Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Cells ; 11(24)2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36552790

RESUMEN

Cancer progression is supported by the cross-talk between tumor cells and the surrounding stroma. In this context, senescent cells in the tumor microenvironment contribute to the development of a pro-inflammatory milieu and the acquisition of aggressive traits by cancer cells. Anticancer treatments induce cellular senescence (therapy-induced senescence, TIS) in both tumor and non-cancerous cells, contributing to many detrimental side effects of therapies. Thus, we focused on the effects of chemotherapy on the stromal compartment of prostate and ovarian cancer. We demonstrated that anticancer chemotherapeutics, regardless of their specific mechanism of action, promote a senescent phenotype in stromal fibroblasts, resulting in metabolic alterations and secretion of paracrine factors, sustaining the invasive and clonogenic potential of both prostate and ovarian cancer cells. The clearance of senescent stromal cells, through senolytic drug treatment, reverts the malignant phenotype of tumor cells. The clinical relevance of TIS was validated in ovarian and prostate cancer patients, highlighting increased accumulation of lipofuscin aggregates, a marker of the senescent phenotype, in the stromal compartment of tissues from chemotherapy-treated patients. These data provide new insights into the potential efficacy of combining traditional anticancer strategies with innovative senotherapy to potentiate anticancer treatments and overcome the adverse effects of chemotherapy.


Asunto(s)
Neoplasias Ováricas , Neoplasias de la Próstata , Humanos , Masculino , Femenino , Neoplasias Ováricas/genética , Próstata/patología , Neoplasias de la Próstata/tratamiento farmacológico , Fenotipo , Microambiente Tumoral
2.
Cell Rep ; 40(7): 111233, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35977477

RESUMEN

5-Fluorouracil (5-FU) is a key component of chemotherapy for colorectal cancer (CRC). 5-FU efficacy is established by intracellular levels of folate cofactors and DNA damage repair strategies. However, drug resistance still represents a major challenge. Here, we report that alterations in serine metabolism affect 5-FU sensitivity in in vitro and in vivo CRC models. In particular, 5-FU-resistant CRC cells display a strong serine dependency achieved either by upregulating endogenous serine synthesis or increasing exogenous serine uptake. Importantly, regardless of the serine feeder strategy, serine hydroxymethyltransferase-2 (SHMT2)-driven compartmentalization of one-carbon metabolism inside the mitochondria represents a specific adaptation of resistant cells to support purine biosynthesis and potentiate DNA damage response. Interfering with serine availability or affecting its mitochondrial metabolism revert 5-FU resistance. These data disclose a relevant mechanism of mitochondrial serine use supporting 5-FU resistance in CRC and provide perspectives for therapeutic approaches.


Asunto(s)
Neoplasias Colorrectales , Neoplasias , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Resistencia a Antineoplásicos/genética , Fluorouracilo/metabolismo , Fluorouracilo/farmacología , Humanos , Mitocondrias/metabolismo , Neoplasias/metabolismo , Nucleótidos/metabolismo , Serina/metabolismo
3.
Cancers (Basel) ; 14(3)2022 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-35158815

RESUMEN

Tumor relapse represents one of the main obstacles to cancer treatment. Many patients experience cancer relapse even decades from the primary tumor eradication, developing more aggressive and metastatic disease. This phenomenon is associated with the emergence of dormant cancer cells, characterized by cell cycle arrest and largely insensitive to conventional anti-cancer therapies. These rare and elusive cells may regain proliferative abilities upon the induction of cell-intrinsic and extrinsic factors, thus fueling tumor re-growth and metastasis formation. The molecular mechanisms underlying the maintenance of resistant dormant cells and their awakening are intriguing but, currently, still largely unknown. However, increasing evidence recently underlined a strong dependency of cell cycle progression to metabolic adaptations of cancer cells. Even if dormant cells are frequently characterized by a general metabolic slowdown and an increased ability to cope with oxidative stress, different factors, such as extracellular matrix composition, stromal cells influence, and nutrient availability, may dictate specific changes in dormant cells, finally resulting in tumor relapse. The main topic of this review is deciphering the role of the metabolic pathways involved in tumor cells dormancy to provide new strategies for selectively targeting these cells to prevent fatal recurrence and maximize therapeutic benefit.

4.
Cancers (Basel) ; 13(14)2021 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-34298765

RESUMEN

Metastatic melanoma is characterized by poor prognosis and a low free-survival rate. Thanks to their high plasticity, melanoma cells are able to migrate exploiting different cell motility strategies, such as the rounded/amoeboid-type motility and the elongated/mesenchymal-type motility. In particular, the amoeboid motility strongly contributes to the dissemination of highly invasive melanoma cells and no treatment targeting this process is currently available for clinical application. Here, we tested Claisened Hexafluoro as a novel inhibitor of the amoeboid motility. Reported data demonstrate that Claisened Hexafluoro specifically inhibits melanoma cells moving through amoeboid motility by deregulating mitochondrial activity and activating the AMPK signaling. Moreover, Claisened Hexafluoro is able to interfere with the adhesion abilities and the stemness features of melanoma cells, thus decreasing the in vivo metastatic process. This evidence may contribute to pave the way for future possible therapeutic applications of Claisened Hexafluoro to counteract metastatic melanoma dissemination.

5.
Biochim Biophys Acta Rev Cancer ; 1874(2): 188442, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33017632

RESUMEN

In mammalian cells, tyrosine phosphorylation is one of the main mechanisms responsible for regulating signal transduction pathways and key cellular functions. Moreover, recent studies demonstrated that tyrosine phosphorylation influences the activity of some metabolic enzymes, even if it remains to be clarified whether tyrosine phosphorylation can be considered a general mechanism involving most of the metabolic enzymes or only a subset of these. To elucidate this aspect, we conducted a two-step analysis. First, we analyzed literature to identify all the metabolic enzymes whose activity is affected by tyrosine phosphorylation. Second, we crossed these data with those obtained from the PhosphoSitePlus database analysis. Collected information was used to depict an exhaustive map showing the real spread of tyrosine phosphorylation among metabolic enzymes. In summary, data reported in this review highlight that tyrosine phosphorylation is not a sporadic event but a widespread post-translational modification, which is essential to promote the metabolic reprogramming of cancer cells.


Asunto(s)
Redes Reguladoras de Genes , Neoplasias/metabolismo , Tirosina/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional
6.
Cancers (Basel) ; 12(10)2020 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-33003469

RESUMEN

Despite a large number of therapeutic options available, malignant melanoma remains a highly fatal disease, especially in its metastatic forms. The oncogenic role of protein tyrosine phosphatases (PTPs) is becoming increasingly clear, paving the way for novel antitumor treatments based on their inhibition. In this review, we present the oncogenic PTPs contributing to melanoma progression and we provide, where available, a description of new inhibitory strategies designed against these enzymes and possibly useful in melanoma treatment. Considering the relevance of the immune infiltrate in supporting melanoma progression, we also focus on the role of PTPs in modulating immune cell activity, identifying interesting therapeutic options that may support the currently applied immunomodulating approaches. Collectively, this information highlights the value of going further in the development of new strategies targeting oncogenic PTPs to improve the efficacy of melanoma treatment.

7.
Nucleic Acids Res ; 48(16): 8943-8958, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32697292

RESUMEN

The epithelial-to-mesenchymal transition (EMT) is a complex transcriptional program induced by transforming growth factor ß1 (TGF-ß1). Histone lysine-specific demethylase 1 (LSD1) has been recognized as a key mediator of EMT in cancer cells, but the precise mechanism that underlies the activation and repression of EMT genes still remains elusive. Here, we characterized the early events induced by TGF-ß1 during EMT initiation and establishment. TGF-ß1 triggered, 30-90 min post-treatment, a nuclear oxidative wave throughout the genome, documented by confocal microscopy and mass spectrometry, mediated by LSD1. LSD1 was recruited with phosphorylated SMAD2/3 to the promoters of prototypic genes activated and repressed by TGF-ß1. After 90 min, phospho-SMAD2/3 downregulation reduced the complex and LSD1 was then recruited with the newly synthesized SNAI1 and repressors, NCoR1 and HDAC3, to the promoters of TGF-ß1-repressed genes such as the Wnt soluble inhibitor factor 1 gene (WIF1), a change that induced a late oxidative burst. However, TGF-ß1 early (90 min) repression of transcription also required synchronous signaling by reactive oxygen species and the stress-activated kinase c-Jun N-terminal kinase. These data elucidate the early events elicited by TGF-ß1 and the priming role of DNA oxidation that marks TGF-ß1-induced and -repressed genes involved in the EMT.


Asunto(s)
ADN/metabolismo , Transición Epitelial-Mesenquimal/genética , Histona Demetilasas/fisiología , Proteína Smad2/fisiología , Factor de Crecimiento Transformador beta1/fisiología , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos
8.
Cancer Cell Int ; 19: 67, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30948927

RESUMEN

BACKGROUND: Low molecular weight protein tyrosine phosphatase (LMW-PTP) is overexpressed in different cancer types and its expression is related to more aggressive disease, reduced survival rate and drug resistance. Morin is a natural polyphenol which negatively modulates, among others, the activity of LMW-PTP, leading to the potentiation of the effects of different antitumoral drugs, representing a potential beneficial treatment against cancer. METHODS: LMW-PTP levels were measured by immunoblot analysis both in CLL cells from patients and in chronic lymphocytic leukemia (CLL)-derived Mec-1 cells. Cell viability was assessed in Mec-1 cells treated with morin alone or in combination with either fludarabine or ibrutinib or following siRNA-mediated LMW-PTP knockdown. Furthermore, the expression levels of VLA-4 and CXCR4 were assessed by both qRT-PCR and flow cytometry and both adhesion to fibronectin-coated plates and migration toward CXCL12 were analyzed in Mec-1 cells treated with morin alone or in combination with fludarabine or ibrutinib. RESULTS: We observed that LMW-PTP is highly expressed in Mec-1 cells as well as in leukemic B lymphocytes purified from CLL patients compared to normal B lymphocytes. Morin treatment strongly decreased LMW-PTP expression levels in Mec-1 cells and potentiated the anticancer properties of both fludarabine and ibrutinib by increasing their apoptotic effects on leukemic cells. Moreover, morin negatively regulates adhesion and CXCL12-dependent migration of Mec-1 cells by affecting VLA-4 integrin expression and CXCR4 receptor recycling. CONCLUSIONS: Morin treatment in CLL-derived Mec-1 cell line synergizes with conventional anticancer drugs currently used in CLL therapy by affecting leukemic cell viability and trafficking.

9.
J Cell Physiol ; 234(6): 8274-8285, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30378132

RESUMEN

Cancer progression is strictly dependent on the relationship between tumor cells and the surrounding stroma, which supports cancer malignancy promoting several crucial steps of tumor progression, including the execution of the epithelial to mesenchymal transition (EMT) associated with enhancement in cell invasion, resistance to both anoikis and chemotherapeutic treatments. Recently it has been highlighted the central role of microRNAs (miRNAs) as regulators of tumor progression. Notably, in several tumors a strong deregulation of miRNAs is observed, supporting proliferation, invasion, and metabolic reprogramming of tumor cells. Here we demonstrated that cancer-associated fibroblasts induce a downregulation of miR-1247 in prostate cancer (PCa) cells. We proved that miR-1247 repression is functional for the achievement of EMT and increased cell invasion as well as stemness traits. These phenomena contribute to promote the metastatic potential of PCa cells as demonstrated by increased lung colonization in in vivo experiments. Moreover, as a consequence of miR-1247 downregulation, we observed a correlated increased expression level of neuropilin-1, a miR-1247 target involved as a coreceptor in the epidermal growth factor receptor signaling. Taken together, our data highlight miR-1247 as a potential target for molecular therapies aimed to block the progression and diffusion of PCa.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , MicroARNs/genética , Neuropilina-1/genética , Neoplasias de la Próstata/genética , Proliferación Celular/genética , Reprogramación Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Metástasis de la Neoplasia , Neoplasias de la Próstata/patología , Células del Estroma/metabolismo , Células del Estroma/patología
10.
Mol Carcinog ; 58(5): 686-698, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30582224

RESUMEN

LMW-PTP has been associated with the development of colorectal cancer (CRC) and with the resistance to chemotherapy in cancer cells. To clarify its role in vivo, we studied LMW-PTP expression in Pirc rats (F344/NTac-Apc am1137 ), genetically prone to CRC and resistant to apoptosis. In the morphologically normal mucosa (NM) of Pirc rats, a dramatic over-expression of LMW-PTP was found compared to wt rats (about 60 times higher). Moreover, LMW-PTP levels further increase in spontaneously developed Pirc colon tumors. To understand if and how LMW-PTP affects resistance to apoptosis, we studied CRC cell lines, sensitive (HT29 and HCT-116), or resistant (HT29R, HCT116R) to 5-Fluorouracil (5-FU): resistant cells over-express LMW-PTP. When resistant cells were challenged with morin, a polyphenol inhibiting LMW-PTP, a fast and dose-related down-regulation of LMW-PTP was observed. 5-FU and morin co-treatment dramatically decreased cell viability, increased apoptosis, and significantly impaired self-renewal ability of all the cancer cell lines we have studied. Similarly, we observed that, in Pirc rats, one-week morin administration (50 mg/kg) down-regulated LMW-PTP and restored the apoptotic response to 5-FU in the NM. Finally, administration of morin for a longer period led to a significant reduction in colon precancerous lesions, together with a down-regulation of LMW-PTP. Taken together, these results document the involvement of LMW-PTP in the process of CRC in vitro and in vivo. Morin treatment may be envisaged as a system to increase the sensitivity to chemotherapy and to prevent carcinogenesis.


Asunto(s)
Carcinogénesis/patología , Colon/patología , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Flavonoides/farmacología , Genes APC , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Animales , Antioxidantes/farmacología , Carcinogénesis/inducido químicamente , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Colon/efectos de los fármacos , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/etiología , Técnicas In Vitro , Masculino , Proteínas Tirosina Fosfatasas/genética , Proteínas Proto-Oncogénicas/genética , Ratas , Ratas Endogámicas F344
11.
Biochim Biophys Acta Gen Subj ; 1862(12): 2533-2544, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30251652

RESUMEN

BACKGROUND: Low Molecular Weight Phosphotyrosine Protein Phosphatase (LMW-PTP) is an enzyme involved not only in tumor onset and progression but also in type 2 diabetes. A recent review shows that LMW-PTP acts on several RTK (receptor tyrosine kinase) such as PDGFR, EGFR, EphA2, Insulin receptor. It is well described also its interaction with cSrc. It is noteworthy that most of these conclusions are based on the use of cell lines expressing low levels of LMW-PTP. The aim of the present study was to discover new LMW-PTP substrates in aggressive human tumors where the over-expression of this phosphatase is a common feature. METHODS: We investigated, by proteomic analysis, the protein phosphorylation pattern of A375 human melanoma cells silenced for LMW-PTP. Two-dimensional electrophoresis (2-DE) analysis, followed by western blot was performed using anti-phosphotyrosine antibodies, in order to identify differentially phosphorylated proteins. RESULTS: Proteomic analysis pointed out that most of the identified proteins belong to the glycolytic metabolism, such as α-enolase, pyruvate kinase, glyceraldehyde-3-phosphate dehydrogenase and triosephosphate isomerase, suggesting an involvement of LMW-PTP in glucose metabolism. Assessment of lactate production and oxygen consumption demonstrated that LMW-PTP silencing enhances glycolytic flux and slow down the oxidative metabolism. In particular, LMW-PTP expression affects PKM2 tyrosine-phosphorylation and nuclear localization, modulating its activity. CONCLUSION: All these findings propose that tumor cells are subjected to metabolic reprogramming after LMW-PTP silencing, enhancing glycolytic flux, probably to compensate the inhibition of mitochondrial metabolism. GENERAL SIGNIFICANCE: Our results highlight the involvement of LMW-PTP in regulating glucose metabolism in A375 melanoma cells.


Asunto(s)
Proteínas Fluorescentes Verdes/metabolismo , Concentración de Iones de Hidrógeno , Neoplasias/metabolismo , Fluorescencia , Humanos , Peso Molecular , Neoplasias/patología
12.
Cancer Med ; 7(5): 1933-1943, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29573568

RESUMEN

Tumor resistance to apoptosis is one the main causes of anticancer treatment failure. Previous studies showed that LMW-PTP overexpression enhances resistance of cancer cells to traditional anticancer drugs. Today, the role of LMW-PTP in inducing resistance to apoptosis in melanoma cells remains to be elucidated. Experimental setting include MTT assay, Annexin V/Pi method, and colony assay to assess whether silencing of LMW-PTP improves the sensitivity of A375 to dacarbazine, 5-FU, and radiotherapy. Pharmacological targeting of LMW-PTP was obtained using Morin, a LMW-PTP inhibitor. The ability of Morin to improve the effectiveness of anticancer drugs and radiotherapy was also studied. Moreover, PC3 cells were used as an alternative cellular model to confirm the data obtained with melanoma cells. We found that LMW-PTP silencing improves the effectiveness of dacarbazine, 5-FU, and radiotherapy. Identical results were obtained in vivo when Morin was used to target LMW-PTP. We demonstrated that Morin synergizes with dacarbazine, improving its cytotoxic activity. However, we showed that the combined treatment, Morin-anticancer drug, does not affect the viability of noncancerous cells. Knockdown of LMW-PTP sensitizes also PC3 cells to docetaxel and radiotherapy. In conclusion, we showed that LMW-PTP targeting improves effectiveness of anticancer drugs used for treatment of melanoma. Moreover, our results suggest that Morin could be used as adjuvant to improve the outcome of patients affected by metastatic melanoma.


Asunto(s)
Resistencia a Antineoplásicos , Flavonoides/farmacología , Melanoma/terapia , Proteínas Tirosina Fosfatasas/genética , Proteínas Proto-Oncogénicas/genética , Tolerancia a Radiación , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Dacarbazina/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Quimioterapia , Fluorouracilo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Humanos , Melanoma/genética , Terapia Molecular Dirigida , Proteínas Tirosina Fosfatasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Tolerancia a Radiación/efectos de los fármacos , Radioterapia , Regulación hacia Arriba/efectos de los fármacos
13.
Oncotarget ; 6(27): 24061-74, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26183399

RESUMEN

Cancer associated fibroblasts (CAFs) are key determinants of cancer progression. In prostate carcinoma (PCa), CAFs induce epithelial-mesenchymal transition (EMT) and metabolic reprogramming of PCa cells towards oxidative phosphorylation (OXPHOS), promoting tumor growth and metastatic dissemination. We herein establish a novel role for pyruvate kinase M2 (PKM2), an established effector of Warburg-like glycolytic behavior, in OXPHOS metabolism induced by CAFs. Indeed, CAFs promote PKM2 post-translational modifications, such as cysteine oxidation and Src-dependent tyrosine phosphorylation, allowing nuclear migration of PKM2 and the formation of a trimeric complex with hypoxia inducible factor-1α (HIF-1α) and the transcriptional repressor Differentially Expressed in Chondrocytes-1 (DEC1). DEC1 recruitment is mandatory for downregulating miR205 expression, thereby fostering EMT execution and metabolic switch toward OXPHOS. Furthermore, the analysis of a cohort of PCa patients reveals a significant positive correlation between PKM2 nuclear localization and cancer aggressiveness, thereby validating our in vitro observations. Crucially, in vitro and in vivo pharmacological targeting of PKM2 nuclear translocation using DASA-58, as well as metformin, impairs metastatic dissemination of PCa cells in SCID mice. Our study indicates that impairing the metabolic tumor:stroma interplay by targeting the PKM2/OXPHOS axis, may be a valuable novel therapeutic approach in aggressive prostate carcinoma.


Asunto(s)
Transporte Activo de Núcleo Celular , Proteínas Portadoras/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias de la Próstata/patología , Piruvato Quinasa/metabolismo , Hormonas Tiroideas/metabolismo , Animales , Sitios de Unión , Carcinoma/metabolismo , Carcinoma/patología , Línea Celular Tumoral , Núcleo Celular/metabolismo , Estudios de Cohortes , Cisteína/química , Fibroblastos/metabolismo , Glucosa/química , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Ácido Láctico/química , Masculino , Metformina/química , Ratones , Ratones SCID , MicroARNs/metabolismo , Metástasis de la Neoplasia , Oxidación-Reducción , Fosforilación Oxidativa , Oxígeno/química , Neoplasias de la Próstata/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Unión a Hormona Tiroide
14.
J Signal Transduct ; 2012: 684592, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22272371

RESUMEN

Increased ROS (cellular reactive oxygen species) are characteristic of both fibrosis and tumour development. ROS induce the trans-differentiation to myofibroblasts, the activated form of fibroblasts able to promote cancer progression. Here, we report the role of ROS produced in response to dysfunctions of mitochondrial complex I, in fibroblast activation and in tumour progression. We studied human fibroblasts with mitochondrial dysfunctions of complex I, leading to hyperproduction of ROS. We demonstrated that ROS level produced by the mutated fibroblasts correlates with their activation. The increase of ROS in these cells provides a greater ability to remodel the extracellular matrix leading to an increased motility and invasiveness. Furthermore, we evidentiated that in hypoxic conditions these fibroblasts cause HIF-1α stabilization and promote a proinvasive phenotype of human melanoma cells through secretion of cytokines. These data suggest a possible role of deregulated mitochondrial ROS production in fibrosis evolution as well as in cancer progression and invasion.

15.
Mol Cancer Res ; 9(2): 149-60, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21205836

RESUMEN

EphA2 kinase regulates cell shape, adhesion, and motility and is frequently overexpressed in several cancers, including melanoma, prostate, breast, and colon cancers and lung carcinoma. Although a function in both tumor onset and metastasis has been proposed, the role played by EphA2 in tumor progression is still debated. In melanoma, EphA2 has been reported to affect cell migration and invasiveness allowing cells to move by a proteolysis-independent strategy, commonly referred as amoeboid motility. With the aim to understand the role of EphA2 in prostate cancer metastatic spreading, we stably silenced EphA2 expression in a model of aggressive metastatic prostate carcinoma. Our results show that EphA2 drives the metastatic program of prostate carcinoma, although its involvement greatly differs among metastatic steps. Indeed, EphA2 expression (i) greatly affects prostate carcinoma cell motility style, guiding an amoeboid movement based on Rho-mediated cell rounding and independent from metalloprotases, (ii) is ineffective on transendothelial migration, adhesion onto extracellular matrix proteins, and on resistance to anoikis, (iii) regulates clonogenic potential of prostate carcinoma, thereby increasing anchorage-independent growth and self-renewal, prostasphere formation, tumor onset, dissemination to bone, and growth of metastatic colonies. Our finding indicate that EphA2-overexpressing prostate carcinoma cells gain an invasive benefit from their amoeboid motility style to escape from primary tumors and then, enhancing their clonogenic potential successfully target bone and grow metastases, thereby acknowledging EphA2 as a target for antimetastatic therapy of aggressive prostate cancers.


Asunto(s)
Movimiento Celular , Neoplasias de la Próstata/patología , Receptor EphA2/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Adhesión Celular , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Células Clonales , Silenciador del Gen , Humanos , Masculino , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Neoplasias de la Próstata/metabolismo
16.
Am J Pathol ; 174(4): 1492-503, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19264906

RESUMEN

Ligand-activated Eph tyrosine kinases regulate cellular repulsion, morphology, adhesion, and motility. EphA2 kinase is frequently up-regulated in several different types of cancers, including prostate, breast, colon, and lung carcinomas, as well as in melanoma. The existing data do not clarify whether EphA2 receptor phosphorylation or its simple overexpression, which likely leads to Eph kinase-independent responses, plays a role in the progression of malignant prostate cancer. In this study, we address the role of EphA2 tyrosine phosphorylation in prostate carcinoma cell adhesion, motility, invasion, and formation of metastases. Tumor cells expressing kinase-deficient EphA2 mutants, as well as an EphA2 variant lacking the cytoplasmic domain, are defective in ephrinA1-mediated cell rounding, retraction fiber formation, de-adhesion from the extracellular matrix, RhoA and Rac1 GTPase regulation, three-dimensional matrix invasion, and in vivo metastasis, suggesting a key role for EphA2 kinase activity. Nevertheless, EphA2 regulation of cell motility and invasion, as well as the formation of bone and visceral tumor colonies, reveals a component of both EphA2 kinase-dependent and -independent features. These results uncover a differential requirement for EphA2 kinase activity in the regulation of prostate carcinoma metastasis outcome, suggesting that although the kinase activity of EphA2 is required for the regulation of cell adhesion and cytoskeletal rearrangement, some distinct kinase-dependent and -independent pathways likely cooperate to drive cancer cell migration, invasion, and metastasis outcome.


Asunto(s)
Invasividad Neoplásica/fisiopatología , Metástasis de la Neoplasia/fisiopatología , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Receptor EphA2/metabolismo , Animales , Western Blotting , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Efrina-A1/metabolismo , Humanos , Inmunoprecipitación , Masculino , Ratones , Ratones Desnudos , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/patología , Fosforilación , Reacción en Cadena de la Polimerasa
17.
Int J Cancer ; 122(7): 1675-8, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18058797

RESUMEN

Recent studies have assessed the role of low molecular weight protein tyrosine phosphatase (LMW-PTP) in cell transformation and tumour onset and progression, observing a significant increase in the expression of LMW-PTP mRNA and protein in human breast, colon, bladder and kidney tumour samples. Moreover, its enhanced expression is generally prognostic of a more aggressive cancer. To better understand the role of this protein during colon carcinogenesis and to study whether its overexpression is also observed in earlier phases of carcinogenesis, we studied its expression in colon tumours, induced in rats by treatment with 1,2-dimethylhydrazine (DMH), an animal model that resemble the sequential formation of histopathological lesions of spontaneous carcinogenesis in humans. The results show a significant increase in LMW-PTP expression in adenocarcinomas, suggesting that this phenomenon is associated with the onset of malignancy. Moreover a significant overexpression of LMW-PTP transcript is associated with tumours originating in the proximal (right) part of the colon, confirming an observation already reported for human colon cancer.


Asunto(s)
Adenocarcinoma/enzimología , Neoplasias del Colon/enzimología , Proteínas Tirosina Fosfatasas/metabolismo , 1,2-Dimetilhidrazina , Adenocarcinoma/inducido químicamente , Animales , Carcinógenos , Neoplasias del Colon/inducido químicamente , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Masculino , Peso Molecular , Ratas , Ratas Endogámicas F344 , Regulación hacia Arriba
18.
Hepatology ; 46(1): 130-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17526025

RESUMEN

UNLABELLED: Adiponectin/ACRP30 is an adipose tissue-derived hormone with antiatherogenic, antidiabetic, and insulin-sensitizing properties. Although the metabolic effects of adiponectin on glucose and lipid metabolism are well known, the signaling pathways triggered by adiponectin receptors remain to be elucidated. We report evidence that in hepatic cells, adiponectin stimulation produces a transient burst of reactive oxygen species (ROS) through activation of the small GTPase Rac1 and 5-lypoxigenase. Furthermore, adiponectin-induced oxidants cause the oxidation/inhibition of protein-tyrosine phosphatase (PTP) 1B, one of the major phosphotyrosine phosphatases involved in the control of insulin receptor phosphorylation. Adiponectin causes increased association of PTP1B to insulin receptor and the oxidation/inhibition of the phosphatase, ultimately provoking the ligand-independent trans-phosphorylation of insulin receptor. We also report evidence that redox signaling plays a key role in both mitogen-activated protein kinase activation and hepatic glucose consumption induced by adiponectin. CONCLUSION: These results point to ROS as critical regulators of the cross-talk between adiponectin and insulin pathways and provide a redox-based molecular mechanism for the insulin-sensitizing function of adiponectin.


Asunto(s)
Adiponectina/farmacología , Receptor de Insulina/fisiología , Línea Celular , Humanos , Peróxido de Hidrógeno/metabolismo , Hígado , Oxidación-Reducción , Monoéster Fosfórico Hidrolasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptor de Insulina/efectos de los fármacos , Activación Transcripcional , Proteína de Unión al GTP rac1/efectos de los fármacos , Proteína de Unión al GTP rac1/metabolismo
19.
J Biol Chem ; 282(27): 19619-28, 2007 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-17449913

RESUMEN

Eph receptors and ephrin ligands are widely expressed in epithelial cells and mediate cell repulsive motility through heterotypic cell-cell interactions. Several Ephs, including EphA2, are greatly overexpressed in certain tumors, in correlation with poor prognosis and high vascularity in cancer tissues. The ability of several Eph receptors to regulate cell migration and invasion likely contribute to tumor progression and metastasis. We report here that in prostatic carcinoma cells ephrinA1 elicits a repulsive response that is executed through a Rho-dependent actino/myosin contractility activation, ultimately leading to retraction of the cell body. This appears to occur through assembly of an EphA2-associated complex involving the two kinases Src and focal adhesion kinase (FAK). EphrinA1-mediated repulsion leads to the selective phosphorylation of Tyr-576/577 of FAK, enhancing FAK kinase activity. The repulsive response elicited by ephrinA1 in prostatic carcinoma cells is mainly driven by a Rho-mediated phosphorylation of myosin light chain II, in which Src and FAK activation are required steps. Consequently, Src and FAK are upstream regulators of the overall response induced by ephrinA1/EphA2, instructing cells to retract the cell body and to move away, probably facilitating dissemination and tissue invasion of ephrin-sensitive carcinomas.


Asunto(s)
Actinas/metabolismo , Miosinas Cardíacas/metabolismo , Movimiento Celular , Efrina-A1/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Cadenas Ligeras de Miosina/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Familia-src Quinasas/metabolismo , Línea Celular Tumoral , Efrina-A2/metabolismo , Humanos , Masculino , Complejos Multiproteicos/metabolismo , Metástasis de la Neoplasia , Fosforilación , Neoplasias de la Próstata/patología , Procesamiento Proteico-Postraduccional , Receptor EphA1/metabolismo
20.
Biochim Biophys Acta ; 1770(5): 753-62, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17296269

RESUMEN

We mutated Trp(134) and Tyr(135) of the yeast LMW-PTP to explore their catalytic roles, demonstrating that the mutations of Trp(134) to Tyr or Ala, and Tyr(135) to Ala, all interfere with the formation of the phosphorylenzyme intermediate, a phenomenon that can be seen by the decrease in the kinetic constant of the chemical step (k(3)). Furthermore, we noted that the Trp(134) to Ala mutation causes a dramatic drop in k(cat)/K(m) and a slight enhancement of the dissociation constant K(s). The conservative mutant W134Y shows a k(cat)/K(m) very close to that of wild type, probably compensating the two-fold decrease of k(3) with an increase in substrate affinity. The Y135A mutation enhances the substrate affinity, but reduces the enzyme phosphorylation rate. The replacement of Trp(134) with alanine interferes with the partition between phosphorylenzyme hydrolysis and phosphotransfer from the phosphorylenzyme to glycerol and abolish the enzyme activation by adenine. Finally, we found that mutation of Trp(134) to Ala causes a dramatic change in the pH-rate profile that becomes similar to that of the D132A mutant, suggesting that an aromatic residue in position 134 is necessary to assist the proper positioning of the proton donor in the transition state of the chemical step.


Asunto(s)
Aminoácidos Aromáticos/genética , Mutagénesis Sitio-Dirigida , Proteínas Tirosina Fosfatasas/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/genética , Alanina/metabolismo , Sustitución de Aminoácidos , Aminoácidos Aromáticos/química , Sitios de Unión , Catálisis , Concentración de Iones de Hidrógeno , Cinética , Modelos Moleculares , Fosforilación , Proteínas Tirosina Fosfatasas/genética , Proteínas de Saccharomyces cerevisiae/genética , Especificidad por Sustrato/genética , Triptófano/química , Triptófano/genética , Tirosina/química , Tirosina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...