Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Ann Surg Oncol ; 28(1): 244-251, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32472412

RESUMEN

BACKGROUND: Extent of tumor load is an important factor in the selection of ovarian cancer patients for cytoreductive surgery (CRS). The Peritoneal Cancer Index (PCI) gives exact information on tumor load but still is not standard in ovarian cancer surgery. The aim of this study was to find a PCI cutoff for incomplete CRS. The secondary aims were to identify reasons for open-close surgery and to compare surgical complications in relation to tumor burden. METHODS: The study included 167 women with stage III or IV ovarian cancer scheduled for CRS. Possible predictors of incomplete surgery were evaluated with receiver operator curves, and a PCI cutoff was identified. Surgical complications were analyzed by one-way analysis of variance and Chi square tests. RESULTS: The median PCI score for all the patients was 22 (range 3-37) but 33 (range 25-37) for the patients with incomplete surgery (n = 19). The PCI predicted incomplete CRS, with an area under the curve of 0.94 (95% confidence interval [CI], 0.91-0.98). Complete CRS was obtained for 67.2% of the patients with a PCI higher than 24, who experienced an increased rate of complications (p = 0.008). Overall major complications were found in 16.9% of the cases. Only 28.6% of the patients with a PCI higher than 33 achieved complete CRS. The reason for open-close surgery (n = 14) was massive carcinomatosis on the small bowel in all cases. CONCLUSION: The study found PCI to be an excellent predictor of incomplete CRS. Due to a lower surgical success rate, the authors suggest that neoadjuvant chemotherapy could be considered if the PCI is higher than 24. Preoperative radiologic assessment should focus on total tumor burden and not necessarily on specific regions.


Asunto(s)
Hipertermia Inducida , Neoplasias Ováricas , Neoplasias Peritoneales , Terapia Combinada , Procedimientos Quirúrgicos de Citorreducción , Femenino , Humanos , Neoplasias Ováricas/cirugía , Neoplasias Peritoneales/cirugía , Estudios Retrospectivos , Tasa de Supervivencia
2.
Cell Mol Life Sci ; 69(2): 325-33, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21739232

RESUMEN

DNA aneuploidy has been identified as a prognostic factor in the majority of epithelial malignancies. We aimed at identifying ploidy-associated protein expression in endometrial cancer of different prognostic subgroups. Comparison of gel electrophoresis-based protein expression patterns between normal endometrium (n = 5), diploid (n = 7), and aneuploid (n = 7) endometrial carcinoma detected 121 ploidy-associated protein forms, 42 differentially expressed between normal endometrium and diploid endometrioid carcinomas, 37 between diploid and aneuploid endometrioid carcinomas, and 41 between diploid endometrioid and aneuploid uterine papillary serous cancer. Proteins were identified by mass spectrometry and evaluated by Ingenuity Pathway Analysis. Targets were confirmed by liquid chromatography/mass spectrometry. Mass spectrometry identified 41 distinct polypeptides and pathway analysis resulted in high-ranked networks with vimentin and Nf-κB as central nodes. These results identify ploidy-associated protein expression differences that overrule histopathology-associated expression differences and emphasize particular protein networks in genomic stability of endometrial cancer.


Asunto(s)
Carcinoma Endometrioide/genética , Cistadenocarcinoma Seroso/genética , Neoplasias Endometriales/genética , Inestabilidad Genómica , Anciano , Anciano de 80 o más Años , Aneuploidia , Carcinoma Endometrioide/metabolismo , Carcinoma Endometrioide/patología , Cistadenocarcinoma Seroso/metabolismo , Cistadenocarcinoma Seroso/patología , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Endometrio/metabolismo , Endometrio/patología , Femenino , Humanos , Persona de Mediana Edad , FN-kappa B/metabolismo , Análisis por Matrices de Proteínas , Proteómica , Vimentina/metabolismo
3.
Cancers (Basel) ; 14(7)2022 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-35406529

RESUMEN

BACKGROUND: Ovarian cancer is the eighth most common cancer among women and has a 5-year survival of only 30-50%. The survival is close to 90% for patients in stage I but only 20% for patients in stage IV. The presently available biomarkers have insufficient sensitivity and specificity for early detection and there is an urgent need to identify novel biomarkers. METHODS: We employed the Explore PEA technology for high-precision analysis of 1463 plasma proteins and conducted a discovery and replication study using two clinical cohorts of previously untreated patients with benign or malignant ovarian tumours (N = 111 and N = 37). RESULTS: The discovery analysis identified 32 proteins that had significantly higher levels in malignant cases as compared to benign diagnoses, and for 28 of these, the association was replicated in the second cohort. Multivariate modelling identified three highly accurate models based on 4 to 7 proteins each for separating benign tumours from early-stage and/or late-stage ovarian cancers, all with AUCs above 0.96 in the replication cohort. We also developed a model for separating the early-stage from the late-stage achieving an AUC of 0.81 in the replication cohort. These models were based on eleven proteins in total (ALPP, CXCL8, DPY30, IL6, IL12, KRT19, PAEP, TSPAN1, SIGLEC5, VTCN1, and WFDC2), notably without MUCIN-16. The majority of the associated proteins have been connected to ovarian cancer but not identified as potential biomarkers. CONCLUSIONS: The results show the ability of using high-precision proteomics for the identification of novel plasma protein biomarker candidates for the early detection of ovarian cancer.

4.
Eur J Surg Oncol ; 47(11): 2915-2924, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34053777

RESUMEN

INTRODUCTION: prediction and importance of severe postoperative complications after ovarian cancer surgery is a strong issue in patient selection and evaluation. Pre- and early peroperative predictors of severe 30-days postoperative complications (Clavien-Dindo class ≥3) after surgery for primary ovarian cancer are not fully established, neither their impact on patients' survival. MATERIALS AND METHODS: A prospective observational study included 256 patients with primary ovarian cancer FIGO stages IIB-IV, operated during 2009-2018 in a primary or interval debulking surgery setting. Patient variables were analysed in relation to severe postoperative complications (Clavien-Dindo class ≥3) and overall survival. RESULTS: High-grade postoperative complications occurred in 24.2% patients. Class 3a complications were observed in 12.5% cases. High-grade complications class ≥3 were observed in 31.6% after primary debulking surgery compared to 12.2% after interval debulking surgery (p = 0.0004). Peritoneal cancer index ≥21 and preoperative albumin concentration ≤33 g/L were independent predictors of high-grade complications. Peritoneal cancer index correlated with the surgical complexity score and completeness of cytoreduction. Increased peritoneal cancer index was a negative predictor of overall survival, but high-grade complications did not influence survival negatively. CONCLUSIONS: Peritoneal cancer index ≥21 was an independent predictor of high-grade complications after ovarian cancer surgery. Increased peritoneal cancer index also impacted overall survival negatively, but high-grade complications did not influence overall survival.


Asunto(s)
Procedimientos Quirúrgicos de Citorreducción , Neoplasias Ováricas/cirugía , Neoplasias Peritoneales/patología , Complicaciones Posoperatorias/patología , Anciano , Femenino , Humanos , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Neoplasias Ováricas/mortalidad , Neoplasias Peritoneales/mortalidad , Complicaciones Posoperatorias/mortalidad , Estudios Prospectivos , Factores de Riesgo , Tasa de Supervivencia
5.
Nanoscale ; 11(20): 10023-10033, 2019 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-31086875

RESUMEN

Protein contents in platelets are frequently changed upon tumor development and metastasis. However, how cancer cells can influence protein-selective redistribution and release within platelets, thereby promoting tumor development, remains largely elusive. With fluorescence-based super-resolution stimulated emission depletion (STED) imaging we reveal how specific proteins, implicated in tumor progression and metastasis, re-distribute within platelets, when subject to soluble activators (thrombin, adenosine diphosphate and thromboxane A2), and when incubated with cancer (MCF-7, MDA-MB-231, EFO21) or non-cancer cells (184A1, MCF10A). Upon cancer cell incubation, the cell-adhesion protein P-selectin was found to re-distribute into circular nano-structures, consistent with accumulation into the membrane of protein-storing alpha-granules within the platelets. These changes were to a significantly lesser extent, if at all, found in platelets incubated with normal cells, or in platelets subject to soluble platelet activators. From these patterns, we developed a classification procedure, whereby platelets exposed to cancer cells, to non-cancer cells, soluble activators, as well as non-activated platelets all could be identified in an automatic, objective manner. We demonstrate that STED imaging, in contrast to electron and confocal microscopy, has the necessary spatial resolution and labelling efficiency to identify protein distribution patterns in platelets and can resolve how they specifically change upon different activations. Combined with image analyses of specific protein distribution patterns within the platelets, STED imaging can thus have a role in future platelet-based cancer diagnostics and therapeutic monitoring. The presented approach can also bring further clarity into fundamental mechanisms for cancer cell-platelet interactions, and into non-contact cell-to-cell interactions in general.


Asunto(s)
Plaquetas/metabolismo , Microscopía Fluorescente , Plaquetas/citología , Plaquetas/ultraestructura , Línea Celular Tumoral , Técnicas de Cocultivo , Fibrinógeno/química , Fibrinógeno/metabolismo , Proteínas de Choque Térmico/química , Proteínas de Choque Térmico/metabolismo , Humanos , Nanoestructuras/química , Selectina-P/química , Selectina-P/metabolismo , Factor A de Crecimiento Endotelial Vascular/química , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Mol Biol Cell ; 16(10): 4765-80, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16055503

RESUMEN

Transforming growth factor-beta (TGFbeta) signaling involves activation of a number of signaling pathways, several of which are controlled by phosphorylation events. Here, we describe a phosphoproteome profiling of MCF-7 human breast epithelial cells treated with TGFbeta1. We identified 32 proteins that change their phosphorylation upon treatment with TGFbeta1; 26 of these proteins are novel targets of TGFbeta1. We show that Smad2 and Smad3 have different effects on the dynamics of TGFbeta1-induced protein phosphorylation. The identified proteins belong to nine functional groups, e.g., proteins regulating RNA processing, cytoskeletal rearrangements, and proteasomal degradation. To evaluate the proteomics findings, we explored the functional importance of TGFbeta1-dependent phosphorylation of one of the targets, i.e., transcription factor-II-I (TFII-I). We confirmed that TGFbeta1 stimulated TFII-I phosphorylation at serine residues 371 and 743. Abrogation of the phosphorylation by replacement of Ser371 and Ser743 with alanine residues resulted in enhanced complex formation between TFII-I and Smad3, and enhanced cooperation between TFII-I and Smad3 in transcriptional regulation, as evaluated by a microarray-based measurement of expression of endogenous cyclin D2, cyclin D3, and E2F2 genes, and by a luciferase reporter assay. Thus, TGFbeta1-dependent phosphorylation of TFII-I may modulate TGFbeta signaling at the transcriptional level.


Asunto(s)
Proteoma/metabolismo , Proteína smad3/metabolismo , Factores de Transcripción TFII/fisiología , Factor de Crecimiento Transformador beta/fisiología , Sustitución de Aminoácidos , Línea Celular Tumoral , Ciclina D2 , Ciclina D3 , Ciclinas/metabolismo , Factor de Transcripción E2F2/metabolismo , Humanos , Fosforilación , Unión Proteica , Transducción de Señal , Proteína Smad2/metabolismo , Transcripción Genética , Factor de Crecimiento Transformador beta1
7.
Biomark Res ; 6: 2, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29344361

RESUMEN

BACKGROUND: Platelets support cancer growth and spread making platelet proteins candidates in the search for biomarkers. METHODS: Two-dimensional (2D) gel electrophoresis, Partial Least Squares Discriminant Analysis (PLS-DA), Western blot, DigiWest. RESULTS: PLS-DA of platelet protein expression in 2D gels suggested differences between the International Federation of Gynaecology and Obstetrics (FIGO) stages III-IV of ovarian cancer, compared to benign adnexal lesions with a sensitivity of 96% and a specificity of 88%. A PLS-DA-based model correctly predicted 7 out of 8 cases of FIGO stages I-II of ovarian cancer after verification by western blot. Receiver-operator curve (ROC) analysis indicated a sensitivity of 83% and specificity of 76% at cut-off >0.5 (area under the curve (AUC) = 0.831, p < 0.0001) for detecting these cases. Validation on an independent set of samples by DigiWest with PLS-DA differentiated benign adnexal lesions and ovarian cancer, FIGO stages III-IV, with a sensitivity of 70% and a specificity of 83%. CONCLUSION: We identified a group of platelet protein biomarker candidates that can quantify the differential expression between ovarian cancer cases as compared to benign adnexal lesions.

8.
Oncotarget ; 9(5): 5716-5735, 2018 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-29464029

RESUMEN

The multifunctional cytokine transforming growth factor ß (TGFß) controls homeostasis and disease during embryonic and adult life. TGFß alters epithelial cell differentiation by inducing epithelial-mesenchymal transition (EMT), which involves downregulation of several cell-cell junctional constituents. Little is understood about the mechanism of tight junction disassembly by TGFß. We found that one of the newly identified gene targets of TGFß, encoding the serine/threonine kinase salt-inducible kinase 1 (SIK), controls tight junction dynamics. We provide bioinformatic and biochemical evidence that SIK can potentially phosphorylate the polarity complex protein Par3, an established regulator of tight junction assembly. SIK associates with Par3, and induces degradation of Par3 that can be prevented by proteasomal and lysosomal inhibition or by mutation of Ser885, a putative phosphorylation site on Par3. Functionally, this mechanism impacts on tight junction downregulation. Furthermore, SIK contributes to the loss of epithelial polarity and examination of advanced and invasive human cancers of diverse origin displayed high levels of SIK expression and a corresponding low expression of Par3 protein. High SIK mRNA expression also correlates with lower chance for survival in various carcinomas. In specific human breast cancer samples, aneuploidy of tumor cells best correlated with cytoplasmic SIK distribution, and SIK expression correlated with TGFß/Smad signaling activity and low or undetectable expression of Par3. Our model suggests that SIK can act directly on the polarity protein Par3 to regulate tight junction assembly.

9.
Oncogene ; 24(14): 2289-97, 2005 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-15735739

RESUMEN

Inactivation of the BRCA1 gene has been found to confer susceptibility to early-onset familial breast and ovarian cancers. BRCA1 regulates DNA repair, chromatin remodeling and affects gene transcription. Transforming growth factor-beta (TGFbeta) is a potent regulator of growth, apoptosis and invasiveness of tumor cells, including breast cancer cells. Here we show that Smad3 which is a component of the TGFbeta signaling pathway, forms a complex with BRCA1 in vitro and in vivo. The interaction is mediated by the MH1 domain of Smad3 and the C-terminal part of BRCA1. We observed a co-localization of Smad3 and BRCA1 in nuclear complexes. We also found that TGFbeta1/Smad3 counteracted BRCA1-dependent repair of DNA double-strand breaks in human breast epithelial cells, as evaluated by BRCA1 nuclear foci formation, single-cell gel electrophoresis and cell survival assays. Thus, TGFbeta1/Smad3 suppresses BRCA1-dependent DNA repair in response to a DNA damaging agent.


Asunto(s)
Daño del ADN , Reparación del ADN/fisiología , Proteínas de Unión al ADN/fisiología , Genes BRCA1 , Transactivadores/fisiología , Factor de Crecimiento Transformador beta/fisiología , Western Blotting , Línea Celular , Supervivencia Celular , Humanos , Inmunohistoquímica , Inmunoprecipitación , Proteína smad3 , Transcripción Genética , Factor de Crecimiento Transformador beta1
10.
FEBS Lett ; 577(1-2): 93-100, 2004 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-15527767

RESUMEN

Smad3 is an important component of transforming growth factor-beta (TGFbeta) intracellular signalling. To identify novel interacting proteins of Smad3, we performed pull-down assays with Smad3 constructs fused to glutathione-S-transferase. Proteins which formed complexes with these constructs were analyzed by two-dimensional gel electrophoresis, and were identified by matrix-assisted laser desorption-ionization time-of-flight mass spectrometry. We identified 14 proteins interacting with the Smad3 construct lacking the N-terminal Mad homology domain 1 (MH1), and 12 proteins interacting with the construct lacking the C-terminal MH2 domain. Proteins involved in signalling processes, in metabolism regulation, novel proteins, and components of cytoskeleton form four groups of interacting proteins. Interactions of AGP7, sex-determining region Y protein, actin beta and sterol regulatory element binding protein-2 (SREBP-2) proteins with Smad3 constructs were confirmed by immunoblotting with specific antibodies. Interaction of Smad3 with SREBP-2 was also confirmed by co-immunoprecipitation of myc-Smad3 and Flag-SREBP-2 upon expression in mammalian cells. We found that SREBP-2 inhibited the transcriptional activity of Smad3 in luciferase reporter assays.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteoma , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Electroforesis en Gel Bidimensional , Humanos , Unión Proteica , Proteína smad3 , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Proteína 2 de Unión a Elementos Reguladores de Esteroles , Transcripción Genética
12.
Proteomics Clin Appl ; 4(1): 17-31, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21137014

RESUMEN

PURPOSE: Cervical cancer is the second most prevalent malignancy of women. Our aim was to identify additional marker protein patterns for objective diagnosis of squamous cervical cancer (SCC). EXPERIMENTAL DESIGN: Collected tissue biopsies of SCC, squamous vaginal cancer (SVC), normal cervical and vaginal mucosa were subjected to 2-DE, SameSpot analysis, MALDI-TOF-MS protein identification, and analysis of the expression of selected proteins by immunohistochemistry. RESULTS: In 148 protein spots selected by the difference in expression 99 proteins were identified. A differential protein pattern for SCC was, e.g. over-expressed (OE) eukaryotic translation initiation factor 3-2ß, neutrophil cytosolic factor 2, annexin A6 (ANXA6), for SVC it was OE cathepsin D, γ-catenin, RAB2A, for both cancers it was OE apolipoprotein E, tropomyosin 3, HSPA8, and underexpressed cytokeratin 13, osteoglycin. In SCC nuclear expression of neutrophil cytosolic factor 2, PRDX2, HSP27 (nine of ten cases), ANXA6 (nine of ten cases) was observed while tropomyosin 4 was expressed only in two of ten cases. There was 81.1% (43/53) agreement between the expression of protein spots and the immune expression of proteins (www.proteinatlas.org). CONCLUSIONS AND CLINICAL RELEVANCE: SCC is characterized by specific tissue marker protein patterns that allow objective detection of the disease. They can become a basis for objective automated cytology-based screening and improve current diagnostics of SCC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de Células Escamosas/diagnóstico , Neoplasias de Células Escamosas/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Neoplasias del Cuello Uterino/diagnóstico , Neoplasias del Cuello Uterino/metabolismo , Biomarcadores de Tumor/aislamiento & purificación , Electroforesis en Gel Bidimensional , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias de Células Escamosas/genética , Proteoma/aislamiento & purificación , Reproducibilidad de los Resultados , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Neoplasias del Cuello Uterino/genética
13.
Proteomics Clin Appl ; 1(9): 1090-101, 2007 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21136760

RESUMEN

Breast, ovarian, endometrial and cervical cancers are diseases with a relatively high lethal outcome, despite recent development of surgical, chemotherapeutic, radiotherapeutic and anti-hormonal treatments. The mortality of women with breast cancer is at the level of 20%, while for ovarian cancer it is up to 75%. For cervical cancer it is 38% and for endometrial cancer 17%. To enhance survival of patients, better diagnostics of these cancers in early and non-invasive stages is essential. Proteomics allows a comprehensive analysis of hundreds of proteins in clinical samples. Studies of tissue samples and body fluids, e.g., blood and plasma, have shown their usefulness in discovery of markers of cancers at early stages. Searches for markers of breast and ovarian cancers have been performed with promising results. Studies of endometrial and cervical cancer markers have been less extensive. This review describes recent reports of markers for early detection of breast, ovarian, endometrial and cervical cancers. A clinical evaluation of the markers discovered with the use of proteomics techniques is discussed.

14.
Int J Cancer ; 118(2): 412-21, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16049973

RESUMEN

Plasma samples of ovarian and breast cancer patients were used to search for markers of cancer using 2-dimensional gel electrophoresis and matrix-assisted laser desorption/ionization time of flight mass spectrometry. Truncated forms of cytosolic serine hydroxymethyl transferase (cSHMT), T-box transcription factor 3 (Tbx3) and utrophin were aberrantly expressed in samples from cancer patients as compared to samples from noncancerous cases. Aberrant expression of proteins was validated by immunoblotting of plasma samples with specific antibodies to cSHMT, Tbx3 and utrophin. A cohort of 79 breast and 39 ovarian cancer patients and 31 individuals with noncancerous conditions was studied. We observed increased expression of truncated cSHMT, Tbx3 and utrophin in plasma samples obtained from patients at early stages of disease. Our data suggest that cSHMT, Tbx3 and utrophin can be used as components of multiparameter monitoring of ovarian and breast cancer (supplementary material for this article can be found on the International Journal of Cancer website at http://www.interscience.wiley.com/jpages/0020-7136/suppmat/index.html).


Asunto(s)
Neoplasias de la Mama/patología , Glicina Hidroximetiltransferasa/sangre , Neoplasias Ováricas/sangre , Proteínas de Dominio T Box/sangre , Utrofina/sangre , Biomarcadores de Tumor/sangre , Estudios de Casos y Controles , Citosol/química , Electroforesis en Gel Bidimensional , Femenino , Perfilación de la Expresión Génica , Glicina Hidroximetiltransferasa/biosíntesis , Humanos , Espectrometría de Masas , Estadificación de Neoplasias/métodos , Proteínas de Dominio T Box/biosíntesis , Utrofina/biosíntesis
15.
Proteomics ; 4(4): 995-1006, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15048981

RESUMEN

Transforming growth factor-beta (TGFbeta) is a potent regulator of angiogenesis affecting proliferation, differentiation and migration of endothelial cells. The effect of TGFbeta on endothelial cells depends on the origin of the cells and on the experimental conditions. Global analysis of TGFbeta signalling is expected to unveil mechanisms of this variability and identify novel targets of the growth factor. Here, we report proteome profiling of human microvascular endothelial cells obtained from dermis, which were treated with TGFbeta1 and compared to nontreated cells. We identified 54 proteins affected by TGFbeta1 using two-dimensional gel electrophoresis and peptide mass fingerprinting. Thirteen of the identified proteins are involved in various signalling processes. Seven proteins are involved in cytoskeleton rearrangements and six are involved in regulation of metabolism. Ten proteins were identical to predicted hypothetical proteins with no assigned functions. In agreement with the effect of TGFbeta1 on components of the cytoskeleton, TGFbeta1 induces actin cytoskeleton rearrangements. TGFbeta1 also affected expression of E2F6, p57Kip2, G(q)alpha, hnRNP A1 and myosin light chain proteins as shown by immunoblotting. Down-regulation of the transcriptional repressor E2F6 by TGFbeta1 correlated with a weak growth-inhibitory activity of TGFbeta1 on HMVEC-d cells. Twenty-five of the identified proteins have not previously been described as being regulated by TGFbeta1, providing new insights into TGFbeta1 signalling in endothelial cells.


Asunto(s)
Citoesqueleto/metabolismo , Células Endoteliales/metabolismo , Proteoma , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Células Cultivadas , Dermis , Electroforesis en Gel Bidimensional , Humanos , Mapeo Peptídico , Factor de Crecimiento Transformador beta1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA