Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Reprod Immunol ; 165: 104297, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39029322

RESUMEN

Recurrent pregnancy loss (RPL) affects 1-2 % of all couples trying to conceive and is a challenging heterogeneous condition. This study aimed to evaluate the prevalence and impact of various risk factors in patients suffering from RPL. We performed a prospective cohort study including patients at the tertiary RPL Unit in the Capital Region of Denmark between 1st January 2000 and 1st January 2023. The main outcome of the study was the first pregnancy after referral and whether the pregnancy was ongoing at least to the 22nd gestational week. A total of 2555 patients were included in the study, out of whom 1892 patients achieved a pregnancy after referral to the RPL Unit. This resulted in 1103 live births (58.3 %) and 718 pregnancy losses (37.9 %). Maternal age, BMI, smoking status and the number of prior pregnancy losses were negatively correlated with the likelihood of achieving pregnancy. Furthermore, maternal age, prior pregnancy losses, antiphospholipid syndrome (APS) and uterine malformations were associated with reduced birth rates. Patients with secondary RPL had a higher birth rate compared to those with primary RPL, and patients with APS treated with low-molecular-weight heparin (LMWH) demonstrated a significantly increased birth rate compared to untreated APS patients. These findings suggest that certain risk factors significantly impact the likelihood of achieving pregnancy and live birth following RPL, which can be used in patient guidance.


Asunto(s)
Aborto Habitual , Nacimiento Vivo , Humanos , Femenino , Embarazo , Aborto Habitual/epidemiología , Aborto Habitual/inmunología , Adulto , Estudios Prospectivos , Factores de Riesgo , Nacimiento Vivo/epidemiología , Síndrome Antifosfolípido/epidemiología , Síndrome Antifosfolípido/inmunología , Edad Materna , Dinamarca/epidemiología , Resultado del Embarazo/epidemiología , Heparina de Bajo-Peso-Molecular/uso terapéutico , Tasa de Natalidad , Estudios de Cohortes
2.
Microb Risk Anal ; 22: 100235, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36248679

RESUMEN

From march 2020 to march 2022 covid-19 has shown a consistent pattern of increasing infections during the Winter and low infection numbers during the Summer. Understanding the effects of seasonal variation on covid-19 spread is crucial for future epidemic modelling and management. In this study, seasonal variation in the transmission rate of covid-19, was estimated based on an epidemic population model of covid-19 in Denmark, which included changes in national restrictions and introduction of the α -variant covid-19 strain, in the period March 2020 - March 2021. Seasonal variation was implemented as a logistic temperature dependent scaling of the transmission rate, and parameters for the logistic relationship was estimated through rejection-based approximate bayesian computation (ABC). The likelihoods used in the ABC were based on national hospital admission data and seroprevalence data stratified into nine and two age groups, respectively. The seasonally induced reduction in the transmission rate of covid-19 in Denmark was estimated to be 27 % , (95% CI [ 24 % ; 31 % ]), when moving from peak Winter to peak Summer. The reducing effect of seasonality on transmission rate per + 1 ∘ C in daily average temperature were shown to vary based on temperature, and were estimated to be - 2.2 % [ - 2.8 % ; - 1.7 % ] pr. 1  ∘ C around 2 ∘ C; 2 % [ - 2.3 % ; - 1.7 % ] pr. 1  ∘ C around 7 ∘ C; and 1.7 % [ - 2.0 % ; - 1.5 % ] pr. 1  ∘ C around a daily average temperature of 11  ∘ C.

3.
Mol Cancer Ther ; 7(9): 2758-67, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18790756

RESUMEN

Matrix metalloproteinases (MMP) have several roles that influence cancer progression and dissemination. However, low molecular weight metalloproteinase inhibitors (MPI) have not yet been tested in transgenic/spontaneous metastasis models. We have tested Galardin/GM6001, a potent MPI that reacts with most MMPs, in the MMTV-PymT transgenic breast cancer model. We followed a cohort of 81 MMTV-PymT transgenic mice that received Galardin, placebo, or no treatment. Galardin treatment was started at age 6 weeks with 100 mg/kg/d, and all mice were killed at age 13.5 weeks. Galardin treatment significantly reduced primary tumor growth. Final tumor burden in Galardin-treated mice was 1.69 cm3 compared with 3.29 cm3 in placebo-treated mice (t test, P = 0.0014). We quantified the total lung metastasis volume in the same cohort of mice. The median metastasis volume was 0.003 mm(3) in Galardin-treated mice compared with 0.56 mm(3) in placebo-treated mice (t test, P < 0.0001). Thus, metastasis burden was reduced more than 100-fold, whereas primary tumor size was reduced only 2-fold. We also found that primary tumors from Galardin-treated mice exhibited a lower histopathologic tumor grade, increased collagen deposition, and increased MMP-2 activity. MMPs are known to have tumor-promoting and tumor-inhibitory effects, and several clinical trials of broad-spectrum MPIs have failed to show promising effects. The very potent antimetastatic effect of Galardin in the MMTV-PymT model does, however, show that it may be possible to find broad-spectrum MPIs with favorable inhibition profiles, or perhaps combinations of monospecific MPIs, for future clinical application.


Asunto(s)
Dipéptidos/farmacología , Neoplasias Pulmonares/secundario , Metástasis Linfática/patología , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/patología , Virus del Tumor Mamario del Ratón/fisiología , Inhibidores de la Metaloproteinasa de la Matriz , Animales , Proliferación Celular , Colágeno/metabolismo , Dipéptidos/química , Dipéptidos/uso terapéutico , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Neoplasias Mamarias Experimentales/genética , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Transgénicos , Carga Tumoral
4.
Oncogene ; 22(28): 4389-97, 2003 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-12853975

RESUMEN

The plasminogen activator inhibitor-1 (PAI-1) blocks the activation of plasmin(ogen), an extracellular protease vital to cancer invasion. PAI-1 is like the corresponding plasminogen activator uPA (urokinase-type plasminogen activator) consistently expressed in human breast cancer. Paradoxically, high levels of PAI-1 as well as uPA are equally associated with poor prognosis in cancer patients. PAI-1 is thought to play a vital role for the controlled extracellular proteolysis during tumor neovascularization. We have studied the effect of PAI-1 deficiency in a transgenic mouse model of metastasizing breast cancer. In these tumors, the expression pattern of uPA and PAI-1 resembles that of human ductal breast cancer and plasminogen is required for efficient metastasis. In a cohort of 63 transgenic mice that were either PAI-1-deficient or wild-type sibling controls, primary tumor growth and vascular density were unaffected by PAI-1 status. PAI-1 deficiency also did not significantly affect the lung metastatic burden. These results agree with the virtual lack of spontaneous phenotype in PAI-1-deficient mice and humans and may reflect that the plasminogen activation reaction is not rate limiting for tumor vascularization and metastasis, or that there is a functional redundancy between PAI-1 and other inhibitors of the uPA/plasmin system, masking the effect of PAI-1 deficiency.


Asunto(s)
Neoplasias Mamarias Experimentales/irrigación sanguínea , Neoplasias Mamarias Experimentales/patología , Neovascularización Patológica/etiología , Inhibidor 1 de Activador Plasminogénico/fisiología , Animales , Femenino , Neoplasias Pulmonares/secundario , Masculino , Virus del Tumor Mamario del Ratón , Ratones , Ratones Transgénicos , Neovascularización Patológica/prevención & control , Inhibidor 1 de Activador Plasminogénico/deficiencia , Inhibidor 2 de Activador Plasminogénico/análisis , Activador de Plasminógeno de Tipo Uroquinasa/análisis
5.
Oncogene ; 22(25): 3964-76, 2003 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-12813470

RESUMEN

Keratinocytes undergo a dramatic phenotypic conversion during reepithelialization of skin wounds to become hyperproliferative, migratory, and invasive. This transient healing response phenotypically resembles malignant transformation of keratinocytes during squamous cell carcinoma progression. Here we present the first analysis of global changes in keratinocyte gene expression during skin wound healing in vivo, and compare these changes to changes in gene expression during malignant conversion of keratinized epithelium. Laser capture microdissection was used to isolate RNA from wound keratinocytes from incisional mouse skin wounds and adjacent normal skin keratinocytes. Changes in gene expression were determined by comparative cDNA array analyses, and the approach was validated by in situ hybridization. The analyses identified 48 candidate genes not previously associated with wound reepithelialization. Furthermore, the analyses revealed that the phenotypic resemblance of wound keratinocytes to squamous cell carcinoma is mimicked at the level of gene expression, but notable differences between the two tissue-remodeling processes were also observed. The combination of laser capture microdissection and cDNA array analysis provides a powerful new tool to unravel the complex changes in gene expression that underlie physiological and pathological remodeling of keratinized epithelium.


Asunto(s)
Carcinoma de Células Escamosas/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Queratinocitos/metabolismo , Neoplasias Cutáneas/genética , Piel/lesiones , Cicatrización de Heridas/genética , Animales , Carcinoma de Células Escamosas/patología , Separación Celular/instrumentación , Separación Celular/métodos , Transformación Celular Neoplásica/genética , Epidermis/lesiones , Perfilación de la Expresión Génica/métodos , Hibridación in Situ , Rayos Láser , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , ARN Mensajero/biosíntesis , ARN Neoplásico/genética , Reproducibilidad de los Resultados , Piel/patología , Neoplasias Cutáneas/patología
6.
Thromb Haemost ; 93(4): 676-81, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15841311

RESUMEN

Breakdown of the extracellular matrix is crucial for cancer invasion and metastasis. It is accomplished by the concerted action of several proteases, including the serine protease plasmin and a number of matrix metalloproteases. The activity of each of these proteases is regulated by an array of activators, inhibitors and cellular receptors. Thus, the generation of plasmin involves the pro-enzyme plasminogen, the urokinase type plasminogen activator uPA and its pro-enzyme pro-uPA, the uPA inhibitor PAI-1, the cell surface uPA receptor uPAR, and the plasmin inhibitor alpha(2)-antiplasmin. Furthermore, the regulation of extracellular proteolysis in cancer involves a complex interplay between cancer cells and non-malignant stromal cells in the expression of the molecular components involved. For some types of cancer, this cellular interplay mimics that observed in the tissue of origin during non-neoplastic tissue remodelling processes. We propose that cancer invasion can be considered as uncontrolled tissue remodelling. Inhibition of extracellular proteases is an attractive approach to cancer therapy. Because proteases have many different functions in the normal organism, efficient inhibition will have toxic side effects. In cancer invasion, like in normal tissue remodelling processes, there appears to be a functional overlap between different extracellular proteases. This redundancy means that combinations of protease inhibitors must be used. Such combination therapy, however, is also likely to increase toxicity. Therefore for each type of cancer, a combination of protease inhibitors that is optimised with respect to both maximal therapeutic effect and minimal toxic side effects need to be identified.


Asunto(s)
Neoplasias/patología , Plasminógeno/metabolismo , Humanos , Invasividad Neoplásica , Neoplasias/etiología , Neoplasias/terapia , Plasminógeno/fisiología , Activadores Plasminogénicos/fisiología , Células del Estroma/metabolismo
7.
Recent Results Cancer Res ; 162: 31-42, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12790319

RESUMEN

Solid tumors co-opt the body's endogenous extracellular proteolytic machinery for their invasion and metastasis. This is supported by a large number of independent observations ranging from histochemical and prognostic studies of cancer patient material to animal experiments. There are several extracellular proteolytic systems that are relevant in the context of cancer, but the plasminogen activation (PA) system and the matrix metalloproteases (MMPs) remain the most thoroughly investigated. Localization studies by immunohistochemistry and in situ mRNA hybridization in tumors of common human cancers have repeatedly identified members of the PA and MMP systems in stromal cells. The cancer cells, of epithelial origin, contribute PA and MMP components in some cases, but their contribution fades in comparison with the overwhelming expression of proteolytic components by fibroblasts, macrophages, endothelial cells, and other stromal cells. Ideal animal models of human cancers should recapitulate this fundamental proteolytic aspect of tumor biology. However, in the transplantable tumor models where PA or MMP components have been studied at the cellular level in vivo, this is most often not the case. Transgenic cancer models may provide a closer parallel to the human situation, in that PA and MMP components are synthesized by the tumor stroma. The pivotal role of stromal cells has been confirmed experimentally in mouse models in which the expression pattern of proteolytic components is strongly reminiscent of human tumors. In these models it is possible to reconstitute the wild-type tumor characteristics of proteolytically deficient tumor-bearing mice by transplantation with wild-type fibroblasts or hemapoietic cells. These studies collectively show that cancer-associated proteolysis is a collaborative effort of malignant cancer cells and various stromal cells--a collaboration in which stromal cells contribute the majority of the active proteolytic components that are necessary for the invasive behavior of the tumors. This cellular division of labor positions the stromal cells as prime targets for future research and possibly therapy. Vascular endothelial cells are already the focus of intense therapeutically relevant research, but tumor-associated fibroblasts, macrophages, neutrophils, lymphendothelial cells, etc. provide additional largely unexplored territory in the ongoing search for efficient countermeasures against invasive cancer.


Asunto(s)
Neoplasias/metabolismo , Células del Estroma/fisiología , Animales , Progresión de la Enfermedad , Humanos , Neoplasias/patología
8.
Clin Exp Metastasis ; 30(3): 277-88, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22996753

RESUMEN

Plasminogen (Plg) plays a central role in tissue remodeling during ontogeny, development, and in pathological tissue remodeling following physical injury, inflammation and cancer. Plg/plasmin is, however, not critical for these processes, as they all occur to a varying extent in its absence, suggesting that there is a functional redundancy with other proteases. To explore this functional overlap in the transgenic MMTV-PyMT breast cancer metastasis model, we have combined Plg deficiency and a pharmacological metalloprotease inhibitor, which is known to reduce metastasis in this model, and has been shown to synergistically inhibit other tissue remodeling events in Plg-deficient mice. While metalloprotease inhibition dramatically reduced metastasis, we found no effect of Plg deficiency on metastasis, either independently or in combination with metalloprotease inhibition. We further show that Plg gene deficiency is of no significant consequence in this metastasis model, when analyzed in two different congenic strains: the FVB strain, and a F1 hybrid of the FVB and C57BL/6J strains. We suggest that the extensive backcrossing performed prior to our studies has eliminated the confounding effect of a known polymorphic metastasis modifier gene region located adjacent to the Plg gene.


Asunto(s)
Eliminación de Gen , Neoplasias Mamarias Experimentales/patología , Plasminógeno/genética , Animales , Femenino , Neoplasias Mamarias Experimentales/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Metástasis de la Neoplasia
9.
J Invest Dermatol ; 128(8): 2092-101, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18337830

RESUMEN

Genetic ablation of plasminogen (Plg) and pharmacological inhibition of metalloproteinase activity by galardin delay skin wound healing in mice, whereas the combined inhibition of these two enzyme systems completely prevents healing. In this study, the impact of plasmin and metalloproteinases as profibrinolytic enzymes has been investigated by comparing skin wound healing in the absence and presence of fibrin. Plg deficiency impairs skin wound healing kinetics, but this delay is only partially restored in the absence of fibrin. This suggests that plasmin-mediated fibrinolysis is the primary, but not the exclusive, requirement for healing of wounds in these mice. In addition, we observe that lack of fibrin reduces Plg activation significantly during wound healing. The profibrinolytic role of metalloproteinases is revealed by the finding that lack of fibrin partially restores the otherwise arrested healing of Plg-deficient wounds after metalloproteinase inhibition. In conclusion, the residual impairment of skin wound healing in the absence of fibrin suggests the existence of a fibrin-independent substrate(s) for plasmin and metalloproteinases. Furthermore, these in vivo data reveal that galardin-sensitive metalloproteinases mediate compensatory fibrinolysis to facilitate wound healing in the absence of plasmin.


Asunto(s)
Fibrinólisis/fisiología , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Plasminógeno/metabolismo , Piel/metabolismo , Cicatrización de Heridas/fisiología , Animales , Dipéptidos/farmacología , Fibrina/metabolismo , Fibrinógeno/genética , Fibrinógeno/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Metaloproteinasa 13 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/genética , Inhibidores de la Metaloproteinasa de la Matriz , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Plasminógeno/genética , Inhibidores de Proteasas/farmacología , Piel/patología
10.
Int J Cancer ; 118(5): 1241-7, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16152618

RESUMEN

We recently undertook expression profiling of all 19 human ADAMTS metalloproteinases (a disintegrin and metalloproteinase with thrombospondin motifs) in malignant and non-neoplastic breast tissue and showed that 11 of the ADAMTS genes are dysregulated in breast carcinoma. We identified a subgroup of ADAMTSs, based on functional and amino acid sequence similarity (ADAMTS1, 4, 5, 8 and 15), to be the focus of further study in breast carcinoma. Further RNA expression analysis by real-time PCR on a different cohort of 229 patients with breast cancer has identified ADAMTS8 as a predictor of poor overall survival (OS) (hazard ratio (HR) = 2.20, 95% C.I. = 1.29-3.74, p = 0.004) and confirmed ADAMTS15 as a predictor of prolonged relapse-free survival (RFS) (HR = 0.54, 95% C.I. = 0.32-0.89, p = 0.016). We explored the differences in survival of the 4 groups that could be categorized based on the expression levels of ADAMTS8 and ADAMTS15. For both RFS and OS, the group with high ADAMTS8 and low ADAMTS15 expression had a particularly poor prognosis. This group had a 3-fold higher chance of recurrence (HR = 3.03, 95% C.I. = 1.49-6.15, p = 0.001) and a greater than 5-fold higher chance of death (HR = 5.40, 95% C.I. = 2.16-13.5, p < 0.001) than the most favorable prognostic group. This prediction of poor prognosis by ADAMTS8 and ADAMTS15 expression was found to be independent of other classical clinicopathological factors. Results observed in FVB-PyMT mice, a robust transgenic model of highly metastatic breast carcinoma, fitted the expectation that relatively high expression levels of ADAMTS8 together with low expression levels of ADAMTS15 seen in human breast carcinoma are associated with a poor clinical outcome. In summary, ADAMTS8 and ADAMTS15 have emerged as novel predictors of survival in patients with breast carcinoma.


Asunto(s)
Proteínas ADAM/metabolismo , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas ADAM/genética , Proteínas ADAMTS , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Humanos , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia
11.
Int J Cancer ; 113(4): 525-32, 2005 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-15472905

RESUMEN

A prominent phenotype of plasmin deficiency in mice is reduced metastasis in the MMTV-PymT transgenic breast cancer model. Proteolytically active plasmin is generated from inactive plasminogen by one of 2 activators, uPA or tPA. We now find that uPA deficiency alone significantly reduces metastasis >7-fold in the MMTV-PymT model. We studied a cohort of 55 MMTV-PymT transgenic mice, either uPA-deficient or wild-type controls. Tumor incidence, latency, growth rate and final primary tumor burden were not significantly affected by uPA deficiency. In contrast, average lung metastasis volume was reduced from 1.58 mm(3) in wild-type controls to 0.21 mm(3) in uPA-deficient mice (p = 0.023). Tumor cell dissemination to brachial lymph nodes was also reduced from 53% (28/53) in wild-type controls to 31% (17/54) in uPA-deficient mice (p = 0.032). Mice without plasminogen display a severe pleiotropic phenotype. By comparison, spontaneous phenotypes are modest in uPA-deficient mice, probably because they still have active tPA. We show that metastasis is strongly and selectively decreased in uPA-deficient mice, suggesting that uPA-directed antimetastatic therapy would be efficacious and have limited side effects.


Asunto(s)
Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Activadores Plasminogénicos/deficiencia , Activador de Plasminógeno de Tipo Uroquinasa/deficiencia , Animales , Antígenos Transformadores de Poliomavirus/genética , Femenino , Humanos , Hibridación in Situ , Incidencia , Neoplasias Pulmonares/enzimología , Metástasis Linfática/patología , Metástasis Linfática/prevención & control , Masculino , Neoplasias Mamarias Experimentales/enzimología , Virus del Tumor Mamario del Ratón/genética , Ratones , Ratones Transgénicos , Tasa de Supervivencia
12.
Int J Cancer ; 113(6): 870-80, 2005 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-15515049

RESUMEN

The plasminogen activation (PA) cascade participates in degradation of extracellular matrix during cancer invasion. We have studied the expression of urokinase-type plasminogen activator (uPA) mRNA, uPA receptor (uPAR) mRNA and immunoreactivity, and type-1 plasminogen activator inhibitor (PAI-1) mRNA and immunoreactivity in 16 prostate adenocarcinomas and 9 benign prostate hyperplasias. uPA mRNA and uPAR mRNA expression were found in 9 and 8 of the adenocarcinomas, respectively, and in 7 and 6 of the benign hyperplasias, respectively. In both malignant and benign lesions, expression of these 2 mRNAs was predominantly seen in cells identified as macrophages, which in most of the carcinomas (approximately 90%) were located in the interstitial tissue between the tumor cell islands, while in most of the benign hyperplasias they were located in the lumen of the glands and were in only a few cases (approximately 30%) found in the interstitial tissue. uPAR immunoreactivity correlated with the mRNA expression and was, in addition, found in neutrophils. PAI-1 mRNA was detected in 13 of the 16 carcinomas and in 8 of the 9 benign hyperplasias, located in scattered fibroblast-like cells in both groups, in some vascular structures and in a few macrophages located in the interstitial tissue of both malignant and benign lesions. A similar expression pattern was found for PAI-1 immunoreactivity. In 8 of the 16 carcinomas, all 3 components were present, and in several areas colocalization was observed in stromal cells in close proximity to cancer cell islands. No immunoreactivity and/or mRNA expression of uPA, uPAR or PAI-1 was observed in cancer cells or in other epithelial cells in any of the cases.


Asunto(s)
Inhibidor 1 de Activador Plasminogénico/genética , Próstata/patología , Neoplasias de la Próstata/genética , Receptores de Superficie Celular/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética , Adenocarcinoma/genética , Adenocarcinoma/patología , Carcinoma/genética , Carcinoma/patología , Humanos , Hibridación in Situ , Macrófagos/patología , Masculino , Hiperplasia Prostática/genética , Hiperplasia Prostática/patología , Neoplasias de la Próstata/patología , ARN Mensajero/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa , Transcripción Genética
13.
Carcinogenesis ; 26(7): 1233-40, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15760918

RESUMEN

Solid tumors synthesize a number of extracellular matrix-degrading proteases that are important for tumor progression. Based on qualitative in situ hybridization studies in human cancer tissue, a range of components involved in proteolysis appear to be expressed by stromal cells rather than cancer cells. We have now used laser capture microdissection and real-time PCR to quantify the mRNA expression of components of matrix-degrading proteolytic systems in cancer and stromal areas of mouse mammary tumors genetically induced by the polyoma virus middle T (PyMT) antigen. We examined the mRNA levels of urokinase plasminogen activator, plasminogen activator inhibitor 1 and the matrix metalloproteases MMP-2, -3, -11, -13 and -14, and found that all these seven genes are predominantly expressed by stromal cells. Our results were qualitatively supported by in situ hybridization analysis of the expression of mRNAs for MMP-2, -3 and -13 in the PyMT tumors. Statistical analyses indicated that the quantitative expression patterns observed in cancer and stromal cells isolated from individual tumors from different PyMT mice are quite reproducible. The methodology described in this study provides excellent tools to study the possible interactions between cancer and stromal cells during the development of breast cancer, and the results suggest that stromal cells are involved in carcinogenesis and tumor progression, which may have important implications for the biology and therapy of cancer.


Asunto(s)
Carcinoma/genética , Neoplasias Mamarias Animales/genética , Péptido Hidrolasas/biosíntesis , Animales , Carcinoma/veterinaria , Femenino , Ratones , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , Reproducibilidad de los Resultados , Células del Estroma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA