Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Life Sci ; 293: 120353, 2022 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-35074406

RESUMEN

AIMS: Piwi-like RNA-mediated gene silencing 4 (PIWIL4) or HIWI2, are seen deregulated in human cancers and possibly play critical roles in tumorigenesis. It is unknown what role HIWI2 plays in the regulation of fibrosarcoma, an early metastatic lethal type of soft tissue sarcoma (STS). The present study aimed to investigate the role of HIWI2 in the tumorigenesis of fibrosarcoma. MAIN METHODS: The expression of HIWI2 in HT1080 fibrosarcoma cells was determined by qRT-PCR and western blotting. The MTT assay, colony formation assay, cell cycle, and PE-AnnexinV/7AAD apoptosis assay using flow cytometry, DNA laddering assay, comet assay, and γH2AX accumulation assay were performed to study the effect of HIWI2 overexpression in HT1080 cells. Further, the effect of silencing of HIWI2 was determined by cell viability assay, transwell migration, and invasion assay. KEY FINDINGS: HIWI2 is under-expressed in STS cell lines and tissues, which is associated with poor disease-free survival, disease-specific survival, and progression-free survival of the patients. Overexpression of HIWI2 in HT1080 cells causes DNA damage by increasing intracellular ROS by inhibiting the expression of antioxidant genes (SOD1, SOD2, GPX1, GPX4, and CAT). Furthermore, an increase in H2AX phosphorylation was observed, which activates p53 that promotes p21 expression and caspase-3 activation, leading to G2/M phase cell cycle arrest and apoptosis. HIWI2 silencing, on the contrary, promotes cell growth, migration, and invasion by activating MMP2 and MMP9. SIGNIFICANCE: These results are the first to show that HIWI2 acts as a tumor suppressor in fibrosarcoma by modulating the ROS/DNA damage/p53 pathway.


Asunto(s)
Daño del ADN/fisiología , Fibrosarcoma/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Proteínas de Unión al ARN/biosíntesis , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/fisiología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/fisiología , Fibrosarcoma/patología , Humanos , Proteínas de Unión al ARN/genética , Proteína p53 Supresora de Tumor/antagonistas & inhibidores
2.
Mol Biol Cell ; 32(20): br2, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34260268

RESUMEN

Heterotrimeric G proteins (αßγ) function at the cytoplasmic surface of a cell's plasma membrane to transduce extracellular signals into cellular responses. However, numerous studies indicate that G proteins also play noncanonical roles at unique intracellular locations. Previous work has established that G protein ßγ subunits (Gßγ) regulate a signaling pathway on the cytoplasmic surface of Golgi membranes that controls the exit of select protein cargo. Now, we demonstrate a novel role for Gßγ in regulating mitotic Golgi fragmentation, a key checkpoint of the cell cycle that occurs in the late G2 phase. We show that small interfering RNA-mediated depletion of Gß1 and Gß2 in synchronized cells causes a decrease in the number of cells with fragmented Golgi in late G2 and a delay of entry into mitosis and progression through G2/M. We also demonstrate that during G2/M Gßγ acts upstream of protein kinase D and regulates the phosphorylation of the Golgi structural protein GRASP55. Expression of Golgi-targeted GRK2ct, a Gßγ-sequestering protein used to inhibit Gßγ signaling, also causes a decrease in Golgi fragmentation and a delay in mitotic progression. These results highlight a novel role for Gßγ in regulation of Golgi structure.


Asunto(s)
Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Subunidades beta de la Proteína de Unión al GTP/metabolismo , Subunidades gamma de la Proteína de Unión al GTP/metabolismo , Aparato de Golgi/fisiología , Ciclo Celular/fisiología , Membrana Celular/metabolismo , Fase G2/fisiología , Aparato de Golgi/metabolismo , Proteínas de la Matriz de Golgi/metabolismo , Células HeLa , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Mitosis/fisiología , Fosforilación , Proteína Quinasa C/metabolismo , Transporte de Proteínas/fisiología , ARN Interferente Pequeño/metabolismo , Transducción de Señal/fisiología
3.
Int J Radiat Oncol Biol Phys ; 111(1): 260-271, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34112559

RESUMEN

PURPOSE: Autophagy and cell-cycle checkpoints act in concert to confer cellular radioresistance. We investigated the functional interaction between radiation-induced autophagy and G2 checkpoint activation in highly radioresistant human pancreatic ductal adenocarcinoma (PDAC) cells. METHODS AND MATERIALS: Four human PDAC cell lines (MIA PaCa-2, KP-4, Panc-1, and SUIT-2) were analyzed. These cells were first irradiated using x-rays, and their cell cycle status, autophagy, and cell cycle checkpoint marker expression and ATP production levels were evaluated. Autophagic flux assays and siRNA knockdown were used to evaluate autophagy activity. Double thymidine block experiments were performed to synchronize the cells. Two inhibitors (MK-1775 and SCH 900776) were used to attenuate G2 checkpoint activation. Cell survival assays and animal experiments were performed to evaluate the radiosensitizing effects of the G2 checkpoint inhibitors. RESULTS: Autophagy and G2/M accumulation were synchronously induced in human PDAC cells with an activated G2 checkpoint at 12 hours after x-ray irradiation of 6 Gy. Radiation-induced autophagy produced the ATP levels required for cell survival. Double thymidine block experiments revealed that no autophagy occurred in cells that were solely in G2 phase. MK-1775 or SCH 900776 exposure attenuated not only G2 checkpoint activation but also postirradiation autophagy, indicating the dependence of radiation-induced autophagy on an activated G2 checkpoint. The inhibitors demonstrated a higher radiosensitizing effect in the PDAC cells than the autophagy inhibitor chloroquine. MK-1775 in combination with x-rays significantly suppressed the tumor growth of MIA PaCa-2 xenografts compared with other treatment groups, including radiation or drug exposure alone, to enhance the radiosensitivity of PDAC cells in vivo. CONCLUSIONS: Biological crosstalk exists between the G2 checkpoint activation and radiation-induced autophagy processes that are believed to independently contribute to the radioresistance of human PDAC cells. These findings have important implications for the development of future radiation therapy strategies for PDAC.


Asunto(s)
Autofagia/efectos de la radiación , Carcinoma Ductal Pancreático/radioterapia , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Neoplasias Pancreáticas/radioterapia , Tolerancia a Radiación , Adenosina Trifosfato/biosíntesis , Animales , Línea Celular Tumoral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Pirazoles/farmacología , Pirimidinonas/farmacología , Quinolinas/farmacología , Tiazoles/farmacología
4.
Mol Immunol ; 135: 304-311, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33964631

RESUMEN

The RIP family plays a key role in mediating cell inflammation, oxidative stress and death. Among them, RIPK1, as an important regulatory factor in the upstream of the NF-κB pathway, is involved in multiple pathways of cell inflammation and death. Epidermal cells constitute the outermost barrier of the human body. Radiation can induce epidermal cell death, inflammation and oxidative stress to cause damage. Therefore, this paper selected HaCaT cell and used CRISPR/Cas technology to construct a cell model of stable knockout of RIPK1 gene, to analyze the effect and regulation of RIPK1 knockout on the function and death of HaCaT cells induced by UVB or TNF-α. The results showed that knockout of RIPK1 had no significant effect on the morphology of HaCaT cells at rest, but it led to slowing cell proliferation and blocking the G2M phase of cell cycle. Compared with HaCaTWT, HaCaTRIP1KO was abnormally sensitive to TNF-α-induced cell death and apoptosis, and may be associated with inhibition of NF-κB pathway. Knocking out RIPK1 led to a more significant inhibition of cell growth by UVB, and up-regulation of the expression of the inflammatory factor IL-1α. P38 MAPK and NF-κB pathways may be involved this process. This study further found that RIPK1 in epidermal cell has a regulatory function on pro-survival signals.


Asunto(s)
Apoptosis/fisiología , Células Epidérmicas/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Rayos Ultravioleta/efectos adversos , Sistemas CRISPR-Cas/genética , Línea Celular Transformada , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Técnicas de Inactivación de Genes , Células HaCaT , Humanos , Interleucina-1alfa/metabolismo , FN-kappa B/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Transducción de Señal/fisiología , Piel/lesiones , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Am J Pathol ; 191(1): 157-167, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33129760

RESUMEN

Colorectal cancer (CRC) is a leading nonfamilial cause of cancer mortality among men and women. Although various genetic and epigenetic mechanisms have been identified, the full molecular mechanisms deriving CRC tumorigenesis are not fully understood. This study demonstrates that cell adhesion molecule transmembrane and immunoglobulin domain containing 1 (TMIGD1) are highly expressed in mouse and human normal intestinal epithelial cells. TMIGD1 knockout mice were developed, and the loss of TMIGD1 in mice was shown to result in the development of adenomas in small intestine and colon. In addition, the loss of TMIGD1 significantly impaired intestinal epithelium brush border membrane, junctional polarity, and maturation. Mechanistically, TMIGD1 inhibits tumor cell proliferation and cell migration, arrests cell cycle at the G2/M phase, and induces expression of p21CIP1 (cyclin-dependent kinase inhibitor 1), and p27KIP1 (cyclin-dependent kinase inhibitor 1B) expression, key cell cycle inhibitor proteins involved in the regulation of the cell cycle. Moreover, TMIGD1 is shown to be progressively down-regulated in sporadic human CRC, and its downregulation correlates with poor overall survival. The findings herein identify TMIGD1 as a novel tumor suppressor gene and provide new insights into the pathogenesis of colorectal cancer and a novel potential therapeutic target.


Asunto(s)
Puntos de Control del Ciclo Celular/fisiología , Neoplasias del Colon/metabolismo , Glicoproteínas de Membrana/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Transformación Celular Neoplásica/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Genes Supresores de Tumor/fisiología , Humanos , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
6.
Mol Med Rep ; 22(6): 5069-5082, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33174040

RESUMEN

Long non­coding RNAs (lncRNAs) affect atherosclerosis by regulating the physiological and pathological processes of endothelial cells; however, the role of lncRNA WEE2 antisense RNA 1 (WEE2­AS1) in arteriosclerosis obliterans (ASO) is not completely understood. The present study aimed to explore the function of lncRNA WEE2­AS1 in human vascular endothelial cells. The results indicated that lncRNA WEE2­AS1 was significantly elevated in plasma and artery tissue samples of patients with ASO compared with healthy controls. The fluorescence in situ hybridization results suggested that lncRNA WEE2­AS1 was expressed in the cytoplasm and nuclei of primary human umbilical vein endothelial cells (HUVECs). The Cell Counting Kit­8 assay results suggested that lncRNA WEE2­AS1 knockdown significantly promoted HUVEC viability, whereas lncRNA WEE2­AS1 overexpression inhibited HUVEC viability compared with the negative control groups. Furthermore, analysis of the cell cycle by flow cytometry indicated that lncRNA WEE2­AS1 knockdown significantly decreased the proportion of cells in the G0/G1 phase and significantly increased the proportion of cells in the G2/M phase compared with the negative control group. However, lncRNA WEE2­AS1 overexpression had no significant effect on cell cycle distribution compared with the negative control group. The western blotting results indicated that lncRNA WEE2­AS1 knockdown significantly reduced the expression levels of phosphorylated cyclin dependent kinase 1, WEE1 homolog 2 and myelin transcription factor 1, but increased the expression level of cell division cycle 25B compared with the negative control group. lncRNA WEE2­AS1 overexpression displayed the opposite effect on protein expression. Collectively, the present study suggested that lncRNA WEE2­AS1 was significantly upregulated in ASO and may serve a role in regulating human vascular endothelial cell viability. Further investigation into lncRNA WEE2­AS1 may broaden the current understanding of the molecular mechanism underlying ASO, and aid with the identification of specific probes and precise targeted drugs for the diagnosis and treatment of ASO.


Asunto(s)
Arteriosclerosis Obliterante/genética , Proteínas de Ciclo Celular/genética , Células Endoteliales/metabolismo , Proteínas Tirosina Quinasas/genética , Anciano , Arteriosclerosis Obliterante/metabolismo , Proteína Quinasa CDC2/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/fisiología , Supervivencia Celular/fisiología , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Proteínas Tirosina Quinasas/metabolismo , ARN sin Sentido/genética , ARN Largo no Codificante/genética
7.
Eur J Dermatol ; 30(4): 329-337, 2020 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-32969792

RESUMEN

BACKGROUND: The pathogenesis of psoriasis is still not fully understood. Dynamin-related protein 1 (Drp1) regulates mitochondrial fission and is implicated in various inflammatory conditions, but research regarding Drp1 in the skin is scarce. OBJECTIVES: To examine Drp1 expression in psoriasis vulgaris lesional skin and the effect of Drp1 expression on keratinocyte proliferation and inflammatory mediator release in vitro. MATERIALS AND METHODS: Skin biopsies were collected, and the expression of Drp1 and TNF-α was investigated in the skin. Serum TNF-α level was also examined. The effect of Drp1 on keratinocyte proliferation and inflammatory mediator release was evaluated in HaCaT cells following Drp1 transfection. The effect of TNF-α on Drp1 expression was also studied in HaCaT cells. RESULTS: Drp1 expression was significantly increased and positively correlated with PASI score and TNF-α expression in skin. In HaCaT cells, Drp1 transfection altered cellular ATP and proliferation, induced G2/M arrest, and affected inflammatory mediator (TNF-α, IL-6 and CXCL8) release. Moreover, TNF-α induced Drp1 expression and recovered cellular ATP content and mediator release. CONCLUSION: Drp1 is significantly elevated in psoriasis vulgaris skin and positively correlates with disease severity, moreover, in vitro, Drp1 regulates keratinocyte function.


Asunto(s)
Proliferación Celular , Dinaminas/metabolismo , Queratinocitos/fisiología , Psoriasis/metabolismo , Psoriasis/fisiopatología , Piel/metabolismo , Adenosina Trifosfato/metabolismo , División Celular , Dinaminas/genética , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Células HaCaT , Humanos , Mediadores de Inflamación/metabolismo , Dinámicas Mitocondriales , Índice de Severidad de la Enfermedad , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba
8.
Mol Cell Proteomics ; 19(11): 1910-1920, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32868396

RESUMEN

Glutathionylation is an important posttranslational modification that protects proteins from further oxidative damage as well as influencing protein structure and activity. In the present study, we demonstrate that the cysteine-42 residue in protein arginine N-methyltransferase 5 (PRMT5) is glutathionylated in aged mice or in cells that have been exposed to oxidative stress. Deglutathionylation of this protein is catalyzed by glutaredoxin-1 (Grx1). Using mutagenesis and subsequent biochemical analyses, we show that glutathionylation decreased the binding affinity of PRMT5 with methylosome protein-50 (MEP50) and reduced the methyltransferase activity of PRMT5. Furthermore, overexpression of PRMT5-C42A mutant caused a significant increase in histone methylation in HEK293T and A549 cells and promoted cell growth, whereas overexpression of the PRMT5-C42D mutant, a mimic of glutathionylated PRMT5, inhibited cell proliferation. Taken together, our results demonstrate a new mechanism of regulation of PRMT5 methyltransferases activity and suggest that PRMT5 glutathionylation is partly responsible for reactive oxygen species-mediated cell growth inhibition.


Asunto(s)
Envejecimiento/metabolismo , Proliferación Celular/efectos de los fármacos , Glutatión/metabolismo , Peróxido de Hidrógeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular/fisiología , Bases de Datos de Proteínas , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Glutarredoxinas/metabolismo , Células HEK293 , Histonas/metabolismo , Humanos , Riñón/enzimología , Riñón/metabolismo , Metilación , Ratones , Unión Proteica , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Procesamiento Proteico-Postraduccional/fisiología , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína-Arginina N-Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/genética , Proteómica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Regulación hacia Arriba
9.
Life Sci Alliance ; 3(11)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32826290

RESUMEN

Pch2 is a meiosis-specific AAA+ protein that controls several important chromosomal processes. We previously demonstrated that Orc1, a subunit of the ORC, functionally interacts with budding yeast Pch2. The ORC (Orc1-6) AAA+ complex loads the AAA+ MCM helicase to origins of replication, but whether and how ORC collaborates with Pch2 remains unclear. Here, we show that a Pch2 hexamer directly associates with ORC during the meiotic G2/prophase. Biochemical analysis suggests that Pch2 uses its non-enzymatic NH2-terminal domain and AAA+ core and likely engages the interface of ORC that also binds to Cdc6, a factor crucial for ORC-MCM binding. Canonical ORC function requires association with origins, but we show here that despite causing efficient removal of Orc1 from origins, nuclear depletion of Orc2 and Orc5 does not trigger Pch2/Orc1-like meiotic phenotypes. This suggests that the function for Orc1/Pch2 in meiosis can be executed without efficient association of ORC with origins of replication. In conclusion, we uncover distinct functionalities for Orc1/ORC that drive the establishment of a non-canonical, meiosis-specific AAA+ assembly with Pch2.


Asunto(s)
Meiosis/fisiología , Proteínas Nucleares/metabolismo , Complejo de Reconocimiento del Origen/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , ADN Helicasas/genética , Replicación del ADN/genética , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Meiosis/genética , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Complejo de Reconocimiento del Origen/fisiología , Profase/fisiología , Unión Proteica/genética , Origen de Réplica/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/fisiología , Saccharomycetales/genética , Saccharomycetales/metabolismo
10.
Development ; 147(13)2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32620578

RESUMEN

In mammalian growing follicles, oocytes are arrested at the diplotene stage (which resembles the G2/M boundary in mitosis), while the granulosa cells (GCs) continue to proliferate during follicular development, reflecting a cell cycle asynchrony between oocytes and GCs. Hypoxanthine (Hx), a purine present in the follicular fluid, has been shown to induce oocytes meiotic arrest, although its role in GC proliferation remains ill-defined. Here, we demonstrate that Hx indiscriminately prevents G2-to-M phase transition in porcine GCs. However, oocyte-derived paracrine factors (ODPFs), particularly GDF9 and BMP15, maintain the proliferation of GCs, partly by activating the ERK1/2 signaling and enabling the G2/M transition that is suppressed by Hx. Interestingly, GCs with lower expression of GDF9/BMP15 receptors appear to be more sensitive to Hx-induced G2/M arrest and become easily detached from the follicular wall. Importantly, Hx-mediated inhibition of G2/M progression instigates GC apoptosis, which is ameliorated in the presence of GDF9 and/or BMP15. Therefore, our data indicate that the counterbalance of intrafollicular factors, particularly Hx and oocyte-derived GDF9/BMP15, fine-tunes the development of porcine follicles by regulating the cell cycle progression of GCs.


Asunto(s)
Células de la Granulosa/metabolismo , Hipoxantina/metabolismo , Oocitos/metabolismo , Animales , Apoptosis/genética , Apoptosis/fisiología , Proteína Morfogenética Ósea 15/genética , Proteína Morfogenética Ósea 15/metabolismo , Proliferación Celular/genética , Proliferación Celular/fisiología , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Factor 9 de Diferenciación de Crecimiento/genética , Factor 9 de Diferenciación de Crecimiento/metabolismo , Porcinos
11.
Biochem Biophys Res Commun ; 528(3): 586-593, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32505357

RESUMEN

Dual-specificity phosphatase 4 (DUSP4), a MAP kinase phosphatase, has been regarded as a tumor suppressor gene in several cancers. However, high-level expression of DUSP4 is occasionally observed in specific cancers and its functional significance in carcinogenesis is not fully understood. In the present study, we showed that downregulation of DUSP4 suppressed the proliferation of cancer cell lines exhibiting high expression of DUSP4 by inducing apoptosis and cell cycle arrest at G2/M phase. Expression microarray analyses and pathway analyses revealed that downregulation of DUSP4 activated the p53 signaling pathway, and might be involved in cell growth suppression. Aberrant accumulation of p53 and induction of p53 downstream target genes were further investigated. Furthermore, cell growth suppression following downregulation of DUSP4 was markedly attenuated in p53-deleted cells established using the CRISPR/Cas9 system. These findings suggest that constitutive expression of DUSP4 in cancer cells contributes to enhanced proliferation through escape from apoptosis and cell cycle arrest. We propose that DUSP4 could be a novel therapeutic target for cancers overexpressing it.


Asunto(s)
Fosfatasas de Especificidad Dual/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Neoplasias/enzimología , Neoplasias/patología , Apoptosis/genética , Apoptosis/fisiología , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Proliferación Celular/fisiología , Supervivencia Celular/genética , Supervivencia Celular/fisiología , Regulación hacia Abajo , Fosfatasas de Especificidad Dual/genética , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Células HCT116 , Humanos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Neoplasias/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
12.
Cancer Gene Ther ; 27(12): 934-947, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32372016

RESUMEN

UNC5B is a known tumor suppressor gene in a variety of cancers. As a transmembrane protein, UNC5B also induces apoptosis in a P53-dependent manner. In this study, we demonstrate that UNC5B inhibits proliferation through G2/M phase arrest by mass spectrometry and bioinformatics analysis in bladder cancer cells. By combing with CDC14A and P53, UNC5B dephosphorylated P53 at Ser-315 site. This dephosphorylation facilitated G2/M phase arrest by reducing the expression of cyclin B1 and increasing the expression of p-CDK1, thus inhibiting tumor proliferation. Knockdown of CDC14A suppressed the G2/M phase arrest induced by UNC5B in vitro, and eliminated the inhibitory effect of UNC5B on tumor proliferation in vivo. Our results show that UNC5B-mediated cell cycle arrest may act as a potential treatment for bladder cancer.


Asunto(s)
Receptores de Netrina/metabolismo , Proteínas Tirosina Fosfatasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias de la Vejiga Urinaria/metabolismo , División Celular/fisiología , Línea Celular Tumoral , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Humanos , Persona de Mediana Edad , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología
13.
J BUON ; 25(1): 472-478, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32277671

RESUMEN

PURPOSE: Osteosarcoma is one of the rare but fatal malignancies. The high metastatic rate, late diagnosis, emergence of drug resistance against drugs such as doxorubicin, and the lack of therapeutic targets obstructs the treatment of osteosarcoma. This study was undertaken to investigate the role and therapeutic potential of miR-187 in human osteosarcoma cells. METHODS: The WST-1 proliferation assay was used for investigation of cell viability. Transfections were carried out by Lipofectamine 2000 reagent. The qRT-PCR was used for expression analysis. DAPI, acridine orange (AO)/ethidium bromide (EB) and Annexin V/propidium iodide (PI) assay were used for apoptosis. Western blot analysis was used for the determination of protein expression. RESULTS: The expression of miR-187 was significantly downregulated in human osteosarcoma cells. Out of all osteosarcoma cell lines the SAOS-2 showed the lowest expression of miR-187 and therefore this cell line was selected for further studies. Overexpression of miR-187 caused significant inhibition in the proliferation of SAOS-2 osteosarcoma cells. The miR-187-triggered growth inhibition was found to be mainly due to induction of G2/M phase cell cycle arrest of the SAOS-2 cells. The G2/M cell cycle arrest was also accompanied by depletion of Cyclin-B1 expression. Additionally, miR-187 enhanced the chemosensitivity of the osteosarcoma cells to doxorubicin. The wound healing and transwell assay showed that miR-187 overexpression resulted in the suppression of migration and invasion of the SAOS-2 osteosarcoma cells. In silico analysis showed that miR-187 exerts its effects by inhibiting mitogen activated protein kinase 7 (MAPK7). The expression of MAPK7 was found to be significantly upregulated in osteosarcoma cells and overexpression of MAPK7 could nullify the effects of miR-187 on the proliferation of the osteosarcoma cells.


Asunto(s)
Neoplasias Óseas/genética , MicroARNs/metabolismo , Proteína Quinasa 7 Activada por Mitógenos/metabolismo , Osteosarcoma/genética , Antibióticos Antineoplásicos/farmacología , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/enzimología , Neoplasias Óseas/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Regulación hacia Abajo , Doxorrubicina/farmacología , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Humanos , Puntos de Control de la Fase M del Ciclo Celular/fisiología , MicroARNs/biosíntesis , MicroARNs/genética , Invasividad Neoplásica , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/enzimología , Osteosarcoma/patología
14.
J BUON ; 25(1): 491-496, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32277674

RESUMEN

PURPOSE: Gliomas are aggressive brain tumors accounting for significant mortality across the globe. Biomarkers for early detection and therapeutic targets for efficient treatment are lacking for glioma. This study was undertaken to investigate the role and therapeutic implications of miR-22 in glioma. METHODS: U-87 glioma cell line was used in this study. qRT-PCR was employed for expression analysis. MTT assay was used for determination of cell viability. Lipofectamine 2000 was used for transfection. Flow cytometry was used for cell analysis. Wound healing assay and transwell assay were used for monitoring cell migration and invasion. Western blot analysis was used for estimation of protein expression. RESULTS: The miR-22 expression was found decreased in glioma cells. Overexpression of miR-22 resulted in arrest of the U-87 glioma cells at G2/M checkpoint of the cell cycle. The percentage of apoptotic U-87 cells in G2/M phase were 13.05% in negative control (NC) and 29.06% in miR-22 mimics transfected cells. The cell cycle arrest promoted by miR-22 overexpression was also associated with depletion of cyclin B1 expression in U-87 cells. Furthermore, miR-22 could also significantly increase the sensitivity of glioma U-87 cells to cisplatin. The TargetScan analysis and dual luciferase assay showed SNAIL1 to be the target of miR-22. The expression of SNAIL1 was also enhanced in all the glioma cells and miR-22 overexpression could cause suppression of the SNAIL1 expression in U-87 cells. Furthermore, SNAIL1 silencing could also cause decline in the viability of the U-87 cells. The wound healing assay showed that miR-5 overexpression caused decrease in the migration of U-87 cells, while the transwell assay showed decline in the invasion of miR-22 mimics transfected U-87 cells. CONCLUSION: Taken together, miR-22 may exhibit therapeutic implications in glioma and may prove useful in glioma treatment.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , MicroARNs/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular/fisiología , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Glioma/genética , Glioma/patología , Humanos , Puntos de Control de la Fase M del Ciclo Celular/fisiología , MicroARNs/genética , Metástasis de la Neoplasia , Factores de Transcripción de la Familia Snail/biosíntesis , Factores de Transcripción de la Familia Snail/genética , Transfección
15.
Int J Mol Med ; 45(5): 1501-1513, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32323739

RESUMEN

Human dental pulp stem cells (hDPSCs) present several advantages, including their ability to be non­invasively harvested without ethical concern. The secretome of hDPSCs can promote the functional recovery of various tissue injuries. However, the protective effects on hypoxia­induced skeletal muscle injury remain to be explored. The present study demonstrated that C2C12 myoblast coculture with hDPSCs attenuated CoCl2­induced hypoxic injury compared with C2C12 alone. The hDPSC secretome increased cell viability and differentiation and decreased G2/M cell cycle arrest under hypoxic conditions. These results were further verified using hDPSC­conditioned medium (hDPSC­CM). The present data revealed that the protective effects of hDPSC­CM depend on the concentration ratio of the CM. In terms of the underlying molecular mechanism, hDPSC­CM activated the Wnt/ß­catenin pathway, which increased the protein levels of Wnt1, phosphorylated­glycogen synthase kinase­3ß and ß­catenin and the mRNA levels of Wnt target genes. By contrast, an inhibitor (XAV939) of Wnt/ß­catenin diminished the protective effects of hDPSC­CM. Taken together, the findings of the present study demonstrated that the hDPSC secretome alleviated the hypoxia­induced myoblast injury potentially through regulating the Wnt/ß­catenin pathway. These findings may provide new insight into a therapeutic alternative using the hDPSC secretome in skeletal muscle hypoxia­related diseases.


Asunto(s)
Pulpa Dental/metabolismo , Hipoxia/metabolismo , Mioblastos/metabolismo , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo , Puntos de Control del Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Supervivencia Celular/fisiología , Células Cultivadas , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Humanos , Músculo Esquelético/metabolismo , Células Madre/metabolismo
16.
Acta Pharmacol Sin ; 41(9): 1223-1233, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32242118

RESUMEN

Receptor-interacting protein 1 (RIP1, also known as RIPK1) is not only a tumor-promoting factor in several cancers but also mediates either apoptosis or necroptosis in certain circumstances. In this study we investigated what role RIP1 plays in human ovarian cancer cells. We showed that knockout (KO) of RIP1 substantially suppressed cell proliferation, accompanied by the G2/M checkpoint arrest in two human ovarian cancer cell lines SKOV3 and A2780. On the other hand, RIP1 KO remarkably attenuated cisplatin-induced cytotoxicity, which was associated with reduction of the apoptosis markers PARP cleavage and the necroptosis marker phospho-MLKL. We found that RIP1 KO suppressed cisplatin-induced ROS accumulation in both SKOV3 and A2780 cells. ROS scavenger BHA, apoptosis inhibitor Z-VAD or necroptosis inhibitor NSA could effectively suppress cisplatin's cytotoxicity in the control cells, suggesting that ROS-mediated apoptosis and necroptosis were involved in cisplatin-induced cell death. In addition, blocking necroptosis with MLKL siRNA effectively attenuated cisplatin-induced cytotoxicity. In human ovarian cancer A2780 cell line xenograft nude mice, RIP1 KO not only significantly suppressed the tumor growth but also greatly attenuated cisplatin's anticancer activity. Our results demonstrate a dual role of RIP1 in human ovarian cancer: it acts as either a tumor-promoting factor to promote cancer cell proliferation or a tumor-suppressing factor to facilitate anticancer effects of chemotherapeutics such as cisplatin.


Asunto(s)
Apoptosis/fisiología , Proliferación Celular/fisiología , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Necroptosis/fisiología , Neoplasias Ováricas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/farmacología , Femenino , Técnicas de Inactivación de Genes , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Necroptosis/efectos de los fármacos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Paclitaxel/farmacología , Especies Reactivas de Oxígeno/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
17.
Biochem Pharmacol ; 175: 113933, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32224138

RESUMEN

Studies have shown that palmatine (PAL) has anti-cancer effects. However, the activity and potential mechanisms of PAL against colorectal cancer remain elusive. The results showed that PAL significantly inhibited the proliferation of colon cancer cells in vitro and in vivo without significant effect on non-tumorigenic colon cells. Target prediction and clinical sample database analysis suggested that PAL may contribute to colon cancer cells phase arrest and apoptosis by targeting aurora kinase A (AURKA). Inhibition and overexpression of AURKA proved that PAL induces G2/M phase arrest and apoptosis in colon cancer cells by targeting AURKA. Moreover, PAL promoted intracellular Reactive oxygen species (ROS) production and decreased mitochondrial membrane potential (ΔΨm). PAL reduced the levels of AURKA, Bcl-xl and Bcl2 proteins, and promoted the expression of pro-apoptotic proteins P53, P73, Caspase3 and Caspase9, as well as the increase of cytochrome c (cyt. c) in cell lysates in vitro and in vivo. Together, our study confirmed that PAL induced G2/M phase arrest and mitochondrial-associated pathway apoptosis in colon cancer cells by targeting AURKA. PAL may provide a novel solution for the treatment of colon cancer by serving as a new AURKA inhibitor.


Asunto(s)
Aurora Quinasa A/antagonistas & inhibidores , Alcaloides de Berberina/administración & dosificación , Neoplasias del Colon , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Mitocondrias/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Aurora Quinasa A/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/metabolismo , Relación Dosis-Respuesta a Droga , Sistemas de Liberación de Medicamentos/métodos , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Células HCT116 , Células HT29 , Humanos , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Mitocondrias/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
18.
Cell Rep ; 30(7): 2083-2093.e5, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32075732

RESUMEN

Cell-autonomous changes in p53 expression govern the duration and outcome of cell-cycle arrest at the G2 checkpoint for DNA damage. Here, we report that mitogen-activated protein kinase (MAPK) signaling integrates extracellular cues with p53 dynamics to determine cell fate at the G2 checkpoint. Optogenetic tools and quantitative cell biochemistry reveal transient oscillations in MAPK activity dependent on ataxia-telangiectasia-mutated kinase after DNA damage. MAPK inhibition alters p53 dynamics and p53-dependent gene expression after checkpoint enforcement, prolonging G2 arrest. In contrast, sustained MAPK signaling induces the phosphorylation of CDC25C, and consequently, the accumulation of pro-mitotic kinases, thereby relaxing checkpoint stringency and permitting cells to evade prolonged G2 arrest and senescence induction. We propose a model in which this MAPK-mediated mechanism integrates extracellular cues with cell-autonomous p53-mediated signals, to safeguard genomic integrity during tissue proliferation. Early steps in oncogene-driven carcinogenesis may imbalance this tumor-suppressive mechanism to trigger genome instability.


Asunto(s)
Daño del ADN , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Expresión Génica , Humanos , Células MCF-7 , Proteínas Quinasas Activadas por Mitógenos/genética , Fosforilación , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
19.
Arch Biochem Biophys ; 684: 108320, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32105659

RESUMEN

Studies have proved the role of GAS5 in the development of different cancers. This study was undertaken to investigate the role and explore therapeutic implications of GAS5 in human cervical cancer. The results showed that GAS5 was significantly (p < 0.05) downregulated in human cervical cancer tissues. The results also showed that cervical cancer progresses with the suppression of GAS5 expression levels. Additionally, the expression of GAS5 was also significantly (p < 0.05) downregulated in human cervical cancer cell lines. Nonetheless, overexpression of GAS5 caused a remarkable decrease in the proliferation of C33A and HeLa cervical cancer cells. The decrease in the proliferation rate was attributed to the induction of apoptosis of C33A and HeLa cells which was accompanied with upregulation of Bax and suppression of Bcl-2. Additionally, GAS5 overexpression also promoted the arrest of C33A and HeLa cells at the G2/M check point of cell cycle via suppression of cyclin B1 and CDK1 expression. The transwell assays showed that GAS5 overexpression significantly (p < 0.05) inhibited the migration and invasion of the C33A and HeLa cervical cancer cells. The bioinformatics analysis as well as the dual luciferase assay showed GAS5 acts as a target of miR-135a. Interestingly, the expression of miR-135a was upregulated in the human cervical cancer cells and its suppression exerted growth inhibitory effects on the C33A and HeLa cells. However, silencing of GAS5 could nullify the effects of miR-135a suppression on the proliferation of C33A and HeLa cells. Taken together, the results of this study point towards the therapeutic implications of GAS5 in the treatment of cervical cancer.


Asunto(s)
Apoptosis/fisiología , Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , ARN Largo no Codificante/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Línea Celular Tumoral , Proliferación Celular/fisiología , Regulación hacia Abajo , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , MicroARNs/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal/fisiología , Regulación hacia Arriba , Neoplasias del Cuello Uterino/genética
20.
Biochem Soc Trans ; 48(1): 245-256, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32010930

RESUMEN

The Golgi complex (GC) has an essential role in the processing and sorting of proteins and lipids. The GC of mammalian cells is composed of stacks of cisternae connected by membranous tubules to create a continuous network, the Golgi ribbon, whose maintenance requires several core and accessory proteins. Despite this complex structural organization, the Golgi apparatus is highly dynamic, and this property becomes particularly evident during mitosis, when the ribbon undergoes a multistep disassembly process that allows its correct partitioning and inheritance by the daughter cells. Importantly, alterations of the Golgi structure are associated with a variety of physiological and pathological conditions. Here, we review the core mechanisms and signaling pathways involved in both the maintenance and disassembly of the Golgi ribbon, and we also report on the signaling pathways that connect the disassembly of the Golgi ribbon to mitotic entry and progression.


Asunto(s)
Puntos de Control de la Fase G2 del Ciclo Celular/fisiología , Proteínas de la Matriz de Golgi/metabolismo , Puntos de Control de la Fase M del Ciclo Celular/fisiología , Red trans-Golgi/metabolismo , Citoesqueleto de Actina/metabolismo , Animales , Humanos , Proteínas de Transporte de Membrana/metabolismo , Microtúbulos/metabolismo , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...