Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Biochem Cell Biol ; 98(6): 676-682, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33167678

RESUMO

Although the abundance of long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) in lung cancer has been well researched, the underlying mechanisms behind its effects were unknown. Here we investigated the molecular events regulating PVT1 in lung cancer. The pro-proliferative property of PVT1 was examined using a xenograft tumor model. Transwell chambers were used to analyze the impact of PVT1 expression on cell invasiveness and migration. In vivo metastasis was examined by tail-vein-injection in mice. Direct binding of miR-128 to PVT1 was investigated using a probe pulldown assay. The relative expression levels of miR-128 and PVT1 were quantified by real-time polymerase chain reaction and Western blotting. We show here that when PVT1 is amplified, there is a poor survival prognosis for patients with lung cancer. Elevated levels of PVT1 promoted lung cancer cell proliferation and metastasis, both in vitro and in vivo. Mechanistically, we found that PVT1 competes endogenously with miR-128 in the regulation of vascular endothelial growth factor C (VEGFC) expression, which is significantly associated with an unfavorable prognosis in lung cancer. We identified that copy number amplification significantly contributes to the high level of PVT1 transcripts in lung cancer, which promotes cell proliferation and metastatic behavior via modulating VEGFC expression by endogenous competition with miR-128.


Assuntos
Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias/biossíntese , RNA Longo não Codificante/metabolismo , RNA Neoplásico/metabolismo , Fator C de Crescimento do Endotélio Vascular/biossíntese , Células A549 , Animais , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Metástase Neoplásica , Proteínas de Neoplasias/genética , RNA Longo não Codificante/genética , RNA Neoplásico/genética , Fator C de Crescimento do Endotélio Vascular/genética
2.
Hepatology ; 67(5): 1807-1822, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29152756

RESUMO

Both activating and inactivating mutations in catenin ß1 (ctnnb1), which encodes ß-catenin, have been implicated in liver tumorigenesis in humans and mice, although the underlying mechanisms are not fully understood. Herein, we show that deletion of endogenous ß-catenin in hepatocytes aggravated hepatocellular carcinoma (HCC) development driven by an oncogenic version of ß-catenin (CAT) in combination with the hepatocyte growth factor receptor MET proto-oncogene receptor tyrosine kinase (MET). Although the mitogenic signaling and cell cycle progression was modestly impaired after CAT/MET transfection, the ß-catenin-deficient livers displayed changes in transcriptomes, increased DNA damage response, expanded Sox9+ cells, and up-regulation of protumorigenic cytokines, including interleukin-6 and transforming growth factor ß1. These events eventually exacerbated CAT/MET-driven hepatocarcinogenesis in ß-catenin-deficient livers, featured by up-regulation of extracellular signal-regulated kinase (Erk), protein kinase B (Akt), and Wnt/ß-catenin signaling and cyclin D1 expression. The resultant mouse tumors showed similar transcriptomes to human HCC samples with concomitant CTNNB1 mutations and MET overexpression. CONCLUSION: These data argue that while dominantly activating mutants of ß-catenin are oncogenic, inhibiting the oncogenic signaling pathway generates a pro-oncogenic microenvironment that may facilitate HCC recurrence following a targeted therapy of the primary tumor. An effective therapeutic strategy must require disruption of the oncogenic signaling in tumor cells and suppression of the secondary tumor-promoting stromal effects in the liver microenvironment. (Hepatology 2018;67:1807-1822).


Assuntos
Carcinoma Hepatocelular/genética , Hepatócitos/metabolismo , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogênicas c-met/genética , beta Catenina/genética , Animais , Carcinogênese/genética , Carcinoma Hepatocelular/metabolismo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Camundongos , Oncogenes , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas c-met/metabolismo , Transdução de Sinais , beta Catenina/metabolismo
3.
Biochem Cell Biol ; 96(1): 11-18, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29024600

RESUMO

Diabetic cardiomyopathy (DCM) is defined as ventricular dysfunction occurring independently of a recognized cause such as hypertension or coronary artery disease. Liver X receptor α (LXRα), a subtype of ligand-activated transcription factors LXRs, has been considered as a potential pharmacological target in the pathogenesis of cardiovascular and metabolic diseases. However, the potential mechanism of how LXRα is regulated in cardiomyocytes is still unclear. This study investigated the effect of activating LXRα with GW3965 on cardiomyocyte apoptosis and its upstream regulator in glucose-induced H9C2 cells. Our data indicated that GW3965 up-regulated the expression of LXRα, inhibited cardiomyocyte apoptosis, and altered the apoptosis-related proteins in glucose-induced H9C2 cells. In addition, GW3965 restored the mitochondrial membrane potential level and decreased the ROS production induced by glucose. Moreover, LXRα was confirmed as a direct target of microRNA-1 (miR-1) that was involved in cardiomyocyte apoptosis of DCM, and overexpression of miR-1 abrogated the inhibiting effect of GW3965 on glucose-induced apoptosis in H9C2 cells. This study highlights an important role of LXRα in the development of DCM and brings new insights into the complex mechanisms involved in the pathogenesis of DCM.


Assuntos
Apoptose/efeitos dos fármacos , Receptores X do Fígado/antagonistas & inibidores , MicroRNAs/farmacologia , Mitocôndrias/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Animais , Células Cultivadas , Receptores X do Fígado/metabolismo , MicroRNAs/genética , Mitocôndrias/metabolismo , Ratos
4.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 46(2): 321-5, 2015 Mar.
Artigo em Chinês | MEDLINE | ID: mdl-25924452

RESUMO

OBJECTIVE: To explore the risk factors for residual/recurrent disease of cervical intraepithelial neoplasia (CIN) 2 or worse after loop electrosurgical excision procedure (LEEP) and the timing point for postoperative follow-up. METHODS: 428 patients with CIN 2 or CIN 3 who were treated with LEEP were retrospectively reviewed. Postoperative follow-up was performed by Pap smear and human papillomavirus (HPV) hybrid capture 2 (HC2) testing. The definition of persistent/recurrent disease was biopsy-proven CIN 2 or worse. RESULTS: 296 patients were CIN 2 and 132 were CIN 3 among 428 patients. The positive rate of HPV HC2 before LEEP was 86.7% (371/428). During follow-up, 26 patients (6.1%) had residual/recurrent disease, the positive LEEP margin, especially the cone top status, was a significant risk factor for persistent/recurrent disease. Other factors such as age, HPV viral load [> or =100 relative light units (RLU)], and HPV typing (type 16/18 vs. other types) did not predict recurrence. HPV HC2 test at 3 months after LEEP can find all the residual/recurrent disease, the sensitivity and negative predictive value of the HPV HC2 test for residual/recurrent disease were both 100% at 3 and 6 months. CONCLUSION: The positive margin of LEEP specimen especially the cone top status was a significant risk factor for residual/recurrent disease after LEEP. HPV test at 3 months during follow-up can offer timely information about residual/recurrent disease and help for the risk control in treatment selection.


Assuntos
Conização , Recidiva Local de Neoplasia/diagnóstico , Neoplasia Residual/diagnóstico , Displasia do Colo do Útero/diagnóstico , Neoplasias do Colo do Útero/diagnóstico , Biópsia , Feminino , Humanos , Recidiva Local de Neoplasia/virologia , Neoplasia Residual/virologia , Teste de Papanicolaou , Papillomaviridae , Estudos Retrospectivos , Fatores de Risco , Neoplasias do Colo do Útero/cirurgia , Neoplasias do Colo do Útero/virologia , Esfregaço Vaginal , Carga Viral , Displasia do Colo do Útero/cirurgia , Displasia do Colo do Útero/virologia
5.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 46(3): 422-5, 462, 2015 May.
Artigo em Chinês | MEDLINE | ID: mdl-26121866

RESUMO

OBJECTIVE: To investigate the relationship of human papillomavirus (HPV) subtypes and multiple infections with different cervical precancerous diseases. METHODS: Retrospective study was done to review 1 226 patients with different cervical lesions who were pathologically diagnosed and scanned for HPV 23 subtypes with positive results from June 2006 to May 2012. These patients were divided into the following groups, chronic cervicitis, cervical condyloma, cervical intraepithelium neoplasia grade I (CIN I), grade II (CIN II), grade III (CINIII). RESULTS: There were significant differences in the proportion of HPV low risk types and high risk types between cervicitis, condyloma, CIN I group and CIN II + III groups (P<0. 05). HPV low risk types in condyloma group were mainly 6 and 11 subtype, while the other four groups were 42 and 43 subtype. The four most prevalence high risk types were 58, 16, 52,18 subtype. The infection rates of HPV16 were significant different in cervicitis (11. 0%), CIN II (20. 3%), and CIN III (20. 2%)(P<0. 01), and the infection rates of HPV58 was quite different between cervicitis (15. 9%) and CIN II (21. 4%) (P<0. 05). HPV multiple infection rate in condyloma (68. 8%) was significant different from that of cervicitis (23. 1%), CINI (26. 1%), CIN II (27. 8%) and CIN III (27. 1%) (P<0. 01); while the rest four groups were not significantly different (P>0. 05). CONCLUSION: There is a unique epidemiologic characteristic of HPV infection in Sichuan Province. The HPV low risk types were mainly 42 and 43, and high risk types were mainly 58, 16, 52, 18. It seems that HPV multiple infection is not the leading cause of progression of cervical disease.


Assuntos
Condiloma Acuminado/virologia , Papillomaviridae/classificação , Infecções por Papillomavirus/virologia , Displasia do Colo do Útero/virologia , Neoplasias do Colo do Útero/virologia , Cervicite Uterina/virologia , Carcinoma de Células Escamosas/virologia , China , Feminino , Papillomavirus Humano 16 , Humanos , Papillomaviridae/patogenicidade , Estudos Retrospectivos
6.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 45(5): 797-800, 2014 Sep.
Artigo em Chinês | MEDLINE | ID: mdl-25341343

RESUMO

OBJECTIVE: To study the expressions of c-myb and Bax proteins and their relationships with clinic-pathological characteristics in endometrial carcinoma. METHODS: The expressions of c-myb and Bax protein were detected by immunohistochemistry in 42 normal endometrial tissues and 68 endometrial carcinoma tissues. Apoptosis index was detected by TUNEL technique. RESULTS: The positive expression rates of c-myb and Bax in endometrial carcinoma were significantly higher than those in normal endometrial tissues (P < 0.05). The expressions of c-myb and Bax in low differentiated endometrial carcinoma were higher than those in high and middle differentiated endometrial carcinoma (P < 0.05). The expressions of c-myb and Bax in stage III and stage IV endometrial carcinoma were higher than those in stage I and stge II endometrial carcinoma (P < 0.05). Apoptosis index in endometrial carcinoma was higher than that in normal endometrial carcinoma. The poorer the cellular differentiation was, the higher the apoptosis index was. There was positive correlation between the expression of Bax and apoptosis index (P < 0.05), but no correlation between the expression of c-myb and apoptosis index (P > 0.05). CONCLUSION: Endometrial carcinoma has high expression of c-myb and Bax, and the expression status of both c-myb and Bax may be related to the malignant degree and prognosis of endometrial carcinoma.


Assuntos
Neoplasias do Endométrio/metabolismo , Proteínas Proto-Oncogênicas c-myb/metabolismo , Proteína X Associada a bcl-2/metabolismo , Apoptose , Diferenciação Celular , Feminino , Humanos , Imuno-Histoquímica , Prognóstico
7.
Front Pharmacol ; 15: 1290183, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38855750

RESUMO

Lung cancer is the leading cause of global cancer-related deaths. Platinum-based chemotherapy is the first-line treatment for the most common type of lung cancer, i.e., non-small-cell lung cancer (NSCLC), but its therapeutic efficiency is limited by chemotherapeutic resistance. Therefore, it is vital to develop effective therapeutic modalities that bypass the common molecular mechanisms associated with chemotherapeutic resistance. Ferroptosis is a form of non-apoptotic regulated cell death characterized by iron-dependent lipid peroxidation (LPO). Ferroptosis is crucial for the proper therapeutic efficacy of lung cancer-associated chemotherapies. If targeted as a novel therapeutic mechanism, ferroptosis modulators present new opportunities for increasing the therapeutic efficacy of lung cancer chemotherapy. Emerging studies have revealed that the pharmacological induction of ferroptosis using natural compounds boosts the efficacy of chemotherapy in lung cancer or drug-resistant cancer. In this review, we first discuss chemotherapeutic resistance (or chemoresistance) in lung cancer and introduce the core mechanisms behind ferroptosis. Then, we comprehensively summarize the small-molecule compounds sourced from traditional medicines that may boost the anti-tumor activity of current chemotherapeutic agents and overcome chemotherapeutic resistance in NSCLC. Cumulatively, we suggest that traditional medicines with ferroptosis-related anticancer activity could serve as a starting point to overcome chemotherapeutic resistance in NSCLC by inducing ferroptosis, highlighting new potential therapeutic regimens used to overcome chemoresistance in NSCLC.

8.
Oncogene ; 43(11): 776-788, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38243080

RESUMO

Rapid development of drug resistance after chemotherapy is a major cause of treatment failure in individuals with pancreatic ductal adenocarcinoma (PDAC). In this study, we illustrate that tumor-derived interleukin 35 (IL-35) mediates the accelerated resistance of PDAC to gemcitabine (GEM). We observe that GEM resistance can spread from GEM-resistant PDAC cells to GEM-sensitive cells, and that IL-35 is responsible for the propagation of chemoresistance, which is supported by sequencing and experimental data. Additionally, we discover that GEM-resistant cells have significantly higher levels of IL-35 expression. Mechanistically, aberrantly expressed IL-35 triggers transcriptional activation of SOD2 expression via GP130-STAT1 signaling, scavenging reactive oxygen species (ROS) and leading to GEM resistance. Furthermore, GEM treatment stimulates IL-35 expression through activation of the NF-κB pathway, resulting in acquired chemoresistance. In the mouse model, a neutralizing antibody against IL-35 enhances the tumor suppressive effect of GEM. Collectively, our data suggests that IL-35 is critical in mediating GEM resistance in pancreatic cancer, and therefore could be a valuable therapeutic target in overcoming PDAC chemoresistance.


Assuntos
Adenocarcinoma , Carcinoma Ductal Pancreático , Resistencia a Medicamentos Antineoplásicos , Interleucinas , Neoplasias Pancreáticas , Animais , Camundongos , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Gencitabina , Interleucinas/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo
9.
Sci Adv ; 10(25): eadj8650, 2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38896624

RESUMO

Pancreatic adenocarcinoma is the fourth leading cause of malignancy-related deaths, with rapid development of drug resistance driven by pancreatic cancer stem cells. However, the mechanisms sustaining stemness and chemotherapy resistance in pancreatic ductal adenocarcinoma (PDAC) remain unclear. Here, we demonstrate that Bicaudal C homolog 1 (BICC1), an RNA binding protein regulating numerous cytoplasmic mRNAs, facilitates chemoresistance and stemness in PDAC. Mechanistically, BICC1 activated tryptophan catabolism in PDAC by up-regulating indoleamine 2,3-dioxygenase-1 (IDO1) expression, a tryptophan-catabolizing enzyme. Increased levels of tryptophan metabolites contribute to NAD+ synthesis and oxidative phosphorylation, leading to a stem cell-like phenotype. Blocking BICC1/IDO1/tryptophan metabolism signaling greatly improves the gemcitabine (GEM) efficacy in several PDAC models with high BICC1 level. These findings indicate that BICC1 is a critical tryptophan metabolism regulator that drives the stemness and chemoresistance of PDAC and thus a potential target for combinatorial therapeutic strategy against chemoresistance.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Células-Tronco Neoplásicas , Neoplasias Pancreáticas , Triptofano , Triptofano/metabolismo , Humanos , Resistencia a Medicamentos Antineoplásicos/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/tratamento farmacológico , Linhagem Celular Tumoral , Animais , Camundongos , Regulação Neoplásica da Expressão Gênica , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Gencitabina , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/genética
10.
J Obstet Gynaecol Can ; 35(9): 816-822, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24099447

RESUMO

OBJECTIVE: To explore the impact of treatment modality on survival in patients with brain metastases from epithelial ovarian cancer. METHODS: We conducted a retrospective review of cases of ovarian cancer with brain metastases treated at institutions in three countries (Canada, China, and India) and conducted a search for studies regarding brain metastases in ovarian cancer reporting survival related to treatment modality. Survival was analyzed according to treatment regimens involving (1) some form of surgical excision or gamma-knife radiation with or without other modalities, (2) other modalities without surgery or gamma-knife radiation, or (3) palliation only. RESULTS: Twelve patients (mean age 56 years) with detailed treatment/outcome data were included; five were from China, four from Canada, and three from India. Median time from diagnosis of ovarian cancer to brain metastasis was 19 months (range 10 to 37 months), and overall median survival time from diagnosis of ovarian cancer was 38 months (13 to 82 months). Median survival time from diagnosis of brain metastasis was 17 months (1 to 45 months). Among patients who had multimodal treatment including gamma-knife radiotherapy or surgical excision, the median survival time after the identification of brain metastasis was 25.6 months, compared with 6.0 months in patients whose treatment did not include this type of focused localized modality (P = 0.006). Analysis of 20 studies also indicated that use of gamma-knife radiotherapy and excisional surgery in multi-modal treatment resulted in improved median survival interval (25 months vs. 6.0 months, P < 0.001). CONCLUSION: In the subset of patients with brain metastases from ovarian cancer, prolonged survival may result from use of multidisciplinary therapy, particularly if metastases are amenable to localized treatments such as gamma-knife radiotherapy and surgical excision.


Objectif : Explorer les effets de la modalité de traitement sur la survie chez les patientes qui présentent des métastases cérébrales attribuables au cancer épithélial de l'ovaire. Méthodes : Nous avons mené une analyse rétrospective des cas de cancer de l'ovaire donnant lieu à des métastases cérébrales qui ont été traités dans des établissements se situant dans trois pays (Canada, Chine et Inde); de plus, nous avons mené une recherche qui visait les études ayant traité des métastases cérébrales associées au cancer de l'ovaire qui faisaient mention des taux de survie liés aux modalités de traitement. La survie a été analysée en fonction de schémas de traitement mettant en jeu (1) une forme quelconque d'excision chirurgicale ou de radiochirurgie par scalpel gamma avec ou sans autres modalités, (2) d'autres modalités sans chirurgie ni radiochirurgie par scalpel gamma ou (3) des modalités palliatives seulement. Résultats : Douze patientes (âge moyen : 56 ans) comptant des données détaillées en ce qui concerne le traitement / les issues ont été admises à l'étude; cinq d'entre elles étaient de la Chine, quatre du Canada et trois de l'Inde. Le délai médian entre le diagnostic de cancer de l'ovaire et l'apparition de métastases cérébrales était de 19 mois (plage de 10 à 37 mois), et la durée de survie médiane globale à la suite du diagnostic de cancer de l'ovaire était de 38 mois (de 13 à 82 mois). La durée de survie médiane à la suite du diagnostic de métastases cérébrales était de 17 mois (de 1 à 45 mois). Chez les patientes ayant subi un traitement multimodal qui faisait appel à la radiochirurgie par scalpel gamma ou à l'excision chirurgicale, la durée de survie médiane à la suite de l'identification des métastases cérébrales était de 25,6 mois, par comparaison avec 6,0 mois chez les patientes dont le traitement ne faisait pas appel à ce type de modalité localisée ciblée (P = 0,006). L'analyse de 20 études a également indiqué que le recours à la radiochirurgie par scalpel gamma et à l'excision chirurgicale dans le cadre d'un traitement multimodal a donné lieu à une amélioration de l'intervalle de survie médian (25 mois vs 6,0 mois, P < 0,001). Conclusion : Dans le sous-ensemble des patientes qui présentent des métastases cérébrales attribuables au cancer de l'ovaire, le recours à un traitement multidisciplinaire pourrait mener à une prolongation de la survie, particulièrement lorsque les métastases se prêtent à des traitements localisés tels que la radiochirurgie par scalpel gamma et l'excision chirurgicale.


Assuntos
Neoplasias Encefálicas/secundário , Neoplasias Encefálicas/terapia , Neoplasias Epiteliais e Glandulares/patologia , Neoplasias Epiteliais e Glandulares/terapia , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Radiocirurgia , Carcinoma Epitelial do Ovário , Terapia Combinada , Feminino , Humanos , Pessoa de Meia-Idade , Estudos Retrospectivos , Análise de Sobrevida
11.
Arch Gynecol Obstet ; 288(1): 113-8, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23296464

RESUMO

OBJECTIVE: Screening programs based on cytology testing led to the incidence reduction of cervical cancer mortality of about 70-80 % in industrialized countries. However, these favorable results have not been replicated in developing areas. Thus, we aim to evaluate the efficacy of TruScreen (Polartechnics, Sydney, Australia) in detecting of precancerous lesions in comparison with cervical cytology test. METHODS: A total of 181 outpatients were screened by TruScreen using the pathological results as the gold standard. The medical records of cytological smear within 6 weeks were obtained from 169 of these participants. The reliability and yield of TruScreen and cytological smear were assessed. The screening results of TruScreen were compared with those obtained from the conventional smear. RESULTS: The sensitivities for histologically confirmed cervical intraepithelial neoplasia (CIN) lesions by TruScreen and Pap, were 67.4 % (95 % CI 53.4-81.5) and 87.9 % (95 % CI 76.7-99.0), respectively. The specificities for histologically confirmed CIN lesions by TruScreen and Pap, were 68.1 % (95 % CI 60.3-75.9) and 74.3 % (95 % CI 70.0-81.4), respectively. In contrast to Pap smear, TruScreen was comparatively efficacious in screening of cervical cancer (χ (2) = 0.0133, P = 0.9081). CONCLUSION: TruScreen is a potential test for initial cervical screening in developing world regions.


Assuntos
Detecção Precoce de Câncer/métodos , Lesões Pré-Cancerosas/diagnóstico , Displasia do Colo do Útero/diagnóstico , Neoplasias do Colo do Útero/diagnóstico , Adulto , Distribuição de Qui-Quadrado , Impedância Elétrica , Estudos de Viabilidade , Feminino , Fluorescência , Humanos , Pessoa de Meia-Idade , Imagem Óptica , Valor Preditivo dos Testes , Esfregaço Vaginal , Adulto Jovem
12.
Signal Transduct Target Ther ; 8(1): 449, 2023 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-38072908

RESUMO

Ferroptosis, a unique modality of cell death with mechanistic and morphological differences from other cell death modes, plays a pivotal role in regulating tumorigenesis and offers a new opportunity for modulating anticancer drug resistance. Aberrant epigenetic modifications and posttranslational modifications (PTMs) promote anticancer drug resistance, cancer progression, and metastasis. Accumulating studies indicate that epigenetic modifications can transcriptionally and translationally determine cancer cell vulnerability to ferroptosis and that ferroptosis functions as a driver in nervous system diseases (NSDs), cardiovascular diseases (CVDs), liver diseases, lung diseases, and kidney diseases. In this review, we first summarize the core molecular mechanisms of ferroptosis. Then, the roles of epigenetic processes, including histone PTMs, DNA methylation, and noncoding RNA regulation and PTMs, such as phosphorylation, ubiquitination, SUMOylation, acetylation, methylation, and ADP-ribosylation, are concisely discussed. The roles of epigenetic modifications and PTMs in ferroptosis regulation in the genesis of diseases, including cancers, NSD, CVDs, liver diseases, lung diseases, and kidney diseases, as well as the application of epigenetic and PTM modulators in the therapy of these diseases, are then discussed in detail. Elucidating the mechanisms of ferroptosis regulation mediated by epigenetic modifications and PTMs in cancer and other diseases will facilitate the development of promising combination therapeutic regimens containing epigenetic or PTM-targeting agents and ferroptosis inducers that can be used to overcome chemotherapeutic resistance in cancer and could be used to prevent other diseases. In addition, these mechanisms highlight potential therapeutic approaches to overcome chemoresistance in cancer or halt the genesis of other diseases.


Assuntos
Antineoplásicos , Ferroptose , Nefropatias , Pneumopatias , Neoplasias , Humanos , Ferroptose/genética , Processamento de Proteína Pós-Traducional/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Neoplasias/patologia , Metilação de DNA , Epigênese Genética/genética , Antineoplásicos/uso terapêutico , Pneumopatias/tratamento farmacológico , Pneumopatias/genética
13.
J Exp Clin Cancer Res ; 42(1): 238, 2023 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-37697370

RESUMO

BACKGROUND: Gemcitabine (GEM)-based chemotherapy is the first-line option for pancreatic ductal adenocarcinoma (PDAC). However, the development of drug resistance limits its efficacy, and the specific mechanisms remain largely unknown. RUNX1, a key transcription factor in hematopoiesis, also involved in the malignant progression of PDAC, but was unclear in the chemoresistance of PDAC. METHODS: Comparative analysis was performed to screen GEM-resistance related genes using our single-cell RNA sequencing(scRNA-seq) data and two public RNA-sequencing datasets (GSE223463, GSE183795) for PDAC. The expression of RUNX1 in PDAC tissues was detected by qRT-PCR, immunohistochemistry (IHC) and western blot. The clinical significance of RUNX1 in PDAC was determined by single-or multivariate analysis and survival analysis. We constructed the stably expressing cell lines with shRUNX1 and RUNX1, and successfully established GEM-resistant cell line. The role of RUNX1 in GEM resistance was determined by CCK8 assay, plate colony formation assay and apoptosis analysis in vitro and in vivo. To explore the mechanism, we performed bioinformatic analysis using the scRNA-seq data to screen for the endoplasm reticulum (ER) stress signaling that was indispensable for RUNX1 in GEM resistance. We observed the cell morphology in ER stress by transmission electron microscopy and validated RUNX1 in gemcitabine resistance depended on the BiP/PERK/eIF2α pathway by in vitro and in vivo oncogenic experiments, using ER stress inhibitor(4-PBA) and PERK inhibitor (GSK2606414). The correlation between RUNX1 and BiP expression was assessed using the scRNA-seq data and TCGA dataset, and validated by RT-PCR, immunostaining and western blot. The mechanism of RUNX1 regulation of BiP was confirmed by ChIP-PCR and dual luciferase assay. Finally, the effect of RUNX1 inhibitor on PDAC was conducted in vivo mouse models, including subcutaneous xenograft and patient-derived xenograft (PDX) mouse models. RESULTS: RUNX1 was aberrant high expressed in PDAC and closely associated with GEM resistance. Silencing of RUNX1 could attenuate resistance in GEM-resistant cell line, and its inhibitor Ro5-3335 displayed an enhanced effect in inhibiting tumor growth, combined with GEM treatment, in PDX mouse models and GEM-resistant xenografts. In detail, forced expression of RUNX1 in PDAC cells suppressed apoptosis induced by GEM exposure, which was reversed by the ER stress inhibitor 4-PBA and PERK phosphorylation inhibitor GSK2606414. RUNX1 modulation of ER stress signaling mediated GEM resistance was supported by the analysis of scRNA-seq data. Consistently, silencing of RUNX1 strongly inhibited the GEM-induced activation of BiP and PERK/eIF2α signaling, one of the major pathways involved in ER stress. It was identified that RUNX1 directly bound to the promoter region of BiP, a primary ER stress sensor, and stimulated BiP expression to enhance the reserve capacity for cell adaptation, which in turn facilitated GEM resistance in PDAC cells. CONCLUSIONS: This study identifies RUNX1 as a predictive biomarker for response to GEM-based chemotherapy. RUNX1 inhibition may represent an effective strategy for overcoming GEM resistance in PDAC cells.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Animais , Camundongos , Gencitabina , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Fatores de Iniciação de Peptídeos , Neoplasias Pancreáticas
14.
Cancer Biol Med ; 20(8)2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37381714

RESUMO

OBJECTIVE: Pancreatic ductal adenocarcinoma (PDAC) is a highly malignant gastrointestinal cancer with a 5-year survival rate of only 9%. Of PDAC patients, 15%-20% are eligible for radical surgery. Gemcitabine is an important chemotherapeutic agent for patients with PDAC; however, the efficacy of gemcitabine is limited due to resistance. Therefore, reducing gemcitabine resistance is essential for improving survival of patients with PDAC. Identifying the key target that determines gemcitabine resistance in PDAC and reversing gemcitabine resistance using target inhibitors in combination with gemcitabine are crucial steps in the quest to improve survival prognosis in patients with PDAC. METHODS: We constructed a human genome-wide CRISPRa/dCas 9 overexpression library in PDAC cell lines to screen key targets of drug resistance based on sgRNA abundance and enrichment. Then, co-IP, ChIP, ChIP-seq, transcriptome sequencing, and qPCR were used to determine the specific mechanism by which phospholipase D1 (PLD1) confers resistance to gemcitabine. RESULTS: PLD1 combines with nucleophosmin 1 (NPM1) and triggers NPM1 nuclear translocation, where NPM1 acts as a transcription factor to upregulate interleukin 7 receptor (IL7R) expression. Upon interleukin 7 (IL-7) binding, IL7R activates the JAK1/STAT5 signaling pathway to increase the expression of the anti-apoptotic protein, BCL-2, and induce gemcitabine resistance. The PLD1 inhibitor, Vu0155069, targets PLD1 to induce apoptosis in gemcitabine-resistant PDAC cells. CONCLUSIONS: PLD1 is an enzyme that has a critical role in PDAC-associated gemcitabine resistance through a non-enzymatic interaction with NPM1, further promoting the downstream JAK1/STAT5/Bcl-2 pathway. Inhibiting any of the participants of this pathway can increase gemcitabine sensitivity.


Assuntos
Adenocarcinoma , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Antimetabólitos Antineoplásicos/farmacologia , Antimetabólitos Antineoplásicos/uso terapêutico , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Gencitabina , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores de Interleucina-7/metabolismo , RNA Guia de Sistemas CRISPR-Cas , Fator de Transcrição STAT5/metabolismo , Fator de Transcrição STAT5/farmacologia , Neoplasias Pancreáticas
15.
Cell Biosci ; 12(1): 183, 2022 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-36371321

RESUMO

BACKGROUND: Exploiting cancer metabolism during nutrient availability holds immense potential for the clinical and therapeutic benefits of hepatocellular carcinoma (HCC) patients. Dietary methionine is a metabolic dependence of cancer development, but how the signal transduction integrates methionine status to achieve the physiological demand of cancer cells remains unknown. METHODS: Low or high levels of dietary methionine was fed to mouse models with patient-derived xenograft or diethyl-nitrosamine induced liver cancer. RNA sequence and metabolomics were performed to reveal the profound effect of methionine restriction on gene expression and metabolite changes. Immunostaining, sphere formation assays, in vivo tumourigenicity, migration and self-renewal ability were conducted to demonstrate the efficacy of methionine restriction and sorafenib. RESULTS: We discovered that mTORC1-c-Myc-SIRT4 axis was abnormally regulated in a methionine-dependent manner and affected the HCC progression. c-Myc rewires methionine metabolism through TRIM32 mediated degradation of SIRT4, which regulates MAT2A activity by ADP-ribosylation on amino acid residue glutamic acid 111. MAT2A is a key enzyme to generate S-adenosylmethionine (SAM). Loss of SIRT4 activates MAT2A, thereby increasing SAM level and dynamically regulating gene expression, which triggers the high proliferation rate of tumour cells. SIRT4 exerts its tumour suppressive function with targeted therapy (sorafenib) by affecting methionine, redox and nucleotide metabolism. CONCLUSIONS: These findings establish a novel characterization of the signaling transduction and the metabolic consequences of dietary methionine restriction in malignant liver tissue of mice. mTORC1, c-Myc, SIRT4 and ADP ribosylation site of MAT2A are promising clinical and therapeutic targets for the HCC treatment.

16.
Ann Transl Med ; 10(8): 471, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35571399

RESUMO

Background: Colposcopy is a critical component of cervical cancer screening services, but the accuracy of colposcopy varies greatly due to the lack of standardized training for colposcopists and pathologists. Thus, to improve the accuracy of colposcopy in the detection of cervical lesions intelligently is urgent. Here, we explored the sensitivity and specificity of a bioimpedance-based neural network algorithm in distinguishing normal and precancerous cervical tissues. Methods: Bioimpedance data were collected using a bioimpedance analyzer (Mscan1.0B, Sealand Technology, Chengdu, China) from the cervices of 102 female patients with abnormal cervical cytology (≥atypical squamous cells of undetermined significance) who required further colposcopy. Finally, the data of 106 samples from 37 patients were included, among which 85were used as the training set and 21 as the validation set. Using the biopsy pathology at each locus as the gold standard, the sensitivity, specificity, predictive value, likelihood ratio, and false positive and false negative rates of the bioimpedance-based neural network in identifying the normal and precancerous cervical tissues were calculated. Results: The bioimpedance method had a sensitivity of 0.90 [95% confidence interval (CI): 0.54 to 0.99], specificity of 0.82 (95% CI: 0.48 to 0.97), positive predictive value of 0.82 (95% CI: 0.48 to 0.97), and a negative predictive value of 0.90 (95% CI: 0.54 to 0.99) in distinguishing normal and precancerous cervical tissues. The Kappa value was 0.72. Conclusions: The bioimpedance method was an intelligent method with relative good sensitivity and specificity in distinguishing benign cervical tissue and precancerous lesions and can therefore be used as an adjunctive test to colposcopy to improve the detection of cervical lesions.

17.
J Exp Clin Cancer Res ; 40(1): 299, 2021 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-34551796

RESUMO

BACKGROUND: Mounting evidence has suggested the essential role of long non-coding RNAs (lncRNAs) in a plethora of malignant tumors, including hepatocellular carcinoma. However, the underlyling mechanisms of lncRNAs remain unidentified in HCC. The present work was aimed to explore the regulatory functions and mechanisms of LncRNA LNCAROD in HCC progression and chemotherapeutic response. METHODS: The expression of LNCAROD in HCC tissues and cell lines were detected by quantitative reverse transcription PCR (qPCR). Cancer cell proliferation, migration, invasion, and chemoresistance were evaluated by cell counting kit 8 (CCK8), colony formation, transwell, and chemosensitivity assays. Methylated RNA immunoprecipitation qRCR (MeRIP-qPCR) was used to determine N6-methyladenosine (m6A) modification level. RNA immunoprecipitation (RIP) and RNA pull down were applied to identify the molecular sponge role of LNCAROD for modulation of miR-145-5p via the competing endogenous RNA (ceRNA) mechanism, as well as the interaction between LNCAROD and serine-and arginine-rich splicing factor 3 (SRSF3). The interaction between insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1) and LNCAROD was also identified by RIP assay. Gain- or-loss-of-function assays were used to identify the function and underlying mechanisms of LNCAROD in HCC. RESULTS: We found that LNCAROD was significantly upregulated and predicted a poorer prognosis in HCC patients. LNCAROD upregulation was maintained by increased m6A methylation-mediated RNA stability. LNCAROD significantly promoted HCC cell proliferation, migration, invasion, and chemoresistance both in vitro and in vivo. Furthermore, mechanistic studies revealed that pyruvate kinase isoform M2 (PKM2)-mediated glycolysis enhancement is critical for the role of LNACROD in HCC. According to bioinformatics prediction and our experimental data, LNCAROD directly binds to SRSF3 to induce PKM switching towards PKM2 and maintains PKM2 levels in HCC by acting as a ceRNA against miR-145-5p. The oncogenic effects of LNCAROD in HCC were more prominent under hypoxia than normoxia due to the upregulation of hypoxia-triggered hypoxia-inducible factor 1α. CONCLUSIONS: In summary, our present study suggests that LNCAROD induces PKM2 upregulation via simultaneously enhancing SRSF3-mediated PKM switching to PKM2 and sponging miR-145-5p to increase PKM2 level, eventually increasing cancer cell aerobic glycolysis to participate in tumor malignancy and chemoresistance, especially under hypoxic microenvironment. This study provides a promising diagnostic marker and therapeutic target for HCC patients.


Assuntos
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Proteínas de Transporte/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Proteínas de Membrana/genética , RNA Longo não Codificante/genética , Hormônios Tireóideos/genética , Processamento Alternativo , Animais , Carcinoma Hepatocelular/mortalidade , Carcinoma Hepatocelular/patologia , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Modelos Animais de Doenças , Glicólise , Xenoenxertos , Humanos , Hipóxia/genética , Hipóxia/metabolismo , Neoplasias Hepáticas/mortalidade , Neoplasias Hepáticas/patologia , Proteínas de Membrana/metabolismo , Camundongos , MicroRNAs/genética , Prognóstico , Interferência de RNA , Hormônios Tireóideos/metabolismo , Proteínas de Ligação a Hormônio da Tireoide
18.
Front Cell Dev Biol ; 9: 642930, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33665193

RESUMO

The hypoxic microenvironment is beneficial to the metastasis but not to the proliferation of cancer cells. However, the mechanisms regarding to hypoxia differentially regulating cancer metastasis and proliferation are largely unknown. In this study, we revealed that hypoxia induced the expression of LIN28A at mRNA level but segregated LIN28A mRNAs in the P-bodies and thus inhibits the production of LIN28A protein. This unexpected finding suggests that there may be non-coding role for LIN28A mRNA in the progression of colon cancer. We further showed that the non-coding LIN28A mRNA promotes the metastasis but not proliferation of colon cancer cells in vitro and in vivo. Mechanistically, we revealed that methionyl aminopeptidase 2 (METAP2) is one of the up-regulated metastasis regulators upon over-expression of non-coding LIN28A identified by mass spectrum, and confirmed that it is non-coding LIN28A mRNA instead of LIN28A protein promotes the expression of METAP2. Moreover, we demonstrated that knockdown of DICER abolished the promotional effects of non-coding LIN28A on the metastasis and METAP2 expression. Conclusively, we showed that hypoxia induces the production of LIN28A mRNAs but segregated them into the P-bodies together with miRNAs targeting both LIN28A and METAP2, and then promotes the metastasis by positively regulating the expression of METAP2. This study uncovered a distinctive role of hypoxia in manipulating the metastasis and proliferation by differently regulating the expression of LIN28A at mRNA and protein level.

19.
Kaohsiung J Med Sci ; 36(6): 405-416, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32003536

RESUMO

Adipose tissue-derived mesenchymal stem cells (ADSCs) are multipotent cells that can differentiate into various cell types. This study aimed to investigate the effect of ghrelin on the neural differentiation of rat ADSCs and underlying molecular mechanisms. Rat ADSCs were isolated and third-passage ADSCs were used in this study. The isolated ADSCs were characterized by flow cytometry analysis for MSCs' surface expression markers as evidenced by positive for CD90, CD44, and CD29 and negative for CD34, CD45, and CD11b/2f/c. The multilineage differentiation of ADSCs was confirmed by adipogenic, osteogenic, and neural differentiation. After induction of neurogenesis, the differentiated cells were identified by development of neuron-like morphology and expression of neural markers including glial fibrillary acidic protein, Nestin, MAP2, and ß-Tubulin III using immunofluorescence and western blot. Ghrelin concentration dependently elevated the proportion of neural-like cells and branching dendrites, as well as upregulated the expression of neural markers. Further, the expression of nuclear ß-catenin, p-GSK-3ß, p-AKT, and p-mTOR was increased by ghrelin, indicating an activation of ß-catenin and AKT/mTOR signaling after the ghrelin treatment. Importantly, inhibition of ß-catenin or AKT/mTOR signaling suppressed ghrelin-induced neurogenesis. Therefore, we demonstrate that ghrelin promotes neural differentiation of ADSCs through the activation of ß-catenin and AKT/mTOR signaling pathways.


Assuntos
Adipócitos/efeitos dos fármacos , Grelina/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/genética , Serina-Treonina Quinases TOR/genética , beta Catenina/genética , Adipócitos/citologia , Adipócitos/metabolismo , Tecido Adiposo/citologia , Tecido Adiposo/metabolismo , Animais , Anticorpos Heterófilos/farmacologia , Diferenciação Celular/efeitos dos fármacos , Regulação da Expressão Gênica , Grelina/genética , Grelina/metabolismo , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Compostos Heterocíclicos com 3 Anéis/farmacologia , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Integrina beta1/genética , Integrina beta1/metabolismo , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Nestina/genética , Nestina/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , beta Catenina/antagonistas & inibidores , beta Catenina/metabolismo
20.
J Cancer Res Clin Oncol ; 145(6): 1387-1403, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31037399

RESUMO

PURPOSE: Aspirin, one of the most commonly used nonsteroidal anti-inflammatory drugs (NAIDS), not only shows cancer chemoprevention effects but also improves cancer therapeutic effects when combined with other therapies. Studies that focus on aspirin regulation of the hallmarks of cancer and the associated molecular mechanisms facilitate a more thorough understanding of aspirin in mediating chemoprevention and may supply additional information for the development of novel cancer therapeutic agents. METHODS: The relevant literatures from PubMed have been reviewed in this article. RESULTS: Current studies have revealed that aspirin regulates almost all the hallmarks of cancer. Within tumor tissue, aspirin suppresses the bioactivities of cancer cells themselves and deteriorates the tumor microenvironment that supports cancer progression. In addition to tumor tissues, blocking of platelet activation also contributes to the ability of aspirin to inhibit cancer progression. In terms of the molecular mechanism, aspirin targets oncogenes and cancer-related signaling pathways and activates certain tumor suppressors. CONCLUSION: Beyond a chemopreventive agent, aspirin is a master regulator of the hallmarks of cancer.


Assuntos
Aspirina/farmacologia , Neoplasias/tratamento farmacológico , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Anticarcinógenos , Metabolismo Energético , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/prevenção & controle , Ativação Plaquetária/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa