Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Transplant ; 2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38642712

RESUMO

Immune checkpoint inhibitors (ICIs) as a downstaging or bridging therapy for liver transplantation (LT) in hepatocellular carcinoma patients are rapidly increasing. However, the evidence about the feasibility and safety of pre-LT ICI therapy is limited and controversial. To this end, a multicenter, retrospective cohort study was conducted in 11 Chinese centers. The results showed that 83 recipients received pre-LT ICI therapy during the study period. The median post-LT follow-up was 8.1 (interquartile range 3.3-14.6) months. During the short follow-up, 23 (27.7%) recipients developed allograft rejection, and 7 of them (30.4%) were diagnosed by liver biopsy. Multivariate logistics regression analysis showed that the time interval between the last administration of ICI therapy and LT (TLAT) ≥ 30 days was an independent protective factor for allograft rejection (odds ratio = 0.096, 95% confidence interval 0.026-0.357; P < .001). Multivariate Cox analysis showed that allograft rejection was an independent risk factor for overall survival (hazard ratio = 9.960, 95% confidence interval 1.006-98.610; P = .043). We conclude that patients who receive a pre-LT ICI therapy with a TLAT shorter than 30 days have a much higher risk of allograft rejection than those with a TLAT longer than 30 days. The presence of rejection episodes might be associated with higher post-LT mortality.

2.
Front Immunol ; 15: 1360955, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38633259

RESUMO

With a shortage of organs for transplant, the use of marginal donors can be an effective measure to meet the shortfall. Myelodysplastic syndromes (MDS) are considered an absolute contraindication for organ donation because of the high invasive potential. Currently, organ transplantation from donors with a past history of MDS has not been reported. In this paper, we report the successful clinical experience of one liver transplantation and two kidney transplantations, with organs donated by a 39-year-old patient diagnosed with a past history of MDS following intracranial hemorrhage. Four and a half years after transplantation, the three recipients are all doing well. However, it is still not clear to what extent organs donated by patients with a past history of MDS can be safely transplanted. This report provides support for the careful use of marginal donors. With effective treatment and full peer assessment, livers and kidneys from donors with a past history of MDS may be safely transplanted.


Assuntos
Transplante de Fígado , Síndromes Mielodisplásicas , Humanos , Adulto , Doadores de Tecidos , Rim , Fígado
3.
Hum Cell ; 37(2): 420-434, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38133876

RESUMO

Hypothermic machine perfusion (HMP) has been demonstrated to be more effective in mitigating ischemia-reperfusion injury (IRI) of donation after circulatory death (DCD) organs than cold storage (CS), yet the underlying mechanism remains obscure. We aimed to propose a novel therapeutic approach to ameliorate IRI in DCD liver transplantation. Twelve clinical liver samples were randomly assigned to HMP or CS treatment and subsequent transcriptomics analysis was performed. By combining in vivo HMP models, we discovered that HMP attenuated inflammation, oxidative stress, and apoptosis in DCD liver through a SEPRINA3-mediated PI3Kδ/AKT signaling cascade. Moreover, in the hypoxia/reoxygenation (H/R) model of BRL-3A, overexpression of SERPINA3 mitigated H/R-induced apoptosis, while SERPINA3 knockdown exacerbated cell injury. Idelalisib (IDE) treatment also reversed the protective effect of SERPINA3 overexpression. Overall, our research provided new insights into therapeutic strategies and identified potential novel molecular targets for therapeutic intervention against DCD liver.


Assuntos
Transplante de Fígado , Traumatismo por Reperfusão , Serpinas , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fígado/metabolismo , Perfusão , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/metabolismo , Serpinas/metabolismo
4.
Oncol Rep ; 50(1)2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37232371

RESUMO

Following the publication of this paper, it was drawn to the Editors' attention by a concerned reader that certain of the tumor images shown in Fig. 4G and H were strikingly similar to tumor images (albeit oriented differently) which had previously appeared in Fig. 8A in another article published in the journal International Journal of Oncology [Tang B, Li Y, Yuan S, Tomlinson S and He S: Upregulation of the δ opioid receptor in liver cancer promotes liver cancer progression both in vitro and in vivo. Int J Oncol 43: 1281­1290, 2013], indicating that results which were purported to have been obtained under different experimental conditions had been derived from the same original source. In view of the fact that these data had already appeared in another publication prior to its submission to Oncology Reports, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a satisfactory reply. The Editor apologizes to the readership for any inconvenience caused. [Oncology Reports 41: 43­56, 2019; DOI: 10.3892/or.2018.6825].

5.
Talanta ; 261: 124669, 2023 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-37210917

RESUMO

Alpha-fetoprotein (AFP) is the best diagnostic marker for hepatocellular carcinoma (HCC) and plays an important role in the general surveillance of the population. Therefore, the establishment of an ultra-sensitive AFP assay is essential for the early screening and clinical diagnosis of HCC. In this work, we designed a signal-off biosensor for ultra-sensitive detection of AFP based on an electrochemiluminescent resonance energy transfer (ECL-RET) strategy using luminol intercalated layered bimetallic hydroxide (Luminol-LDH) as an ECL donor and Pt nanoparticles-grown on copper sulfide nanospheres (CuS@Pt) as ECL acceptor. The (Au NPs/Luminol-LDH)n multilayer nanomembrane synthesized by our intercalation and layer-by-layer electrostatic assembly process not only effectively immobilizes luminol but also significantly enhances the ECL signal. The CuS@Pt composite has well visible light absorption ability and can burst the light emitted from luminol by ECL-RET. The biosensor showed good linearity in the range from 10-5 ng mL-1 to 100 ng mL-1 and a minimum detection limit of 2.6 fg mL-1. Therefore, the biosensor provides a novel and efficient strategy for the detection of AFP, which is important for the early screening and clinical diagnosis of HCC.


Assuntos
Técnicas Biossensoriais , Carcinoma Hepatocelular , Neoplasias Hepáticas , Nanopartículas Metálicas , Humanos , Luminol , alfa-Fetoproteínas , Carcinoma Hepatocelular/diagnóstico , Neoplasias Hepáticas/diagnóstico , Medições Luminescentes , Transferência de Energia , Técnicas Eletroquímicas , Limite de Detecção , Ouro
6.
Molecules ; 28(5)2023 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-36903458

RESUMO

Hepatocellular carcinoma (HCC) is the fourth most common cause of cancer-related deaths in the world. However, there are currently few clinical diagnosis and treatment options available, and there is an urgent need for novel effective approaches. More research is being undertaken on immune-associated cells in the microenvironment because they play a critical role in the initiation and development of HCC. Macrophages are specialized phagocytes and antigen-presenting cells (APCs) that not only directly phagocytose and eliminate tumor cells, but also present tumor-specific antigens to T cells and initiate anticancer adaptive immunity. However, the more abundant M2-phenotype tumor-associated macrophages (TAMs) at tumor sites promote tumor evasion of immune surveillance, accelerate tumor progression, and suppress tumor-specific T-cell immune responses. Despite the great success in modulating macrophages, there are still many challenges and obstacles. Biomaterials not only target macrophages, but also modulate macrophages to enhance tumor treatment. This review systematically summarizes the regulation of tumor-associated macrophages by biomaterials, which has implications for the immunotherapy of HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Macrófagos Associados a Tumor , Materiais Biocompatíveis , Macrófagos , Microambiente Tumoral
7.
Front Genet ; 13: 1065693, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36479252

RESUMO

Background: The inadequate early detection strategies makes hepatocellular carcinoma (HCC) patients with poor prognisis. Therefore, more effective detection methods are urgently needed for early detection and early intervention of HCC. Methods: 17 cases of suspected HCC patients and 11 cases of HBV-related decompensated cirrhosis (HBV-DeCi) patients were enrolled. For each patient, 5 ml blood sample was separated into circulating tumor cells (CTCs) and plasma, CTCs were stained with Diff staining for counting. Plasma was used for extracting cell free DNA (cfDNA) and then analyzed by qMSP assay. Ct values were recorded for GNB4 and Riplet as target genes and ß-actin as an endogenous reference gene. Finally, clinical efficacy of CTC count combined with GNB4/Riplet methylation detection for early diagnosis of HCC was analyzed. Results: The CTC of HCC patients has pleomorphic characteristics, but it is difficult to distinguish from other blood cells with non-obviously pleomorphic of CTC. Although a small number of CTCs can also be detected in HBV-DeCi patients (control group), the number is significantly lower than that in HCC patients, the sensitivity and specificity of CTC for HCC detection were 70.6% and 90.9% (AUC = 0.81). The Ct values of GNB4 and Riplet methylation were significantly different between HCC patients and control group patients. When CTC combined with two genes, the AUC value was significantly increased to 0.98, the sensitivity was 88.2%, and the specificity was 100%. Conclusion: Our study has developed a novel test that CTC count combined with GNB4/Riplet methylation detection and showed its high performance for early diagnosis of HCC.

9.
Front Cell Dev Biol ; 9: 781867, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34858996

RESUMO

Background: Hepatocellular carcinoma (HCC) cells exhibit the stemness property, which makes the patient with HCC prone to tumor recurrence and metastasis. Despite the prominent regulatory role of long non-coding RNAs (lncRNAs) in tumor stemness, the roles and molecular mechanisms of LINC00106 in HCC are poorly understood. Methods: LINC00106, let7f and periostin expression levels in tissue specimens and cell lines were assessed through qRT-PCR and immunohistochemistry (IHC). Various in vivo and in vitro assays, namely sphere/colony formation, proportion of side population cells (SP%), invasion, migration, western blot, and murine xenograft model were employed for assessing the stemness and metastatic properties of HCC cells. Luciferase reporter assays, RNA-seq, RNA pull-down, RNA immunoprecipitation (RIP) were conducted to clarificate the target gene and analyze the underlying mechanisms. Results: LINC00106 was prominently upregulated in tissues and cell lines of HCC. Patients having a high LINC00106 level exhibited a poor outcome. Under in vivo and in vitro conditions, the stemness and metastatic properties of HCC cells were augmented by LINC00106. Additionally, LINC00106 was found to sponge let7f to upregulate periostin, which lead to the activation of periostin-associated PI3K-AKT signaling pathway. Moreover, m6A methylation was found to cause LINC00106 upregulation while maintaining LINC00106 RNA transcript stability. Conclusion: m6A methylation triggers the upregulation of LINC00106, which promotes the stemness and metastasis properties in HCC cells by sponging let7f, thereby resulting in periostin activation. The findings indicate the potential of LINC00106 as a diagnostic marker and therapeutic target for HCC.

10.
J Mol Histol ; 51(6): 685-700, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33070277

RESUMO

Renal ischemia-reperfusion (IR) injury is a common issue in urological surgery, and the renal tubules, particularly the proximal tubules, are extremely vulnerable to IR injury. In this work, we detected the differently expressed genes (DEGs) between normal rabbit kidneys and IR kidneys by RNA-sequencing, then identified that matrix metalloproteinase-7 (MMP7) played an important role in the progress of IR injury. Indeed, A time-dependent promotion of renal injury was detected in rabbit model, as demonstrated by the increased levels of MMP2/7/9, and the decreased of tight junction protein-1 (TJP1). Furtherly, similar results were confirmed in human renal proximal tubule epithelial (HK-2) cells model. Notably, downregulation of MMP7 affected the activity of MMP2/9 by suppressing expression of cleaved-MMP2/9 not the pro-MMP2/9 protein, which directly alleviated the degradation of TJP1 in HK-2 model. On the contrary, MMP7 had not been affected by inhibiting MMP2/9. In addition, coimmunoprecipitation assay showed that knockdown MMP7 restrained the interaction between MMP2/9 and TJP1. Collectively, this study suggested that MMP7 could serve as early biomarkers for renal tubular injury, and revealed that MMP7 could destroy the integrity of tubular epithelium through degrading TJP1 by activating MMP2/9.


Assuntos
Epitélio/metabolismo , Túbulos Renais/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 7 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/metabolismo , Animais , Biomarcadores , Modelos Animais de Doenças , Suscetibilidade a Doenças , Epitélio/ultraestrutura , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Imuno-Histoquímica , Túbulos Renais/ultraestrutura , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 7 da Matriz/genética , Metaloproteinase 9 da Matriz/genética , Inibidores de Metaloproteinases de Matriz/farmacologia , Coelhos , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/patologia
11.
Am J Transplant ; 20(7): 1916-1921, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32282986

RESUMO

Over 1 000 000 cases of coronavirus disease 2019 (COVID-19) have been confirmed since the worldwide outbreak began. Not enough data on infected solid organ transplant (SOT) recipients are available, especially data about the management of immunosuppressants. We report two cases of COVID-19 in two transplant recipients, with different treatments and prognoses. The first patient received liver transplantation due to hepatitis B virus-related hepatocellular carcinoma and was confirmed to have COVID-19 9 days later. Following a treatment regimen consisting of discontinued immunosuppressant use and low-dose methylprednisolone-based therapy, the patient developed acute rejection but eventually recovered. The other patient had undergone a renal transplant from a living-related donor 17 years ago, and was admitted to the hospital because of persistent fever. This patient was also diagnosed with COVID-19. His treatment regimen consisted of reduced immunosuppressant use. No signs of rejection were observed during the regimen. In the end, the patient successfully recovered from COVID-19. These effectively treated cases can provide a basis for immunosuppressant management of COVID-19-positive SOT recipients.


Assuntos
Infecções por Coronavirus/complicações , Infecções por Coronavirus/terapia , Imunossupressores/uso terapêutico , Transplante de Órgãos , Pneumonia Viral/complicações , Pneumonia Viral/terapia , Transplantados , Adulto , Betacoronavirus , COVID-19 , Carcinoma Hepatocelular/complicações , Carcinoma Hepatocelular/cirurgia , Hepatite B/complicações , Hepatite B/cirurgia , Vírus da Hepatite B , Humanos , Falência Renal Crônica/complicações , Falência Renal Crônica/cirurgia , Transplante de Rim , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/cirurgia , Transplante de Fígado , Masculino , Metilprednisolona/administração & dosagem , Pessoa de Meia-Idade , Pandemias , Prognóstico , SARS-CoV-2 , Resultado do Tratamento
12.
Oxid Med Cell Longev ; 2020: 1079129, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32064020

RESUMO

Inflammation and oxidative stress are pivotal mechanisms for the pathogenesis of ischemia and reperfusion injury (IRI). Vagus nerve stimulation (VNS) may participate in maintaining oxidative homeostasis and response to external stimulus or injury. We investigated whether the in vivo VNS can protect the liver from IRI. In this study, hepatic IRI were induced by ligating the vessels supplying the left and middle lobes of the liver, which underwent 1 h occlusion followed with 24 h reperfusion. VNS was initiated 15 min after ischemia and continued 30 min. Hepatic function, histology, and apoptosis rates were evaluated after 24 h reperfusion. Compared with the IRI group, VNS significantly improved hepatic function. The protective effect was accompanied by a reduction in histological damage in the ischemic area, and the apoptosis rate of hepatocytes has considerable reduction. To find the underlying mechanism, proteomic analysis was performed and differential expression of glutathione synthetase (GSS) and glutathione S-transferase (GST) was observed. Subsequently, test results indicated that VNS upregulated the expression of mRNA and protein of GSS and GST. Meanwhile, VNS increased the plasma levels of glutathione and glutathione peroxidases. We found that VNS alleviated hepatic IRI by upregulating the antioxidant glutathione via the GSS/glutathione/GST signaling pathway.


Assuntos
Glutationa/sangue , Hepatócitos/metabolismo , Hepatopatias/terapia , Traumatismo por Reperfusão/terapia , Estimulação do Nervo Vago , Animais , Antioxidantes/metabolismo , Apoptose/genética , Citocinas/metabolismo , Glutationa/biossíntese , Glutationa/metabolismo , Glutationa Peroxidase/sangue , Glutationa Sintase/genética , Glutationa Sintase/metabolismo , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Hepatócitos/enzimologia , Fígado/enzimologia , Fígado/lesões , Fígado/metabolismo , Fígado/patologia , Hepatopatias/enzimologia , Hepatopatias/metabolismo , Hepatopatias/patologia , Masculino , Estresse Oxidativo , Proteômica , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/enzimologia , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Transdução de Sinais/genética
13.
Biomater Sci ; 7(12): 5247-5257, 2019 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-31602445

RESUMO

Fibrous chitin dressing (FCD) prepared from a NaOH-urea aqueous solution of chitin via a physical process was used to study its effect on wound healing using a full-thickness cutaneous wound model in rats and mice. It was demonstrated that wounds in rats covered with the FCD showed faster collagen (especially type I collagen) growth and speedier healing than those with Gauze (12 days versus 16 days). The ability of FCD to promote wound healing was also observed on wild-type (WT) mice. For MyD88-knockout mice, however, FCD displayed no beneficial but an adverse effect on wound healing: the healing time for wounds treated with FCD was even longer than those treated with gauze. Importantly, in vivo studies indicated that FCD-treated mice, compared to gauze-treated ones, exhibited markedly higher expressions of MyD88, IKBα, TGF-ß, P-TßR II, TßR II and P-Smad2/3 in wild-type mice. For MyD88 knockout mice, however, the expressions of those molecules were inhibited and lowered in FCD-treated ones than those treated with gauze. In vitro studies confirmed that chitin increased the expression of TGF-ß, P-TßRII and P-Smad2/3 while the expressions of those molecules were significantly inhibited with CD14 antibody (p < 0.05). These results indicated that FCD accelerated wound healing through a MyD88-dependent pathway, followed by a TGF-ß/Smad pathway. This work not only demonstrated the superior wound healing effect of chitin-derived dressing, but also provided for the first time the underlying molecular mechanism, further establishing chitin as an important biomedical material for potential clinical applications.


Assuntos
Bandagens , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Quitina/química , Quitina/farmacologia , Cicatrização/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo I/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Fatores de Crescimento do Endotélio Vascular/metabolismo , Proteínas Smad/metabolismo , Hidróxido de Sódio/química , Fator de Crescimento Transformador beta/metabolismo , Ureia/química , Fator A de Crescimento do Endotélio Vascular/metabolismo , Água/química
14.
Oncol Lett ; 17(6): 5159-5168, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31186731

RESUMO

Deubiquitinating enzymes regulate protein activity and cell homeostasis by removing ubiquitin moieties from various substrates. Ubiquitin carboxyl-terminal hydrolase 22 (USP22) is a member of the deubiquitinating protease family and is associated with the development of several tumor types. A previous study demonstrated that USP22 is highly expressed in liver cancer, and its high expression is associated with resistance to chemotherapy. However, the role of USP22 in hepatitis B virus (HBV)-associated liver cancer has not yet been elucidated. The current study demonstrated that USP22 was highly expressed in the tissues of patients with HBV-associated liver cancer, and its high expression was associated with clinicopathological characteristics, including tumor size, clinical stage and prognosis. Further results indicated that USP22 may regulate the proliferative and apoptotic abilities of HepG2.2.15 cells. Additionally, investigation into the underlying mechanism, using small interfering RNA, revealed that the downregulation of USP22 inhibited proliferation and promoted apoptosis though the phosphoinositide 3-kinase/protein kinase B signaling pathway. Therefore, USP22 has the potential to be used as an independent predictor of patient prognosis, as well as a therapeutic target for the treatment of HBV-associated liver cancer.

15.
Transl Cancer Res ; 8(4): 1268-1278, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35116869

RESUMO

BACKGROUND: The precise role of heat shock protein 27 (HSP27), as a type of small molecular protein in HSPs, in pancreatic ductal adenocarcinoma (PDAC) remains to be elucidated. The aim of the present study was to investigate the expression and function of HSP27 in PDAC cells. METHODS: We first detected the expression of HSP27 in PDAC tissues. Combining with the clinical pathology characteristics of PDAC patients, the relationship between them was analyzed. Then, we knocked down HSP27 using short interfering RNA (siRNA) and observed its biological functions using scratch assay and matrigel invasion and migration assays in ASPC-1 and PANC-1 cells. In mechanism, we verified the ß-catenin/MMP-3 pathway associated proteins in ASPC-1 and PANC-1 cells. RESULTS: We found that HSP27 was highly expression in PDAC tissues, and was positively correlated with tumor differentiation, TNM staging and poor prognosis of PDAC patients. In vitro, we down-regulated the expression of HSP27 in ASPC-1 and PANC-1 cells and found that the invasion and migration ability of PDAC cells were significantly depressed, meanwhile, the activation of the ß-catenin/MMP-3 pathway was inhibited. CONCLUSIONS: HSP27 may be used as a tumor biomarker for diagnosis of PDAC, and HSP27 can promote the invasion and migration of PDAC by activating the ß-catenin/MMP3 Pathway. Therefore, inhibition of HSP27 has therapeutic potential for the treatment of PDAC.

16.
Oncol Rep ; 41(1): 43-56, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30365128

RESUMO

Liver metastasis is one of the major causes of death in patients with colorectal cancer, and although treatment has improved recently, the long­term survival rate of patients has not improved significantly. In the present study, we used immunohistochemistry to determine that phosphoprotein enriched in astrocytes­15 kDa (PEA15) was highly expressed in colorectal cancer tissues and liver metastatic cancer tissues. It was also highly expressed in metastatic colorectal cancer patients compared to non­metastatic patients. Through clinicopathological data of patients with liver metastasis of colorectal cancer, we found that high expression of PEA15 was positively correlated with TNM staging, liver metastasis and poor prognosis of colorectal cancer patients. Using confocal immunofluorescence microscopy, western blotting and cell proliferation, migration and invasion assays, we also determined that PEA15 could promote cancer cell proliferation in vitro and in vivo, epithelial mesenchymal transition (EMT) and the characteristics of cancer stem cells in vitro, thus promoting the abilities of invasion and migration. In addition, we revealed that PEA15 promoted the liver metastasis of colorectal cancer cells in a xenograft tumor metastasis model. In addition, concerning the mechanism, we used gene chip analysis to determine that PEA15 upregulated the ERK/MAPK signaling pathway in colorectal cancer cells. Therefore, we concluded that PEA15 may be a potential biomarker for liver metastasis of colorectal cancer therapy. Collectively, PEA15 promoted the development of liver metastasis of colorectal cancer through the ERK/MAPK signaling pathway.


Assuntos
Neoplasias Colorretais/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/secundário , Sistema de Sinalização das MAP Quinases , Fosfoproteínas/metabolismo , Regulação para Cima , Animais , Proteínas Reguladoras de Apoptose , Células CACO-2 , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HT29 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Transplante de Neoplasias , Fosfoproteínas/genética , Prognóstico
17.
Cell Commun Signal ; 16(1): 30, 2018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29898735

RESUMO

In this study, we investigated the role of microRNA-644a (miR-644a) in the growth and survival of hepatocellular carcinoma (HCC) cells. MiR-644a levels were lower in HCC tissues than in adjacent peri-cancerous tissues (n = 135). MiR-644a expression was inversely correlated with heat shock factor 1 (HSF1) expression, tumour diameter and TNM stage. Moreover, HepG2 and SMMC-7721 cell lines showed lower miR-644a expression than normal L-O2 hepatocytes. MiR-644a overexpression in HepG2 and SMMC-7721 cells increased apoptosis by downregulating HSF1. Dual luciferase reporter assays confirmed the presence of a miR-644a binding site in the 3'-untranslated region (3'-UTR) of HSF1. Xenograft tumours derived from SMMC-7721 cells transfected with a miR-664a mimic showed less growth than tumours derived from untransfected controls. Protein chip analysis revealed that miR-644a-overexpressing SMMC-7721 and HepG2 cells strongly expressed pro-apoptotic BH3-only proteins, such as BID, BAD, BIM, SMAC, Apaf-1 and cleaved caspases-3 and -9. These findings suggest miR-644a promotes apoptosis in HCC cells by inhibiting HSF1.


Assuntos
Apoptose/genética , Carcinoma Hepatocelular/patologia , Regulação para Baixo/genética , Fatores de Transcrição de Choque Térmico/genética , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Regiões 3' não Traduzidas/genética , Animais , Carcinoma Hepatocelular/genética , Proliferação de Células/genética , Transformação Celular Neoplásica , Progressão da Doença , Feminino , Humanos , Neoplasias Hepáticas/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Carga Tumoral/genética
18.
Mol Med Rep ; 16(5): 6214-6221, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28901476

RESUMO

Hepatic ischemia/reperfusion (I/R) injury is a common pathophysiological process that occurs following liver surgery, which is associated with oxidative stress, and can cause acute liver injury and lead to liver failure. Recently, the development of drugs for the prevention of hepatic I/R injury has garnered interest in the field of liver protection research. Previous studies have demonstrated that [D­Ala2, D­Leu5]­Enkephalin (DADLE) exerts protective effects against hepatic I/R injury. To further clarify the specific mechanism underlying the effects of DADLE on hepatic I/R injury, the present study aimed to observe the effects of various doses of DADLE on hepatic I/R injury in mice. The results indicated that DADLE, at a concentration of 5 mg/kg, significantly reduced the levels of alanine aminotransferase and aspartate aminotransferase in the serum, and the levels of malondialdehyde in the liver homogenate. Conversely, the levels of glutathione, catalase and superoxide dismutase in the liver homogenate were increased. In addition, DADLE was able to promote nuclear factor, erythroid 2 like 2 (Nrf2) nuclear translocation and upregulate the expression of heme oxygenase (HO)­1, which is a factor downstream of Nrf2, thus improving hepatic I/R injury in mice. In conclusion, the present study demonstrated that DADLE was able to significantly improve hepatic I/R injury in mice, and the specific mechanism may be associated with the Nrf2/HO­1 signaling pathway.


Assuntos
Leucina Encefalina-2-Alanina , Heme Oxigenase-1 , Hepatopatias , Fígado , Proteínas de Membrana , Fator 2 Relacionado a NF-E2 , Traumatismo por Reperfusão , Transdução de Sinais , Animais , Masculino , Camundongos , Aspartato Aminotransferases/metabolismo , Modelos Animais de Doenças , Leucina Encefalina-2-Alanina/farmacologia , Heme Oxigenase-1/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Hepatopatias/tratamento farmacológico , Hepatopatias/metabolismo , Malondialdeído/metabolismo , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Transdução de Sinais/efeitos dos fármacos , Superóxido Dismutase/metabolismo , Regulação para Cima/efeitos dos fármacos
19.
J Exp Clin Cancer Res ; 36(1): 64, 2017 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-28482903

RESUMO

BACKGROUND: As a relatively conservative transcriptional regulator in biological evolution, heat shock factor 1 (HSF1) is activated by, and regulates the expression of heat shock proteins (HSPs) in response to a variety of stress conditions. HSF1 also plays a key role in regulating the development of various tumors; however, its role in pancreatic cancer and the specific underlying mechanism are not clear. METHODS: We first examined HSF1 expression in pancreatic cancer tissues by immunohistochemistry, and then studied its clinical significance. We then constructed HSF1-siRNA to investigate the potential of HSF1 to regulate apoptosis, proliferation and the cell cycle of pancreatic cancer cells and the underlying mechanism both in vitro and in vivo. Protein chip analysis was used subsequently to explore the molecular regulation pathway. Finally, second mitochondria-derived activator of caspase (SMAC)-siRNA was used to validate the signaling pathway. RESULTS: HSF1 was highly expressed in pancreatic cancer tissues and the level of upregulation was found to be closely related to the degree of pancreatic cancer differentiation and poor prognosis. After HSF1-silencing, we found that pancreatic cancer cell proliferation decreased both in vitro and in vivo and the apoptotic cell ratio increased, while the mitochondrial membrane potential decreased, and the cells were arrested at the G0/G1 phase. In terms of the molecular mechanism, we confirmed that HSF1 regulated SMAC to inhibit mitochondrial apoptosis in pancreatic cancer cells, and to promote the occurrence of pancreatic tumors. SMAC silencing reversed the effects of HSF1 silencing. CONCLUSION: Our study provides evidence that HSF1 functions as a novel oncogene in pancreatic tumors and is implicated as a target for the diagnosis and treatment of pancreatic cancer.


Assuntos
Apoptose , Transformação Celular Neoplásica/metabolismo , Fatores de Transcrição de Choque Térmico/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais , Adulto , Idoso , Animais , Proteínas Reguladoras de Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Transformação Celular Neoplásica/genética , Modelos Animais de Doenças , Feminino , Expressão Gênica , Fatores de Transcrição de Choque Térmico/genética , Xenoenxertos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Mitocôndrias/genética , Modelos Biológicos , Gradação de Tumores , Estadiamento de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia
20.
Biomed Rep ; 6(5): 525-531, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28515911

RESUMO

The aim of the present study was to investigate the association between tubulin folding cofactor B (TBCB) expression and ischemia-reperfusion injury (IRI) in mice. A total of 48 C57BL/6 mice were randomly divided into a control group (Sham, n=6) and an ischemia-reperfusion group (n=42). The ischemia-reperfusion group was further divided into 6 subgroups as per different times after reperfusion (2, 4, 6, 8, 12 and 24 h), with 7 mice per subgroup. A hepatic IRI model was established in mice by clamping the hepatic hilum. Morphology, serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), interleukin 6 (IL-6) and tumor necrosis factor-α (TNF-α), and the expression level of TBCB were detected. Compared with the control group, the livers from the ischemia-reperfusion group were significantly changed, particularly at 12 h following ischemia-reperfusion, with obvious hepatic cell degeneration and necrosis. The ALT, AST, IL-6 and TNF-α levels in the sera of the mice in the hepatic ischemia-reperfusion group were increased at all time points following ischemia-reperfusion, and were the highest at 12 h, demonstrating statistically significant differences when compared with the control group (P<0.05). Furthermore, the expression levels of TBCB, TNF-α and IL-6 were significantly increased at all time-points following ischemia-reperfusion, and were the most significant at 12 h. At 24 h following ischemia-reperfusion, the expression levels had decreased. The present study indicated that TBCB expression is associated with TNF-α and IL-6 expression levels in mice with hepatic ischemia-reperfusion, and may be key in the development of liver injury during ischemia-reperfusion in mice.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA