Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Int J Mol Sci ; 23(2)2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35055098

RESUMEN

Heterogeneous nuclear ribonucleoproteins (hnRNPs) control gene expression by acting at multiple levels and are often deregulated in epithelial tumors; however, their roles in the fine regulation of cellular reprogramming, specifically in epithelial-mesenchymal transition (EMT), remain largely unknown. Here, we focused on the hnRNP-Q (also known as SYNCRIP), showing by molecular analysis that in hepatocytes it acts as a "mesenchymal" gene, being induced by TGFß and modulating the EMT. SYNCRIP silencing limits the induction of the mesenchymal program and maintains the epithelial phenotype. Notably, in HCC invasive cells, SYNCRIP knockdown induces a mesenchymal-epithelial transition (MET), negatively regulating their mesenchymal phenotype and significantly impairing their migratory capacity. In exploring possible molecular mechanisms underlying these observations, we identified a set of miRNAs (i.e., miR-181-a1-3p, miR-181-b1-3p, miR-122-5p, miR-200a-5p, and miR-let7g-5p), previously shown to exert pro- or anti-EMT activities, significantly impacted by SYNCRIP interference during EMT/MET dynamics and gathered insights, suggesting the possible involvement of this RNA binding protein in their transcriptional regulation.


Asunto(s)
Carcinoma Hepatocelular/etiología , Transformación Celular Neoplásica/genética , Transición Epitelial-Mesenquimal/genética , Hepatocitos/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/genética , Neoplasias Hepáticas/etiología , Animales , Biomarcadores , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Susceptibilidad a Enfermedades , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Hepatocitos/patología , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Ratones , MicroARNs/genética , Fenotipo , Interferencia de ARN , Proteínas de Unión al ARN
2.
Liver Int ; 35(2): 302-10, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24766136

RESUMEN

The complex spatial and paracrine relationships between the various liver histotypes are essential for proper functioning of the hepatic parenchymal cells. Only within a correct tissue organization, in fact, they stably maintain their identity and differentiated phenotype. The loss of histotype identity, which invariably occurs in the primary hepatocytes in culture, or in vivo in particular pathological conditions (fibrosis and tumours), is mainly because of the phenomenon of epithelial-to-mesenchymal transition (EMT). The EMT process, that occurs in the many epithelial cells, appears to be driven by a number of general, non-tissue-specific, master transcriptional regulators. The reverse process, the mesenchymal-to-epithelial transition (MET), as yet much less characterized at a molecular level, restores specific epithelial identities, and thus must include tissue-specific master elements. In this review, we will summarize the so far unveiled events of EMT/MET occurring in liver cells. In particular, we will focus on hepatocyte and describe the pivotal role in the control of EMT/MET dynamics exerted by a tissue-specific molecular mini-circuitry. Recent evidence, indeed, highlighted as two transcriptional factors, the master gene of EMT Snail, and the master gene of hepatocyte differentiation HNF4α, exhorting a direct reciprocal repression, act as pivotal elements in determining opposite cellular outcomes. The different balances between these two master regulators, further integrated by specific microRNAs, in fact, were found responsible for the EMT/METs dynamics as well as for the preservation of both hepatocyte and stem/precursor cells identity and differentiation. Overall, these findings impact the maintenance of stem cells and differentiated cells both in in vivo EMT/MET physio-pathological processes as well as in culture.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Regulación de la Expresión Génica/fisiología , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/citología , Modelos Biológicos , Fenotipo , Factores de Transcripción/metabolismo , Factor Nuclear 4 del Hepatocito/uso terapéutico , Hepatocitos/fisiología , Humanos , MicroARNs/metabolismo , Factores de Transcripción de la Familia Snail
3.
J Hepatol ; 58(1): 65-72, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22960426

RESUMEN

BACKGROUND & AIMS: The tumor fate derives from cell autonomous properties and niche microenvironmental cues. The transforming growth factor ß (TGFß) is a major microenvironmental factor for hepatocellular carcinoma (HCC) influencing tumor dedifferentiation, induction of epithelial-to-mesenchymal transition (EMT) and acquisition of metastatic properties. The loss of the transcriptional factor HNF4α is a predominant mechanism through which HCCs progress to a more aggressive phenotype; its re-expression, reducing tumor formation and repressing EMT program, has been suggested as a therapeutic tool for HCC gene therapy. We investigated the influence of TGFß on the anti-EMT and tumor suppressor HNF4α activity. METHODS: Cell motility and invasion were analyzed by wound healing and invasion assays. EMT was evaluated by RT-qPCR and immunofluorescence. ChIP and EMSA assays were utilized for investigation of the HNF4α DNA binding activity. HNF4α post-translational modifications (PTMs) were assessed by 2-DE analysis. GSK3ß activity was modulated by chemical inhibition and constitutive active mutant expression. RESULTS: We demonstrated that the presence of TGFß impairs the efficiency of HNF4α as tumor suppressor. We found that TGFß induces HNF4α PTMs that correlate with the early loss of HNF4α DNA binding activity on target gene promoters. Furthermore, we identified the GSK3ß kinase as one of the TGFß targets mediating HNF4α functional inactivation: GSK3ß chemical inhibition results in HNF4α DNA binding impairment while a constitutively active GSK3ß mutant impairs the TGFß-induced inhibitory effect on HNF4α tumor suppressor activity. CONCLUSIONS: Our data identify in the dominance of TGFß a limit for the HNF4α-mediated gene therapy of HCC.


Asunto(s)
Carcinoma Hepatocelular , Terapia Genética , Glucógeno Sintasa Quinasa 3/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Neoplasias Hepáticas , Factor de Crecimiento Transformador beta/metabolismo , Animales , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/secundario , Carcinoma Hepatocelular/terapia , Línea Celular Transformada , Línea Celular Tumoral , Movimiento Celular/fisiología , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Genes Supresores de Tumor/fisiología , Glucógeno Sintasa Quinasa 3 beta , Células Hep G2 , Factor Nuclear 4 del Hepatocito/genética , Hepatocitos/citología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/terapia , Ratones , Factor de Crecimiento Transformador beta/genética , Microambiente Tumoral/genética , Microambiente Tumoral/fisiología
4.
J Exp Clin Cancer Res ; 42(1): 147, 2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37308974

RESUMEN

The epithelial-to-mesenchymal transition (EMT) is a trans-differentiation process that endows epithelial cells with mesenchymal properties, including motility and invasion capacity; therefore, its aberrant reactivation in cancerous cells represents a critical step to gain a metastatic phenotype. The EMT is a dynamic program of cell plasticity; many partial EMT states can be, indeed, encountered and the full inverse mesenchymal-to-epithelial transition (MET) appears fundamental to colonize distant secondary sites. The EMT/MET dynamics is granted by a fine modulation of gene expression in response to intrinsic and extrinsic signals. In this complex scenario, long non-coding RNAs (lncRNAs) emerged as critical players. This review specifically focuses on the lncRNA HOTAIR, as a master regulator of epithelial cell plasticity and EMT in tumors. Molecular mechanisms controlling its expression in differentiated as well as trans-differentiated epithelial cells are highlighted here. Moreover, current knowledge about HOTAIR pleiotropic functions in regulation of both gene expression and protein activities are described. Furthermore, the relevance of the specific HOTAIR targeting and the current challenges of exploiting this lncRNA for therapeutic approaches to counteract the EMT are discussed.


Asunto(s)
ARN Largo no Codificante , Diferenciación Celular , Plasticidad de la Célula , Células Epiteliales , Transición Epitelial-Mesenquimal , Humanos , Animales
5.
Cell Death Dis ; 14(1): 32, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36650140

RESUMEN

YES-associated protein (YAP) is a transcriptional cofactor with a key role in the regulation of several physio-pathological cellular processes, by integrating multiple cell autonomous and microenvironmental cues. YAP is the main downstream effector of the Hippo pathway, a tumor-suppressive signaling able to transduce several extracellular signals. The Hippo pathway acts restraining YAP activity, since its activation induces YAP phosphorylation and cytoplasmic sequestration. However, recent observations indicate that YAP activity can be also modulated by Hippo independent/integrating pathways, still largely unexplored. In this study, we demonstrated the role of the extracellular signal-regulated kinase 5 (ERK5)/mitogen-activated protein kinase in the regulation of YAP activity. By means of ERK5 inhibition/silencing and overexpression experiments, and by using as model liver stem cells, hepatocytes, and hepatocellular carcinoma (HCC) cell lines, we provided evidence that ERK5 is required for YAP-dependent gene expression. Mechanistically, ERK5 controls the recruitment of YAP on promoters of target genes and its physical interaction with the transcriptional partner TEAD; moreover, it mediates the YAP activation occurring in cell adhesion, migration, and TGFß-induced EMT of liver cells. Furthermore, we demonstrated that ERK5 signaling modulates YAP activity in a LATS1/2-independent manner. Therefore, our observations identify ERK5 as a novel upstream Hippo-independent regulator of YAP activity, thus unveiling a new target for therapeutic approaches aimed at interfering with its function.


Asunto(s)
Hepatocitos , Proteína Quinasa 7 Activada por Mitógenos , Proteínas Señalizadoras YAP , Humanos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Carcinoma Hepatocelular/patología , Proliferación Celular , Neoplasias Hepáticas/patología , Proteína Quinasa 7 Activada por Mitógenos/genética , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Fosfoproteínas/metabolismo , Proteínas Señalizadoras YAP/genética , Proteínas Señalizadoras YAP/metabolismo , Hepatocitos/metabolismo , Células Madre
6.
Hepatology ; 53(6): 2063-74, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21384409

RESUMEN

UNLABELLED: The concept that cellular terminal differentiation is stably maintained once development is complete has been questioned by numerous observations showing that differentiated epithelium may undergo an epithelial-to-mesenchymal transition (EMT) program. EMT and the reverse process, mesenchymal-to-epithelial transition (MET), are typical events of development, tissue repair, and tumor progression. In this study, we aimed to clarify the molecular mechanisms underlying these phenotypic conversions in hepatocytes. Hepatocyte nuclear factor 4α (HNF4α) was overexpressed in different hepatocyte cell lines and the resulting gene expression profile was determined by real-time quantitative polymerase chain reaction. HNF4α recruitment on promoters of both mesenchymal and EMT regulator genes was determined by way of electrophoretic mobility shift assay and chromatin immunoprecipitation. The effect of HNF4α depletion was assessed in silenced cells and in the context of the whole liver of HNF4 knockout animals. Our results identified key EMT regulators and mesenchymal genes as new targets of HNF4α. HNF4α, in cooperation with its target HNF1α, directly inhibits transcription of the EMT master regulatory genes Snail, Slug, and HMGA2 and of several mesenchymal markers. HNF4α-mediated repression of EMT genes induces MET in hepatomas, and its silencing triggers the mesenchymal program in differentiated hepatocytes both in cell culture and in the whole liver. CONCLUSION: The pivotal role of HNF4α in the induction and maintenance of hepatocyte differentiation should also be ascribed to its capacity to continuously repress the mesenchymal program; thus, both HNF4α activator and repressor functions are necessary for the identity of hepatocytes.


Asunto(s)
Diferenciación Celular/fisiología , Células Epiteliales/patología , Factor Nuclear 4 del Hepatocito/fisiología , Hepatocitos/patología , Mesodermo/patología , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Factor Nuclear 1-alfa del Hepatocito/fisiología , Factor Nuclear 4 del Hepatocito/genética , Humanos , Neoplasias Hepáticas/patología , Ratones , Ratones Noqueados , Modelos Animales , Fenotipo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/fisiología
7.
Cells ; 11(10)2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35626752

RESUMEN

Epithelial-to-mesenchymal transition (EMT) is a dynamic program of cell plasticity aberrantly reactivated in cancer. The crosstalk between tumor cells and the tumoral microenvironment (TME) has a pivotal importance for the induction of the EMT and the progression toward a malignant phenotype. Notably, exosomes are key mediators of this crosstalk as vehicles of specific molecular signals that include the class of circular RNAs (circRNAs). This review specifically focuses on the role of exosome-associated circRNAs as key regulators of EMT in cancer. The relevance of these molecules in regulating the intercellular communication in TME and tumor progression is highlighted. Moreover, the here-presented evidence indicates that exosome-associated circRNA modulation should be taken in account for cancer diagnostic and therapeutic approaches.


Asunto(s)
Exosomas , Neoplasias , Transición Epitelial-Mesenquimal/genética , Exosomas/genética , Humanos , Neoplasias/genética , ARN Circular/genética , Microambiente Tumoral/genética
8.
Cell Death Dis ; 13(11): 965, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36396626

RESUMEN

Histone acetylation/deacetylation play an essential role in modifying chromatin structure and in regulating cell plasticity in eukaryotic cells. Therefore, histone deacetylase (HDAC) pharmacological inhibitors are promising tools in the therapy of fibrotic diseases and in cancer. Peritoneal fibrosis is a pathological process characterized by many cellular and molecular alterations, including the acquisition of invasive/pro-fibrotic abilities by mesothelial cells (MCs) through induction of mesothelial to mesenchymal transition (MMT). The aim of this study was to characterize the molecular mechanism of the antifibrotic role of HDAC1 inhibition. Specifically, treatment with MS-275, an HDAC1-3 inhibitor previously known to promote MMT reversal, induced the expression of several TGFBRI mRNA-targeting miRNAs. Among them, miR-769-5p ectopic expression was sufficient to promote MMT reversal and to limit MC migration and invasion, whereas miR-769-5p silencing further enhanced mesenchymal gene expression. These results were confirmed by HDAC1 genetic silencing. Interestingly, miR-769-5p silencing maintained mesenchymal features despite HDAC1 inhibition, thus indicating that it is necessary to drive MMT reversal induced by HDAC1 inhibition. Besides TGFBRI, miR-769-5p was demonstrated to target SMAD2/3 and PAI-1 expression directly. When analyzing molecular mechanisms underlying miR-769-5p expression, we found that the transcription factor Wilms' tumor 1 (WT1), a master gene controlling MC development, binds to the miR-769-5p promoter favoring its expression. Interestingly, both WT1 expression and binding to miR-769-5p promoter were increased by HDAC1 inhibition and attenuated by TGFß1 treatment. Finally, we explored the significance of these observations in the cell-to-cell communication: we evaluated the ability of miR-769-5p to be loaded into extracellular vesicles (EVs) and to promote MMT reversal in recipient mesenchymal-like MCs. Treatment of fibrotic MCs with EVs isolated from miR-769-5p over-expressing MCs promoted the down-regulation of specific mesenchymal targets and the reacquisition of an epithelial-like morphology. In conclusion, we highlighted an HDAC1-WT1-miR-769-5p axis potentially relevant for therapies aimed at counteracting organ fibrosis.


Asunto(s)
Transición Epitelial-Mesenquimal , MicroARNs , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Epitelio/metabolismo , MicroARNs/metabolismo
9.
Nat Cell Biol ; 4(1): 11-9, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11780126

RESUMEN

Phosphorylation of p53 at Ser 46 was shown to regulate p53 apoptotic activity. Here we demonstrate that homeodomain-interacting protein kinase-2 (HIPK2), a member of a novel family of nuclear serine/threonine kinases, binds to and activates p53 by directly phosphorylating it at Ser 46. HIPK2 localizes with p53 and PML-3 into the nuclear bodies and is activated after irradiation with ultraviolet. Antisense inhibition of HIPK2 expression reduces the ultraviolet-induced apoptosis. Furthermore, HIPK2 and p53 cooperate in the activation of p53-dependent transcription and apoptotic pathways. These data define a new functional interaction between p53 and HIPK2 that results in the targeted subcellular localization of p53 and initiation of apoptosis.


Asunto(s)
Apoptosis/genética , Proteínas Portadoras/metabolismo , Proteínas Nucleares , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de la radiación , Proteínas Portadoras/genética , Proteínas Portadoras/efectos de la radiación , Núcleo Celular/genética , Núcleo Celular/metabolismo , Núcleo Celular/ultraestructura , Activación Enzimática/efectos de la radiación , Genes Supresores de Tumor , Humanos , Ratones , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Oligonucleótidos Antisentido , Fosforilación , Proteína de la Leucemia Promielocítica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/efectos de la radiación , Serina , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor , Rayos Ultravioleta
10.
Skeletal Radiol ; 40(5): 609-16, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-20931188

RESUMEN

PURPOSE: To describe the potential value of high-resolution sonography for evaluation of the musculocutaneous nerve (MCN). MATERIALS AND METHODS: The normal anatomy of the MCN was evaluated on three cadaveric limbs and correlated with the US images obtained in 15 healthy subjects. Seven consecutive patients with MCN neuropathy were then evaluated with sonography using 17.5 and 12.5-MHz broadband linear array transducers. All patients had abnormal nerve conduction studies and underwent correlative MR imaging on a 1.5-T system. RESULTS: One-to-one comparison between cadaveric specimens and sonographic images showed that the MCN can be reliably identified from the axilla through the elbow, including the lateral antebrachial cutaneous (LAbC) nerve. In the patients group with MCN neuropathy, sonography allowed detection of a wide spectrum of abnormalities. In 5/7 cases, a spindle neuroma was depicted in continuity with the nerve. In one case, US identified focal swelling of the nerve and in another case US was negative. The neuroma was hyperintense on T2-weighted sequences in 75% of cases. In one patient, the nerve showed Gd-enhancement on fat-suppressed T1-weighted sequences. The nerve was never detected on unenhanced T1-scans. Owing to its small-size and out-of-plane course, the MCN may be more reliably depicted with sonography rather than with MR imaging. CONCLUSIONS: US is promising for evaluating traumatic injuries of the MCN. By providing unique information on the entire course of the nerve, US can be used as a valuable complement of clinical and electrophysiologic findings.


Asunto(s)
Nervio Musculocutáneo/diagnóstico por imagen , Síndromes de Compresión Nerviosa/diagnóstico por imagen , Adulto , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Radiografía , Ultrasonografía
11.
Semin Musculoskelet Radiol ; 14(5): 512-22, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21072729

RESUMEN

Traumatic injury to peripheral nerves is a significant cause of morbidity and disability. Until reinnervation occurs, electrodiagnostic studies cannot differentiate severe axonotmetic lesions (Sunderland class 4) from complete nerve transection or neurotmesis (Sunderland class 5). This limitation is relevant clinically because in cases of neurotmesis an improved outcome may be achieved with an early surgical repair (within 1 week after trauma). High-resolution ultrasound (US) is an efficient modality to visualize injured nerves and is becoming increasingly important among radiologists and surgeons. Magnetic resonance (MR) imaging is complementary to high-resolution US, especially in evaluating deep-seated and proximal nerve segments. This article describes the imaging features of traumatic peripheral nerve lesions. The role of diagnostic imaging in stretching injuries, contusion trauma, penetrating wounds, and after surgery is discussed. A multimodality diagnostic approach including physical examination, electrophysiology, and US and MR imaging allows an accurate evaluation of most peripheral nerves. Imaging assessment of peripheral nerves trauma is useful for the diagnosis, follow-up, and postoperative evaluation.


Asunto(s)
Nervios Periféricos/diagnóstico por imagen , Nervios Periféricos/patología , Enfermedades del Sistema Nervioso Periférico/diagnóstico por imagen , Enfermedades del Sistema Nervioso Periférico/patología , Complicaciones Posoperatorias/diagnóstico por imagen , Complicaciones Posoperatorias/patología , Contusiones/diagnóstico por imagen , Contusiones/patología , Contusiones/cirugía , Humanos , Imagen por Resonancia Magnética/métodos , Traumatismos de los Nervios Periféricos , Enfermedades del Sistema Nervioso Periférico/cirugía , Ultrasonografía , Heridas Penetrantes/diagnóstico por imagen , Heridas Penetrantes/patología , Heridas Penetrantes/cirugía
12.
Semin Musculoskelet Radiol ; 14(3): 344-56, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20539959

RESUMEN

Neuropathies about the ankle and foot may be the cause of chronic pain and disability. In most cases, these conditions derive from mechanical or dynamic compression of a segment of a nerve within a narrow osteofibrous tunnel, an opening in a fibrous structure, or a passageway close to a ligament or a muscle. Although the evaluation of nerve disorders primarily relies on neurological examination and electrophysiology, diagnostic imaging is currently used as a complement to help define the site and etiology of nerve compression and exclude other disease possibly underlying the patient' symptoms. In this article, a review of the anatomical and pathological features of nerve entrapments in the distal lower extremity is presented on ultrasound and magnetic resonance imaging, according to the nerve involved.


Asunto(s)
Enfermedades del Pie/diagnóstico por imagen , Enfermedades del Pie/patología , Pie/inervación , Síndromes de Compresión Nerviosa/diagnóstico por imagen , Síndromes de Compresión Nerviosa/patología , Tobillo/diagnóstico por imagen , Tobillo/inervación , Tobillo/patología , Pie/diagnóstico por imagen , Pie/patología , Humanos , Nervio Peroneo/diagnóstico por imagen , Nervio Peroneo/patología , Nervio Sural/diagnóstico por imagen , Nervio Sural/patología , Nervio Tibial/diagnóstico por imagen , Nervio Tibial/patología , Ultrasonografía
13.
Cell Signal ; 20(11): 2113-8, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18760348

RESUMEN

Extracellular signal-regulated protein kinase 5 (ERK5) is a mitogen-activated protein kinase, specifically activated by MEK5, and involved in the regulation of many cellular functions including proliferation, survival, differentiation and apoptosis. MEK5/ERK5 module is an important element of different signal transduction pathways. The aim of this study was to investigate whether ERK5 participates to the signalling of the multifunctional cytokine TGFbeta, known to play an important role in the regulation of hepatic growth. Here, we reported that ERK5 is phosphorylated and activated by TGFbeta in hepatocytes, with a rapid and sustained kinetic, through a Src-dependent pathway. Moreover, we demonstrated that ERK5 participates to the TGFbeta-induced Snail protein regulation being required for its stabilization. We also found that the functional inactivation of ERK5 impedes the TGFbeta-mediated glycogen synthase kinase-3beta inactivation suggesting this as mechanism responsible for ERK5-mediated Snail stabilization. Thus, results presented in this study uncovered for the first time a role for ERK5 in the TGFbeta-induced cellular responses.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/enzimología , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Animales , Línea Celular , Activación Enzimática/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta , Ratones , Factores de Transcripción de la Familia Snail , Termodinámica , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Familia-src Quinasas/metabolismo
14.
Front Pharmacol ; 10: 942, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31543815

RESUMEN

The cytokine transforming growth factor ß (TGFß) plays a crucial role in the induction of both epithelial-to-mesenchymal transition (EMT) program and fibro-cirrhotic process in the liver, where it contributes also to organ inflammation following several chronic injuries. All these pathological situations greatly increase the risk of hepatocellular carcinoma (HCC) and contribute to tumor progression. In particular, late-stage HCCs are characterized by constitutive activation of TGFß pathway and by an EMT molecular signature leading to the acquisition of invasive and metastatic properties. In these pathological conditions, the cytokine has been shown to induce the transcriptional downregulation of HNF1α, a master regulator of the epithelial/hepatocyte differentiation and of the EMT reverse process, the mesenchymal-to-epithelial transition (MET). Therefore, the restoration of HNF1α expression/activity has been proposed as targeted therapeutic strategy for liver fibro-cirrhosis and late-stage HCCs. In this study, TGFß is found to trigger an early functional inactivation of HNF1α during EMT process that anticipates the effects of the transcriptional downregulation of its own gene. Mechanistically, the cytokine, while not affecting the HNF1α DNA-binding capacity, impaired its ability to recruit CBP/p300 acetyltransferases on target gene promoters and, consequently, its transactivating function. The loss of HNF1α capacity to bind to CBP/p300 and HNF1α functional inactivation have been found to correlate with a change of its posttranslational modification profile. Collectively, the results obtained in this work unveil a new level of HNF1α functional inactivation by TGFß and contribute to shed light on the early events triggering EMT in hepatocytes. Moreover, these data suggest that the use of HNF1α as anti-EMT tool in a TGFß-containing microenvironment may require the design of new therapeutic strategies overcoming the TGFß-induced HNF1α inactivation.

15.
Cell Death Dis ; 10(10): 768, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31601778

RESUMEN

Yes-associated protein (YAP) is a transcriptional co-factor involved in many cell processes, including development, proliferation, stemness, differentiation, and tumorigenesis. It has been described as a sensor of mechanical and biochemical stimuli that enables cells to integrate environmental signals. Although in the liver the correlation between extracellular matrix elasticity (greatly increased in the most of chronic hepatic diseases), differentiation/functional state of parenchymal cells and subcellular localization/activation of YAP has been previously reported, its role as regulator of the hepatocyte differentiation remains to be clarified. The aim of this study was to evaluate the role of YAP in the regulation of epithelial/hepatocyte differentiation and to clarify how a transducer of general stimuli can integrate tissue-specific molecular mechanisms determining specific cell outcomes. By means of YAP silencing and overexpression we demonstrated that YAP has a functional role in the repression of epithelial/hepatocyte differentiation by inversely modulating the expression of Snail (master regulator of the epithelial-to-mesenchymal transition and liver stemness) and HNF4α (master regulator of hepatocyte differentiation) at transcriptional level, through the direct occupancy of their promoters. Furthermore, we found that Snail, in turn, is able to positively control YAP expression influencing protein level and subcellular localization and that HNF4α stably represses YAP transcription in differentiated hepatocytes both in cell culture and in adult liver. Overall, our data indicate YAP as a new member of the HNF4/Snail epistatic molecular circuitry previously demonstrated to control liver cell state. In this model, the dynamic balance between three main transcriptional regulators, that are able to control reciprocally their expression/activity, is responsible for the induction/maintenance of different liver cell differentiation states and its modulation could be the aim of therapeutic protocols for several chronic liver diseases.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular/genética , Células Epiteliales/metabolismo , Factor Nuclear 4 del Hepatocito/metabolismo , Hepatocitos/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Regulación hacia Abajo , Células Epiteliales/citología , Transición Epitelial-Mesenquimal/genética , Silenciador del Gen , Factor Nuclear 4 del Hepatocito/genética , Hepatocitos/citología , Humanos , Ratones , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción/genética , Regulación hacia Arriba , Proteínas Señalizadoras YAP
16.
Eur J Med Chem ; 163: 722-735, 2019 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-30576903

RESUMEN

In aggressive tumors, alkylglyceronephosphate synthase (AGPS) controls cellular ether phospholipid utilization and metabolism to promote cancer cell proliferation and motility. SAR studies on the first-in-class AGPS inhibitor 1, discovered by our group, led to the 2,6-difluoro analog 2i which showed higher binding affinity than 1in vitro. In 231MFP cancer cells, 2i reduced ether lipids levels and cell migration rate. When tested in PC-3 and MDA-MB-231 cancer cells, 2i specifically impaired epithelial to mesenchymal transition (EMT) by modulating E-cadherin, Snail and MMP2 expression levels. Moreover, the combination of siRNAs against AGPS and 2i provided no additive effect, confirming that the modulation of 2i on EMT specifically relies on AGPS inhibition. Finally, this compound also affected cancer cell proliferation especially in MDA-MB-231 cells expressing higher AGPS level, whereas it provided negligible effects on MeT5A, a non-tumorigenic cell line, thus showing cancer specificity.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Transición Epitelial-Mesenquimal/efectos de los fármacos , Metabolismo de los Lípidos/efectos de los fármacos , Neoplasias/patología , Cadherinas/metabolismo , Línea Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Metaloproteinasa 2 de la Matriz/metabolismo , Neoplasias/tratamiento farmacológico , Factores de Transcripción de la Familia Snail/metabolismo , Relación Estructura-Actividad
17.
Artículo en Inglés | MEDLINE | ID: mdl-29971255

RESUMEN

Hepatocellular carcinoma (HCC), is one of the most frequent human cancer and is characterized by a high mortality rate. The aggressiveness appears strictly related to the liver pathological background on which cancer develops. Inflammation and the consequent fibro/cirrhosis, derived from chronic injuries of several origins (viral, toxic and metabolic) and observable in almost all oncological patients, represents the most powerful risk factor for HCC and, at the same time, an important obstacle to the efficacy of systemic therapy. Multiple microenvironmental cues, indeed, play a pivotal role in the pathogenesis, evolution and recurrence of HCC as well as in the resistance to standard therapies observed in most of patients. The identification of altered pathways in cancer cells and of microenvironmental changes, strictly connected in pathogenic feedback loop, may permit to plan new therapeutic approaches targeting tumor cells and their permissive microenvironment, simultaneously.

18.
Stem Cells Int ; 2016: 5481493, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27057172

RESUMEN

In many cell types, several cellular processes, such as differentiation of stem/precursor cells, maintenance of differentiated phenotype, motility, adhesion, growth, and survival, strictly depend on the stiffness of extracellular matrix that, in vivo, characterizes their correspondent organ and tissue. In the liver, the stromal rigidity is essential to obtain the correct organ physiology whereas any alteration causes liver cell dysfunctions. The rigidity of the substrate is an element no longer negligible for the cultivation of several cell types, so that many data so far obtained, where cells have been cultured on plastic, could be revised. Regarding liver cells, standard culture conditions lead to the dedifferentiation of primary hepatocytes, transdifferentiation of stellate cells into myofibroblasts, and loss of fenestration of sinusoidal endothelium. Furthermore, standard cultivation of liver stem/precursor cells impedes an efficient execution of the epithelial/hepatocyte differentiation program, leading to the expansion of a cell population expressing only partially liver functions and products. Overcoming these limitations is mandatory for any approach of liver tissue engineering. Here we propose cell lines as in vitro models of liver stem cells and hepatocytes and an innovative culture method that takes into account the substrate stiffness to obtain, respectively, a rapid and efficient differentiation process and the maintenance of the fully differentiated phenotype.

19.
Stem Cells Int ; 2016: 3543678, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26941801

RESUMEN

Peritoneal dialysis is a form of renal replacement alternative to the hemodialysis. During this treatment, the peritoneal membrane acts as a permeable barrier for exchange of solutes and water. Continual exposure to dialysis solutions, as well as episodes of peritonitis and hemoperitoneum, can cause acute/chronic inflammation and injury to the peritoneal membrane, which undergoes progressive fibrosis, angiogenesis, and vasculopathy, eventually leading to discontinuation of the peritoneal dialysis. Among the different events controlling this pathological process, epithelial to mesenchymal transition of mesothelial cells plays a main role in the induction of fibrosis and in subsequent functional deterioration of the peritoneal membrane. Here, the main extracellular inducers and cellular players are described. Moreover, signaling pathways acting during this process are elucidated, with emphasis on signals delivered by TGF-ß family members and by Toll-like/IL-1ß receptors. The understanding of molecular mechanisms underlying fibrosis of the peritoneal membrane has both a basic and a translational relevance, since it may be useful for setup of therapies aimed at counteracting the deterioration as well as restoring the homeostasis of the peritoneal membrane.

20.
PLoS One ; 11(11): e0167158, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27893804

RESUMEN

In all mammals, the adult liver shows binucleated as well as mononucleated polyploid hepatocytes. The hepatic polyploidization starts after birth with an extensive hepatocyte binucleation and generates hepatocytes of several ploidy classes. While the functional significance of hepatocyte polyploidy is becoming clearer, how it is triggered and maintained needs to be clarified. Aim of this study was to identify a major inducer of hepatocyte binucleation/polyploidization and the cellular and molecular mechanisms involved. We found that, among several cytokines analyzed, known to be involved in early liver development and/or mass control, TGFbeta1 was capable to induce, together with the expected morphological changes, binucleation in hepatocytes in culture. Most importantly, the pharmacological inhibition of TGFbeta signaling in healthy mice during weaning, when the physiological binucleation occurs, induced a significant decrease of hepatocyte binucleation rate, without affecting cell proliferation and hepatic index. The TGFbeta-induced hepatocyte binucleation resulted from a cytokinesis failure, as assessed by video microscopy, and is associated with a delocalization of the cytokinesis regulator RhoA-GTPase from the mid-body of dividing cells. The use of specific chemical inhibitors demonstrated that the observed events are Src-dependent. Finally, the restoration of a fully epithelial phenotype by TGFbeta withdrawal gave rise to a cell progeny capable to maintain the polyploid state. In conclusion, we identified TGFbeta as a major inducer of hepatocyte binucleation both in vitro and in vivo, thus ascribing a novel role to this pleiotropic cytokine. The production of binucleated/tetraploid hepatocytes is due to a cytokinesis failure controlled by the molecular axis TGFbeta/Src/RhoA.


Asunto(s)
Núcleo Celular/genética , Citocinesis/fisiología , Hepatocitos/citología , Hígado/citología , Poliploidía , Factor de Crecimiento Transformador beta/farmacología , Familia-src Quinasas/metabolismo , Animales , Núcleo Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Citocinesis/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Familia-src Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA