator A de Crescimento do Endotélio Vascular , Peixe-Zebra , Doença de Alzheimer/genética , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Humanos , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Proteína Smad3/metabolismo , Proteína Smad3/genética , Astrócitos/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Pericitos/metabolismo , Pericitos/patologia , Masculino , Células-Tronco Pluripotentes Induzidas/metabolismo , Feminino , Idoso , Transcriptoma , Encéfalo/metabolismo , Encéfalo/patologia , Encéfalo/irrigação sanguínea , Idoso de 80 Anos ou mais , Modelos Animais de Doenças
2.
JCI Insight ; 8(13)2023 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-37427593

RESUMO

Glioblastomas (GBM) are aggressive tumors that lack effective treatments. Here, we show that the Rho family guanine nucleotide exchange factor Syx promotes GBM cell growth both in vitro and in orthotopic xenografts derived from patients with GBM. Growth defects upon Syx depletion are attributed to prolonged mitosis, increased DNA damage, G2/M cell cycle arrest, and cell apoptosis, mediated by altered mRNA and protein expression of various cell cycle regulators. These effects are phenocopied by depletion of the Rho downstream effector Dia1 and are due, at least in part, to increased phosphorylation, cytoplasmic retention, and reduced activity of the YAP/TAZ transcriptional coactivators. Furthermore, targeting Syx signaling cooperates with radiation treatment and temozolomide (TMZ) to decrease viability in GBM cells, irrespective of their inherent response to TMZ. The data indicate that a Syx-RhoA-Dia1-YAP/TAZ signaling axis regulates cell cycle progression, DNA damage, and therapy resistance in GBM and argue for its targeting for cancer treatment.


Assuntos
Glioblastoma , Humanos , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Linhagem Celular Tumoral , Transdução de Sinais , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Dano ao DNA , Divisão Celular
3.
Autophagy ; 19(6): 1711-1732, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36469690

RESUMO

The ubiquitin (Ub) kinase-ligase pair PINK1-PRKN mediates the degradation of damaged mitochondria by macroautophagy/autophagy (mitophagy). PINK1 surveils mitochondria and upon stress accumulates on the mitochondrial surface where it phosphorylates serine 65 of Ub to activate PRKN and to drive mitochondrial turnover. While loss of either PINK1 or PRKN is genetically linked to Parkinson disease (PD) and activating the pathway seems to have great therapeutic potential, there is no formal proof that stimulation of mitophagy is always beneficial. Here we used biochemical and cell biological methods to study single nucleotide variants in the activation loop of PINK1 to modulate the enzymatic function of this kinase. Structural modeling and in vitro kinase assays were used to investigate the molecular mechanism of the PINK1 variants. In contrast to the PD-linked PINK1G411S mutation that diminishes Ub kinase activity, we found that the PINK1G411A variant significantly boosted Ub phosphorylation beyond levels of PINK1 wild type. This resulted in augmented PRKN activation, mitophagy rates and increased viability after mitochondrial stress in midbrain-derived, gene-edited neurons. Mechanistically, the G411A variant stabilizes the kinase fold of PINK1 and transforms Ub to adopt the preferred, C-terminally retracted conformation for improved substrate turnover. In summary, we identify a critical role of residue 411 for substrate receptivity that may now be exploited for drug discovery to increase the enzymatic function of PINK1. The genetic substitution of Gly411 to Ala increases mitophagy and may be useful to confirm neuroprotection in vivo and might serve as a critical positive control during therapeutic development.Abbreviations: ATP: adenosine triphosphate; CCCP: carbonyl cyanide m-chlorophenyl hydrazone; Ub-CR: ubiquitin with C-terminally retracted tail; CTD: C-terminal domain (of PINK1); ELISA: enzyme-linked immunosorbent assay; HCI: high-content imaging; IB: immunoblot; IF: immunofluorescence; NPC: neuronal precursor cells; MDS: molecular dynamics simulation; PD: Parkinson disease; p-S65-Ub: ubiquitin phosphorylated at Ser65; RMSF: root mean scare fluctuation; TOMM: translocase of outer mitochondrial membrane; TVLN: ubiquitin with T66V and L67N mutation, mimics Ub-CR; Ub: ubiquitin; WT: wild-type.


Assuntos
Doença de Parkinson , Proteínas Quinases , Humanos , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Doença de Parkinson/genética , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/metabolismo , Autofagia , Ubiquitina/metabolismo
4.
J Nanobiotechnology ; 18(1): 162, 2020 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-33160390

RESUMO

BACKGROUND: Cancer cell-derived extracellular vesicles (EVs) have previously been shown to contribute to pre-metastatic niche formation. Specifically, aggressive tumors secrete pro-metastatic EVs that travel in the circulation to distant organs to modulate the microenvironment for future metastatic spread. Previous studies have focused on the interface between pro-metastatic EVs and epithelial/endothelial cells in the pre-metastatic niche. However, EV interactions with circulating components such as low-density lipoprotein (LDL) have been overlooked. RESULTS: This study demonstrates that EVs derived from brain metastases cells (Br-EVs) and corresponding regular cancer cells (Reg-EVs) display different interactions with LDL. Specifically, Br-EVs trigger LDL aggregation, and the presence of LDL accelerates Br-EV uptake by monocytes, which are key components in the brain metastatic niche. CONCLUSIONS: Collectively, these data are the first to demonstrate that pro-metastatic EVs display distinct interactions with LDL, which impacts monocyte internalization of EVs.


Assuntos
Neoplasias Encefálicas/metabolismo , Vesículas Extracelulares/metabolismo , Lipoproteínas LDL/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias da Mama , Linhagem Celular Tumoral , Células Endoteliais , Humanos , Macrófagos , Monócitos , Células THP-1 , Microambiente Tumoral
5.
Blood Adv ; 4(10): 2143-2157, 2020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32421811

RESUMO

Patients with chronic lymphocytic leukemia (CLL) are characterized by monoclonal expansion of CD5+CD23+CD27+CD19+κ/λ+ B lymphocytes and are clinically noted to have profound immune suppression. In these patients, it has been recently shown that a subset of B cells possesses regulatory functions and secretes high levels of interleukin 10 (IL-10). Our investigation identified that CLL cells with a CD19+CD24+CD38hi immunophenotype (B regulatory cell [Breg]-like CLL cells) produce high amounts of IL-10 and transforming growth factor ß (TGF-ß) and are capable of transforming naive T helper cells into CD4+CD25+FoxP3+ T regulatory cells (Tregs) in an IL-10/TGF-ß-dependent manner. A strong correlation between the percentage of CD38+ CLL cells and Tregs was observed. CD38hi Tregs comprised more than 50% of Tregs in peripheral blood mononuclear cells (PBMCs) in patients with CLL. Anti-CD38 targeting agents resulted in lethality of both Breg-like CLL and Treg cells via apoptosis. Ex vivo, use of anti-CD38 monoclonal antibody (mAb) therapy was associated with a reduction in IL-10 and CLL patient-derived Tregs, but an increase in interferon-γ and proliferation of cytotoxic CD8+ T cells with an activated phenotype, which showed an improved ability to lyse patient-autologous CLL cells. Finally, effects of anti-CD38 mAb therapy were validated in a CLL-patient-derived xenograft model in vivo, which showed decreased percentage of Bregs, Tregs, and PD1+CD38hiCD8+ T cells, but increased Th17 and CD8+ T cells (vs vehicle). Altogether, our results demonstrate that targeting CD38 in CLL can modulate the tumor microenvironment; skewing T-cell populations from an immunosuppressive to immune-reactive milieu, thus promoting immune reconstitution for enhanced anti-CLL response.


Assuntos
Linfócitos B Reguladores , Leucemia Linfocítica Crônica de Células B , Linfócitos T CD8-Positivos , Humanos , Imunossupressores , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Linfócitos T Reguladores , Microambiente Tumoral
6.
Small ; 16(10): e1904064, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32067382

RESUMO

Extracellular vesicles secreted from adipose-derived mesenchymal stem cells (ADSCs) have therapeutic effects in inflammatory diseases. However, production of extracellular vesicles (EVs) from ADSCs is costly, inefficient, and time consuming. The anti-inflammatory properties of adipose tissue-derived EVs and other biogenic nanoparticles have not been explored. In this study, biogenic nanoparticles are obtained directly from lipoaspirate, an easily accessible and abundant source of biological material. Compared to ADSC-EVs, lipoaspirate nanoparticles (Lipo-NPs) take less time to process (hours compared to months) and cost less to produce (clinical-grade cell culture facilities are not required). The physicochemical characteristics and anti-inflammatory properties of Lipo-NPs are evaluated and compared to those of patient-matched ADSC-EVs. Moreover, guanabenz loading in Lipo-NPs is evaluated for enhanced anti-inflammatory effects. Apolipoprotein E and glycerolipids are enriched in Lipo-NPs compared to ADSC-EVs. Additionally, the uptake of Lipo-NPs in hepatocytes and macrophages is higher. Lipo-NPs and ADSC-EVs have comparable protective and anti-inflammatory effects. Specifically, Lipo-NPs reduce toll-like receptor 4-induced secretion of inflammatory cytokines in macrophages. Guanabenz-loaded Lipo-NPs further suppress inflammatory pathways, suggesting that this combination therapy can have promising applications for inflammatory diseases.


Assuntos
Tecido Adiposo , Vesículas Extracelulares , Inflamação , Nanopartículas , Tecido Adiposo/química , Anti-Inflamatórios/economia , Anti-Inflamatórios/uso terapêutico , Humanos , Inflamação/terapia , Células-Tronco Mesenquimais/metabolismo
7.
Hepatol Commun ; 4(2): 298-313, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32025612

RESUMO

By exploiting their biological functions, the use of biological nanoparticles such as extracellular vesicles can provide an efficient and effective approach for hepatic delivery of RNA-based therapeutics for the treatment of liver cancers such as hepatocellular cancer (HCC). Targeting liver cancer stem cells (LCSC) within HCC provide an untapped opportunity to improve outcomes by enhancing therapeutic responses. Cells with tumor-initiating capabilities such as LCSC can be identified by expression of markers such as epithelial cell adhesion molecule (EpCAM) on their cell surface. EpCAM is a target of Wnt/ß-catenin signaling, a fundamental pathway in stem-cell growth. Moreover, mutations in the ß-catenin gene are frequently observed in HCC and can be associated with constitutive activation of the Wnt/ß-catenin pathway. However, targeting these pathways for the treatment of HCC has been challenging. Using RNA nanotechnology, we developed engineered biological nanoparticles capable of specific and effective delivery of RNA therapeutics targeting ß-catenin to LCSC. Extracellular vesicles isolated from milk were loaded with small interfering RNA to ß-catenin and decorated with RNA scaffolds to incorporate RNA aptamers capable of binding to EpCAM. Cellular uptake of these EpCAM-targeting therapeutic milk-derived nanovesicles in vitro resulted in loss of ß-catenin expression and decreased proliferation. The uptake and therapeutic efficacy of these engineered biological nanotherapeutics was demonstrated in vivo using tumor xenograft mouse models. Conclusion: ß-catenin can be targeted directly to control the proliferation of hepatic cancer stem cells using small interfering RNA delivered using target-specific biological nanoparticles. Application of this RNA nanotechnology-based approach to engineer biological nanotherapeutics provides a platform for developing cell-surface molecule-directed targeted therapeutics.

8.
Mol Oncol ; 9(9): 1783-98, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26105207

RESUMO

Src-family kinase (SFK) signaling impacts multiple tumor-related properties, particularly in the context of the brain tumor glioblastoma. Consequently, the pan-SFK inhibitor dasatinib has emerged as a therapeutic strategy, despite physiologic limitations to its effectiveness in the brain. We investigated the importance of individual SFKs (Src, Fyn, Yes, and Lyn) to glioma tumor biology by knocking down individual SFK expression both in culture (LN229, SF767, GBM8) and orthotopic xenograft (GBM8) contexts. We evaluated the effects of these knockdowns on tumor cell proliferation, migration, and motility-related signaling in culture, as well as overall survival in the orthotopic xenograft model. The four SFKs differed significantly in their importance to these properties. In culture, Src, Fyn, and Yes knockdown generally reduced growth and migration and altered motility-related phosphorylation patterns while Lyn knockdown did so to a lesser extent. However the details of these effects varied significantly depending on the cell line: in no case were conclusions about the role of a particular SFK applicable to all of the measures or all of the cell types examined. In the orthotopic xenograft model, mice implanted with non-target or Src or Fyn knockdown cells showed no differences in survival. In contrast, mice implanted with Yes knockdown cells had longer survival, associated with reduced tumor cell proliferation. Those implanted with Lyn knockdown cells had shorter survival, associated with higher overall tumor burden. Together, our results suggest that Yes signaling directly affects tumor cell biology in a pro-tumorigenic manner, while Lyn signaling affects interactions between tumor cells and the microenvironment in an anti-tumor manner. In the context of therapeutic targeting of SFKs, these results suggest that pan-SFK inhibitors may not produce the intended therapeutic benefit when Lyn is present.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Dasatinibe/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Inibidores de Proteínas Quinases/uso terapêutico , Quinases da Família src/genética , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias Encefálicas/enzimologia , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dasatinibe/farmacologia , Técnicas de Silenciamento de Genes , Glioblastoma/enzimologia , Glioblastoma/patologia , Humanos , Camundongos Nus , Inibidores de Proteínas Quinases/farmacologia , Quinases da Família src/metabolismo
10.
PLoS One ; 10(3): e0122209, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25816016

RESUMO

Folate receptor alpha (FOLR1) has been identified as a potential prognostic and therapeutic target in a number of cancers. A correlation has been shown between intense overexpression of FOLR1 in breast tumors and poor prognosis, yet there is limited examination of the distribution of FOLR1 across clinically relevant breast cancer subtypes. To explore this further, we used RNA-seq data from multiple patient cohorts to analyze the distribution of FOLR1 mRNA across breast cancer subtypes comprised of estrogen receptor positive (ER+), human epidermal growth factor receptor positive (HER2+), and triple negative (TNBC) tumors. FOLR1 expression varied within breast tumor subtypes; triple negative/basal tumors were significantly associated with increased expression of FOLR1 mRNA, compared to ER+ and HER2+ tumors. However, subsets of high level FOLR1 expressing tumors were observed in all clinical subtypes. These observations were supported by immunohistochemical analysis of tissue microarrays, with the largest number of 3+ positive tumors and highest H-scores of any subtype represented by triple negatives, and lowest by ER+ tumors. FOLR1 expression did not correlate to common clinicopathological parameters such as tumor stage and nodal status. To delineate the importance of FOLR1 overexpression in triple negative cancers, RNA-interference was used to deplete FOLR1 in overexpressing triple negative cell breast lines. Loss of FOLR1 resulted in growth inhibition, whereas FOLR1 overexpression promoted folate uptake and growth advantage in low folate conditions. Taken together, our data suggests patients with triple negative cancers expressing high FOLR1 expression represent an important population of patients that may benefit from targeted anti-FOLR1 therapy. This may prove particularly helpful for a large number of patients who would typically be classified as triple negative and who to this point have been left without any targeted treatment options.


Assuntos
Receptor 1 de Folato/metabolismo , Neoplasias de Mama Triplo Negativas/genética , Linhagem Celular Tumoral , Feminino , Receptor 1 de Folato/genética , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Regulação para Cima
11.
Mol Cell Biol ; 33(24): 4909-18, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24126053

RESUMO

The role of RhoA in promoting directed cell migration has been complicated by studies showing that it is activated both in the front and the rear of migrating cells. We report here that the RhoA-specific guanine nucleotide exchange factor Syx is required for the polarity of actively migrating brain and breast tumor cells. This function of Syx is mediated by the selective activation of the RhoA downstream effector Dia1, the subsequent reorganization of microtubules, and the downregulation of focal adhesions and actin stress fibers. The data argue that directed cell migration requires the precise spatiotemporal regulation of Dia1 and ROCK activities in the cell. The recruitment of Syx to the cell membrane and the subsequent selective activation of Dia1 signaling, coupled with the suppression of ROCK and activation of cofilin-mediated actin reorganization, plays a key role in establishing cell polarity during directed cell migration.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Movimento Celular , Fatores de Troca do Nucleotídeo Guanina/fisiologia , Quinases Associadas a rho/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular Tumoral , Polaridade Celular , Forma Celular , Adesões Focais/metabolismo , Forminas , Técnicas de Silenciamento de Genes , Humanos , Microtúbulos/metabolismo , Microtúbulos/ultraestrutura , Fenótipo , Estabilidade Proteica , Transporte Proteico , RNA Interferente Pequeno/genética
12.
PLoS One ; 8(2): e56505, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23457577

RESUMO

Anti-VEGF antibody therapy with bevacizumab provides significant clinical benefit in patients with recurrent glioblastoma multiforme (GBM). Unfortunately, progression on bevacizumab therapy is often associated with a diffuse disease recurrence pattern, which limits subsequent therapeutic options. Therefore, there is an urgent need to understand bevacizumab's influence on glioma biology and block it's actions towards cell invasion. To explore the mechanism(s) of GBM cell invasion we have examined a panel of serially transplanted human GBM lines grown either in short-term culture, as xenografts in mouse flank, or injected orthotopically in mouse brain. Using an orthotopic xenograft model that exhibits increased invasiveness upon bevacizumab treatment, we also tested the effect of dasatinib, a broad spectrum SFK inhibitor, on bevacizumab-induced invasion.We show that 1) activation of Src family kinases (SFKs) is common in GBM, 2) the relative invasiveness of 17 serially transplanted GBM xenografts correlates strongly with p120 catenin phosphorylation at Y228, a Src kinase site, and 3) SFK activation assessed immunohistochemically in orthotopic xenografts, as well as the phosphorylation of downstream substrates occurs specifically at the invasive tumor edge. Further, we show that SFK signaling is markedly elevated at the invasive tumor front upon bevacizumab administration, and that dasatinib treatment effectively blocked the increased invasion induced by bevacizumab.Our data are consistent with the hypothesis that the increased invasiveness associated with anti-VEGF therapy is due to increased SFK signaling, and support testing the combination of dasatinib with bevacizumab in the clinic.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Glioma/patologia , Terapia de Alvo Molecular , Inibidores de Proteínas Quinases/farmacologia , Quinases da Família src/antagonistas & inibidores , Animais , Bevacizumab , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Dasatinibe , Ativação Enzimática/efeitos dos fármacos , Humanos , Camundongos , Invasividade Neoplásica , Pirimidinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia , Quinases da Família src/metabolismo
13.
J Cell Biol ; 199(7): 1103-15, 2012 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-23253477

RESUMO

Vascular endothelial growth factor (VEGF) and Ang1 (Angiopoietin-1) have opposing effects on vascular permeability, but the molecular basis of these effects is not fully known. We report in this paper that VEGF and Ang1 regulate endothelial cell (EC) junctions by determining the localization of the RhoA-specific guanine nucleotide exchange factor Syx. Syx was recruited to junctions by members of the Crumbs polarity complex and promoted junction integrity by activating Diaphanous. VEGF caused translocation of Syx from cell junctions, promoting junction disassembly, whereas Ang1 maintained Syx at the junctions, inducing junction stabilization. The VEGF-induced translocation of Syx from EC junctions was caused by PKD1 (protein kinase D1)-mediated phosphorylation of Syx at Ser(806), which reduced Syx association to its junctional anchors. In support of the pivotal role of Syx in regulating EC junctions, syx(-/-) mice had defective junctions, resulting in vascular leakiness, edema, and impaired heart function.


Assuntos
Angiopoietina-1/fisiologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Junções Intercelulares/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia , Animais , Permeabilidade Capilar , Proteínas de Transporte/metabolismo , Cães , Forminas , Técnicas de Silenciamento de Genes , Fatores de Troca do Nucleotídeo Guanina/genética , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Células Madin Darby de Rim Canino , Proteínas de Membrana , Camundongos , Camundongos Knockout , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Estabilidade Proteica , Transporte Proteico , Interferência de RNA , Transdução de Sinais , Volume Sistólico , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/patologia
14.
Biochim Biophys Acta ; 1826(1): 23-31, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22440943

RESUMO

In the context of cancer, E-cadherin has traditionally been categorized as a tumor suppressor, given its essential role in the formation of proper intercellular junctions, and its downregulation in the process of epithelial-mesenchymal transition (EMT) in epithelial tumor progression. Germline or somatic mutations in the E-cadherin gene (CDH1) or downregulation by epigenetic mechanisms have been described in a small subset of epithelial cancers. However, recent evidence also points toward a promoting role of E-cadherin in several aspects of tumor progression. This includes preserved (or increased) E-cadherin expression in microemboli of inflammatory breast carcinoma, a possible "mesenchymal to epithelial transition" (MET) in ovarian carcinoma, collective cell invasion in some epithelial cancers, a recent association of E-cadherin expression with a more aggressive brain tumor subset, as well as the intriguing possibility of E-cadherin involvement in specific signaling networks in the cytoplasm and/or nucleus. In this review we address a lesser-known, positive role for E-cadherin in cancer.


Assuntos
Caderinas/genética , Neoplasias/genética , Neoplasias/patologia , Animais , Caderinas/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Humanos , Neoplasias/metabolismo , Transdução de Sinais
15.
PLoS One ; 5(10): e13665, 2010 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-21060868

RESUMO

BACKGROUND: Cadherins are essential components of the adherens junction complexes that mediate cell-cell adhesion and regulate cell motility. During tissue morphogenesis, changes in cadherin expression (known as cadherin switching) are a common mechanism for altering cell fate. Cadherin switching is also common during epithelial tumor progression, where it is thought to promote tumor invasion and metastasis. E-cadherin is the predominant cadherin expressed in epithelial tissues, but its expression is very limited in normal brain. METHODOLOGY/PRINCIPAL FINDINGS: We identified E-cadherin expression in a retrospective series of glioblastomas exhibiting epithelial or pseudoepithelial differentiation. Unlike in epithelial tissues, E-cadherin expression in gliomas correlated with an unfavorable clinical outcome. Western blotting of two panels of human GBM cell lines propagated either as xenografts in nude mice or grown under conventional cell culture conditions confirmed that E-cadherin expression is rare. However, a small number of xenograft lines did express E-cadherin, its expression correlating with increased invasiveness when the cells were implanted orthotopically in mouse brain. In the conventionally cultured SF767 glioma cell line, E-cadherin expression was localized throughout the plasma membrane rather than being restricted to areas of cell-cell contact. ShRNA knockdown of E-cadherin in these cells resulted in decreased proliferation and migration in vitro. CONCLUSIONS/SIGNIFICANCE: Our data shows an unexpected correlation between the abnormal expression of E-cadherin in a subset of GBM tumor cells and the growth and migration of this aggressive brain tumor subtype.


Assuntos
Neoplasias Encefálicas/metabolismo , Caderinas/metabolismo , Glioblastoma/metabolismo , Animais , Western Blotting , Neoplasias Encefálicas/patologia , Diferenciação Celular , Divisão Celular , Linhagem Celular Tumoral , Glioblastoma/patologia , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Estudos Retrospectivos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
16.
Neurosurg Focus ; 21(4): E13, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17112191

RESUMO

Cell-cell adhesion is a crucial process occurring during normal tissue development. Cadherins are calcium-dependent cell-surface adhesion molecules involved in cell-cell adhesion. They reorganize the actin cytoskeleton via interaction with the catenins. Modulation of the cadherin/catenin system plays a role in cell motility. Dysregulation of the cadherin/catenin assembly has been implicated in various cancers. In this review, the authors summarize all studies focusing on the role of cadherins and catenins in glioma formation. With the emergence of recent data regarding gliomas' putative cell of origin, elucidation of the role of cadherins/catenins in gliomagenesis will become important in devising new therapeutic approaches against such deadly cancers.


Assuntos
Neoplasias Encefálicas/fisiopatologia , Caderinas/fisiologia , Cateninas/fisiologia , Glioma/fisiopatologia , Adesão Celular/fisiologia , Transformação Celular Neoplásica , Humanos
17.
Ann N Y Acad Sci ; 1069: 1-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16855130

RESUMO

The development of glucocorticoid (GC) resistance is a serious problem that complicates the treatment of immune-related diseases, such as asthma, ulcerative colitis, and hematologic cancers. hGRalpha and hGRbeta are two isoforms of the human glucocorticoid receptor, which differ in the structural composition of the carboxy-terminal end of the ligand-binding domain and therefore in their ability to bind glucocorticoid ligand and in their physiological function. hGRalpha is the classically functional GR, while hGRbeta seems to act mainly as a dominant negative to the function of hGRalpha. Because of the ability of hGRbeta to antagonize the action of hGRalpha, it has been hypothesized that changes in the expression of hGRbeta may underlie the development of glucocorticoid resistance. In this article we review what is known about the expression and physiological action of hGRbeta in normal cells and tissue as well as in several disease states. Taken together, the evidence suggests that the ratio of hGRalpha:hGRbeta expression is indeed critical to the glucocorticoid responsiveness of various cells. This ratio can be altered by changing the expression level of hGRalpha, hGRbeta, or both receptors simultaneously. Higher ratios correlate with glucocorticoid sensitivity, while lower ratios correlate with glucocorticoid resistance. Thus hGRbeta can be an important modulator of glucocorticoid responsiveness.


Assuntos
Resistência a Medicamentos/efeitos dos fármacos , Resistência a Medicamentos/fisiologia , Glucocorticoides/farmacologia , Receptores de Glucocorticoides/metabolismo , Animais , Doença , Regulação da Expressão Gênica , Humanos , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/genética
18.
Mol Cell Neurosci ; 25(3): 536-47, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15033181

RESUMO

In neurons and neuroendocrine cells, tyrosine hydroxylase (TH) gene expression is induced by stimuli that elevate cAMP, by depolarization, and by hypoxia. Using these stimuli, we examined TH promoter mutants, cAMP response element binding protein (CREB) phosphorylation site mutants, and transcriptional interference with dominant negative transcription factors to assess the relative contributions of CREB/AP-1 family members to the regulation of basal and inducible TH transcription in PC12 cells. We found that basal transcription depends on transcription factor activity at the partial dyad (-17 bp), CRE (-45 bp), and AP1 (-205 bp) elements. Induced transcription is regulated primarily by activity at the CRE, with only small contributions from the AP1 or hypoxia response element 1 (HRE1; -225 bp) elements, regardless of inducing stimulus. CREB, ATF-1, and CREMtau all mediate CRE-dependent transcription, with CREB and CREMtau being more effective than ATF-1. Phosphorylation of CREB on Ser133, but not on Ser142 or Ser143, is required for induced transcription, regardless of inducing stimulus.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/biossíntese , Transcrição Gênica/fisiologia , Tirosina 3-Mono-Oxigenase/biossíntese , Animais , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , AMP Cíclico/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Células PC12 , Ratos , Transcrição Gênica/efeitos dos fármacos , Tirosina 3-Mono-Oxigenase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA