Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
Cancer Cell Int ; 24(1): 236, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38970092

RESUMEN

Chemotherapy is currently one of the most effective methods in clinical cancer treatment. However, chemotherapy resistance is an important reason for poor chemotherapy efficacy and prognosis, which has become an urgent problem to be solved in the field of cancer chemotherapy. Therefore, it is very important to deeply study and analyze the mechanism of cancer chemotherapy resistance and its regulatory factors. Long non-coding RNA nuclear paraspeckle assembly transcript 1 (LncRNA NEAT1) has been shown to be closely associated with chemotherapy resistance in cancer. NEAT1 induces cancer cell resistance to chemotherapeutic drugs by regulating cell apoptosis, cell cycle, drug transport and metabolism, DNA damage repair, EMT, autophagy, cancer stem cell characteristics, and metabolic reprogramming. This indicates that NEAT1 may be an important target to overcome chemotherapy resistance and is expected to be a potential biomarker to predict the effect of chemotherapy. This article summarizes the expression characteristics and clinical characteristics of NEAT1 in different cancers, and deeply discusses the regulatory role of NEAT1 in cancer chemotherapy resistance and related molecular mechanisms, aiming to clarify NEAT1 as a new target to overcome cancer chemotherapy resistance and the feasibility of chemotherapy sensitizers, with a view to providing a potential therapeutic direction for overcoming the dilemma of cancer resistance in the future.

2.
Cell Biochem Funct ; 42(2): e3961, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38425124

RESUMEN

A substantial number of long noncoding RNAs (lncRNAs) have been identified as potent regulators of human disease. Human leukocyte antigen complex group 18 (HCG18) is a new type of lncRNA that has recently been proven to play an important role in the occurrence and development of various diseases. Studies have found that abnormal expression of HCG18 is closely related to the clinicopathological characteristics of many diseases. More importantly, HCG18 was also found to promote disease progression by affecting a series of cell biological processes. This article mainly discusses the expression characteristics, clinical characteristics, biological effects and related regulatory mechanisms of HCG18 in different human diseases, providing a scientific theoretical basis for its early clinical application.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , MicroARNs/metabolismo
3.
J Cell Mol Med ; 27(7): 991-1005, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36915230

RESUMEN

Cyclin D1 (CCND1), a mediator of cell cycle control, has a G870A polymorphism which results in the formation of two splicing variants: full-length CCND1 (CCND1a) and C-terminally truncated CCND1 species (CCND1b). However, the role of CCND1a and CCND1b variants in cancer chemoresistance remains unknown. Therefore, this study aimed to explore the molecular mechanism of alternative splicing of CCND1 in breast cancer (BC) chemoresistance. To address the contribution of G870A polymorphism to the production of CCND1 variants in BC chemoresistance, we sequenced the G870A polymorphism and analysed the expressions of CCND1a and CCND1b in MCF-7 and MCF-7/ADM cells. In comparison with MCF-7 cells, MCF-7/ADM cells with the A allele could enhance alternative splicing with the increase of SC-35, upregulate the ratio of CCND1b/a at both mRNA and protein levels, and activate the CDK4/CyclinD1-pRB-E2F1 pathway. Furthermore, CCND1b expression and the downstream signalling pathway were analysed through Western blotting and cell cycle in MCF-7/ADM cells with knockdown of CCND1b. Knockdown of CCND1b downregulated the ratio of CCND1b/a, demoted cell proliferation, decelerated cell cycle progression, inhibited the CDK4/CyclinD1-pRB-E2F1 pathway and thereby decreased the chemoresistance of MCF-7/ADM cells. Finally, CCND1 G870A polymorphism, the alternative splicing of CCDN1 was detected through Sequenom Mass ARRAY platform, Sanger sequencing, semi-quantitative RT-PCR, Western blotting and immunohistochemistry in clinical BC specimens. The increase of the ratio of CCND1b/a caused by G870A polymorphism was involved in BC chemoresistance. Thus, these findings revealed that CCND1b/a ratio caused by the polymorphism is involved in BC chemoresistance via CDK4/CyclinD1-pRB-E2F1 pathway.


Asunto(s)
Neoplasias de la Mama , Femenino , Humanos , Empalme Alternativo/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Ciclina D1/genética , Quinasa 4 Dependiente de la Ciclina/genética , Resistencia a Antineoplásicos/genética , Factor de Transcripción E2F1/genética , Polimorfismo Genético , Proteína de Retinoblastoma/metabolismo
4.
J Cell Physiol ; 238(8): 1836-1849, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37334439

RESUMEN

Quiescent cancer cells are major impediments to effective radiotherapy (RT) and exhibit limited sensitivity to traditional photon therapy. Herein, the functional role and underlying mechanism of carbon ions in overcoming the radioresistance of quiescent cervical cancer HeLa cells were determined. Briefly, serum withdrawal was used to induce synchronized quiescence in HeLa cells. Quiescent HeLa cells displayed strong radioresistance and DNA repair potential. After irradiation with carbon ions, the DNA damage repair pathway may markedly rely on error-prone nonhomologous end-joining in proliferating cells, whereas the high-precision homologous recombination pathway is more relevant in quiescent cells. This phenomenon could be explained by the ionizing radiation (IR)-induced cell cycle re-entry of quiescent cancer cells. There are three strategies for eradicating quiescent cancer cells using high-linear energy transfer (LET) carbon ions: direct cell death through complex DNA damage; apoptosis via an enhanced mitochondria-mediated intrinsic pathway; forced re-entry of quiescent cancer cells into the cell cycle, thereby improving their susceptibility to IR. Silencing ß-catenin signaling is essential for maintaining the dormant state in quiescent cells. Herein, carbon ions activated the ß-catenin pathway in quiescent cells, and inhibition of this pathway improved the resistance of quiescent HeLa cells to carbon ions by alleviating DNA damage, improving DNA damage repair, maintaining quiescent depth, and inhibiting apoptosis. Collectively, carbon ions conquer the radioresistance of quiescent HeLa cells by activating ß-catenin signaling, which provides a theoretical basis for improved therapeutic effects in patients with middle-advanced-stage cervical cancer with radioresistance.


Asunto(s)
Neoplasias del Cuello Uterino , beta Catenina , Femenino , Humanos , Células HeLa , beta Catenina/genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/radioterapia , Reparación del ADN , Carbono , Iones/farmacología , Daño del ADN , Tolerancia a Radiación/genética
5.
FASEB J ; 36(3): e22229, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35199870

RESUMEN

The radioresistance induced by hypoxia is the major obstacle in the successful treatment of cancer radiotherapy. p21 was initially identified as a widespread inhibitor of cyclin-dependent kinases, through which mediates the p53-dependent cell cycle G1 phase arrest in response to a variety of stress stimuli. In this study, we discovered a novel function of p21, which participated in the regulation of metabolic pathways under hypoxia. We found that p21 was upregulated in glioblastoma (GBM) cells under hypoxic conditions, which enhanced the radioresistance of GBM cells. In principle, HIF-1α is bound directly to the hypoxia response elements (HREs) of the p21 promoter to enhance its transcription activity, in turn, p21 also promoted the transcription of HIF-1α at the mRNA level and maintained HIF-1α function under oxygen deficiency. The positive correlation between p21 and HIF-1α augmented Glut1/LDHA-mediated glycolysis and aggravated the radioresistance of GBM cells. Thus, our results constructed a positive feedback circuit comprising p21/HIF-1α that might play a key role in enhancing the radioresistance of GBM under hypoxia.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Glioblastoma/metabolismo , Glucólisis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Hipoxia Tumoral , Animales , Neoplasias Encefálicas/radioterapia , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Retroalimentación Fisiológica , Femenino , Glioblastoma/radioterapia , Transportador de Glucosa de Tipo 1/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , L-Lactato Deshidrogenasa/metabolismo , Ratones , Tolerancia a Radiación
6.
FASEB J ; 34(12): 15647-15658, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33058239

RESUMEN

Poly (rC)-binding protein 1 (PCBP1), an RNA- or DNA-binding protein with a relative molecular weight of 38 kDa, which is characterized by downregulation in many cancer types. Numerous cases have indicated that PCBP1 could be considered as a tumor suppressor to inhibit tumorigenesis, development, and metastasis. In the current review, we described the multilevel regulatory roles of PCBP1, including gene transcription, alternative splicing, and translation of many cancer-related genes. Additionally, we also provided a brief overview about the inhibitory effect of PCBP1 on most common tumors. More importantly, we summarized the current research status about PCBP1 in hypoxic microenvironment, autophagy, apoptosis, and chemotherapy of cancer cells, aiming to clarify the molecular mechanisms of PCBP1 in cancer. Taken together, in-depth study of PCBP1 in cancer may provide new ideas for cancer therapy.


Asunto(s)
Proteínas de Unión al ADN/genética , Neoplasias/genética , Proteínas de Unión al ARN/genética , Animales , Apoptosis/genética , Autofagia/genética , Carcinogénesis/genética , Expresión Génica/genética , Humanos , Microambiente Tumoral/genética
7.
Int J Mol Sci ; 22(2)2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33467535

RESUMEN

Cell can integrate the caspase family and mammalian target of rapamycin (mTOR) signaling in response to cellular stress triggered by environment. It is necessary here to elucidate the direct response and interaction mechanism between the two signaling pathways in regulating cell survival and determining cell fate under cellular stress. Members of the caspase family are crucial regulators of inflammation, endoplasmic reticulum stress response and apoptosis. mTOR signaling is known to mediate cell growth, nutrition and metabolism. For instance, over-nutrition can cause the hyperactivation of mTOR signaling, which is associated with diabetes. Nutrition deprivation can inhibit mTOR signaling via SH3 domain-binding protein 4. It is striking that Ras GTPase-activating protein 1 is found to mediate cell survival in a caspase-dependent manner against increasing cellular stress, which describes a new model of apoptosis. The components of mTOR signaling-raptor can be cleaved by caspases to control cell growth. In addition, mTOR is identified to coordinate the defense process of the immune system by suppressing the vitality of caspase-1 or regulating other interferon regulatory factors. The present review discusses the roles of the caspase family or mTOR pathway against cellular stress and generalizes their interplay mechanism in cell fate determination.


Asunto(s)
Apoptosis/fisiología , Caspasas/metabolismo , Proliferación Celular/fisiología , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/metabolismo , Animales , Retículo Endoplásmico/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Humanos , Inflamación/metabolismo
8.
J Cell Physiol ; 235(3): 1903-1914, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31332789

RESUMEN

Transforming growth factor ß (TGF-ß) is part of the transforming growth factor ß superfamily which is involved in many physiological processes and closely related to the carcinogenesis. Here, we discuss the TGF-ß structure, function, and its canonical Smads signaling pathway. Importantly, TGF-ß has been proved that it plays both tumor suppressor as well as an activator role in tumor progression. In an early stage, TGF-ß inhibits cell proliferation and is involved in cell apoptosis. In an advanced tumor, TGF-ß signaling pathway induces tumor invasion and metastasis through promoting angiogenesis, epithelial-mesenchymal transition, and immune escape. Furthermore, we are centered on updated research results into the inhibitors as drugs which have been studied in preclinical or clinical trials in tumor carcinogenesis to prevent the TGF-ß synthesis and block its signaling pathways such as antibodies, antisense molecules, and small-molecule tyrosine kinase inhibitors. Thus, it is highlighting the crucial role of TGF-ß in tumor therapy and may provide opportunities for the new antitumor strategies in patients with cancer.


Asunto(s)
Neoplasias/metabolismo , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Transducción de Señal/efectos de los fármacos
10.
Anal Bioanal Chem ; 412(2): 481-488, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31728594

RESUMEN

Monitoring and imaging glutathione (GSH) in living systems is an essential tool to determine the key roles of GSH in biological pathways, but most fluorescent sensors can only be used in vitro because of their potential biotoxicity. Here, a peptide-based fluorescent sensor, FP, has been successfully designed and synthesized based on the biocompatibility of the peptide backbone and low toxicity. The design strategy of FP contains a specific spatial structure of the peptide sequence which selectively binds to Cu2+, triggering fluorescence quenching. Interestingly, the fluorescence of FP can be fully restored by GSH, due to the strong binding between Cu2+ and the GSH sulfhydryl groups. Finally, the sensor is highly sensitive and selective for imaging GSH both in vitro and in vivo with low toxicity. Thus, FP with its strong "on-off-on" fluorescence changes is a powerful way to image GSH both in cells and zebrafish larvae to study the GSH pathway.


Asunto(s)
Colorantes Fluorescentes/química , Glutatión/metabolismo , Péptidos/química , Animales , Cromatografía Líquida de Alta Presión , Células HeLa , Humanos , Límite de Detección , Microscopía Fluorescente , Espectrometría de Fluorescencia , Espectrometría de Masa por Ionización de Electrospray , Pez Cebra/embriología
11.
Ecotoxicol Environ Saf ; 201: 110831, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32535367

RESUMEN

The risk of exposure to ionizing radiation (IR) environments has increased with the development of nuclear technology. IR exposure induces excessive apoptosis of the spermatogonia, which leads to male infertility. Spermatogonia apoptosis may be involved in ribosomal stress triggered by DNA damage following exposure to IR because ribosomal proteins (RPs) directly interact with mouse double minute 2 homolog (MDM2) to induce apoptosis. This study aimed to use comparative proteomics and transcriptomics approach to screen the differential RPs and ribosomal mRNAs in mouse testes following high linear energy transfer (LET) carbon ion radiation (CIR). The expression of ribosomal large subunit protein 27a (Rpl27a) decreased at both protein and mRNA levels in the spermatogonia in vivo. After 6 h of CIR, the immunofluorescence signal of 8-oxo-dG and phosphorylated ataxia-telangiectasia-mutated protein (ATM)/histone H2Ax increased, but that of Rpl27a decreased in the spermatogonia of p53 wild-type and knockout mouse testes. Moreover, the nucleolin was scattered throughout the nucleoplasm after CIR. These results suggested that CIR-induced DNA damage might trigger ribosomal stress, and the reduction in the expression of Rpl27a was associated with DNA damage in the spermatogonia. Similarly, in vitro, the immunofluorescence signal of 8-oxo-dG increased in the GC-1 cells after CIR. Moreover, the expression of Rpl27a was regulated by DNA damage because the co-transfection of ATM and Rpl27a or inhibition of ATM-treated CIR could restore the expression of Rpl27a. Furthermore, the reduction in the expression of Rpl27a led to weakened binding of E2F transcription factor 1 (E2F1) and p53 to MDM2, causing p53 activation and E2F1 degradation in p53 wild-type and knockdown GC-1 cells. This study proposed that heavy ion radiation-induced DNA damage mediated spermatogonia apoptosis via the Rpl27a-Rpl5-MDM2-p53/E2F1 signaling pathway. The results provided the underlying molecular mechanisms of spermatogonia apoptosis following exposure to high LET radiation.


Asunto(s)
Apoptosis/efectos de la radiación , Daño del ADN , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Radiación Ionizante , Proteínas Ribosómicas/metabolismo , Espermatogonias/efectos de la radiación , Animales , Apoptosis/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Iones Pesados , Humanos , Masculino , Ratones , Proteínas Proto-Oncogénicas c-mdm2/genética , Proteínas Ribosómicas/genética , Transducción de Señal , Espermatogonias/metabolismo , Espermatogonias/patología , Proteína p53 Supresora de Tumor/genética
12.
J Cell Physiol ; 234(12): 22439-22449, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31087336

RESUMEN

The mitochondrial proteins involved in spermatogenic cells apoptosis in zebrafish after carbon ion radiation (CIR) were screened. The relative biological effectiveness (RBE) of CIR in zebrafish testes was investigated. Apoptosis of testicular cells was measured within 24 hr following 1 and 4 Gy CIR. Immunoblotting was used to assess the levels of mitochondrial apoptotic proteins in testes, and proliferative and apoptotic spermatogenic cells were detected by immunofluorescence after CIR. Label-free quantitative (LFQ) and parallel reaction monitoring-based target proteomics (PRM) were combined to screen and validate differential mitochondrial proteins in testes between 4 Gy and control groups at 24 hr after CIR. The RBE of CIR in zebrafish testes was 1.48 ± 0.04, and induction of apoptosis by CIR was higher than that of X-rays in testicular cells. Mitochondrial apoptotic pathways play a crucial role in spermatogenic cells apoptosis after CIR, with 60 differential mitochondrial proteins identified. Among 20 target proteins, 12 were significantly upregulated, 2 were significantly downregulated in the 4 Gy CIR group. The results of PRM were consistent with label-free analysis. This is the first study to screen the differential mitochondrial proteins and provide useful information to understand the underlying mechanisms of spermatogenic cell apoptosis in zebrafish following CIR.


Asunto(s)
Apoptosis/efectos de la radiación , Carbono , Radioterapia de Iones Pesados/efectos adversos , Iones Pesados , Espermatogonias/efectos de los fármacos , Testículo/citología , Animales , Proliferación Celular/efectos de la radiación , Regulación de la Expresión Génica/efectos de la radiación , Masculino , Mitocondrias/metabolismo , Mitocondrias/efectos de la radiación , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Proteómica , Testículo/efectos de la radiación , Rayos X , Pez Cebra
13.
J Cell Physiol ; 234(8): 13182-13190, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30536619

RESUMEN

Prostate cancer (CaP) is the second most common cancer in men worldwide in 2012, and radiation therapy is one of the most common definitive treatment options for localized CaP. However, radioresistance is a major challenge for the current radiotherapy, accumulating evidences suggest microRNAs (miRNAs), as an important regulator in cellular ionizing radiation (IR) responses, are closely correlated with radiosensitivity in many cancers. Here, we identified microRNA-16-5p(miR-16-5p) is significantly upregulated in CaP LNCaP cells following IR and can enhance radiosensitivity through modulating Cyclin D1/E1-pRb-E2F1 pathway. To identify the expression profile of miRNAs in CaP cells exposed to IR, we performed human miRNA probe hybridization chip analysis and miR-16-5p was found to be significantly overexpressed in all treatment groups that irradiated with different doses of X-rays and heavy ions (12 C6+ ). Furthermore, overexpression of miR-16-5p suppressed cell proliferation, reduced cell viability, and induced cell cycle arrest at G0/G1 phase, resulting in enhanced radiosensitivity in LNCaP cells. Additionally, miR-16-5p specifically targeted the Cyclin D1/E1-3'-UTR in LNCaP cells and affected the expression of Cyclin D1/E1 in both mRNA and protein levels. Taken together, miR-16-5p enhanced radiosensitivity of CaP cells, the mechanism may be through modulating Cyclin D1/Cyclin E1/pRb/E2F1 pathway to cause cell cycle arrest at G0/G1 phase. These findings provided new insight into the correlation between miR-16-5p, cell cycle arrest, and radiosensitivity in CaP, revealed a previously unrecognized function of miR-16-5p-Cyclin D1/E1-pRb-E2F1 regulation in response to IR and may offer an alternative therapy to improve the efficiency of conventional radiotherapy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , MicroARNs/metabolismo , Neoplasias de la Próstata/genética , Tolerancia a Radiación/genética , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Ciclina D1/genética , Ciclina D1/metabolismo , Ciclina E/genética , Ciclina E/metabolismo , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Humanos , Masculino , MicroARNs/genética , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/metabolismo , Neoplasias de la Próstata/metabolismo , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Transducción de Señal/genética
14.
J Cell Physiol ; 233(2): 1312-1320, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28500630

RESUMEN

Deletion of p53, most common genetic alteration, is observed in human tumors and reported to lead to improve in cell radioresistance. Heavy-ion irradiation (IR) could induce p53-/- cancer cells apoptosis. However, little is known regarding the molecular mechanism in this type of cell apoptosis. The present studies have focused on mechanisms state of signaling pathways as an activator of the cell fate decisions induced by heavy ion IR without p53. Carbon ion IR could induce up-regulation of E2F1 expression in cancer cells. This phenomenon was not observed in X-ray IR group. Up-regulation of E2F1 could cause a higher reduction in clonogenic survival, low level of cellular activity, G2 /M phase arrest, promotion of apoptosis rate, up-regulation of phosphor-Rb, Bax, and cleaved-caspase 3 proteins expressions without p53. Changes of E2F1 expressions could partly alter radioresistance in cancer cells. The results were suggested that heavy ion IR could induce p53-/- cancer cells apoptosis via E2F1 signal pathway. Our study provides a scientific rationale for the clinical use of heavy ion as radiotherapy in patients with p53-deficient tumors, which are often resistant to radiotherapy.


Asunto(s)
Apoptosis/efectos de la radiación , Carcinoma Hepatocelular/tratamiento farmacológico , Caspasa 3/metabolismo , Factor de Transcripción E2F1/metabolismo , Radioterapia de Iones Pesados , Neoplasias Hepáticas/radioterapia , Tolerancia a Radiación , Proteína X Asociada a bcl-2/metabolismo , Células A549 , Carcinoma Hepatocelular/enzimología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Relación Dosis-Respuesta en la Radiación , Factor de Transcripción E2F1/genética , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Regulación Neoplásica de la Expresión Génica , Células Hep G2 , Humanos , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Transducción de Señal/efectos de la radiación , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Regulación hacia Arriba
15.
Tumour Biol ; 37(4): 4831-40, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26520443

RESUMEN

miR-449a, a novel tumor suppressor, is deregulated in various malignancies, including prostate cancer. Overexpression of miR-449a induces cell cycle arrest, apoptosis, and senescence, but its role in response to ionizing radiation and underlying molecular mechanism are still unknown. Here, we report that miR-449a enhances radiation-induced G2/M phase arrest and apoptosis through modulating pRb/E2F1 and sensitizes prostate cancer cells to X-ray radiation. In wild-type Rb PC-3 cells, overexpression of miR-449a enhances radiation-induced G2/M arrest and apoptosis and promotes the sensitivity to X-ray radiation. While mutant Rb DU-145 cells are resistant to the X-ray radiation despite in the presence of miR-449a. The cell cycle distribution of DU-145 cells is not significantly altered by miR-449a in the response to ionizing radiation. Furthermore, elevated miR-449a downregulates cell cycle regulator CDC25A and oncogene HDAC1. By targeting genes involved in controlling pRb/E2F1 activity, miR-449a regulates cell cycle progression and apoptosis and consequently enhances the radiosensitivity of PC-3 cells. Thus, miR-449a, as a miRNA component of the Rb pathway, promotes the radiosensitivity of PC-3 cells through regulating pRb/E2F1.


Asunto(s)
Factor de Transcripción E2F1/genética , MicroARNs/genética , Neoplasias de la Próstata/radioterapia , Tolerancia a Radiación/genética , Proteína de Retinoblastoma/genética , Apoptosis/efectos de la radiación , Puntos de Control del Ciclo Celular/efectos de la radiación , Línea Celular Tumoral , Proliferación Celular/efectos de la radiación , Factor de Transcripción E2F1/biosíntesis , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Masculino , MicroARNs/biosíntesis , Próstata/efectos de la radiación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteína de Retinoblastoma/biosíntesis , Transducción de Señal/efectos de la radiación , Rayos X
16.
J Cell Physiol ; 229(1): 100-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23804302

RESUMEN

Mitochondria are a major source of reactive oxygen species (ROS) and are also the target of cellular ROS. ROS damage to mitochondria leads to dysfunction that further enhances the production of mitochondrial ROS. This feed-forward vicious cycle between mitochondria and ROS induces cell death. Within a few minutes of radiation exposure, NADPH oxidase is activated to elevate the ROS level. Activated NADPH oxidase might induce the feed-forward cycle of mitochondria and this is a possible mechanism for cancer cell death induced by heavy ion irradiation. We found that after 4 Gy of (12) C(6+) ion radiation of HepG2 cells, the NADPH oxidase membrane subunit gp91(phox) was not involved in enzyme activation through increased expression; however, the subunit p47(phox) was involved in activation by being translocated to the membrane. (12) C(6+) ion radiation clearly decreased the ΔΨm of HepG2 cells, increasing mitochondrial DNA damage and inducing cell death. Pretreatment with apocynin (APO, an NADPH oxidase inhibitor) effectively prevented the ΔΨm decrease, mitochondrial DNA damage, and cell death induced by radiation. However, these protective effects were not observed with APO treatment after irradiation exposure. These data demonstrated that NADPH oxidase activation was an initiator in mitochondrial damage. Once mitochondria entered the feed-forward cycle, cell fate was no longer controlled by NADPH oxidase. Only antioxidants that targeted mitochondria such as MitoQ could break the cycle and release cells from death.


Asunto(s)
Carcinoma Hepatocelular/metabolismo , Radioterapia de Iones Pesados/efectos adversos , Neoplasias Hepáticas/metabolismo , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/toxicidad , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antioxidantes/metabolismo , Apoptosis/efectos de los fármacos , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/radioterapia , Activación Enzimática , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/radioterapia , Glicoproteínas de Membrana/metabolismo , Mitocondrias Hepáticas/efectos de los fármacos , Mitocondrias Hepáticas/efectos de la radiación , NADPH Oxidasa 2 , Compuestos Organofosforados/administración & dosificación , Oxidación-Reducción , Especies Reactivas de Oxígeno/efectos de la radiación , Ubiquinona/administración & dosificación , Ubiquinona/análogos & derivados
17.
Cell Death Dis ; 15(2): 160, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38383492

RESUMEN

Dysregulation of anti-apoptotic and pro-apoptotic protein isoforms arising from aberrant splicing is a crucial hallmark of cancers and may contribute to therapeutic resistance. Thus, targeting RNA splicing to redirect isoform expression of apoptosis-related genes could lead to promising anti-cancer phenotypes. Glioblastoma (GBM) is the most common type of malignant brain tumor in adults. In this study, through RT-PCR and Western Blot analysis, we found that BCLX pre-mRNA is aberrantly spliced in GBM cells with a favored splicing of anti-apoptotic Bcl-xL. Modulation of BCLX pre-mRNA splicing using splice-switching oligonucleotides (SSOs) efficiently elevated the pro-apoptotic isoform Bcl-xS at the expense of the anti-apoptotic Bcl-xL. Induction of Bcl-xS by SSOs activated apoptosis and autophagy in GBM cells. In addition, we found that ionizing radiation could also modulate the alternative splicing of BCLX. In contrast to heavy (carbon) ion irradiation, low energy X-ray radiation-induced an increased ratio of Bcl-xL/Bcl-xS. Inhibiting Bcl-xL through splicing regulation can significantly enhance the radiation sensitivity of 2D and 3D GBM cells. These results suggested that manipulation of BCLX pre-mRNA alternative splicing by splice-switching oligonucleotides is a novel approach to inhibit glioblastoma tumorigenesis alone or in combination with radiotherapy.


Asunto(s)
Glioblastoma , Precursores del ARN , Humanos , Empalme Alternativo/genética , Apoptosis/genética , Proteína bcl-X/genética , Proteína bcl-X/metabolismo , Glioblastoma/genética , Glioblastoma/radioterapia , Oligonucleótidos/metabolismo , Isoformas de Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Precursores del ARN/genética , Precursores del ARN/metabolismo , Empalme del ARN/genética
18.
Cell Death Discov ; 10(1): 16, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38195680

RESUMEN

Radiopharmaceuticals play a vital role in cancer therapy. The carrier of radiopharmaceuticals can precisely locate and guide radionuclides to the target, where radionuclides kill surrounding tumor cells. Effective application of radiopharmaceuticals depends on the selection of an appropriate carrier. Herein, different types of carriers of radiopharmaceuticals and the characteristics are briefly described. Subsequently, we review radiolabeled monoclonal antibodies (mAbs) and their derivatives, and novel strategies of radiolabeled mAbs and their derivatives in the treatment of lymphoma and colorectal cancer. Furthermore, this review outlines radiolabeled peptides, and novel strategies of radiolabeled peptides in the treatment of neuroendocrine neoplasms, prostate cancer, and gliomas. The emphasis is given to heterodimers, bicyclic peptides, and peptide-modified nanoparticles. Last, the latest developments and applications of radiolabeled nucleic acids and small molecules in cancer therapy are discussed. Thus, this review will contribute to a better understanding of the carrier of radiopharmaceuticals and the application in cancer therapy.

19.
Mutat Res ; 745-746: 26-33, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23535216

RESUMEN

The effects of carbon ion irradiation and ferulic acid (FA) on the induction of oxidative stress and alteration of gene expression were studied in zebrafish (Danio rerio) embryos. Zebrafish embryos at 8 hpf were divided into seven groups: the control group; the 1Gy, 3Gy and 7Gy irradiation groups; and three FA-pre-treated irradiation groups. In the irradiated groups, a significant increase in the teratogenesis of the zebrafish embryos and oxidative stress was accompanied by increased malondialdehyde (MDA) content, decreased glutathione (GSH) content and alterations in antioxidant enzyme activities (such as catalase [CAT] and superoxide dismutase [SOD]). Moreover, the mRNA levels for Cu/Zn-sod, Mn-sod, cat and gpx, the genes encoding these antioxidant proteins, were altered significantly. However, the mRNA expression patterns were not in accordance with those of the antioxidant enzymes and were more sensitive under low-dose irradiation. In addition, we detected the mRNA expression of ucp-2 and bcl-2, which are located at the mitochondrial inner membrane and related to reactive oxidative species (ROS) production. In the irradiated groups, the mRNA level of ucp-2 was significantly increased, whereas the mRNA level of bcl-2 was significantly decreased. Supplementation with FA, an antioxidant, was better able to reduce the irradiation-induced oxidative damage marked by changes in mortality, morphology, antioxidant enzyme activities and the MDA and GSH content, as well as in the mRNA expression levels. Overall, this study provided helpful information about the transcriptional effects of irradiation to better understand the mechanism of carbon ion-induced oxidative stress and FA-induced radioprotective effects.


Asunto(s)
Ácidos Cumáricos/farmacología , Regulación del Desarrollo de la Expresión Génica/efectos de la radiación , Estrés Oxidativo/efectos de la radiación , Protectores contra Radiación/farmacología , Pez Cebra/embriología , Pez Cebra/genética , Animales , Carbono , Catalasa/genética , Catalasa/metabolismo , Relación Dosis-Respuesta en la Radiación , Embrión no Mamífero/efectos de la radiación , Femenino , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glutatión/genética , Glutatión/metabolismo , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Peroxidación de Lípido/efectos de la radiación , Masculino , Malondialdehído/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
20.
Mutat Res ; 755(2): 148-55, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23827780

RESUMEN

This paper investigates the mechanism of action of heavy ion radiation (HIR) on mouse testes. The testes of male mice subjected to whole body irradiation with carbon ion beam (0.5 and 4Gy) were analyzed at 7days after irradiation. A two-dimensional gel electrophoresis approach was employed to investigate the alteration of protein expression in the testes. Spot detection and matching were performed using the PDQuest 8.0 software. A difference of more than threefold in protein quantity (normalized spot volume) is the standard for detecting differentially expressed protein spots. A total of 11 differentially expressed proteins were found. Protein identification was performed using matrix-assisted laser desorption/ionization tandem time-of-flight mass spectrometry (MALDI-TOF-TOF). Nine specific proteins were identified by searching the protein sequence database of the National Center for Biotechnology Information. These proteins were found involved in molecular chaperones, metabolic enzymes, oxidative stress, sperm function, and spermatogenic cell proliferation. HIR decreased glutathione activity and increased malondialdehyde content in the testes. Given that Pin1 is related to the cell cycle and that proliferation is affected by spermatogenesis, we analyzed testicular histological changes and Pin1 protein expression through immunoblotting and immunofluorescence. Alterations of multiple pathways may be associated with HIR toxicity to the testes. Our findings are essential for studies on the development, biology, and pathology of mouse testes after HIR in space or radiotherapy.


Asunto(s)
Carbono/toxicidad , Perfilación de la Expresión Génica/métodos , Iones Pesados/efectos adversos , Biosíntesis de Proteínas/efectos de la radiación , Proteómica/métodos , Testículo/efectos de la radiación , Animales , Proteínas de Ciclo Celular/biosíntesis , Proteínas de Ciclo Celular/genética , Diferenciación Celular/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Electroforesis en Gel Bidimensional , Glutatión/análisis , Peroxidación de Lípido/efectos de la radiación , Masculino , Malondialdehído/análisis , Ratones , Microscopía Fluorescente , Chaperonas Moleculares/biosíntesis , Chaperonas Moleculares/genética , Peptidilprolil Isomerasa de Interacción con NIMA , Estrés Oxidativo/genética , Estrés Oxidativo/efectos de la radiación , Isomerasa de Peptidilprolil/biosíntesis , Isomerasa de Peptidilprolil/genética , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espermatogénesis/genética , Técnica de Sustracción , Testículo/metabolismo , Testículo/ultraestructura , Irradiación Corporal Total
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA