Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Pathol ; 263(4-5): 466-481, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38924548

RESUMEN

The E3 ubiquitin ligase thyroid hormone receptor interacting protein 12 (TRIP12) has been implicated in pancreatic adenocarcinoma (PDAC) through its role in mediating the degradation of pancreas transcription factor 1a (PTF1a). PTF1a is a transcription factor essential for the acinar differentiation state that is notably diminished during the early steps of pancreatic carcinogenesis. Despite these findings, the direct involvement of TRIP12 in the onset of pancreatic cancer has yet to be established. In this study, we demonstrated that TRIP12 protein was significantly upregulated in human pancreatic preneoplastic lesions. Furthermore, we observed that TRIP12 overexpression varied within PDAC samples and PDAC-derived cell lines. We further demonstrated that TRIP12 was required for PDAC-derived cell growth and for the expression of E2F-targeted genes. Acinar-to-ductal cell metaplasia (ADM) is a reversible process that reflects the high plasticity of acinar cells. ADM becomes irreversible in the presence of oncogenic Kras mutations and leads to the formation of preneoplastic lesions. Using two genetically modified mouse models, we showed that a loss of TRIP12 prevented acini from developing ADM in response to pancreatic injury. With two additional mouse models, we further discovered that a depletion of TRIP12 prevented the formation of KrasG12D-induced preneoplastic lesions and impaired metastasis formation in the presence of mutated KrasG12D and Trp53R172H genes. In summary our study identified an overexpression of TRIP12 from the early stages of pancreatic carcinogenesis and proposed this E3 ubiquitin ligase as a novel regulator of acinar plasticity with an important dual role in initiation and metastatic steps of PDAC. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Asunto(s)
Células Acinares , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Ubiquitina-Proteína Ligasas , Animales , Neoplasias Pancreáticas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/enzimología , Humanos , Células Acinares/patología , Células Acinares/metabolismo , Células Acinares/enzimología , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Carcinoma Ductal Pancreático/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/enzimología , Metaplasia/patología , Metaplasia/metabolismo , Plasticidad de la Célula , Carcinogénesis/genética , Carcinogénesis/metabolismo , Ratones , Línea Celular Tumoral , Proliferación Celular , Ratones Noqueados , Regulación Neoplásica de la Expresión Génica , Lesiones Precancerosas/patología , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/enzimología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Transformación Celular Neoplásica/metabolismo , Proteínas Portadoras
2.
J Cell Sci ; 134(3)2021 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-33536247

RESUMEN

The class I phosphoinositide 3-kinase (PI3K) catalytic subunits p110α and p110ß are ubiquitously expressed but differently targeted in tumours. In cancer, PIK3CB (encoding p110ß) is seldom mutated compared with PIK3CA (encoding p110α) but can contribute to tumorigenesis in certain PTEN-deficient tumours. The underlying molecular mechanisms are, however, unclear. We have previously reported that p110ß is highly expressed in endometrial cancer (EC) cell lines and at the mRNA level in primary patient tumours. Here, we show that p110ß protein levels are high in both the cytoplasmic and nuclear compartments in EC cells. Moreover, high nuclear:cytoplasmic staining ratios were detected in high-grade primary tumours. High levels of phosphatidylinositol (3,4,5)-trisphosphate [PtdIns(3,4,5)P3] were measured in the nucleus of EC cells, and pharmacological and genetic approaches showed that its production was partly dependent upon p110ß activity. Using immunofluorescence staining, p110ß and PtdIns(3,4,5)P3 were localised in the nucleolus, which correlated with high levels of 47S pre-rRNA. p110ß inhibition led to a decrease in both 47S rRNA levels and cell proliferation. In conclusion, these results present a nucleolar role for p110ß that may contribute to tumorigenesis in EC.This article has an associated First Person interview with Fatemeh Mazloumi Gavgani, joint first author of the paper.


Asunto(s)
Neoplasias Endometriales , Fosfatidilinositol 3-Quinasa , Proliferación Celular/genética , Neoplasias Endometriales/genética , Femenino , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Regulación hacia Arriba/genética
3.
Nat Rev Mol Cell Biol ; 11(5): 329-41, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20379207

RESUMEN

Phosphoinositide 3-kinases (PI3Ks) function early in intracellular signal transduction pathways and affect many biological functions. A further level of complexity derives from the existence of eight PI3K isoforms, which are divided into class I, class II and class III PI3Ks. PI3K signalling has been implicated in metabolic control, immunity, angiogenesis and cardiovascular homeostasis, and is one of the most frequently deregulated pathways in cancer. PI3K inhibitors have recently entered clinical trials in oncology. A better understanding of how the different PI3K isoforms are regulated and control signalling could uncover their roles in pathology and reveal in which disease contexts their blockade could be most beneficial.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Animales , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Mutación/genética , Neoplasias/enzimología , Neoplasias/genética , Fosfatidilinositol 3-Quinasas/química , Fosfatidilinositol 3-Quinasas/genética
4.
Biochem J ; 478(6): 1199-1225, 2021 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-33740047

RESUMEN

PI3Ks are important lipid kinases that produce phosphoinositides phosphorylated in position 3 of the inositol ring. There are three classes of PI3Ks: class I PI3Ks produce PIP3 at plasma membrane level. Although D. melanogaster and C. elegans have only one form of class I PI3K, vertebrates have four class I PI3Ks called isoforms despite being encoded by four different genes. Hence, duplication of these genes coincides with the acquisition of coordinated multi-organ development. Of the class I PI3Ks, PI3Kα and PI3Kß, encoded by PIK3CA and PIK3CB, are ubiquitously expressed. They present similar putative protein domains and share PI(4,5)P2 lipid substrate specificity. Fifteen years after publication of their first isoform-selective pharmacological inhibitors and genetically engineered mouse models (GEMMs) that mimic their complete and specific pharmacological inhibition, we review the knowledge gathered in relation to the redundant and selective roles of PI3Kα and PI3Kß. Recent data suggest that, further to their redundancy, they cooperate for the integration of organ-specific and context-specific signal cues, to orchestrate organ development, physiology, and disease. This knowledge reinforces the importance of isoform-selective inhibitors in clinical settings.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Fosfatidilinositoles/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Humanos , Fosforilación , Transducción de Señal , Especificidad por Sustrato
5.
Genes Dev ; 28(23): 2621-35, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25452273

RESUMEN

Increased PI 3-kinase (PI3K) signaling in pancreatic ductal adenocarcinoma (PDAC) correlates with poor prognosis, but the role of class I PI3K isoforms during its induction remains unclear. Using genetically engineered mice and pharmacological isoform-selective inhibitors, we found that the p110α PI3K isoform is a major signaling enzyme for PDAC development induced by a combination of genetic and nongenetic factors. Inactivation of this single isoform blocked the irreversible transition of exocrine acinar cells into pancreatic preneoplastic ductal lesions by oncogenic Kras and/or pancreatic injury. Hitting the other ubiquitous isoform, p110ß, did not prevent preneoplastic lesion initiation. p110α signaling through small GTPase Rho and actin cytoskeleton controls the reprogramming of acinar cells and regulates cell morphology in vivo and in vitro. Finally, p110α was necessary for pancreatic ductal cancers to arise from Kras-induced preneoplastic lesions by increasing epithelial cell proliferation in the context of mutated p53. Here we identify an in vivo context in which p110α cellular output differs depending on the epithelial transformation stage and demonstrate that the PI3K p110α is required for PDAC induced by oncogenic Kras, the key driver mutation of PDAC. These data are critical for a better understanding of the development of this lethal disease that is currently without efficient treatment.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/fisiopatología , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/fisiopatología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Animales , Animales Modificados Genéticamente , Proliferación Celular , Células Epiteliales/citología , Silenciador del Gen , Humanos , Ratones , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Transducción de Señal
6.
Int J Mol Sci ; 23(18)2022 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-36142542

RESUMEN

Despite decades of effort in understanding pancreatic ductal adenocarcinoma (PDAC), there is still a lack of innovative targeted therapies for this devastating disease. Herein, we report the expression of apelin and its receptor, APJ, in human pancreatic adenocarcinoma and its protumoral function. Apelin and APJ protein expression in tumor tissues from patients with PDAC and their spatiotemporal pattern of expression in engineered mouse models of PDAC were investigated by immunohistochemistry. Apelin signaling function in tumor cells was characterized in pancreatic tumor cell lines by Western blot as well as proliferation, migration assays and in murine orthotopic xenograft experiments. In premalignant lesions, apelin was expressed in epithelial lesions whereas APJ was found in isolated cells tightly attached to premalignant lesions. However, in the invasive stage, apelin and APJ were co-expressed by tumor cells. In human tumor cells, apelin induced a long-lasting activation of PI3K/Akt, upregulated ß-catenin and the oncogenes c-myc and cyclin D1 and promoted proliferation, migration and glucose uptake. Apelin receptor blockades reduced cancer cell proliferation along with a reduction in pancreatic tumor burden. These findings identify the apelin signaling pathway as a new actor for PDAC development and a novel therapeutic target for this incurable disease.


Asunto(s)
Adenocarcinoma , Receptores de Apelina/metabolismo , Apelina/metabolismo , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Adenocarcinoma/patología , Animales , Carcinoma Ductal Pancreático/genética , Ciclina D1/metabolismo , Glucosa , Humanos , Ratones , Oncogenes , Neoplasias Pancreáticas/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Neoplasias Pancreáticas
7.
Phys Rev Lett ; 125(12): 128103, 2020 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-33016731

RESUMEN

While many cellular mechanisms leading to chemotherapeutic resistance have been identified, there is an increasing realization that tumor-stroma interactions also play an important role. In particular, mechanical alterations are inherent to solid cancer progression and profoundly impact cell physiology. Here, we explore the influence of compressive stress on the efficacy of chemotherapeutics in pancreatic cancer spheroids. We find that increased compressive stress leads to decreased drug efficacy. Theoretical modeling and experiments suggest that mechanical stress decreases cell proliferation which in turn reduces the efficacy of chemotherapeutics that target proliferating cells. Our work highlights a mechanical form of drug resistance and suggests new strategies for therapy.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Modelos Biológicos , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Resistencia a Antineoplásicos , Humanos , Estrés Mecánico , Gemcitabina
8.
Arterioscler Thromb Vasc Biol ; 38(6): 1346-1357, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29650694

RESUMEN

OBJECTIVE: Estrogens exert beneficial effect on the blood vascular system. However, their role on the lymphatic system has been poorly investigated. We studied the protective effect of the 17ß estradiol-the most potent endogenous estrogen-in lymphedema-a lymphatic dysfunction, which results in a massive fluid and fat accumulation in the limb. APPROACH AND RESULTS: Screening of DNA motifs able to mobilize ERs (estrogen receptors) and quantitative real-time polymerase chain reaction analysis revealed that estradiol promotes transcriptional activation of lymphangiogenesis-related gene expression including VEGF (vascular endothelial growth factor)-D, VEGFR (VEGF receptor)-3, lyve-1, and HASs (hyaluronan synthases). Using an original model of secondary lymphedema, we observed a protective effect of estradiol on lymphedema by reducing dermal backflow-a representative feature of the pathology. Blocking ERα by tamoxifen-the selective estrogen modulator-led to a remodeling of the lymphatic network associated with a strong lymphatic leakage. Moreover, the protection of lymphedema by estradiol treatment was abrogated by the endothelial deletion of the receptor ERα in Tie2-Cre; ERαlox/lox mice, which exhibit dilated lymphatic vessels. This remodeling correlated with a decrease in lymphangiogenic gene expression. In vitro, blocking ERα by tamoxifen in lymphatic endothelial cells decreased cell-cell junctions, inhibited migration and sprouting, and resulted in an inhibition of Erk but not of Akt phosphorylation. CONCLUSIONS: Estradiol protection from developing lymphedema is mediated by an activation of its receptor ERα and is antagonized by tamoxifen. These findings reveal a new facet of the estrogen influence in the management of the lymphatic system and provide more evidence that secondary lymphedema is worsened by hormone therapy.


Asunto(s)
Linfedema del Cáncer de Mama/prevención & control , Estradiol/administración & dosificación , Receptor alfa de Estrógeno/agonistas , Terapia de Reemplazo de Hormonas , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Animales , Linfedema del Cáncer de Mama/metabolismo , Linfedema del Cáncer de Mama/patología , Linfedema del Cáncer de Mama/fisiopatología , Modelos Animales de Enfermedad , Implantes de Medicamentos , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Vasos Linfáticos/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Ovariectomía , Fosforilación , Moduladores Selectivos de los Receptores de Estrógeno/toxicidad , Tamoxifeno/toxicidad
9.
PLoS Genet ; 11(7): e1005304, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26132308

RESUMEN

The organismal roles of the ubiquitously expressed class I PI3K isoform p110ß remain largely unknown. Using a new kinase-dead knockin mouse model that mimics constitutive pharmacological inactivation of p110ß, we document that full inactivation of p110ß leads to embryonic lethality in a substantial fraction of mice. Interestingly, the homozygous p110ß kinase-dead mice that survive into adulthood (maximum ~26% on a mixed genetic background) have no apparent phenotypes, other than subfertility in females and complete infertility in males. Systemic inhibition of p110ß results in a highly specific blockade in the maturation of spermatogonia to spermatocytes. p110ß was previously suggested to signal downstream of the c-kit tyrosine kinase receptor in germ cells to regulate their proliferation and survival. We now report that p110ß also plays a germ cell-extrinsic role in the Sertoli cells (SCs) that support the developing sperm, with p110ß inactivation dampening expression of the SC-specific Androgen Receptor (AR) target gene Rhox5, a homeobox gene critical for spermatogenesis. All extragonadal androgen-dependent functions remain unaffected by global p110ß inactivation. In line with a crucial role for p110ß in SCs, selective inactivation of p110ß in these cells results in male infertility. Our study is the first documentation of the involvement of a signalling enzyme, PI3K, in the regulation of AR activity during spermatogenesis. This developmental pathway may become active in prostate cancer where p110ß and AR have previously been reported to functionally interact.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Fertilidad/fisiología , Infertilidad Masculina/genética , Receptores Androgénicos/metabolismo , Células de Sertoli/metabolismo , Animales , Blastocisto/citología , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasa Clase I/genética , Femenino , Proteínas de Homeodominio/genética , Infertilidad Femenina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mórula/citología , Receptores Androgénicos/genética , Transducción de Señal/genética , Espermatogénesis/genética , Factores de Transcripción/genética , Transcripción Genética/genética
10.
Br J Cancer ; 113(11): 1590-8, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26512875

RESUMEN

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignancies with a mortality that is almost identical to incidence. Because early detected PDAC is potentially curable, blood-based biomarkers that could detect currently developing neoplasia would improve patient survival and management. PDAC develops from pancreatic intraepithelial neoplasia (PanIN) lesions, graded from low grade (PanIN1) to high grade (PanIN3). We made the hypothesis that specific proteomic signatures from each precancerous stage exist and are detectable in plasma. METHODS: We explored the peptide profiles of microdissected PanIN cells and of plasma samples corresponding to the different PanIN grade from genetically engineered mouse models of PDAC using capillary electrophoresis coupled to mass spectrometry (CE-MS) and Chip-MS/MS. RESULTS: We successfully characterised differential peptides profiles from PanIN microdissected cells. We found that plasma from tumor-bearing mice and age-matched controls exhibit discriminative peptide signatures. We also determined plasma peptide signatures corresponding to low- and high-grade precancerous step present in the mice pancreas using the two mass spectrometry technologies. Importantly, we identified biomarkers specific of PanIN3. CONCLUSIONS: We demonstrate that benign and advanced PanIN lesions display distinct plasma peptide patterns. This strongly supports the perspectives of developing a non-invasive screening test for prediction and early detection of PDAC.


Asunto(s)
Biomarcadores de Tumor/sangre , Carcinoma in Situ/sangre , Carcinoma Ductal Pancreático/sangre , Proteínas de Neoplasias/sangre , Neoplasias Pancreáticas/sangre , Péptidos/sangre , Lesiones Precancerosas/sangre , Animales , Biomarcadores de Tumor/análisis , Carcinoma in Situ/química , Carcinoma in Situ/patología , Carcinoma Ductal Pancreático/química , Modelos Animales de Enfermedad , Ratones , Proteínas de Neoplasias/análisis , Neoplasias Pancreáticas/química , Péptidos/análisis , Lesiones Precancerosas/química , Lesiones Precancerosas/patología , Análisis por Matrices de Proteínas , Proteoma/análisis
11.
Nature ; 453(7195): 662-6, 2008 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-18449193

RESUMEN

Phosphoinositide 3-kinases (PI3Ks) signal downstream of multiple cell-surface receptor types. Class IA PI3K isoforms couple to tyrosine kinases and consist of a p110 catalytic subunit (p110alpha, p110beta or p110delta), constitutively bound to one of five distinct p85 regulatory subunits. PI3Ks have been implicated in angiogenesis, but little is known about potential selectivity among the PI3K isoforms and their mechanism of action in endothelial cells during angiogenesis in vivo. Here we show that only p110alpha activity is essential for vascular development. Ubiquitous or endothelial cell-specific inactivation of p110alpha led to embryonic lethality at mid-gestation because of severe defects in angiogenic sprouting and vascular remodelling. p110alpha exerts this critical endothelial cell-autonomous function by regulating endothelial cell migration through the small GTPase RhoA. p110alpha activity is particularly high in endothelial cells and preferentially induced by tyrosine kinase ligands (such as vascular endothelial growth factor (VEGF)-A). In contrast, p110beta in endothelial cells signals downstream of G-protein-coupled receptor (GPCR) ligands such as SDF-1alpha, whereas p110delta is expressed at low level and contributes only minimally to PI3K activity in endothelial cells. These results provide the first in vivo evidence for p110-isoform selectivity in endothelial PI3K signalling during angiogenesis.


Asunto(s)
Movimiento Celular , Células Endoteliales/citología , Células Endoteliales/enzimología , Neovascularización Fisiológica , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Células Cultivadas , Fosfatidilinositol 3-Quinasa Clase I , Femenino , Humanos , Ratones , Fosfatidilinositol 3-Quinasas/genética , Interferencia de ARN , Ratas , Transducción de Señal/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Heridas y Lesiones , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
12.
Theranostics ; 14(1): 249-264, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38164151

RESUMEN

Rationale: 17ß-estradiol (E2) can directly promote the growth of ERα-negative cancer cells through activation of endothelial ERα in the tumor microenvironment, thereby increasing a normalized tumor angiogenesis. ERα acts as a transcription factor through its nuclear transcriptional AF-1 and AF-2 transactivation functions, but membrane ERα plays also an important role in endothelium. The present study aims to decipher the respective roles of these two pathways in ERα-negative tumor growth. Moreover, we delineate the actions of tamoxifen, a Selective Estrogen Receptor Modulator (SERM) in ERα-negative tumors growth and angiogenesis, since we recently demonstrated that tamoxifen impacts vasculature functions through complex modulation of ERα activity. Methods: ERα-negative B16K1 cancer cells were grafted into immunocompetent mice mutated for ERα-subfunctions and tumor growths were analyzed in these different models in response to E2 and/or tamoxifen treatment. Furthermore, RNA sequencings were analyzed in endothelial cells in response to these different treatments and validated by RT-qPCR and western blot. Results: We demonstrate that both nuclear and membrane ERα actions are required for the pro-tumoral effects of E2, while tamoxifen totally abrogates the E2-induced in vivo tumor growth, through inhibition of angiogenesis but promotion of vessel normalization. RNA sequencing indicates that tamoxifen inhibits the E2-induced genes, but also initiates a specific transcriptional program that especially regulates angiogenic genes and differentially regulates glycolysis, oxidative phosphorylation and inflammatory responses in endothelial cells. Conclusion: These findings provide evidence that tamoxifen specifically inhibits angiogenesis through a reprogramming of endothelial gene expression via regulation of some transcription factors, that could open new promising strategies to manage cancer therapies affecting the tumor microenvironment of ERα-negative tumors.


Asunto(s)
Neoplasias , Tamoxifeno , Ratones , Animales , Tamoxifeno/farmacología , Receptor alfa de Estrógeno/genética , Receptor alfa de Estrógeno/metabolismo , Células Endoteliales/metabolismo , Angiogénesis , Expresión Génica , Endotelio/metabolismo , Línea Celular Tumoral , Microambiente Tumoral/genética
13.
Biochem J ; 442(1): 151-9, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22150431

RESUMEN

The PI3K (phosphoinositide 3-kinase) pathway is commonly activated in cancer as a consequence of inactivation of the tumour suppressor PTEN (phosphatase and tensin homologue deleted on chromosome 10), a major negative regulator of PI3K signalling. In line with this important role of PTEN, mice that are heterozygous for a PTEN-null allele (PTEN+/− mice) spontaneously develop a variety of tumours in multiple organs. PTEN is a phosphatase with selectivity for PtdIns(3,4,5)P3, which is produced by the class I isoforms of PI3K (p110α, p110ß, p110γ and p110δ). Previous studies indicated that PTEN-deficient cancer cell lines mainly depend on p110ß, and that p110ß, but not p110α, controls mouse prostate cancer development driven by PTEN loss. In the present study, we investigated whether the ubiquitously expressed p110α can also functionally interact with PTEN in cancer. Using genetic mouse models that mimic systemic administration of p110α- or p110ß-selective inhibitors, we confirm that inactivation of p110ß, but not p110α, inhibits prostate cancer development in PTEN+/− mice, but also find that p110α inactivation protects from glomerulonephritis, pheochromocytoma and thyroid cancer induced by PTEN loss. This indicates that p110α can modulate the impact of PTEN loss in disease and tumourigenesis. In primary and immortalized mouse fibroblast cell lines, both p110α and p110ß controlled steady-state PtdIns(3,4,5)P3 levels and Akt signalling induced by heterozygous PTEN loss. In contrast, no correlation was found in primary mouse tissues between PtdIns(3,4,5)P3 levels, PI3K/PTEN genotype and cancer development. Taken together, our results from the present study show that inactivation of either p110α or p110ß can counteract the impact of PTEN inactivation. The potential implications of these findings for PI3K-targeted therapy of cancer are discussed.


Asunto(s)
Fosfohidrolasa PTEN/antagonistas & inhibidores , Fosfatidilinositol 3-Quinasas/metabolismo , Animales , Transformación Celular Neoplásica , Fosfatidilinositol 3-Quinasa Clase I , Isoenzimas/metabolismo , Linfoma/etiología , Masculino , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Neoplasias de la Próstata/etiología , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-akt/fisiología , Neoplasias de la Tiroides/prevención & control
14.
Cancers (Basel) ; 15(3)2023 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-36765741

RESUMEN

Phosphatidylinositol-3-kinase (PI3K) enzymes, producing signaling phosphoinositides at plasma and intracellular membranes, are key in intracellular signaling and vesicular trafficking pathways. PI3K is a family of eight enzymes divided into three classes with various functions in physiology and largely deregulated in cancer. Here, we will review the recent evidence obtained during the last 5 years on the roles of PI3K class I, II and III isoforms in tumor biology and on the anti-tumoral action of PI3K inhibitors in preclinical cancer models. The dependency of tumors to PI3K isoforms is dictated by both genetics and context (e.g., the microenvironment). The understanding of class II/III isoforms in cancer development and progression remains scarce. Nonetheless, the limited available data are consistent and reveal that there is an interdependency between the pathways controlled by all PI3K class members in their role to promote cancer cell proliferation, survival, growth, migration and metabolism. It is unknown whether this feature contributes to partial treatment failure with isoform-selective PI3K inhibitors. Hence, a better understanding of class II/III functions to efficiently inhibit their positive and negative interactions with class I PI3Ks is needed. This research will provide the proof-of-concept to develop combination treatment strategies targeting several PI3K isoforms simultaneously.

15.
Life Sci Alliance ; 6(9)2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37364915

RESUMEN

In living organisms, cells sense mechanical forces (shearing, tensile, and compressive) and respond to those physical cues through a process called mechanotransduction. This process includes the simultaneous activation of biochemical signaling pathways. Recent studies mostly on human cells revealed that compressive forces selectively modulate a wide range of cell behavior, both in compressed and in neighboring less compressed cells. Besides participating in tissue homeostasis such as bone healing, compression is also involved in pathologies, including intervertebral disc degeneration or solid cancers. In this review, we will summarize the current scattered knowledge of compression-induced cell signaling pathways and their subsequent cellular outputs, both in physiological and pathological conditions, such as solid cancers.


Asunto(s)
Mecanotransducción Celular , Neoplasias , Humanos , Transducción de Señal
16.
Lab Chip ; 23(20): 4445-4455, 2023 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-37740366

RESUMEN

Conventional culture conditions are oftentimes insufficient to study tissues, organisms, or 3D multicellular assemblies. They lack both dynamic chemical and mechanical control over the microenvironment. While specific microfluidic devices have been developed to address chemical control, they often do not allow the control of compressive forces emerging when cells proliferate in a confined environment. Here, we present a generic microfluidic device to control both chemical and mechanical compressive forces. This device relies on the use of sliding elements consisting of microfabricated rods that can be inserted inside a microfluidic device. Sliding elements enable the creation of reconfigurable closed culture chambers for the study of whole organisms or model micro-tissues. By confining the micro-tissues, we studied the biophysical impact of growth-induced pressure and showed that this mechanical stress is associated with an increase in macromolecular crowding, shedding light on this understudied type of mechanical stress. Our mechano-chemostat allows the long-term culture of biological samples and can be used to study both the impact of specific conditions as well as the consequences of mechanical compression.


Asunto(s)
Microfluídica , Estrés Mecánico , Presión
17.
Blood ; 115(10): 2008-13, 2010 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-20065293

RESUMEN

During platelet activation, phosphoinositide 3-kinases (PI3Ks) produce lipid second messengers participating in the regulation of functional responses. Here, we generated a megakaryocyte-restricted p110beta null mouse model and demonstrated a critical role of PI3Kbeta in platelet activation via an immunoreceptor tyrosine-based activation motif, the glyco-protein VI-Fc receptor gamma-chain complex, and its contribution in response to G-protein-coupled receptors. Interestingly, the production of phosphatidylinositol 3,4,5-trisphosphate and the activation of protein kinase B/Akt were strongly inhibited in p110beta null platelets stimulated either via immunoreceptor tyrosine-based activation motif or G-protein-coupled receptors. Functional studies showed an important delay in fibrin clot retraction and an almost complete inability of these platelets to adhere onto fibrinogen under flow condition, suggesting that PI3Kbeta is also acting downstream of alpha(IIb)beta(3). In vivo studies showed that these mice have a normal bleeding time and are not protected from acute pulmonary thromboembolism but are resistant to thrombosis after FeCl(3) injury of the carotid, suggesting that PI3Kbeta is a potential target for antithrombotic drugs.


Asunto(s)
Plaquetas/metabolismo , Proteína Oncogénica v-akt/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/fisiología , Trombosis/genética , Animales , Tiempo de Sangría , Plaquetas/enzimología , Plaquetas/patología , Linaje de la Célula/genética , Células Cultivadas , Cloruros , Fosfatidilinositol 3-Quinasa Clase I , Modelos Animales de Enfermedad , Activación Enzimática/genética , Compuestos Férricos , Eliminación de Gen , Predisposición Genética a la Enfermedad , Isoenzimas/genética , Megacariocitos/metabolismo , Megacariocitos/fisiología , Ratones , Ratones Transgénicos , Fosfatos de Fosfatidilinositol/metabolismo , Agregación Plaquetaria/genética , Trombosis/inducido químicamente , Trombosis/enzimología , Trombosis/patología
18.
Proc Natl Acad Sci U S A ; 105(24): 8292-7, 2008 Jun 17.
Artículo en Inglés | MEDLINE | ID: mdl-18544649

RESUMEN

The p110 isoforms of phosphoinositide 3-kinase (PI3K) are acutely regulated by extracellular stimuli. The class IA PI3K catalytic subunits (p110alpha, p110beta, and p110delta) occur in complex with a Src homology 2 (SH2) domain-containing p85 regulatory subunit, which has been shown to link p110alpha and p110delta to Tyr kinase signaling pathways. The p84/p101 regulatory subunits of the p110gamma class IB PI3K lack SH2 domains and instead couple p110gamma to G protein-coupled receptors (GPCRs). Here, we show, using small-molecule inhibitors with selectivity for p110beta and cells derived from a p110beta-deficient mouse line, that p110beta is not a major effector of Tyr kinase signaling but couples to GPCRs. In macrophages, both p110beta and p110gamma contributed to Akt activation induced by the GPCR agonist complement 5a, but not by the Tyr kinase ligand colony-stimulating factor-1. In fibroblasts, which express p110beta but not p110gamma, p110beta mediated Akt activation by the GPCR ligands stromal cell-derived factor, sphingosine-1-phosphate, and lysophosphatidic acid but not by the Tyr kinase ligands PDGF, insulin, and insulin-like growth factor 1. Introduction of p110gamma in these cells reduced the contribution of p110beta to GPCR signaling. Taken together, these data show that p110beta and p110gamma can couple redundantly to the same GPCR agonists. p110beta, which shows a much broader tissue distribution than the leukocyte-restricted p110gamma, could thus provide a conduit for GPCR-linked PI3K signaling in the many cell types where p110gamma expression is low or absent.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Fosfatidilinositol 3-Quinasa Clase I , Complemento C5a/farmacología , Fibroblastos/enzimología , Prueba de Complementación Genética , Isoenzimas/genética , Isoenzimas/metabolismo , Ligandos , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/enzimología , Ratones , Ratones Mutantes , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Transducción de Señal
19.
Clin Res Hepatol Gastroenterol ; 45(1): 101473, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32593694

RESUMEN

Pancreatic ductal adenocarcinoma PDAC is a complex disease with an important diversity of genetic alterations found between patients. KRAS mutation is considered as a major oncogenic driver in this cancer (around 90% of the patients), but there exists different KRAS mutation types. The type of KRAS mutation was recently shown to be of importance to detect signalling vulnerabilities in a subset of PDAC patients. We comment on these innovative results and discuss their importance when designing clinical trials with PI3K targeted therapies in this cancer.


Asunto(s)
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/genética , Humanos , Mutación , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética
20.
Mol Cancer Ther ; 20(12): 2433-2445, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34552006

RESUMEN

The PI3K pathway is highly active in human cancers. The four class I isoforms of PI3K are activated by distinct mechanisms leading to a common downstream signaling. Their downstream redundancy is thought to be responsible for treatment failures of PI3K inhibitors. We challenged this concept, by mapping the differential phosphoproteome evolution in response to PI3K inhibitors with different isoform-selectivity patterns in pancreatic cancer, a disease currently without effective therapy. In this cancer, the PI3K signal was shown to control cell proliferation. We compared the effects of LY294002 that inhibit with equal potency all class I isoenzymes and downstream mTOR with the action of inhibitors with higher isoform selectivity toward PI3Kα, PI3Kß, or PI3Kγ (namely, A66, TGX-221 and AS-252424). A bioinformatics global pathway analysis of phosphoproteomics data allowed us to identify common and specific signals activated by PI3K inhibitors supported by the biological data. AS-252424 was the most effective treatment and induced apoptotic pathway activation as well as the highest changes in global phosphorylation-regulated cell signal. However, AS-252424 treatment induced reactivation of Akt, therefore decreasing the treatment outcome on cell survival. Reversely, AS-252424 and A66 combination treatment prevented p-Akt reactivation and led to synergistic action in cell lines and patient organoids. The combination of clinically approved α-selective BYL-719 with γ-selective IPI-549 was more efficient than single-molecule treatment on xenograft growth. Mapping unique adaptive signaling responses to isoform-selective PI3K inhibition will help to design better combinative treatments that prevent the induction of selective compensatory signals.


Asunto(s)
Neoplasias Pancreáticas/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3/uso terapéutico , Proteómica/métodos , Animales , Línea Celular Tumoral , Resistencia a Medicamentos , Humanos , Ratones , Neoplasias Pancreáticas/patología , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA