Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int Immunol ; 35(7): 303-312, 2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-36719100

RESUMEN

Dupuytren's contracture (DC) is an inflammatory fibrosis characterized by fibroproliferative disorders of the palmar aponeurosis, for which there is no effective treatment. Although several genome-wide association studies have identified risk alleles associated with DC, the functional linkage between these alleles and the pathogenesis remains elusive. We here focused on two single nucleotide polymorphisms (SNPs) associated with DC, rs16879765 and rs17171229, in secreted frizzled related protein 4 (SFRP4). We investigated the association of SRFP4 with the IL-6 amplifier, which amplifies the production of IL-6, growth factors and chemokines in non-immune cells and aggravates inflammatory diseases via NF-κB enhancement. Knockdown of SFRP4 suppressed activation of the IL-6 amplifier in vitro and in vivo, whereas the overexpression of SFRP4 induced the activation of NF-κB-mediated transcription activity. Mechanistically, SFRP4 induced NF-κB activation by directly binding to molecules of the ubiquitination SFC complex, such as IkBα and ßTrCP, followed by IkBα degradation. Furthermore, SFRP4 expression was significantly increased in fibroblasts derived from DC patients bearing the risk alleles. Consistently, fibroblasts with the risk alleles enhanced activation of the IL-6 amplifier. These findings indicate that the IL-6 amplifier is involved in the pathogenesis of DC, particularly in patients harboring the SFRP4 risk alleles. Therefore, SFRP4 is a potential therapeutic target for various inflammatory diseases and disorders, including DC.


Asunto(s)
Contractura de Dupuytren , Humanos , Contractura de Dupuytren/genética , Contractura de Dupuytren/patología , Polimorfismo de Nucleótido Simple , Estudio de Asociación del Genoma Completo , FN-kappa B/metabolismo , Interleucina-6/metabolismo , Fibroblastos/metabolismo , Inflamación/genética , Inflamación/metabolismo , Proteínas Proto-Oncogénicas/metabolismo
2.
Exp Dermatol ; 30(1): 62-67, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32970880

RESUMEN

Type XVII collagen (COL17) is a transmembrane protein expressed in the basal epidermis. COL17 serves as a niche for epidermal stem cells, and although its reduction has been implicated in altering cell polarity and ageing of the epidermis, it is unknown how COL17 affects epidermal cell polarity. Here, we uncovered COL17 as a binding partner of the aPKC-PAR complex, which is a key regulating factor of cell polarity. Immunoprecipitation-immunoblot assay and protein-protein binding assay revealed that COL17 interacts with aPKC and PAR3. COL17 deficiency or epidermis-specific aPKCλ deletion destabilized PAR3 distribution in the epidermis, while aPKCζ knockout did not. Asymmetrical cell division was pronounced in COL17-null neonatal paw epidermis. These results show that COL17 is pivotal for maintaining epidermal cell polarity. Our study highlights the previously unrecognized role of COL17 in the basal keratinocytes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Polaridad Celular , Epidermis/metabolismo , Colágenos no Fibrilares/metabolismo , Proteína Quinasa C/metabolismo , Animales , Autoantígenos/genética , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Colágenos no Fibrilares/genética , Isoformas de Proteínas/metabolismo , Colágeno Tipo XVII
3.
Proc Natl Acad Sci U S A ; 116(35): 17450-17459, 2019 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-31399545

RESUMEN

Although KRAS and TP53 mutations are major drivers of pancreatic ductal adenocarcinoma (PDAC), the incurable nature of this cancer still remains largely elusive. ARF6 and its effector AMAP1 are often overexpressed in different cancers and regulate the intracellular dynamics of integrins and E-cadherin, thus promoting tumor invasion and metastasis when ARF6 is activated. Here we show that the ARF6-AMAP1 pathway is a major target by which KRAS and TP53 cooperatively promote malignancy. KRAS was identified to promote eIF4A-dependent ARF6 mRNA translation, which contains a quadruplex structure at its 5'-untranslated region, by inducing TEAD3 and ETV4 to suppress PDCD4; and also eIF4E-dependent AMAP1 mRNA translation, which contains a 5'-terminal oligopyrimidine-like sequence, via up-regulating mTORC1. TP53 facilitated ARF6 activation by platelet-derived growth factor (PDGF), via its known function to promote the expression of PDGF receptor ß (PDGFRß) and enzymes of the mevalonate pathway (MVP). The ARF6-AMAP1 pathway was moreover essential for PDGF-driven recycling of PD-L1, in which KRAS, TP53, eIF4A/4E-dependent translation, mTOR, and MVP were all integral. We moreover demonstrated that the mouse PDAC model KPC cells, bearing KRAS/TP53 mutations, express ARF6 and AMAP1 at high levels and that the ARF6-based pathway is closely associated with immune evasion of KPC cells. Expression of ARF6 pathway components statistically correlated with poor patient outcomes. Thus, the cooperation among eIF4A/4E-dependent mRNA translation and MVP has emerged as a link by which pancreatic driver mutations may promote tumor cell motility, PD-L1 dynamics, and immune evasion, via empowering the ARF6-based pathway and its activation by external ligands.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Antígeno B7-H1/metabolismo , Evasión Inmune/genética , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína p53 Supresora de Tumor/genética , Factor 6 de Ribosilación del ADP , Sitios de Unión , Biomarcadores de Tumor , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Modelos Moleculares , Mutación , Neoplasias Pancreáticas/mortalidad , Neoplasias Pancreáticas/patología , Pronóstico , Unión Proteica , ARN Mensajero/genética , Receptores del Factor de Crecimiento Derivado de Plaquetas/metabolismo , Transducción de Señal
4.
Cell Commun Signal ; 16(1): 94, 2018 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-30509302

RESUMEN

BACKGROUND: TP53 mutations in cancer cells often evoke cell invasiveness, whereas fibroblasts show invasiveness in the presence of intact TP53. AMAP1 (also called DDEF1 or ASAP1) is a downstream effector of ARF6 and is essential for the ARF6-driven cell-invasive phenotype. We found that AMAP1 levels are under the control of p53 (TP53 gene product) in epithelial cells but not in fibroblasts, and here addressed that molecular basis of the epithelial-specific function of p53 in suppressing invasiveness via targeting AMAP1. METHODS: Using MDA-MB-231 cells expressing wild-type and p53 mutants, we identified miRNAs in which their expression is controlled by normal-p53. Among them, we identified miRNAs that target AMAP1 mRNA, and analyzed their expression levels and epigenetic statuses in epithelial cells and nonepithelial cells. RESULTS: We found that normal-p53 suppresses AMAP1 mRNA in cancer cells and normal epithelial cells, and that more than 30 miRNAs are induced by normal-p53. Among them, miR-96 and miR-182 were found to target the 3'-untranslated region of AMAP1 mRNA. Fibroblasts did not express these miRNAs at detectable levels. The ENCODE dataset demonstrated that the promoter region of the miR-183-96-182 cistron is enriched with H3K27 acetylation in epithelial cells, whereas this locus is enriched with H3K27 trimethylation in fibroblasts and other non-epithelial cells. miRNAs, such as miR-423, which are under the control of p53 but not associated with AMAP1 mRNA, demonstrated similar histone modifications at their gene loci in epithelial cells and fibroblasts, and were expressed in these cells. CONCLUSION: Histone modifications of certain miRNA loci, such as the miR-183-96-182 cistron, are different between epithelial cells and non-epithelial cells. Such epithelial-specific miRNA regulation appears to provide the molecular basis for the epithelial-specific function of p53 in suppressing ARF6-driven invasiveness.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Células Epiteliales/metabolismo , Sitios Genéticos/genética , Código de Histonas/genética , MicroARNs/genética , Proteína p53 Supresora de Tumor/genética , Secuencia de Bases , Línea Celular Tumoral , Células Epiteliales/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación , Invasividad Neoplásica , ARN Mensajero/genética
5.
Cancers (Basel) ; 10(6)2018 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-29799511

RESUMEN

In addition to its classical roles as a tumor suppressor, p53 has also been shown to act as a guardian of epithelial integrity by inducing the microRNAs that target transcriptional factors driving epithelial⁻mesenchymal transition. On the other hand, the ENCODE project demonstrated an enrichment of putative motifs for the binding of p53 in epithelial-specific enhancers, such as CDH1 (encoding E-cadherin) enhancers although its biological significance remained unknown. Recently, we identified two novel modes of epithelial integrity (i.e., maintenance of CDH1 expression): one involves the binding of p53 to a CDH1 enhancer region and the other does not. In the former, the binding of p53 is necessary to maintain permissive histone modifications around the CDH1 transcription start site, whereas in the latter, p53 does not bind to this region nor affect histone modifications. Furthermore, these mechanisms likely coexisted within the same tissue. Thus, the mechanisms involved in epithelial integrity appear to be much more complex than previously thought. In this review, we describe our findings, which may instigate further experimental scrutiny towards understanding the whole picture of epithelial integrity as well as the related complex asymmetrical functions of p53. Such understanding will be important not only for cancer biology but also for the safety of regenerative medicine.

6.
Cell Commun Signal ; 16(1): 1, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29329590

RESUMEN

BACKGROUND: The small GTPase Arf6 and its downstream effector AMAP1 (also called ASAP1/DDEF1) constitute a signaling pathway promoting cell invasion, in which AMAP1 interacts with several different proteins, including PRKD2, EPB41L5, paxillin, and cortactin. Components of this pathway are often overexpressed in human breast cancer cells, to be correlated with poor prognosis of the patients, whereas overexpression of the Arf6 pathway did not correlate with the four main molecular classes of human breast tumors. In this pathway, receptor tyrosine kinases, including EGFR and Her2, activate Arf6 via GEP100. MMTV-PyMT mice and MMTV-Neu mice are well-established models of human breast cancer, and exhibit the early dissemination and the lung metastasis, by utilizing protein tyrosine phosphorylation for oncogenesis. PyMT-tumors and Neu-tumors are known to have overlapping gene expression profiles, which primarily correspond to the luminal B-type of human mammary tumors, although they differ in the time necessary for tumor onset and metastasis. Given the common usage of protein tyrosine phosphorylation, as well as the frequent use of these animal models for studying breast cancer at the molecular level, we here investigated whether mammary tumors in these mouse models utilize the Arf6-based pathway for invasion. METHODS: Expression levels of Arf6, AMAP1, and GEP100 were analyzed in PyMT-tumors and Neu-tumors by western blotting. Expression of Arf6 and AMAP1 was also analyzed by immunohistochemistry. The involvement of AMAP1 in invasion, and the possible correlation of its high expression levels with cancer mesenchymal properties were also investigated. RESULTS: We found that PyMT-tumors, but not Neu-tumors, frequently overexpress AMAP1 and use it for invasion, whereas both types of tumors expressed Arf6 and GEP100 at different levels. High levels of the AMAP1 expression among PyMT-tumor cells were frequently correlated with loss of the epithelial marker CK8 and also with expression of the mesenchymal marker vimentin both at the primary sites and at sites of the lung metastases. CONCLUSIONS: PyMT-tumors appear to frequently utilize the Arf6-based invasive machinery, whereas Neu-tumors do not. Our results suggest that MMTV-PyMT mice, rather than MMTV-Neu mice, are useful to study the Arf6-based mammary tumor malignancies, as a representative model of human breast cancer.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antígenos Transformadores de Poliomavirus/genética , Neoplasias de la Mama/patología , Virus del Tumor Mamario del Ratón/genética , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/antagonistas & inhibidores , Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Antígenos Transformadores de Poliomavirus/metabolismo , Neoplasias de la Mama/metabolismo , Modelos Animales de Enfermedad , Femenino , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/secundario , Ratones , Invasividad Neoplásica , Regiones Promotoras Genéticas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
7.
Sci Rep ; 8(1): 1595, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29371630

RESUMEN

TP53 mutation (i.e., loss of normal-p53) may evoke epithelial-mesenchymal transition (EMT), which was previously attributed to loss of certain miRNAs. However, not all epithelial cells undergo EMT upon TP53 mutation, and the p53-miRNA axis may not fully explain p53 function in epithelial integrity. We here show two modes of epithelial integrity: one involves p53-binding to a nucleotide region and the other does not. In the former, p53 binds to the CDH1 (encoding E-cadherin) locus to antagonize EZH2-mediated H3K27 trimethylation (H3K27me3) to maintain high levels of acetylation of H3K27 (H3K27ac). In the latter, the same locus is not highly acetylated at H3K27, and does not allow p53-binding, nor needs to antagonize EZH2. We moreover demonstrated that although the CDH1 locus in the p53-independent cells, but not in fibroblasts, becomes high-H3K27ac by butyrate and allows p53-biniding, their CDH1 expression does not become dependent on p53. Our results identified novel modes of the epithelial integrity, in which the same epithelial-specific gene locus exhibits different requirement for p53 with different histone modifications among different epithelial cells to warrant its expression.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Epigénesis Genética , Células Epiteliales/clasificación , Células Epiteliales/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular , Humanos
8.
Cell Commun Signal ; 15(1): 36, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28969640

RESUMEN

BACKGROUND: The small GTPase ARF1 mediates membrane trafficking mostly from the Golgi, and is essential for the G protein-coupled receptor (GPCR)-mediated chemotaxis of neutrophils. In this process, ARF1 is activated by the guanine nucleotide exchanger GBF1, and is inactivated by the GTPase-activating protein GIT2. Neutrophils generate the Gßγ-PAK1-αPIX-GIT2 linear complex during GPCR-induced chemotaxis, in which αPIX activates RAC1/CDC42, which then employs PAK1. However, it has remained unclear as to why GIT2 is included in this complex. RESULTS: We investigated the association between ARF1 and RAC1/CDC42 during the fMLP-stimulated chemotaxis of HL60 cells. We found that the silencing of GBF1 significantly impaired the recruitment of RAC1 to the leading edges, but not PAK1, αPIX, RAC2, or CDC42. A significant population of RAC1 colocalized with ARF1 at the leading edges in stimulated cells, whereas fMLP activated both ARF1 and ARF5. Consistently, the silencing of ARF1, but not ARF5, impaired the recruitment of RAC1, whereas the silencing of RAC1 did not affect the recruitment of ARF1 to the leading edges. CONCLUSIONS: Our results indicated that the activation of ARF1 triggers the plasma membrane recruitment of RAC1 in GPCR-mediated chemotaxis, which is essential for cortical actin remodeling. Thus, membrane remodeling at the leading edges appears to precede actin remodeling in chemotaxis. Together with the fact that GIT2, which inactivates ARF1, is an integral component of the machinery activating RAC1, we proposed a model in which the ARF1-RAC1 linkage enables the regulation of ARF1 by repetitive on/off cycles during GPCR-mediated neutrophil chemotaxis.


Asunto(s)
Factor 1 de Ribosilacion-ADP/metabolismo , Membrana Celular/metabolismo , Quimiotaxis , Neutrófilos/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Factor 1 de Ribosilacion-ADP/genética , Actinas/metabolismo , Línea Celular Tumoral , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Neutrófilos/fisiología , Proteína de Unión al GTP cdc42/metabolismo , Quinasas p21 Activadas/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína RCA2 de Unión a GTP
9.
Cell Commun Signal ; 14(1): 28, 2016 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-27871329

RESUMEN

BACKGROUND: Squamous cell carcinoma of the tongue (tongue SCC) is a major subtype of head and neck squamous cell carcinoma (HNSCC), which is an intractable cancer under current therapeutics. ARF6 and its effector AMAP1 are often overexpressed in different types of cancers, such as breast cancer and renal cancer, and in these cancers, AMAP1 binds to EPB41L5 to promote invasion, metastasis, and drug resistance. EPB41L5 is a mesenchymal-specific protein, normally induced during epithelial-mesenchymal transition (EMT) to promote focal adhesion dynamics. Similarly to breast cancer and renal cancer, the acquisition of mesenchymal phenotypes is the key process that drives the malignancy of HNSCC. We previously showed that the overexpression of AMAP1 in tongue SCC is statistically correlated with the poor outcome of patients. In this study, we examined whether tongue SCC also expresses EPB41L5 at high levels. RESULTS: Immunohistochemical staining of clinical specimens of tongue SCC demonstrated that high expression levels of EPB41L5 statistically correlate with poor disease-free survival and poor overall survival rates of patients. The tongue SCC cell line SCC-9, which overexpress Arf6 and AMAP1, also expressed EPB41L5 at high levels to promote invasiveness, whereas the weakly invasive SCC-25 cells did not express EPB41L5 at notable levels. Among the different EMT-associated transcriptional factors, ZEB1 was previously found to be most crucial in inducing EPB41L5 in breast cancer and renal cancer. In contrast, expression levels of ZEB1 did not correlate with the expression levels of EPB41L5 in tongue SCC, whereas KLF8 and FOXO3 levels showed positive correlations with EPB41L5 levels. Moreover, silencing of EPB41L5 only marginally improved the drug resistance of SCC-9 cells, even when coupled with ionizing radiation. CONCLUSION: Our results indicate that activation of the cancer mesenchymal program in tongue SCC, which leads to EPB41L5 expression, closely correlates with the poor prognosis of patients. However, ZEB1 was not the major inducer of EPB41L5 in tongue SCC, unlike in breast cancer and renal cancer. Thus, processes that trigger the mesenchymal program of tongue SCC, which drives their malignancies, seem to be substantially different from those of other cancers.


Asunto(s)
Factores de Ribosilacion-ADP/genética , Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de la Membrana/genética , Neoplasias de la Lengua/genética , Lengua/patología , Factor 6 de Ribosilación del ADP , Factores de Ribosilacion-ADP/análisis , Carcinoma de Células Escamosas/diagnóstico , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Humanos , Proteínas de la Membrana/análisis , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Pronóstico , Lengua/metabolismo , Neoplasias de la Lengua/diagnóstico , Neoplasias de la Lengua/patología , Regulación hacia Arriba
10.
Mol Cell Oncol ; 3(4): e1185564, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27652329

RESUMEN

The mevalonate pathway results in the prenylation of small GTPases, which are pivotal for oncogenesis and cancer malignancies. However, inhibitors of this pathway, such as statins, have not necessarily produced favorable results in clinical trials. We recently identified properties of statin responders, together with the underlying molecular mechanisms and simple biomarkers to predict these responders.

11.
J Cell Biol ; 213(1): 81-95, 2016 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-27044891

RESUMEN

Drug resistance, metastasis, and a mesenchymal transcriptional program are central features of aggressive breast tumors. The GTPase Arf6, often overexpressed in tumors, is critical to promote epithelial-mesenchymal transition and invasiveness. The metabolic mevalonate pathway (MVP) is associated with tumor invasiveness and known to prenylate proteins, but which prenylated proteins are critical for MVP-driven cancers is unknown. We show here that MVP requires the Arf6-dependent mesenchymal program. The MVP enzyme geranylgeranyl transferase II (GGT-II) and its substrate Rab11b are critical for Arf6 trafficking to the plasma membrane, where it is activated by receptor tyrosine kinases. Consistently, mutant p53, which is known to support tumorigenesis via MVP, promotes Arf6 activation via GGT-II and Rab11b. Inhibition of MVP and GGT-II blocked invasion and metastasis and reduced cancer cell resistance against chemotherapy agents, but only in cells overexpressing Arf6 and components of the mesenchymal program. Overexpression of Arf6 and mesenchymal proteins as well as enhanced MVP activity correlated with poor patient survival. These results provide insights into the molecular basis of MVP-driven malignancy.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Resistencia a Antineoplásicos/fisiología , Ácido Mevalónico/metabolismo , Metástasis de la Neoplasia/patología , Proteína p53 Supresora de Tumor/metabolismo , Factor 6 de Ribosilación del ADP , Antineoplásicos/farmacología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/fisiología , Transición Epitelial-Mesenquimal/fisiología , Células HEK293 , Humanos , Células MCF-7 , Invasividad Neoplásica/patología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/fisiología , gamma-Glutamiltransferasa/metabolismo , Proteínas de Unión al GTP rab/metabolismo
12.
Nat Commun ; 7: 10656, 2016 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-26854204

RESUMEN

Acquisition of mesenchymal properties by cancer cells is critical for their malignant behaviour, but regulators of the mesenchymal molecular machinery and how it is activated remain elusive. Here we show that clear cell renal cell carcinomas (ccRCCs) frequently utilize the Arf6-based mesenchymal pathway to promote invasion and metastasis, similar to breast cancers. In breast cancer cells, ligand-activated receptor tyrosine kinases employ GEP100 to activate Arf6, which then recruits AMAP1; and AMAP1 then binds to the mesenchymal-specific protein EPB41L5, which promotes epithelial-mesenchymal transition and focal adhesion dynamics. In renal cancer cells, lysophosphatidic acid (LPA) activates Arf6 via its G-protein-coupled receptors, in which GTP-Gα12 binds to EFA6. The Arf6-based pathway may also contribute to drug resistance. Our results identify a specific mesenchymal molecular machinery of primary ccRCCs, which is triggered by a product of autotaxin and it is associated with poor outcome of patients.


Asunto(s)
Factores de Ribosilacion-ADP/metabolismo , Carcinoma de Células Renales/metabolismo , Transición Epitelial-Mesenquimal , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Neoplasias Renales/metabolismo , Lisofosfolípidos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores del Ácido Lisofosfatídico/metabolismo , Factor 6 de Ribosilación del ADP , Adulto , Anciano , Anciano de 80 o más Años , Amidas/farmacología , Animales , Antineoplásicos/farmacología , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Inhibidores Enzimáticos/farmacología , Femenino , Factores de Intercambio de Guanina Nucleótido , Células HEK293 , Humanos , Inmunohistoquímica , Indoles/farmacología , Isoxazoles/farmacología , Neoplasias Renales/patología , Masculino , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Metástasis de la Neoplasia , Trasplante de Neoplasias , Propionatos/farmacología , Piridinas/farmacología , Pirroles/farmacología , Transducción de Señal , Sirolimus/análogos & derivados , Sirolimus/farmacología , Sunitinib , Triazoles/farmacología
13.
PLoS One ; 10(1): e0116135, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25617756

RESUMEN

Despite the recent advances in the treatment of multiple myeloma (MM), MM patients with high-risk cytogenetic changes such as t(4;14) translocation or deletion of chromosome 17 still have extremely poor prognoses. With the goal of helping these high-risk MM patients, we previously developed a novel phthalimide derivative, TC11. Here we report the further characterization of TC11 including anti-myeloma effects in vitro and in vivo, a pharmacokinetic study in mice, and anti-osteoclastogenic activity. Intraperitoneal injections of TC11 significantly delayed the growth of subcutaneous tumors in human myeloma-bearing SCID mice. Immunohistochemical analyses showed that TC11 induced apoptosis of MM cells in vivo. In the pharmacokinetic analyses, the Cmax was 2.1 µM at 1 h after the injection of TC11, with 1.2 h as the half-life. TC11 significantly inhibited the differentiation and function of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated osteoclasts in mouse osteoclast cultures using M-CSF and RANKL. We also revealed that TC11 induced the apoptosis of myeloma cells accompanied by α-tubulin fragmentation. In addition, TC11 and lenalidomide, another phthalimide derivative, directly bound to nucleophosmin 1 (NPM1), whose role in MM is unknown. Thus, through multiple molecular interactions, TC11 is a potentially effective drug for high-risk MM patients with bone lesions. The present results suggest the possibility of the further development of novel thalidomide derivatives by drug designing.


Asunto(s)
Mieloma Múltiple/patología , Ftalimidas/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Semivida , Xenoinjertos/efectos de los fármacos , Humanos , Lenalidomida , Masculino , Ratones Endogámicos ICR , Ratones SCID , Mieloma Múltiple/genética , Proteínas Nucleares/química , Nucleofosmina , Osteoclastos/efectos de los fármacos , Ftalimidas/química , Ftalimidas/farmacocinética , Talidomida/análogos & derivados , Talidomida/química , Talidomida/farmacología
14.
Cancer Res ; 73(16): 5100-9, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23811942

RESUMEN

NFATc1 is a transcription factor that regulates T-cell development, osteoclastogenesis, and macrophage function. Given that T cells, osteoclasts, and macrophages in the tumor microenvironment are thought to modulate tumor progression, tumor cells may acquire NFATc1 expression through fusion with these NFATc1-expressing normal cells. We here revealed that a small proportion of tumor cells in human carcinoma specimens expressed NFATc1. To investigate the consequences of NFATc1 acquisition by tumor cells, we established A549 and MCF7 cell lines expressing a constitutively active form of NFATc1 (NFATc1CA) in an inducible manner. The expression of NFATc1CA promoted cancer cell invasion in association with changes in cell morphology. Analysis of gene expression and RNA interference experiments revealed that NFATc1CA suppressed E-cadherin expression by upregulating the transcriptional repressors Snail and Zeb1 in a manner independent of TGF-ß signaling. Induced expression of NFATc1CA also downregulated E-cadherin expression and increased invasive activity in tumor xenografts in vivo. Our results thus suggest that the acquisition of NFATc1 expression contributes to tumor progression.


Asunto(s)
Cadherinas/metabolismo , Factores de Transcripción NFATC/biosíntesis , Factores de Transcripción NFATC/genética , Neoplasias/metabolismo , Neoplasias/patología , Animales , Antígenos CD , Cadherinas/genética , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Factores de Transcripción NFATC/metabolismo , Invasividad Neoplásica , Neoplasias/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc
15.
PLoS One ; 8(3): e60528, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23555988

RESUMEN

Podosomes are cellular "feet," characterized by F-actin-rich membrane protrusions, which drive cell migration and invasion into the extracellular matrix. Small GTPases that regulate the actin cytoskeleton, such as Cdc42 and Rac are central regulators of podosome formation. The adaptor protein IRSp53 contains an I-BAR domain that deforms membranes into protrusions and binds to Rac, a CRIB motif that interacts with Cdc42, an SH3 domain that binds to many actin cytoskeletal regulators with proline-rich peptides including VASP, and the C-terminal variable region by splicing. However, the role of IRSp53 and VASP in podosome formation had been unclear. Here we found that the knockdown of IRSp53 by RNAi attenuates podosome formation and migration in Src-transformed NIH3T3 (NIH-Src) cells. Importantly, the differences in the IRSp53 C-terminal splicing isoforms did not affect podosome formation. Overexpression of IRSp53 deletion mutants suggested the importance of linking small GTPases to SH3 binding partners. Interestingly, VASP physically interacted with IRSp53 in NIH-Src cells and was essential for podosome formation. These data highlight the role of IRSp53 as a linker of small GTPases to VASP for podosome formation.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Movimiento Celular , Proteínas de Microfilamentos/metabolismo , Células 3T3 NIH/citología , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas/metabolismo , Seudópodos/metabolismo , Actinas/metabolismo , Animales , Ratones , Proteínas de Unión al GTP Monoméricas/metabolismo , Células 3T3 NIH/metabolismo , Proteínas del Tejido Nervioso/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Regulación hacia Arriba , Dominios Homologos src , Familia-src Quinasas/metabolismo
16.
Cell Mol Life Sci ; 70(18): 3341-53, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23296124

RESUMEN

Osteoclasts are bone-resorbing cells of monocytic origin. An imbalance between bone formation and resorption can lead to osteoporosis or osteopetrosis. Osteoclastogenesis is triggered by RANKL- and IP3-induced Ca(2+) influx followed by activation of NFATc1, a master transcription factor for osteoclastogenic gene regulation. During differentiation, osteoclasts undergo cytoskeletal remodeling to migrate and attach to the bone surface. Simultaneously, they fuse with each other to form multinucleated cells. These processes require PI3-kinase-dependent cytoskeletal protein activation to initiate cytoskeletal remodeling, resulting in the formation of circumferential podosomes and fusion-competent protrusions. In multinucleated osteoclasts, circumferential podosomes mature into stabilized actin rings, which enables the formation of a ruffled border where intensive membrane trafficking is executed. Membrane lipids, especially phosphoinositides, are key signaling molecules that regulate osteoclast morphology and act as second messengers and docking sites for multiple important effectors. We examine the critical roles of phosphoinositides in the signaling cascades that regulate osteoclast functions.


Asunto(s)
Membrana Celular/metabolismo , Regulación de la Expresión Génica , Lípidos de la Membrana/metabolismo , Osteoclastos/citología , Actinas/metabolismo , Animales , Calcio/metabolismo , Movimiento Celular , Citoesqueleto/metabolismo , Humanos , Ratones , Ratones Transgénicos , Osteopetrosis/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositoles/metabolismo , Fosforilación , Ligando RANK/metabolismo , Transducción de Señal , Transcripción Genética , Regulación hacia Arriba , Familia-src Quinasas/metabolismo
17.
J Cell Biol ; 197(4): 553-68, 2012 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-22584907

RESUMEN

Osteoclasts fuse to form multinucleated cells during osteoclastogenesis. This process is mediated by dynamic rearrangement of the plasma membrane and cytoskeleton, and it requires numerous factors, many of which have been identified. The underlying mechanism remains obscure, however. In this paper, we show that Tks5, a master regulator of invadopodia in cancer cells, is crucial for osteoclast fusion downstream of phosphoinositide 3-kinase and Src. Expression of Tks5 was induced during osteoclastogenesis, and prevention of this induction impaired both the formation of circumferential podosomes and osteoclast fusion without affecting cell differentiation. Tyrosine phosphorylation of Tks5 was attenuated in Src-/- osteoclasts, likely accounting for defects in podosome organization and multinucleation in these cells. Circumferential invadopodia formation in B16F0 melanoma cells was also accompanied by Tks5 phosphorylation. Co-culture of B16F0 cells with osteoclasts in an inflammatory milieu promoted the formation of melanoma-osteoclast hybrid cells. Our results thus reveal an unexpected link between circumferential podosome/invadopodium formation and cell-cell fusion in and beyond osteoclasts.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Diferenciación Celular , Fosfoproteínas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Animales , Fusión Celular , Línea Celular Tumoral , Humanos , Ratones , Osteoclastos/metabolismo , Proteínas de Unión a Fosfato , Fosfoproteínas/genética
18.
Commun Integr Biol ; 5(5): 511-5, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23739834

RESUMEN

Podosomes and invadopodia seen in osteoclasts and cancer cells, respectively, are actin-rich membrane protrusions. We recently demonstrated that an adaptor protein, Tks5, which is an established regulator of invadopodia in cancer cells, drives osteoclast-osteoclast fusion as well as osteoclast-cancer cell fusion by generating circumferential podosomes/invadopodia. This finding revealed an unexpected potential of podosomes/invadopodia to act as fusion-competent protrusions. Fusion of biological membranes involves the intricate orchestration of various proteins and lipids. Recent literature suggests the importance of membrane curvature formation in lipid bilayer fusion. In this study, we investigated the expression of Bin-Amphiphysin-Rvs161/167 (BAR) domain superfamily proteins, which have membrane deforming activity, during osteoclastogenesis. We found that IRTKS was specifically induced during osteoclast fusion and interacted with Tks5, suggesting the role of IRTKS in the formation of fusion-competent protrusions via its BAR domain.

19.
Oncotarget ; 1(5): 320-8, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21307399

RESUMEN

Invadopodia are extracellular matrix (ECM)-degrading protrusions formed by invasive cancer cells. Podosomes are structures functionally similar to invadopodia that are found in oncogene-transformed fibroblasts and monocyte-derived cells, including macrophages and osteoclasts. These structures are thought to play important roles in the pericellular remodeling of ECM during cancer invasion and metastasis. Much effort has been directed toward identification of the molecular components and regulators of invadopodia/podosomes, which could be therapeutic targets in the treatment of malignant cancers. However, it remains largely unknown how these components are assembled into invadopodia/podosomes and how the assembly process is spatially and temporally regulated. This review will summarize recent progress on the molecular mechanisms of invadopodia/podosome formation, with strong emphasis on the roles of lipid rafts and phosphoinositides.


Asunto(s)
Extensiones de la Superficie Celular/metabolismo , Lípidos de la Membrana/metabolismo , Microdominios de Membrana/metabolismo , Neoplasias/metabolismo , Animales , Caveolina 1/metabolismo , Extensiones de la Superficie Celular/patología , Matriz Extracelular/metabolismo , Humanos , Microdominios de Membrana/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/patología , Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositoles/metabolismo , Transducción de Señal
20.
Cell Adh Migr ; 3(2): 195-7, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19262173

RESUMEN

Cell-to-extracellular matrix (ECM) adhesion plays important roles in various biological events, such as proliferation, differentiation and migration. Distinct from other types of adhesion structures (focal complexes, focal adhesions and so on), podosomes and invadopodia are thought to have additional functions beyond attachment, possibly including invasion into the ECM. for podosomes and invadopodia to invade into the ECM, molecules involved in adhesion, actin polymerization and ECM degradation must be recruited to sites of action. Our recent study demonstrated that podosomes form near newly formed focal adhesions via the minimally expressed phosphoinositide PtdIns(3,4) P2-mediated recruitment of the Tks5-Grb2 scaffold, followed by the accumulation of N-WASP. Although this study demonstrated details of molecular interplay during the transformation of focal adhesion, its regulation in the in vivo invasion process remains to be clarified. Here, we discuss the molecular bases of the transformation of focal adhesions to podosomes/invadopodia based on current understanding.


Asunto(s)
Adhesiones Focales/fisiología , Fosfatos de Fosfatidilinositol/fisiología , Animales , Células Cultivadas , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...