Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
bioRxiv ; 2024 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-38328202

RESUMO

Glioblastoma (GBM) is the most common primary tumor of the central nervous system. One major challenge in GBM treatment is the resistance to chemotherapy and radiotherapy observed in subpopulations of cancer cells, including GBM stem-like cells (GSCs). These cells hold the ability to self-renew or differentiate following treatment, participating in tumor recurrence. The gap junction protein connexin43 (Cx43) has complex roles in oncogenesis and we have previously demonstrated an association between Cx43 and GBM chemotherapy resistance. Here, we report, for the first time, increased direct interaction between non-junctional Cx43 with microtubules in the cytoplasm of GSCs. We hypothesize that non-junctional Cx43/microtubule complexing is critical for GSC maintenance and survival and sought to specifically disrupt this interaction while maintaining other Cx43 functions, such as gap junction formation. Using a Cx43 mimetic peptide of the carboxyl terminal tubulin-binding domain of Cx43 (JM2), we successfully ablated Cx43 interaction with microtubules in GSCs. Importantly, administration of JM2 significantly decreased GSC survival in vitro , and limited GSC-derived tumor growth in vivo . Together, these results identify JM2 as a novel peptide drug to ablate GSCs in GBM treatment.

2.
Materials (Basel) ; 15(22)2022 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-36431555

RESUMO

The characteristics of local strain distribution and evolution of duplex stainless steel during the tensile process were studied using the digital image correlation (DIC) technique. In addition, the finite element inversion of nanoindentation experiments of austenitic and ferrite phases in duplex stainless steel was carried out to obtain the stress-strain response of the two phases. Further, based on the representative volume element (RVE) and the material parameters obtained from the finite element inversion method, the local stress and strain behavior of duplex stainless steel at microscale was simulated numerically. The results fit well with the experiments, showing that the austenite phase is softer than ferrite phase, with the larger strain zone concentrated in the austenite phase and the larger stress zone concentrated in the ferrite phase. The grain boundaries are prone to obvious stress and strain concentrations. The local stress and strain distributions are influenced by the shape and interaction of the grains, while the distribution features become more obvious as the load increases. The research results effectively reveal the two-phase interaction and local failure mechanism of duplex stainless steel, and may provide a reference for material preparation and safety design of related structures.

3.
Oncogenesis ; 11(1): 2, 2022 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-35022385

RESUMO

Circumventing chemoresistance is crucial for effectively treating cancer including glioblastoma, a lethal brain cancer. The gap junction protein connexin 43 (Cx43) renders glioblastoma resistant to chemotherapy; however, targeting Cx43 is difficult because mechanisms underlying Cx43-mediated chemoresistance remain elusive. Here we report that Cx43, but not other connexins, is highly expressed in a subpopulation of glioblastoma and Cx43 mRNA levels strongly correlate with poor prognosis and chemoresistance in this population, making Cx43 the prime therapeutic target among all connexins. Depleting Cx43 or treating cells with αCT1-a Cx43 peptide inhibitor that sensitizes glioblastoma to the chemotherapy temozolomide-inactivates phosphatidylinositol-3 kinase (PI3K), whereas overexpression of Cx43 activates this signaling. Moreover, αCT1-induced chemo-sensitization is counteracted by a PI3K active mutant. Further research reveals that αCT1 inactivates PI3K without blocking the release of PI3K-activating molecules from membrane channels and that Cx43 selectively binds to the PI3K catalytic subunit ß (PIK3CB, also called PI3Kß or p110ß), suggesting that Cx43 activates PIK3CB/p110ß independent of its channel functions. To explore the therapeutic potential of simultaneously targeting Cx43 and PIK3CB/p110ß, αCT1 is combined with TGX-221 or GSK2636771, two PIK3CB/p110ß-selective inhibitors. These two different treatments synergistically inactivate PI3K and sensitize glioblastoma cells to temozolomide in vitro and in vivo. Our study has revealed novel mechanistic insights into Cx43/PI3K-mediated temozolomide resistance in glioblastoma and demonstrated that targeting Cx43 and PIK3CB/p110ß together is an effective therapeutic approach for overcoming chemoresistance.

4.
Mol Cancer Ther ; 21(2): 322-335, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34789563

RESUMO

MET-targeted therapies are clinically effective in MET-amplified and MET exon 14 deletion mutant (METex14) non-small cell lung cancers (NSCLCs), but their efficacy is limited by the development of drug resistance. Structurally distinct MET tyrosine kinase inhibitors (TKIs) (type I/II) have been developed or are under clinical evaluation, which may overcome MET-mediated drug resistance mechanisms. In this study, we assess secondary MET mutations likely to emerge in response to treatment with single-agent or combinations of type I/type II MET TKIs using TPR-MET transformed Ba/F3 cell mutagenesis assays. We found that these inhibitors gave rise to distinct secondary MET mutant profiles. However, a combination of type I/II TKI inhibitors (capmatinib and merestinib) yielded no resistant clones in vitro The combination of capmatinib/merestinib was evaluated in vivo and led to a significant reduction in tumor outgrowth compared with either MET inhibitor alone. Our findings demonstrate in vitro and in vivo that a simultaneous treatment with a type I and type II MET TKI may be a clinically viable approach to delay and/or diminish the emergence of on target MET-mediated drug-resistance mutations.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Simulação de Acoplamento Molecular/métodos , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Feminino , Humanos , Camundongos , Inibidores de Proteínas Quinases/farmacologia
5.
Ann Transl Med ; 9(10): 887, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34164521

RESUMO

BACKGROUND: Obesity is associated with many adverse effects on female fertility. Obese women have a higher likelihood of developing ovulatory dysfunction due to dysregulation of the hypothalamic-pituitary-ovarian axis. However, the effect of obesity on ovarian function during early pregnancy needs to be further assessed. METHODS: C57BL6/J mice were given a high-fat diet (HFD) for 12 weeks to induce obesity. An in vitro high-fat model was established by treating the human ovarian granulosa cell line KGN with oleic acid and palmitic acid. Ovarian morphology of obese mice in early pregnancy was assessed by hematoxylin and eosin staining and ovarian function was assessed by enzyme-linked immunosorbent assay, western blotting, and immunohistochemistry. Oil Red O staining and transmission electron microscopy were used to detect fatty acid accumulation. Specific markers relating to the ovarian functional mechanism were assessed by real-time PCR, western blotting, lactate detection, adenosine triphosphate (ATP) detection, biochemical analyses, and enzyme-linked immunosorbent assay. RESULTS: The results of this study showed that during early pregnancy, the number of corpus lutea, serum estradiol and progesterone levels, and the expression of the steroid biosynthesis-related protein CYP19A1 (aromatase), CYP11A1 (cholesterol side chain cleavage enzyme), and StAR (steroidogenic acute regulatory protein), were significantly increased in HFD mice. Mice fed an HFD also showed a significant increase in ovarian lipid accumulation on day 7 of pregnancy. Genes involved in fatty acid synthesis (Acsl4 and Elovl5), and fatty acid uptake and transport (Slc27a4), together with the ß-oxidation rate-limiting enzyme Cpt1a, were significantly upregulated in HFD mice. Specifically, there was abnormal elevation of ATP and aberrant expression of tricarboxylic acid cycle (TCA)- and electron transport chain (ETC)-related genes in the ovaries of pregnant HFD mice. KGN cells treated with etomoxir targeting ß-oxidation of fatty acid showed decreased TCA cycle and ETC related gene expression. The elevation of ATP and estradiol and progesterone levels was reversed. CONCLUSIONS: During early pregnancy, HFD-induced obesity increases fatty acid ß-oxidation, which in turn increases TCA cycle and ETC related gene expression, leading to increased ATP production and ovarian dysfunction.

6.
J Endocrinol ; 245(3): 357-368, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32208360

RESUMO

Embryo implantation is essential for normal pregnancy. Decidualization is known to facilitate embryo implantation and maintain pregnancy. Uterine stromal cells undergo transformation into decidual cells after embryo attachment to the endometrium. Pyruvate kinase M2 (PKM2) is a rate limiting enzyme in the glycolysis process which catalyzes phosphoenolpyruvic acid into pyruvate. However, little is known regarding the role of PKM2 during endometrial decidualization. In this study, PKM2 was found to be mainly located in the uterine glandular epithelium and luminal epithelium on day 1 and day 4 of pregnancy and strongly expressed in the decidual zone after embryo implantation. PKM2 was dramatically increased with the onset of decidualization. Upon further exploration, PKM2 was found to be more highly expressed at the implantation sites than at the inter-implantation sites on days 5 to 7 of pregnancy. PKM2 expression was also significantly increased after artificial decidualization both in vivo and in vitro. After PKM2 expression was knocked down by siRNA, the number of embryo implantation sites in mice on day 7 of pregnancy was significantly reduced, and the decidualization markers BMP2 and Hoxa10 were also obviously downregulated in vivo and in vitro. Downregulated PKM2 could also compromise cell proliferation in primary endometrial stromal cells and in Ishikawa cells. The migration rate of Ishikawa cells was also obviously suppressed by si-PKM2 according to the wound healing assay. In conclusion, PKM2 might play an important role in decidualization during early pregnancy, and cell proliferation might be one pathway for PKM2 regulated decidualization.


Assuntos
Endométrio/metabolismo , Piruvato Quinase/metabolismo , Animais , Proliferação de Células/fisiologia , Decídua/metabolismo , Implantação do Embrião/fisiologia , Feminino , Camundongos , Gravidez , Células Estromais/metabolismo , Útero/metabolismo , Cicatrização/genética , Cicatrização/fisiologia
7.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 33(5): 537-541, 2019 May 15.
Artigo em Chinês | MEDLINE | ID: mdl-31090344

RESUMO

OBJECTIVE: To explore the effectiveness and safety of closed reduction combined with Taylor three-dimensional space stent fixation in treatment of supracondylar femoral fractures in children. METHODS: Between July 2008 and July 2016, 20 patients with supracondylar femoral fractures were treated with closed reduction combined with Taylor three-dimensional space stent fixation. There were 14 males and 6 females, with an average age of 10.3 years (range, 6-14 years). The cause of injury was traffic accident in 5 cases, falling from high place in 6 cases, and falling in 9 cases. All fractures were closed fractures. Among them, 12 cases were flexion type and 8 cases were straight type. According to AO classification, 12 cases were rated as type A1 and 8 cases as type A2. The fractures were over 0.5-5.0 cm (mean, 2.5 cm) of the epiphysis line. The time from injury to surgery was 2-8 days (mean, 3.5 days). Postoperative knee joint function was evaluated based on the Kolment evaluation criteria. RESULTS: All children were followed up 6-24 months (mean, 18.1 months). There was no complication such as nail infection, vascular nerve injury, external fixation looseing, fracture displacement, or re-fracture. All fractures healed and the fracture healing time was 4-6 weeks with an average of 4.5 weeks. The stent removal time was 8-12 weeks (mean, 9.5 weeks). The gait and knee function recovered, and there was no abnormality of the epiphysis. At last follow-up, the knee joint function were excellent in 18 cases and good in 2 cases according to the Kolment evaluation criteria, and the excellent and good rate was 100%. CONCLUSION: Closed reduction combined with Taylor three-dimensional space stent fixation is an effective treatment for the children with supracondylar femoral fractures, with small trauma and rapid recovery. It can avoid damaging the tarsal plate, be high fracture healing rate, and promote the recovery of limb function.


Assuntos
Fraturas do Colo Femoral , Fraturas Fechadas , Stents , Adolescente , Criança , Feminino , Fixação Interna de Fraturas , Consolidação da Fratura , Humanos , Masculino , Resultado do Tratamento
8.
Cancer Discov ; 9(1): 34-45, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30297358

RESUMO

KRAS-driven lung cancers frequently inactivate TP53 and/or STK11/LKB1, defining tumor subclasses with emerging clinical relevance. Specifically, KRAS-LKB1 (KL)-mutant lung cancers are particularly aggressive, lack PD-L1, and respond poorly to immune checkpoint blockade (ICB). The mechanistic basis for this impaired immunogenicity, despite the overall high mutational load of KRAS-mutant lung cancers, remains obscure. Here, we report that LKB1 loss results in marked silencing of stimulator of interferon genes (STING) expression and insensitivity to cytoplasmic double-strand DNA (dsDNA) sensing. This effect is mediated at least in part by hyperactivation of DNMT1 and EZH2 activity related to elevated S-adenylmethionine levels and reinforced by DNMT1 upregulation. Ectopic expression of STING in KL cells engages IRF3 and STAT1 signaling downstream of TBK1 and impairs cellular fitness, due to the pathologic accumulation of cytoplasmic mitochondrial dsDNA associated with mitochondrial dysfunction. Thus, silencing of STING avoids these negative consequences of LKB1 inactivation, while facilitating immune escape. SIGNIFICANCE: Oncogenic KRAS-mutant lung cancers remain treatment-refractory and are resistant to ICB in the setting of LKB1 loss. These results begin to uncover the key underlying mechanism and identify strategies to restore STING expression, with important therapeutic implications because mitochondrial dysfunction is an obligate component of this tumor subtype.See related commentary by Corte and Byers, p. 16.This article is highlighted in the In This Issue feature, p. 1.


Assuntos
Adenocarcinoma/genética , Deleção de Genes , Neoplasias Pulmonares/genética , Proteínas de Membrana/metabolismo , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Quinases Proteína-Quinases Ativadas por AMP , Adenocarcinoma/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , Fator Regulador 3 de Interferon/metabolismo , Neoplasias Pulmonares/metabolismo , Proteínas de Membrana/genética , Mutação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Fator de Transcrição STAT1/metabolismo
9.
Sci Rep ; 8(1): 13621, 2018 09 11.
Artigo em Inglês | MEDLINE | ID: mdl-30206363

RESUMO

Glioblastoma is the most common malignant brain cancer with a dismal prognosis. The difficulty in treating glioblastoma is largely attributed to the lack of effective therapeutic targets. In our previous work, we identified casein kinase 1 ε (CK1ε, also known as CSNK1E) as a potential survival factor in glioblastoma. However, how CK1ε controls cell survival remains elusive and whether targeting CK1ε is a possible treatment for glioblastoma requires further investigation. Here we report that CK1ε was expressed at the highest level among six CK1 isoforms in glioblastoma and enriched in high-grade glioma, but not glia cells. Depletion of CK1ε remarkably inhibited the growth of glioblastoma cells and suppressed self-renewal of glioblastoma stem cells, while having limited effect on astrocytes. CK1ε deprivation activated ß-catenin and induced apoptosis, which was further counteracted by knockdown of ß-catenin. The CK1ε inhibitor IC261, but not PF-4800567, activated ß-catenin and blocked the growth of glioblastoma cells and glioblastoma stem cells. Congruently, IC261 elicited a robust growth inhibition of human glioblastoma xenografts in mice. Together, our results demonstrate that CK1ε regulates the survival of glioblastoma cells and glioblastoma stem cells through ß-catenin signaling, underscoring the importance of targeting CK1ε as an effective treatment for glioblastoma.


Assuntos
Caseína Quinase I/metabolismo , Glioblastoma/enzimologia , Proteínas de Neoplasias/metabolismo , Transdução de Sinais , Animais , Caseína Quinase I/antagonistas & inibidores , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Indóis/farmacologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/antagonistas & inibidores , Floroglucinol/análogos & derivados , Floroglucinol/farmacologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , beta Catenina/metabolismo
10.
Cancer Cell ; 34(3): 439-452.e6, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30205046

RESUMO

Despite extensive efforts, oncogenic KRAS remains resistant to targeted therapy. Combined downstream RAL-TBK1 and MEK inhibition induces only transient lung tumor shrinkage in KRAS-driven genetically engineered mouse models (GEMMs). Using the sensitive KRAS;LKB1 (KL) mutant background, we identify YAP1 upregulation and a therapy-induced secretome as mediators of acquired resistance. This program is reversible, associated with H3K27 promoter acetylation, and suppressed by BET inhibition, resensitizing resistant KL cells to TBK1/MEK inhibition. Constitutive YAP1 signaling promotes intrinsic resistance in KRAS;TP53 (KP) mutant lung cancer. Intermittent treatment with the BET inhibitor JQ1 thus overcomes resistance to combined pathway inhibition in KL and KP GEMMs. Using potent and selective TBK1 and BET inhibitors we further develop an effective therapeutic strategy with potential translatability to the clinic.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antineoplásicos Imunológicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/imunologia , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Resistencia a Medicamentos Antineoplásicos/imunologia , Células HEK293 , Humanos , Imunidade Inata/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/imunologia , Fator de Crescimento Insulin-Like I/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Transgênicos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Fosfoproteínas/imunologia , Fosfoproteínas/metabolismo , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/imunologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Fatores de Transcrição , Proteínas de Sinalização YAP
11.
Sci Rep ; 8(1): 2822, 2018 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-29434216

RESUMO

Dysregulated autophagy is central to the pathogenesis and therapeutic development of cancer. However, how autophagy is regulated in cancer is not well understood and genes that modulate cancer autophagy are not fully defined. To gain more insights into autophagy regulation in cancer, we performed a large-scale RNA interference screen in K562 human chronic myeloid leukemia cells using monodansylcadaverine staining, an autophagy-detecting approach equivalent to immunoblotting of the autophagy marker LC3B or fluorescence microscopy of GFP-LC3B. By coupling monodansylcadaverine staining with fluorescence-activated cell sorting, we successfully isolated autophagic K562 cells where we identified 336 short hairpin RNAs. After candidate validation using Cyto-ID fluorescence spectrophotometry, LC3B immunoblotting, and quantitative RT-PCR, 82 genes were identified as autophagy-regulating genes. 20 genes have been reported previously and the remaining 62 candidates are novel autophagy mediators. Bioinformatic analyses revealed that most candidate genes were involved in molecular pathways regulating autophagy, rather than directly participating in the autophagy process. Further autophagy flux assays revealed that 57 autophagy-regulating genes suppressed autophagy initiation, whereas 21 candidates promoted autophagy maturation. Our RNA interference screen identifies identified genes that regulate autophagy at different stages, which helps decode autophagy regulation in cancer and offers novel avenues to develop autophagy-related therapies for cancer.


Assuntos
Autofagia/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Proteínas Reguladoras de Apoptose/metabolismo , Cadaverina/análogos & derivados , Cadaverina/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Citometria de Fluxo , Corantes Fluorescentes , Ensaios de Triagem em Larga Escala , Humanos , Células K562 , Microscopia de Fluorescência , Interferência de RNA , RNA Interferente Pequeno , Espectrometria de Fluorescência
12.
Neuro Oncol ; 20(4): 494-505, 2018 03 27.
Artigo em Inglês | MEDLINE | ID: mdl-29016844

RESUMO

Background: Glioblastoma (GBM) is difficult to treat. Phosphoinositide 3-kinase (PI3K) is an attractive therapeutic target for GBM; however, targeting this pathway to effectively treat GBM is not successful because the roles of PI3K isoforms remain to be defined. The aim of this study is to determine whether PIK3CB/p110ß, but not other PI3K isoforms, is a biomarker for GBM recurrence and important for cell survival. Methods: Gene expression and clinical relevance of PI3K genes in GBM patients were analyzed using online databases. Expression/activity of PI3K isoforms was determined using immunoblotting. PI3K genes were inhibited using short hairpin RNAs or isoform-selective inhibitors. Cell viability/growth was assessed by the MTS assay and trypan blue exclusion assay. Apoptosis was monitored using the caspase activity assay. Mouse GBM xenograft models were used to gauge drug efficacy. Results: PIK3CB/p110ß was the only PI3K catalytic isoform that significantly correlated with high incidence rate, risk, and poor survival of recurrent GBM. PIK3CA/p110α, PIK3CB/p110ß, and PIK3CD/p110δ were differentially expressed in GBM cell lines and primary tumor cells derived from patient specimens, whereas PIK3CG/p110γ was barely detected. PIK3CB/p110ß protein levels presented a stronger association with the activities of PI3K signaling than other PI3K isoforms. Blocking p110ß deactivated PI3K signaling, whereas inhibition of other PI3K isoforms had no effect. Specific inhibitors of PIK3CB/p110ß, but not other PI3K isoforms, remarkably suppressed viability and growth of GBM cells and xenograft tumors in mice, with minimal cytotoxic effects on astrocytes. Conclusions: PIK3CB/p110ß is a biomarker for GBM recurrence and selectively important for GBM cell survival.


Assuntos
Biomarcadores Tumorais/metabolismo , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Glioblastoma/patologia , Recidiva Local de Neoplasia/patologia , Animais , Apoptose , Biomarcadores Tumorais/genética , Proliferação de Células , Classe I de Fosfatidilinositol 3-Quinases/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos SCID , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Prognóstico , Transdução de Sinais , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Ultrasound Med ; 36(2): 285-293, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-28039877

RESUMO

OBJECTIVES: Liver biopsy remains the reference standard for the assessment of liver fibrosis, but this procedure is invasive and can lead to complications. Thus, studies to determine the optimal noninvasive test are warranted. This study compared several noninvasive tests and their combinations for evaluating liver fibrosis stages in patients with chronic hepatitis B. METHODS: The shear wave velocity (SWV) and laboratory indicators were collected from 174 patients with chronic hepatitis B. Formulas were applied to calculate the serum fibrosis model, including the aspartate aminotransaminase-to-platelet ratio index (APRI), fibrosis-4 index (FIB-4) and aspartate aminotransferase-to-alanine aminotransferase ratio (AAR). The diagnostic performance of all noninvasive tests was assessed in comparison with percutaneous liver biopsy, based on a receiver operating characteristic curve analysis. RESULTS: The SWV (area under the receiver operating characteristic curve [AUC], 0.82) and APRI (AUC = 0.77) performed better than the FIB-4 (AUC = 0.62), and the AAR (AUC = 0.47) was not suitable for evaluating substantial liver fibrosis (stage ≥F2). The SWV (AUC = 0.96) was the best indicator, being superior to the APRI (AUC = 0.75) and FIB-4 (AUC = 0.74), and the AAR (AUC = 0.45) was not suitable for assessing cirrhosis (F4). Combining the SWV and APRI, the AUC improved to 0.85 for substantial liver fibrosis, and the sensitivity increased to 100% for cirrhosis. CONCLUSIONS: The SWV, APRI, and FIB-4 were valid tests for evaluating substantial liver fibrosis and cirrhosis. The combination of these tests with several noninvasive indicators is expected to enhance the assessment of liver fibrosis stages.


Assuntos
Algoritmos , Hepatite B Crônica/sangue , Hepatite B Crônica/diagnóstico por imagem , Cirrose Hepática/sangue , Cirrose Hepática/diagnóstico por imagem , Ultrassonografia , Adolescente , Adulto , Alanina Transaminase/sangue , Aspartato Aminotransferases/sangue , Plaquetas , Estudos de Coortes , Feminino , Hepatite B Crônica/complicações , Humanos , Fígado/diagnóstico por imagem , Cirrose Hepática/complicações , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Índice de Gravidade de Doença , Adulto Jovem
14.
Oncotarget ; 7(52): 86406-86419, 2016 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-27863440

RESUMO

The dismal prognosis of glioblastoma is, at least in part, attributable to the difficulty in eradicating glioblastoma stem cells (GSCs). However, whether this difficulty is caused by the differential responses of GSCs to drugs remains to be determined. To address this, we isolated and characterized ten GSC lines from established cell lines, xenografts, or patient specimens. Six lines formed spheres in a regular culture condition, whereas the remaining four lines grew as monolayer. These adherent lines formed spheres only in plates coated with poly-2-hydroxyethyl methacrylate. The self-renewal capabilities of GSCs varied, with the cell density needed for sphere formation ranging from 4 to 23.8 cells/well. Moreover, a single non-adherent GSC either remained quiescent or divided into two cells in four-seven days. The stem cell identity of GSCs was further verified by the expression of nestin or glial fibrillary acidic protein. Of the two GSC lines that were injected in immunodeficient mice, only one line formed a tumor in two months. The protein levels of NOTCH1 and platelet derived growth factor receptor alpha positively correlated with the responsiveness of GSCs to γ-secretase inhibitor IX or imatinib, two compounds that inhibit these two proteins, respectively. Furthermore, a combination of temozolomide and a connexin 43 inhibitor robustly inhibited the growth of GSCs. Collectively, our results demonstrate that patient-derived GSCs exhibit different growth rates in culture, possess differential capabilities to form a tumor, and have varied responses to targeted therapies. Our findings underscore the importance of patient-derived GSCs in glioblastoma research and therapeutic development.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Animais , Neoplasias Encefálicas/química , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Separação Celular , Proteína Glial Fibrilar Ácida/análise , Glioblastoma/química , Glioblastoma/patologia , Humanos , Camundongos , Receptor Notch1/análise , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/análise
15.
Methods Mol Biol ; 1465: 95-109, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27581142

RESUMO

Autophagy is a catabolic process whereby cellular components are degraded to fuel cells for longer survival during stress. Hence, autophagy plays a vital role in determining cell fate and is central for homeostasis and pathogenesis of many human diseases including chronic myeloid leukemia (CML). It has been well established that autophagy is important for the leukemogenesis as well as drug resistance in CML. Thus, autophagy is an intriguing therapeutic target. However, current approaches that detect autophagy lack reliability and often fail to provide quantitative measurements. To overcome this hurdle and facilitate the development of autophagy-related therapies, we have recently developed an autophagy assay termed as the Cyto-ID fluorescence spectrophotometric assay. This method uses a cationic fluorescence dye, Cyto-ID, which specifically labels autophagic compartments and is detected by a spectrophotometer to permit a large-scale and quantitative analysis. As such, it allows rapid, reliable, and quantitative detection of autophagy and estimation of autophagy flux. In this chapter, we further provide technical details of this method and step-by-step protocols for measuring autophagy or autophagy flux in CML cell lines as well as primary hematopoietic cells.


Assuntos
Autofagia , Corantes Fluorescentes/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Animais , Contagem de Células , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Células K562 , Camundongos , Espectrometria de Fluorescência
16.
Cancer Res ; 76(1): 139-49, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26542214

RESUMO

Resistance of glioblastoma (GBM) to the front-line chemotherapeutic agent temozolomide (TMZ) continues to challenge GBM treatment efforts. The repair of TMZ-induced DNA damage by O-6-methylguanine-DNA methyltransferase (MGMT) confers one mechanism of TMZ resistance. Paradoxically, MGMT-deficient GBM patients survive longer despite still developing resistance to TMZ. Recent studies indicate that the gap junction protein connexin 43 (Cx43) renders GBM cells resistant to TMZ through its carboxyl terminus (CT). In this study, we report insights into how Cx43 promotes TMZ resistance. Cx43 levels were inversely correlated with TMZ sensitivity of GBM cells, including GBM stem cells. Moreover, Cx43 levels inversely correlated with patient survival, including as observed in MGMT-deficient GBM patients. Addition of the C-terminal peptide mimetic αCT1, a selective inhibitor of Cx43 channels, sensitized human MGMT-deficient and TMZ-resistant GBM cells to TMZ treatment. Moreover, combining αCT1 with TMZ-blocked AKT/mTOR signaling, induced autophagy and apoptosis in TMZ-resistant GBM cells. Our findings suggest that Cx43 may offer a biomarker to predict the survival of patients with MGMT-independent TMZ resistance and that combining a Cx43 inhibitor with TMZ could enhance therapeutic responses in GBM, and perhaps other TMZ-resistant cancers.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Materiais Biomiméticos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Conexina 43/antagonistas & inibidores , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Peptídeos/farmacologia , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Conexina 43/metabolismo , Dacarbazina/administração & dosagem , Dacarbazina/farmacologia , Sinergismo Farmacológico , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Peptídeos/administração & dosagem , Transdução de Sinais , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Nano Lett ; 15(4): 2329-35, 2015 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-25734907

RESUMO

Nanoparticle-based therapy represents a novel and promising approach to treat glioblastoma, the most common and lethal malignant brain cancer. Although similar therapies have achieved significant cytotoxicity in cultured glioblastoma or glioblastoma stem cells (GSCs), the lack of an appropriate approach to monitor interactions between cells and nanoparticle-based therapies impedes their further clinical application in human patients. To address this critical issue, we first obtained NOTCH1 positive GSCs from patient-derived primary cultures. We then developed a new imaging approach to directly observe the dynamic nature of nanoparticles at the molecular level using in situ transmission electron microscopy (TEM). Utilizing these tools we were able to visualize real-time movements of nanoparticles interacting with GSCs for the first time. Overall, we show strong proof-of-concept results that real-time visualization of nanoparticles in single cells can be achieved at the nanoscale using TEM, thereby providing a powerful platform for the development of nanotherapeutics.


Assuntos
Glioblastoma/ultraestrutura , Dispositivos Lab-On-A-Chip , Microscopia Eletrônica de Transmissão/instrumentação , Imagem Molecular/instrumentação , Nanopartículas/ultraestrutura , Células-Tronco Neoplásicas/química , Linhagem Celular Tumoral , Sistemas Computacionais , Desenho de Equipamento , Análise de Falha de Equipamento , Glioblastoma/química , Humanos , Aumento da Imagem/instrumentação , Nanopartículas/química , Células-Tronco Neoplásicas/ultraestrutura
18.
Autophagy ; 11(3): 560-72, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25714620

RESUMO

The lack of a rapid and quantitative autophagy assay has substantially hindered the development and implementation of autophagy-targeting therapies for a variety of human diseases. To address this critical issue, we developed a novel autophagy assay using the newly developed Cyto-ID fluorescence dye. We first verified that the Cyto-ID dye specifically labels autophagic compartments with minimal staining of lysosomes and endosomes. We then developed a new Cyto-ID fluorescence spectrophotometric assay that makes it possible to estimate autophagy flux based on measurements of the Cyto-ID-stained autophagic compartments. By comparing to traditional autophagy approaches, we found that this assay yielded a more sensitive, yet less variable, quantification of the stained autophagic compartments and the estimate of autophagy flux. Furthermore, we tested the potential application of this autophagy assay in high throughput research by integrating it into an RNA interference (RNAi) screen and a small molecule screen. The RNAi screen revealed WNK2 and MAP3K6 as autophagy-modulating genes, both of which inhibited the MTOR pathway. Similarly, the small molecule screen identified sanguinarine and actinomycin D as potent autophagy inducers in leukemic cells. Moreover, we successfully detected autophagy responses to kinase inhibitors and chloroquine in normal or leukemic mice using this assay. Collectively, this new Cyto-ID fluorescence spectrophotometric assay provides a rapid, reliable quantification of autophagic compartments and estimation of autophagy flux with potential applications in developing autophagy-related therapies and as a test to monitor autophagy responses in patients being treated with autophagy-modulating drugs.


Assuntos
Autofagia , Corantes Fluorescentes/química , Espectrometria de Fluorescência/métodos , Animais , Sobrevivência Celular , Cloroquina/química , Dactinomicina/química , Endossomos/química , Regulação Leucêmica da Expressão Gênica , Células HEK293 , Células HeLa , Humanos , Mesilato de Imatinib/química , Células K562 , Leucemia/metabolismo , Lisossomos/química , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Microscopia de Fluorescência , Transplante de Neoplasias , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA
19.
Methods Mol Biol ; 1176: 59-72, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25030919

RESUMO

RNA interference (RNAi) screening is a powerful technique for understanding the molecular biology of cancer and searching drug targets. Genes and their upstream activators that are essential for the survival of cancer cells often dictate cancer formation/progression. Hence, they are preferable therapeutic targets. Identifying these genes using RNAi is, however, problematic because knocking them down leads to cell death. Here we describe a diphtheria toxin (DT) negative selection method to circumvent the problem of cell death in RNAi screening. DT fails to kill mouse cells due to the lack of functional DT receptor (DTR). Thus, we first prepare a construct encoding a human functional DTR driven by the promoter of mouse Atf5, a gene essential for the survival of malignant glioma. Then a DT-sensitive mouse malignant glioma cell line is established by over-expressing this DTR. Finally, an RNAi screen is performed in this cell line and genes that activate Atf5 expression are identified. The negative selection approach described here allows RNAi screening to be used for identifying genes controlling cell survival in cancers or perhaps other human diseases with potential in therapeutic intervention.


Assuntos
Toxina Diftérica/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Interferência de RNA , RNA Interferente Pequeno/genética , Fatores Ativadores da Transcrição/genética , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Relação Dose-Resposta a Droga , Biblioteca Gênica , Genes Reporter , Glioma/genética , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/genética , Humanos , Camundongos , Regiões Promotoras Genéticas
20.
J Am Heart Assoc ; 1(6): e002238, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23316315

RESUMO

BACKGROUND: This study tested the hypothesis that type 2 diabetes restricts multipotency of db/db mesenchymal stem cells (MSCs), promotes their terminal differentiation into adipocytes rather than endothelial cells, thereby promotes adipocytic infiltration into ischemic muscles, and reduces their capacity to participate in postischemic neovascularization. METHODS AND RESULTS: To test this hypothesis, we transplanted MSCs from db/db or wild-type (WT) mice into WT recipients after induction of hind limb ischemia. WT recipients of db/db MSCs demonstrated adipocyte infiltration of ischemic muscle and impaired neovascularization; WT recipients of WT MSCs showed no intramuscular adipocyte infiltration and had significantly enhanced neovascularization (P<0.05; n=6). Confocal microscopy showed that the percentage of MSCs that differentiated into an adipocyte phenotype was greater and into an endothelial cell was less in WT recipients transplanted with db/db MSCs than those transplanted with WT MSCs (P<0.05; n=6). In vitro, db/db MSCs exhibited greater oxidant stress, greater adipocyte differentiation, and less endothelial differentiation than WT MSCs, and these differences were reversed by treatment with N-acetylcysteine or Nox4 siRNA (P<0.05; n=6). Insulin increased Nox4 expression, oxidant stress, and adipocyte differentiation in WT MSCs, and these insulin-induced effects were reversed by Nox4 siRNA (P<0.05; n=6). Reversal of db/db MSC oxidant stress by in vivo pretreatment with Nox4 siRNA before transplantation reversed their impaired capacity to augment postischemic neovascularization. CONCLUSIONS: Type 2 diabetes-induced oxidant stress restricts the multipotency of MSCs and impairs their capacity to increase blood flow recovery after the induction of hind-limb ischemia. Reversal of MSC oxidant stress might permit greater leverage of the therapeutic potential of MSC transplantation in the setting of diabetes.


Assuntos
Adipócitos/fisiologia , Diabetes Mellitus Tipo 2/fisiopatologia , Isquemia/fisiopatologia , Células-Tronco Mesenquimais/fisiologia , Neovascularização Fisiológica/fisiologia , Animais , Diferenciação Celular , Movimento Celular , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Expressão Gênica , Membro Posterior/irrigação sanguínea , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Isquemia/etiologia , Masculino , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Células-Tronco Multipotentes/efeitos dos fármacos , Células-Tronco Multipotentes/fisiologia , NADPH Oxidase 4 , NADPH Oxidases/efeitos dos fármacos , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Estresse Oxidativo/efeitos dos fármacos , RNA Interferente Pequeno/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA