Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Cancer ; 20(1): 78, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34039363

RESUMEN

BACKGROUND: Distant metastasis is the major cause of death in patients with colorectal cancer (CRC). Previously, we identified KITENIN as a metastasis-enhancing gene and suggested that the oncogenic KITENIN complex is involved in metastatic dissemination of KITENIN-overexpressing CRC cells. Here, we attempted to find substances targeting the KITENIN complex and test their ability to suppress distant metastasis of CRC. METHODS: We screened a small-molecule compound library to find candidate substances suppressing the KITENIN complex in CRC cells. We selected a candidate compound and examined its effects on the KITENIN complex and distant metastasis through in vitro assays, a molecular docking model, and in vivo tumor models. RESULTS: Among several compounds, we identified DKC1125 (Disintegrator of KITENIN Complex #1125) as the best candidate. DKC1125 specifically suppressed KITENIN gain of function. After binding KH-type splicing regulatory protein (KSRP), DKC1125 degraded KITENIN and Dvl2 by recruiting RACK1 and miRNA-124, leading to the disintegration of the functional KITENIN-KSRP-RACK1-Dvl2 complex. A computer docking model suggested that DKC1125 specifically interacted with the binding pocket of the fourth KH-domain of KSRP. KITENIN-overexpressing CRC cells deregulated certain microRNAs and were resistant to 5-fluorouracil, oxaliplatin, and cetuximab. DKC1125 restored sensitivity to these drugs by normalizing expression of the deregulated microRNAs, including miRNA-124. DKC1125 effectively suppressed colorectal liver metastasis in a mouse model. Interestingly, the combination of DKC1125 with 5-fluorouracil suppressed metastasis more effectively than either drug alone. CONCLUSION: DKC1125 targets the KITENIN complex and could therefore be used as a novel therapeutic to suppress liver metastasis in CRC expressing high levels of KITENIN.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Portadoras/efectos de los fármacos , Neoplasias Colorrectales/patología , Proteínas de la Membrana/efectos de los fármacos , Proteínas de Unión al ARN/efectos de los fármacos , Proteínas de Unión al ARN/metabolismo , Transactivadores/efectos de los fármacos , Transactivadores/metabolismo , Animales , Antineoplásicos/química , Descubrimiento de Drogas , Humanos , Ratones , Simulación del Acoplamiento Molecular , Metástasis de la Neoplasia/patología , Proteínas de Unión al ARN/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores
2.
Mol Carcinog ; 56(3): 1068-1081, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27648936

RESUMEN

Understanding the complex biological functions of E3-ubiquitin ligases may facilitate the development of mechanism-based anti-cancer drugs. We recently identified that the KITENIN/ErbB4-Dvl2-c-Jun axis works as a novel unconventional downstream signal of epidermal growth factor (EGF) in colorectal cancer (CRC) tissues. Here we addressed whether E3-ubiquitin ligases are required for operation of this axis. We found that Nrdp1, an E3-ligase for ErbB3/ErbB4, interacted with KITENIN (KAI1 C-terminal interacting tetraspanin) to form a functional KITENIN/ErbB4/Nrdp1 complex and is responsible for down-regulating Dvl2 within this complex. Interestingly, ErbB4 was resistant to degradation by Nrdp1 in KITENIN/Nrdp1-co-transfected CRC cells, and KITENIN bound to the C-terminal coiled-coil domain of Nrdp1. Chemical blockade of ErbB kinase did not block the action of EGF to increase in total/phospho-ErbB4 and phospho-ERK in KITENIN/ErbB4-cotransfected cells, whereas it blocked the action of EGF in ErbB4 alone-transfected CRC cells. In human CRC tissues, higher expressions of ErbB4 and KITENIN and lower expression of Dvl2 was observed in stage IV samples than in stage I, but a low level of Nrdp1 was expressed in both stages and it did not differ significantly by stage. These results indicated that Nrdp1 is necessary for the reduction in Dvl2 to generate c-Jun in the EGF-KITENIN/ErbB4-c-Jun axis, but more importantly, elevated KITENIN protects KITENIN-bound ErbB4 from Nrdp1-mediated degradation via physical collaboration between the KITENIN/ErbB4 complex and Nrdp1, but not via modulation of ErbB kinase activity. Thus, KITENIN functions in the maintenance of a higher expression level of ErbB4 in advanced CRC tissues, independent of ubiquitin-mediated degradation via Nrdp1. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Proteínas Portadoras/metabolismo , Neoplasias Colorrectales/patología , Proteínas de la Membrana/metabolismo , Receptor ErbB-4/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Células CACO-2 , Proteínas Portadoras/genética , Línea Celular Tumoral , Neoplasias Colorrectales/metabolismo , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Células HEK293 , Células HT29 , Humanos , Proteínas de la Membrana/genética , Estadificación de Neoplasias , Fosforilación , Proteolisis , Receptor ErbB-4/genética
3.
Int J Mol Sci ; 18(12)2017 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-29207493

RESUMEN

Glycoprotein 90K (also known as LGALS3BP or Mac-2BP) is a tumor-associated protein, and high 90K levels are associated with poor prognosis in some cancers. To clarify the role of 90K as an indicator for poor prognosis and metastasis in epithelial cancers, the present study investigated the effect of 90K on an adherens junctional protein, E-cadherin, which is frequently absent or downregulated in human epithelial cancers. Treatment of certain cancer cells with 90K significantly reduced E-cadherin levels in a cell-population-dependent manner, and these cells showed decreases in cell adhesion and increases in invasive cell motility. Mechanistically, 90K-induced E-cadherin downregulation occurred via ubiquitination-mediated proteasomal degradation. 90K interacted with the E-cadherin-p120-catenin complex and induced its dissociation, altering the phosphorylation status of p120-catenin, whereas it did not associate with ß-catenin. In subconfluent cells, 90K decreased membrane-localized p120-catenin and the membrane fraction of the p120-catenin. Particularly, 90K-induced E-cadherin downregulation was diminished in p120-catenin knocked-down cells. Taken together, 90K upregulation promotes the dissociation of the E-cadherin-p120-catenin complex, leading to E-cadherin proteasomal degradation, and thereby destabilizing adherens junctions in less confluent tumor cells. Our results provide a potential mechanism to explain the poor prognosis of cancer patients with high serum 90K levels.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Biomarcadores de Tumor/metabolismo , Cadherinas/metabolismo , Proteínas Portadoras/metabolismo , Cateninas/metabolismo , Glicoproteínas/metabolismo , Neoplasias/metabolismo , Antígenos CD , Células CACO-2 , Cateninas/genética , Adhesión Celular , Recuento de Células , Línea Celular Tumoral , Membrana Celular/metabolismo , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Humanos , Células MCF-7 , Masculino , Invasividad Neoplásica , Fosforilación , Pronóstico , Proteolisis , Catenina delta
4.
BMC Complement Altern Med ; 16(1): 288, 2016 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-27527352

RESUMEN

BACKGROUND: Recent studies report that inflammatory diseases of the large intestine are associated with colorectal cancer. Geijigajakyak Decoction (GJD) has antispasmodic and anti-inflammatory effects on the gastrointestinal tract. Thus, in light of the connection between chronic bowel inflammation and colorectal cancer (CRC), we asked whether GJD inhibits colorectal tumorigenesis. METHODS: The effects of GJD on the viability and proliferation of CRC cells were evaluated using MTT and BrdU assays, respectively. The motility of CRC cells was examined by a Transwell migration/invasion assay and immunoblot analysis was used to examine the signaling pathways associated with migration. A syngeneic Balb/c mice allograft model, in which CT26 cells were injected into the dorsum, was used to evaluate the anti-tumor effects of GJD in vivo. RESULTS: GJD had no cytotoxic effects against HCT116 CRC cells, although it did inhibit their proliferation. GJD inhibited the migration of HCT116 cells, and suppressed the invasion of HCT116, Caco2, and CSC221 CRC cells. In addition, GJD downregulated the expression of p-JNK and p-p38 MAPK, which are downstream signaling molecules associated with invasiveness. Furthermore, oral administration of GJD (333 mg/kg, twice a day) inhibited tumor growth in a mouse xenograft model. CONCLUSIONS: GJD inhibited the motility of human CRC cells and suppressed tumorigenesis in a mouse model. These results suggest that GJD warrants further study as a potential adjuvant anti-cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/metabolismo , Medicamentos Herbarios Chinos/farmacología , Animales , Células HCT116 , Humanos , Ratones , Ratones Endogámicos BALB C , Invasividad Neoplásica , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Mol Ther ; 22(9): 1653-64, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24909917

RESUMEN

MicroRNAs are increasingly implicated in the modulation of the progression of various cancers. We previously observed that KAI1 C-terminal interacting tetraspanin (KITENIN) is highly expressed in sporadic human colorectal cancer (CRC) tissues and hence the functional KITENIN complex acts to promote progression of CRC. However, it remains unknown that microRNAs target KITENIN and whether KITENIN-targeting microRNAs modulate CRC cell motility and colorectal tumorigenesis. Here, through bioinformatic analyses and functional studies, we showed that miR-124, miR-27a, and miR-30b negatively regulate KITENIN expression and suppress the migration and invasion of several CRC cell lines via modulation of KITENIN expression. Through in vitro and in vivo induction of mature microRNAs using a tetracycline-inducible system, miR-124 was found to effectively inhibit the invasion of CT-26 colon adenocarcinoma cells and tumor growth in a syngeneic mouse xenograft model. Constitutive overexpression of precursor miR-124 in CT-26 cells suppressed in vivo tumorigenicity and resulted in decreased expression of KITENIN as well as that of MYH9 and SOX9, which are targets of miR-124. Thus, our findings identify that KITENIN-targeting miR-124, miR-27a, and miR-30b function as endogenous inhibitors of CRC cell motility and demonstrate that miR-124 among KITENIN-targeting microRNAs plays a suppressor role in colorectal tumorigenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Neoplasias del Colon/patología , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Animales , Células CACO-2 , Proteínas Portadoras/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Doxiciclina/administración & dosificación , Doxiciclina/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Humanos , Proteínas de la Membrana/genética , Ratones , MicroARNs/genética , Invasividad Neoplásica , Trasplante de Neoplasias
6.
Clin Transl Med ; 12(7): e871, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35853101

RESUMEN

The stability of a protein, as well as its function and versatility, can be enhanced through oligomerization. KITENIN (KAI1 C-terminal interacting tetraspanin) is known to promote the malignant progression of colorectal cancer (CRC). How KITENIN maintains its structural integrity and stability are largely unknown, however. Here we investigated the mechanisms regulating the stability of KITENIN with the aim of developing therapeutics blocking its oncogenic functions. We found that KITENIN formed a homo-oligomeric complex and that the intracellular C-terminal domain (KITENIN-CTD) was needed for this oligomerization. Expression of the KITENIN-CTD alone interfered with the formation of the KITENIN homodimer, and the amino acid sequence from 463 to 471 within the KITENIN-CTD was the most effective. This sequence coupled with a cell-penetrating peptide was named a KITENIN dimerization-interfering peptide (KDIP). We next studied the mechanisms by which KDIP affected the stability of KITENIN. The KITENIN-interacting protein myosin-X (Myo10), which has oncogenic activity in several cancers, functioned as an effector to stabilize the KITENIN homodimer in the cis formation. Treatment with KDIP resulted in the disintegration of the homodimer via downregulation of Myo10, which led to increased binding of RACK1 to the exposed RACK1-interacting motif (463-471 aa), and subsequent autophagy-dependent degradation of KITENIN and reduced CRC cell invasion. Intravenous injection of KDIP significantly reduced the tumour burden in a syngeneic mouse tumour model and colorectal liver metastasis in an intrasplenic hepatic metastasis model. Collectively, our present results provide a new cancer therapeutic peptide for blocking colorectal liver metastasis, which acts by inducing the downregulation of Myo10 and specifically targeting the stability of the oncogenic KITENIN protein.


Asunto(s)
Neoplasias Colorrectales , Proteínas de la Membrana , Péptidos , Animales , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Dimerización , Neoplasias Hepáticas Experimentales/metabolismo , Neoplasias Hepáticas Experimentales/secundario , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Ratones , Miosinas/química , Miosinas/metabolismo , Proteínas Oncogénicas/química , Proteínas Oncogénicas/metabolismo , Péptidos/farmacología , Estabilidad Proteica/efectos de los fármacos
7.
Gut ; 59(7): 907-17, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20581239

RESUMEN

BACKGROUND AND AIMS: 90K, a tumour-associated glycoprotein, interacts with galectins and has roles in host defence by augmenting the immune response, but the serum 90K level was suggested to indicate poor prognosis in several cancers. The cellular mechanisms of 90K action on colorectal cancer (CRC) cell motility and its effect on CRC progression were investigated. METHODS: The impact of 90K was analysed by combining cell cultures, in vitro assays, and immunohistochemistry. RESULTS: Secreted 90K suppresses CRC cell invasion, but this action of 90K is masked through binding with extracellular galectins. A novel pathway is identified comprising a secretory 90K and a CD9/CD82 tetraspanin web; in this pathway, 90K interacts with CD9/CD82, suppresses the Wnt/beta-catenin signal via a novel proteasomal-ubiquitination mechanism of beta-catenin that is dependent on ISG15 (interferon-stimulated gene-15) modification (ISGylation) but not on glycogen synthase kinase 3beta (GSK-3beta) and Siah/Adenomatous polyposis coli (APC). In a syngeneic mouse colon tumour model, tumour growth and lung metastasis were increased with 90K knockdown. In colon tissues from stage IV human CRC and invading cancer cells of corresponding metastatic liver tissues, in which beta-catenin and galectin expression was higher, immunostained 90K and CD9/CD82 were lower than in adjacent hepatic tissues or colon tissues from stage I. CONCLUSIONS: 90K itself has antitumour activity in CRC cells via suppression of Wnt signalling with a novel mechanism of ISGylation-dependent ubiquitination of beta-catenin when it interacts with CD9/CD82, but is downregulated in advanced CRC tissues. The data suggest a strategy of strengthening this novel pathway with concomitant knockdown of galectins as a potential therapeutic approach to CRC progression.


Asunto(s)
Antígenos CD/metabolismo , Neoplasias Colorrectales/metabolismo , Glicoproteínas/metabolismo , Proteína Kangai-1/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , beta Catenina/metabolismo , Animales , Antígenos de Neoplasias , Biomarcadores de Tumor , Proteínas Portadoras , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/patología , Medios de Cultivo Condicionados/farmacología , Citocinas/fisiología , Modelos Animales de Enfermedad , Regulación hacia Abajo/fisiología , Galectinas/metabolismo , Glicoproteínas/fisiología , Humanos , Lactosa/farmacología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/prevención & control , Neoplasias Pulmonares/secundario , Ratones , Proteínas de Neoplasias/fisiología , Transducción de Señal/fisiología , Tetraspanina 29 , Células Tumorales Cultivadas , Ubiquitina/metabolismo , Ubiquitinas/fisiología , Proteínas Wnt/metabolismo
8.
BMC Cancer ; 10: 197, 2010 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-20462455

RESUMEN

BACKGROUND: Coactivator-associated arginine methyltransferase 1 (CARM1) functions as a transcriptional coactivator of androgen receptor (AR)-mediated signaling. Correspondingly, overexpression of CARM1 has been associated with the development of prostate cancer (PCa) and its progression to androgen-independent PCa. In our preliminary study, however, the promoting effects of CARM1, with regard to androgen-stimulated AR target gene expression were minimal. These results suggested that the AR target gene expression associated with CARM1 may result primarily from non-hormone dependent activity. The goal of this study was to confirm the pattern of expression of CARM1 in human tumors and determine the mechanism of action in CARM1 overexpressed tumors. METHODS: Tissue microarray was used to determine the pattern of expression of CARM1 in human cancers by immunohistochemistry. CARM1 expression was also evaluated in prostate and colorectal surgical specimens and the clinical records of all cases were reviewed. In addition, a reporter transcription assay using the prostate-specific antigen (PSA) promoter was used to identify the signaling pathways involved in non-hormone-mediated signal activation associated with CARM1. RESULTS: The tissue microarray showed that CARM1 was particularly overexpressed in the colorectal cancers while CARM1 expression was not prevalent in the prostate and breast cancers. Further studies using surgical specimens demonstrated that CARM1 was highly overexpressed in 75% of colorectal cancers (49 out of 65) but not in the androgen-independent PCa. In addition, CARM1's coactivating effect on the entire PSA promoter was very limited in both androgen-dependent and androgen-independent PCa cells. These results suggest that there are other factors associated with CARM1 expression in PSA regulation. Indeed, CARM1 significantly regulated both p53 and NF-kappaB target gene transcription. CONCLUSIONS: The results of this study suggest that, in addition to its role in activation of steroid receptors, CARM1 functions as a transcriptional modulator by altering the activity of many transcriptional factors, especially with regard to androgen independent PCa and colorectal cancers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Colorrectales/enzimología , Neoplasias de la Próstata/enzimología , Proteína-Arginina N-Metiltransferasas/metabolismo , Andrógenos/metabolismo , Animales , Biomarcadores de Tumor/genética , Neoplasias de la Mama/enzimología , Línea Celular Tumoral , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Femenino , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Mutación , FN-kappa B/genética , FN-kappa B/metabolismo , Estadificación de Neoplasias , Regiones Promotoras Genéticas , Antígeno Prostático Específico/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteína-Arginina N-Metiltransferasas/genética , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Análisis de Matrices Tisulares , Transcripción Genética , Transfección , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
9.
Oncogene ; 38(49): 7416-7432, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31420606

RESUMEN

The cell surface receptor tyrosine kinase (RTK) exists in a dynamic state, however, it remains unknown how single membrane-spanning RTK proteins are retained in the plasma membrane before their activation. This study was undertaken to investigate how RTK proteins are anchored in the plasma membrane before they bind with their respective extracellular ligands for activation through protein-protein interaction, co-localization, and functional phenotype studies. Here we show that unconventional myosin-I MYO1D functions to hold members of the EGFR family (except ErbB3) at the plasma membrane. MYO1D binds only with unphosphorylated EGFRs and anchors them to underlying actin cytoskeleton at the plasma membrane. The C-terminal end region of the MYO1D tail domain containing a ß-meander motif is critical for direct binding with kinase domain of the EGFR family, and expression of the tail domain alone suppresses the oncogenic action of full-length MYO1D. Overexpressed MYO1D increases colorectal and breast cancer cell motility and viability through upregulating EGFR level, and thereby promotes colorectal tumor progression in a syngeneic mouse model. MYO1D is upregulated in human colorectal cancer tissues from advanced stages. Collectively, molecular motor MYO1D plays a distinct role in the dynamic regulation of EGFR family levels by holding them at the plasma membrane before their activation. Overexpressed MYO1D contributes to colorectal carcinogenesis possibly as a novel oncogene and thus may serve as an additional target for suppression of RTK signaling in cancer treatment.


Asunto(s)
Carcinogénesis/patología , Membrana Celular/metabolismo , Neoplasias Colorrectales/patología , Miosinas/fisiología , Citoesqueleto de Actina/metabolismo , Animales , Apoptosis , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Ligandos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Dominios Proteicos , Transducción de Señal , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Mol Cell Endocrinol ; 289(1-2): 29-37, 2008 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-18502031

RESUMEN

The proprotein convertase subtilisin/kexin (PCSKs), a family of subtilisin-like proteases, is the processing enzymes for the activation of many hormone precursors. The present study was designed to identify the PCSK isoform expressed in the ovary and to examine its expression in gonadotropin-stimulated rat ovary. Northern blot analysis of ovaries obtained from prepubertal rats revealed an increased expression of Pcsk5 messenger RNA (mRNA) during development with the highest levels at 21 days of age. Treatment of immature rats with PMSG further increased ovarian Pcsk5 expression, and in situ hybridization analysis revealed the localization of Pcsk5 mRNA in theca-interstitial cells of follicles in different sizes. Interestingly, treatment of PMSG-primed rats with hCG resulted in a transient stimulation of ovarian Pcsk5 mRNA levels within 3-6 h. In addition to theca-interstitial cells, hCG treatment induced the expression of Pcsk5 in granulosa cells of preovulatory follicles. Pcsk1, 2 and 4 mRNAs were not detected whereas Pcsk7 mRNA was slightly expressed. Injection of a progestin antagonist RU486 or an inhibitor of 3beta-hydroxysteroid dehydrogenase epostane at 1h before hCG treatment inhibited hCG-induced Pcsk5 mRNA levels. Treatment with LH stimulated both Pcsk5 mRNA and protein levels in preovulatory follicles cultured in vitro. In addition, forskolin but not TPA stimulated Pcsk5 mRNA levels. RNase protection assay revealed that the soluble Pcsk5A variant was the predominant form stimulated by gonadotropins in the ovary. Finally, the predicted proprotein substrates cleaved by PCSK5 were analyzed in preovulatory follicles using regular expressions. The present study demonstrates PCSK5A as the gonadotropin-regulated PCSK isoform in the ovary, and its possible contribution to ovulation by processing pro-TGFbeta and matrix metalloproteinase family.


Asunto(s)
Folículo Ovárico/metabolismo , Proproteína Convertasa 5/metabolismo , Factores de Edad , Envejecimiento , Animales , Gonadotropina Coriónica/metabolismo , Gonadotropina Coriónica/farmacología , Femenino , Hormona Folículo Estimulante/metabolismo , Hormona Folículo Estimulante/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células de la Granulosa/metabolismo , Hormona Luteinizante/metabolismo , Hormona Luteinizante/farmacología , Folículo Ovárico/crecimiento & desarrollo , Ovario/citología , Ovario/embriología , Ovario/metabolismo , Proproteína Convertasa 5/genética , ARN Mensajero/análisis , Ratas , Ratas Sprague-Dawley , Células Tecales/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
13.
Clin Cancer Res ; 22(5): 1284-94, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26527747

RESUMEN

PURPOSE AND EXPERIMENTAL DESIGN: The molecular events in the malignant progression of colon adenoma after loss of adenomatous polyposis coli (APC) are not fully understood. KITENIN (KAI1 C-terminal interacting tetraspanin) increases the invasiveness of colorectal cancer cells, and we identified a novel EGFR-independent oncogenic signal of EGF that works under coexpressed KITENIN and ErbB4. Here we tested whether elevated KITENIN and ErbB4 contribute to further progression of intestinal adenoma following APC loss. RESULTS: The intestinal tissues of villin-KITENIN transgenic mice in which villin-driven KITENIN expression induces increased c-Jun expression exhibit mild epithelial cell proliferation but no epithelial lineage changes compared with those of nontransgenic mice. Among the four ErbB4 isoforms, JM-a/CYT-2 and JM-b/CYT-2 exhibited the highest AP-1 activity when cells coexpressing KITENIN and each isoform were stimulated by EGF. Interestingly, predominant overexpression of the ErB4-CYT-2 mRNA as well as increased EGFR expression were observed in intestinal adenoma of APC(min/+) mice, which makes the microenvironment of activated EGF signaling. When we crossed villin-KITENIN mice with APC(min/+) mice, intestinal tumor tissues in the crossed mice showed the characteristics of early-stage invading adenocarcinoma. In patients with colorectal cancer, ErbB4-CYT-2 mRNA expression was significantly greater in tumor tissues than in normal adjacent tissues, but no significant differences in tumor tissue expression were found between different colorectal cancer stages. Furthermore, the mRNA expression of KITENIN and that of ErbB4-CYT-2 were positively correlated in human colorectal cancer tissue. CONCLUSIONS: Elevated coexpression of KITENIN and ErbB4-CYT-2 promotes the transition of colon adenoma to adenocarcinoma within an APC loss-associated tumor microenvironment.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/biosíntesis , Biomarcadores de Tumor/biosíntesis , Proteínas Portadoras/biosíntesis , Neoplasias Colorrectales/genética , Proteínas de la Membrana/biosíntesis , Receptor ErbB-4/biosíntesis , Adenocarcinoma/genética , Adenocarcinoma/patología , Adenoma/genética , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Biomarcadores de Tumor/genética , Proteínas Portadoras/genética , Proliferación Celular/genética , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Factor de Crecimiento Epidérmico/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/biosíntesis , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Isoformas de Proteínas/biosíntesis , Isoformas de Proteínas/genética , Receptor ErbB-4/genética , Microambiente Tumoral/genética
14.
Oncotarget ; 6(5): 3240-53, 2015 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-25605251

RESUMEN

KITENIN (KAI1 COOH-terminal interacting tetraspanin) promotes tumor invasion and metastasis in various cancers. This study assessed the association between KITENIN expression and advanced glioma grade in patients. In vitro assays revealed that KITENIN knockdown inhibited the invasion and migration of glioma cells, whereas KITENIN overexpression promoted their invasion and migration. In orthotopic mouse tumor models, mice transplanted with KITENIN-transfected glioma cells had significantly shorter survival than mice transplanted with mock-transfected cells. Patients with low KITENIN expression showed a significantly longer progression-free survival than patients with high KITENIN expression. KITENIN induced the expression of the epithelial-mesenchymal transition (EMT) markers (N-cadherin, ZEB1, ZEB2, SNAIL and SLUG) as well as the glioma stemness markers (CD133, ALDH1 and EPH-B1). Taken together, these findings showed that high levels of KITENIN increased glioma invasiveness and progression, associated with the up-regulation of EMT and stemness markers.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Proteínas Portadoras/metabolismo , Movimiento Celular , Transición Epitelial-Mesenquimal , Glioma/metabolismo , Proteínas de la Membrana/metabolismo , Células Madre Neoplásicas/metabolismo , Animales , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/cirugía , Proteínas Portadoras/genética , Línea Celular Tumoral , Femenino , Glioma/genética , Glioma/mortalidad , Glioma/patología , Glioma/cirugía , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica , Células Madre Neoplásicas/patología , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transfección , Regulación hacia Arriba
15.
Clin Cancer Res ; 20(15): 4115-28, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24893630

RESUMEN

PURPOSE: EGF-stimulated signaling via EGF receptor (EGFR) is important in colorectal tumorigenesis and drug targeting. However, anti-EGFR therapy is not effective in a subset of patients with colorectal cancer, suggesting that unidentified EGF-stimulated pathways might play roles in colorectal cancer. Previously, we identified KAI1 C-terminal interacting tetraspanin (KITENIN) as a metastasis-enhancing gene and found it to be highly expressed in sporadic colorectal cancer tissues. We recently found that EGF further increases KITENIN-induced elevated AP-1 activity. Here we attempted to clarify this novel EGF-stimulated molecular pathway and its roles in colorectal cancer. EXPERIMENTAL DESIGN: We analyzed how EGF modulates the downstream signaling pathway of oncogenic KITENIN in colorectal cancer cells. Biological alterations following EGF treatment were identified in KITENIN-overexpressed colorectal cancer cells with or without alteration of EGFR activity. RESULTS: We identified the KITENIN/ErbB4-Dvl2-c-Jun axis as a novel downstream signal of EGF that is switched on under elevated KITENIN conditions in an EGFR-independent manner. This unconventional EGF signal upregulates c-Jun and enhances invasion and anchorage-independent growth of colorectal cancer cells. In addition, tumor tissues from metastatic patients with colorectal cancer who showed initial poor responses to cetuximab/chemotherapy expressed higher levels of KITENIN than did responders to therapy. CONCLUSIONS: Our results highlight the role of an EGFR-independent EGF signal in mediating the invasiveness and tumorigenesis of colorectal cancer cells. This unconventional pathway might be related to the limited clinical efficacy of anti-EGFR agents in a subset of patients with colorectal cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas Portadoras/metabolismo , Neoplasias Colorrectales/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Hepáticas/secundario , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Receptor ErbB-4/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis , Biomarcadores de Tumor/genética , Western Blotting , Proteínas Portadoras/genética , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Proteínas Dishevelled , Resistencia a Antineoplásicos/efectos de los fármacos , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Proteínas de la Membrana/genética , Invasividad Neoplásica , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor ErbB-4/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas
16.
Yonsei Med J ; 52(3): 463-8, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21488189

RESUMEN

PURPOSE: KAI1 COOH-terminal interacting tetraspanin (KITENIN) has been found to act as a promoter of metastasis in murine models of colon cancer and squamous cell carcinoma (SCC). The suppression of tumor progression and metastasis of established colon cancer in mice was observed after intravenous delivery of small interfering RNA (siRNA) targeting KITENIN. The purpose of this study was to investigate the efficacy of gene therapy targeting KITENIN in human head and neck SCC. MATERIALS AND METHODS: SNU-1041, a well-established human hypopharyngeal SCC cell line, was used. KITENIN expression in SNU-1041 was measured by Western blot analysis. The cells were prepared, maintained in culture dishes with media, and divided into two groups: the si-KITENIN group and the scrambled group (control). The siRNA targeting KITENIN (si-KITENIN) and scrambled DNA were transfected into the SNU-1041 cells in each group. The effect of gene therapy was compared by in vitro experiments to evaluate invasion, migration, and proliferation. RESULTS: KITENIN was strongly expressed in the SNU-1041 cells, and the number of invaded cells was reduced more in the si-KITENIN group than in the scrambled group (p<0.001). The speed for the narrowing gap, made through adherent cells, was lower in the si-KITENIN group (p<0.001), and the number of viable proliferating cells was reduced in the si-KITENIN group compared to the scrambled group (p<0.001, the third day). KITENIN protein expression was no longer identified in the si-KITENIN group. CONCLUSION: Gene therapy using an anti-KITENIN strategy might be effective for head and neck squamous carcinoma.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Proteínas Portadoras/antagonistas & inhibidores , Terapia Genética , Neoplasias de Cabeza y Cuello/terapia , Proteínas de la Membrana/antagonistas & inhibidores , ARN Interferente Pequeño , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Proteínas Portadoras/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Proteínas de la Membrana/genética
17.
Laryngoscope ; 120(5): 953-8, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20422689

RESUMEN

OBJECTIVES/HYPOTHESIS: To investigate the expression of KAI1 COOH-terminal interacting tetraspanin (KITENIN) in patients with laryngeal cancers and to examine the correlation between its expression and various clinical and pathological variables. STUDY DESIGN: Cross-sectional study with planned data collection. METHODS: Tumor specimens were collected from 32 patients with laryngeal squamous carcinoma (collection of consecutive 32 tumor samples; 14 early stage, 18 advanced stage). Expression of KITENIN in the tissues obtained was determined by Western blot analysis and immunohistochemical staining. The patient characteristics including age, gender, tumor location, histology, stage, tumor extent, lymph node metastasis, and survival were obtained by review of the hospital records. RESULTS: KITENIN expression was significantly increased in laryngeal cancer tissues compared to adjacent normal tissue mucosa, as well as in metastatic lymph nodes compared to nonmetastatic lymph nodes. High KITENIN expression was significantly associated with advanced stage, tumor extent, and lymph node metastasis (P = .016, .016, and .005, respectively). There was no difference in the overall survival and disease-free survival between the low- and high-KITENIN expression groups among patients with laryngeal cancer. CONCLUSIONS: These results suggest that KITENIN expression may be associated with tumor progression in patients with laryngeal cancer. Further studies are needed to determine whether KITENIN expression adds prognostic value to conventional factors, such as the stage and status of metastasis, in a large series with a long period of follow-up.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Células Escamosas/patología , Proteínas Portadoras/análisis , Neoplasias Laríngeas/patología , Proteínas de la Membrana/análisis , Anciano , Biopsia , Western Blotting , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/cirugía , Supervivencia sin Enfermedad , Femenino , Humanos , Técnicas para Inmunoenzimas , Neoplasias Laríngeas/mortalidad , Neoplasias Laríngeas/cirugía , Laringe/patología , Metástasis Linfática/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico
18.
FEBS Lett ; 583(4): 711-7, 2009 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-19166844

RESUMEN

KAI1 C-terminal interacting tetraspanin (KITENIN) is reported to promote metastasis in mouse colon cancer models. We investigated the role of KITENIN on the progression of squamous cell carcinoma (SCC). In a preliminary clinical study using resected tissues from head and neck SCC patients, KITENIN was highly expressed in tumors and metastatic lymph nodes, while KAI1 was more increased in adjacent mucosa than in tumor. KITENIN-transfected mouse squamous cancer (SCC VII/KITENIN) cells showed significantly higher invasion, migration, and proliferation than empty vector-transfected cells. In syngeneic mouse squamous tumor models, more increased tumor volume and enhanced lung metastasis were found in SCC VII/KITENIN cells-injected mice. Thus, KITENIN increases invasion and migration of squamous cancer cells and thereby promotes distant metastasis in mouse squamous tumor models.


Asunto(s)
Carcinoma de Células Escamosas/secundario , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Neoplasias de Cabeza y Cuello/secundario , Neoplasias Pulmonares/secundario , Proteínas de la Membrana/metabolismo , Animales , Carcinoma de Células Escamosas/genética , Proteínas Portadoras/genética , Línea Celular Tumoral , Medio de Cultivo Libre de Suero , Femenino , Vectores Genéticos , Neoplasias de Cabeza y Cuello/genética , Humanos , Neoplasias Pulmonares/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C3H , Invasividad Neoplásica/genética , Metástasis de la Neoplasia , ARN Mensajero/metabolismo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA