Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Sci Rep ; 13(1): 18283, 2023 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-37880340

RESUMO

Tissue engineering is a promising alternative to current full thickness circumferential esophageal replacement methods. The aim of our study was to develop a clinical grade Decellularized Human Esophagus (DHE) for future clinical applications. After decontamination, human esophagi from deceased donors were placed in a bioreactor and decellularized with sodium dodecyl sulfate (SDS) and ethylendiaminetetraacetic acid (EDTA) for 3 days. The esophagi were then rinsed in sterile water and SDS was eliminated by filtration on an activated charcoal cartridge for 3 days. DNA was removed by a 3-hour incubation with DNase. A cryopreservation protocol was evaluated at the end of the process to create a DHE cryobank. The decellularization was efficient as no cells and nuclei were observed in the DHE. Sterility of the esophagi was obtained at the end of the process. The general structure of the DHE was preserved according to immunohistochemical and scanning electron microscopy images. SDS was efficiently removed, confirmed by a colorimetric dosage, lack of cytotoxicity on Balb/3T3 cells and mesenchymal stromal cell long term culture. Furthermore, DHE did not induce lymphocyte proliferation in-vitro. The cryopreservation protocol was safe and did not affect the tissue, preserving the biomechanical properties of the DHE. Our decellularization protocol allowed to develop the first clinical grade human decellularized and cryopreserved esophagus.


Assuntos
Matriz Extracelular , Alicerces Teciduais , Camundongos , Animais , Humanos , Alicerces Teciduais/química , Engenharia Tecidual/métodos , Criopreservação , Dodecilsulfato de Sódio/química , Esôfago
2.
Surgery ; 171(2): 384-392, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34392978

RESUMO

BACKGROUND: Tissue engineering is an attractive alternative to conventional esophageal replacement techniques using intra-abdominal organs which are associated with a substantial morbidity. The objective was to evaluate the feasibility of esophageal replacement by an allogenic decellularized esophagus in a porcine model. Secondary objectives were to evaluate the benefit of decellularized esophagus recellularization with autologous bone marrow mesenchymal stromal cells and omental maturation of the decellularized esophagus. METHODS: Eighteen pigs divided into 4 experimental groups according to mesenchymal stromal cells recellularization and omental maturation underwent a 5-cm long circumferential replacement of the thoracic esophagus. Turbo green florescent protein labelling was used for in vivo mesenchymal stromal cells tracking. The graft area was covered by a stent for 3 months. Clinical and histologic outcomes were analyzed over a 6-month period. RESULTS: The median follow-up was 112 days [5; 205]. Two animals died during the first postoperative month, 2 experienced an anastomotic leakage, 13 experienced a graft area stenosis following stent migration of which 3 were sacrificed as initially planned after successful endoscopic treatment. The stent could be removed in 2 animals: the graft area showed a continuous mucosa without stenosis. After 3 months, the graft area showed a tissue specific regeneration with a mature epithelium and muscular cells. Clinical and histologic results were similar across experimental groups. CONCLUSION: Circumferential esophageal replacement by a decellularized esophagus was feasible and allowed tissue remodeling toward an esophageal phenotype. We could not demonstrate any benefit provided by the omental maturation of the decellularized esophagus nor its recellularization with mesenchymal stromal cells.


Assuntos
Esôfago/anatomia & histologia , Esôfago/cirurgia , Engenharia Tecidual/métodos , Alicerces Teciduais , Animais , Estudos de Viabilidade , Feminino , Transplante de Células-Tronco Mesenquimais , Modelos Animais , Omento/citologia , Stents , Suínos , Transplante Autólogo
3.
Int J Mol Sci ; 22(19)2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34638998

RESUMO

During transformation, myelodysplastic syndromes (MDS) are characterized by reducing apoptosis of bone marrow (BM) precursors. Mouse models of high risk (HR)-MDS and acute myelogenous leukemia (AML) post-MDS using mutant NRAS and overexpression of human BCL-2, known to be poor prognostic indicators of the human diseases, were created. We have reported the efficacy of the BCL-2 inhibitor, ABT-737, on the AML post-MDS model; here, we report that this BCL-2 inhibitor also significantly extended survival of the HR-MDS mouse model, with reductions of BM blasts and lineage negative/Sca1+/KIT+ (LSK) cells. Secondary transplants showed increased survival in treated compared to untreated mice. Unlike the AML model, BCL-2 expression and RAS activity decreased following treatment and the RAS:BCL-2 complex remained in the plasma membrane. Exon-specific gene expression profiling (GEP) of HR-MDS mice showed 1952 differentially regulated genes upon treatment, including genes important for the regulation of stem cells, differentiation, proliferation, oxidative phosphorylation, mitochondrial function, and apoptosis; relevant in human disease. Spliceosome genes, found to be abnormal in MDS patients and downregulated in our HR-MDS model, such as Rsrc1 and Wbp4, were upregulated by the treatment, as were genes involved in epigenetic regulation, such as DNMT3A and B, upregulated upon disease progression and downregulated upon treatment.


Assuntos
Compostos de Bifenilo/administração & dosagem , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/metabolismo , Nitrofenóis/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/metabolismo , Sulfonamidas/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Medula Óssea/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Perfilação da Expressão Gênica/métodos , Estimativa de Kaplan-Meier , Camundongos , Camundongos Transgênicos , Proteínas Monoméricas de Ligação ao GTP/genética , Síndromes Mielodisplásicas/mortalidade , Piperazinas/administração & dosagem , Proteínas Proto-Oncogênicas c-bcl-2/genética , Células-Tronco/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos
5.
Sci Rep ; 11(1): 8355, 2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33863936

RESUMO

In recent years, 3D cell culture models such as spheroid or organoid technologies have known important developments. Many studies have shown that 3D cultures exhibit better biomimetic properties compared to 2D cultures. These properties are important for in-vitro modeling systems, as well as for in-vivo cell therapies and tissue engineering approaches. A reliable use of 3D cellular models still requires standardized protocols with well-controlled and reproducible parameters. To address this challenge, a robust and scaffold-free approach is proposed, which relies on multi-trap acoustic levitation. This technology is successfully applied to Mesenchymal Stem Cells (MSCs) maintained in acoustic levitation over a 24-h period. During the culture, MSCs spontaneously self-organized from cell sheets to cell spheroids with a characteristic time of about 10 h. Each acoustofluidic chip could contain up to 30 spheroids in acoustic levitation and four chips could be ran in parallel, leading to the production of 120 spheroids per experiment. Various biological characterizations showed that the cells inside the spheroids were viable, maintained the expression of their cell surface markers and had a higher differentiation capacity compared to standard 2D culture conditions. These results open the path to long-time cell culture in acoustic levitation of cell sheets or spheroids for any type of cells.


Assuntos
Acústica , Técnicas de Cultura de Células/métodos , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Esferoides Celulares , Engenharia Tecidual/métodos , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Fluoresceínas , Humanos , Propídio , Fatores de Tempo
6.
Blood ; 137(22): 3050-3063, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-33512437

RESUMO

The extrafollicular immune response is essential to generate a rapid but transient wave of protective antibodies during infection. Despite its importance, the molecular mechanisms controlling this first response are poorly understood. Here, we demonstrate that enhanced Cxcr4 signaling caused by defective receptor desensitization leads to exacerbated extrafollicular B-cell response. Using a mouse model bearing a gain-of-function mutation of Cxcr4 described in 2 human hematologic disorders, warts, hypogammaglobulinemia, infections, and myelokathexis (WHIM) syndrome and Waldenström macroglobulinemia, we demonstrated that mutant B cells exhibited enhanced mechanistic target of rapamycin signaling, cycled more, and differentiated more potently into plasma cells than wild-type B cells after Toll-like receptor (TLR) stimulation. Moreover, Cxcr4 gain of function promoted enhanced homing and persistence of immature plasma cells in the bone marrow, a phenomenon recapitulated in WHIM syndrome patient samples. This translated in increased and more sustained production of antibodies after T-independent immunization in Cxcr4 mutant mice. Thus, our results establish that fine-tuning of Cxcr4 signaling is essential to limit the strength and length of the extrafollicular immune response.


Assuntos
Mutação com Ganho de Função , Doenças Hematológicas/imunologia , Plasmócitos/imunologia , Receptores CXCR4/imunologia , Transdução de Sinais/imunologia , Animais , Doenças Hematológicas/genética , Humanos , Camundongos , Camundongos Transgênicos , Receptores CXCR4/genética , Transdução de Sinais/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/imunologia
7.
Front Immunol ; 11: 579776, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33329550

RESUMO

Mechanisms driving acute graft-versus-host disease (aGVHD) onset in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) are still poorly understood. To provide a detailed characterization of tissue-infiltrating T lymphocytes (TL) and search for eventual site-specific specificities, we developed a xenogeneic model of aGVHD in immunodeficient mice. Phenotypic characterization of xenoreactive T lymphocytes (TL) in diseased mice disclosed a massive infiltration of GVHD target organs by an original CD4+CD8+ TL subset. Immunophenotypic and transcriptional profiling shows that CD4+CD8+ TL comprise a major PD1+CD62L-/+ transitional memory subset (>60%) characterized by low level expression of cytotoxicity-related transcripts. CD4+CD8+ TL produce high IL-10 and IL-13 levels, and low IL-2 and IFN-γ, suggestive of regulatory function. In vivo tracking of genetically labeled CD4+ or CD8+ TL subsequently found that CD4+CD8+ TL mainly originate from chronically activated cytotoxic TL (CTL). On the other hand, phenotypic profiling of CD3+ TL from blood, duodenum or rectal mucosa in a cohort of allo-HSCT patients failed to disclose abnormal expansion of CD4+CD8+ TL independent of aGVHD development. Collectively, our results show that acquisition of surface CD4 by xenoreactive CD8+ CTL is associated with functional diversion toward a regulatory phenotype, but rule out a central role of this subset in the pathogenesis of aGVHD in allo-HSCT patients.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Doença Enxerto-Hospedeiro/imunologia , Transplante de Células-Tronco Hematopoéticas , Linfócitos T Citotóxicos/imunologia , Animais , Citocinas/metabolismo , Feminino , Humanos , Memória Imunológica , Masculino , Camundongos , Camundongos SCID , Receptor de Morte Celular Programada 1/metabolismo , Transplante Heterólogo
8.
Acta Biomater ; 116: 186-200, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32911108

RESUMO

While human bone morphogenetic protein-2 (BMP-2) is a promising growth factor for bone regeneration, a major challenge in biomedical applications is finding an optimal carrier for its delivery at the site of injury. Because of their natural affinities for growth factors (including BMP-2) as well as their role in instructing cell function, cultured cell-derived extracellular matrices (ECM) are of special interest. We hereby hypothesized that a "bony matrix" containing mineralized, osteogenic ECM is a potential efficacious carrier of BMP-2 for promoting bone formation and, therefore, compared the efficacy of the decellularized ECM derived from osteogenic-differentiated human mesenchymal stem cells (hMSCs) to the one obtained from ECM from undifferentiated hMSCs. Our results provided evidence that both ECMs can bind BMP-2 and promote bone formation when implanted ectopically in mice. The osteoinductive potential of BMP-2, however, was greater when loaded within an osteogenic MSC-derived ECM; this outcome was correlated with higher sequestration capacity of BMP-2 over time in vivo. Interestingly, although the BMP-2 mainly bound onto the mineral crystals contained within the osteogenic MSC derived-ECM, these mineral components were not involved in the observed higher osteoinductivity, suggesting that the organic components were the critical components for the matrix efficacy as BMP-2 carrier.


Assuntos
Células-Tronco Mesenquimais , Animais , Proteína Morfogenética Óssea 2 , Regeneração Óssea , Diferenciação Celular , Células Cultivadas , Matriz Extracelular , Camundongos , Osteogênese
9.
J Tissue Eng Regen Med ; 13(12): 2191-2203, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31670903

RESUMO

In pathologies of the esophagus such as esophageal atresia, cancers, and caustic injuries, methods for full thickness esophageal replacement require the sacrifice of healthy intra-abdominal organs such as the stomach and the colon and are associated with high morbidity, mortality, and poor functional results. To overcome these problems, tissue engineering methods are developed to create a substitute with scaffolds and cells. The aim of this study was to develop a simple and safe decellularization process in order to obtain a clinical grade esophageal extracellular matrix. Following the decontamination step, porcine esophagi were decellularized in a bioreactor with sodium dodecyl sulfate and ethylenediaminetetraacetic acid for 3 days and were rinsed with deionized water. DNA was eliminated by a 3-hr DNase treatment. To remove any residual detergent, the matrix was then incubated with an absorbing resin. The resulting porcine esophageal matrix was characterized by the assessment of the efficiency of the decellularization process (DNA quantification), evaluation of sterility and absence of cytotoxicity, and its composition and biomechanical properties, as well as the possibility to be reseeded with mesenchymal stem cells. Complete decellularization with the preservation of the general structure, composition, and biomechanical properties of the native esophageal matrix was obtained. Sterility was maintained throughout the process, and the matrix showed no cytotoxicity. The resulting matrix met clinical grade criteria and was successfully reseeded with mesenchymal stem cells..


Assuntos
Esôfago/química , Matriz Extracelular/química , Teste de Materiais , Células-Tronco Mesenquimais/metabolismo , Engenharia Tecidual , Alicerces Teciduais/química , Animais , Células-Tronco Mesenquimais/citologia , Suínos
10.
Proc Natl Acad Sci U S A ; 116(13): 6298-6307, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30846549

RESUMO

Natural regulatory T cells (nTregs) ensure the control of self-tolerance and are currently used in clinical trials to alleviate autoimmune diseases and graft-versus-host disease after hematopoietic stem cell transfer. Based on CD39/CD26 markers, blood nTreg analysis revealed the presence of five different cell subsets, each representing a distinct stage of maturation. Ex vivo added microenvironmental factors, including IL-2, TGFß, and PGE2, direct the conversion from naive precursor to immature memory and finally from immature to mature memory cells, the latest being a no-return stage. Phenotypic and genetic characteristics of the subsets illustrate the structural parental maturation between subsets, which further correlates with the expression of regulatory factors. Regarding nTreg functional plasticity, both maturation stage and microenvironmental cytokines condition nTreg activities, which include blockade of autoreactive immune cells by cell-cell contact, Th17 and IL-10 Tr1-like activities, or activation of TCR-stimulating dendritic cell tolerization. Importantly, blood nTreg CD39/CD26 profile remained constant over a 2-y period in healthy persons but varied from person to person. Preliminary data on patients with autoimmune diseases or acute myelogenous leukemia illustrate the potential use of the nTreg CD39/CD26 profile as a blood biomarker to monitor chronic inflammatory diseases. Finally, we confirmed that naive conventional CD4 T cells, TCR-stimulated under a tolerogenic conditioned medium, could be ex vivo reprogrammed to FOXP3 lineage Tregs, and further found that these cells were exclusively committed to suppressive function under all microenvironmental contexts.


Assuntos
Microambiente Celular/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/fisiologia , Apirase/sangue , Doenças Autoimunes/sangue , Doenças Autoimunes/imunologia , Linfócitos T CD4-Positivos/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Dinoprostona/metabolismo , Dipeptidil Peptidase 4/sangue , Fatores de Transcrição Forkhead/metabolismo , Humanos , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Leucemia Mieloide , Células Th17/imunologia , Fator de Crescimento Transformador beta/metabolismo
11.
Cancer Biol Ther ; 18(8): 560-570, 2017 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-28692326

RESUMO

BACKGROUND: Eukaryote initiation factor 2 subunit ß (eIF2ß) plays a crucial role in regulation protein synthesis, which mediates the interaction of eIF2 with mRNA. eIF2ß contains evolutionarily conserved polylysine stretches in amino-terminal region and a zinc finger motif in the carboxy-terminus. METHODS: The gene eIF2ß was cloned under tetracycline transcription control and the polylysine stretches were deleted by site-directed mutagenesis (eIF2ßΔ3K). The plasmid was transfected into HEK 293 TetR cells. These cells were analyzed for their proliferative and translation capacities as well as cell death rate. Experiments were performed using gene reporter assays, western blotting, flow cytometry, cell sorting, cell proliferation assays and confocal immunofluorescence. RESULTS: eIF2ßΔ3K affected negatively the protein synthesis, cell proliferation and cell survival causing G2 cell cycle arrest and increased cell death, acting in a negative dominant manner against the native protein. Polylysine stretches are also essential for eIF2ß translocated from the cytoplasm to the nucleus, accumulating in the nucleolus and eIF2ßΔ3K did not make this translocation. DISCUSSION: eIF2ß is involved in the protein synthesis process and should act in nuclear processes as well. eIF2ßΔ3K reduces cell proliferation and causes cell death. Since translation control is essential for normal cell function and survival, the development of drugs or molecules that inhibit translation has become of great interest in the scenario of proliferative disorders. In conclusion, our results suggest the dominant negative eIF2ßΔ3K as a therapeutic strategy for the treatment of proliferative disorders and that eIF2ß polylysine stretch domains are promising targets for this.


Assuntos
Proliferação de Células/genética , Fator de Iniciação 2B em Eucariotos/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , Polilisina/genética , Biossíntese de Proteínas/genética , Deleção de Sequência/genética , Apoptose/genética , Sítios de Ligação , Núcleo Celular/metabolismo , Sobrevivência Celular/genética , Citoplasma/metabolismo , Fator de Iniciação 2B em Eucariotos/metabolismo , Células HEK293 , Humanos , Terapia de Alvo Molecular/métodos , Mutagênese Sítio-Dirigida , Neoplasias/terapia , Ligação Proteica , Transporte Proteico , RNA Mensageiro/metabolismo
12.
Dev Cell ; 41(1): 82-93.e4, 2017 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-28366282

RESUMO

Global transcriptome reprogramming during spermatogenesis ensures timely expression of factors in each phase of male germ cell differentiation. Spermatocytes and spermatids require particularly extensive reprogramming of gene expression to switch from mitosis to meiosis and to support gamete morphogenesis. Here, we uncovered an extensive alternative splicing program during this transmeiotic differentiation. Notably, intron retention was largely the most enriched pattern, with spermatocytes showing generally higher levels of retention compared with spermatids. Retained introns are characterized by weak splice sites and are enriched in genes with strong relevance for gamete function. Meiotic intron-retaining transcripts (IRTs) were exclusively localized in the nucleus. However, differently from other developmentally regulated IRTs, they are stable RNAs, showing longer half-life than properly spliced transcripts. Strikingly, fate-mapping experiments revealed that IRTs are recruited onto polyribosomes days after synthesis. These studies reveal an unexpected function for regulated intron retention in modulation of the timely expression of select transcripts during spermatogenesis.


Assuntos
Diferenciação Celular/genética , Íntrons/genética , Meiose/genética , Espermatozoides/citologia , Espermatozoides/metabolismo , Processamento Alternativo/genética , Animais , Núcleo Celular/genética , Ontologia Genética , Masculino , Camundongos Endogâmicos C57BL , Estabilidade de RNA/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sequências Reguladoras de Ácido Nucleico/genética , Espermatogênese/genética , Transcrição Gênica , Transcriptoma/genética
13.
Oncotarget ; 6(12): 9766-80, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25825981

RESUMO

EMMPRIN/CD147 is mainly known for its protease inducing function but a role in promoting tumor angiogenesis has also been demonstrated. This study provides evidence that EMMPRIN is a new coreceptor for the VEGFR-2 tyrosine kinase receptor in both endothelial and tumor cells, as it directly interacts with it and regulates its activation by its VEGF ligand, signalling and functional consequences both in vitro and in vivo. Computational docking analyses and mutagenesis studies identified a molecular binding site in the extracellular domain of EMMPRIN located close to the cell membrane and containing the amino acids 195/199. EMMPRIN is overexpressed in cancer and hence is able to further potentiate VEGFR-2 activation, suggesting that a combinatory therapy of an antiangiogenic drug together with an inhibitor of EMMPRIN/VEGFR-2 interaction may have a greater impact on inhibiting angiogenesis and malignancy.


Assuntos
Basigina/metabolismo , Regulação Neoplásica da Expressão Gênica , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Inibidores da Angiogênese/química , Animais , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Movimento Celular , Sistema Livre de Células , Simulação por Computador , Feminino , Inativação Gênica , Humanos , Ligantes , Camundongos , Camundongos Nus , Microvasos/citologia , Mutagênese , Mutagênese Sítio-Dirigida , Transplante de Neoplasias , Neovascularização Patológica , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/metabolismo , Transdução de Sinais
14.
Exp Dermatol ; 24(6): 443-8, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25807898

RESUMO

EMMPRIN is known to promote tumor invasion through extracellular matrix (ECM) degradation. Here we report that EMMPRIN can regulate melanoma cell adhesion to the ECM through an interaction with ß1 integrin involving kindlin-3. In this study, EMMPRIN knockdown in the human melanoma cell line M10 using siRNA decreased cell invasion and significantly increased cell adhesion and spreading. A morphological change from a round to a spread shape was observed associated with enhanced phalloidin-labelled actin staining. In situ proximity ligation assay and co-immunoprecipitation revealed that EMMPRIN silencing increased the interaction of ß1 integrin with kindlin-3, a focal adhesion protein. This was associated with an increase in ß1 integrin activation and a decrease in the phosphorylation of the downstream integrin kinase FAK. Moreover, the expression at both the transcript and protein level of kindlin-3 and of ß1 integrin was inversely regulated by EMMPRIN. EMMPRIN did not regulate either talin expression or its interaction with ß1 integrin. These results are consistent with our in vivo demonstration that EMMPRIN inhibition increased ß1 integrin activation and its interaction with kindlin-3. To conclude, these findings reveal a new role of EMMPRIN in tumor cell migration through ß1 integrin/kindlin-3-mediated adhesion pathway.


Assuntos
Basigina/fisiologia , Adesão Celular/fisiologia , Integrina beta1/fisiologia , Melanoma/patologia , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias Cutâneas/patologia , Animais , Basigina/efeitos dos fármacos , Basigina/genética , Linhagem Celular Tumoral , Forma Celular/fisiologia , Matriz Extracelular/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Xenoenxertos , Humanos , Técnicas In Vitro , Melanoma/fisiopatologia , Camundongos , Camundongos Nus , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/fisiologia , Neoplasias Cutâneas/fisiopatologia
15.
Clin Cancer Res ; 21(4): 916-24, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25501128

RESUMO

PURPOSE: Developing strategies to overcome resistance to sunitinib is a major challenge in human renal cell carcinoma (RCC). We hypothesized that sunitinib-induced tumor necrosis-associated hypoxia could interact with renal cancer stem cells in patients with metastatic RCC. EXPERIMENTAL DESIGN: We studied tissue samples from 7 patients with primary metastatic RCC, before and after sunitinib treatment, and from six xenograft models derived from human RCC. Two xenograft models were responders to sunitinib, the four others were nonresponders. CD133/CXCR4-coexpressing cells derived from the two responder xenograft models were used for in vitro studies. RESULTS: In the seven primary RCCs, we identified a significantly larger number of CD133/CXCR4-coexpressing cells in perinecrotic versus perivascular areas. Their numbers also significantly increased after treatment, in perinecrotic areas. We reproduced these clinical and pathologic results in all six RCC xenograft models with again a preferential perinecrotic distribution of CD133-expressing cells. Necrosis occurred at day 7 in the two responder models treated with sunitinib, whereas it occurred at day 21 in the untreated controls and in the four nonresponder models. Strikingly, when we studied the six RCC xenograft models at the time necrosis, whether spontaneous or sunitinib-induced, occurred, necrosis area correlated with stem-cell number in all 120 xenografted RCCs. When studied under experimental hypoxia, the number of CD133/CXCR4-coexpressing cells and their tumorigenic potency increased whereas their sensitivity to sunitinib decreased. CONCLUSIONS: In human RCC, sunitinib was able to generate resistance to its own therapeutic effect via induced hypoxia in perinecrotic areas where cancer stem cells were found in increased numbers.


Assuntos
Carcinoma de Células Renais/patologia , Resistencia a Medicamentos Antineoplásicos/fisiologia , Neoplasias Renais/patologia , Células-Tronco Neoplásicas/patologia , Animais , Antineoplásicos/uso terapêutico , Carcinoma de Células Renais/tratamento farmacológico , Humanos , Indóis/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Camundongos , Camundongos Nus , Necrose , Células-Tronco Neoplásicas/efeitos dos fármacos , Pirróis/uso terapêutico , Sunitinibe , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Oncotarget ; 5(19): 8970-85, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25344860

RESUMO

Kindlin-3 (FERMT-3) is known to be central in hemostasis and thrombosis control and its deficiency disrupts platelet aggregation and causes Leukocyte Adhesion Deficiency disease. Here we report that Kindlin-3 has a tumor suppressive role in solid cancer. Our present genetic and functional data show that Kindlin-3 is downregulated in several solid tumors by a mechanism involving gene hypermethylation and deletions. In vivo experiments demonstrated that Kindlin-3 knockdown in 2 tumor cell models (breast cancer and melanoma) markedly increases metastasis formation, in accord with the in vitro increase of tumor cell malignant properties. The metastatic phenotype was supported by a mechanism involving alteration in ß3-integrin activation including decreased phosphorylation, interaction with talin and the internalization of its active form leading to less cell attachment and more migration/invasion. These data uncover a novel and unexpected tumor suppressor role of Kindlin-3 which can influence integrins targeted therapies development.


Assuntos
Neoplasias da Mama/patologia , Genes Supressores de Tumor , Melanoma/patologia , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Adesão Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Metilação de DNA , Decitabina , Feminino , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Integrina beta3/metabolismo , Melanoma/tratamento farmacológico , Melanoma/genética , Proteínas de Membrana/biossíntese , Proteínas de Membrana/metabolismo , Camundongos , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/metabolismo , Transplante de Neoplasias , Fosforilação , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Interferente Pequeno , Talina/genética
17.
Int J Clin Exp Pathol ; 7(6): 2950-62, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25031714

RESUMO

Renal-cell carcinomas (RCC) are often resistant to conventional cytotoxic agents. Xenograft models are used for in vivo preclinical studies and drug development. The validity of these studies is highly dependent on the phenotypic and genotypic stability of the models. Here we assessed the stability of six aggressive human RCC xenografted in nude/NMRI mice. We compared the initial samples (P0), first (P1) and fifth (P5) passages for the following criteria: histopathology, immunohistochemistry for CK7, CD10, vimentin and p53, DNA allelic profiles using 10 microsatellites and CGH-array. Next we evaluated the response to sunitinib in primary RCC and corresponding xenografted RCC. We observed a good overall stability between primary RCC and corresponding xenografted RCC at P1 and P5 regarding histopathology and immunohistochemistry except for cytokeratin 7 (one case) and p53 (one case) expression. Out of 44 groups with fully available microsatellite data (at P0, P1 and P5), 66% (29 groups) showed no difference from P0 to P5 while 34% (15 groups) showed new or lost alleles. Using CGH-array, overall genomic alterations at P5 were not different from those of initial RCC. The xenografted RCC had identical response to sunitinib therapy compared to the initial human RCC from which they derive. These xenograft models of aggressive human RCC are clinically relevant, showing a good histological and molecular stability and are suitable for studies of basic biology and response to therapy.


Assuntos
Carcinoma de Células Renais/patologia , Modelos Animais de Doenças , Neoplasias Renais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Idoso , Animais , Carcinoma de Células Renais/genética , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Renais/genética , Masculino , Camundongos , Camundongos Nus , Pessoa de Meia-Idade
18.
Eur J Immunol ; 44(10): 3068-80, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25041786

RESUMO

Acute myeloid leukemia (AML) cells are killed by allogeneic NK cells. However, autologous NK cells from AML patients express decreased levels of activating receptors, and show reduced cytotoxicity. Here, we investigated how interactions between NK and AML cells might cause loss of NK-cell activity in patients. Our results show that AML cell lines and primary blasts alter the NK-cell phenotype, reducing their cytotoxic potential upon prolonged contact. Downregulation of NK-cell-activating receptors was contact-dependent and correlated with conjugate formation. Time-lapse imaging of HL60 AML cell line and NK-cell interactions showed a high proportion of noncytolytic contacts. Studies of NK-cell immunological synapses revealed a defect in lytic synapse formation. Namely, despite correct F-actin and LFA-1 recruitment, polarization of lytic granules toward primary blasts or AML cell lines was reduced. The NK-AML cell line synapses showed impairment of CD3ζ recruitment. Attempts to correct these synapse defects by cytokine stimulation of NK cells improved conjugate formation, but not granule polarization. Pretreatment of AML cell lines with the immunomodulating molecule lenalidomide significantly enhanced granule polarization. We speculate that combining immunomodulatory drugs and cytokines could increase AML cell sensitivity to autologous NK cells and reinforce the activity of allogeneic NK cells in adoptive immunotherapy.


Assuntos
Citotoxicidade Imunológica/imunologia , Sinapses Imunológicas/imunologia , Células Matadoras Naturais/imunologia , Leucemia Mieloide Aguda/imunologia , Linhagem Celular Tumoral , Citometria de Fluxo , Humanos , Microscopia de Fluorescência , Imagem com Lapso de Tempo
19.
J Cell Biol ; 204(6): 931-45, 2014 Mar 17.
Artigo em Inglês | MEDLINE | ID: mdl-24637324

RESUMO

The promyelocytic leukemia (PML) protein organizes PML nuclear bodies (NBs), which are stress-responsive domains where many partner proteins accumulate. Here, we clarify the basis for NB formation and identify stress-induced partner sumoylation as the primary NB function. NB nucleation does not rely primarily on intermolecular interactions between the PML SUMO-interacting motif (SIM) and SUMO, but instead results from oxidation-mediated PML multimerization. Oxidized PML spherical meshes recruit UBC9, which enhances PML sumoylation, allow partner recruitment through SIM interactions, and ultimately enhance partner sumoylation. Intermolecular SUMO-SIM interactions then enforce partner sequestration within the NB inner core. Accordingly, oxidative stress enhances NB formation and global sumoylation in vivo. Some NB-associated sumoylated partners also become polyubiquitinated by RNF4, precipitating their proteasomal degradation. As several partners are protein-modifying enzymes, NBs could act as sensors that facilitate and confer oxidative stress sensitivity not only to sumoylation but also to other post-translational modifications, thereby explaining alterations of stress response upon PML or NB loss.


Assuntos
Proteínas Nucleares/metabolismo , Estresse Oxidativo , Sumoilação , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Células CHO , Células COS , Núcleo Celular/metabolismo , Senescência Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Células HeLa , Humanos , Camundongos , Proteína da Leucemia Promielocítica , Transporte Proteico , Espécies Reativas de Oxigênio/metabolismo , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases
20.
Blood ; 122(16): 2864-76, 2013 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-23943652

RESUMO

Myelodysplastic syndrome (MDS) transforms into an acute myelogenous leukemia (AML) with associated increased bone marrow (BM) blast infiltration. Using a transgenic mouse model, MRP8[NRASD12/hBCL-2], in which the NRAS:BCL-2 complex at the mitochondria induces MDS progressing to AML with dysplastic features, we studied the therapeutic potential of a BCL-2 homology domain 3 mimetic inhibitor, ABT-737. Treatment significantly extended lifespan, increased survival of lethally irradiated secondary recipients transplanted with cells from treated mice compared with cells from untreated mice, with a reduction of BM blasts, Lin-/Sca-1(+)/c-Kit(+), and progenitor populations by increased apoptosis of infiltrating blasts of diseased mice assessed in vivo by technicium-labeled annexin V single photon emission computed tomography and ex vivo by annexin V/7-amino actinomycin D flow cytometry, terminal deoxynucleotidyltransferase-mediated dUTP nick end labeling, caspase 3 cleavage, and re-localization of the NRAS:BCL-2 complex from mitochondria to plasma membrane. Phosphoprotein analysis showed restoration of wild-type (WT) AKT or protein kinase B, extracellular signal-regulated kinase 1/2 and mitogen-activated protein kinase patterns in spleen cells after treatment, which showed reduced mitochondrial membrane potential. Exon specific gene expression profiling corroborates the reduction of leukemic cells, with an increase in expression of genes coding for stem cell development and maintenance, myeloid differentiation, and apoptosis. Myelodysplastic features persist underscoring targeting of BCL-2-mediated effects on MDS-AML transformation and survival of leukemic cells.


Assuntos
Compostos de Bifenilo/farmacologia , Leucemia Mieloide Aguda/metabolismo , Nitrofenóis/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sulfonamidas/farmacologia , Proteínas ras/metabolismo , Animais , Antígenos Ly/metabolismo , Linhagem da Célula , Membrana Celular/metabolismo , Proliferação de Células , Transformação Celular Neoplásica , Transplante de Células , Modelos Animais de Doenças , Citometria de Fluxo , Regulação Leucêmica da Expressão Gênica , Sistema de Sinalização das MAP Quinases , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Transgênicos , Mitocôndrias/metabolismo , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-kit/metabolismo , Células-Tronco/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...