Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Eur J Public Health ; 34(3): 566-571, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38519451

RESUMEN

BACKGROUND: The objective is to estimate the importance of the decrease of smoking habits in Sweden for the occurrence of lung cancer. METHODS: The change in smoking habits in the general population was retrieved from surveys and on taxation of sale of cigarettes. We used data from the Swedish Cancer Register on incidence of lung cancer between 1970 and 2021, stratified for sex, age and cell type, and compared the occurrence overtime in ages between 40 and 84 years. RESULTS: The sale of cigarettes peaked in 1980 to 1800 cigarettes per person and decreased to 600 per person in 2021. The change in incidence rates of squamous cell cancer and other cell types varied over time, sex, and age in a pattern that partly seems to be explained by change in the prevalence of daily smokers. The incidence of adenocarcinoma was similar in men and women 1970-2021 and increased, e.g. for women and men 75-79 years of age from around 20 cases in early 1970s to around 120 cases per 100 000 person-years in the 2020s. CONCLUSIONS: Our data indicate that the risk of lung cancer several years after smoking cessation is less favourable than previously studies have indicated. There is a similar increase in the incidence of adenocarcinoma in men and women which is hard to explain only with changing smoking habits. The change from non-filter to filter cigarettes in the 1960s-1970s may be a contributing factor.


Asunto(s)
Neoplasias Pulmonares , Fumar , Humanos , Suecia/epidemiología , Neoplasias Pulmonares/epidemiología , Masculino , Femenino , Anciano , Persona de Mediana Edad , Adulto , Anciano de 80 o más Años , Incidencia , Fumar/epidemiología , Sistema de Registros , Prevalencia , Cese del Hábito de Fumar/estadística & datos numéricos
2.
Int J Cancer ; 154(10): 1814-1827, 2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38282121

RESUMEN

Adenoid cystic carcinoma (ACC) and basal cell adenoma (BCA) share many histological characteristics and often need a differential diagnosis in clinical pathology. Recently, we found homeobox protein engrailed-1 (EN1) was a potential diagnostic marker for ACC in an organoids library of salivary gland tumors (SGTs). Here we aim to confirm EN1 as a differential diagnostic marker for ACC, and further investigate the regulatory mechanism and biological function of EN1 in tumor progression. The transcriptional analysis, quantitative polymerase chain reaction, Western blot and immunohistochemistry staining were performed and revealed that EN1 was specifically and highly expressed in ACC, and accurately differentiated ACC from BCA. Furthermore, TGFß signaling pathway was found associated with ACC, and the regulation of EN1 through TGFß was detected in the human ACC cell lines and patient-derived organoids (PDOs). TGFß-induced EN1 was important in promoting tumor budding in the PDOs model. Interestingly, a high level of EN1 and TGFß1 in the budding tips was observed in ACC clinical samples, and the expression of EN1 and TGFß1 in ACC was significantly associated with the clinical stage. In summary, our study verified EN1 is a good diagnostic marker to differentiate ACC from BCA. TGFß-induced EN1 facilitates the tumor budding of ACC, which might be an important mechanism related to the malignant phenotype of ACC.


Asunto(s)
Adenoma , Carcinoma Adenoide Quístico , Neoplasias de las Glándulas Salivales , Humanos , Adenoma/patología , Biomarcadores de Tumor/genética , Carcinoma Adenoide Quístico/patología , Proteínas de Homeodominio , Neoplasias de las Glándulas Salivales/patología , Factor de Crecimiento Transformador beta
3.
bioRxiv ; 2023 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-37292721

RESUMEN

The majority of the world population carry the gastric pathogen Helicobacter pylori. Fortunately, most individuals experience only low-grade or no symptoms, but in many cases the chronic inflammatory infection develops into severe gastric disease, including duodenal ulcer disease and gastric cancer. Here we report on a protective mechanism where H. pylori attachment and accompanying chronic mucosal inflammation can be reduced by antibodies that are present in a vast majority of H. pylori carriers. These antibodies block binding of the H. pylori attachment protein BabA by mimicking BabA's binding to the ABO blood group glycans in the gastric mucosa. However, many individuals demonstrate low titers of BabA blocking antibodies, which is associated with an increased risk for duodenal ulceration, suggesting a role for these antibodies in preventing gastric disease.

