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.
Sci Data ; 10(1): 570, 2023 08 26.
Artigo em Inglês | MEDLINE | ID: mdl-37634014

RESUMO

Many studies have shown that cellular morphology can be used to distinguish spiked-in tumor cells in blood sample background. However, most validation experiments included only homogeneous cell lines and inadequately captured the broad morphological heterogeneity of cancer cells. Furthermore, normal, non-blood cells could be erroneously classified as cancer because their morphology differ from blood cells. Here, we constructed a dataset of microscopic images of organoid-derived cancer and normal cell with diverse morphology and developed a proof-of-concept deep learning model that can distinguish cancer cells from normal cells within an unlabeled microscopy image. In total, more than 75,000 organoid-drived cells from 3 cholangiocarcinoma patients were collected. The model achieved an area under the receiver operating characteristics curve (AUROC) of 0.78 and can generalize to cell images from an unseen patient. These resources serve as a foundation for an automated, robust platform for circulating tumor cell detection.


Assuntos
Linhagem Celular Tumoral , Neoplasias , Humanos , Área Sob a Curva , Aprendizado Profundo , Microscopia , Linhagem Celular Tumoral/classificação , Linhagem Celular Tumoral/patologia , Neoplasias/diagnóstico por imagem , Neoplasias/patologia
2.
Mol Cancer Res ; 21(3): 240-252, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36490322

RESUMO

Since its establishment in 2015, the transcriptomics-based consensus molecular subtype (CMS) classification has unified our understanding of colorectal cancer. Each of the four CMS exhibited distinctive high-level molecular signatures that correlated well with prognosis and treatment response. Nonetheless, many key aspects of colorectal cancer progression and intra-subtype heterogeneity remain unresolved. This is partly because the bulk transcriptomic data used to define CMS contain substantial interference from non-tumor cells. Here, we propose a concise panel of 62 genes that not only accurately recapitulates all key characteristics of the four original CMS but also identifies three additional subpopulations with unique molecular signatures. Validation on independent cohorts confirms that the new CMS4 intra-subtypes coincide with single-cell-derived intrinsic subtypes and that the panel consists of many immune cell-type markers that can capture the status of tumor microenvironment. Furthermore, a 2D embedding of CMS structure based on the proposed gene panel provides a high-resolution view of the functional pathways and cell-type markers that underlie each CMS intra-subtype and the continuous progression from CMS2 to CMS4 subtypes. Our gene panel and 2D visualization refined the delineation of colorectal cancer subtypes and could aid further discovery of molecular mechanisms in colorectal cancer. IMPLICATIONS: : Well-selected gene panel and representation can capture both the continuum of cancer cell states and tumor microenvironment status.


Assuntos
Neoplasias Colorretais , Humanos , Neoplasias Colorretais/patologia , Perfilação da Expressão Gênica , Transcriptoma , Biomarcadores Tumorais/genética , Microambiente Tumoral/genética
3.
Sci Rep ; 12(1): 18686, 2022 11 04.
Artigo em Inglês | MEDLINE | ID: mdl-36333384

RESUMO

Circulating tumor cells (CTCs) have been shown as a surrogate for cancer progression and prognostication. We aimed to determine an association between CTCs and survival of hepatocellular carcinoma (HCC) patients. Peripheral blood was obtained from 73 HCC patients to enumerate for epithelial CTCs/8 mL blood. CTCs were detected by immunoaffinity-based method using epithelial cell adhesion molecule (EpCAM) and mucin1 (MUC1). The CTCs detection rates of BCLC stages A, B, and C patients were 65.4% (17/26), 77.3% (17/22), and 96% (24/25), respectively, p = 0.018. Patients with CTCs < 5 cells/8 mL had significantly longer survival than those with CTCs ≥ 5 cells/8 mL (>36 vs. 4.6 months, p < 0.001). In multivariate analysis, CTP B, BCLC B, BCLC C, AFP ≥ 400 ng/mL, and CTC ≥ 5 cells/8 mL were independently associated with survival, with adjusted HRs (95%CI) of 4.1 (2.0-8.4), 3.5 (1.1-11.4), 4.7 (1.4-15.4), 2.4 (1.1-5.0), and 2.6 (1.2-8.4); p < 0.001, 0.036, 0.011, 0.025 and 0.012, respectively. The combination of CTCs ≥ 5 cells/8 mL and AFP ≥ 400 ng/mL provided additively increased HR to 5.3 (2.5-11.1), compared to HRs of 4.0 (2.0-8.0) and 3.5 (1.8-6.7) for CTCs ≥ 5 cells/8 mL and AFP ≥ 400 ng/mL, p < 0.001, respectively. The larger number of peripheral CTCs is correlated with higher tumor aggressive features and poorer survival of HCC patients. CTCs can potentially become novel prognostic biomarker in HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Células Neoplásicas Circulantes , Humanos , Carcinoma Hepatocelular/metabolismo , Células Neoplásicas Circulantes/patologia , Neoplasias Hepáticas/patologia , Prognóstico , alfa-Fetoproteínas , Molécula de Adesão da Célula Epitelial , Biomarcadores Tumorais/metabolismo
4.
PLoS One ; 17(2): e0263141, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35120168

RESUMO

Corneal grafts are the imperative clinical treatment for canine corneal blindness. To serve the growing demand, this study aimed to generate tissue-engineered canine cornea in part of the corneal epithelium and underlying stroma based on canine limbal epithelial stem cells (cLESCs) seeded silk fibroin/gelatin (SF/G) film and canine corneal stromal stem cells (cCSSCs) seeded SF/G scaffold, respectively. Both cell types were successfully isolated by collagenase I. SF/G corneal films and stromal scaffolds served as the prospective substrates for cLESCs and cCSSCs by promoting cell adhesion, cell viability, and cell proliferation. The results revealed the upregulation of tumor protein P63 (P63) and ATP-binding cassette super-family G member 2 (Abcg2) of cLESCs as well as Keratocan (Kera), Lumican (Lum), aldehyde dehydrogenase 3 family member A1 (Aldh3a1) and Aquaporin 1 (Aqp1) of differentiated keratocytes. Moreover, immunohistochemistry illustrated the positive staining of tumor protein P63 (P63), aldehyde dehydrogenase 3 family member A1 (Aldh3a1), lumican (Lum) and collagen I (Col-I), which are considerable for native cornea. This study manifested a feasible platform to construct tissue-engineered canine cornea for functional grafts and positively contributed to the body of knowledge related to canine corneal stem cells.


Assuntos
Materiais Biocompatíveis/química , Epitélio Corneano/patologia , Regeneração , Células-Tronco/citologia , Células Estromais/citologia , Células 3T3 , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Aquaporina 1/metabolismo , Proliferação de Células , Colágeno Tipo I/metabolismo , Transplante de Córnea , Cães , Proteínas do Olho/metabolismo , Fibroblastos/citologia , Fibroínas/química , Gelatina/química , Genes Supressores de Tumor , Imuno-Histoquímica , Técnicas In Vitro , Lumicana/metabolismo , Camundongos , Resistência à Tração , Engenharia Tecidual , Alicerces Teciduais
5.
Cancer Gene Ther ; 29(5): 445-455, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34548635

RESUMO

Cholangiocarcinoma (CCA), a lethal malignancy of the biliary epithelium, is the second most common primary liver cancer. The poor prognosis of CCA is due to the high rate of tumour invasion and distant metastasis. We found that the RNA-binding protein LIN28B, a known regulator of microRNA biogenesis, stem cell maintenance, and oncogenesis, is expressed in a subpopulation of CCA patients. To further investigate the potential role of LIN28B in CCA pathogenesis, we studied the effect of LIN28B overexpression in the cholangiocyte cell line MMNK-1 and cholangiocarcinoma cell lines HuCCT-1 and KKU-214. Here, we show that enhanced LIN28B expression promoted cancer stem cell-like properties in CCA, including enhanced cell migration, epithelial-to-mesenchymal transition (EMT), increased cell proliferation and spheroid formation. Proteomic analysis revealed TGF-ß-induced protein (TGFBI) as a novel LIN28B target gene, and further analysis showed upregulation of other components of the TGF-ß signalling pathway, including TGF-ß receptor type I (TGFBRI) expression and cytokine TGFB-I, II and III secretion. Importantly, the small molecule TGF-ß inhibitor SB431542 negated the effects of LIN28B on both cell migration and clonogenic potential. Overexpression of TGFBI alone promoted cholangiocarcinoma cell migration and EMT changes, but not spheroid formation, suggesting that TGFBI partially contributes to LIN28B-mediated aggressive cell behaviour. These observations are consistent with a model in which TGF-ß and LIN28B work together to form a positive feedback loop during cholangiocarcinoma metastasis and provide a therapeutic intervention opportunity.


Assuntos
Neoplasias dos Ductos Biliares , Colangiocarcinoma , Proteínas de Ligação a RNA , Fator de Crescimento Transformador beta , Neoplasias dos Ductos Biliares/genética , Neoplasias dos Ductos Biliares/metabolismo , Ductos Biliares Intra-Hepáticos/metabolismo , Ductos Biliares Intra-Hepáticos/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Transformação Celular Neoplásica , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Transição Epitelial-Mesenquimal/genética , Retroalimentação , Humanos , Proteômica , Proteínas de Ligação a RNA/genética , Fator de Crescimento Transformador beta/metabolismo
6.
Platelets ; 33(5): 700-708, 2022 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-34749590

RESUMO

Janus kinase 2 (JAK2) gene mutations are the main drivers for polycythemia vera (PV) and essential thrombocythemia (ET). The mechanisms of single altered gene causing two different diseases are unclear. Additionally, novel treatments specifically targeting mutated JAK2 proteins are needed. In this study, the induced pluripotent stem cells (iPSCs) were virally transduced to express wild-type JAK2 (JAK2WT), JAK2p.V617F (JAK2V617F) or JAK2p.N542_E543del (JAK2exon12) under a doxycycline-inducible system. The modified iPSCs which were differentiated into megakaryocytes in the presence vs. absence of doxycycline were compared to ensure that the differences were solely from mutated JAK2 expressions. The JAK2V617-expressing iPSCs yielded significantly higher numbers of megakaryocytes consistent with the ET phenotype, while there was no enhancement by JAK2exon12 expression compatible with the pure erythrocytosis in humans. Capillary Western analyses revealed significantly greater JAK2 phosphorylation in iPSCs carrying JAK2V617F but not in JAK2WT and JAK2exon12 iPSCs. Activation of STAT3, STAT5 and AKT was increased by JAK2V617F, while they were decreased in JAK2exon12 iPSCs. Notably, interferon alpha and/or arsenic trioxide inhibited megakaryocytes proliferation and reduced JAK2, STAT3, STAT5 and AKT phosphorylation in mutant JAK2-expressing iPSCs compared with those without induction. In conclusion, JAK2V617F expression in iPSCs preferentially promoted megakaryocytes with a signaling profile distinctive from JAK2exon12 expression. Treatments with interferon alpha or arsenic trioxide preferentially suppressed the mutated over wild-type JAK2 signaling. This iPSC model is helpful in mechanistic studies and novel therapy screen for myeloproliferative neoplasm.


Assuntos
Células-Tronco Pluripotentes Induzidas , Janus Quinase 2 , Transdução de Sinais , Trombocitemia Essencial , Trióxido de Arsênio/farmacologia , Doxiciclina , Humanos , Interferon-alfa/farmacologia , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Megacariócitos/metabolismo , Mutação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Trombocitemia Essencial/genética
7.
Sci Rep ; 11(1): 5255, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33664283

RESUMO

Activating mutations affecting the JAK-STAT signal transduction is the genetic driver of myeloproliferative neoplasms (MPNs) which comprise polycythemia vera (PV), essential thrombocythemia (ET) and myelofibrosis. The JAK2p.V617F mutation can produce both erythrocytosis in PV and thrombocytosis in ET, while JAK2 exon 12 mutations cause only erythrocytosis. We hypothesized that these two mutations activated different intracellular signals. In this study, the induced pluripotent stem cells (iPSCs) were used to model JAK2-mutated MPNs. Normal iPSCs underwent lentiviral transduction to overexpress JAK2p.V617F or JAK2p.N542_E543del (JAK2exon12) under a doxycycline-inducible system. The modified iPSCs were differentiated into erythroid cells. Compared with JAK2V617F-iPSCs, JAK2exon12-iPSCs yielded more total CD71+GlycophorinA+ erythroid cells, displayed more mature morphology and expressed more adult hemoglobin after doxycycline induction. Capillary Western immunoassay revealed significantly higher phospho-STAT1 but lower phospho-STAT3 and lower Phospho-AKT in JAK2exon12-iPSCs compared with those of JAK2V617F-iPSCs in response to erythropoietin. Furthermore, interferon alpha and arsenic trioxide were tested on these modified iPSCs to explore their potentials for MPN therapy. Both agents preferentially inhibited proliferation and promoted apoptosis of the iPSCs expressing mutant JAK2 compared with those without doxycycline induction. In conclusion, the modified iPSC model can be used to investigate the mechanisms and search for new therapy of MPNs.


Assuntos
Janus Quinase 2/genética , Policitemia Vera/genética , Policitemia/genética , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT3/genética , Eritropoese/genética , Éxons , Regulação da Expressão Gênica/genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Mutação/genética , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Policitemia/patologia , Policitemia Vera/patologia , Mielofibrose Primária , Fatores de Transcrição STAT/genética , Trombocitemia Essencial/genética , Trombocitemia Essencial/patologia , Trombocitose/genética , Trombocitose/patologia
8.
Angew Chem Int Ed Engl ; 60(8): 3934-3939, 2021 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-33063327

RESUMO

Polypeptides generated from proteolytic processing of protein precursors, or proteolytic proteoforms, play an important role in diverse biological functions and diseases. However, their often-small size and intricate post-translational biogenesis preclude the use of simple genetic tagging in their cellular studies. Herein, we develop a labeling strategy for this class of proteoforms, based on residue-specific genetic code expansion labeling with a molecular beacon design. We demonstrate the utility of such a design by creating a molecular beacon reporter to detect amyloid-ß peptides, known to be involved in the pathogenesis of Alzheimer's disease, as they are produced from amyloid precursor protein (APP) along the endocytic pathway of living cells.


Assuntos
Peptídeos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Lisina/análogos & derivados , Aminoacil-tRNA Sintetases/genética , Aminoacil-tRNA Sintetases/metabolismo , Peptídeos beta-Amiloides/química , Precursor de Proteína beta-Amiloide/genética , Proteínas Arqueais/genética , Proteínas Arqueais/metabolismo , Código Genético , Células HEK293 , Humanos , Lisina/química , Lisina/metabolismo , Methanosarcina/enzimologia , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Processamento de Proteína Pós-Traducional
9.
Angew Chem Weinheim Bergstr Ger ; 133(8): 3980-3985, 2021 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-38504667

RESUMO

Polypeptides generated from proteolytic processing of protein precursors, or proteolytic proteoforms, play an important role in diverse biological functions and diseases. However, their often-small size and intricate post-translational biogenesis preclude the use of simple genetic tagging in their cellular studies. Herein, we develop a labeling strategy for this class of proteoforms, based on residue-specific genetic code expansion labeling with a molecular beacon design. We demonstrate the utility of such a design by creating a molecular beacon reporter to detect amyloid-ß peptides, known to be involved in the pathogenesis of Alzheimer's disease, as they are produced from amyloid precursor protein (APP) along the endocytic pathway of living cells.

10.
Sci Rep ; 10(1): 8472, 2020 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-32439978

RESUMO

Platelet demand has increased around the world. However, the inadequacy of donors, the risk of transfusion-transmitted infections and associated reactions, and the refractory nature of platelet transfusions are among the limitations of allogeneic platelet transfusions. To alleviate these problems, we propose generating platelets in a laboratory that do not induce alloimmunity to human leukocyte antigen (HLA) class I, which is a major cause of immune reaction in platelet transfusion refractoriness. Induced pluripotent stem cells (iPSCs) were generated from peripheral blood mononuclear cells (PBMCs) of a healthy Thai woman. We then knocked out the ß2-microglobulin (ß2m) gene in the cells using paired CRISPR/Cas9 nickases and sequentially differentiated the cells into haematopoietic stem cells (HSCs), megakaryocytes (MKs) and platelets. Silencing of HLA class I expression was observed on the cell surface of ß2m-knockout iPSCs, iPSC-derived HSCs, MKs and platelets. The HLA-universal iPSC-derived platelets were shown to be activated, and they aggregated after stimulation. In addition, our in vivo platelet survival experiments demonstrated that human platelets were detectable at 2 and 24 hours after injecting the ß2m-KO MKs. In summary, we successfully generated functional iPSC-derived platelets in vitro without HLA class I expression by knocking out the ß2m gene using paired CRISPR/Cas9 nickases.


Assuntos
Plaquetas/citologia , Células-Tronco Hematopoéticas/citologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Leucócitos Mononucleares/citologia , Megacariócitos/citologia , Animais , Plaquetas/metabolismo , Diferenciação Celular , Feminino , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/metabolismo , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
11.
Exp Anim ; 68(1): 35-47, 2019 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-30089733

RESUMO

Stem cells are promising cell source for treatment of multiple diseases as well as myocardial infarction. Rabbit model has essentially used for cardiovascular diseases and regeneration but information on establishment of induced pluripotent stem cells (iPSCs) and differentiation potential is fairly limited. In addition, there is no report of cardiac differentiation from iPSCs in the rabbit model. In this study, we generated rabbit iPSCs by reprogramming rabbit fibroblasts using the 4 transcription factors (OCT3/4, SOX2, KLF4, and c-Myc). Three iPSC lines were established. The iPSCs from all cell lines expressed genes (OCT3/4, SOX2, KLF4 and NANOG) and proteins (alkaline phosphatase, OCT-3/4 and SSEA-4) essentially described for pluripotency (in vivo and in vitro differentiation). Furthermore, they also had ability to form embryoid body (EB) resulting in three-germ layer differentiation. However, ability of particular cell lines and cell numbers at seeding markedly influenced on EB formation and also their diameters. The cell density at 20,000 cells per EB was selected for cardiac differentiation. After plating, the EBs attached and cardiac-like beating areas were seen as soon as 11 days of culture. The differentiated cells expressed cardiac progenitor marker FLK1 (51 ± 1.48%) on day 5 and cardiac troponin-T protein (10.29 ± 1.37%) on day 14. Other cardiac marker genes (cardiac ryanodine receptors (RYR2), α-actinin and PECAM1) were also expressed. This study concluded that rabbit iPSCs remained their in vitro pluripotency with capability of differentiation into mature-phenotype cardiomyocytes. However, the efficiency of cardiac differentiation is still restricted.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Miócitos Cardíacos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Coelhos , Fosfatase Alcalina/fisiologia , Animais , Linhagem Celular , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/fisiologia , Proteína Homeobox Nanog/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Antígenos Embrionários Estágio-Específicos/fisiologia
12.
Biosci Rep ; 38(5)2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30068696

RESUMO

Lentiviral vectors have emerged as the most efficient system to stably transfer and insert genes into cells. By adding a tetracycline (Tet)-inducible promoter, transgene expression delivered by a lentiviral vector can be expressed whenever needed and halted when necessary. Here we have constructed a doxycycline (Dox)-inducible lentiviral vector which efficiently introduces a designed zinc finger protein, 2-long terminal repeat zinc-finger protein (2LTRZFP), into hematopoietic cell lines and evaluated its expression in pluripotent stem cells. As a result this lentiviral inducible system can regulate 2LTRZFP expression in the SupT1 T-cell line and in pluripotent stem cells. Using this vector, no basal expression was detected in the T-cell line and its induction was achieved with low Dox concentrations. Remarkably, the intracellular regulatory expression of 2LTRZFP significantly inhibited HIV-1 integration and replication in HIV-inoculated SupT1 cells. This approach could provide a potential tool for gene therapy applications, which efficiently control and reduce the side effect of therapeutic genes expression.


Assuntos
Terapia Genética/métodos , Vetores Genéticos , Repetição Terminal Longa de HIV/genética , HIV-1/genética , Integração Viral/fisiologia , Relação Dose-Resposta a Droga , Doxiciclina/administração & dosagem , Doxiciclina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Infecções por HIV/genética , Repetição Terminal Longa de HIV/efeitos dos fármacos , HIV-1/patogenicidade , Humanos , Lentivirus/genética , Células-Tronco Pluripotentes/virologia , Tetraciclina/farmacologia , Transgenes , Integração Viral/efeitos dos fármacos , Integração Viral/genética , Dedos de Zinco
13.
J Neurogastroenterol Motil ; 22(3): 367-82, 2016 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-27184041

RESUMO

Irritable bowel syndrome is one of the most common functional gastrointestinal (GI) disorders that significantly impair quality of life in patients. Current available treatments are still not effective and the pathophysiology of this condition remains unclearly defined. Recently, research on intestinal stem cells has greatly advanced our understanding of various GI disorders. Alterations in conserved stem cell regulatory pathways such as Notch, Wnt, and bone morphogenic protein/TGF- ß have been well documented in diseases such as inflammatory bowel diseases and cancer. Interaction between intestinal stem cells and various signals from their environment is important for the control of stem cell self-renewal, regulation of number and function of specific intestinal cell types, and maintenance of the mucosal barrier. Besides their roles in stem cell regulation, these signals are also known to have potent effects on immune cells, enteric nervous system and secretory cells in the gut, and may be responsible for various aspects of pathogenesis of functional GI disorders, including visceral hypersensitivity, altered gut motility and low grade gut inflammation. In this article, we briefly summarize the components of these signaling pathways, how they can be modified by extrinsic factors and novel treatments, and provide evidenced support of their roles in the inflammation processes. Furthermore, we propose how changes in these signals may contribute to the symptom development and pathogenesis of irritable bowel syndrome.

14.
Exp Biol Med (Maywood) ; 240(12): 1640-7, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26080460

RESUMO

We investigate antitumor efficacy and 2D and 3D intratumoral distribution of 7-ethyl-10-hydroxycamptothecin (SN-38) from polymeric depots inside U-87MG xenograft tumor model in nude mice. Results showed that polymeric depots could be used to administer and controlled release of a large amount of SN-38 directly to the brain tumor model. SN-38 released from depots suppressed tumor growth, where the extent of suppression greatly depended on doses and the number of depot injections. Tumor suppression of SN-38 from depots was three-fold higher in animals which received double injections of depots at high dose (9.7 mg of SN-38) compared to single injection (2.2 mg). H&E staining of tumor sections showed that the area of tumor cell death/survival of the former group was two-fold higher than those of the latter group. Fluorescence imaging based on self-fluorescent property of SN-38 was used to evaluate the intratumoral distribution of this drug compared to histological results. The linear correlation between fluorescence intensity and the amount of SN-38 allowed quantitative determination of SN-38 in tumor tissues. Results clearly showed direct correlation between the amount of SN-38 in tumor sections and cancer cell death. Moreover, 3D reconstruction representing the distribution of SN-38 in tumors was obtained. Results from this study suggest the rationale for intratumoral drug administration and release of drugs inside tumor, which is necessary to design drug delivery systems with efficient antitumor activity.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Camptotecina/análogos & derivados , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/análise , Antineoplásicos Fitogênicos/farmacocinética , Neoplasias Encefálicas/química , Camptotecina/administração & dosagem , Camptotecina/análise , Camptotecina/farmacocinética , Camptotecina/uso terapêutico , Preparações de Ação Retardada , Relação Dose-Resposta a Droga , Sistemas de Liberação de Medicamentos , Feminino , Irinotecano , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Thromb Haemost ; 113(4): 792-805, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25518736

RESUMO

Wiskott-Aldrich syndrome (WAS) is an X-linked recessive disorder characterised by microthrombocytopenia, complex immunodeficiency, autoimmunity, and haematologic malignancies. It is caused by mutations in the gene encoding WAS protein (WASP), a regulator of actin cytoskeleton and chromatin structure in various blood cell lineages. The molecular mechanisms underlying microthrombocytopenia caused by WASP mutations remain elusive. Murine models of WASP deficiency exhibited only mild thrombocytopenia with normal-sized platelets. Here we report on the successful generation of induced pluripotent stem cell (iPSC) lines from two patients with different mutations in WASP (c.1507T>A and c.55C>T). When differentiated into early CD34+ haematopoietic and megakaryocyte progenitors, the WAS-iPSC lines were indistinguishable from the wild-type iPSCs. However, all WAS-iPSC lines exhibited defects in platelet productionin vitro. WAS-iPSCs produced platelets with more irregular shapes and smaller sizes. Immunofluorescence and electron micrograph showed defects in cytoskeletal rearrangement, F-actin distribution, and proplatelet formation. Proplatelet defects were more pronounced when using culture systems with stromal feeders comparing to feeder-free culture condition. Overexpression of WASP in the WAS-iPSCs using a lentiviral vector improved proplatelet structures and increased the platelet size. Our findings substantiate the use of iPSC technology to elucidate the disease mechanisms of WAS in thrombopoiesis.


Assuntos
Plaquetas/metabolismo , Citoesqueleto/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Células Progenitoras de Megacariócitos/metabolismo , Megacariócitos/metabolismo , Trombopoese , Síndrome de Wiskott-Aldrich/metabolismo , Actinas/metabolismo , Antígenos CD34/metabolismo , Plaquetas/ultraestrutura , Linhagem da Célula , Forma Celular , Tamanho Celular , Técnicas de Cocultura , Citoesqueleto/ultraestrutura , Células Alimentadoras , Predisposição Genética para Doença , Humanos , Células-Tronco Pluripotentes Induzidas/ultraestrutura , Células Progenitoras de Megacariócitos/ultraestrutura , Megacariócitos/ultraestrutura , Mutação , Fenótipo , Trombopoese/genética , Transfecção , Síndrome de Wiskott-Aldrich/sangue , Síndrome de Wiskott-Aldrich/diagnóstico , Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo
16.
J Biomater Sci Polym Ed ; 25(12): 1240-52, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24933469

RESUMO

Neuronal activities play critical roles in both neurogenesis and neural regeneration. In that sense, electrically conductive and biocompatible biomaterial scaffolds can be applied in various applications of neural tissue engineering. In this study, we fabricated a novel biomaterial for neural tissue engineering applications by coating electrospun poly(lactic acid) (PLA) nanofibers with a conducting polymer, polypyrole (PPy), via admicellar polymerization. Optimal conditions for polymerization and preparation of PPy-coated electrospun PLA nanofibers were obtained by comparing results from scanning electron microscopy, X-ray photoelectron spectrometer, and surface conductivity tests. In vitro cell culture experiments showed that PPy-coated electrospun PLA fibrous scaffold is not toxic. The scaffold could support attachment and migration of neural progenitor cells. Neurons derived from progenitor exhibited long neurite outgrowth under electrical stimulation. Our study concluded that PPy-coated electrospun PLA fibers had a good biocompatibility with neural progenitor cells and may serve as a promising material for controlling progenitor cell behaviors and enhancing neural repair.


Assuntos
Materiais Biocompatíveis/farmacologia , Condutividade Elétrica , Ácido Láctico/química , Neurônios/citologia , Neurônios/efeitos dos fármacos , Polímeros/química , Pirróis/química , Alicerces Teciduais/química , Animais , Materiais Biocompatíveis/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Hipocampo/citologia , Nanofibras/química , Nanotecnologia/métodos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Poliésteres , Ratos , Propriedades de Superfície , Engenharia Tecidual
17.
Dev Biol ; 268(1): 220-31, 2004 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15031118

RESUMO

Neural stem cells proliferate and maintain multipotency when cultured in the presence of FGF2, but subsequent lineage commitment by the cells is nevertheless influenced by the exposure to FGF2. Here we show that FGF2 effects on neural stem cells are mediated, in part, by beta-catenin. Conversely, the effects of beta-catenin in neural stem cells depend in part upon whether there is concurrent fibroblast growth factor (FGF) signaling. FGF2 increases beta-catenin signaling through several different mechanisms including increased expression of beta-catenin mRNA, increased nuclear translocation of beta-catenin, increased phosphorylation of GSK-3beta, and tyrosine phosphorylation of beta-catenin. Overexpression of beta-catenin in the presence of FGF2 helps to maintain neural progenitor cells in a proliferative state. However, overexpression of beta-catenin in the absence of FGF2 enhances neuronal differentiation. Further, chromatin immunoprecipitation (ChIP) assays demonstrate that both beta-catenin and Lef1 bind directly to the neurogenin promoter, and luciferase reporter assays demonstrate that beta-catenin is directly involved in the regulation of neurogenin 1 and possibly other proneural genes when neural stem cells are cultured in the presence of FGF2. We suggest that the balance between the mitogenic effects and the proneural effects of beta-catenin is determined by the presence of FGF signaling.


Assuntos
Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Sistema Nervoso Central/embriologia , Proteínas do Citoesqueleto/fisiologia , Fator 2 de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Transativadores/fisiologia , Animais , Western Blotting , Linhagem Celular Tumoral , Sistema Nervoso Central/citologia , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Feminino , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Imuno-Histoquímica , Camundongos , Fosforilação , Testes de Precipitina , Gravidez , Transporte Proteico , RNA Mensageiro/genética , Transativadores/genética , Transativadores/metabolismo , beta Catenina
18.
J Neurosci Res ; 69(6): 803-9, 2002 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12205674

RESUMO

Treatment of cultured ventricular zone (VZ) progenitor cells with bone morphogenetic protein-4 (BMP4) promoted cell death in a dose-dependent manner. VZ progenitor cells became progressively more resistant to the proapoptotic effects of BMP4 between E10 and E16, and, by E18 and thereafter, BMP4 treatment no longer led to progenitor cell death. BMP4 treatment of E13 progenitor cells promoted expression of msx2 and p21(CIP1/WAF1) (p21) and inhibition of expression of either gene prevented BMP4-mediated apoptosis. Treatment of E18 cells with BMP4 failed to induce apoptosis but still induced expression of low levels of msx2 and p21. Knockout of bax significantly reduced but did not prevent BMP4-mediated death of E13 murine progenitor cells. These observations indicate that msx2 and p21 mediate the proapoptotic effects of BMP4 on VZ progenitor cells and that each gene is necessary but insufficient to promote apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/farmacologia , Ciclinas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neurônios/citologia , Proteínas Proto-Oncogênicas c-bcl-2 , Células-Tronco/citologia , Animais , Proteína Morfogenética Óssea 4 , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ventrículos Cerebrais/citologia , Ventrículos Cerebrais/embriologia , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/genética , Proteínas de Ligação a DNA/genética , Relação Dose-Resposta a Droga , Feminino , Expressão Gênica/fisiologia , Proteínas de Homeodomínio , Gravidez , Proteínas Proto-Oncogênicas/fisiologia , Ratos , Ratos Sprague-Dawley , Células-Tronco/efeitos dos fármacos , Proteína X Associada a bcl-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA