Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37905142

RESUMO

Glioblastoma (GBM) is the most aggressive form of primary brain tumor. Complete surgical resection of GBM is almost impossible due to the infiltrative nature of the cancer. While no evidence for recent selection events have been found after diagnosis, the selective forces that govern gliomagenesis are strong, shaping the tumor's cell composition during the initial progression to malignancy with late consequences for invasiveness and therapy response. We present a mathematical model that simulates the growth and invasion of a glioma, given its ploidy level and the nature of its brain tissue micro-environment (TME), and use it to make inferences about GBM initiation and response to standard-of-care treatment. We approximate the spatial distribution of resource access in the TME through integration of in-silico modelling, multi-omics data and image analysis of primary and recurrent GBM. In the pre-malignant setting, our in-silico results suggest that low ploidy cancer cells are more resistant to starvation-induced cell death. In the malignant setting, between first and second surgery, simulated tumors with different ploidy compositions progressed at different rates. Whether higher ploidy predicted fast recurrence, however, depended on the TME. Historical data supports this dependence on TME resources, as shown by a significant correlation between the median glucose uptake rates in human tissues and the median ploidy of cancer types that arise in the respective tissues (Spearman r = -0.70; P = 0.026). Taken together our findings suggest that availability of metabolic substrates in the TME drives different cell fate decisions for cancer cells with different ploidy and shapes GBM disease initiation and relapse characteristics.

2.
Sci Rep ; 9(1): 7587, 2019 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-31110245

RESUMO

Mechanisms of transmesothelial invasion of ovarian cancer are still poorly understood. Here we examined whether this phenomenon may be determined by an expression of intercellular junctions in peritoneal mesothelial cells (PMCs). Analysis of ovarian tumors showed that cancer cells are localized below an intact layer of PMCs. The PMCs located near the invaded cancer cells displayed low expression of connexin 43, E-cadherin, occludin, and desmoglein, as well as expressed SA-ß-Gal, a marker of senescence. Experiments in vitro showed that senescent PMCs exhibited decreased levels of the four tested intercellular junctions, and that the invasion of ovarian cancer cells through the PMCs increased proportionally to the admixture of senescent cells. Intervention studies showed that the expression of connexin 43, E-cadherin, occludin, and desmoglein in senescent PMCs could be restored upon the blockade of p38 MAPK, NF-κB, AKT, JNK, HGF, and TGF-ß1. When these molecules were neutralized, the efficiency of the transmesothelial cancer cell invasion was diminished. Collectively, our findings show that the integrity of the peritoneal mesothelium, which is determined by the expression of junctional proteins, is critical for the invasion of ovarian cancer. They also indicate a mechanism by which senescent PMCs may promote the invasive potential of cancer cells.


Assuntos
Carcinoma Epitelial do Ovário/patologia , Epitélio/patologia , Junções Intercelulares/patologia , Invasividade Neoplásica/patologia , Neoplasias Ovarianas/patologia , Peritônio/patologia , Adulto , Células Cultivadas , Senescência Celular , Feminino , Humanos , Pessoa de Meia-Idade , Adulto Jovem
3.
Biomed Res Int ; 2017: 2592496, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29085834

RESUMO

Here we examined whether malignant ascites may determine ovarian tumor angiogenesis, and if so whether ascites generated by highly aggressive serous and undifferentiated cancers are more proangiogenic than those from less aggressive clear cell and endometrioid tumors. Angiogenesis was analyzed according to expression of CD31, CD34, and connexin 43. Proliferation and migration of endothelial cells were tested using fluorescence-based methods. The quantification of angiogenic agents and hypoxia-inducible factor 1α (HIF-1α) was performed using specific immunoassays. Results showed that the expression of CD31 and CD34 in serous and undifferentiated tumors was greater, whereas endothelial expression of connexin 43 was lower than in clear cell and endometrioid lesions. Serous cancers that formed in the presence of ascites displayed increased expression of connexin 43 in vascular smooth muscles as compared with tumors developed in the fluid's absence. Endothelial cells exposed to ascites from serous and undifferentiated tumors proliferated and migrated more vigorously than cells subjected to ascites from clear cell and endometrioid cancers. They also exhibited an increased level of HIF-1α and produced increased amounts of multiple proangiogenic agents. Our results indicate that high vascularization of aggressive ovarian tumors may be associated with profound angiogenic capabilities of ascites generated by these tumors.


Assuntos
Ascite/metabolismo , Regulação Neoplásica da Expressão Gênica , Proteínas de Neoplasias/biossíntese , Neovascularização Patológica/metabolismo , Neoplasias Ovarianas/metabolismo , Ascite/patologia , Feminino , Humanos , Neovascularização Patológica/patologia , Neoplasias Ovarianas/patologia
4.
Free Radic Biol Med ; 110: 270-279, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28652056

RESUMO

The cancer-promoting activity of senescent peritoneal mesothelial cells (HPMCs) has already been well evidenced both in vitro and in vivo. Here we sought to determine if ovarian cancer cells may activate senescence in HPMCs. The study showed that conditioned medium (CM) from ovarian cancer cells (OVCAR-3, SKOV-3, A2780) inhibited growth and promoted the development of senescence phenotype (increased SA-ß-Gal, γ-H2A.X, 53BP1, and decreased Cx43) in HPMCs. An analysis of tumors isolated from the peritoneum of patients with ovarian cancer revealed an abundance of senescent HPMCs in proximity to cancerous tissue. The presence of senescent HPMCs was incidental when fragments of peritoneum free from cancer were evaluated. An analysis of the cells' secretome followed by intervention studies with exogenous proteins and neutralizing antibodies revealed hepatocyte growth factor (HGF) as the mediator of the pro-senescence impact of the cancer cells. The activity of cancerous CM and HGF was associated with an induction of mitochondrial oxidative stress. Signaling pathways involved in the senescence of HPMCs elicited by the cancer-derived CM and HGF included p38 MAPK, AKT and NF-κB. HPMCs that senesced prematurely in response to the cancer-derived CM promoted adhesion of ovarian cancer cells, however this effect was effectively prevented by the cell protection against oxidative stress. Collectively, our findings indicate that ovarian cancer cells can elicit HGF-dependent senescence in HPMCs, which may contribute to the formation of a metastatic niche for these cells within the peritoneal cavity.


Assuntos
Meios de Cultivo Condicionados/farmacologia , Células Epiteliais/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Fator de Crescimento de Hepatócito/genética , Neoplasias Ovarianas/genética , Anticorpos Neutralizantes/farmacologia , Linhagem Celular Tumoral , Senescência Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Epitélio/efeitos dos fármacos , Epitélio/metabolismo , Epitélio/patologia , Feminino , Fator de Crescimento de Hepatócito/antagonistas & inibidores , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Estresse Oxidativo , Cavidade Peritoneal/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Cell Death Dis ; 7(12): e2565, 2016 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-28032864

RESUMO

Although both incidence and aggressiveness of ovarian malignancy rise with age, the exact reason for this tendency, in particular the contribution of senescent cells, remains elusive. In this project we found that the patient's age determines the frequency of intraperitoneal metastases of ovarian cancer. Moreover, we documented that senescent human peritoneal mesothelial cells (HPMCs) stimulate proliferation, migration and invasion of ovarian cancer cells in vitro, and that this effect is related to both the activity of soluble agents released to the environment by these cells and direct cell-cell contact. The panel of mediators of the pro-cancerous activity of senescent HPMCs appeared to be cancer cell line-specific. The growth of tumors in a mouse peritoneal cavity was intensified when the cancer cells were co-injected together with senescent HPMCs. This effect was reversible when the senescence of HPMCs was slowed down by the neutralization of p38 MAPK. The analysis of lesions excised from the peritoneum of patients with ovarian cancer showed the abundance of senescent HPMCs in close proximity to the cancerous tissue. Collectively, our findings indicate that senescent HPMCs which accumulate in the peritoneum in vivo may create a metastatic niche facilitating intraperitoneal expansion of ovarian malignancy.


Assuntos
Envelhecimento/patologia , Epitélio/patologia , Neoplasias Ovarianas/patologia , Peritônio/patologia , Adulto , Fatores Etários , Animais , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/patologia , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Progressão da Doença , Epitélio/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Endogâmicos C57BL , Camundongos SCID , Pessoa de Meia-Idade , Metástase Neoplásica , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/genética , Fenótipo , Solubilidade , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
6.
Clin Exp Metastasis ; 33(1): 15-27, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26433963

RESUMO

It is believed that senescent cells contribute to the progression of primary and metastatic tumors, however, the exact mechanisms of this activity remain elusive. In this report we show that senescent human peritoneal mesothelial cells (HPMCs) alter the secretory profile of ovarian cancer cells (A2780, OVCAR-3, SKOV-3) by increasing the release of four angiogenic agents: CXCL1, CXCL8, HGF, and VEGF. Proliferation and migration of endothelial cells subjected to conditioned medium generated by: cancer cells modified by senescent HPMCs; cancer cells co-cultured with senescent HPMCs; and by early-passage HPMCs from aged donors, were markedly intensified. The same was the case for the vascularization, size and number of tumors that developed in the mouse peritoneum upon injection of ovarian cancer cells with senescent HPMCs. When the identified pro-angiogenic proteins were neutralized in conditioned medium from the cancer cells, both aspects of endothelial cell behavior intensified in vitro in response to senescent HPMCs were markedly reduced. The search for mediators of senescent HPMC activity using specific neutralizing antibodies and recombinant exogenous proteins showed that the intensified angiogenic potential of cancer cells was elicited by IL-6 and TGF-ß1. At the transcriptional level, increased proliferation and migration of endothelial cells exposed to cancer cells modified by senescent HPMCs was regulated by HIF-1α, NF-κB/p50 and AP-1/c-Jun. Collectively, our findings indicate that senescent HPMCs may promote the progression of ovarian cancer cells by reprogramming their secretory phenotype towards increased production of pro-angiogenic agents and subsequent increase in the angiogenic capabilities of the vascular endothelium.


Assuntos
Senescência Celular/fisiologia , Epitélio/patologia , Neovascularização Patológica/patologia , Neoplasias Ovarianas/patologia , Peritônio/patologia , Animais , Movimento Celular , Proliferação de Células , Progressão da Doença , Feminino , Xenoenxertos , Humanos , Camundongos , Camundongos SCID , Fenótipo
7.
Oncotarget ; 6(30): 29178-95, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26284488

RESUMO

Gastrointestinal cancers metastasize into the peritoneal cavity in a process controlled by peritoneal mesothelial cells (HPMCs). In this paper we examined if senescent HPMCs can intensify the progression of colorectal (SW480) and pancreatic (PSN-1) cancers in vitro and in vivo. Experiments showed that senescent HPMCs stimulate proliferation, migration and invasion of SW480 cells, and migration of PSN-1 cells. When SW480 cells were injected i.p. with senescent HPMCs, the dynamics of tumor formation and vascularization were increased. When xenografts were generated using PSN-1 cells, senescent HPMCs failed to favor their growth. SW480 cells subjected to senescent HPMCs displayed up-regulated expression of transcripts for various pro-cancerogenic agents as well as increased secretion of their products. Moreover, they underwent an epithelial-mesenchymal transition in the Smad 2/3-Snail1-related pathway. The search for mediators of senescent HPMC activity showed that increased SW480 cell proliferation was stimulated by IL-6, migration by CXCL8 and CCL2, invasion by IL-6, MMP-3 and uPA, and epithelial-mesenchymal transition by TGF-ß1. Secretion of these agents by senescent HPMCs was increased in an NF-κB- and p38 MAPK-dependent mechanism. Collectively, our findings indicate that in the peritoneum senescent HPMCs may create a metastatic niche in which critical aspects of cancer progression become intensified.


Assuntos
Senescência Celular , Neoplasias Colorretais/patologia , Neoplasias Pancreáticas/patologia , Comunicação Parácrina , Neoplasias Peritoneais/secundário , Peritônio/patologia , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Transição Epitelial-Mesenquimal , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos SCID , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/metabolismo , Peritônio/metabolismo , Transdução de Sinais , Fatores de Tempo , Microambiente Tumoral
8.
Cancer Lett ; 355(2): 310-5, 2014 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-25301450

RESUMO

The role of mesothelial cells in the intraperitoneal spread of ovarian cancer is still elusive. In particular, it is unclear whether these cells constitute a passive barrier preventing cancer cell progression or perhaps act as an active promoter of this process. In this report we show that omental human peritoneal mesothelial cells (HPMCs) stimulate adhesion and proliferation of ovarian cancer cells (A2780, OVCAR-3, SKOV-3). The latter was associated with the paracrine activity of GRO-1, IL-6, and IL-8 released to the environment by HPMCs. Furthermore, the growth dynamics of ovarian cancer xenografts produced in response to i.p. injection of ovarian cancer cells together with HPMCs was remarkably greater than for implantation of cancer cells alone. A layer of peritoneal mesothelium was consistently present in close proximity to the tumor mass in every xenograft model. In conclusion, our results indicate that HPMCs play a supporting role in the intraperitoneal invasiveness of ovarian malignancy, whose effect may be attributed to their ability to stimulate adhesion and proliferation of cancer cells.


Assuntos
Epitélio/patologia , Neoplasias Ovarianas/patologia , Peritônio/patologia , Animais , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Quimiocina CXCL1/metabolismo , Progressão da Doença , Epitélio/metabolismo , Feminino , Xenoenxertos , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Camundongos , Neoplasias Ovarianas/metabolismo , Peritônio/metabolismo
9.
Skin Res Technol ; 19(4): 432-7, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23521510

RESUMO

BACKGROUND: The main purpose of this study was the exact comparison of B-scan images obtained from 16 patients suffering from AD with histological. METHODS: Sixteen patients diagnosed with AD aged between 14 and 57 years, were chosen for the study. In all patients before the introduction of proper treatment regimen, the high frequency skin ultrasonography (Derma Scan Cortex Technology, version 3, Hadsund, Germany) has been performed and then a 5 mm wide punch biopsies have been taken from the area of scanned lesions. Control group consisted of 15 healthy individuals without any signs of atopic and chronic diseases. RESULTS: There was 7.8 ± 0.4% mean skin echogenicity from lesional antecubital skin in AD patients (min value 7.2%, max value 8.5%). The skin hypoechoic band was detected in fifteen out of sixteen patients (93.7%). There were statistically significant correlations between the hypoechoic band thickness and the following parameters: degree of epidermal hyperplasia, the degree of epidermal hyperkeratosis, the degree of parakeratosis and the degree of spongiosis as well as the intensity of inflammatory infiltrates. Skin echogenicity strongly correlated negatively with the intensity of inflammatory infiltrates. CONCLUSION: On the basis of analysis of 16 specimens we can conclude that all pathological changes of AD influence ultrasound image and in addition, the USG picture depends on the phase of disease process.


Assuntos
Dermatite Atópica/diagnóstico por imagem , Epiderme/diagnóstico por imagem , Índice de Gravidade de Doença , Ultrassonografia/métodos , Adolescente , Adulto , Biópsia , Dermatite Atópica/patologia , Epiderme/patologia , Feminino , Humanos , Hiperplasia/diagnóstico por imagem , Hiperplasia/patologia , Ceratose/diagnóstico por imagem , Ceratose/patologia , Masculino , Pessoa de Meia-Idade , Paraceratose/diagnóstico por imagem , Paraceratose/patologia , Ultrassonografia/instrumentação , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...