4.
Sci Signal ; 15(760): eabp9521, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36378749

RESUMEN

Transforming growth factor-ß (TGF-ß) signaling has important roles during embryonic development and in tissue homeostasis. TGF-ß ligands exert cellular effects by binding to type I (TßRI) and type II (TßRII) receptors and inducing both SMAD-dependent and SMAD-independent intracellular signaling pathways, the latter of which includes the activation of the tyrosine kinase Src. We investigated the mechanism by which TGF-ß stimulation activates Src in human and mouse cells. Before TGF-ß stimulation, inactive Src was complexed with TßRII. Upon TGF-ß1 stimulation, TßRII associated with and phosphorylated TßRI at Tyr182. Binding of Src to TßRI involved the interaction of the Src SH2 domain with phosphorylated Tyr182 and the interaction of the Src SH3 domain with a proline-rich region in TßRI and led to the activation of Src kinase activity and Src autophosphorylation. TGF-ß1-induced Src activation required the kinase activities of TßRII and Src but not that of TßRI. Activated Src also phosphorylated TßRI on several tyrosine residues, which may stabilize the binding of Src to the receptor. Src activation was required for the ability of TGF-ß to induce fibronectin production and migration in human breast carcinoma cells and to induce α-smooth muscle actin and actin reorganization in mouse fibroblasts. Thus, TGF-ß induces Src activation by stimulating a direct interaction with TßRI that depends on tyrosine phosphorylation of TßRI by TßRII.


Asunto(s)
Receptores de Factores de Crecimiento Transformadores beta , Factor de Crecimiento Transformador beta1 , Humanos , Ratones , Animales , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Serina-Treonina Quinasas , Actinas , Factor de Crecimiento Transformador beta/metabolismo , Tirosina
5.
EBioMedicine ; 82: 104155, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35853811

RESUMEN

BACKGROUND: Transforming growth factor ß (TGFß) is overexpressed in several advanced cancer types and promotes tumor progression. We have reported that the intracellular domain (ICD) of TGFß receptor (TßR) I is cleaved by proteolytic enzymes in cancer cells, and then translocated to the nucleus in a manner dependent on the endosomal adaptor proteins APPL1/2, driving an invasiveness program. How cancer cells evade TGFß-induced growth inhibition is unclear. METHODS: We performed microarray analysis to search for genes regulated by APPL1/2 proteins in castration-resistant prostate cancer (CRPC) cells. We investigated the role of TßRI and TRAF6 in mitosis in cancer cell lines cultured in 10% FBS in the absence of exogenous TGFß. The molecular mechanism of the ubiquitination of AURKB by TRAF6 in mitosis and the formation of AURKB-TßRI complex in cancer cell lines and tissue microarrays was also studied. FINDINGS: During mitosis and cytokinesis, AURKB-TßRI complexes formed in midbodies in CRPC and KELLY neuroblastoma cells. TRAF6 induced polyubiquitination of AURKB on K85 and K87, protruding on the surface of AURKB to facilitate its activation. AURKB-TßRI complexes in patient's tumor tissue sections correlated with the malignancy of prostate cancer. INTERPRETATION: The AURKB-TßRI complex may become a prognostic biomarker for patients with risk of developing aggressive PC. FUNDING: Swedish Medical Research Council (2019-01598, ML; 2015-02757 and 2020-01291, CHH), the Swedish Cancer Society (20 0964, ML), a regional agreement between Umeå University and Region Västerbotten (ALF; RV-939377, -967041, -970057, ML). The European Research Council (787472, CHH). KAW 2019.0345, and the Kempe Foundation SMK-1866; ML. National Microscopy Infrastructure (NMI VR-RFI 2016-00968).


Asunto(s)
Aurora Quinasa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias de la Próstata Resistentes a la Castración , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Factor 6 Asociado a Receptor de TNF , Línea Celular Tumoral , Citocinesis , Humanos , Ligasas , Masculino , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/patología , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitina/metabolismo
6.
Biomolecules ; 12(5)2022 04 25.
Artículo en Inglés | MEDLINE | ID: mdl-35625561

RESUMEN

Transforming growth factor ß (TGF-ß) is a multifunctional cytokine regulating homeostasis and immune responses in adult animals and humans. Aberrant and overactive TGF-ß signaling promotes cancer initiation and fibrosis through epithelial-mesenchymal transition (EMT), as well as the invasion and metastatic growth of cancer cells. TGF-ß is a key factor that is active during hypoxic conditions in cancer and is thereby capable of contributing to angiogenesis in various types of cancer. Another potent role of TGF-ß is suppressing immune responses in cancer patients. The strong tumor-promoting effects of TGF-ß and its profibrotic effects make it a focus for the development of novel therapeutic strategies against cancer and fibrosis as well as an attractive drug target in combination with immune regulatory checkpoint inhibitors. TGF-ß belongs to a family of cytokines that exert their function through signaling via serine/threonine kinase transmembrane receptors to intracellular Smad proteins via the canonical pathway and in combination with co-regulators such as the adaptor protein and E3 ubiquitin ligases TNF receptor-associated factor 4 (TRAF4) and TNF receptor-associated factor 6 (TRAF6) to promote non-canonical pathways. Finally, the outcome of gene transcription initiated by TGF-ß is context-dependent and controlled by signals exerted by other growth factors such as EGF and Wnt. Here, we discuss the synergistic cooperation between TGF-ß and hypoxia in development, fibrosis and cancer.


Asunto(s)
Neoplasias , Factor de Crecimiento Transformador beta , Animales , Fibrosis , Humanos , Hipoxia , Neoplasias/metabolismo , Proteínas Smad/metabolismo , Factor 4 Asociado a Receptor de TNF/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
7.
J Clin Pathol ; 74(4): 216-222, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32467322

RESUMEN

Renal cell carcinoma (RCC) includes diverse tumour types characterised by various genetic abnormalities. The genetic changes, like mutations, deletions and epigenetic alterations, play a crucial role in the modification of signalling networks, tumour pathogenesis and prognosis. The most prevalent RCC type, clear cell RCC (ccRCC), is asymptomatic in the early stages and has a poorer prognosis compared with the papillary and the chromophobe types RCCs. Generally, ccRCC is refractory to chemotherapy and radiation therapy. Loss of von Hippel-Lindau (VHL) gene and upregulation of hypoxia-inducible factors (HIF), the signature of most sporadic ccRCC, promote multiple growth factors. Hence, VHL/HIF and a variety of pathways, including phosphatase and TEnsin homolog on chromosome 10/phosphatidylinositol-3-kinase (PI3K)/AKT, are closely connected and contribute to the ontogeny of ccRCC. In the recent decade, multiple targeting agents have been developed based on blocking major signalling pathways directly or indirectly involved in ccRCC tumour progression, metastasis, angiogenesis and survival. However, most of these drugs have limitations; either metastatic ccRCC develops resistance to these agents, or despite blocking receptors, tumour cells use alternate signalling pathways. This review compiles the state of knowledge about the PI3K/AKT signalling pathway confined to ccRCC and its cross-talks with VHL/HIF pathway.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Biomarcadores de Tumor , Carcinoma de Células Renales/enzimología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Renales/enzimología , Fosfatidilinositol 3-Quinasa/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/patología , Terapia Molecular Dirigida , Fosfohidrolasa PTEN/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Resultado del Tratamiento , Hipoxia Tumoral , Microambiente Tumoral
8.
Transl Oncol ; 13(12): 100857, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32866936

RESUMEN

HopQ is an outer-membrane protein of Helicobacter pylori that binds to human carcinoembryonic antigen-related cell-adhesion molecules (CEACAMs) with high specificity. We aimed to investigate fluorescence targeting of CEACAM-expressing colorectal tumors in patient-derived orthotopic xenograft (PDOX) models with fluorescently labeled recombinant HopQ (rHopQ). Western blotting, flow cytometry and ELISA were performed to determine the efficiency of rHopQ binding to CEACAMs. rHopQ was conjugated to IR800DyeCW (rHopQ-IR800). Nude mice received orthotopic implantation of colon cancer tumors. Three weeks later, mice were administered 25 µg or 50 µg HopQ-IR800 and imaged 24 or 48 h later. Intravital images were analyzed for tumor-to-background ratio (TBR). Flow cytometry and ELISA demonstrated binding of HopQ to CEACAM1, 3 and 5. Dose-response intravital imaging in PDOX models demonstrated optimal results 48 h after administration of 50 µg rHopQ-IR800 (TBR = 3.576) in our protocol. Orthotopic models demonstrated clear tumor margins of primary tumors and small regional metastases with a mean TBR = 3.678 (SD ±â€¯1.027). rHopQ showed specific binding to various CEACAMs in PDOX models. rHopQ may be useful for CEACAM-positive tumor and metastasis detection for pre-surgical diagnosis, intra-operative imaging and fluorescence-guided surgery.

9.
iScience ; 23(9): 101470, 2020 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-32888405

RESUMEN

Transforming growth factor ß (TGF-ß) enhances migration and invasion of cancer cells, causing life-threatening metastasis. Smad7 expression is induced by TGF-ß to control TGF-ß signaling in a negative feedback manner. Here we report an additional function of Smad7, i.e., to enhance TGF-ß induction of c-Jun and HDAC6 via binding to their regulatory regions, promoting migration and invasion of prostate cancer cells. Lysine 102 in Smad7 is crucial for binding to specific consensus sites in c-Jun and HDAC6, even when endogenous Smad2, 3, and 4 were silenced by siRNA. A correlation between the mRNA expression of Smad7 and HDAC6, Smad7 and c-Jun, and c-Jun and HDAC6 was found in public databases from analyses of prostate cancer tissues. High expression of Smad7, HDAC6, and c-Jun correlated with poor prognosis for patients with prostate cancer. The knowledge that Smad7 can activate transcription of proinvasive genes leading to prostate cancer progression provides clinically relevant information.

10.
Cell Cycle ; 18(17): 2141-2156, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31339433

RESUMEN

To investigate the significance of expression of HIF-1α, HIF-2α, and SNAIL1 proteins; and TGF-ß signaling pathway proteins in ccRCC, their relation with clinicopathological parameters and patient's survival were examined. We also investigated potential crosstalk between HIF-α and TGF-ß signaling pathway, including the TGF-ß type 1 receptor (ALK5-FL) and the intracellular domain of ALK5 (ALK5-ICD). Tissue samples from 154 ccRCC patients and comparable adjacent kidney cortex samples from 38 patients were analyzed for HIF-1α/2α, TGF-ß signaling components, and SNAIL1 proteins by immunoblot. Protein expression of HIF-1α and HIF-2α were significantly higher, while SNAIL1 had similar expression levels in ccRCC compared with the kidney cortex. HIF-2α associated with poor cancer-specific survival, while HIF-1α and SNAIL1 did not associate with survival. Moreover, HIF-2α positively correlated with ALK5-ICD, pSMAD2/3, and PAI-1; HIF-1α positively correlated with pSMAD2/3; SNAIL1 positively correlated with ALK5-FL, ALK5-ICD, pSMAD2/3, PAI-1, and HIF-2α. Intriguingly, in vitro experiments performed under normoxic conditions revealed that ALK5 interacts with HIF-1α and HIF-2α, and promotes their expression and the expression of their target genes GLUT1 and CA9, in a VHL dependent manner. We found that ALK5 induces expression of HIF-1α and HIF-2α, through its kinase activity. Under hypoxic conditions, HIF-α proteins correlated with the activated TGF-ß signaling pathway. In conclusion, we reveal that ALK5 plays a pivotal role in synergistic crosstalk between TGF-ß signaling and hypoxia pathway, and that the interaction between ALK5 and HIF-α contributes to tumor progression.


Asunto(s)
Carcinoma de Células Renales/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Inhibidor 1 de Activador Plasminogénico/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Adulto , Carcinoma de Células Renales/patología , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Transducción de Señal/genética , Factores de Transcripción de la Familia Snail/genética
11.
EBioMedicine ; 45: 192-207, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31262711

RESUMEN

BACKGROUND: Tumour necrosis factor receptor associated factor 6 (TRAF6) promotes inflammation in response to various cytokines. Aberrant Wnt3a signals promotes cancer progression through accumulation of ß-Catenin. Here we investigated a potential role for TRAF6 in Wnt signaling. METHODS: TRAF6 expression was silenced by siRNA in human prostate cancer (PC3U) and human colorectal SW480 cells and by CRISPR/Cas9 in zebrafish. Several biochemical methods and analyses of mutant phenotype in zebrafish were used to analyse the function of TRAF6 in Wnt signaling. FINDINGS: Wnt3a-treatment promoted binding of TRAF6 to the Wnt co-receptors LRP5/LRP6 in PC3U and LNCaP cells in vitro. TRAF6 positively regulated mRNA expression of ß-Catenin and subsequent activation of Wnt target genes in PC3U cells. Wnt3a-induced invasion of PC3U and SW480 cells were significantly reduced when TRAF6 was silenced by siRNA. Database analysis revealed a correlation between TRAF6 mRNA and Wnt target genes in patients with prostate cancer, and high expression of LRP5, TRAF6 and c-Myc correlated with poor prognosis. By using CRISPR/Cas9 to silence TRAF6 in zebrafish, we confirm TRAF6 as a key molecule in Wnt3a signaling for expression of Wnt target genes. INTERPRETATION: We identify TRAF6 as an important component in Wnt3a signaling to promote activation of Wnt target genes, a finding important for understanding mechanisms driving prostate cancer progression. FUND: KAW 2012.0090, CAN 2017/544, Swedish Medical Research Council (2016-02513), Prostatacancerförbundet, Konung Gustaf V:s Frimurarestiftelse and Cancerforskningsfonden Norrland. The funders did not play a role in manuscript design, data collection, data analysis, interpretation nor writing of the manuscript.


Asunto(s)
Inflamación/genética , Neoplasias de la Próstata/genética , Factor 6 Asociado a Receptor de TNF/genética , Proteína Wnt3A/genética , Proteínas de Pez Cebra/genética , Animales , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Inflamación/patología , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Masculino , Neoplasias de la Próstata/patología , Vía de Señalización Wnt/genética , Pez Cebra , beta Catenina/genética
12.
Oncogene ; 38(22): 4215-4231, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30705401

RESUMEN

Prostate cancer disseminates primarily into the adjacent lymph nodes, which is related to a poor outcome. Atypical protein kinase C ζ (PKCζ) is highly expressed in aggressive prostate cancer and correlates with Gleason score, clinical stage, and poor prognosis. Here, we report the molecular mechanisms of PKCζ in lymphatic metastasis during prostate cancer progression. Using zinc-finger nuclease technology or PKCζ shRNA lentiviral particles, and orthotopic mouse xenografts, we show that PKCζ-knockout or knockdown from aggressive prostate cancer (PC3 and PC3U) cells, decreasesd tumor growth and lymphatic metastasis in vivo. Intriguingly, PKCζ-knockout or knockdown impaired the activation of AKT, ERK, and NF-κB signaling in prostate cancer cells, thereby impairing the expression of lymphangiogenic factors and macrophage recruitment, resulting in aberrant lymphangiogenesis. Moreover, PKCζ regulated the expression of hyaluronan synthase enzymes, which is important for hyaluronan-mediated lymphatic drainage and tumor dissemination. Thus, PKCζ plays a crucial oncogenic role in the lymphatic metastasis of prostate cancer and is predicted to be a novel therapeutic target for prostate cancer.


Asunto(s)
Metástasis Linfática/patología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteína Quinasa C/metabolismo , Células A549 , Animales , Línea Celular Tumoral , Humanos , Hialuronano Sintasas/metabolismo , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Linfangiogénesis/fisiología , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Sistema de Señalización de MAP Quinasas/fisiología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Desnudos , FN-kappa B/metabolismo , Células PC-3 , Próstata/metabolismo , Próstata/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/fisiología
13.
Oncotarget ; 9(23): 16297-16310, 2018 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-29662646

RESUMEN

To evaluate the role of pVHL in the regulation of TGF-ß signaling pathways in clear cell renal cell carcinoma (ccRCC) as well as in non-ccRCC; the expression of pVHL, and the TGF-ß pathway components and their association with clinicopathological parameters and patient's survival were explored. Tissue samples from 143 ccRCC and 58 non-ccRCC patients were examined by immunoblot. ccRCC cell lines were utilized for mechanistic in-vitro studies. Expression levels of pVHL were significantly lower in ccRCC compared with non-ccRCC. Non-ccRCC and ccRCC pVHL-High expressed similar levels of pVHL. Expression of the TGF-ß type I receptor (ALK5) and intra-cellular domain were significantly higher in ccRCC compared with non-ccRCC. In non-ccRCC, expressions of ALK5-FL, ALK5-ICD, pSMAD2/3, and PAI-1 had no association with clinicopathological parameters and survival. In ccRCC pVHL-Low, ALK5-FL, ALK5-ICD, pSMAD2/3, and PAI-1 were significantly related with tumor stage, size, and survival. In ccRCC pVHL-High, the expression of PAI-1 was associated with stage and survival. In-vitro studies revealed that pVHL interacted with ALK5 to downregulate its expression through K48-linked poly-ubiquitination and proteasomal degradation, thus negatively controlling TGF-ß induced cancer cell invasiveness. The pVHL status controls the ALK5 and can thereby regulate the TGF-ß pathway, aggressiveness of tumors, and survival of the ccRCC and non-ccRCC patients.

14.
Prostate ; 78(6): 446-456, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29383751

RESUMEN

BACKGROUND: Transforming growth factor ß (TGFß) functions as a double-edged sword in prostate cancer tumorigenesis. In initial stages of the disease, TGFß acts as a growth inhibitor upon tumor cells, whereas it in later stages of disease rather promotes invasion and metastatic potential. One well-known cellular source of TGFß in the bone metastatic site is the bone-forming osteoblasts. Here we have studied the effects by osteoblast-derived factors on metastatic potential in several human prostate cancer cell lines. METHODS: Effects on metastatic potential in prostate cancer cells by osteoblast-derived factors were studied in vitro using several methods, including Transwell migration and evaluation of formation of pro-migratory protrusions. Confocal microscopy was used to evaluate possible changes in differentiation state in tumor cells by analysis of markers for epithelial-to-mesenchymal transition (EMT). The Matrigel-on-top 3D culture method was used for further assessment of metastatic characteristics in tumor cells by analysis of formation of filopodium-like protrusions (FLPs). RESULTS: Osteoblast-derived factors increased migration of PC-3U cells, an effect less prominent in cells overexpressing a mutated type I TGFß receptor (TßRI) preventing non-canonical TRAF6-dependent TGFß signaling. Osteoblast-derived factors also increased the formation of long protrusions and loss of cell-cell contacts in PC-3U cells, suggesting induction of a more aggressive phenotype. In addition, treatment with TGFß or osteoblast-derived factors of PC-3U cells in Matrigel-on-top 3D cultures promoted formation of FLPs, previously shown to be essential for metastatic establishment. CONCLUSIONS: These findings suggests that factors secreted from osteoblasts, including TGFß, can induce several cellular traits involved in metastatic potential of PC-3U cells, further strengthening the role for bone cells to promote metastatic tumor cell behavior.


Asunto(s)
Osteoblastos/metabolismo , Neoplasias de la Próstata/patología , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Línea Celular Tumoral , Movimiento Celular/fisiología , Forma de la Célula/fisiología , Medios de Cultivo Condicionados , Transición Epitelial-Mesenquimal , Humanos , Masculino , Neoplasias de la Próstata/metabolismo
15.
Oncotarget ; 8(58): 97703-97726, 2017 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-29228645

RESUMEN

Transforming growth factor ß (TGFß) is a key regulator of epithelial-to-mesenchymal transition (EMT) during embryogenesis and in tumors. The effect of TGFß, on ΕΜΤ, is conveyed by induction of the pro-invasive transcription factor Snail1. In this study, we report that TGFß stimulates Snail1 sumoylation in aggressive prostate, breast and lung cancer cells. Sumoylation of Snail1 lysine residue 234 confers its transcriptional activity, inducing the expression of classical EMT genes, as well as TGFß receptor I (TßRI) and the transcriptional repressor Hes1. Mutation of Snail1 lysine residue 234 to arginine (K234R) abolished sumoylation of Snail1, as well as its migratory and invasive properties in human prostate cancer cells. An increased immunohistochemical expression of Snail1, Sumo1, TßRI, Hes1, and c-Jun was observed in aggressive prostate cancer tissues, consistent with their functional roles in tumorigenesis.

16.
Cell Rep ; 20(8): 1893-1905, 2017 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-28834752

RESUMEN

Cellular blebbing, caused by local alterations in cell-surface tension, has been shown to increase the invasiveness of cancer cells. However, the regulatory mechanisms balancing cell-surface dynamics and bleb formation remain elusive. Here, we show that an acute reduction in cell volume activates clathrin-independent endocytosis. Hence, a decrease in surface tension is buffered by the internalization of the plasma membrane (PM) lipid bilayer. Membrane invagination and endocytosis are driven by the tension-mediated recruitment of the membrane sculpting and GTPase-activating protein GRAF1 (GTPase regulator associated with focal adhesion kinase-1) to the PM. Disruption of this regulation by depleting cells of GRAF1 or mutating key phosphatidylinositol-interacting amino acids in the protein results in increased cellular blebbing and promotes the 3D motility of cancer cells. Our data support a role for clathrin-independent endocytic machinery in balancing membrane tension, which clarifies the previously reported role of GRAF1 as a tumor suppressor.


Asunto(s)
Clatrina/metabolismo , Endocitosis/fisiología , Seudópodos/fisiología , Fenómenos Biológicos , Humanos , Invasividad Neoplásica
17.
Sci Signal ; 10(486)2017 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-28676490

RESUMEN

Transforming growth factor-ß (TGF-ß) is a pluripotent cytokine that regulates cell fate and plasticity in normal tissues and tumors. The multifunctional cellular responses evoked by TGF-ß are mediated by the canonical SMAD pathway and by noncanonical pathways, including mitogen-activated protein kinase (MAPK) pathways and the phosphatidylinositol 3'-kinase (PI3K)-protein kinase B (AKT) pathway. We found that TGF-ß activated PI3K in a manner dependent on the activity of the E3 ubiquitin ligase tumor necrosis factor receptor-associated factor 6 (TRAF6). TRAF6 polyubiquitylated the PI3K regulatory subunit p85α and promoted the formation of a complex between the TGF-ß type I receptor (TßRI) and p85α, which led to the activation of PI3K and AKT. Lys63-linked polyubiquitylation of p85α on Lys513 and Lys519 in the iSH2 (inter-Src homology 2) domain was required for TGF-ß-induced activation of PI3K-AKT signaling and cell motility in prostate cancer cells and activated macrophages. Unlike the activation of SMAD pathways, the TRAF6-mediated activation of PI3K and AKT was not dependent on the kinase activity of TßRI. In situ proximity ligation assays revealed that polyubiquitylation of p85α was evident in aggressive prostate cancer tissues. Thus, our data reveal a molecular mechanism by which TGF-ß activates the PI3K-AKT pathway to drive cell migration.


Asunto(s)
Movimiento Celular , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Apoptosis , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Lisina/metabolismo , Masculino , Ratones , Ratones Noqueados , Fosforilación , Neoplasias de la Próstata/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Transducción de Señal , Ubiquitinación
18.
Cell Host Microbe ; 21(3): 376-389, 2017 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-28279347

RESUMEN

The BabA adhesin mediates high-affinity binding of Helicobacter pylori to the ABO blood group antigen-glycosylated gastric mucosa. Here we show that BabA is acid responsive-binding is reduced at low pH and restored by acid neutralization. Acid responsiveness differs among strains; often correlates with different intragastric regions and evolves during chronic infection and disease progression; and depends on pH sensor sequences in BabA and on pH reversible formation of high-affinity binding BabA multimers. We propose that BabA's extraordinary reversible acid responsiveness enables tight mucosal bacterial adherence while also allowing an effective escape from epithelial cells and mucus that are shed into the acidic bactericidal lumen and that bio-selection and changes in BabA binding properties through mutation and recombination with babA-related genes are selected by differences among individuals and by changes in gastric acidity over time. These processes generate diverse H. pylori subpopulations, in which BabA's adaptive evolution contributes to H. pylori persistence and overt gastric disease.


Asunto(s)
Adhesinas Bacterianas/metabolismo , Adhesión Bacteriana , Mucosa Gástrica/microbiología , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Mucosa Gástrica/patología , Infecciones por Helicobacter/patología , Concentración de Iones de Hidrógeno
19.
Oncotarget ; 7(24): 35917-35931, 2016 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-27166254

RESUMEN

The molecular mechanisms whereby transforming growth factor-ß (TGF-ß) promotes clear cell renal cell carcinoma (ccRCC) progression is elusive. The cell membrane bound TGF-ß type I receptor (ALK5), was recently found to undergo proteolytic cleavage in aggressive prostate cancer cells, resulting in liberation and subsequent nuclear translocation of its intracellular domain (ICD), suggesting that ALK5-ICD might be a useful cancer biomarker. Herein, the possible correlation between ALK5 full length (ALK5-FL) and ALK5-ICD protein, phosphorylated Smad2/3 (pSmad2/3), and expression of TGF-ß target gene PAI-1, was investigated in a clinical ccRCC material, in relation to tumor grade, stage, size and cancer specific survival. Expression of ALK5-FL, ALK5-ICD, pSmad2/3 and PAI-1 protein levels were significantly higher in higher stage and associated with adverse survival. ALK5-ICD, pSmad2/3 and PAI-1 correlated with higher grade, and ALK5-FL, pSmad2/3 and PAI-1 protein levels were significantly correlated with larger tumor size. Moreover, the functional role of the TGF-ß - ALK5-ICD pathway were investigated in two ccRCC cell lines by treatment with ADAM/MMP2 inhibitor TAPI-2, which prevented TGF-ß-induced ALK5-ICD generation, nuclear translocation, as well as cell invasion. The present study demonstrated that canonical TGF-ß Smad2/3 pathway and generation of ALK5-ICD correlates with poor survival and invasion of ccRCC in vitro.


Asunto(s)
Carcinoma de Células Renales/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Renales/genética , Factor de Crecimiento Transformador beta/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Femenino , Humanos , Estimación de Kaplan-Meier , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Inhibidor 1 de Activador Plasminogénico/genética , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad2/genética , Proteína Smad2/metabolismo , Proteína smad3/genética , Proteína smad3/metabolismo
20.
Oncotarget ; 7(1): 279-92, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26583432

RESUMEN

The multifunctional cytokine transforming growth factor-ß (TGFß) is produced by several types of cancers, including prostate cancer, and promote tumour progression in autocrine and paracrine manners. In response to ligand binding, the TGFß type I receptor (TßRI) activates Smad and non-Smad signalling pathways. The ubiquitin-ligase tumour necrosis factor receptor-associated factor 6 (TRAF6) was recently linked to regulate intramembrane proteolytic cleavage of the TßRI in cancer cells. Subsequently, the intracellular domain (ICD) of TßRI enters in an unknown manner into the nucleus, where it promotes the transcription of pro-invasive genes, such as MMP2 and MMP9. Here we show that the endocytic adaptor molecules APPL1 and APPL2 are required for TGFß-induced nuclear translocation of TßRI-ICD and for cancer cell invasiveness of human prostate and breast cancer cell lines. Moreover, APPL proteins were found to be expressed at high levels in aggressive prostate cancer tissues, and to be associated with TßRI in a TRAF6-dependent manner. Our results suggest that the APPL-TßRI complex promotes prostate tumour progression, and may serve as a prognostic marker.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Núcleo Celular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Sitios de Unión/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Immunoblotting , Masculino , Microscopía Confocal , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA