Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Neuro Oncol ; 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38853689

RESUMEN

BACKGROUND: The FDA approval of oncolytic herpes simplex-1 virus (oHSV) therapy underscores its therapeutic promise and safety as a cancer immunotherapy. Despite this promise, the current efficacy of oHSV is significantly limited to a small subset of patients largely due to the resistance in tumor and tumor microenvironment (TME). METHODS: RNA sequencing (RNA-Seq) was used to identify molecular targets of oHSV resistance. Intracranial human and murine glioma or breast cancer brain metastasis (BCBM) tumor-bearing mouse models were employed to elucidate the mechanism underlying oHSV therapy-induced resistance. RESULTS: Transcriptome analysis identified IGF2 as one of the top secreted proteins following oHSV treatment. Moreover, IGF2 expression was significantly upregulated in 10 out of 14 recurrent GBM patients after treatment with oHSV, rQNestin34.5v.2 (71.4%) (p=0.0020) (ClinicalTrials.gov, NCT03152318). Depletion of IGF2 substantially enhanced oHSV-mediated tumor cell killing in vitro and improved survival of mice bearing BCBM tumors in vivo. To mitigate the oHSV-induced IGF2 in the TME, we constructed a novel oHSV, oHSV-D11mt, secreting a modified IGF2R domain 11 (IGF2RD11mt) that acts as IGF2 decoy receptor. Selective blocking of IGF2 by IGF2RD11mt significantly increased cytotoxicity, reduced oHSV-induced neutrophils/PMN-MDSCs infiltration, and reduced secretion of immune suppressive/proangiogenic cytokines, while increased CD8+cytotoxic T lymphocytes (CTLs) infiltration, leading to enhanced survival in GBM or BCBM tumor-bearing mice. CONCLUSION: This is the first study reporting that oHSV-induced secreted IGF2 exerts a critical role in resistance to oHSV therapy, which can be overcome by oHSV-D11mt as a promising therapeutic advance for enhanced viro-immunotherapy.

2.
Cell Death Dis ; 14(8): 546, 2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37612293

RESUMEN

KIAA1324 is a transmembrane protein largely reported as a tumor suppressor and favorable prognosis marker in various cancers, including gastric cancer. In this study, we report the role of N-linked glycosylation in KIAA1324 as a functional post-translational modification (PTM). Loss of N-linked glycosylation eliminated the potential of KIAA1324 to suppress cancer cell proliferation and migration. Furthermore, we demonstrated that KIAA1324 undergoes fucosylation, a modification of the N-glycan mediated by fucosyltransferase, and inhibition of fucosylation also significantly suppressed KIAA1324-induced cell growth inhibition and apoptosis of gastric cancer cells. In addition, KIAA1324-mediated apoptosis and tumor regression were inhibited by the loss of N-linked glycosylation. RNA sequencing (RNAseq) analysis revealed that genes most relevant to the apoptosis and cell cycle arrest pathways were modulated by KIAA1324 with the N-linked glycosylation, and Gene Regulatory Network (GRN) analysis suggested novel targets of KIAA1324 for anti-tumor effects in the transcription level. The N-linked glycosylation blockade decreased protein stability through rapid proteasomal degradation. The non-glycosylated mutant also showed altered localization and lost apoptotic activity that inhibits the interaction between GRP78 and caspase 7. These data demonstrate that N-linked glycosylation of KIAA1324 is essential for the suppressive role of KIAA1324 protein in gastric cancer progression and indicates that KIAA1324 may have anti-tumor effects by targeting cancer-related genes with N-linked glycosylation. In conclusion, our study suggests the PTM of KIAA1324 including N-linked glycosylation and fucosylation is a necessary factor to consider for cancer prognosis and therapy improvement.


Asunto(s)
Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , Glicosilación , Procesamiento Proteico-Postraduccional , Fucosiltransferasas
3.
Cell Death Dis ; 13(4): 374, 2022 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-35440077

RESUMEN

Triple-negative breast cancer (TNBC) is a heterogeneous disease characterized by poor response to standard therapies and therefore unfavorable clinical outcomes. Better understanding of TNBC and new therapeutic strategies are urgently needed. ROR nuclear receptors are multifunctional transcription factors with important roles in circadian pathways and other processes including immunity and tumorigenesis. Nobiletin (NOB) is a natural compound known to display anticancer effects, and our previous studies showed that NOB activates RORs to enhance circadian rhythms and promote physiological fitness in mice. Here, we identified several TNBC cell lines being sensitive to NOB, by itself or in combination. Cell and xenograft experiments showed that NOB significantly inhibited TNBC cell proliferation and motility in vitro and in vivo. ROR loss- and gain-of-function studies showed concordant effects of the NOB-ROR axis on MDA-MB-231 cell growth. Mechanistically, we found that NOB activates ROR binding to the ROR response elements (RRE) of the IκBα promoter, and NOB strongly inhibited p65 nuclear translocation. Consistent with transcriptomic analysis indicating cancer and NF-κB signaling as major pathways altered by NOB, p65-inducible expression abolished NOB effects, illustrating a requisite role of NF-κB suppression mediating the anti-TNBC effect of NOB. Finally, in vivo mouse xenograft studies showed that NOB enhanced the antitumor efficacy in mammary fat pad implanted TNBC, as a single agent or in combination with the chemotherapy agent Docetaxel. Together, our study highlights an anti-TNBC mechanism of ROR-NOB via suppression of NF-κB signaling, suggesting novel preventive and chemotherapeutic strategies against this devastating disease.


Asunto(s)
Flavonas , Neoplasias de la Mama Triple Negativas , Animales , Línea Celular Tumoral , Proliferación Celular , Flavonas/farmacología , Flavonas/uso terapéutico , Humanos , Quinasa I-kappa B/metabolismo , Ratones , FN-kappa B/metabolismo , Transducción de Señal , Neoplasias de la Mama Triple Negativas/patología , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Clin Cancer Res ; 28(7): 1460-1473, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35022322

RESUMEN

PURPOSE: Oncolytic herpes simplex virus-1 (oHSV) infection of brain tumors activates NOTCH, however the consequences of NOTCH on oHSV-induced immunotherapy is largely unknown. Here we evaluated the impact of NOTCH blockade on virus-induced immunotherapy. EXPERIMENTAL DESIGN: RNA sequencing (RNA-seq), TCGA data analysis, flow cytometry, Luminex- and ELISA-based assays, brain tumor animal models, and serum analysis of patients with recurrent glioblastoma (GBM) treated with oHSV was used to evaluate the effect of NOTCH signaling on virus-induced immunotherapy. RESULTS: TCGA data analysis of patients with grade IV glioma and oHSV treatment of experimental brain tumors in mice showed that NOTCH signaling significantly correlated with a higher myeloid cell infiltration. Immunofluorescence staining and RNA-seq uncovered a significant induction of Jag1 (NOTCH ligand) expression in infiltrating myeloid cells upon oHSV infection. Jag1-expressing macrophages further spread NOTCH activation in the tumor microenvironment (TME). NOTCH-activated macrophages increased the secretion of CCL2, which further amplified myeloid-derived suppressor cells. CCL2 and IL10 induction was also observed in serum of patients with recurrent GBM treated with oHSV (rQnestin34.5; NCT03152318). Pharmacologic blockade of NOTCH signaling rescued the oHSV-induced immunosuppressive TME and activated a CD8-dependent antitumor memory response, resulting in a therapeutic benefit. CONCLUSIONS: NOTCH-induced immunosuppressive myeloid cell recruitment limited antitumor immunity. Translationally, these findings support the use of NOTCH inhibition in conjunction with oHSV therapy.


Asunto(s)
Glioblastoma , Células Supresoras de Origen Mieloide , Viroterapia Oncolítica , Virus Oncolíticos , Animales , Línea Celular Tumoral , Glioblastoma/patología , Humanos , Inmunoterapia , Ratones , Células Supresoras de Origen Mieloide/metabolismo , Recurrencia Local de Neoplasia/terapia , Viroterapia Oncolítica/métodos , Virus Oncolíticos/genética , Simplexvirus , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Clin Transl Gastroenterol ; 12(11): e00431, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34797250

RESUMEN

INTRODUCTION: Chronic pancreatitis is associated with an increased risk of developing pancreatic cancer, and patients with inherited forms of pancreatitis are at greatest risk. We investigated whether clinical severity of pancreatitis could also be an indicator of cancer risk independent of etiology by performing targeted DNA sequencing to assess the mutational burden in 55 cancer-associated genes. METHODS: Using picodroplet digital polymerase chain reaction and next-generation sequencing, we reported the genomic profiles of pancreases from severe clinical cases of chronic pancreatitis that necessitated palliative total pancreatectomy with islet autotransplantation. RESULTS: We assessed 57 tissue samples from 39 patients with genetic and idiopathic etiologies and found that despite the clinical severity of disease, there was no corresponding increase in mutational burden. The average allele frequency of somatic variants was 1.19% (range 1.00%-5.97%), and distinct regions from the same patient displayed genomic heterogeneity, suggesting that these variants are subclonal. Few oncogenic KRAS mutations were discovered (7% of all samples), although we detected evidence of frequent cancer-related variants in other genes such as TP53, CDKN2A, and SMAD4. Of note, tissue samples with oncogenic KRAS mutations and samples from patients with PRSS1 mutations harbored an increased total number of somatic variants, suggesting that these patients may have increased genomic instability and could be at an increased risk of developing pancreatic cancer. DISCUSSION: Overall, we showed that even in those patients with chronic pancreatitis severe enough to warrant total pancreatectomy with islet autotransplantation, pancreatic cancer-related mutational burden is not appreciably increased.


Asunto(s)
Carcinoma Ductal Pancreático/genética , Mutación , Neoplasias Pancreáticas/genética , Pancreatitis Crónica/genética , Adulto , Edad de Inicio , Niño , Femenino , Humanos , Trasplante de Islotes Pancreáticos , Masculino , Pancreatectomía , Pancreatitis Crónica/complicaciones , Pancreatitis Crónica/cirugía , Gravedad del Paciente , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas p21(ras)/genética , Tripsina/genética
6.
Sci Rep ; 11(1): 9219, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33911148

RESUMEN

Tumor suppressive microRNAs (miRNAs) are increasingly implicated in the development of anti-tumor therapy by reprogramming gene network that are aberrantly regulated in cancer cells. This study aimed to determine the therapeutic potential of putative tumor suppressive miRNA, miR-138, against glioblastoma (GBM). Whole transcriptome and miRNA expression profiling analyses on human GBM patient tissues identified miR-138 as one of the significantly downregulated miRNAs with an inverse correlation with CD44 expression. Transient overexpression of miR-138 in GBM cells inhibited cell proliferation, cell cycle, migration, and wound healing capability. We unveiled that miR-138 negatively regulates the expression of CD44 by directly binding to the 3' UTR of CD44. CD44 inhibition by miR-138 resulted in an inhibition of glioblastoma cell proliferation in vitro through cell cycle arrest as evidenced by a significant induction of p27 and its translocation into nucleus. Ectopic expression of miR-138 also increased survival rates in mice that had an intracranial xenograft tumor derived from human patient-derived primary GBM cells. In conclusion, we demonstrated a therapeutic potential of tumor suppressive miR-138 through direct downregulation of CD44 for the treatment of primary GBM.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/patología , Regulación Neoplásica de la Expresión Génica , Glioblastoma/patología , Receptores de Hialuranos/metabolismo , MicroARNs/genética , Animales , Apoptosis , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Ciclo Celular , Movimiento Celular , Proliferación Celular , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Receptores de Hialuranos/genética , Ratones , Ratones Endogámicos NOD , Ratones SCID , Pronóstico , Tasa de Supervivencia , Transcriptoma , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancers (Basel) ; 12(4)2020 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-32340193

RESUMEN

Vascularization is a common pathology for many solid tumors, and therefore anti-angiogenic strategies are being investigated as a therapeutic target for treatment. Numerous studies are also being conducted regarding the effects of oncolytic viruses, including ImlygicTM, an FDA approved oncolytic herpes simplex virus-1 (oHSV) for the treatment of highly vascularized tumors such as Kaposi sarcoma (NCT04065152), and brain tumors. To our knowledge, the effects of combining oncolytic HSV with angiogenesis inhibition on endothelial cell activation has not been previously described. Here, we tested the effects of Rapid Antiangiogenesis Mediated By Oncolytic Virus (RAMBO), an oHSV which expresses a potent anti-angiogenic gene Vasculostatin on endothelial cell activation in heavily vascularized solid tumors. oHSV treatment induces endothelial cell activation, which inhibits virus propagation and oncolysis in adjacent tumor cells in vitro. Consistently, this was also observed in intravital imaging of intracranial tumor-bearing mice in vivo where infected tumor endothelial cells could efficiently clear the virus without cell lysis. Quantitative real-time PCR (Q-PCR), leukocyte adhesion assay, and fluorescent microscopy imaging data, however, revealed that RAMBO virus significantly decreased expression of endothelial cell activation markers and leukocyte adhesion, which in turn increased virus replication and cytotoxicity in endothelial cells. In vivo RAMBO treatment of subcutaneously implanted sarcoma tumors significantly reduced tumor growth in mice bearing sarcoma compared to rHSVQ. In addition, histological analysis of RAMBO-treated tumor tissues revealed large areas of necrosis and a statistically significant reduction in microvessel density (MVD). This study provides strong preclinical evidence of the therapeutic benefit for the use of RAMBO virus as a treatment option for highly vascularized tumors.

8.
Sci Rep ; 10(1): 2935, 2020 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32076068

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive malignancies. TGF-ß is strongly expressed in both the epithelial and stromal compartments of PDAC, and dysregulation of TGF-ß signalling is a frequent molecular disturbance in PDAC progression and metastasis. In this study, we investigated whether blockade of TGF-ß signalling synergizes with nal-IRI/5-FU/LV, a chemotherapy regimen for malignant pancreatic cancer, in an orthotopic pancreatic tumour mouse model. Compared to nal-IRI/5-FU/LV treatment, combining nal-IRI/5-FU/LV with vactosertib, a TGF-ß signalling inhibitor, significantly improved long-term survival rates and effectively suppressed invasion to surrounding tissues. Through RNA-sequencing analysis, we identified that the combination treatment results in robust abrogation of tumour-promoting gene signatures and positive enrichment of tumour-suppressing and apoptotic gene signatures. Particularly, the expression of tumour-suppressing gene Ccdc80 was induced by vactosertib and further induced by vactosertib in combination with nal-IRI/5-FU/LV. Ectopic expression of CCDC80 suppressed migration and colony formation concomitant with decreased expression of epithelial-to-mesenchymal transition (EMT) markers in pancreatic cancer cells. Collectively, these results indicate that combination treatment of vactosertib with nal-IRI/5-FU/LV improves overall survival rates in a mouse model of pancreatic cancer by suppressing invasion through CCDC80. Therefore, combination therapy of nal-IRI/5-FU/LV with vactosertib could provide clinical benefits to pancreatic cancer patients.


Asunto(s)
Fluorouracilo/uso terapéutico , Irinotecán/uso terapéutico , Leucovorina/uso terapéutico , Nanopartículas/química , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Compuestos de Anilina/farmacología , Compuestos de Anilina/uso terapéutico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Carcinogénesis/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fluorouracilo/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Irinotecán/farmacología , Leucovorina/farmacología , Liposomas , Ratones Endogámicos C57BL , Invasividad Neoplásica , Neoplasias Pancreáticas/genética , Análisis de Supervivencia , Transcriptoma/genética , Triazoles/farmacología , Triazoles/uso terapéutico , Ensayo de Tumor de Célula Madre , Regulación hacia Arriba/efectos de los fármacos
10.
J Cancer Prev ; 23(1): 1-9, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29629343

RESUMEN

BACKGROUND: Smad3 linker phosphorylation plays essential roles in tumor progression and metastasis. We have previously reported that the mutation of Smad3 linker phosphorylation sites (Smad3-Erk/Pro-directed kinase site mutant constructs [EPSM]) markedly reduced the tumor progression while increasing the lung metastasis in breast cancer. METHODS: We performed high-throughput RNA-Sequencing of the human prostate cancer cell lines infected with adenoviral Smad3-EPSM to identify the genes regulated by Smad3-EPSM. RESULTS: In this study, we identified genes which are differentially regulated in the presence of Smad3-EPSM. We first confirmed that Smad3-EPSM strongly enhanced a capability of cell motility and invasiveness as well as the expression of epithelial-mesenchymal transition marker genes, CDH2, SNAI1, and ZEB1 in response to TGF-ß1 in human pancreatic and prostate cancer cell lines. We identified GADD45B, CTGF, and JUNB genes in the expression profiles associated with cell motility and invasiveness induced by the Smad3-EPSM. CONCLUSIONS: These results suggested that inhibition of Smad3 linker phosphorylation may enhance cell motility and invasiveness by inducing expression of GADD45B, CTGF, and JUNB genes in various cancers.

11.
Biochem Biophys Res Commun ; 494(3-4): 706-713, 2017 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-29097203

RESUMEN

Smad3 linker phosphorylation is a candidate target for several kinases that play important roles in cancer cell initiation, proliferation and progression. Also, Smad3 is an essential intracellular mediator of TGF-ß1-induced transcriptional responses during carcinogenesis. Therefore, it is highly advantageous to identify and develop inhibitors targeting Smad3 linker phosphorylation for the treatment of cancers. Galangin (3,5,7-trihydroxyflavone) has been known to be an active flavonoid showing a cytotoxic effect on several cancer cells. However, the mechanism of action of galangin in various cancers remains unclear, and there has been no report concerning regulation of Smad3 phosphorylation by galangin. In the present study, we show that galangin significantly induced apoptosis and inhibited cell proliferation in the presence of TGF-ß1 in both human prostate and pancreatic cancer cell lines. Particularly, galangin effectively inhibits phosphorylation of the Thr-179 site at Smad3 linker region through suppression of CDK4 phosphorylation. Thus, galangin can be a promising candidate as a selective inhibitor to suppress phosphorylation of Smad3 linker region.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Flavonoides/administración & dosificación , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Antineoplásicos/administración & dosificación , Apoptosis/efectos de los fármacos , Sitios de Unión , Línea Celular Tumoral , Humanos , Neoplasias/patología , Fosforilación/efectos de los fármacos , Unión Proteica , Treonina/metabolismo , Resultado del Tratamiento
12.
Mol Cell Biol ; 35(16): 2841-50, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26055326

RESUMEN

Epidermal growth factor (EGF) regulates various cellular events, including proliferation, differentiation, migration, and tumorigenesis. For the maintenance of homeostasis, EGF signaling should be tightly regulated to prevent the aberrant activation. Smad7 has been known as inhibitory Smad that blocks the signal transduction of transforming growth factor ß. In the process of cell proliferation or transformation, Smad7 has been shown the opposite activities as a promoter or suppressor depending on cell types or microenvironments. We found that the overexpression of Smad7 in human HaCaT keratinocyte cells and mouse skin tissues elevated EGF receptor (EGFR) activity by impairing ligand-induced ubiquitination and degradation of activated receptor, which is induced by the E3 ubiquitin ligase c-Cbl. The C-terminal MH2 region but not MH1 region of Smad7 is critical for interaction with c-Cbl to inhibit the ubiquitination of EGFR. Interestingly, wild-type Smad7, but not Smad6 or mutant Smad7, destabilized the EGF-induced complex formation of c-Cbl and EGFR. These data suggest a novel role for Smad7 as a promoter for prolonging the EGFR signal in keratinocyte and skin tissue by reducing its ligand-induced ubiquitination and degradation.


Asunto(s)
Receptores ErbB/metabolismo , Queratinocitos/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteína smad7/metabolismo , Ubiquitinación , Animales , Sitios de Unión , Línea Celular , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutación , Estructura Terciaria de Proteína , Interferencia de ARN , ARN Interferente Pequeño/genética , Transducción de Señal , Piel/citología , Piel/metabolismo , Proteína smad7/química , Proteína smad7/genética , Regulación hacia Arriba
13.
Cancer Res ; 75(15): 3087-97, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26045166

RESUMEN

Recent advances in genome and transcriptome analysis have contributed to the identification of many potential cancer-related genes. Furthermore, biological and clinical investigations of the candidate genes provide us with a better understanding of carcinogenesis and development of cancer treatment. Here, we report a novel role of KIAA1324 as a tumor suppressor in gastric cancer. We observed that KIAA1324 was downregulated in most gastric cancers from transcriptome sequencing data and found that histone deacetylase was involved in the suppression of KIAA1324. Low KIAA1324 levels were associated with poor prognosis in gastric cancer patients. In the xenograft model, KIAA1324 significantly reduced tumor formation of gastric cancer cells and decreased development of preformed tumors. KIAA1324 also suppressed proliferation, invasion, and drug resistance and induced apoptosis in gastric cancer cells. Through protein interaction analysis, we identified GRP78 (glucose-regulated protein 78 kDa) as a KIAA1324-binding partner. KIAA1324 blocked oncogenic activities of GRP78 by inhibiting GRP78-caspase-7 interaction and suppressing GRP78-mediated AKT activation, thereby inducing apoptosis. In conclusion, our study reveals a tumor suppressive role of KIAA1324 via inhibition of GRP78 oncoprotein activities and provides new insight into the diagnosis and treatment of gastric cancer.


Asunto(s)
Proteínas de Choque Térmico/metabolismo , Proteínas de Neoplasias/metabolismo , Neoplasias Gástricas/patología , Anciano , Animales , Apoptosis/genética , Estudios de Casos y Controles , Línea Celular Tumoral , Chaperón BiP del Retículo Endoplásmico , Regulación Neoplásica de la Expresión Génica , Proteínas de Choque Térmico/genética , Humanos , Estimación de Kaplan-Meier , Proteínas de la Membrana , Ratones , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Estructura Terciaria de Proteína , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidad , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cell Mol Life Sci ; 72(3): 583-596, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25063542

RESUMEN

Genomic instability is one of the representative causes in genetic disorder, where the proper cellular response to DNA damage is essential in maintaining genomic stability. ATM and the Mre11-Rad50-Nbs1 (MRN) complex play critical roles in the cellular response to DNA damage such as DNA double-strand break (DSB). In this study, we report that Smad7 is indispensible in DNA damage response as a novel component of MRN complex. Smad7 enhances cell survival against DNA damage by accelerating ATM dependent DNA repair signaling. In Smad7-deficient mouse embryonic fibroblast cells, the loss of Smad7 decreases ATM activation and inhibits recruitment of ATM to the sites of DSBs. Smad7 interacts with Nbs1, a member of MRN complex, and enhances the interaction between ATM and Nbs1 upon DNA damage response, leading to phosphorylation of downstream substrates. Ectopic expression of Smad7 in the skin of mice enhances the phosphorylation of ATM upon X-irradiation. We found that effect of Smad7 on enhancing DNA repair is independent of its inhibitory activity of TGF-ß signaling. Taken together, our results highlight a critical function of Smad7 in DSB response and establish the novel mechanism in which Smad7 facilitates the recruitment of ATM to the MRN complex through direct interaction with Nbs1.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Inestabilidad Genómica/fisiología , Complejos Multiproteicos/metabolismo , Proteína smad7/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Ácido Anhídrido Hidrolasas , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Western Blotting , Proteínas de Ciclo Celular/metabolismo , Ensayo Cometa , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Fibroblastos , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Inmunoprecipitación , Proteína Homóloga de MRE11 , Ratones , Proteínas Nucleares/metabolismo , Fosforilación , Piel/metabolismo , Proteína smad7/deficiencia
15.
Genes Chromosomes Cancer ; 54(3): 142-55, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25521327

RESUMEN

Homozygous deletion is a frequent mutational mechanism of silencing tumor suppressor genes in cancer. Therefore, homozygous deletions have been analyzed for identification of tumor suppressor genes that can be utilized as biomarkers or therapeutic targets for cancer treatment. In this study, to elucidate potential tumor suppressor genes involved in gastric cancer (GC), we analyzed the entire set of large homozygous deletions in six human GC cell lines through genome- and transcriptome-wide approaches. We identified 51 genes in homozygous deletion regions of chromosomes and confirmed the deletion frequency in tumor tissues of 219 GC patients from The Cancer Genome Atlas database. We evaluated the effect of homozygous deletions on the mRNA level and found significantly affected genes in chromosome bands 9p21, 3p22, 5p14, and 6q15. Among the genes in 9p21, we investigated the potential tumor suppressive effect of KLHL9. We demonstrated that ectopic expression of KLHL9 inhibited cell proliferation and tumor formation in KLHL9-deficient SNU-16 cell line. In addition, we observed that homozygous focal deletions generated truncated transcripts of TGFBR2, CTNNA1, and STXBP5. Ectopic expression of two kinds of TGFBR2-reverse GADL1 fusion genes suppressed TGF-ß signaling, which may lead to the loss of sensitivity to TGF-ß tumor suppressive activity. In conclusion, our findings suggest that novel tumor suppressor genes that are aberrantly expressed through homozygous deletions may play important roles in gastric tumorigenesis.


Asunto(s)
Deleción Cromosómica , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Neoplasias Gástricas/genética , Animales , Línea Celular Tumoral , Cromosomas Humanos Par 3 , Cromosomas Humanos Par 5 , Cromosomas Humanos Par 6 , Cromosomas Humanos Par 9 , Femenino , Humanos , Ratones , Ratones Desnudos , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética
16.
J Cell Sci ; 127(Pt 2): 411-21, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24259667

RESUMEN

In most human cancers the Myc proto-oncogene is highly activated. Dysregulation of Myc oncoprotein contributes to tumorigenesis in numerous tissues and organs. Thus, targeting Myc stability could be a crucial step for cancer therapy. Here we report Smad7 as a key molecule regulating Myc stability and activity by recruiting the F-box protein, Skp2. Ectopic expression of Smad7 downregulated the protein level of Myc without affecting the transcription level, and significantly repressed its transcriptional activity, leading to inhibition of cell proliferation and tumorigenic activity. Furthermore, Smad7 enhanced ubiquitylation of Myc through direct interaction with Myc and recruitment of Skp2. Ablation of Smad7 resulted in less sensitivity to the growth inhibitory effect of TGF-ß by inducing stable Myc expression. In conclusion, these findings that Smad7 functions in Myc oncoprotein degradation and enhances the cytostatic effect of TGF-ß signaling provide a possible new therapeutic approach for cancer treatment.


Asunto(s)
Citostáticos/farmacología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Proteínas Inhibidoras de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica/efectos de los fármacos , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Estabilidad Proteica/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proto-Oncogenes Mas , Transcripción Genética/efectos de los fármacos , Ubiquitina/metabolismo
17.
J Mol Med (Berl) ; 91(11): 1273-84, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24072041

RESUMEN

UNLABELLED: Renal fibrosis is a common consequence of unilateral ureteral obstruction, which provides a useful model to investigate the pathogenesis of obstructive nephropathy and progressive renal fibrosis. Transforming growth factor (TGF-ß1) has been recognized as a key mediator in renal fibrosis by stimulating matrix-producing fibrogenic cells and promoting extracellular matrix deposition. Therefore, considerable efforts have been made to regulate TGF-ß signaling for antifibrotic therapy. Here, we investigated the mode of action of glucosamine hydrochloride (GS-HCl) on TGF-ß1-induced renal fibrosis. In the obstructed kidneys and TGF-ß1-treated renal cells, GS-HCl significantly decreased renal expression of α-smooth muscle actin, collagen I, and fibronectin. By investigating the inhibitory mechanism of GS-HCl on renal fibrosis, we found that GS-HCl suppressed TGF-ß signaling by inhibiting N-linked glycosylation of the type II TGF-ß receptor (TßRII), leading to an inefficient trafficking of TßRII to the membrane surface. Defective N-glycosylation of TßRII further suppressed the TGF-ß1-binding to TßRII, thereby decreasing TGF-ß signaling. Notably, GS-HCl treatment significantly reduced TGF-ß1-induced up-regulation of Smad2/3 phosphorylation and transcriptional activity in vivo and in vitro. Taken together, GS-HCl-mediated regulation of TGF-ß signaling exerted an antifibrotic effect, thereby ameliorating renal fibrosis. Our study suggests that GS-HCl would be a promising agent for therapeutic intervention for preventing TGF-ß1-induced renal fibrosis in kidney diseases. KEY MESSAGE: Glucosamine-mediated attenuation of TGF-ß signaling ameliorates renal fibrosis in vivo TGF-ß1-induced fibrogenic action is reduced by glucosamine in vitro N-glycosylation of the type II TGF-ß receptor is suppressed by glucosamine Glucosamine-induced defective N-glycosylation of TßRII decreases TGF-ß signaling.


Asunto(s)
Fibrosis/etiología , Fibrosis/prevención & control , Glucosamina/uso terapéutico , Enfermedades Renales/etiología , Enfermedades Renales/prevención & control , Riñón/patología , Obstrucción Ureteral/complicaciones , Animales , Línea Celular , Fibrosis/patología , Glicosilación/efectos de los fármacos , Humanos , Riñón/efectos de los fármacos , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
18.
Cell Transplant ; 21(1): 299-312, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-21669031

RESUMEN

Bone marrow mononuclear cells (BMMNCs) can be used to treat patients with myocardial infarction, since BMMNCs can differentiate in vitro toward cardiomyogenic lineages when treated with transforming growth factor-ß1 (TGF-ß1). However, the in vitro cardiomyogenic differentiation culture process is costly and laborious, and the patients should wait during the culture period. In this study, we hypothesize that BMMNCs implanted in cardiomyogenically undifferentiated state to myocardial infarction site would differentiate cardiomyogenically in situ when exogenous TGF-ß1 is delivered to the cell implantation site. Heparin-conjugated poly(lactic-co-glycolic acid) nanospheres (HCPNs) suspended in fibrin gel were used as a TGF-ß1 delivery system. BMMNCs were labeled with a green fluorescent dye (PKH67) and implanted into the infarction border zone of rat myocardium using fibrin gel containing HCPNs and TGF-ß1. BMMNC implantation using fibrin gel and HCPNs without TGF-ß1 served as a control. Four weeks after implantation, the expression of cardiomyogenic marker proteins by the implanted BMMNCs was dramatically greater in the TGF-ß1 delivery group than in the control group. This method can significantly improve the stem cell therapy technology for myocardial regeneration, since it can remove in vitro cell culture step for cardiomyogenic differentiation prior to cell implantation.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Infarto del Miocardio/terapia , Miocitos Cardíacos/citología , Factor de Crecimiento Transformador beta1/farmacología , Animales , Células de la Médula Ósea/metabolismo , Diferenciación Celular , Células Cultivadas , Sistemas de Liberación de Medicamentos , Fibrina , Leucocitos Mononucleares , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley
19.
J Biomater Sci Polym Ed ; 21(5): 635-45, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20338097

RESUMEN

Previously, we developed an apatite-coated non-porous poly(lactic-co-glycolic acid) (PLGA) microsphere (ANPM) as an injectable bone substitute. We hypothesized that an apatite-coated porous PLGA microsphere (APPM) would have enhanced osteogenic potential compared to that of an ANPM. To test the hypothesis, critical-sized bone defects were made in mouse calvaria, and APPMs and ANPMs were implanted in the defects for 8 weeks. New bone formed around both types of bone substitutes implanted in mouse calvarial defects. Importantly, the portion of bone-like tissue area in the implant cross-sectional area was significantly higher in the APPM group than in the ANPM group (36.9% versus 14.6%, P < 0.001). Fluorochrome-labeling analysis showed that bone regeneration occurred in the pores of implanted APPMs. The results show that APPM may be useful as a bone substitute in orthopedic applications.


Asunto(s)
Apatitas/química , Sustitutos de Huesos , Durapatita/uso terapéutico , Glicolatos/química , Microesferas , Animales , Regeneración Ósea , Sustitutos de Huesos/química , Sustitutos de Huesos/metabolismo , Sustitutos de Huesos/uso terapéutico , Durapatita/química , Inyecciones/métodos , Ácido Láctico , Masculino , Ratones , Ratones Endogámicos ICR , Ácido Poliglicólico , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , Porosidad , Cráneo/patología , Cráneo/cirugía
20.
J Biomed Mater Res A ; 93(2): 666-72, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-19609878

RESUMEN

We hypothesized that bone morphogenetic protein-2 (BMP-2) would significantly enhance in vivo bone formation efficacy of osteogenically undifferentiated human cord blood mesenchymal stem cells (hCBMSCs). To test this hypothesis, poly(lactic-co-glycolic acid)/hydroxyapatite (PLGA/HA) scaffolds (group 1), BMP-2-loaded PLGA/HA scaffolds (group 2), undifferentiated hCBMSCs seeded on PLGA/HA scaffolds (group 3), undifferentiated hCBMSCs seeded on BMP-2-loaded PLGA/HA scaffolds (group 4), and osteogenically differentiated hCBMSCs seeded on PLGA/HA scaffolds (group 5) were implanted into dorsal, subcutaneous spaces of athymic mice for 8 weeks. Histological analysis showed that group 4 exhibited the largest bone formation area. RT-PCR analysis showed that human mRNA expression of osteoblastic markers such as ALP and osteocalcin in group 4 was higher than that of the other groups. Mouse osteoblastic markers of the host cells in the implants were also expressed more in group 4 than in the other groups. This study demonstrated that hCBMSCs that were not differentiated osteogenically in vitro prior to transplantation regenerate bone negligibly in vivo and that transplantation of osteogenically undifferentiated hCBMSCs with BMP-2 delivery results in much more extensive bone formation in vivo than that of undifferentiated or osteogenically differentiated hCBMSCs.


Asunto(s)
Regeneración Ósea/fisiología , Diferenciación Celular/fisiología , Sangre Fetal/citología , Células Madre Mesenquimatosas/fisiología , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Durapatita/química , Durapatita/metabolismo , Femenino , Humanos , Implantes Experimentales , Ácido Láctico/química , Ácido Láctico/metabolismo , Ensayo de Materiales , Células Madre Mesenquimatosas/citología , Ratones , Ratones Desnudos , Osteoblastos/metabolismo , Osteogénesis/fisiología , Ácido Poliglicólico/química , Ácido Poliglicólico/metabolismo , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , ARN Mensajero/genética , ARN Mensajero/metabolismo , Propiedades de Superficie , Andamios del Tejido/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...