Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Biochem Biophys Res Commun ; 681: 232-241, 2023 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-37788590

RESUMEN

Inflammation is implicated in the development of diabetic complications including vascular pathology. Centrosome is known to play a role in cell secretion. We have reported that diabetes can trigger centrosome amplification (CA). Thus, in the present study, we investigated the relationship between CA and the release of proinflammatory cytokines interleukin-1ß, tumor necrosis factor-α and interleukin-6 in hCMEC/D3 human endothelial cells treated with advanced glycation end products (AGEs). We found that AGEs induced CA via PLK4 and increased the biosynthesis of the three cytokines via NF-κB. Importantly, treatment of the cells with AGEs also increased the release of the three cytokines. Inhibiting CA by knockdown of polo like kinase 4 (PLK4) attenuated the cytokine release but not their biosynthesis. Knockdown of the cytokines inhibited the CA, while addition of the cytokines individually to the cell culture increased the protein level of PLK4 and CA to a moderate level. Addition of the three cytokines together into the cell culture markedly enhanced the CA, to a level higher than that in the AGEs-treated group. In conclusion, our results provide the direct evidence that the cytokines can induce CA, and suggest that there is a mutual promoting cycle between CA and cytokine release in the treated samples. It is proposed that the cycle of CA-cytokine release is a candidate biological link between diabetes and its complications such as vascular pathologies.


Asunto(s)
Citocinas , Diabetes Mellitus , Humanos , Productos Finales de Glicación Avanzada/metabolismo , Células Endoteliales/metabolismo , FN-kappa B/metabolismo , Centrosoma/metabolismo , Proteínas Serina-Treonina Quinasas
2.
Ecotoxicol Environ Saf ; 254: 114726, 2023 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-36898312

RESUMEN

Fetal growth restriction (FGR) is one of the most common obstetric diseases, and affects approximately 10 % of all pregnancies worldwide. Maternal cadmium (Cd) exposure is one of the factors that may increase the risk of the development of FGR. However, its underlying mechanisms remain largely unknown. In this study, using Cd-treated mice as an experimental model, we analyzed the levels of some nutrients in the circulation and the fetal livers by biochemical assays; the expression patterns of several key genes involved in the nutrient uptake and transport, and the metabolic changes in the maternal livers were also examined by quantitative real-time PCR and gas chromatography-time of flight-mass spectrometry method. Our results showed that, the Cd treatment specifically reduced the levels of total amino acids in the peripheral circulation and the fetal livers. Concomitantly, Cd upregulated the expressions of three amino acid transport genes (SNAT4, SNAT7 and ASCT1) in the maternal livers. The metabolic profiling of maternal livers also revealed that, several amino acids and their derivatives were also increased in response to the Cd treatment. Further bioinformatics analysis indicated that the experimental treatment activated the metabolic pathways, including the alanine, aspartate and glutamate metabolism, valine, leucine and isoleucine biosynthesis, arginine and proline metabolism. These findings suggest that maternal Cd exposure activate the amino acid metabolism and increase the amino acid uptake in the maternal liver, which reduces the supply of amino acids to the fetus via the circulation. We suspect that this underlies the Cd-evoked FGR.


Asunto(s)
Aminoácidos , Cadmio , Embarazo , Humanos , Femenino , Ratones , Animales , Aminoácidos/metabolismo , Cadmio/metabolismo , Placenta/metabolismo , Exposición Materna/efectos adversos , Hígado/metabolismo
3.
Am J Physiol Cell Physiol ; 318(1): C48-C62, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31618077

RESUMEN

We recently published that type 2 diabetes promotes cell centrosome amplification via upregulation of Rho-associated protein kinase 1 (ROCK1) and 14-3-3 protein-σ (14-3-3σ). This study further investigates the molecular mechanisms underlying diabetes-associated centrosome amplification. We found that treatment of cells with high glucose, insulin, and palmitic acid levels increased the intracellular and extracellular protein levels of Wingless-type MMTV integration site family member 6 (Wnt6) as well as the cellular level of ß-catenin. The treatment also activated ß-catenin and promoted its nuclear translocation. Treatment of cells with siRNA species for Wnt6, Frizzled-4 (FZD4), or ß-catenin as well as introduction of antibodies against Wnt6 or FZD4 to the cell culture medium could all attenuate the treatment-triggered centrosome amplification. Moreover, we showed that secreted Wnt6-FZD4-ß-catenin was the signaling pathway that was upstream of ROCK1 and 14-3-3σ. We found that advanced glycation end products (AGEs) were also able to increase the cellular and extracellular levels of Wnt6, the cellular protein level of ß-catenin, and centrosome amplification. Treatment of the cells with siRNA species for Wnt6 or FZD4 as well as introduction of antibodies against Wnt6 or FZD4 to the cell culture could all inhibit the AGEs-elicited centrosome amplification. In colon tissues from a diabetic mouse model, the protein levels of Wnt6 and 14-3-3σ were increased. In conclusion, our results showed that the pathophysiological factors in type 2 diabetes, including AGEs, were able to induce centrosome amplification. It is suggested that secreted Wnt6 binds to FZD4 to activate the canonical Wnt6 signaling pathway, which is upstream of ROCK1 and 14-3-3σ, and that this is the cell signaling pathway underlying diabetes-associated centrosome amplification.


Asunto(s)
Centrosoma/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Receptores Frizzled/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Proteínas 14-3-3/metabolismo , Animales , Biomarcadores de Tumor/metabolismo , Glucemia/metabolismo , Centrosoma/patología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patología , Exorribonucleasas/metabolismo , Femenino , Receptores Frizzled/genética , Productos Finales de Glicación Avanzada/farmacología , Células HCT116 , Humanos , Insulina/sangre , Ratones Endogámicos ICR , Ácido Palmítico/farmacología , Unión Proteica , Ratas , Proteínas Wnt/genética , Quinasas Asociadas a rho/metabolismo
4.
Acta Biochim Biophys Sin (Shanghai) ; 52(1): 72-83, 2020 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-31844893

RESUMEN

Type 2 diabetes increases the risk for cancer. Centrosome amplification can initiate tumorigenesis. We have described that type 2 diabetes increases the centrosome amplification of peripheral blood mononuclear cells, with high glucose, insulin, and palmitic acid as the triggers, which suggests that centrosome amplification is a candidate biological mechanism linking diabetes to cancer. In this study, we aimed to further investigate the signaling pathways of the diabetes-associated centrosome amplification and to examine whether and how resveratrol inhibits the centrosome amplification. The results showed that treatment with high glucose, insulin, and palmitic acid, alone or in combination, could increase the protein levels of phospho-protein kinase C alpha (p-PKCα), phospho-p38 mitogen-activated protein kinases (p-p38), c-myc, and c-jun, as well as the mRNA levels of c-myc and c-jun. PKCα inhibitor could inhibit the treatment-induced increase in the protein levels of p-p38, c-myc, and c-jun. Inhibitor or siRNA of p38 was also able to inhibit the treatment-induced increase in the levels of p-p38, c-myc, and c-jun. Meanwhile, knockdown of c-myc or c-jun did not alter the treatment-induced increase in the phosphorylation of PKCα or p38. Importantly, inhibition of the phosphorylation of PKCα or p38 and knockdown of c-myc or c-jun could attenuate the centrosome amplification. In diabetic mice, the levels of p-PKCα, p-p38, c-myc, and c-jun were all increased in the colon tissues. Interestingly, resveratrol, but not metformin, was able to attenuate the treatment-induced increase in the levels of p-PKCα, p-p38, c-myc, and c-jun, as well as the centrosome amplification. In conclusion, our results suggest that PKCα-p38 to c-myc/c-jun is the signaling pathway of the diabetes-associated centrosome amplification, and resveratrol attenuates the centrosome amplification by inhibiting this signaling pathway.


Asunto(s)
Centrosoma/efectos de los fármacos , Diabetes Mellitus Experimental/tratamiento farmacológico , Proteína Quinasa C-alfa/metabolismo , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Resveratrol/farmacología , Resveratrol/uso terapéutico , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Centrosoma/metabolismo , Colon/metabolismo , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Técnicas de Silenciamiento del Gen , Glucosa/farmacología , Células HCT116 , Humanos , Insulina/farmacología , Ratones , Ácido Palmítico/farmacología , Fosforilación/efectos de los fármacos , Proteína Quinasa C-alfa/genética , Proteínas Proto-Oncogénicas c-myc/genética , ARN Interferente Pequeño/genética , Estreptozocina/efectos adversos , Estreptozocina/farmacología , Transfección , Proteínas Quinasas p38 Activadas por Mitógenos/genética
5.
Proteomics ; 19(7): e1800197, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30688006

RESUMEN

It has been reported recently that type 2 diabetes promotes centrosome amplification via 14-3-3σ/ROCK1 complex. In the present study, 14-3-3σ interacting proteins are characterized and their roles in the centrosome amplification by high glucose, insulin, and palmitic acid are investigated. Co-immunoprecipitation in combination with MS analysis identified 134 proteins that interact with 14-3-3σ, which include heat shock 70 kDa protein 4 (Hsp74). Gene ontology analyses reveal that many of them are enriched in binding activity. Kyoto Encyclopedia of Genes and Genomes analysis shows that the top three enriched pathways are ribosome, carbon metabolism, and biosynthesis of amino acids. Molecular and functional investigations show that the high glucose, insulin, and palmitic acid increase the expression and binding of 14-3-3σ and Hsp74 as well as centrosome amplification, all of which are inhibited by knockdown of 14-3-3σ or Hsp74. Moreover, molecular docking analysis shows that the interaction between the 14-3-3σ and the Hsp74 is mainly through hydrophobic contacts and a lesser degree ionic interactions and hydrogen bond by different amino acids residues. In conclusion, the results suggest that the experimental treatment triggers centrosome amplification via upregulations of expression and binding of 14-3-3σ and Hsp74.


Asunto(s)
Proteínas 14-3-3/metabolismo , Proteínas Portadoras/metabolismo , Centrosoma/metabolismo , Glucosa/farmacología , Proteínas HSP70 de Choque Térmico/metabolismo , Insulina/farmacología , Ácido Palmítico/farmacología , Western Blotting , Centrosoma/efectos de los fármacos , Biología Computacional/métodos , Células HCT116 , Humanos , Espectrometría de Masas , Microscopía Confocal , Proteínas Mitocondriales , Simulación del Acoplamiento Molecular , Unión Proteica/efectos de los fármacos
6.
J Cell Physiol ; 234(7): 11511-11523, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30478982

RESUMEN

We have recently reported that type 2 diabetes promotes centrosome amplification via enhancing the expression, biding, and centrosome translocation of rho-associated coiled-coil containing protein kinase 1 (ROCK1)/14-3-3σ complex in HCT116 cells. In the functional proteomic study, we further investigated the molecular pathways underlying the centrosome amplification using HCT116 cells. We found that treatment of HCT116 cells with high glucose, insulin, and palmitic acid triggered the centrosome amplification and increased the expressions of proliferating cell nuclear antigen (PCNA), nucleophosmin (NPM), and 14-3-3σ. Individual knockdown of PCNA, NPM, or 14-3-3σ inhibited the centrosome amplification. Knockdown of PCNA inhibited the treatment-increased expression of ROCK1, whereas knockdown of ROCK1 did not affect the PCNA expression. High glucose, insulin, and palmitic acid also increased the expressions of c-Jun N-terminal kinase-1 (JNK1) and signal transducer and activator of transcription 3 (Stat3), individual knockdown of which upregulated the treatment-increased expression of 14-3-3σ and promoted the centrosome amplification. In contrast, overexpression of JNK1 inhibited the centrosome amplification. Knockdown of Stat3 enhanced the centrosome translocation of 14-3-3σ. Moreover, we showed that knockdown of JNK1 inhibited the treatment-increased expression of Stat3. Knockdown of PCNA, JNK1, or Stat3 did not have an effect on NPM and vice versa. In conclusion, our results suggest that PCNA and JNK1-Stat3 pathways respectively promotes and feedback inhibits the centrosome amplification by targeting at the ROCK1/14-3-3σ complex, and NPM serves as an independent signal for the centrosome amplification.


Asunto(s)
Proteínas 14-3-3/metabolismo , Biomarcadores de Tumor/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Exorribonucleasas/metabolismo , Proteína Quinasa 8 Activada por Mitógenos/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Factor de Transcripción STAT3/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteínas 14-3-3/genética , Biomarcadores de Tumor/genética , Centrosoma/metabolismo , Neoplasias del Colon , Exorribonucleasas/genética , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Proteína Quinasa 8 Activada por Mitógenos/genética , Factor de Transcripción STAT3/genética , Quinasas Asociadas a rho/genética
7.
J Cell Physiol ; 234(10): 18230-18248, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30883760

RESUMEN

There is evidence that cadmium can initiate carcinogenesis. However, the underlying mechanisms remain unknown. There is also evidence that moderate centrosome amplification can initiate tumorigenesis. The present study investigated whether cadmium could trigger cell centrosome amplification, and examined the underlying molecular mechanisms. We found that cadmium was able to cause cell centrosome amplification at the subtoxic concentrations, in a dose-dependent manner. It could cause centrosome amplification via the signaling of reactive oxygen species (ROS). Proteomic analysis revealed that cadmium caused differential expressions of three proteins, which included HSPA1A which is associated with endoplasmic reticulum (ER) stress. Western blot analysis confirmed that cadmium upregulated HSPA1A. Further analyses showed that cadmium upregulated Bip and decreased the phosphorylation of ASK1 as well as increased the phosphorylation of MKK7 and c-Jun N-terminal kinases (JNK). Knockdown of JNK2 using small interfering RNA inhibited the cadmium-induced centrosome amplification but not the level of ROS. N-acetylcysteine did not inhibit the cadmium-activated ER stress pathway. In conclusion, our results suggest that cadmium can induce cell centrosome amplification via ROS as well as ER stress through the Bip-TRAF2-ASK1-MKK7-JNK signaling route, in parallel. More studies are required to clarify whether centrosome amplification underlies cadmium-induced carcinogenesis.


Asunto(s)
Cadmio/farmacología , Centrosoma/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Línea Celular Tumoral , Células HCT116 , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Fosforilación/efectos de los fármacos , Proteómica/métodos , Regulación hacia Arriba/efectos de los fármacos
8.
J Cell Physiol ; 234(11): 20694-20703, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30989671

RESUMEN

We have recently published that type 2 diabetes can induce cell centrosome amplification due to the action of high glucose, palmitic acid, and insulin, and ROCK1 and 14-3-3σ are signal mediators. In this study, we further investigated the molecular mechanisms of the centrosome amplification in colon cancer HCT116 cells. Treatment of the cells with high glucose, palmitic acid, and insulin increased the expression of peroxisome proliferator-activated receptor γ (PPARγ) as well as the spindle and kinetochore associated protein 1 (SKA1), knockdown of each of which resulted in the inhibition of the treatment-triggered centrosome amplification. Knockdown of PPARγ inhibited the treatment-evoked increase in the SKA1 level, whereas knockdown of SKA1 did not modify the treatment-increased PPARγ level. We found a predicted binding site for PPARγ in the promoter region of the SKA1 gene from the JASPAR database. Experimental results showed that the treatment increased the messenger RNA level of SKA1, which could be inhibited by PPARγ chemical inhibitor or small interfering RNA. Moreover, we were able to show that PPARγ could bind to the binding site in the SKA1 gene promoter, which was increased by the experimental treatment. In conclusion, it is suggested that the pathophysiological factors in type 2 diabetes, high glucose, palmitic acid, and insulin, induce the cell centrosome amplification through the PPARγ-SKA1 pathway, in which PPARγ increases the expression of SKA1 via directly enhancing the SKA1 gene transcription.


Asunto(s)
Centrosoma/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , PPAR gamma/metabolismo , Animales , Centrosoma/efectos de los fármacos , Proteínas Cromosómicas no Histona/genética , Colon/metabolismo , Diabetes Mellitus Experimental , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Células HCT116 , Humanos , Insulina/farmacología , Ratones , PPAR gamma/genética , Ácido Palmítico/farmacología , Interferencia de ARN , ARN Interferente Pequeño
9.
Hereditas ; 156: 34, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31708719

RESUMEN

BACKGROUND: Cadmium (Cd) is a ubiquitous environmental toxicant for aquatic animals. The freshwater crab, Sinopotamon henanense (S. henanense), is a useful model for monitoring Cd exposure since it is widely distributed in sediments whereby it tends to accumulate several toxicants, including Cd. In the recent years, the toxic effects of Cd in the hepatopancreas of S. henanense have been demonstrated by a series of biochemical analysis and ultrastructural observations as well as the deep sequencing approaches and gene expression profile analysis. However, the post-transcriptional regulatory network underlying Cd toxicity in S.henanense is still largely unknown. RESULTS: The miRNA transcriptional profile of the hepatopancreas of S. henanense was used to investigate the expression levels of miRNAs in response to Cd toxicity. In total, 464 known miRNAs and 191 novel miRNAs were identified. Among these 656 miRNAs, 126 known miRNAs could be matched with the miRNAs of Portunus trituberculatus, Eriocheir sinensis and Scylla paramamosain. Furthermore, a total of 24 conserved miRNAs were detected in these four crab species. Fifty-one differentially expressed miRNAs were identified in the Cd-exposed group, with 31 up-regulated and 20 down-regulated. Eight of the differentially expressed miRNAs were randomly selected and verified by the quantitative real-time PCR (qRT-PCR), and there was a general consistency (87.25%) between the qRT-PCR and miRNA transcriptome data. A total of 5258 target genes were screened by bioinformatics prediction. GO term analysis showed that, 17 GO terms were significantly enriched, which were mainly related to the regulation of oxidoreductase activity. KEGG pathway analysis showed that 18 pathways were significantly enriched, which were mainly associated with the biosynthesis, modification and degradation of proteins. CONCLUSION: In response to Cd toxicity, in the hepatopancreas of S. henanense, the expressions of significant amount of miRNAs were altered, which may be an adaptation to resist the oxidative stress induced by Cd. These results provide a basis for further studies of miRNA-mediated functional adaptation of the animal to combat Cd toxicity.


Asunto(s)
Braquiuros/efectos de los fármacos , Braquiuros/genética , Cadmio/toxicidad , Hepatopáncreas/efectos de los fármacos , Hepatopáncreas/metabolismo , MicroARNs/genética , Animales , Braquiuros/metabolismo , Cadmio/metabolismo , Biología Computacional , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Ontología de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Transcriptoma
10.
Fish Shellfish Immunol ; 71: 177-190, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29017939

RESUMEN

Toll signaling is essential for expression of immune genes which are important for defense against bacterial, fungal and viral infections in invertebrates. Although several toll genes have been identified in the crustaceans, none of them has been investigated in freshwater crab Sinopotamon henanense. Moreover, the effect of cadmium (Cd) on toll gene expression has never been examined on the freshwater crabs which live in the sediments and are prone to heavy metal bioaccumulation. Our transcriptomic analysis of hepatopancreas tissue reveals that toll3 gene expression has been decreased when treated with Cd. In this study, we cloned one toll gene (hereby designated Shtoll3) from the crab. The full-length cDNA of Shtoll3 was 4488 bp, with an ORF of 3693 bp encoding a putative protein of 1230 amino acids, a 5'-untranslated region of 414 bp and a 3'-untranslated region of 781 bp. Phylogenetic analysis showed that ShToll3 was clustered into the group of DmToll8. The tissue distribution results showed that Shtoll3 was expressed widely in different tissues, with the highest in gills, and the lowest in hemocytes. Shtoll3 expression was down-regulated only in midguts after Aeromonas hydrophila infection. With Cd presence, Shtoll3 expression in response to A. hydrophila were up-regulated in midguts and gills, which was further confirmed by western blotting analysis. Moreover, the mRNA level of two antimicrobial peptides (AMPs) crustin and c-lys, which possibly responded to Cd and A. hydrophila stimulation through Shtoll3, were analysised. Thus, we conclude that Cd changes the susceptibility of Shtoll3 to A. hydrophila infection in gills and midguts. This suggest that Shtoll3 may contribute to the innate immune defense of S. henanense to A. hydrophila and Cd can modify the immune function in epithelium.


Asunto(s)
Braquiuros/genética , Braquiuros/inmunología , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/genética , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Secuencia de Aminoácidos , Animales , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Proteínas de Artrópodos/inmunología , Secuencia de Bases , Perfilación de la Expresión Génica , Filogenia , Alineación de Secuencia , Receptor Toll-Like 3/química
11.
Toxicol Lett ; 392: 84-93, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38185225

RESUMEN

Cadmium (Cd) is a prevalent heavy metal contaminant that can cause centrosome amplification (CA) and cancer. Since CA can initiate tumorigenesis, it is plausible that cadmium initiates tumorigenesis via CA. The present study investigated the signaling pathways underlying CA by Cd. Our findings confirmed that sub-toxic concentrations of Cd could induce CA in the HCT116 colon cancer cells, and revealed that reactive oxygen species (ROS), GCLM, CCDC85C and PLK4 were the signaling molecules that formed a pathway of ROS-GCLM-CCDC85C-PLK4. Cd not only increased the protein levels of CCDC85C and PLK4, but also promoted their distribution to the centrosomes. Molecular docking analysis revealed that CCDC85C and PLK4 had the binding potential. Indeed, antibodies against CCDC85C and PLK4 were able to pull down PLK4 and CCDC85C, respectively. Knockdown of CCDC85C decreased the Cd-promoted centrosomal distribution of PLK4. Similarly, knockdown of PLK4 reduced the centrosomal distribution of CCDC85C. Our results suggest that Cd activates ROS-GCLM pathway that triggers the expression of and binding between CCDC85C and PLK4, and promotes the translocation of CCDC85C-PLK4 complex to the centrosomes, which eventually leads to CA.


Asunto(s)
Cadmio , Neoplasias del Colon , Humanos , Cadmio/toxicidad , Especies Reactivas de Oxígeno/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Simulación del Acoplamiento Molecular , Centrosoma/metabolismo , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Carcinogénesis
12.
Toxicol Lett ; 380: 12-22, 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-36963620

RESUMEN

BACKGROUND: Hexavalent chromium can promote centrosome amplification (CA) as well as tumorigenesis. Since CA can lead to tumorigenesis, it is plausible that the chromium promotes the development of cancer via CA. In the present study, we investigated the signaling pathways of the chromium-induced CA. RESULTS: Our results showed that sub-toxic concentration of chromium was able to cause CA in HCT116 cells, and decrease the expression of TMOD2 and NPM2. Furthermore, TMOD2 and NPM2 interacted to each other via their C-terminal and the N-terminal, respectively, which was inhibited by the chromium. Overexpression of TMOD2 and NPM2 increased their binding and significantly attenuated the CA. Moreover, TMOD2 and NPM2 were co-localized with the centrosomes. The chromium inhibited the centrosomeal localization of NPM2, which was reversed by the overexpression of TMOD2, C-terminal of TMOD2, but not the N-terminal of NPM2. CONCLUSION: Our results suggest that the chromium induces CA via inhibiting the binding between TMOD2 and NPM2 as well as the dissociation of NPM2 from centrosomes.


Asunto(s)
Carcinogénesis , Centrosoma , Cromo , Humanos , Transformación Celular Neoplásica , Cromo/toxicidad
13.
Front Pharmacol ; 13: 982860, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36199691

RESUMEN

In the study, we investigated the anti-cancer effect of clinopodiside A and the underlying mechanisms using T24 bladder cancer cells as an experimental model. We found that the compound inhibited the growth of the bladder cancer cells in vitro and in vivo in a in a concentration- and dose-dependent manner, respectively, which showed a combinational effect when used together with cisplatin. In the bladder cancer cells, clinopodiside A caused autophagy, which was mediated by the signaling of BLK and RasGRP2, independently. Inhibition of the autophagy by chemical inhibitor 3-methyladenine or by the inhibition of the signaling molecules attenuated the cytotoxicity of clinopodiside A. Further analyses showed that clinopodiside A acted in synergism with cisplatin which itself could trigger both autophagy and apoptosis, which occurred with concomitant enhancements in autophagy and the cisplatin-evoked apoptosis. In conclusion, our results suggest that clinopodiside A inhibits the growth of the bladder cancer cells via BLK- and RasGRP2-mediated autophagy. The synergistic effect between clinopodiside A and cisplatin is attributed to the increases in autophagy and autophagy-promoted apoptosis. Clinopodiside A is a promising investigational drug for the treatment of cancer, at least blabber, which can be used alone or in combination with clinical drug(s).

14.
Biochim Biophys Acta Rev Cancer ; 1876(1): 188566, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33992724

RESUMEN

Accumulated evidence from genetically modified cell and animal models indicates that centrosome amplification (CA) can initiate tumorigenesis with metastatic potential and enhance cell invasion. Multiple human diseases are associated with CA and carcinogenesis as well as metastasis, including infection with oncogenic viruses, type 2 diabetes, toxicosis by environmental pollution and inflammatory disease. In this review, we summarize (1) the evidence for the roles of CA in tumorigenesis and tumor cell invasion; (2) the association between diseases and carcinogenesis as well as metastasis; (3) the current knowledge of CA in the diseases; and (4) the signaling pathways of CA. We then give our own thinking and discuss perspectives relevant to CA in carcinogenesis and cancer metastasis in human diseases. In conclusion, investigations in this area might not only identify CA as a biological link between these diseases and the development of cancer but also prove the causal role of CA in cancer and progression under pathophysiological conditions, potentially taking cancer research into a new era.


Asunto(s)
Transformación Celular Neoplásica/patología , Centrosoma/patología , Neoplasias/patología , Animales , Movimiento Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Centrosoma/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Invasividad Neoplásica , Metástasis de la Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Transducción de Señal
15.
Oncol Rep ; 46(1)2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34080666

RESUMEN

Type 2 diabetes increases the risk various types of cancer and is associated with a poor prognosis therein. There is also evidence that the disease is associated with cancer metastasis. Centrosome amplification can initiate tumorigenesis with metastasis in vivo and increase the invasiveness of cancer cells in vitro. Our previous study reported that type 2 diabetes promotes centrosome amplification via the upregulation and centrosomal translocation of Rho­associated protein kinase 1 (ROCK1), which suggests that centrosome amplification is a candidate biological link between type 2 diabetes and cancer development. In the present study, functional proteomics analysis was used to further investigate the molecular pathways underlying centrosome amplification by targeting ROCK1 binding partners. High glucose, insulin and palmitic acid were used to induce centrosome amplification, and immunofluorescent staining was employed to visualize centrosomal alterations. Combined with immunoprecipitation, mass spectrometry­based proteomics analysis was used to identify ROCK1 binding proteins, and protein complex disruption was achieved by siRNA­knockdown. In total, 1,148 ROCK1 binding proteins were identified, among which 106 proteins were exclusively associated with the treated samples, 193 were only associated with the control samples, and 849 were found in both the control and treated samples. Of the proteins with evidence of centrosomal localization, Dynactin subunit 2 (DCTN2) was confirmed to be localized to the centrosomes. Treating the cells with high glucose, insulin and palmitic acid increased the protein levels of ROCK1 and DCTN2, promoted their binding with each other, and triggered centrosome amplification. Disruption of the protein complex by knocking down ROCK1 or DCTN2 expression partially attenuated centrosome amplification, while simultaneous knockdown of both proteins completely inhibited centrosome amplification. These results suggested ROCK1­DCTN2 binding as a signal for the regulation of centrosome homeostasis, which is key for diabetes­associated centrosome amplification, and enriches our knowledge of centrosome biology. Therefore, the ROCK1­DCTN2 complex may serve as a target for inhibiting centrosome amplification both in research or future therapeutic development.


Asunto(s)
Centrosoma/metabolismo , Neoplasias del Colon/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Complejo Dinactina/metabolismo , Quinasas Asociadas a rho/metabolismo , Línea Celular Tumoral , Glucosa/farmacología , Células HCT116 , Humanos , Insulina/farmacología , Modelos Biológicos , Ácido Palmítico/farmacología , Unión Proteica , Proteómica , Regulación hacia Arriba
16.
Am J Cancer Res ; 11(12): 6004-6023, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35018239

RESUMEN

Patients with diabetes have increased risk of cancer and poor response to anti-cancer treatment. Increased protein synthesis is associated with endoplasmic reticulum (ER) stress which can trigger the unfolded protein response (UPR) to restore homeostasis, failure of which can lead to dysregulated cellular growth. We hypothesize that hyperglycemia may have legacy effect in promoting survival of cancer cells through dysregulation of UPR. Using HCT116 colorectal cancer cells as a model, we demonstrated the effects of high glucose (25 mM) on promoting cell growth which persisted despite return to normal glucose medium (5.6 mM). Using the Affymetrix gene expression microarray in HCT116 cells programmed by high glucose, we observed activation of genes related to cell proliferation and cell cycle progression and suppression of genes implicated in UPR including BiP and CHOP. These gene expression changes were validated in HCT116 cancer cells using quantitative real-time PCR and Western blot analysis. We further examined the effects of thapsigargin, an anti-cancer prodrug, which utilized ER stress pathway to induce apoptosis. High glucose attenuated thapsigargin-induced UPR and growth inhibition in HCT116 cells, which persisted despite return to normal glucose medium. Western blot analysis showed activation of caspase-3 in thapsigargin-treated cells in both normal and high glucose medium, albeit with lower levels of cleaved caspase-3 in cells exposed to high glucose, suggesting reduced apoptosis. Flow cytometry analysis confirmed fewer apoptotic cells under thapsigargin treatment in cells exposed to high glucose. Our results suggested that hyperglycemia altered gene expression involved in UPR with increased cell proliferation and facilitated survival of HCT116 cells under thapsigargin-induced ER stress by reducing the apoptotic response.

17.
Toxicology ; 423: 112-122, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31152847

RESUMEN

Cadmium (Cd) is one of the environmental pollutants, which has multiple toxic effects on fetuses and placentas. Placental fatty acid (FA) uptake and transport are critical for the fetal and placental development. We aimed to analyze the triglyceride (TG) level, the expression patterns of several key genes involved in FA uptake and transport, and the molecular mechanisms for the altered gene expressions in placentas in response to Cd treatment. Our results showed that the placental TG level was significantly decreased in the Cd-exposed placentas. Fatty acid transporting protein 1 (FATP1), FATP6 and fatty acid binding protein 3 (FABP3) were significantly down-regulated in the placentas from Cd-exposed mice. The expression level of phospho-p38 MAPK was increased by Cd treatment, while the protein level of total p38 MAPK remained unchanged. The expression levels of peroxisome proliferator-activated receptor-γ (PPAR-γ) and the hypoxia-inducible factor-1α (HIF-1α) were significantly decreased in the Cd-exposed placentas. The methylation levels of the promoter regions of FATP1, FATP6 and FABP3 showed no significant differences between the treatment and control groups. In addition, the circulating non-esterified fatty acid (NEFA), total cholesterol (TC), and TG levels were not decreased in the maternal serum from the Cd-exposed mice. Therefore, our results suggest Cd exposure dose not reduce the maternal FA supply, but reduces the placental TG level. Cd treatment also downregulates the placental expressions of FATP1, FATP6 and FABP3, respectively associated with p38-MAPK, p38 MAPK/PPAR-γ and HIF-1α pathways.


Asunto(s)
Cadmio/toxicidad , Proteínas de Transporte de Ácidos Grasos/metabolismo , Retardo del Crecimiento Fetal/metabolismo , Intercambio Materno-Fetal , Placenta/efectos de los fármacos , Animales , Línea Celular , Regulación hacia Abajo , Proteínas de Transporte de Ácidos Grasos/genética , Femenino , Retardo del Crecimiento Fetal/inducido químicamente , Retardo del Crecimiento Fetal/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones Endogámicos C57BL , PPAR gamma/genética , PPAR gamma/metabolismo , Ácido Palmítico/farmacología , Placenta/metabolismo , Embarazo , ARN Mensajero/metabolismo , Triglicéridos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
J Diabetes Res ; 2018: 3274084, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30271790

RESUMEN

Type 2 diabetes increases the risk for all-site cancers including colon cancer. Diabetic patients present typical pathophysiological features including an increased level of advanced glycation end products (AGEs), which comes from a series of nonenzymatic reactions between sugars and biological macromolecules, positively associated with the occurrence of diabetic complications. MDM2 is an oncogene implicated in cancer development. The present study investigated whether diabetes promoted MDM2 expression in colon cells and the underlying mechanisms. Our results showed that AGE increased the protein level of MDM2 in a cell model and promoted binding between MDM2 and Rb as well as p53, which led to degradation of Rb and p53. KLF5 was able to bind to the regulatory sequence of the MDM2 gene, and knockdown of the KLF5 protein level inhibited the AGE-triggered MDM2 overexpression, which indicated that KLF5 was the transcription factor for MDM2. In a mouse model of diabetes, we found that AGE level was increased in serum. The protein levels of both KLF5 and MDM2 were increased. KLF5 was able to bind to the regulatory sequence of the MDM2 gene. In conclusion, our results suggest that diabetes increases the level of AGE which enhances the expression of MDM2 via transcription factor KLF5 in colon cells. MDM2 overexpression is a candidate biological link between type 2 diabetes and colon cancer development.


Asunto(s)
Colon/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Productos Finales de Glicación Avanzada/farmacología , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Línea Celular Tumoral , Colon/citología , Colon/metabolismo , Citometría de Flujo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Proto-Oncogénicas c-mdm2/genética , Transducción de Señal/efectos de los fármacos
19.
Int J Biochem Cell Biol ; 39(3): 497-504, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17074529

RESUMEN

Diabetes mellitus is one of the most common non-communicable diseases, and if uncontrolled, targets multi-organ systems with serious debilitating and life-threatening sequela. Most diabetic cases fall under the Type 2 category, characterized by relatively late onset, development of insulin resistance and/or deficiency, and amyloidosis. Type 1 diabetes, on the other hand, manifests early during childhood and has an autoimmune component to it that causes a severe deficiency in the circulating levels of insulin. Despite the heterogeneity in etiology and clinical presentation, hyperglycemia is the most common metabolic abnormality in diabetic patients. At the molecular level, pancreatic beta-cell loss by apoptosis appears to play an important role in the development of insulin deficiency and the onset and/or progression of the disease. Here, we provide a short review on the apoptotic death circuitry in the pathogenesis of diabetes.


Asunto(s)
Apoptosis/fisiología , Diabetes Mellitus/patología , Autoinmunidad , Diabetes Mellitus/fisiopatología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/fisiopatología , Diabetes Mellitus Tipo 2/patología , Diabetes Mellitus Tipo 2/fisiopatología , Predisposición Genética a la Enfermedad , Humanos , Insulina/deficiencia , Células Secretoras de Insulina/patología , Células Secretoras de Insulina/fisiología , Mitosis , Modelos Biológicos
20.
Sci Rep ; 7(1): 15627, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-29142221

RESUMEN

Elemental selenium nanoparticles (SeNPs) have multiple biological activities. In this study, we investigated the protective effects of biogenic SeNPs (BioSeNPs) on CCl4-induced liver damage in mice. The results showed that: (i) when compared to sodium selenite (SS), BioSeNPs has a similar tissue distribution after intragastrical administration to mice; (ii) BioSeNPs and SS showed comparable efficacy in increasing the activities of glutathione peroxidase and thioredoxin reductase in liver cell lines, mice blood and liver; (iii) pretreatment with BioSeNPs inhibiting the elevation of activities of various enzymes significantly which included aspartate aminotransferase, alanine aminotransferase, alkaline phosphatase, lactate dehydrogenase and liver lipid peroxide (p < 0.05 or p < 0.01) in CCl4-treated mice; (iv) activities of antioxidant enzymes (superoxide dismutase, catalase, and glutathione peroxidase) were significantly increased (p < 0.05 or p < 0.01) after a pretreatment with BioSeNPs in CCl4-treated mice; (v) histopathological damages in the liver from CCl4-treated mice were ameliorated by a pretreatment with BioSeNPs. In conclusion, these results have shown that BioSeNPs is able to protect the liver from CCl4-induced hepatic damage via increasing the antioxidant capacity and inhibiting oxidative damage. BioSeNPs may have the potential to be used as a trace element food supplement inducing antioxidant bioactivities.


Asunto(s)
Intoxicación por Tetracloruro de Carbono/tratamiento farmacológico , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Nanopartículas del Metal/administración & dosificación , Selenio/administración & dosificación , Animales , Antioxidantes/administración & dosificación , Tetracloruro de Carbono/toxicidad , Intoxicación por Tetracloruro de Carbono/metabolismo , Intoxicación por Tetracloruro de Carbono/patología , Línea Celular , Enfermedad Hepática Inducida por Sustancias y Drogas/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Glutatión Peroxidasa/genética , Humanos , Nanopartículas del Metal/química , Ratones , Selenio/metabolismo , Selenito de Sodio/administración & dosificación , Reductasa de Tiorredoxina-Disulfuro/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA