Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Biol Macromol ; 274(Pt 2): 133401, 2024 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-38925184

RESUMEN

Porcine epidemic diarrhea virus (PEDV) is one of the most devastating diseases affecting the pig industry globally. Due to the emergence of novel strains, no effective vaccines are available for prevention and control. Investigating the pathogenic mechanisms of PEDV may provide insights for creating clinical interventions. This study constructed and expressed eukaryotic expression vectors containing PEDV proteins (except NSP11) with a 3' HA tag in Vero cells. The subcellular localization of PEDV proteins was examined using endogenous protein antibodies to investigate their involvement in the viral life cycle, including endocytosis, intracellular trafficking, genome replication, energy metabolism, budding, and release. We systematically analyzed the potential roles of all PEDV viral proteins in the virus life cycle. We found that the endosome sorting complex required for transport (ESCRT) machinery may be involved in the replication and budding processes of PEDV. Our study provides insight into the molecular mechanisms underlying PEDV infection. IMPORTANCE: The global swine industry has suffered immense losses due to the spread of PEDV. Currently, there are no effective vaccines available for clinical protection. Exploring the pathogenic mechanisms of PEDV may provide valuable insights for clinical interventions. This study investigated the involvement of viral proteins in various stages of the PEDV lifecycle in the state of viral infection and identified several previously unreported interactions between viral and host proteins. These findings contribute to a better understanding of the pathogenic mechanisms underlying PEDV infection and may serve as a basis for further research and development of therapeutic strategies.

2.
Front Microbiol ; 15: 1380578, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38577683

RESUMEN

Introduction: Porcine epidemic diarrhea (PED) is an acute, highly contagious, and high-mortality enterophilic infectious disease caused by the porcine epidemic diarrhea virus (PEDV). PEDV is globally endemic and causes substantial economic losses in the swine industry. The PEDV E protein is the smallest structural protein with high expression levels that interacts with the M protein and participates in virus assembly. However, how the host proteins interact with E proteins in PEDV replication remains unknown. Methods: We identified host proteins that interact with the PEDV E protein using a combination of PEDV E protein-labeled antibody co-immunoprecipitation and tandem liquid-chromatography mass-spectroscopy (LC-MS/MS). Results: Bioinformatical analysis showed that in eukaryotes, ribosome biogenesis, RNA transport, and amino acid biosynthesis represent the three main pathways that are associated with the E protein. The interaction between the E protein and isocitrate dehydrogenase [NAD] ß-subunit (NAD-IDH-ß), DNA-directed RNA polymerase II subunit RPB9, and mRNA-associated protein MRNP 41 was validated using co-immunoprecipitation and confocal assays. NAD-IDH-ß overexpression significantly inhibited viral replication. Discussion: The antiviral effect of NAD-IDH-ß suggesting that the E protein may regulate host metabolism by interacting with NAD-IDH-ß, thereby reducing the available energy for viral replication. Elucidating the interaction between the PEDV E protein and host proteins may clarify its role in viral replication. These results provide a theoretical basis for the study of PEDV infection mechanism and antiviral targets.

3.
PLoS Pathog ; 20(3): e1012103, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38489378

RESUMEN

Alphacoronaviruses are the primary coronaviruses responsible for causing severe economic losses in the pig industry with the potential to cause human outbreaks. Currently, extensive studies have reported the essential role of endosomal sorting and transport complexes (ESCRT) in the life cycle of enveloped viruses. However, very little information is available about which ESCRT components are crucial for alphacoronaviruses infection. By using RNA interference in combination with Co-immunoprecipitation, as well as fluorescence and electron microscopy approaches, we have dissected the role of ALIX and TSG101 for two porcine alphacoronavirus cellular entry and replication. Results show that infection by two porcine alphacoronaviruses, including porcine epidemic diarrhea virus (PEDV) and porcine enteric alphacoronavirus (PEAV), is dramatically decreased in ALIX- or TSG101-depleted cells. Furthermore, PEDV entry significantly increases the interaction of ALIX with caveolin-1 (CAV1) and RAB7, which are crucial for viral endocytosis and lysosomal transport, however, does not require TSG101. Interestingly, PEAV not only relies on ALIX to regulate viral endocytosis and lysosomal transport, but also requires TSG101 to regulate macropinocytosis. Besides, ALIX and TSG101 are recruited to the replication sites of PEDV and PEAV where they become localized within the endoplasmic reticulum and virus-induced double-membrane vesicles. PEDV and PEAV replication were significantly inhibited by depletion of ALIX and TSG101 in Vero cells or primary jejunal epithelial cells, indicating that ALIX and TSG101 are crucial for PEDV and PEAV replication. Collectively, these data highlight the dual role of ALIX and TSG101 in the entry and replication of two porcine alphacoronaviruses. Thus, ESCRT proteins could serve as therapeutic targets against two porcine alphacoronaviruses infection.


Asunto(s)
Alphacoronavirus , Proteínas de Unión al Calcio , Virus de la Diarrea Epidémica Porcina , Animales , Alphacoronavirus/metabolismo , Línea Celular , Chlorocebus aethiops , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Células Epiteliales/metabolismo , Virus de la Diarrea Epidémica Porcina/metabolismo , Porcinos , Células Vero , Replicación Viral , Proteínas de Unión al Calcio/metabolismo
4.
Front Microbiol ; 14: 1273589, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37904874

RESUMEN

Porcine epidemic diarrhea (PED) is an enterophilic infectious disease caused by the porcine epidemic diarrhea virus (PEDV), which can lead to dehydration-like diarrhea in piglets with a mortality rate of up to 100%, causing huge economic losses to the global pig industry. In this study, we isolated two PEDV strains, FS202201 and JY202201, from diarrheal samples collected from two new PED outbreak farms in 2022. We performed phylogenetic analysis of the S gene and whole gene sequence. The effects of the different mutations on viral pathogenicity were investigated using piglet challenge experiments. The results showed that both strains belong to the G2c subtype, a widely prevalent virulent strain. Compared with FS202201, JY202201 harbored substitution and deletion mutations in nsp1. Both FS202201 and JY202201 infected piglets showed severe diarrhea and significant intestinal tissue lesions at an infection dose of 104 TCID50/mL, with a mortality rate of 50%; however, JY202201 required an additional day to reach mortality stabilization. An infection dose of 103 TCID50/mL reduced diarrhea and intestinal tissue lesions in piglets, with mortality rates of the two strains at 16.7% and 0%, respectively. In addition, PEDV was detected in the heart, liver, spleen, lungs, kidneys, mesenteric lymph nodes, stomach, large intestine, duodenum, jejunum, and ileum, with the highest levels in the intestinal tissues. In conclusion, this study enriches the epidemiology of PEDV and provides a theoretical basis for the study of its pathogenic mechanism and prevention through virus isolation, identification, and pathogenicity research on newly identified PED in the main transmission hub area of PEDV in China (Guangdong).

5.
J Virol ; 97(4): e0021023, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-36975780

RESUMEN

Porcine enteric alphacoronavirus (PEAV) is a new bat HKU2-like porcine coronavirus, and its endemic outbreak has caused severe economic losses to the pig industry. Its broad cellular tropism suggests a potential risk of cross-species transmission. A limited understanding of PEAV entry mechanisms may hinder a rapid response to potential outbreaks. This study analyzed PEAV entry events using chemical inhibitors, RNA interference, and dominant-negative mutants. PEAV entry into Vero cells depended on three endocytic pathways: caveolae, clathrin, and macropinocytosis. Endocytosis requires dynamin, cholesterol, and a low pH. Rab5, Rab7, and Rab9 GTPases (but not Rab11) regulate PEAV endocytosis. PEAV particles colocalize with EEA1, Rab5, Rab7, Rab9, and Lamp-1, suggesting that PEAV translocates into early endosomes after internalization, and Rab5, Rab7, and Rab9 regulate trafficking to lysosomes before viral genome release. PEAV enters porcine intestinal cells (IPI-2I) through the same endocytic pathway, suggesting that PEAV may enter various cells through multiple endocytic pathways. This study provides new insights into the PEAV life cycle. IMPORTANCE Emerging and reemerging coronaviruses cause severe human and animal epidemics worldwide. PEAV is the first bat-like coronavirus to cause infection in domestic animals. However, the PEAV entry mechanism into host cells remains unknown. This study demonstrates that PEAV enters into Vero or IPI-2I cells through caveola/clathrin-mediated endocytosis and macropinocytosis, which does not require a specific receptor. Subsequently, Rab5, Rab7, and Rab9 regulate PEAV trafficking from early endosomes to lysosomes, which is pH dependent. The results advance our understanding of the disease and help to develop potential new drug targets against PEAV.


Asunto(s)
Alphacoronavirus , Caveolas , Clatrina , Pinocitosis , Internalización del Virus , Proteínas de Unión al GTP rab , Alphacoronavirus/fisiología , Proteínas de Unión al GTP rab/metabolismo , Endosomas/metabolismo , Infecciones por Coronavirus/metabolismo , Concentración de Iones de Hidrógeno , Dinaminas/metabolismo , Caveolas/metabolismo , Colesterol/metabolismo , Clatrina/metabolismo , Pinocitosis/fisiología , Células Vero , Chlorocebus aethiops , Animales
6.
J Inorg Biochem ; 224: 111583, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34428638

RESUMEN

Inorganic mercury is a ubiquitous toxic pollutant in the environment. Exposure to inorganic mercury can cause various poisonous effects, including kidney injury. However, no safe and effective treatment for kidney injury caused by inorganic mercury has been found and used. Luteolin (Lut) possesses various beneficial bioactivities. Here, our research aims to investigate the protective effect of Lut on renal injury induced by mercury chloride (HgCl2) and identify the underlying autophagy regulation mechanism. Twenty-eight 6-8 weeks old Wistar rats were randomly assigned to four groups: control, HgCl2, HgCl2 + Lut, and Lut. We performed the determination of oxidative stress and renal function indicators, histopathological analysis, the terminal deoxynucleotidyl transferase-mediated deoxyuracil nucleoside triphosphate nick-end labeling assay to detect apoptosis, western blot detection of autophagy-related protein levels, and atomic absorption method to detect mercury content. Our results showed that Lut ameliorated oxidative stress, apoptosis and restored the autophagy and renal function caused by HgCl2 in rats. Concretely, the level of nuclear factor E2-related factor, renal adenosine monophosphate-activated protein kinase (AMPK) expression, and autophagy regulation-related proteins levels were down-regulated, and the mammalian target of rapamycin (mTOR) expression was up-regulated by HgCl2 treatment. However, Lut treatment reversed the above changes. Notably, Lut reduced the accumulation of HgCl2 in the kidneys and promoted the excretion of HgCl2 through urine. Collectively, our results demonstrate that Lut can attenuate inorganic mercury-induced renal injury via activating the AMPK/mTOR autophagy pathway. Therefore, Lut may be a potential biological medicine to protect against renal damage induced by HgCl2.


Asunto(s)
Autofagia/efectos de los fármacos , Riñón/lesiones , Luteolina/farmacología , Cloruro de Mercurio/toxicidad , Sustancias Protectoras/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis/efectos de los fármacos , Riñón/metabolismo , Masculino , Mercurio/toxicidad , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Serina-Treonina Quinasas TOR/metabolismo
7.
BMC Cancer ; 20(1): 840, 2020 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-32883234

RESUMEN

BACKGROUND: Genomic instability is a hallmark of cancer. The G1 checkpoint allows cells to repair damaged DNA that may lead to genomic instability. The high-risk human papillomavirus (HPV) E7 gene can abrogate the G1 checkpoint, yet the mechanism is still not fully understood. Our recent study showed that WDHD1 (WD repeat and high mobility group [HMG]-box DNA-binding protein 1) plays a role in regulating G1 checkpoint of E7 expressing cells. In this study, we explored the mechanism by which WDHD1 regulates G1 checkpoint in HPV E7 expressing cells. METHODS: NIKS and RPE1 derived cell lines were used. Real-time PCR, Rescue experiment, FACS and BrdU labeling experiments were performed to examine role of GCN5 in G1 checkpoint abrogation in HPV-16 E7 expressing cells. RESULTS: In this study, we observed that WDHD1 facilitates G1 checkpoint abrogation by modulating GCN5 in HPV E7 expressing cells. Notably, depletion of WDHD1 caused G1 arrest while overexpression of GCN5 rescued the inhibitory effects of WDHD1 knockdown on G1/S progression. Furthermore, siWDHD1 significantly decreased cell cycle proliferation and DNA synthesis that was correlated with Akt phosphorylation (p-Akt), which was reversed by GCN5 overexpression in HPV E7 expressing cells. CONCLUSIONS: In summary, our data identified a WDHD1/GCN5/Akt pathway leading to the abrogation of G1 checkpoint in the presence of damaged DNA, which may cause genomic instability and eventually HPV induced tumorigenesis.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/genética , Papillomavirus Humano 16/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Carcinogénesis/genética , Línea Celular , Proliferación Celular/genética , Daño del ADN/genética , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Inestabilidad Genómica/genética , Humanos , Infecciones por Papillomavirus/virología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transfección , Factores de Transcripción p300-CBP/genética
8.
Hum Reprod ; 35(6): 1377-1390, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32469403

RESUMEN

STUDY QUESTION: Do changes in tumor necrosis factor-α-induced protein 8 (TNFAIP8)-like 2 (TIPE2) levels in endometrium of patients with adenomyosis alter the proliferation, migration and invasion ability of endometrial cells? SUMMARY ANSWER: TIPE2 expression levels were low in eutopic and ectopic endometrium of adenomyosis patients, and TIPE2 inhibited the migration and invasion of endometrial cells, mainly by targeting ß-catenin, to reverse the epithelial-mesenchymal transition (EMT). WHAT IS KNOWN ALREADY: Adenomyosis is a benign disease, but it has some pathophysiological characteristics similar to the malignant tumor. TIPE2 is a novel negative immune regulatory molecule, and it also participates in the development of malignant tumors. STUDY DESIGN, SIZE, DURATION: Control endometrium (n = 48 women with non-endometrial diseases) and eutopic/ectopic endometrium from patients with adenomyosis (n = 50), human endometrial cancer cell lines, and primary endometrial cells from the eutopic endometrium of adenomyosis patients were used in the study. PARTICIPANTS/MATERIALS, SETTING, METHODS: The expression level of TIPE2 mRNA and protein in the eutopic/ectopic endometrial tissues of adenomyosis patients and control endometrium was determined by quantitative RT-PCR (qRT-PCR), western blot and immunohistochemistry. The effects of TIPE2 overexpression and knockdown on the proliferation, migration and invasion of endometrial cell lines and primary adenomyotic endometrial cells were determined using a cell counting kit-8, 5-ethynyl-2'-deoxyuridine assay, colony-forming assay, transwell migration assay and matrigel invasion assay. The expression of EMT-related markers and signal molecules was detected by western blot. The interaction between TIPE2 and ß-catenin was detected by co-immunoprecipitation and laser confocal microscopy. MAIN RESULTS AND THE ROLE OF CHANCE: The mRNA and protein expression levels of TIPE2 in the eutopic and ectopic endometrial tissues of adenomyosis patients were significantly downregulated compared with the control endometrium (P Ë‚ 0.01). TIPE2 could bind to ß-catenin and inhibit the nuclear translocation of ß-catenin, downregulate the expression of stromal cell markers, upregulate the expression of glandular epithelial cell markers, decrease the occurrence of epithelial-mesenchymal transition (EMT) and suppress the migration and invasion of endometrial cells (P Ë‚ 0.01). LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: In this study, the experiments were performed only in eutopic and ectopic endometrial tissues, endometrial cancer cell lines and primary adenomyotic endometrial cells. A mouse model of adenomyosis will be constructed to detect the effects of TIPE2 in vivo. WIDER IMPLICATIONS OF THE FINDINGS: These results suggest that TIPE2 is involved in the development of adenomyosis, which provides a potential new diagnostic and therapeutic strategy for the treatment of adenomyosis. STUDY FUNDINGS/COMPETING INTEREST(S): This present study was supported by grants from the National Natural Science Foundation of China (81471437, 81771554), Natural Science Foundation of Shandong (ZR2018MH013), Science and technology development plan provided by Health and Family Planning Committee in Shandong (2014-25). The authors declare that they have no conflicts of interest.


Asunto(s)
Adenomiosis , Endometriosis , China , Endometrio , Transición Epitelial-Mesenquimal , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , beta Catenina/genética
9.
Artículo en Inglés | MEDLINE | ID: mdl-31555603

RESUMEN

Condyloma acuminatum (CA) is a benign epithelium hyperplasia mainly caused by human papillomavirus (HPV), which is now the second most common viral sexually transmitted infection (STI) in China. In total, 90% of CA patients are caused by the low-risk HPV 6 and 11. Aside from low-risk HPV infection there are likely other factors within the local microenvironment that contribute to CA and there has been related research before. In this study, 62 vaginal specimens were analyzed using 16S rRNA gene sequencing. The diversity of the vaginal microbiota was higher and the composition was different with LR-HPV infection. While the relative abundance of dominant Firmicutes was lower, Actinobacteria, Proteobacteria, and Fusobacteria phyla were significantly higher; at the genus level Gardnerella, Bifidobacterium, Sneathia, Hydrogenophilus, Burkholderia, and Atopobium were higher. This study firstly confirmed a more accurate and comprehensive understanding of the relationship between low-risk HPV infection and vaginal microbiota, in order to provide a theoretical basis for further research on the occurrence and development of CA.


Asunto(s)
Microbiota/fisiología , Infecciones por Papillomavirus/microbiología , Vagina/microbiología , Adulto , Bacterias/clasificación , Bacterias/genética , Biodiversidad , Condiloma Acuminado/microbiología , Condiloma Acuminado/virología , ADN Bacteriano/genética , Femenino , Humanos , Microbiota/genética , Papillomaviridae , Infecciones por Papillomavirus/virología , ARN Ribosómico 16S/genética , Enfermedades de Transmisión Sexual/microbiología , Enfermedades de Transmisión Sexual/virología
10.
Oncol Rep ; 42(2): 849-856, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31233196

RESUMEN

Programmed cell death 4 (PDCD4) has been identified as a tumor­suppressor gene that inhibits neoplastic transformation, tumor progression, and protein translation. It has been reported that multiple factors participate in the regulation of PDCD4 mRNA and protein. The endometrium is regulated by changing concentrations of ovarian hormones, such as estrogen and progesterone, and shows periodical changes. However, whether ovarian hormones regulate PDCD4 expression remains unclear. This study aimed to explore the effect and mechanism of estrogen or progesterone on PDCD4 mRNA and protein expression in human endometrial cancer cells. The expression of PDCD4 mRNA and protein in Ishikawa and HEC­1­A cells was detected by quantitative real­time PCR and western blot analysis. The signaling pathway­related proteins were detected by western blot analysis. The results showed that PDCD4 mRNA levels exhibited no significant changes after treatment with estrogen or progesterone in both Ishikawa and HEC­1­A cell lines. Estrogen also had no obvious effect on PDCD4 protein expression. However, progesterone effectively decreased the expression of PDCD4 protein and the PI3K/AKT pathway may be involved in the downregulation of PDCD4 protein induced by progesterone. These results suggest that the downregulation of PDCD4 induced by progesterone affects the therapeutic efficacy of progesterone in human endometrial cancer or endometriosis, which may have important implications for progesterone treatment in the clinic.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Neoplasias Endometriales/tratamiento farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/química , Progesterona/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas de Unión al ARN/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Apoptosis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Humanos , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Progestinas/farmacología , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Células Tumorales Cultivadas
11.
Reprod Biol Endocrinol ; 15(1): 68, 2017 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-28851386

RESUMEN

BACKGROUND: Missed abortion is a common occurrence for otherwise healthy women. Immunological factor is one of the most important reasons. Tumor necrosis factor-α-induced protein-8 like-2 (TIPE2) is a novel negative immune regulator related to several human diseases. However, the expression level and clinical significance of TIPE2 in missed abortion remain unclear. METHODS: The expression of TIPE2 mRNA and protein in decidua and chorion from 36 missed abortion patients and 36 healthy controls was detected using quantitative real-time PCR, western blot and immunohistochemistry. In addition, serum TNF-ɑ and IL-10 levels were measured using flow cytometry. Serum estradiol and progesterone levels were measured by radioimmunoassay test. The correlations of TIPE2 protein levels with TNF-ɑ, IL-10, estradiol and progesterone were further analyzed. RESULTS: TIPE2 protein levels were significantly lower in decidual tissues of missed abortion patients than those in healthy controls. The patients with missed abortion had significantly higher levels of serum TNF-ɑ, and lower levels of serum IL-10, estradiol and progesterone compared with healthy controls. The TIPE2 protein levels were positively related to serum IL-10 levels. CONCLUSION: Our data indicate TIPE2 could play important roles in maintaining the maternal-fetal tolerance and decreased TIPE2 expression in the decidua may be related to the development of missed abortion.


Asunto(s)
Aborto Retenido/genética , Decidua/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Aborto Retenido/sangre , Aborto Retenido/diagnóstico , Adulto , Biomarcadores/sangre , Biomarcadores/metabolismo , Estudios de Casos y Controles , Estradiol/sangre , Femenino , Humanos , Interleucina-10/sangre , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Relaciones Materno-Fetales , Embarazo , Primer Trimestre del Embarazo/sangre , Primer Trimestre del Embarazo/genética , Pronóstico , Factor de Necrosis Tumoral alfa/sangre
12.
Springerplus ; 5(1): 988, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27398268

RESUMEN

Endometrial cancer is one of the most common malignancies in the female genital tract. Programmed cell death 5 (PDCD5) is a newly identified apoptosis related gene and plays an important role in the development of some human tumors. However, the expression and clinical significance of PDCD5 in endometrial cancer have not been fully elucidated. Here, we evaluated the expression of PDCD5 in endometrioid endometrial carcinoma and control endometrium by qRT-PCR, western blot and immunohistochemistry, and analyzed the associations of PDCD5 expression with clinicopathological parameters of patients. In addition, we detected the expression of PDCD5 in control endometrial glandular epithelial cells and endometrioid endometrial carcinoma-derived cell line KLE by immunocytochemistry. The results showed that PDCD5 protein mainly expressed in the cytoplasm of glandular epithelial cells and endometrial carcinoma cells, and there was a low level of PDCD5 expression in the nuclei of the above cells. Furthermore, PDCD5 protein level was significantly lower in endometrial carcinoma samples than that in control endometrium. The decreased PDCD5 expression was correlated with the tumor differentiation degree. It is clear that PDCD5 protein expression was lower in middle and low differentiated endometrial carcinoma compared with control endometrium and high differentiated endometrial carcinoma. However, there were no significant differences of PDCD5 expression between the proliferative phase and the secretory phase of control endometrium, as well as between high differentiated endometrial carcinoma and controls. The results were verified in control glandular epithelial cells and KLE cells by immunocytochemistry. Therefore, PDCD5 may play a key role in the pathogenesis of endometrial cancer and may be a novel target for diagnosis and treatment of endometrial cancer.

13.
Med Oncol ; 33(7): 73, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27277757

RESUMEN

Ovarian cancer is the leading cause of death among gynecological malignancies, and high grade serous ovarian carcinoma is the most common and most aggressive subtype. Recently, it was demonstrated that cAMP mediates protein kinase A-independent effects through Epac (exchange protein directly activated by cAMP) proteins. Epac proteins, including Epac1 and Epac2, are implicated in several diverse cellular responses, such as insulin secretion, exocytosis, cellular calcium handling and formation of cell-cell junctions. Several reports document that Epac1 could play vital roles in promoting proliferation, invasion and migration of some cancer cells. However, the expression levels and roles of Epac1 in ovarian cancer have not been investigated. In the present study, we detected the expression levels of Epac1 mRNA and protein in three kinds of ovarian cancer cells SKOV3, OVCAR3 and CAOV3. Furthermore, the effect of Epac1 knockdown on the proliferation and apoptosis of SKOV3 and OVCAR3 cells was evaluated in vitro and in vivo. The results showed that there was higher expression of Epac1 mRNA and protein in SKOV3 and OVCAR3 cells. Epac1 knockdown inhibited the proliferation of SKOV3 and OVCAR3 cells in vitro and in vivo. Decreased proliferation may be due to downregulation of Epac1-induced G1 phase arrest by inactivating the AKT/Cyclin D1/CDK4 pathway, but not to alterations in the MAPK pathway or to apoptosis. Taken together, our data provide new insight into the essential role of Epac1 in regulating growth of ovarian cancer cells and suggest that Epac1 might represent an attractive therapeutic target for treatment of ovarian cancer.


Asunto(s)
Quinasa 4 Dependiente de la Ciclina/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Neoplasias Ováricas/patología , Transducción de Señal/fisiología , Animales , Apoptosis/fisiología , Western Blotting , Línea Celular Tumoral , Proliferación Celular/fisiología , Ciclina D1/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Inmunohistoquímica , Técnicas In Vitro , Ratones , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA