Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Int Immunopharmacol ; 122: 110580, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37418984

RESUMEN

Lung adenocarcinoma (LUAD) is a malignant respiratory disease, resulting in a heavy social burden. Epidermal growth factor receptor-tyrosine kinase inhibitor (EGFR-TKI) resistance and tumor immune microenvironment are important directions in the treatment of LUAD. In this study, we confirmed the role of ADAM metallopeptidase domain 12 (ADAM12) in LUAD development and progression. Our bioinformatic analysis was conducted to screen ADAM12 was correlated with EGFR-TKI and immune infiltration in LUAD patients. Our results showed that the transcription and post-transcription level of ADAM12 is significantly increased in tumor samples compared to normal samples, and ADAM12 correlated with poor prognosis in LUAD patients. High level of ADAM12 accelerated the LUAD progression via promoting proliferation, cell cycle, apoptosis escaping, immune escaping, EGFR-TKI resistance, angiogenesis, invasion and migration based on experiment validation in vitro and in vivo, which could be attenuated by ADAM12 knockdown. Further mechanistic studies suggested that the PI3K/Akt/mTOR and RAS signaling pathways were activated after ADAM12 knockdown. Therefore, ADAM12 might be validated as a possible molecular therapy target and prognostic marker for patients with LUAD.


Asunto(s)
Adenocarcinoma del Pulmón , Neoplasias Pulmonares , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Neoplasias Pulmonares/patología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Resistencia a Antineoplásicos , Línea Celular Tumoral , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Proliferación Celular , Microambiente Tumoral , Proteína ADAM12/genética , Proteína ADAM12/metabolismo
2.
J Transl Med ; 21(1): 56, 2023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36717944

RESUMEN

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is a major worldwide health problem due to its high prevalence and mortality rate. A disintegrin and metalloproteinase 12 (ADAM12) is aberrantly expressed in various cancers and plays an important role in tumor progression. However, its explicit effect and molecular mechanism in ccRCC remain unclear. METHODS: We investigated the dysregulation of ADAM12 in ccRCC through public databases and bioinformatics analyses. The expression of ADAM12 was further verified in ccRCC tissues by RT-qPCR and immunohistochemistry (IHC). The relationship between ADAM12 expression and clinicopathological characteristics was analyzed statistically. The effects of ADAM12 on the proliferation, migration and invasion of ccRCC cells were examined by in vitro and in vivo experiments. RESULTS: ADAM12 was significantly upregulated in ccRCC tissues and associated with poor prognosis in ccRCC patients. ADAM12 promoted ccRCC cell proliferation, migration and invasion in vitro and the growth of subcutaneous tumors in vivo. Knockdown of ADAM12 successfully suppressed its oncogenic function. Mechanistically, its overexpression induced epithelial-mesenchymal transition (EMT) by downregulating E-cadherin and upregulating N-cadherin and Snail. Moreover, ADAM12 participated in the epidermal growth factor receptor (EGFR) pathway and activated the downstream signal ERK1/2 by shedding the EGFR ligand, thereby upregulating target genes including c-Myc, enhancing cell survival and invasion ability, and promoting tumor progression, metastasis and the induction of EMT. CONCLUSIONS: High expression of ADAM12 induced EMT and promoted cell proliferation, migration, and invasion by activating the EGFR/ERK signaling pathway in ccRCC.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/patología , Transición Epitelial-Mesenquimal/genética , Línea Celular Tumoral , Transducción de Señal/genética , Proliferación Celular/genética , Neoplasias Renales/patología , Receptores ErbB/metabolismo , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Proteína ADAM12/genética , Proteína ADAM12/metabolismo
3.
Dig Liver Dis ; 55(3): 412-421, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-35853821

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is an extremely aggressive malignant tumor associated with high migratory and invasive potential. The present study intends to explore regulatory mechanism of p53/microRNA (miR)-29c-3p/A disintegrin and metalloproteinase 12 (ADAM12) axis in HCC based on clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) technology. METHODS: Putative miR-29c-3p binding sites on ADAM12 3'UTR were verified by a luciferase assay. The binding affinity of p53 to miR-29c-3p was assessed based on CRISPR/Cas9 technology to construct a p53 knockout (p53-/-) HCCLM3 cell line. Furthermore, the effect of p53/miR-29c-3p/ADAM12 was assessed on maligant phenotypes in vitro and tumor formation and metastasis in nude mice. RESULTS: ADAM12 was highly expressed but miR-29c-3p was poorly expressed in HCC. miR-29c-3p inhibited migratory and invasive abilities of HCC cells by targeting ADAM12 expression. p53 was found to target and upregulate miR-29c-3p, thus downregulating ADAM12 and conferring inhibitory effect on HCC cell activities. Moreover, ADAM12 knockout or p53 overexpression reduced HCC tumor formation and metastasis, which were reversed by further silencing of miR-29c-3p. CONCLUSION: The identification of the p53/miR-29c-3p/ADAM12 axis in migration and invasion of HCC may potentially further our understanding of mechanisms underpinning HCC, and also bear translational value as novel molecular targets.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroARNs , Animales , Ratones , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/patología , Ratones Desnudos , MicroARNs/genética , Proteína p53 Supresora de Tumor/genética , Proteína ADAM12/metabolismo
4.
Clin Transl Oncol ; 25(5): 1425-1435, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36512304

RESUMEN

BACKGROUND: Gemcitabine (GEM)-based chemotherapy regimens is widely used in bladder cancer (BC) patients. However, GEM resistance may occur and result in treatment failure and disease progression. A disintegrin and metalloprotease 12 (ADAM12) plays a critical role in many cancers. However, the role of ADAM12 in GEM resistance of BC remains unclear. METHODS: We analyzed the relationship between ADAM12 expression and tumor characteristics using the data downloaded from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database. Then, we established GEM resistant BC cell lines and used quantitative real-time PCR, western blot, cell counting kit-8, immunohistochemistry, and xenograft mouse model to investigate the role of ADAM12 in GEM resistance. RESULTS: In general, ADAM12 was found to be upregulated in GEM resistant BC cells. ADAM12 knockdown increased the chemosensitivity of BC cells. We further proved that ADAM12 could promote GEM resistance by activating the epidermal growth factor receptor (EGFR) signaling pathway in BC. Furthermore, the epithelial-mesenchymal transition (EMT) phenotype was observed in GEM resistant BC cells. ADAM12 induced EMT process and promotes tumor progression in BC. CONCLUSION: Our findings suggested that ADAM12 was a key gene for GEM resistance and positively correlated with malignancy of BC. It might serve as a novel and valuable therapeutic target for BC.


Asunto(s)
Antineoplásicos , Resistencia a Antineoplásicos , Transición Epitelial-Mesenquimal , Gemcitabina , Neoplasias de la Vejiga Urinaria , Animales , Humanos , Ratones , Proteína ADAM12/genética , Proteína ADAM12/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/fisiología , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Transición Epitelial-Mesenquimal/fisiología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Gemcitabina/farmacología , Gemcitabina/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Transducción de Señal/genética , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
5.
Curr Med Sci ; 42(6): 1131-1139, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36542326

RESUMEN

OBJECTIVE: Pituitary adenomas (PAs) can adapt an aggressive phenotype by invading adjacent brain structures with rapid cellular proliferation. Previous studies demonstrated that excessive expression of metalloproteases ADAM12 and MMP-14 is instrumental for the active proliferation and invasiveness of PA cells in vitro and of tumors in vivo. However, the mechanisms regulating ADAM12 and MMP-14 expression in PAs remain unclear. METHODS: Target gene prediction and transcriptomic profiling of invasive vs. noninvasive human PA samples were performed to identify miRNA species potentially involved in the regulation of ADAM12 and MMP14. For cellular analyses of miRNA functions, two mouse PA cell lines (AtT20 and TtT/GF) were transfected with miR-149-3p and miR-149-5p, respectively. The effects of miR-149 (3p and 5p) on expression levels of ADAM12 and MMP14 were determined by Western blotting followed by an analysis of proliferation and colony formation assays, scratch migration assays, and invasion assays. RESULTS: A significant downregulation of miRNA-149 was observed in invasive vs. noninvasive PA (0.32 vs. 0.09, P<0.0001). In AtT-20 and TtT/GF mouse PAs cells, transfection of mimic miRNA-149 (3p and 5p) caused a significantly reduced cell proliferation and matrigel invasion, whilst the effect on cell migration was less pronounced. Both strands of miRNA-149 (3p and 5p) markedly reduced protein levels of ADAM12 and MMP-14 by at least 40% in both cell lines. CONCLUSION: This study proved that the invasiveness of PA cells is, at least partly, regulated by miRNA-149-dependent expression of ADAM12 and MMP-14.


Asunto(s)
MicroARNs , Neoplasias Hipofisarias , Ratones , Animales , Humanos , Neoplasias Hipofisarias/genética , Metaloproteinasa 14 de la Matriz/genética , Metaloproteinasa 14 de la Matriz/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Proliferación Celular/genética , Movimiento Celular/genética , Proteína ADAM12/genética , Proteína ADAM12/metabolismo
6.
BMC Cancer ; 22(1): 394, 2022 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-35413826

RESUMEN

BACKGROUND: Recently it has been recognized that stromal markers could be used as a clinically relevant biomarker for therapy response and prognosis. Here, we report on a serum marker for stromal activation, A Disintegrin and Metalloprotease 12 (ADAM12) in colorectal cancer (CRC). METHODS: Using gene expression databases we investigated ADAM12 expression in CRC and delineated the source of ADAM12 expression. The clinical value of ADAM12 was retrospectively assessed in the CAIRO2 trial in metastatic CRC with 235 patients (31% of total cohort), and an independent rectal cancer cohort (n = 20). RESULTS: ADAM12 is expressed by activated CRC associated fibroblasts. In the CAIRO2 trial cohort, ADAM12 serum levels were prognostic (ADAM12 low versus ADAM12 high; median OS 25.3 vs. 17.1 months, HR 1.48 [95% CI 1.11-1.96], P = 0.007). The prognostic potential was specifically high for metastatic rectal cancer (HR 1.78 [95% CI 1.06-3.00], P = 0.030) and mesenchymal subtype tumors (HR 2.12 [95% CI 1.25-3.60], P = 0.004). ADAM12 also showed potential for predicting recurrence in an exploratory analysis of non-metastatic rectal cancers. CONCLUSIONS: Here we describe a non-invasive marker for activated stroma in CRC which associates with poor outcome, especially for primary cancers located in the rectum.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias del Colon , Neoplasias Colorrectales , Neoplasias del Recto , Proteína ADAM12/genética , Proteína ADAM12/metabolismo , Biomarcadores , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Neoplasias Colorrectales/patología , Humanos , Pronóstico , Estudios Retrospectivos
7.
Eur J Histochem ; 65(3)2021 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-34587717

RESUMEN

Oral squamous cell carcinoma (OSCC) is the most common malignant tumor in the head and neck, and radiotherapy is the main approach for this disease, while irradiation resistance is a huge challenge that influences radiosensitivity. This study aims to determine the role and function of miR-29a-3p and ADAM12 in the radiosensitivity of OSCC cells. The expression pattern of ADAM12 in OSCC cells was searched in TCGA database. The binding of miR-29a-3p and ADAM12 was predicted by Starbase and verified using dual luciferase reporter gene assay. The RNA or protein expressions of miR-29a-3p and ADAM12 were measured by RT-qPCR or western blot. OSCC cell lines were treated by various γ-ray irradiation dosages before the alteration on miR-29a-3p expression and on the cell viability, proliferation, migration and cell apoptosis was detected. ADAM12 was highly expressed in OSCC cells, whose expression in resistant cells was positively correlated with irradiation dosage. Overexpression of ADAM12 in OSCC cells lead to increased cell proliferation and migration ability as well as inhibited cell apoptosis. miRNAs potentially binding ADAM12 in PITA, microT, miRmap and targetscan were screened, among which miR-29a-3p had the maximum differential expression levels in OSCC cells determined by RT-qPCR. Overexpression of miR-29a-3p resulted in suppressed cell viability, proliferation, migration ability and increased cell apoptosis, while this expression pattern can be partially counteracted by ADAM12 overexpression in OSCC cells. miR-29a-3p through targeting and inhibiting AMDM12 enhances the radiosensitivity of OSCC cells.


Asunto(s)
Proteína ADAM12/antagonistas & inhibidores , Neoplasias de Cabeza y Cuello/metabolismo , MicroARNs/metabolismo , Tolerancia a Radiación/fisiología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Proteína ADAM12/genética , Proteína ADAM12/metabolismo , Apoptosis/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Técnicas de Silenciamiento del Gen , Humanos
8.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33952697

RESUMEN

Breast cancer patients with increased expression of hypoxia-inducible factors (HIFs) in primary tumor biopsies are at increased risk of metastasis, which is the major cause of breast cancer-related mortality. The mechanisms by which intratumoral hypoxia and HIFs regulate metastasis are not fully elucidated. In this paper, we report that exposure of human breast cancer cells to hypoxia activates epidermal growth factor receptor (EGFR) signaling that is mediated by the HIF-dependent expression of a disintegrin and metalloprotease 12 (ADAM12), which mediates increased ectodomain shedding of heparin-binding EGF-like growth factor, an EGFR ligand, leading to EGFR-dependent phosphorylation of focal adhesion kinase. Inhibition of ADAM12 expression or activity decreased hypoxia-induced breast cancer cell migration and invasion in vitro, and dramatically impaired lung metastasis after orthotopic implantation of MDA-MB-231 human breast cancer cells into the mammary fat pad of immunodeficient mice.


Asunto(s)
Proteína ADAM12/genética , Proteína ADAM12/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Regulación Neoplásica de la Expresión Génica , Hipoxia/metabolismo , Proteína ADAM12/deficiencia , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular , Receptores ErbB/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Humanos , Pulmón/patología , Neoplasias Pulmonares/patología , Ratones , Ratones SCID , Metástasis de la Neoplasia/genética , Transducción de Señal , Microambiente Tumoral
9.
Pathol Res Pract ; 221: 153449, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33930608

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is a kind of malignant tumor of digestive system severely affecting human health. The occurrence of CRC is a polygenic and multi-step complex process involving genetic and epigenetic alterations. ADAM12 (a disintegrin and metalloproteases 12), is a gene that was commonly hypermethylated in esophageal cancer using whole-genome methylation microarray in our previous study. METHODS: We detected the methylation frequencies of the CpG island in ADAM12 promoter using bisulfite-pyrosequencing in CRC cell lines and tissue samples. The expression of ADAM12 was detected by quantitative real-time PCR (qRT-PCR) and immunohistochemistry (IHC). A systematic and comprehensive analysis of relationship of DNA hypermethylation and ADAM12 expression in CRC was performed in our samples and TCGA database. RESULTS: The expression of ADAM12 in hypermethylated cell lines was significantly lower than that in hypomethylated cell lines, and demethylation agent 5-Aza-dC could demethylate ADAM12 promoter region and reactivate ADAM12 expression effectively. In 74 pairs of colorectal cancer and normal tissues, bisulfite-pyrosequencing results showed significantly hypermethylation of ADAM12 in CRC compared with adjacent normal mucosa, accompanied with lower expression of ADAM12 in CRC tissues compared to that of the normal tissues. In addition, there was a statistically significant negative correlation between ADAM12 protein expression and methylation levels (rho =-0.28, p = 0.015). CONCLUSION: Promoter hypermethylation was probably a mechanism of ADAM12 epigenetic silencing in CRC.


Asunto(s)
Proteína ADAM12/genética , Neoplasias Colorrectales/patología , Metilación de ADN/genética , Regulación Neoplásica de la Expresión Génica/genética , Regiones Promotoras Genéticas/genética , Proteína ADAM12/metabolismo , Adulto , Anciano , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad
10.
Int J Mol Sci ; 22(5)2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33804570

RESUMEN

Accumulating evidence indicates that an elevated ephrin-A1 expression is positively correlated with a worse prognosis in some cancers such as colon and liver cancer. The detailed mechanism of an elevated ephrin-A1 expression in a worse prognosis still remains to be fully elucidated. We previously reported that ADAM12-cleaved ephrin-A1 enhanced lung vascular permeability and thereby induced lung metastasis. However, it is still unclear whether or not cleaved forms of ephrin-A1 are derived from primary tumors and have biological activities. We identified the ADAM12-mediated cleavage site of ephrin-A1 by a Matrix-assisted laser desorption ionization mass spectrometry and checked levels of ephrin-A1 in the serum and the urine derived from the primary tumors by using a mouse model. We found elevated levels of tumor-derived ephrin-A1 in the serum and the urine in the tumor-bearing mice. Moreover, inhibition of ADAM-mediated cleavage of ephrin-A1 or antagonization of the EphA receptors resulted in a significant reduction of lung metastasis. The results suggest that tumor-derived ephrin-A1 is not only a potential biomarker to predict lung metastasis from the primary tumor highly expressing ephrin-A1 but also a therapeutic target of lung metastasis.


Asunto(s)
Proteína ADAM12/metabolismo , Carcinoma Pulmonar de Lewis/patología , Efrina-A1/metabolismo , Receptor EphA2/metabolismo , Proteína ADAM12/genética , Animales , Permeabilidad Capilar , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Efrina-A1/genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Receptor EphA2/genética , Células Tumorales Cultivadas
11.
Int J Mol Sci ; 23(1)2021 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-35008854

RESUMEN

Both Type 1 diabetes mellitus (DM1) and type 2 diabetes mellitus (DM2) are associated with an increased risk of limb amputation in peripheral arterial disease (PAD). How diabetes contributes to poor PAD outcomes is poorly understood but may occur through different mechanisms in DM1 and DM2. Previously, we identified a disintegrin and metalloproteinase gene 12 (ADAM12) as a key genetic modifier of post-ischemic perfusion recovery. In an experimental PAD, we showed that ADAM12 is regulated by miR-29a and this regulation is impaired in ischemic endothelial cells in DM1, contributing to poor perfusion recovery. Here we investigated whether miR-29a regulation of ADAM12 is altered in experimental PAD in the setting of DM2. We also explored whether modulation of miR-29a and ADAM12 expression can improve perfusion recovery and limb function in mice with DM2. Our result showed that in the ischemic limb of mice with DM2, miR-29a expression is poorly downregulated and ADAM12 upregulation is impaired. Inhibition of miR-29a and overexpression of ADAM12 improved perfusion recovery, reduced skeletal muscle injury, improved muscle function, and increased cleaved Tie 2 and AKT phosphorylation. Thus, inhibition of miR-29a and or augmentation of ADAM12 improves experimental PAD outcomes in DM2 likely through modulation of Tie 2 and AKT signalling.


Asunto(s)
Proteína ADAM12/metabolismo , Diabetes Mellitus Experimental/fisiopatología , Isquemia/complicaciones , MicroARNs/metabolismo , Músculo Esquelético/lesiones , Músculo Esquelético/fisiopatología , Enfermedad Arterial Periférica/fisiopatología , Recuperación de la Función , Animales , Capilares/patología , Diabetes Mellitus Experimental/genética , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Regulación hacia Abajo/genética , Células Progenitoras Endoteliales/metabolismo , Conducta Alimentaria , Isquemia/fisiopatología , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , Músculo Esquelético/patología , Perfusión , Enfermedad Arterial Periférica/genética , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regulación hacia Arriba/genética
12.
FEBS Open Bio ; 10(11): 2489-2498, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33030286

RESUMEN

Selective estrogen receptor modulators (SERMs) significantly affect survival and invasiveness of rodent pituitary adenoma (PA) cells. The impact of three clinically relevant SERMs (bazedoxifene, clomiphene, raloxifene) on invasiveness and on gene and protein expression of invasion-related proteases [matrix metalloproteinase-14 (MMP-14) and A disintegrin and metalloproteinase-12 (ADAM12)] was analyzed in murine PA cells (AtT-20 and TtT/GF). All SERMs significantly decreased cell invasiveness. Moreover, SERMs significantly decreased expression of ADAM12 mRNA in both cell lines and of MMP-14 mRNA in TtT/GF cells. Invasion rates of AtT-20 and TtT/GF significantly decreased after ADAM12 gene silencing, and the invasion rate of TtT/GF cells significantly decreased after MMP-14 gene silencing. All SERMs affected ADAM12 protein expression in AtT-20 cells whereas bazedoxifene and raloxifene decreased MMP-14 protein expression in TtT/GF cells. We conclude that SERMs attenuate invasiveness of murine PA cells by downregulating expression levels of invasion-related proteases MMP-14 and ADAM12.


Asunto(s)
Proteína ADAM12/genética , Adenoma/patología , Metaloproteinasa 14 de la Matriz/genética , Neoplasias Hipofisarias/patología , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Proteína ADAM12/metabolismo , Adenoma/genética , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Concentración 50 Inhibidora , Metaloproteinasa 14 de la Matriz/metabolismo , Ratones , Invasividad Neoplásica , Neoplasias Hipofisarias/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
13.
J Chem Neuroanat ; 109: 101846, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32622867

RESUMEN

ADAM (a disintergin and metalloprotease) 12 is a member of the large family of multidomain metalloprotease-disintegrins, which possess cell-binding and metalloprotease properties. The enzyme is responsible for the shedding of a number of membrane-bound proteins (heparin-binding-EGF, insulin-like growth factor 2-binding proteins 3 and 5, oxytocinase, glycoprotein non-metastatic melanoma protein B and basigin). In rat and human CNS, ADAM12 is predominantly localized in white and gray matter oligodendrocytes. In addition it can be detected in astrocytes, neurons and endothelial cells. Its function in healthy brain is not well established yet, but prominent roles in CNS development, myelination and high cognitive abilities are discussed. There is increasing evidence that ADAM12 is involved in numerous major diseases of the CNS, which are summarized in the present review (brain tumors, multiple sclerosis, amyotrophic lateral sclerosis, Alzheimer´s disease, stroke, schizophrenia, autism and bipolar disorder).


Asunto(s)
Proteína ADAM12/metabolismo , Neoplasias Encefálicas/diagnóstico , Encéfalo/metabolismo , Cognición/fisiología , Animales , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/metabolismo , Humanos , Oligodendroglía/metabolismo , Ratas
14.
Int J Oncol ; 56(5): 1162-1174, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32319603

RESUMEN

ADAM metallopeptidase domain 12 (ADAM12) has been demonstrated to mediate cell proliferation and apoptosis resistance in several types of cancer cells. However, the effect of ADAM12 silencing on the proliferation and apoptosis of choriocarcinoma cells remains unknown. The present study revealed that ADAM12 silencing significantly inhibited cellular activity and proliferation in the human choriocarcinoma JEG3 cell line and increased the rate of apoptosis. In addition, ADAM12 silencing significantly increased the expression levels of the autophagy proteins microtubule­associated protein­light­chain 3 (LC3B) and autophagy related 5 (ATG5) and the fluorescence density of LC3B in JEG­3 cells. However, the suppression of autophagy by 3­methyladenine could block ADAM12 silencing­induced cellular apoptosis. ADAM12 silencing reduced the levels of the inflammatory factors interleukin­1ß, interferon­Î³ and TNF­α, and inactivated nuclear p65­NF­κB and p­mTOR in JEG­3 cells. The downregulation of p­mTOR expression by ADAM12 silencing was rescued in 3­methyladenine­treated JEG­3 cells, indicating that mTOR might participate in the autophagy­mediated pro­apoptotic effect of ADAM12 silencing. In conclusion, ADAM12 silencing promoted cellular apoptosis in human choriocarcinoma JEG3 cells, which might be associated with autophagy and the mTOR response. These findings indicate that ADAM12 silencing might be a potential novel therapeutic target for choriocarcinoma.


Asunto(s)
Proteína ADAM12/genética , Proteína ADAM12/metabolismo , Coriocarcinoma/genética , ARN Interferente Pequeño/farmacología , Neoplasias Uterinas/genética , Adenina/análogos & derivados , Adenina/farmacología , Apoptosis , Autofagia , Proteína 5 Relacionada con la Autofagia/metabolismo , Línea Celular Tumoral , Proliferación Celular , Coriocarcinoma/metabolismo , Femenino , Silenciador del Gen , Humanos , Proteínas Asociadas a Microtúbulos/metabolismo , Embarazo , Neoplasias Uterinas/metabolismo
15.
Biomed Res Int ; 2020: 4351671, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32025520

RESUMEN

Cervical cancer is one of the malignant tumors that seriously threaten women's health. The mechanism of development needs to be deeply studied. In recent years, lncRNA has been identified as one of the important factors affecting the malignant progression of tumors. In this study, we illustrated the important mechanism of lncRNA CAR10 in the development of cervical cancer. We found that CAR10 is significantly increased in4 cervical cancer tissues and cells, which can promote the proliferation of cervical cancer cells in vitro and in vivo, indicating that CAR10 is involved in the progression of cervical cancer as an oncogene. Further studies showed that CAR10 is a target gene of miR-125b-5p, and miR-125b-5p can inhibit the effect of CAR10 on the proliferation of cervical cancer cells. In addition, we also found that 3-phosphoinositide-dependent protein kinase 1 (PDPK1) is also a target gene of miR-125b-5p, and CAR10 can upregulate the expression level of PDPK1. The results showed that CAR10 acts as a ceRNA to upregulate the expression of PDPK1 by sponging miR-125b-5p. Knockdown of PDPK1 can inhibit the effect of CAR10 on cervical cancer cells. Our study demonstrates that, based on ceRNA mechanism, CAR10/miR-125b-5p/PDPK1 network can regulate the proliferation of cervical cancer cells and play an important role in the development of cervical cancer. In addition, our study also suggests that intervention of CAR10/miR-125b-5p/PDPK1 network may be a new strategy for targeted therapy of cervical cancer.


Asunto(s)
Proteínas Quinasas Dependientes de 3-Fosfoinosítido/metabolismo , Proteína ADAM12/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Proteínas Quinasas Dependientes de 3-Fosfoinosítido/genética , Proteína ADAM12/genética , Animales , Apoptosis , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , ARN Largo no Codificante/genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
16.
Cells ; 8(7)2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31319505

RESUMEN

Background: The odontogenic keratocyst (OKC) is an odontogenic cyst that shows aggressive and intriguing biological behavior. It is suggested that a hypoxic environment occurs in OKC, which led us to investigate the immunoexpression and location of hypoxia-inducible factor 1-alpha (HIF-1α) and other hypoxia-related proteins. Methods: Twenty cases of OKC were evaluated for the expression of Notch homolog 1 (NOTCH1), HIF-1α, disintegrin and metalloproteinase domain-containing protein 12 (ADAM-12), and heparin-binding epidermal growth factor-like growth factor (HBEGF) by immunohistochemistry and compared to eight control cases of calcifying odontogenic cystic (COC), orthokeratinized odontogenic cyst (OOC), and normal oral mucosa (OM) in basal and parabasal layers. Results: In OKC, all the proteins tested were expressed significantly higher in both basal (except for NOTCH1 and HBEGF in OOC) and suprabasal epithelial layers compared to controls. Looking at the epithelial layers within OKC, we observed an increased NOTCH1 and HIF-1α expression in parabasal layers. Conclusions: These results suggest that hypoxia occurs more intensively in OKC compared to COC, OM, and OOC. Hypoxia appeared to be stronger in parabasal layers as observed by higher HIF-1α expression in upper cells. Overexpression of NOTCH1, ADAM-12, and HBEGF in OKC was observed, which suggests that microenvironmental hypoxia could potentially regulate the expression of hypoxia-related proteins, and consequently, its clinical and biological behavior.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Quistes Odontogénicos/metabolismo , Receptor Notch1/metabolismo , Transducción de Señal , Proteína ADAM12/genética , Proteína ADAM12/metabolismo , Hipoxia de la Célula , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Quistes Odontogénicos/patología , Oxígeno/metabolismo , Receptor Notch1/genética , Regulación hacia Arriba
17.
Breast J ; 25(4): 691-695, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31079422

RESUMEN

Currently, tumor biopsies are used for breast cancer molecular subtyping. Biopsies are associated with various pathological changes and are thought to contribute to the dissemination of tumor cells. Extracellular vesicles shed by tumor cells into circulation exhibit the molecular signature of the parent cells. Herein, we show that proteomic analysis of circulating EV can discriminate BC patients from healthy subjects and indicate stage of the disease. Also, we performed a correlation between the BC molecular subtype using plasma EV and immunohistochemistry of tumor biopsies. Circulating EV may represent a useful, non-invasive tool to study the molecular makeup of BC tumors.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias de la Mama/patología , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patología , Proteína ADAM12/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Neoplasias de la Mama/sangre , Estudios de Casos y Controles , Línea Celular Tumoral , Receptor alfa de Estrógeno/metabolismo , Femenino , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Inhibidor 1 de Activador Plasminogénico/metabolismo , Proteómica , Receptor ErbB-2/metabolismo , Receptores de Progesterona/metabolismo , beta Catenina/metabolismo
18.
Int J Mol Sci ; 20(8)2019 04 22.
Artículo en Inglés | MEDLINE | ID: mdl-31013576

RESUMEN

The transmembrane glycoprotein basigin, a member of the immunoglobulin superfamily, stimulates matrix metalloproteinase (MMP)-mediated extracellular matrix (ECM) degradation and thereby drives cancer cell invasion. Basigin is proteolytically shed from the cell surface and high concentrations of soluble basigin in the blood dictates poor prognosis in cancer patients. A positive correlation between basigin and a disintegrin and metalloproteinase (ADAM)-12 in serum from prostate cancer patients has been reported. Yet, the functional relevance of this correlation is unknown. Here, we show that ADAM12 interacts with basigin and cleaves it in the juxtamembrane region. Specifically, overexpression of ADAM12 increases ectodomain shedding of an alkaline phosphatase-tagged basigin reporter protein from the cell surface. Moreover, CRISPR/Cas9-mediated knockout of ADAM12 in human HeLa carcinoma cells results in reduced shedding of the basigin reporter, which can be rescued by ADAM12 re-expression. We detected endogenous basigin fragments, corresponding to the expected size of the ADAM12-generated ectodomain, in conditioned media from ADAM12 expressing cancer cell-lines, as well as serum samples from a healthy pregnant donor and five bladder cancer patients, known to contain high ADAM12 levels. Supporting the cancer relevance of our findings, we identified several cancer-associated mutations in the basigin membrane proximal region. Subsequent in vitro expression showed that some of these mutants are more prone to ADAM12-mediated shedding and that the shed ectodomain can enhance gelatin degradation by cancer cells. In conclusion, we identified ADAM12 as a novel basigin sheddase with a potential implication in cancer.


Asunto(s)
Proteína ADAM12/metabolismo , Basigina/metabolismo , Proteína ADAM12/química , Proteína ADAM12/genética , Secuencia de Aminoácidos , Basigina/química , Basigina/genética , Sistemas CRISPR-Cas , Línea Celular , Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Humanos , Mutación , Especificidad por Sustrato
19.
Nucleic Acids Res ; 47(7): 3407-3421, 2019 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-30753595

RESUMEN

The proper tissue-specific regulation of gene expression is essential for development and homeostasis in metazoans. However, the illegitimate expression of normally tissue-restricted genes-like testis- or placenta-specific genes-is frequently observed in tumors; this promotes transformation, but also allows immunotherapy. Two important questions are: how is the expression of these genes controlled in healthy cells? And how is this altered in cancer? To address these questions, we used an unbiased approach to test the ability of 350 distinct genetic or epigenetic perturbations to induce the illegitimate expression of over 40 tissue-restricted genes in primary human cells. We find that almost all of these genes are remarkably resistant to reactivation by a single alteration in signaling pathways or chromatin regulation. However, a few genes differ and are more readily activated; one is the placenta-expressed gene ADAM12, which promotes invasion. Using cellular systems, an animal model, and bioinformatics, we find that a non-canonical but druggable TGF-ß/KAT2A/TAK1 axis controls ADAM12 induction in normal and cancer cells. More broadly, our data show that illegitimate gene expression in cancer is an heterogeneous phenomenon, with a few genes activatable by simple events, and most genes likely requiring a combination of events to become reactivated.


Asunto(s)
Regulación de la Expresión Génica/genética , Neoplasias/genética , Especificidad de Órganos/genética , Transcripción Genética/genética , Proteína ADAM12/genética , Proteína ADAM12/metabolismo , Línea Celular , Línea Celular Tumoral , Cromatina/genética , Cromatina/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Histona Acetiltransferasas/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN , Transducción de Señal/genética , Factor de Crecimiento Transformador beta1/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo
20.
J Bone Miner Res ; 34(5): 911-922, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30615802

RESUMEN

Pamapimod (PAM) is a novel selective p38 mitogen-activated protein (MAP) kinase inhibitor proved to be effective in rheumatoid arthritis in phase 2 clinical trial. However, its effect on osteoclast-associated osteoporosis and the underlying mechanisms remain unclear. In this study, we showed that PAM suppressed receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation via inhibition of p38 phosphorylation and subsequent c-Fos and nuclear factor of activated T cells c1 (NFATc1) expression. In addition, the downregulated NFATc1 leads to reduced expression of its targeting gene disintegrin and metalloproteinase domain-containing protein 12 (ADAM12), which was further proven to be critical for osteoclastic bone resorption. Therefore, we treated ovariectomized (OVX) mice with PAM and revealed a protective effect of PAM on osteoporosis in vivo. In conclusion, our results demonstrated PAM can prevent OVX-induced bone loss through suppression of p38/NFATc1-induced osteoclast formation and NFATc1/ADAM12-associated bone resorption. © 2018 American Society for Bone and Mineral Research.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Osteoclastos/metabolismo , Osteoporosis/tratamiento farmacológico , Piridonas/farmacología , Pirimidinas/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteína ADAM12/metabolismo , Animales , Estrógenos/metabolismo , Femenino , Ratones , Factores de Transcripción NFATC/metabolismo , Osteoclastos/patología , Osteoporosis/metabolismo , Osteoporosis/patología , Ovariectomía , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA