Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Scand J Public Health ; : 14034948241228482, 2024 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-38436303

RESUMEN

AIMS: Connecting cohorts with biobanks is a Finnish biobank collaboration, creating an infrastructure for the study of healthy ageing. We aimed to develop a model for data integration and harmonisation between different biobanks with procedures for joint access. METHODS: The heart of the collaboration is the integrated datasets formed by using data from three biobanks: (a) Arctic Biobank, hosting regional birth cohorts and cohorts of elderly; (b) hospital-affiliated Borealis Biobank of Northern Finland; and (c) THL Biobank, hosting population-based cohorts. The datasets were created by developing a data dictionary, harmonising cohort data and with a joint pseudonymisation process. RESULTS: The connecting cohorts with biobanks resource at its widest consists altogether of almost 1.4 million individuals from collaborating biobanks. Utilising data from 107,000 cohort participants, we created harmonised datasets that contain attributes describing metabolic risk and frailty for studies of healthy ageing. These data can be complemented with medical data available from Biobank Borealis and with samples taken at hospital settings for approximately 38,000 cohort participants. In addition, the harmonised connecting cohorts with biobanks datasets can be expanded with supplementary data and samples from the collaborating biobanks. CONCLUSIONS: The connecting cohorts with biobanks datasets provide a unique resource for research on ageing-related personalised healthcare and for real-world evidence studies. Following the FAIR principles on findability, accessibility, interoperability, and reusability, the reused and harmonised datasets are findable and made accessible for researchers. The same approach can be further utilised to develop additional datasets for other research topics.

2.
Exp Cell Res ; 376(1): 18-26, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30710501

RESUMEN

Pro-tumorigenic activities of matrix metalloproteinase (MMP) 9 have been linked to many cancers, but recently the tumour-suppressing role of MMP9 has also been elucidated. The multifaceted evidence on this subject prompted us to examine the role of MMP9 in the behaviour of oral tongue squamous cell carcinoma (OTSCC) cells. We used gelatinase-specific inhibitor, CTT2, and short hairpin (sh) RNA gene silencing to study the effects of MMP9 on proliferation, motility and invasion of an aggressive OTSCC cell line, HSC-3. We found that the migration and invasion of HSC-3 cells were increased by CTT2 and shRNA silencing of MMP9. Proliferation, in turn, was decreased by MMP9 inhibition. Furthermore, arresten-overexpressing HSC-3 cells expressed increased levels of MMP9, but exhibited decreased motility compared with controls. Interestingly, these cells restored their migratory capabilities by CTT2 inhibition of MMP9. Hence, although higher MMP9 expression could give rise to an increased tumour growth in vivo due to increased proliferation, in some circumstances, it may participate in yet unidentified molecular mechanisms that reduce the cell movement in OTSCC.


Asunto(s)
Carcinoma de Células Escamosas/genética , Metaloproteinasa 9 de la Matriz/genética , Neoplasias de la Boca/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Gelatinasas/antagonistas & inhibidores , Gelatinasas/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias de la Boca/patología , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Péptidos Cíclicos/farmacología
3.
Int J Mol Sci ; 20(18)2019 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-31514474

RESUMEN

Matrix metalloproteinases (MMPs) have traditionally been considered as tumor promoting enzymes as they degrade extracellular matrix components, thus increasing the invasion of cancer cells. It has become evident, however, that MMPs can also cleave and alter the function of various non-matrix bioactive molecules, leading to both tumor promoting and suppressive effects. We applied systematic review guidelines to study MMP8 in cancer including the use of MMP8 as a prognostic factor or as a target/anti-target in cancer treatment, and its molecular mechanisms. A total of 171 articles met the inclusion criteria. The collective evidence reveals that in breast, skin and oral tongue cancer, MMP8 inhibits cancer cell invasion and proliferation, and protects patients from metastasis via cleavage of non-structural substrates. Conversely, in liver and gastric cancers, high levels of MMP8 worsen the prognosis. Expression and genetic alterations of MMP8 can be used as a prognostic factor by examination of the tumor and serum/plasma. We conclude, that MMP8 has differing effects on cancers depending on their tissue of origin. The use of MMP8 as a prognostic factor alone, or with other factors, seems to have potential. The molecular mechanisms of MMP8 in cancer further emphasize its role as an important regulator of bioactive molecules.


Asunto(s)
Metaloproteinasa 8 de la Matriz/metabolismo , Neoplasias/enzimología , Animales , Biomarcadores de Tumor/metabolismo , Epigénesis Genética , Humanos , Metaloproteinasa 8 de la Matriz/sangre , Neoplasias/sangre , Neoplasias/genética , Polimorfismo de Nucleótido Simple/genética , Pronóstico
4.
Exp Cell Res ; 344(2): 229-40, 2016 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-27090016

RESUMEN

The invasion of carcinoma cells is a crucial feature in carcinogenesis. The penetration efficiency not only depends on the cancer cells, but also on the composition of the tumor microenvironment. Our group has developed a 3D invasion assay based on human uterine leiomyoma tissue. Here we tested whether human, porcine, mouse or rat hearts as well as porcine tongue tissues could be similarly used to study carcinoma cell invasion in vitro. Three invasive human oral tongue squamous cell carcinoma (HSC-3, SCC-25 and SCC-15), melanoma (G-361) and ductal breast adenocarcinoma (MDA-MB-231) cell lines, and co-cultures of HSC-3 and carcinoma-associated or normal oral fibroblasts were assayed. Myoma tissue, both native and lyophilized, promoted invasion and growth of the cancer cells. However, the healthy heart or tongue matrices were unable to induce the invasion of any type of cancer cells tested. Moreover, when studied in more detail, small molecular weight fragments derived from heart tissue rinsing media inhibited HSC-3 horizontal migration. Proteome analysis of myoma rinsing media, on the other hand, revealed migration enhancing factors. These results highlight the important role of matrix composition for cancer invasion studies in vitro and further demonstrate the unique properties of human myoma organotypic model.


Asunto(s)
Matriz Extracelular/metabolismo , Neoplasias/patología , Microambiente Tumoral , Animales , Línea Celular Tumoral , Membrana Celular/patología , Movimiento Celular , Colágeno/metabolismo , Liofilización , Humanos , Ratones , Miocardio/patología , Mioma/patología , Invasividad Neoplásica , Ratas , Receptores de Superficie Celular/metabolismo , Solubilidad , Sus scrofa , Lengua/patología
5.
Exp Cell Res ; 336(1): 130-40, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26112215

RESUMEN

The turnover of extracellular matrix liberates various cryptic molecules with novel biological activities. Endostatin is an endogenous angiogenesis inhibitor that is derived from the non-collagenous domain of collagen XVIII. Although there are a large number of studies on its anti-tumor effects, the molecular mechanisms are not yet completely understood, and the reasons why endostatin has not been successful in clinical trials are unclear. Research has mostly focused on its anti-angiogenic effect in tumors. Here, we aimed to elucidate how endostatin affects the behavior of aggressive tongue HSC-3 carcinoma cells that were transfected to overproduce endostatin. Endostatin inhibited the invasion of HSC-3 cells in a 3D collagen-fibroblast model. However, it had no effect on invasion in a human myoma organotypic model, which lacks vital fibroblasts. Recombinant endostatin was able to reduce the Transwell migration of normal fibroblasts, but had no effect on carcinoma associated fibroblasts. Surprisingly, endostatin increased the proliferation and decreased the apoptosis of cancer cells in organotypic models. Also subcutaneous tumors overproducing endostatin grew bigger, but showed less local invasion in nude mice xenografts. We conclude that endostatin affects directly to HSC-3 cells increasing their proliferation, but its net effect on cancer invasion seem to depend on the cellular composition and interactions of tumor microenvironment.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Carcinoma de Células Escamosas/patología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Endostatinas/farmacología , Neoplasias de la Lengua/patología , Microambiente Tumoral/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/irrigación sanguínea , Carcinoma de Células Escamosas/tratamiento farmacológico , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Miofibroblastos/efectos de los fármacos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Mioma/irrigación sanguínea , Mioma/tratamiento farmacológico , Mioma/patología , Invasividad Neoplásica , Neovascularización Patológica/tratamiento farmacológico , Neoplasias de la Lengua/irrigación sanguínea , Neoplasias de la Lengua/tratamiento farmacológico , Células Tumorales Cultivadas , Neoplasias Uterinas/irrigación sanguínea , Neoplasias Uterinas/tratamiento farmacológico , Neoplasias Uterinas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
6.
BMC Cancer ; 15: 25, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25633184

RESUMEN

BACKGROUND: Caveolin-1 (CAV1) may be upregulated by hypoxia and acts in a tumor-dependent manner. We investigated CAV1 in tongue squamous cell carcinoma (TSCC) and its association with clinical outcomes, and studied in vitro possible ways for CAV1 accumulation in the tumor microenvironment (TME). METHODS: TSCC cases (N = 64) were immunohistochemically stained for CAV1. Scores were separately assessed in the tumor and TME and plotted for association with recurrence and survival (univariate analysis with log-rank test). In vitro studies were performed on a 3D myoma organotypic model, a mimicker of TME. Prior to monoculturing HSC-3 tongue cancer cells, the model underwent modifications in oxygenation level (1%O2 hypoxia to upregulate CAV1) and/or in the amount of natural soluble factors [deleted by 14-day rinsing (rinsed myoma, RM), to allow only HSC-3-derived factors to act]. Controls included normoxia (21%O2) and naturally occurring soluble factors (intact myoma, IM). HSC-3 cells were also co-cultured with CaDEC12 cells (fibroblasts exposed to human tongue cancer). CAV1 expression and cellular distribution were examined in different cellular components in hypoxic and rinsed myoma assays. Twist served as a marker for the process of epithelial-mesenchymal transition (EMT). Exosomes isolated from HSC-3 media were investigated for containing CAV1. RESULTS: Expression of CAV1 in TSCC had a higher score in TME than in the tumor cells and a negative impact on recurrence (p = 0.01) and survival (p = 0.003). Monocultures of HSC-3 revealed expression of CAV1 mainly in the TME-like myoma assay, similar to TSCC. CAV1+, alpha-smooth muscle actin (αSMA) + and Twist + CAF-like cells were observed surrounding the invading HSC-3, possibly reflecting EMT. RM findings were similar to IM, inferring action of HSC-3 derived factors, and no differences were seen when hypoxia was induced. HSC-3-CaDEC12 co-cultures revealed CAV1+, αSMA+ and cytokeratin-negative CAF-like cells, raising the possibility of CaDEC12 cells gaining a CAF phenotype. HSC-3-derived exosomes were loaded with CAV1. CONCLUSIONS: Accumulation of CAV1-TME in TSCC had a negative prognostic value. In vitro studies showed the presence of CAV1 in cancer cells undergoing EMT and in fibroblasts undergoing trans-differentiation to CAFs. CAV1 delivery to the TME involved cancer cell-derived exosomes.


Asunto(s)
Caveolina 1/metabolismo , Neoplasias de la Lengua/metabolismo , Neoplasias de la Lengua/mortalidad , Microambiente Tumoral , Adulto , Anciano , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/mortalidad , Caveolina 1/genética , Técnicas de Cultivo de Célula , Técnicas de Cocultivo , Femenino , Estudios de Seguimiento , Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Neoplasias de la Lengua/genética , Neoplasias de la Lengua/patología , Células Tumorales Cultivadas , Microambiente Tumoral/genética
7.
Exp Cell Res ; 325(2): 58-64, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24462456

RESUMEN

The research on oral cancer has focused mainly on the cancer cells, their genetic changes and consequent phenotypic modifications. However, it is increasingly clear that the tumor microenvironment (TME) has been shown to be in a dynamic state of inter-relations with the cancer cells. The TME contains a variety of components including the non-cancerous cells (i.e., immune cells, resident fibroblasts and angiogenic vascular cells) and the ECM milieu [including fibers (mainly collagen and fibronectin) and soluble factors (i.e., enzymes, growth factors, cytokines and chemokines)]. Thus, it is currently assumed that TME is considered a part of the cancerous tissue and the functionality of its key components constitutes the setting on which the hallmarks of the cancer cells can evolve. Therefore, in terms of controlling a malignancy, one should control the growth, invasion and spread of the cancer cells through modifications in the TME components. This mini review focuses on the TME as a diagnostic approach and reports the recent insights into the role of different TME key components [such as carcinoma-associated fibroblasts (CAFs) and inflammation (CAI) cells, angiogenesis, stromal matrix molecules and proteases] in the molecular biology of oral carcinoma. Furthermore, the impact of TME components on clinical outcomes and the concomitant need for development of new therapeutic approaches will be discussed.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Microambiente Tumoral/efectos de los fármacos , Animales , Humanos
8.
Exp Cell Res ; 319(4): 376-89, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23262025

RESUMEN

Invasion is an important hallmark of cancer involving interactions between the tumor microenvironment and the cancer cells. Hypoxia, low oxygen level, is related to increased invasion and metastasis in many cancers. The aim was to elucidate the effect of hypoxia on invasion of oral squamous cell carcinoma cells (OSCCs), and the applicability of a novel 3-dimentional myoma organotypic invasion model in hypoxia experiments. OSCC cell lines (primary oral carcinoma derived cells UT-SCC-43A, recurrent oral carcinoma cells UT-SCC-43B and aggressive tongue carcinoma cells HSC-3) were studied for their migration and invasion capabilities under normoxia, hypoxia, and in the presence a hypoxia-mimicker cobalt chloride. As expected, the recurrent UT-SCC-43B cells were significantly more aggressive than the primary tumor derived cells. In contrast to tongue carcinoma HSC-3 cells, they only mildly responded to hypoxia in the migration or invasion assays, indicating a cell line specific response of hypoxia on the invasive potential. The modification of the organotypic human tissue-derived matrix via the removal of various yet unidentified soluble factors by rinsing the tissue resulting in stripped matrix substantially changed the invasion pattern of HSC-3 cells and the outcomes of hypoxic treatments. Only in the stripped tissue hypoxia significantly increased invasion, whereas in native intact tissue the induced invasion was not observed. This demonstrates the importance of the soluble factors to the invasion pattern and to the hypoxia response. A metastasis and poor prognosis marker, hypoxia-regulated lysyl oxidase (LOX), was present in the myoma tissue, but could be removed by rinsing. The inhibition of LOX resulted in a decrease in invasion area, but only very mildly in invasion depth. Thus, it may have a role in the modulation of the invasion pattern. Another hypoxia-related poor prognosis marker carbonic anhydrase 9 (CAIX) was induced in HSC-3 cells both by the hypoxic exposure and interestingly in invading HSC-3 cells inside the tissue even in normoxic conditions. In conclusion, this suggests that the intact myoma organotypic model offers optimally hypoxic surroundings, thus being an excellent human tumor microenvironment mimicker.


Asunto(s)
Carcinoma de Células Escamosas/patología , Hipoxia/patología , Neoplasias de la Boca/patología , Microambiente Tumoral/fisiología , Anciano , Aminopropionitrilo/farmacología , Carcinoma de Células Escamosas/metabolismo , Ensayos de Migración Celular , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Hipoxia/complicaciones , Leiomioma/metabolismo , Leiomioma/patología , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/metabolismo , Invasividad Neoplásica , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Proteína-Lisina 6-Oxidasa/metabolismo , Proteína-Lisina 6-Oxidasa/fisiología , Células Tumorales Cultivadas , Microambiente Tumoral/efectos de los fármacos , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología , Cicatrización de Heridas
9.
BMJ Open ; 14(6): e081947, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38866570

RESUMEN

INTRODUCTION: A better understanding of the earliest stages of Alzheimer's disease (AD) could expedite the development or administration of treatments. Large population biobanks hold the promise to identify individuals at an elevated risk of AD and related dementias based on health registry information. Here, we establish the protocol for an observational clinical recall and biomarker study called TWINGEN with the aim to identify individuals at high risk of AD by assessing cognition, health and AD-related biomarkers. Suitable candidates were identified and invited to participate in the new study among THL Biobank donors according to TWINGEN study criteria. METHODS AND ANALYSIS: A multi-centre study (n=800) to obtain blood-based biomarkers, telephone-administered and web-based memory and cognitive parameters, questionnaire information on lifestyle, health and psychological factors, and accelerometer data for measures of physical activity, sedentary behaviour and sleep. A subcohort is being asked to participate in an in-person neuropsychological assessment (n=200) and wear an Oura ring (n=50). All participants in the TWINGEN study have genome-wide genotyping data and up to 48 years of follow-up data from the population-based older Finnish Twin Cohort (FTC) study of the University of Helsinki. The data collected in TWINGEN will be returned to THL Biobank from where it can later be requested for other biobank studies such as FinnGen that supported TWINGEN. ETHICS AND DISSEMINATION: This recall study consists of FTC/THL Biobank/FinnGen participants whose data were acquired in accordance with the Finnish Biobank Act. The recruitment protocols followed the biobank protocols approved by Finnish Medicines Agency. The TWINGEN study plan was approved by the Ethics Committee of Hospital District of Helsinki and Uusimaa (number 16831/2022). THL Biobank approved the research plan with the permission no: THLBB2022_83.


Asunto(s)
Enfermedad de Alzheimer , Bancos de Muestras Biológicas , Biomarcadores , Humanos , Finlandia , Biomarcadores/sangre , Femenino , Anciano , Masculino , Estudios de Cohortes , Persona de Mediana Edad , Pruebas Neuropsicológicas , Cognición , Factores de Riesgo , Proyectos de Investigación
10.
ScientificWorldJournal ; 2013: 920595, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23365550

RESUMEN

One hallmark of cancer is the degradation of the extracellular matrix (ECM), which is caused by proteinases. In oral cancers, matrix metalloproteinases (MMPs), especially MMP-9, are associated with this degradation. MMPs break down the ECM allowing cancer to spread; they also release various factors from their cryptic sites, including cytokines. These factors modulate cell behavior and enhance cancer progression by regulating angiogenesis, migration, proliferation, and invasion. The development of early metastases is typical for oral cancer, and increased MMP-9 expression is associated with a poor disease prognosis. However, many studies fail to relate MMP-9 expression with metastasis formation. Contrary to earlier models, recent studies show that MMP-9 plays a protective role in oral cancers. Therefore, the role of MMP-9 is complicated and may fluctuate throughout the different types and stages of oral cancers.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Metaloproteinasa 9 de la Matriz/metabolismo , Modelos Biológicos , Neoplasias de la Boca/enzimología , Boca/enzimología , Animales , Humanos
11.
Comput Med Imaging Graph ; 108: 102276, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37611486

RESUMEN

Submucosal invasion depth is a significant prognostic factor when assessing lymph node metastasis and cancer itself to plan proper treatment for the patient. Conventionally, oncologists measure the invasion depth by hand which is a laborious, subjective, and time-consuming process. The manual pathological examination by measuring accurate carcinoma cell invasion with considerable inter-observer and intra-observer variations is still challenging. The increasing use of medical imaging and artificial intelligence reveals a significant role in clinical medicine and pathology. In this paper, we propose an approach to study invasive behavior and measure the invasion depth of carcinoma from stained histopathology images. Specifically, our model includes adaptive stain normalization, color decomposition, and morphological reconstruction with adaptive thresholding to separate the epithelium with blue ratio image. Our method splits the image into multiple non-overlapping meaningful segments and successfully finds the homogeneous segments to measure accurate invasion depth. The invasion depths are measured from the inner epithelium edge to outermost pixels of the deepest part of particles in image. We conduct our experiments on skin melanoma tissue samples as well as on organotypic invasion model utilizing myoma tissue and oral squamous cell carcinoma. The performance is experimentally compared to three closely related reference methods and our method provides a superior result in measuring invasion depth. This computational technique will be beneficial for the segmentation of epithelium and other particles for the development of novel computer-aided diagnostic tools in biobank applications.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de la Boca , Humanos , Inteligencia Artificial , Neoplasias de la Boca/diagnóstico por imagen , Epidermis , Células Epidérmicas , Colorantes
12.
medRxiv ; 2023 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-37965200

RESUMEN

Introduction: A better understanding of the earliest stages of Alzheimer's disease (AD) could expedite the development or administration of treatments. Large population biobanks hold the promise to identify individuals at an elevated risk of AD and related dementias based on health registry information. Here, we establish the protocol for an observational clinical recall and biomarker study called TWINGEN with the aim to identify individuals at high risk of AD by assessing cognition, health and AD-related biomarkers. Suitable candidates were identified and invited to participate in the new study among Finnish biobank donors according to TWINGEN study criteria. Methods and analysis: A multi-center study (n=800) to obtain blood-based biomarkers, telephone-administered and web-based memory and cognitive parameters, questionnaire information on lifestyle, health and psychological factors, and accelerometer data for measures of physical activity, sedentary behavior and sleep. A sub-cohort are being asked to participate in an in-person neuropsychological assessment (n=200) and wear an Oura ring (n=50). All participants in the TWINGEN study have genome-wide genotyping data and up to 48 years of follow-up data from the population-based older Finnish Twin Cohort (FTC) study of the University of Helsinki. TWINGEN data will be transferred to Finnish Institute of Health and Welfare (THL) biobank and we aim to further to transfer it to the FinnGen study where it will be combined with health registry data for prediction of AD. Ethics and dissemination: This recall study consists of FTC/THL/FinnGen participants whose data were acquired in accordance with the Finnish Biobank Act. The recruitment protocols followed the biobank protocols approved by Finnish Medicines Agency. The TWINGEN study plan was approved by the Ethics Committee of Hospital District of Helsinki and Uusimaa (number 16831/2022). THL Biobank approved the research plan with the permission no: THLBB2022_83.

13.
Cancer Invest ; 30(8): 583-92, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22909050

RESUMEN

Enhanced proteolysis and altered tight junction (TJ) proteins associate with carcinoma invasion. We hypothesized that trypsin-2, a tumor-associated serine proteinase, induces tongue carcinoma invasion by activating pro-membrane type-1 matrix metalloproteinase (MT1-MMP) and disturbing the TJs. The effects of invasion were analyzed using trypsin-2 over-expressing human tongue squamous cell carcinoma cells (Try2-HSC-3) in vitro and in vivo. The invasion of Try2-HSC-3 cells was increased in mouse xenografts and human organotypic model. Trypsin-2 activated proMT1-MMP, as well as altered the expression of TJ protein claudin-7. In conclusion, trypsin-2 over-expression enhanced tongue carcinoma cell invasion by various genetic and proteolytic mechanisms.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Metaloproteinasa 14 de la Matriz/metabolismo , Precursores de Proteínas/metabolismo , Uniones Estrechas/metabolismo , Neoplasias de la Lengua/metabolismo , Neoplasias de la Lengua/patología , Tripsina/metabolismo , Tripsinógeno/metabolismo , Animales , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Movimiento Celular/genética , Claudinas/genética , Claudinas/metabolismo , Activación Enzimática , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Desnudos , Invasividad Neoplásica , Precursores de Proteínas/genética , Neoplasias de la Lengua/genética , Tripsina/genética , Tripsinógeno/genética , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Comput Biol Med ; 144: 105301, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35255294

RESUMEN

In the recent decade, medical image registration and fusion process has emerged as an effective application to follow up diseases and decide the necessary therapies based on the conditions of patient. For many of the considerable diagnostic analyses, it is common practice to assess two or more different histological slides or images from one tissue sample. A specific area analysis of two image modalities requires an overlay of the images to distinguish positions in the sample that are organized at a similar coordinate in both images. In particular cases, there are two common challenges in digital pathology: first, dissimilar appearances of images resulting due to staining variances and artifacts; second, large image size. In this paper, we develop algorithm to overcome the fact that scanners from different manufacturers have variations in the images. We propose whole slide image registration algorithm where adaptive smoothing is employed to smooth the stained image. A modified scale-invariant feature transform is applied to extract common information and a joint distance helps to match keypoints correctly by eliminating position transformation error. Finally, the registered image is obtained by utilizing correct correspondences and the interpolation of color intensities. We validate our proposal using different images acquired from surgical resection samples of lung cancer (adenocarcinoma). Extensive feature matching with apparently increasing correct correspondences and registration performance on several images demonstrate the superiority of our method over state-of-the-art methods. Our method potentially improves the matching accuracy that might be beneficial for computer-aided diagnosis in biobank applications.


Asunto(s)
Algoritmos , Procesamiento de Imagen Asistido por Computador , Artefactos , Diagnóstico por Computador , Humanos , Procesamiento de Imagen Asistido por Computador/métodos , Coloración y Etiquetado
15.
Comput Med Imaging Graph ; 90: 101901, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33862354

RESUMEN

Medical imaging provides the means for diagnosing many of the medical phenomena currently studied in clinical medicine and pathology. The variations of color and intensity in stained histological slides affect the quantitative analysis of the histopathological images. Moreover, stain normalization utilizing color for the classification of pixels into different stain components is challenging. The staining also suffers from variability, which complicates the automatization of tissue area segmentation with different staining and the analysis of whole slide images. We have developed a Retinex model based stain normalization technique in terms of area segmentation from stained tissue images to quantify the individual stain components of the histochemical stains for the ideal removal of variability. The performance was experimentally compared to reference methods and tested on organotypic carcinoma model based on myoma tissue and our method consistently has the smallest standard deviation, skewness value, and coefficient of variation in normalized median intensity measurements. Our method also achieved better quality performance in terms of Quaternion Structure Similarity Index Metric (QSSIM), Structural Similarity Index Metric (SSIM), and Pearson Correlation Coefficient (PCC) by improving robustness against variability and reproducibility. The proposed method could potentially be used in the development of novel research as well as diagnostic tools with the potential improvement of accuracy and consistency in computer aided diagnosis in biobank applications.


Asunto(s)
Colorantes , Procesamiento de Imagen Asistido por Computador , Diagnóstico por Computador , Reproducibilidad de los Resultados , Coloración y Etiquetado
16.
Am J Pathol ; 175(3): 1281-91, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19679876

RESUMEN

Carcinoma cell invasion is traditionally studied in three-dimensional organotypic models composed of type I collagen and fibroblasts. However, carcinoma cell behavior is affected by the various cell types and the extracellular matrix (ECM) in the tumor microenvironment. In this study, a novel organotypic model based on human uterine leiomyoma tissue was established and characterized to create a more authentic environment for carcinoma cells. Human tongue squamous cell carcinoma cells (HSC-3) were cultured on top of either collagen or myoma. Organotypic sections were examined by immunohistochemistry and in situ hybridization. The maximal invasion depth of HSC-3 cells was markedly increased in myomas compared with collagen. In myomas, various cell types and ECM components were present, and the HSC-3 cells only expressed ECM molecules in the myoma model. Organotypic media were analyzed by radioimmunoassay, zymography, or Western blotting. During carcinoma cell invasion, matrix metalloprotease-9 production and collagen degradation were enhanced particularly in the myoma model. To evaluate the general applicability of the myoma model, several oral carcinoma, breast carcinoma, and melanoma cell lines were cultured on myomas and found to invade in highly distinct patterns. We conclude that myoma tissue mimics the native tumor microenvironment better than previous organotypic models and possibly enhances epithelial-to-mesenchymal transition. Thus, the myoma model provides a promising tool for analyzing the behavior of carcinoma cells.


Asunto(s)
Carcinoma de Células Escamosas/patología , Comunicación Celular , Leiomioma/patología , Modelos Biológicos , Neoplasias de la Lengua/patología , Neoplasias Uterinas/patología , Neoplasias de la Mama/patología , Carcinoma de Células Escamosas/metabolismo , Técnicas de Cocultivo , Colágeno , Femenino , Humanos , Leiomioma/metabolismo , Metaloproteinasa 9 de la Matriz/biosíntesis , Melanoma/patología , Neoplasias de la Boca/patología , Invasividad Neoplásica , Técnicas de Cultivo de Órganos , Neoplasias de la Lengua/metabolismo , Neoplasias Uterinas/metabolismo
17.
Exp Cell Res ; 314(18): 3292-305, 2008 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-18775695

RESUMEN

Physiological and pathological turnover of basement membranes liberates biologically active cryptic molecules. Several collagen-derived fragments possess anti-angiogenic activity. Arresten is the 26-kDa non-collagenous domain of type IV collagen alpha1 chain. It functions as an efficient inhibitor of angiogenesis and tumor growth in mouse models, but its anti-angiogenic mechanism is not completely known. Here we show that arresten significantly increases apoptosis of endothelial cells in vitro by decreasing the amount of anti-apoptotic molecules of the Bcl-family; Bcl-2 and Bcl-xL. Although the pro-apoptotic effect of arresten is endothelial cell specific in vitro, in mouse tumors arresten induced apoptosis both in endothelial and tumor cells. The tumor cell apoptosis is likely an indirect effect due to the inhibition of blood vessel growth into the tumor. The active site of arresten was localized by deletion mutagenesis within the C-terminal half of the molecule. We have previously shown that arresten binds to alpha1beta1 integrin on human umbilical vein endothelial cells. However, the microvascular endothelial cells (MLECs) are more important in the context of tumor vasculature. We show here that arresten binds also to the microvascular endothelial cells via alpha1beta1 integrin. Furthermore, it has no effect on Matrigel neovascularization or the viability of integrin alpha1 null MLECs. Tumors implanted on integrin alpha1 deficient mice show no integrin alpha1 expression in the host-derived vascular endothelium, and thus arresten does not inhibit the tumor growth. Collectively, this data sheds more light into the anti-angiogenic mechanism of arresten.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Colágeno Tipo IV/química , Células Endoteliales/efectos de los fármacos , Integrina alfa1beta1/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Supresoras de Tumor/farmacología , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/metabolismo , Animales , Aorta/citología , Apoptosis/efectos de los fármacos , Western Blotting , Bovinos , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Colágeno Tipo IV/genética , Células Endoteliales/citología , Humanos , Integrina alfa1beta1/genética , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Estructura Terciaria de Proteína/fisiología , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Arteria Pulmonar/citología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteína bcl-X/efectos de los fármacos , Proteína bcl-X/metabolismo
18.
J Clin Invest ; 115(10): 2801-10, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16151532

RESUMEN

Human noncollagenous domain 1 of the alpha1 chain of type IV collagen [alpha1(IV)NC1], or arresten, is derived from the carboxy terminal of type IV collagen. It was shown to inhibit angiogenesis and tumor growth in vivo; however, the mechanisms involved are not known. In the present study we demonstrate that human alpha1(IV)NC1 binds to alpha1beta1 integrin, competes with type IV collagen binding to alpha1beta1 integrin, and inhibits migration, proliferation, and tube formation by ECs. Also, alpha1(IV)NC1 pretreatment inhibited FAK/c-Raf/MEK/ERK1/2/p38 MAPK activation in ECs but had no effect on the PI3K/Akt pathway. In contrast, alpha1(IV)NC1 did not affect proliferation, migration, or the activation of FAK/c-Raf/MEK1/2/p38/ERK1 MAPK pathway in alpha1 integrin receptor knockout ECs. Consistent with this, alpha1(IV)NC1 elicited significant antiangiogenic effects and tumor growth inhibition in vivo but failed to do the same in alpha1 integrin receptor knockout mice. This suggests a highly specific, alpha1beta1 integrin-dependent antiangiogenic activity of alpha1(IV)NC1. In addition, alpha1(IV)NC1 inhibited hypoxia-induced expression of hypoxia-inducible factor 1alpha and VEGF in ECs cultured on type IV collagen by inhibiting ERK1/2 and p38 activation. This unravels a hitherto unknown function of human alpha1(IV)NC1 and suggests a critical role for integrins in hypoxia and hypoxia-induced angiogenesis. Collectively, the above data indicate that alpha1(IV)NC1 is a potential therapeutic candidate for targeting tumor angiogenesis.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Colágeno Tipo IV/metabolismo , Células Endoteliales/metabolismo , Sangre Fetal/metabolismo , Integrina alfa1beta1/metabolismo , Sistema de Señalización de MAP Quinasas , Neovascularización Patológica/metabolismo , Inhibidores de la Angiogénesis/genética , Inhibidores de la Angiogénesis/farmacología , Hipoxia de la Célula/efectos de los fármacos , Colágeno Tipo IV/genética , Colágeno Tipo IV/farmacología , Células Endoteliales/citología , Activación Enzimática , Sangre Fetal/citología , Eliminación de Gen , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/biosíntesis , Integrina alfa1beta1/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neovascularización Patológica/tratamiento farmacológico , Proteínas Quinasas/metabolismo , Estructura Terciaria de Proteína/genética , Factor A de Crecimiento Endotelial Vascular/biosíntesis
19.
Front Biosci ; 13: 6537-53, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508679

RESUMEN

The tumor microenvironment is a mixture of extracellular matrix molecules, tumor cells, endothelial cells, fibroblasts and immune cells. Tumor growth and metastasis formation are dependent on the growth of blood vessels into the tumor mass. The tumor microenvironment contributes to this pathological angiogenic process. The extracellular matrix and basement membranes are a source for endogenous angiogenesis inhibitors, such as endostatin. On the other hand, many extracellular matrix molecules can promote angiogenesis by stabilizing blood vessels and sequestering pro-angiogenic growth factors. The majority of stromal cells in carcinomas are fibroblasts. Carcinoma-associated fibroblasts show a distinct phenotype from normal fibroblasts. The mechanisms how the tumor-associated fibroblasts regulate angiogenesis are not fully known, but they are suggested to be an important source for growth factors and cytokines recruiting endothelial cells. The immune cells, particularly macrophages and neutrophils are another source for angiogenesis-regulating chemokines, growth factors and proteases. Taken together, the tumor microenvironment is a complex unorganized tissue of various cell types and extracellular matrix that can regulate the pathological angiogenic switch.


Asunto(s)
Neoplasias/irrigación sanguínea , Neoplasias/tratamiento farmacológico , Neovascularización Patológica/fisiopatología , Inhibidores de la Angiogénesis/uso terapéutico , Matriz Extracelular/patología , Humanos , Integrinas/fisiología , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/prevención & control , Neoplasias/patología , Neovascularización Patológica/prevención & control
20.
Methods Mol Biol ; 1731: 213-221, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29318556

RESUMEN

Here we describe a protocol to utilize human benign leiomyoma tissue in in vitro 3D model that enables an assessment of cell invasion. The chapter also describes detailed instructions for image analysis to quantify the results. Leiomyoma is a benign tumor of the uterus which mimics authentic components of the tumor microenvironment including fibroblasts, vessels, collagen fibers, and extracellular protein composition. The leiomyoma invasion model represents a superior 3D model for cell invasion studies compared to the other non-human organotypic models.


Asunto(s)
Procesamiento de Imagen Asistido por Computador/métodos , Leiomioma/patología , Técnicas de Cultivo de Tejidos/métodos , Técnicas de Cocultivo/instrumentación , Técnicas de Cocultivo/métodos , Progresión de la Enfermedad , Femenino , Fibroblastos , Humanos , Procesamiento de Imagen Asistido por Computador/instrumentación , Leiomioma/diagnóstico por imagen , Leiomioma/cirugía , Invasividad Neoplásica/diagnóstico por imagen , Técnicas de Cultivo de Tejidos/instrumentación , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda