Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cell Signal ; 108: 110691, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37116554

RESUMO

OBJECTIVE: To explore the role and mechanism of hyperforin (one of the active components of Sophora flavescens) in renal fibrosis. METHODS: The active compounds and target proteins of Sophora flavescens were first screened through TCMSP (https://tcmsp-e.com/). The renal fibrosis-related genes were analyzed through GeneCards (https://www.genecards.org/). The differentially expressed genes (DEGs) in renal fibrosis in GEO dataset GSE156181 were obtained. Metascape was applied for target protein enrichment analysis. TGF-ß1-stimulated renal tubular epithelial cells were used for renal fibrosis cell model establishment. The unilateral ureteral obstruction (UUO) mouse model was used for the renal fibrosis in vivo model. Cell viability was detected using an MTT assay. Immunofluorescence staining was employed to detect cell morphology changes and the expression of α-SMA and collagen I. Hematoxylin and eosin (H&E) and Masson staining were employed to determine the renal morphologic change. qRT-PCR or Western blotting was applied to determine the expression levels of the target proteins. RESULTS: After intersecting the analysis results of TCMSP, GeneCards, and dataset GSE156181, hyperforin targeting ICAM1 was identified. Metascape pathway enrichment analysis results revealed that the effective compounds of Sophora flavescens were tightly associated with extracellular matrix (ECM) remodeling and inflammatory response. MTT assay demonstrated that hyperforin had no toxic effect on cells. Immunofluorescence staining results evidenced that hyperforin could partially restore TGF-ß1-induced epithelial-mesenchymal transition (EMT), the PI3K/AKT pathway activation, and ICAM1 upregulation, and these effects of hyperforin could be reversed by ICAM1 overexpression. While the PI3K/AKT pathway activator IGF-1 effectively reversed the EMT inhibition effect of hyperforin on renal tubular epithelial cells. Moreover, the UUO mouse model further confirmed that hyperforin reduced renal fibrosis. CONCLUSION: Hyperforin inhibited renal fibrosis via the PI3K/AKT/ICAM1 axis.


Assuntos
Nefropatias , Obstrução Ureteral , Camundongos , Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Rim/patologia , Nefropatias/metabolismo , Fibrose , Obstrução Ureteral/metabolismo , Transição Epitelial-Mesenquimal/genética
2.
Cell Cycle ; 22(4): 433-449, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36196456

RESUMO

Severe hydronephrosis increases the risk of urinary tract infection and irretrievable renal fibrosis. While TGFß1-mediated fibrotic changes in proximal tubular epithelial cells and fatty acid oxidation (FAO) deregulation contribute to renal fibrosis and hydronephrosis. Firstly, a few elements were analyzed in this paper, including differentially-expressed long non-coding RNAs (lncRNAs), and miRNAs correlated to CPT1A, RXRA, and NCOA1. This paper investigated TGFß1 effects on lncRNA FABP5P3, CPT1A, RXRA, and NCOA1 expression and fibrotic changes in HK-2 cells and FABP5P3 overexpression effects on TGFß1-induced changes. Moreover, this paper predicted and proved that miR-22 binding to lncRNA FABP5P3, 3'UTR of CPT1A, RXRA, and NCOA1 was validated. The dynamic effects of the FABP5P3/miR-22 axis on TGFß1-induced changes were investigated. A Renal fibrosis model was established in unilateral ureteral obstruction (UUO) mice, and FABP5P3 effects were investigated. Eventually, this paper concluded that TGFß1 inhibited lncRNA FABP5P3, CPT1A, RXRA, and NCOA1 expression, induced fibrotic changes in HK-2 cells, and induced metabolic reprogramming within HK-2 cells, especially lower FAO. FABP5P3 overexpression partially reversed TGFß1-induced changes. miR-22 targeted lncRNA FABP5P3, CPT1A, RXRA, and NCOA1. LncRNA FABP5P3 counteracted miR-22 inhibition of CPT1A, NCOA1, and RXRA through competitive binding. TGFß1 stimulation induced the activation of TGFß/SMAD and JAG/Notch signaling pathways; Nocth2 knockdown reversed TGFß1 suppression on lncRNA FABP5P3. FABP5P3 overexpression attenuated renal fibrosis in unilateral ureteral obstruction mice. The LncRNA FABP5P3/miR-22 axis might be a potent target for improving the FAO deregulation and fibrotic changes in proximal TECs under TGFß1 stimulation.


Assuntos
Hidronefrose , Nefropatias , MicroRNAs , RNA Longo não Codificante , Obstrução Ureteral , Animais , Camundongos , Células Epiteliais/metabolismo , Ácidos Graxos/metabolismo , Fibrose , Hidronefrose/metabolismo , Hidronefrose/patologia , Nefropatias/metabolismo , MicroRNAs/genética , RNA Longo não Codificante/genética , Fator de Crescimento Transformador beta1/metabolismo , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia , Humanos
3.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 47(5): 555-561, 2022 May 28.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-35753725

RESUMO

Kidney stone is a disease with complex etiology and high incidence, and the most common chemical composition type of it is calcium oxalate stone. The formation of calcium oxalate stones includes crystal formation, crystal growth and aggregation, crystal interaction with renal tubular epithelial cells, and crystal invasion of renal interstitial extracellular matrix and so on. In these processes, crystal-cell interactions are essential for kidney crystal retention and kidney stone formation. Recently many studies have found that the interaction between crystal and renal tubular epithelial cells is closely related to various key binding molecules, endoplasmic reticulum stress of tubular cells, extracellular matrix proteins, and various lithotriptic drugs. Understanding the mechanism of crystal-cell interaction is of great significance for the prevention and early treatment of calcium oxalate stones.


Assuntos
Oxalato de Cálcio , Cálculos Renais , Oxalato de Cálcio/análise , Comunicação Celular , Células Epiteliais/metabolismo , Humanos , Cálculos Renais/química , Túbulos Renais/química , Túbulos Renais/metabolismo
4.
J Cell Mol Med ; 23(11): 7773-7784, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31496041

RESUMO

We previously identified testis developmental related gene 1 (TDRG1), a gene implicated in proliferation of TCam-2 seminoma cells. Recent evidence has revealed that autophagy influences the chemosensitivity of cancer cells to chemotherapy. However, whether TDRG1 protein regulates autophagy in seminoma cells and influences their sensitivity to cis-dichlorodiammine platinum (CDDP) remains unknown. In this study, we used TCam-2 cells and male athymic BALB/c nude mice with xenografts of TCam-2 cells to investigate autophagy, cell viability, apoptosis and the p110ß/Rab5/Vps34 (PI3-kinase Class III) pathway under the conditions of TDRG1 overexpression or knockdown and with or without CDDP treatment. We found that TDRG1 upregulation promoted autophagy in both TCam-2 cells and seminoma xenografts via p110ß/Rab5/Vps34 activation. Inhibition of autophagy reduced cell viability and promoted apoptosis during CDDP treatment of TCam-2 cells. Similarly, TDRG1 knockdown inhibited autophagy, reduced cell viability and promoted apoptosis during CDDP treatment of TCam-2 cells. TDRG1 knockdown inhibited tumour growth and promoted apoptosis in TCam-2 cell xenografts, whereas TDRG1 overexpression had the opposite effect. According to these results, we propose that high expression of TDRG1 promotes autophagy through the p110ß/Rab5/Vps34 pathway in TCam-2 cells. TDRG1 overexpression promotes autophagy and leads to CDDP resistance, whereas TDRG1 knockdown inhibits autophagy and promotes chemosensitivity to CDDP both in vivo and in vitro. This study has uncovered a novel role of TDRG1 in reducing chemoresistance during CDDP treatment and provides potential therapeutic strategies for the treatment of human seminoma.


Assuntos
Autofagia/genética , Cisplatino/farmacologia , Proteínas/metabolismo , Seminoma/genética , Seminoma/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Cisplatino/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas/genética , RNA Longo não Codificante , Seminoma/tratamento farmacológico , Transdução de Sinais/efeitos dos fármacos , Proteínas rab5 de Ligação ao GTP/genética
5.
Exp Mol Pathol ; 107: 110-117, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30594602

RESUMO

BACKGROUND: Renal cell carcinoma (RCC) is the most common form of kidney cancer. Recent studies reported that Tescalcin was overexpressed in various tumor types. However, the status of Tescalcin protein expression in RCC and its biological function is uncertain. This study was designed to investigate the expression of Tescalcin in human RCC and its biological function. METHODS: shRNA transfection was performed to abrogates the expression of Tescalcin. Quantitative real time PCR and western blotting assays were used to determine mRNA and protein expression levels, respectively. The cell viability was analyzed by MTT and colony formation. Cell flow cytometry was used to assess pHi value and cell apoptosis. Cell invasive and migratory ability was measured with modified Boyden chamber assay. Xenograft model was setup to evaluate tumor growth. RESULTS: Tescalcin was overexpressed in RCC tissues compared with matched normal tissues. It was also overexpressed in RCC cell lines relative that of normal cells. Suppression Tescalcin with specific shRNA resulted in the inhibition of cell proliferation, migration, invasion and apoptosis of RCC cells. Additionally, silencing of Tescalcin also caused the inhibition of the tumor growth in nude mice. Mechanistic study showed that Tescalcin regulated cell proliferation, migration and invasion via NHE1/pHi axis as well as AKT/NF-κB signaling pathway. CONCLUSIONS: These findings demonstrate that atopic expression of Tescalcin facilitates the survival, migration and invasion of RCC cells via NHE1/pHi axis as well as AKT/ NF-κB signaling pathway, providing new perspectives for the future study of Tescalcin as a therapeutic target for RCC.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Carcinoma de Células Renais/patologia , Neoplasias Renais/patologia , NF-kappa B/metabolismo , Trocador 1 de Sódio-Hidrogênio/metabolismo , Animais , Proliferação de Células/fisiologia , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica/fisiologia , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica/fisiopatologia , Transdução de Sinais/fisiologia
6.
Cancer Med ; 7(12): 6247-6257, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30430771

RESUMO

The role of TDRG1 in tumorigenesis and the progression of seminoma, as well as its role in regulating chemosensitivity of seminoma to cisplatin through the PI3K/Akt/mTOR signaling pathway, has been previously defined. However, the detailed mechanism underlying TDRG1 expression and concomitant chemoresistance conditions are unknown. Furthermore, it has been reported that non-protein-coding RNAs play an important role in a variety of vital processes including cellular chemosensitivity. However, the role of non-protein-coding RNAs in regulating the chemosensitivity of seminoma remains unknown. In this study, using microarray analysis, we found that long non-coding RNA H19 was upregulated while miRNA-106b-5p was downregulated in an established cisplatin-resistant TCam-2 cell line. Moreover, H19 acts as a miRNA-106b-5p sponge and thus impairs the function of miRNA-106b-5p on its target gene, TDRG1. Based on these findings, we propose that H19 promotes the expression of TDRG1 by sequestering miRNA-106b-5p and uses this mechanism to facilitate cell survival in cisplatin-based chemotherapeutic conditions. These findings elucidate the mechanisms, at least partially, applied to deregulate TDRG1 and cisplatin sensitivity, and may provide new therapeutic possibilities for chemoresistant seminoma.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , MicroRNAs , Proteínas/genética , RNA Longo não Codificante , Seminoma/genética , Neoplasias Testiculares/genética , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Seminoma/tratamento farmacológico , Neoplasias Testiculares/tratamento farmacológico
7.
Oncol Rep ; 38(3): 1373-1382, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28731173

RESUMO

Anacardic acid, which is commonly seen in plants of Anacardiaceae, is an important composition of cashew, ginkgo leaf and fruit, and it has been suggested in previous research to show antitumor activity. The main aim of the present study was to evaluate the anticancer effects of anacardic acid on cell apoptosis of prostatic cancer and molecular mechanisms of this phenomenon. In this study we found that anacardic acid inhibited cell proliferation, induced apoptosis and caspase-3/9 activities and Bax protein expression of prostatic cancer. Anacardic acid induced the ER stress inducing factors (BiP, CHOP, p-eIF2α), autophagy, LC3, Beclin-1, Atg 7 and DAPK3 protein expression, and suppressed p-Akt and p-mTOR protein expression of prostatic cancer. Si-CHOP was used to inhibit ER stress in prostatic cancer by anacardic acid, which showed that the cell proliferation was increased, apoptosis, and caspase-3/9 activities and Bax protein expression was suppressed, autophagy, LC3, Beclin-1, Atg 7 and DAPK3 protein expression was reduced, and p-Akt and p-mTOR protein expression was promoted. DAPK3 inhibited p-Akt and p-mTOR protein expression, enhanced the anticancer effects of anacardic acid on prostatic cancer through autophagy. For the first time, the present study showed that anacardic acid induces cell apoptosis of prostatic cancer through autophagy by ER stress/DAPK3/Akt signaling pathway.


Assuntos
Ácidos Anacárdicos/administração & dosagem , Proteínas Quinases Associadas com Morte Celular/genética , Neoplasias da Próstata/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/genética , Ácidos Anacárdicos/química , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Autofagia/genética , Proliferação de Células/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Transdução de Sinais/efeitos dos fármacos
8.
Cancer Biol Ther ; 17(7): 741-50, 2016 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-27104982

RESUMO

We previously identified TDRG1 (testis developmental related gene 1), a novel gene with exclusive expression in testis, promoted the proliferation and progression of cultured human seminoma cells through PI3K/Akt/mTOR signaling. As increasing evidence reveal that aberrant activation of this signaling is involved in cisplatin resistance. Then, in this study, we further explored whether TDRG1 regulated the chemosensitivity of seminoma TCam-2 cells to cisplatin. Our researches showed TDRG1 could regulate the viability of TCam-2 cells following cisplatin treatment in vitro through control of both cell apoptosis and cell cycle. Mechanistically, we observed TDRG1 positively regulated the expression levels of the key elements in PI3K/Akt/mTOR pathway including p-PI3K, p-Akt and p-mTOR and also affected the translocation of nuclear p-Akt in TCam-2 cells during cisplatin treatment. Meanwhile, the levels of Bad, cytochrome c, caspase-9 ratio (activated/total), caspase-3 ratio (activated/total) and cleaved-PARP were negatively modulated by TDRG1, which meant the involvement of mitochondria-mediated apoptotic pathway. Furthermore, we found the effect of TDRG1 knockdown or TDRG1 overexpression could be reversed by IGF-1, a PI3K signaling activator, or LY294002, a inhibitor of this pathway, respectively. Similar effects of TDRG1 on cisplatin chemosensitivity and associated molecular mechanism were also confirmed in vivo by employing xenograft assays. In addition, the positive correlation between TDRG1 and p-PI3K, or p-Akt, was found in tumor tissues from seminoma patients. In conclusion, we uncover that TDRG1 regulates chemosensitivity of TCam-2 cells to cisplatin through PI3K/Akt/mTOR signaling and mitochondria-mediated apoptotic pathway both in vitro and in vivo.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Imuno-Histoquímica/métodos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Seminoma/genética , Serina-Treonina Quinases TOR/metabolismo , Neoplasias Testiculares/química , Neoplasias Testiculares/genética , Animais , Antineoplásicos/farmacologia , Apoptose , Cisplatino/farmacologia , Humanos , Masculino , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Proteínas/metabolismo , RNA Longo não Codificante , Seminoma/patologia , Transdução de Sinais , Neoplasias Testiculares/patologia , Transfecção
9.
Onco Targets Ther ; 9: 409-20, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26855590

RESUMO

Human testis development-related gene 1 (TDRG1) is a recently identified gene that is expressed exclusively in the testes and promotes the development of testicular germ cell tumors. In this study, the role of TDRG1 in the development of testicular seminoma, which is the most common testicular germ cell tumor, was further investigated. Based on polymerase chain reaction, Western blotting, and immunohistochemistry tests, both gene and protein expression levels of TDRG1 were significantly upregulated in testicular seminoma tissues compared with normal testicular tissues. Additionally, the levels of phosphoinositide-3 kinase (PI3K)/p110 and Akt phosphorylation were dramatically upregulated in testicular seminoma tissues. Accordingly, in our cell experiment, seminoma TCam-2 cells were subjected to different treatments: the TDRG1 knockout, TDRG1 overexpression, PI3K inhibition (LY294002 administration), or PI3K activation (insulin-like growth factor-1 administration). Cell proliferation, the proliferation index, apoptosis rate, cell adhesive capacity, and cell invasion capability were assessed. Cells with both TDRG1 knockout and PI3K inhibition exhibited decreased cell proliferation, proliferation indexes, cell adhesion capacity, and cell invasion capability and increased apoptosis rates. Most of these effects were reversed by TDRG1 overexpression or PI3K activation, indicating that both TDRG1- and PI3K-mediated signaling promote proliferation and invasion of testicular seminoma cells. The knockout of TDRG1 significantly decreased the phosphorylation levels of PI3K/p85, PI3K/p110, Akt, and mammalian target of rapamycin (mTOR; Ser(2448)). Except for PI3K/p110, TDRG1 overexpression had the opposite effects on phosphorylation levels. Phosphorylated mTOR at Ser(2481) and Thr(2446) was not affected by TDRG1 or PI3K in our tests. Thus, these results indicate that TDRG1 promotes the development and migration of seminoma cells via the regulation of the PI3K/Akt/mTOR signaling pathway; this contributes to an understanding of the precise mechanisms underlying the development and migration of seminomas and lays a theoretical foundation for the development of appropriate therapies.

10.
Zhonghua Nan Ke Xue ; 21(7): 615-8, 2015 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-26333223

RESUMO

OBJECTIVE: To study the causes of orchiectomy in different age groups. METHODS: We retrospectively reviewed the clinical data about 291 cases of orchiectomy performed between March 1993 and October 2014 and analyzed the causes of surgery and their distribution in different age groups. RESULTS: The main causes of orchiectomy were testicular torsion (45.8%), cryptorchidism (32.5%) and testicular tumor (16.9%) in the patients aged 0-25 years, testicular tumor (42.4%), cryptorchidism (25.9%) and tuberculosis (10.6%) in those aged 26-50 years. Prostate cancer was the leading cause in those aged 51-75 years (77.6%) or older (84.0%)), and testicular tumor was another cause in the 51-75 years old men (10.2%). Prostate cancer, testicular tumor, cryptorchidism, and testicular torsion were the first four causes of orchiectomy between 1993 and 2009. From 2010 to 2014, however, testicular tumor rose to the top while prostate cancer dropped to the fourth place. CONCLUSION: The causes of orchiectomy vary in different age groups. The proportion of castration for prostate cancer patients significantly reduced in the past five years, which might be attributed to the improvement of comprehensive health care service.


Assuntos
Criptorquidismo/cirurgia , Orquiectomia , Neoplasias da Próstata/cirurgia , Torção do Cordão Espermático/cirurgia , Neoplasias Testiculares/cirurgia , Adolescente , Adulto , Fatores Etários , Idoso , Causalidade , Criança , Pré-Escolar , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Orquiectomia/estatística & dados numéricos , Estudos Retrospectivos , Tuberculose dos Genitais Masculinos/cirurgia , Adulto Jovem
11.
Oncol Lett ; 10(1): 61-66, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26170977

RESUMO

Testis developmental related gene 1 (TDRG1) is a novel human testis-specific gene. TDRG1 is differentially expressed in cancerous tissue compared with normal testicular tissue and demonstrates a unique expression pattern in normal testes; therefore, this gene may be involved in the occurrence and development of testicular germ cell tumors (TGCT). In the present study, the expression level of TDRG1 was downregulated in human TGCT NTERA-2 cells by RNA interference (RNAi) in order to investigate the association between TDRG1 and TGCT. The TDRG1 mRNA and protein expression levels in NTERA-2 cells were significantly inhibited following transfection with specific RNAi plasmids. The ability to proliferate (inhibited by 15.4% at day 3 and 26.1% at day 5; P<0.001) and invade (reduced by 49.1%; P=0.01) in vitro was suppressed in cells in which the expression level of TDRG1 was reduced, and a corresponding increase in the apoptotic potential was observed (the early apoptotic potential and total apoptotic potential were increased by 75%; P=0.019 and 54.8%; P=0.009, respectively). The results of the present study indicated that the biological behavior of NTERA-2 cells is associated with TDRG1 expression levels, and that this gene may be a novel target candidate in the treatment of TGCT.

12.
Exp Ther Med ; 9(3): 737-743, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25667621

RESUMO

Renal interstitial fibrosis (RIF) occurs as a result of chronic kidney disease (CKD) and is a common pathway leading to end-stage renal failure. Renal tissue hypoxia and ischemia are present during CKD. Adenosine (ADO) is an important signaling molecule induced under ischemic and hypoxic conditions. In the present study, the association between ADO and RIF was investigated using a mouse model, with the aim of obtaining important information relevant to the prevention and treatment of RIF. A unilateral ureteral obstruction (UUO) model of RIF was established in mice. A total of 44 male mice were randomly divided into sham, model and intervention groups, and samples were collected on days 1, 3, 7, and 14 after modeling. These were collected to detect hypoxia and changes in ADO concentration in obstructed renal tissue as well as to analyze the pathological changes and degree of RIF in the renal tissue. Changes in the levels of collagen deposition and profibrogenic factors in renal tissues were analyzed following intervention with an ADO receptor blocker. Following the UUO procedure, continuous hypoxia was present in the obstructed renal tissue, accompanied by an increased ADO concentration. Tubular injury and interstitial fibrosis progressively increased over time following the UUO procedure. The mRNA expression levels of tissue tumor growth factor ß1 (TGF-ß1) and α1(I) procollagen were significantly increased. Subsequent to the ADO pathway being blocked by 8-(p-sulfophenyl)-theophylline, tubular injury and interstitial fibrosis were reduced and the expression of related cytokines was decreased. Increased ADO levels were induced by hypoxia, causing the development of RIF. Following the blocking of the ADO pathway, renal damage was deferred and renal functions were protected.

13.
Med Oncol ; 32(3): 84, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25701463

RESUMO

Uroplakin 1A (UPK1A) is a specific marker of mammalian urothelium and one of major proteins contained in urothelial plaques. Many recent studies reported that UPK1A could be useful marker for diagnosis, detection and prognostic prediction of transitional cell carcinoma. However, relatively little is known about its exact roles in bladder transitional cell carcinoma (BTCC). We tried to explore the roles UPK1A plays in BTCC via the transfection of its antisense nucleotides (AS) into T24 cells to observe their changes of proliferation and apoptosis. After AS was successfully transfected into T24 cells, the percentages of proliferating T24 cells at 24 and 48 h after the treatment were 57.2 ± 6.8 and 44.7 ± 5.2%, significantly lower than that of control group, as shown by MTT (p < 0.05 and 0.01). At 24 h after transfection of AS, the percentage of apoptotic T24 cells was 26.87% measured by flow cytometry, significantly higher than that of control group (p < 0.01). Similarly, Hoechst 33258 staining showed that the percentage of apoptotic nuclei of T24 cells after 24 h treated by AS was 28.9%, significantly higher than that of control (p < 0.05). The most common and typical morphological changes of apoptosis, including shrink, pyknosis and karyorrhexis of T24 cells nuclei and DNA fragmentation were seen from Hoechst 33258 staining and DNA agarose gel electrophoresis. Taken together, inhibition of UPK1A can suppress proliferation and enhance apoptosis of BTCC T24 cells, suggesting it a potential target to treat this disease.


Assuntos
Carcinoma de Células de Transição/patologia , Neoplasias da Bexiga Urinária/patologia , Uroplaquina Ia/metabolismo , Apoptose/genética , Carcinoma de Células de Transição/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo , Citometria de Fluxo , Humanos , Oligonucleotídeos Antissenso/genética , Neoplasias da Bexiga Urinária/metabolismo , Uroplaquina Ia/genética
14.
Mol Med Rep ; 11(6): 4397-402, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25672943

RESUMO

Fibroblasts are the effector cells of collagen secretion in renal interstitial fibrosis (RIF), and their proliferation and activation are essential for the development of RIF. Hypoxic ischemia in local tissues has been identified in chronic kidney diseases (CKDs), with adenosine (ADO) as a key signaling molecule. The current study investigated the association between ADO and the biological behavior of renal fibroblasts by establishing an in vitro hypoxia cell model. This aimed to provide experimental evidence for the prevention and treatment of RIF. NIH3T3 fibroblasts were exposed to hypoxia, and the subtypes of the ADO receptor (AR) on the cell surface were identified by a TaqMan probe­based assay. Cells were divided into the following four groups: i) Control; ii) 5'­N­ethylcarboxamidoadenosine (NECA); iii) PT, NECA + 8­phenyltheophylline (PT); and iv) MRS, NECA + N­(4­cyanophenyl)­2­[4­(2,3,6,7­tetrahydro­2,6­dioxo­1,3­dipropyl­1H­purin­8­yl)phenoxy]­acetamide (MRS1754). The mRNA levels of transforming growth factor­ß1 (TGF­ß1), procollagen α1 (I) and α­smooth muscle actin (α­SMA) were measured following 24, 48, and 72 h of hypoxia. Cell proliferation was evaluated by a 3­(4,5­dimethylthiazol­2­yl)­2,5­diphenyltetrazolium bromide assay at 0, 12, 24, 48 and 72 h. The results demonstrated that A2BR was the predominant AR subtype present in hypoxia­stimulated fibroblasts. NECA significantly induced fibroblast proliferation and upregulated the expression of TGF­ß1, procollagen α1 (I) and α­SMA mRNA, while 8­PT and MRS1754 inhibited fibroblast proliferation and downregulated the expression of TGF­ß1, procollagen α1 (I) and α­SMA mRNA. The blockage of A2BR in hypoxia significantly inhibited the proliferation and activation of fibroblasts, and reduced the production of profibrotic cytokines, thus preventing the generation and development of fibrosis.


Assuntos
Fibroblastos/metabolismo , Hipóxia/metabolismo , Rim/citologia , Rim/metabolismo , Receptor A2B de Adenosina/metabolismo , Actinas/genética , Actinas/metabolismo , Animais , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Expressão Gênica , Hipóxia/genética , Camundongos , Células NIH 3T3 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor A2B de Adenosina/genética , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
15.
Int J Clin Exp Pathol ; 8(12): 15926-32, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26884865

RESUMO

Anacardic acid (6-pentadecylsalicylic acid, AA), a natural compound isolated from the traditional medicine Amphipterygiumadstringens, has been reported as potential antitumor agents in various cancers including prostate cancer (PC). However, the effects and mechanism of AA on the radiosensitivity of prostate cancer remains unknown. The results indicated that AA exhibited strong antitumor activity in PC cell lines, either as a single agentor in combination with radiation. AA significantly induced the downregulation of H2AX and p-H2AX expression, increase of cell apoptosis and decreasing of cell invasion, which were reversed by overexpressed H2AX. These results suggest that AA sensitize prostate cancer cells to radiation therapy by repressing H2AX expression.


Assuntos
Ácidos Anacárdicos/farmacologia , Histonas/metabolismo , Neoplasias da Próstata/radioterapia , Radiossensibilizantes/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Regulação para Baixo , Humanos , Masculino , Invasividade Neoplásica , Fosforilação , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia
16.
PLoS One ; 9(10): e109470, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25279731

RESUMO

Alterations of mitochondrial DNA (mtDNA) have been associated with the risk of a number of human cancers; however, the relationship between mtDNA copy number in peripheral blood leukocytes (PBLs) and the risk of prostate cancer (PCa) has not been investigated. In a case-control study of 196 PCa patients and 196 age-paired healthy controls in a Chinese Han population, the association between mtDNA copy number in PBLs and PCa risk was evaluated. The relative mtDNA copy number was measured using quantitative real-time PCR; samples from three cases and two controls could not be assayed, leaving 193 cases and 194 controls for analysis. PCa patients had significantly higher mtDNA copy numbers than controls (medians 0.91 and 0.82, respectively; P<0.001). Dichotomized at the median value of mtDNA copy number in the controls, high mtDNA copy number was significantly associated with an increased risk of PCa (adjusted odds ratio= 1.85, 95% confidence interval: 1.21-2.83). A significant dose-response relationship was observed between mtDNA copy number and risk of PCa in quartile analysis (Ptrend = 0.011). Clinicopathological analysis showed that high mtDNA copy numbers in PCa patients were significantly associated with high Gleason score and advanced tumor stage, but not serum prostate-specific antigen level (P = 0.002, 0.012 and 0.544, respectively). These findings of the present study indicate that increased mtDNA copy number in PBLs is significantly associated with an increased risk of PCa and may be a reflection of tumor burden.


Assuntos
Variações do Número de Cópias de DNA/genética , DNA Mitocondrial/genética , Leucócitos/patologia , Mitocôndrias/patologia , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Adenocarcinoma/genética , Adenocarcinoma/patologia , Idoso , Estudos de Casos e Controles , Seguimentos , Humanos , Leucócitos/metabolismo , Masculino , Mitocôndrias/metabolismo , Gradação de Tumores , Estadiamento de Neoplasias , Prognóstico , Reação em Cadeia da Polimerase em Tempo Real , Estudos Retrospectivos , Fatores de Risco , Carga Tumoral
17.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 38(11): 1135-40, 2013 Nov.
Artigo em Chinês | MEDLINE | ID: mdl-24316933

RESUMO

OBJECTIVE: To construct a p53-fused dual luciferase reporter and to test whether this reporter can mimic wild-type p53 activities in a high-throughput screen. METHODS: A restriction endonuclease site was added to each terminus and the stop codon of the wild-type full-length p53 open reading frame (ORF) was removed by PCR. A restriction endonuclease site was added to each terminus and the start codon of the firefly luciferase ORF was removed by PCR. The two modified ORFs were inserted upstream of the IRES-induced renilla luciferase ORF in a CMV-derived vector. The p53 fusion protein was expressed in cells to test its MDM2-mediated degradation, subcellular localization, and induction of p53-responsive promoter. RESULTS: The p53-fused dual luciferase reporter was successfully constructed. After transfection into the host cells, the reporter expressing the p53 fusion protein that was degraded by oncoprotein MDM2, was mainly located inside the nucleus, and induced the p53-responsive promoter, respectively. CONCLUSION: The p53-fused dual luciferase reporter (p53FL/IRES/RL) can identify modulators of P53 protein level in a high-throughput screen of genetic or chemical libraries.


Assuntos
Genes Reporter , Genes p53 , Vetores Genéticos , Humanos , Luciferases de Vaga-Lume , Luciferases de Renilla , Fases de Leitura Aberta , Regiões Promotoras Genéticas , Transfecção
18.
J Chemother ; 25(4): 229-38, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23816136

RESUMO

BACKGROUND: Few researches on increase of chemotherapy sensitivity by microRNA (miRNA) were reported. We aim to investigate exact role of miR-381 in chemotherapy sensitivity of 5-fluorouracil (5-FU) in renal cancer cells. METHODS: We investigated the cell survival, cell-cycle and apoptosis of 786-O and HK-2 cells treated with miR-381 and 5-FU. IC50 of 5-FU was calculated. To study apoptosis and G2/M arrest, we determined pHH3, mitotic index and caspase-3/7 activity. RESULTS: We showed that miR-381 combined with 5-FU inhibited proliferation and potentiated the anti-tumour efficacies of 5-FU at tolerated concentration in vitro. miR-381 combined with 5-FU led to Cdc2 activation, mitotic catastrophe, and cell apoptosis through inhibitory WEE1. WEE1 was also validated as the direct target of miR-381. IC50 of 5-FU decreased significantly in the presence of miR-381. CONCLUSION: miR-381 increases sensitivity of 786-O cells to 5-FU by inhibitory WEE1 and increase of Cdc2 activity.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Proteínas de Ciclo Celular/antagonistas & inibidores , Fluoruracila/farmacologia , Neoplasias Renais/tratamento farmacológico , MicroRNAs/farmacologia , Terapia de Alvo Molecular/métodos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptose/efeitos dos fármacos , Proteína Quinase CDC2 , Carcinoma de Células Renais/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Ciclina B/efeitos dos fármacos , Quinases Ciclina-Dependentes , Sinergismo Farmacológico , Fluoruracila/uso terapêutico , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Renais/genética , MicroRNAs/uso terapêutico , Proteínas Nucleares/genética , Proteínas Tirosina Quinases/genética , Regulação para Cima
19.
J Int Med Res ; 41(4): 964-74, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23867448

RESUMO

OBJECTIVE: To study the effects of methyl jasmonate on multidrug resistance in a mouse model of hepatocellular carcinoma. METHODS: Multidrug resistant H22 (H22/FAP) hepatocellular carcinoma cells were produced in vitro by continuous exposure to increasing doses of doxorubicin, cisplatin and 5-fluorouracil (FAP regimen). Cell toxicity was measured using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolum bromide (MTT) assay. Survival time was calculated for BALB/c mice that received intraperitoneal injections of H22/FAP cells followed by treatment with methyl jasmonate or verapamil in combination with FAP for 7 days. Adenosine triphosphate (ATP) hydrolysis was used to measure the activity of permeability-glycoprotein (P-gp) ATPase activity in plasma membranes. RESULTS: The MTT assay showed that methyl jasmonate significantly enhanced the cytotoxicity of the FAP regimen in multidrug resistant H22/FAP cells. Methyl jasmonate (10 mg/kg and 5 mg/kg) combined with FAP significantly increased survival time in BALB/c mice by 44.25% and 48.01%, respectively, compared with FAP. Methyl jasmonate increased P-gp ATPase activity. CONCLUSION: The combined use of methyl jasmonate and the FAP regimen might be a novel strategy for overcoming the multidrug resistance often observed in hepatocellular carcinoma.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Acetatos/farmacologia , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Ciclopentanos/farmacologia , Resistência a Múltiplos Medicamentos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Neoplasias Hepáticas/tratamento farmacológico , Oxilipinas/farmacologia , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino , Doxorrubicina , Sinergismo Farmacológico , Injeções Intraperitoneais , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais , Análise de Sobrevida , Tegafur , Uracila , Verapamil/farmacologia
20.
Clinics (Sao Paulo) ; 68(6): 825-33, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23778472

RESUMO

OBJECTIVES: MiRNAs are intrinsic RNAs that interfere with protein translation. Few studies on the synergistic effects of miRNAs have been reported. Both miR-424 and miR-381 have been individually reported to be involved in carcinogenesis. They share a common putative target, WEE1, which is described as an inhibitor of G2/M progression. Here, we studied the synergistic effects of miR-424 and miR-381 on renal cancer cells. METHODS: The viability of 786-O cells was analyzed after transfection with either a combination of miR-424 and miR-381 or each miRNA alone. We investigated cell cycle progression and apoptosis with flow cytometry. To confirm apoptosis and the abrogation of G2/M arrest, we determined the level of pHH3, which is an indicator of mitosis, and caspase-3/7 activity. The expression levels of WEE1, Cdc25, γH2AX, and Cdc2 were manipulated to investigate the roles of these proteins in the miRNA-induced anti-tumor effects. To verify that WEE1 was a direct target of both miR-424 and miR-381, we performed a dual luciferase reporter assay. RESULTS: We showed that the combination of these miRNAs synergistically inhibited proliferation, abrogated G2/M arrest, and induced apoptosis. This combination led to Cdc2 activation through WEE1 inhibition. This regulation was more effective when cells were treated with both miRNAs than with either miRNA alone, indicating synergy between these miRNAs. WEE1 was verified to be a direct target of each miRNA according to the luciferase reporter assay. CONCLUSIONS: These data clearly demonstrate that these two miRNAs might synergistically act as novel modulators of tumorigenesis by down-regulating WEE1 expression in renal cell cancer cells.


Assuntos
Carcinoma de Células Renais/genética , Proteínas de Ciclo Celular/metabolismo , Ciclina B/metabolismo , Neoplasias Renais/genética , MicroRNAs/farmacologia , Proteínas Nucleares/metabolismo , Proteínas Tirosina Quinases/metabolismo , Western Blotting , Proteína Quinase CDC2 , Linhagem Celular Tumoral , Fenômenos Fisiológicos Celulares , Transformação Celular Neoplásica , Quinases Ciclina-Dependentes , Regulação para Baixo , Citometria de Fluxo , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Tempo , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA