Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Exp Hematol Oncol ; 13(1): 68, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39014491

RESUMO

Nuclear factor-kappaB (NF-ĸB) plays a crucial role in both innate and adaptive immune systems, significantly influencing various physiological processes such as cell proliferation, migration, differentiation, survival, and stemness. The function of NF-ĸB in cancer progression and response to chemotherapy has gained increasing attention. This review highlights the role of NF-ĸB in inflammation control, biological mechanisms, and therapeutic implications in cancer treatment. NF-ĸB is instrumental in altering the release of inflammatory factors such as TNF-α, IL-6, and IL-1ß, which are key in the regulation of carcinogenesis. Specifically, in conditions including colitis, NF-ĸB upregulation can intensify inflammation, potentially leading to the development of colorectal cancer. Its pivotal role extends to regulating the tumor microenvironment, impacting components such as macrophages, fibroblasts, T cells, and natural killer cells. This regulation influences tumorigenesis and can dampen anti-tumor immune responses. Additionally, NF-ĸB modulates cell death mechanisms, notably by inhibiting apoptosis and ferroptosis. It also has a dual role in stimulating or suppressing autophagy in various cancers. Beyond these functions, NF-ĸB plays a role in controlling cancer stem cells, fostering angiogenesis, increasing metastatic potential through EMT induction, and reducing tumor cell sensitivity to chemotherapy and radiotherapy. Given its oncogenic capabilities, research has focused on natural products and small molecule compounds that can suppress NF-ĸB, offering promising avenues for cancer therapy.

2.
Adv Biol (Weinh) ; 8(7): e2300288, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38717793

RESUMO

Organelles, substructures in the cytoplasm with specific morphological structures and functions, interact with each other via membrane fusion, membrane transport, and protein interactions, collectively termed organelle interaction. Organelle interaction is a complex biological process involving the interaction and regulation of several organelles, including the interaction between mitochondria-endoplasmic reticulum, endoplasmic reticulum-Golgi, mitochondria-lysosomes, and endoplasmic reticulum-peroxisomes. This interaction enables intracellular substance transport, metabolism, and signal transmission, and is closely related to the occurrence, development, and treatment of many diseases, such as cancer, neurodegenerative diseases, and metabolic diseases. Herein, the mechanisms and regulation of organelle interactions are reviewed, which are critical for understanding basic principles of cell biology and disease development mechanisms. The findings will help to facilitate the development of novel strategies for disease prevention, diagnosis, and treatment opportunities.


Assuntos
Organelas , Humanos , Organelas/metabolismo , Animais , Doenças Neurodegenerativas/terapia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Neoplasias/terapia , Neoplasias/metabolismo , Neoplasias/patologia , Retículo Endoplasmático/metabolismo , Mitocôndrias/metabolismo
3.
Pharmacol Res ; 205: 107228, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38810904

RESUMO

Coronavirus disease 2019 (COVID-19) affected people worldwide, and fever is one of the major symptoms of this disease. Although Acetaminophen (APAP) is a common fever-reducing medication, it can also mediate liver injury. However, the role of PGC-1α in regulating mitochondrial quality control by lactate dehydrogenase B (LDHB), a vital enzyme catalyzing the conversion of lactate to pyruvate, in APAP-induced hepatotoxicity, is unclear. Here, gene expression omnibus data of patients with APAP-induced liver injury were used to explore gene expression profiles. AML12 cells and C57/BL6 mice were used to establish models of APAP-induced acute liver injury. SIRT1 and PGC-1α were overexpressed in vitro via lentiviral transfection to establish stable cell lines. The results showed that APAP treatment decreased SIRT1/PGC-1α/LDHB expression and increased protein lactylation, mitochondrial lactate levels, and pathological damage in liver mitochondria. PGC-1α upregulation or activation ameliorated APAP-induced damage in the cells and liver. Furthermore, PGC-1α overexpression increased LDHB synthesis, reduced lactylation, and induced a switch from lactate to pyruvate production. These results suggest that PGC-1α and LDHB play a role in APAP-induced liver injury by regulating mitochondrial quality control and lactate metabolic reprogramming. Therefore, the PGC-1α/LDHB axis is a potential therapeutic target for APAP-induced liver injury.


Assuntos
Acetaminofen , Doença Hepática Induzida por Substâncias e Drogas , L-Lactato Desidrogenase , Camundongos Endogâmicos C57BL , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Animais , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/patologia , Camundongos , Humanos , Masculino , L-Lactato Desidrogenase/metabolismo , Ácido Láctico/metabolismo , Proteínas Mitocondriais/metabolismo , Proteínas Mitocondriais/genética , Linhagem Celular , Mitocôndrias Hepáticas/metabolismo , Mitocôndrias Hepáticas/efeitos dos fármacos , Sirtuína 1/metabolismo , Sirtuína 1/genética , Isoenzimas
4.
Genes Dis ; 10(4): 1596-1612, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37397525

RESUMO

Non-alcoholic steatohepatitis (NASH), a progressive form of non-alcoholic fatty liver disease (NAFLD), is characterised by chronic liver inflammation, which can further progress into complications such as liver cirrhosis and NASH-associated hepatocellular carcinoma (HCC) and therefore has become a growing health problem worldwide. The type I interferon (IFN) signaling pathway plays a pivotal role in chronic inflammation; however, the molecular mechanisms underlying NAFLD/NASH from the perspective of innate immune response has not yet been fully explored. In this study, we elucidated the mechanisms of how innate immune response modulates NAFLD/NASH pathogenesis, and demonstrated that hepatocyte nuclear factor-1alpha (HNF1A) was suppressed and the type I IFN production pathway was activated in liver tissues of patients with NAFLD/NASH. Further experiments suggested that HNF1A negatively regulates the TBK1-IRF3 signaling pathway by promoting autophagic degradation of phosphorylated-TBK1, which constrains IFN production, thereby inhibiting the activation of type I IFN signaling. Mechanistically, HNF1A interacts with the phagophore membrane protein LC3 through its LIR-docking sites, and mutations of LIRs (LIR2, LIR3, LIR4, and LIRs) block the HNF1A-LC3 interaction. In addition, HNF1A was identified not only as a novel autophagic cargo receptor but also to specifically induce K33-linked ubiquitin chains on TBK1 at Lys670, thereby resulting in autophagic degradation of TBK1. Collectively, our study illustrates the crucial function of the HNF1A-TBK1 signaling axis in NAFLD/NASH pathogenesis via cross-talk between autophagy and innate immunity.

5.
Biosens Bioelectron ; 222: 114956, 2023 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36525708

RESUMO

Convenient, ultrasensitive, and accurate detection of rare variants is essential for early cancer diagnosis and precision medicine, however, despite years of efforts, tools that have all these qualities remain elusive. Here, we developed a one-step CRISPR/Cas12a-based digital diagnostic platform for accurately quantifying mutant alleles, referred to as the CRISPR ASsoaciated Mutation Allele Rapid Test (CASMART). The platform accurately quantifies the variant allele frequency of EGFR L858R within 1 h at 42 °C and can detect mutant targets as low as 0.3 copies/µL (0.498 aM) in mock multiplex cfDNA samples. We further investigated the applicability of CASMART using human genomic samples with confirmed EGFR L858R mutations previously measured variant allele frequency by next-generation sequencing. Comparison across platforms revealed equivalent detection performance (Pearson's correlation coefficient, R2 = 0.9208) and high quantification accuracy for mutation allele frequency (intraclass correlation coefficient = 0.959). Our one-step approach enables easy and accurate variant allele frequency measurement of rare mutant alleles without PCR instrumentation, while the assay time was reduced by approximately half compared to the digital PCR with the shortest turnaround. The CASMART is an alternative to conventional single nucleotide polymorphism detection methods with great potential as a next-generation biosensor for rapidly quantifying the variant allele fraction, especially in resource-limited settings.


Assuntos
Técnicas Biossensoriais , Sistemas CRISPR-Cas , Humanos , Alelos , Sistemas CRISPR-Cas/genética , Mutação , Receptores ErbB/genética
6.
Cancer Cell Int ; 22(1): 143, 2022 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-35366902

RESUMO

BACKGROUND: Pancreatic adenocarcinoma (PAAD) is a severe malignant with a 5-year survival rate of approximately 9%. Oleanolic acid is a well-known natural triterpenoid which exhibits pharmacological activities. We previously synthesized a series of oleanolic acid derivatives and evaluated the tumor-suppressive activity of olean-28,13ß-lactam (B28) in prostate cancer. However, the detailed mechanism remains to be understood. METHODS: The anti-tumor activity of B28 in PAAD was confirmed by RTCA, colony formation assay and flow cytometry. GO and KEGG enrichment analyses were performed to analyze the differentially expressed genes (DEGs) obtained by RNA sequencing. The effects of B28 on cell bioenergetics were evaluated by seahorse analyzer. Lenti-virus packaged plasmids were performed to knockdown or overexpress target genes. Alteration of mitochondrial membrane potential, ROS and GSH/GSSG were measured by corresponding detection kits according to the manufacturer's protocol. RESULTS: We evaluated and confirmed the promising anti-tumor activity of B28 in vitro. RNA-seq profile indicated that multiple metabolic pathways were interrupted in B28 treated PAAD cells. Next, we demonstrated that B28 induces cellular bioenergetics crisis to inhibit PAAD cells growth and induce cell death. We further validated that cell cycle arrest, inhibition of cell growth, cell apoptosis and cell bioenergetics disruption were functionally rescued by ROS scavenger NAC. Mechanistically, we found glutamine metabolism was inhibited due to B28 administration. Moreover, we validated that down-regulation of GLS1 contributes to ROS generation and bioenergetics interruption induced by B28. Furthermore, we elucidated that YTHDF1-GLS1 axis is the potential downstream target of B28 to induce PAAD cell metabolic crisis and cell death. Finally, we also confirmed the anti-tumor activity of B28 in vivo. CONCLUSIONS: Current study demonstrates B28 disrupts YTDFH1-GLS1 axis to induce ROS-dependent cell bioenergetics crisis and cell death which finally suppress PAAD cell growth, indicating that this synthesized olean-28,13ß-lactam maybe a potent agent for PAAD intervention.

7.
Mol Ther Nucleic Acids ; 26: 1351-1363, 2021 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-34853732

RESUMO

Aberrant expression of long non-coding RNAs (lncRNAs) has been reported in multiple cancers. However, the underlying mechanisms mediated by super-enhancers remain elusive. Here we sought to define the role of a novel lncRNA termed lncRNA-DAW in tumorigenesis. Our results revealed that lncRNA-DAW was driven by a liver-specific super-enhancer and transcriptionally activated by HNF4G, leading to frequent elevation in hepatocellular carcinoma (HCC) specimens. Ectopic expression of lncRNA-DAW promoted both in vivo and in vitro tumor growth. By using RNA sequencing, Wnt2 was screened out as a downstream effector of lncRNA-DAW. We next found that lncRNA-DAW physically interacted with EZH2, a negative regulator of Wnt2. This interplay subsequently potentiated CDK1-EZH2 interaction, leading to the phosphorylation and ubiquitination of EZH2. The lncRNA-DAW-mediated EZH2 degradation facilitated the de-repression of Wnt2 transcription, which eventually activated the Wnt/ß-catenin pathway. Furthermore, we verified that Wnt2 potentiated in vitro and in vivo cancer cell growth by activating the Wnt/ß-catenin pathway. Finally, Wnt2 amplification was confirmed as a common event in liver cancer, and the expression of lncRNA-DAW was positively correlated with Wnt2 in HCC specimens. Collectively, we are the first to identify lncRNA-DAW as a novel candidate oncogene in liver cancer, and this lncRNA may serve as a novel clinical diagnosis biomarker for liver cancer.

8.
Front Cell Dev Biol ; 9: 697832, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34350183

RESUMO

Cannabidiol (CBD), a phytochemical derived from Cannabis sativa L., has been demonstrated to exhibit promising anti-tumor properties in multiple cancer types. However, the effects of CBD on hepatocellular carcinoma (HCC) cells remain unknown. We have shown that CBD effectively suppresses HCC cell growth in vivo and in vitro, and induced HCC cell pyroptosis in a caspase-3/GSDME-dependent manner. We further demonstrated that accumulation of integrative stress response (ISR) and mitochondrial stress may contribute to the initiation of pyroptotic signaling by CBD. Simultaneously, CBD can repress aerobic glycolysis through modulation of the ATF4-IGFBP1-Akt axis, due to the depletion of ATP and crucial intermediate metabolites. Collectively, these observations indicate that CBD could be considered as a potential compound for HCC therapy.

9.
Aesthetic Plast Surg ; 45(2): 749-757, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32350561

RESUMO

This study aimed to investigate the value of miR-222 in hypertrophic scars (HS). Specific mechanisms were used to measure the level of miR-222, while MTT assay, flow cytometry, western blot and qRT-PCR were employed to detect the relative proteins after fibroblasts were transfected with the miR-222 mimic/inhibitor. The direct target of miR-222 was determined by Dual-Luciferase Reporter assay. Furthermore, qRT-PCR and western blot were employed to detect the matrix metalloproteinase 1 (MMP1) RNA/protein after fibroblasts were transfected with the miR-222 mimic/inhibitor. These results revealed that miR-222 was significantly upregulated in HS fibroblasts. The overexpression of miR-222 enhanced the HS fibroblast proliferation, increased the cell population in the S phase, inhibited the cell apoptosis, enhanced the expression levels of Col1A1, Col3A1 mRNA/protein, proliferating cell nuclear antigen (PCNA), cyclin D1, cyclin E1 and CDK1 and reduced the expression levels of cleaved caspase-3/9. However, the miR-222 suppression triggered opposite effects. Furthermore, miR-222 played a regulatory role in HS by negatively regulating its target gene MMP1 by binding with its 3'-untranslated region. The overexpression of MMP1 reduced the expression levels of PCNA and cyclin D1, but enhanced the expression levels of cleaved caspase-3. Therefore, MiR-222 and MMP1 have potential value for HS. NO LEVEL ASSIGNED: This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .


Assuntos
Cicatriz Hipertrófica , MicroRNAs , Apoptose , Proliferação de Células , Cicatriz Hipertrófica/genética , Cicatriz Hipertrófica/patologia , Fibroblastos/patologia , Humanos , Metaloproteinase 1 da Matriz/genética , MicroRNAs/genética
10.
J Cancer ; 11(6): 1505-1515, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32047557

RESUMO

Activated pancreatic stellate cells (PSCs) are the main effector cells in the process of fibrosis, a major pathological feature in pancreatic diseases that including chronic pancreatitis and pancreatic cancer. During tumorigenesis, quiescent PSCs change into an active myofibroblast-like phenotype which could create a favorable tumor microenvironment and facilitate cancer progression by increasing proliferation, invasiveness and inducing treatment resistance of pancreatic cancer cells. Many cellular signals are revealed contributing to the activation of PSCs, such as transforming growth factor-ß, platelet derived growth factor, mitogen-activated protein kinase (MAPK), Smads, nuclear factor-κB (NF-κB) pathways and so on. Therefore, investigating the role of these factors and signaling pathways in PSCs activation will promote the development of PSCs-specific therapeutic strategies that may provide novel options for pancreatic cancer therapy. In this review, we systematically summarize the current knowledge about PSCs activation-associated stimulating factors and signaling pathways and hope to provide new strategies for the treatment of pancreatic diseases.

11.
Cell Commun Signal ; 17(1): 145, 2019 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-31718671

RESUMO

BACKGROUND: Notch1 signalling is a stem-cell-related pathway that is essential for embryonic development, tissue regeneration and organogenesis. However, the role of Notch1 in the formation of myofibroblasts and fibrosis in kidneys following injury remains unknown. METHODS: The activity of Notch1 signalling was evaluated in fibrotic kidneys in CKD patients and in ureteral obstructive models in vivo and in cultured fibroblasts and TECs in vitro. In addition, the crosstalk of Notch1 with TGF-ß1/Smad2/3 signalling was also investigated. RESULTS: Notch1 activity was elevated in fibrotic kidneys of rat models and patients with chronic kidney disease (CKD). Further study revealed that epithelial and interstitial Notch1 activity correlated with an α-SMA-positive myofibroblastic phenotype. In vitro, injury stimulated epithelial Notch1 activation and epithelial-mesenchymal transition (EMT), resulting in matrix deposition in tubular epithelial cells (TECs). Additionally, interstitial Notch1 activation in association with fibroblast-myofibroblast differentiation (FMD) in fibroblasts mediated a myofibroblastic phenotype. These TGF-ß1/Smad2/3-dependent phenotypic transitions were abolished by Notch1 knockdown or a specific antagonist, DAPT, and were exacerbated by Notch1 overexpression or an activator Jagged-1-Fc chimaera protein. Interestingly, as a major driving force behind the EMT and FMD, TGF-ß1, also induced epithelial and interstitial Notch1 activity, indicating that TGF-ß1 may engage in crosstalk with Notch1 signalling to trigger fibrogenesis. CONCLUSION: These findings suggest that epithelial and interstitial Notch1 activation in kidneys following injury contributes to the myofibroblastic phenotype and fibrosis through the EMT in TECs and to the FMD in fibroblasts by targeting downstream TGF-ß1/Smad2/3 signalling.


Assuntos
Diaminas/farmacologia , Células Epiteliais/efeitos dos fármacos , Fibrose/tratamento farmacológico , Miofibroblastos/efeitos dos fármacos , Receptor Notch1/antagonistas & inibidores , Tiazóis/farmacologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibrose/metabolismo , Fibrose/patologia , Humanos , Masculino , Miofibroblastos/metabolismo , Fenótipo , Ratos , Ratos Sprague-Dawley , Receptor Notch1/metabolismo , Transdução de Sinais/efeitos dos fármacos
12.
Phytomedicine ; 62: 152976, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31177020

RESUMO

BACKGROUND: Sedum sarmentosum Bunge extract (SSBE) has been used traditionally to treat liver inflammatory diseases in the Asian area. PURPOSE: The aim of this study is to evaluate the anti-inflammatory activity of SSBE on renal injury. METHODS: We investigated whether SSBE has an anti-inflammatory effect by suppressing M1-macrophage polarization in rats with unilateral ureteral obstruction (UUO) and in cultured macrophages. In addition, the effect of SSBE on the activities of interferon regulatory factor-5 (IRF5) and NF-κB p65 were further examined. RESULTS: Oral administration of SSBE (100 mg kg-1) markedly inhibited the infiltration of CD68-positive macrophages and reduced tubulointerstitial damage in kidney tissues following injury. In addition, SSBE reduced the expression of proinflammatory cytokine (MIF), chemokine (MCP-1), interleukin (IL-6), IFN-γ, and TNF-α, which are involved in the infiltration and activation of macrophages. Moreover, SSBE treatment also decreased the synthesis and release of MCP-1 and MIF in tubular epithelial cells after injury. Further study revealed that SSBE downregulated the levels of IL-12 and iNOS, indicating a crucial role of SSBE on the inhibition of M1 macrophage polarization in kidney injury. In cultured macrophages, lipopolysaccharide (LPS) induced the polarization of macrophage towards M1 phenotype, but was inhibited by SSBE treatment. Notably, SSBE reduced the activities of interferon regulatory factor 5 (IRF5) and NF-κB p65 in injured kidneys and in LPS-treated macrophages, which was independent of TLR4/MyD88. As a result, SSBE reduced the expression of HIF-1α and the induction of GLUT1, and thereby inhibited anaerobic glycolysis in macrophages. CONCLUSION: SSBE exerts a marked anti-inflammatory effect and alleviates kidney injury, at least in part, by suppressing M1-macrophage polarization.


Assuntos
Inflamação/tratamento farmacológico , Extratos Vegetais/farmacologia , Sedum/química , Animais , Rim/imunologia , Rim/lesões , Lipopolissacarídeos/farmacologia , Ativação de Macrófagos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Masculino , Óxido Nítrico Sintase Tipo II/efeitos dos fármacos , Óxido Nítrico Sintase Tipo II/metabolismo , Extratos Vegetais/química , Ratos , Ratos Sprague-Dawley , Fator de Transcrição RelA/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo
13.
Melanoma Res ; 29(3): 254-262, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30640294

RESUMO

Long noncoding RNA X-inactive specific transcript (XIST) was confirmed to participate in the development of many cancers. However, the function of XIST in malignant melanoma (MM) remained largely unknown. In the current study, we found that the XIST expression level was upregulated in MM tissues and cell lines. In addition, the growth rate of MM cells transfected with silencing XIST was significantly decreased compared with that with silencing normal control. XIST knockdown inhibited proliferation and migration in MM cells and increased the oxaliplatin sensitivity of oxaliplatin-resistant MM cells. Bioinformatics analysis showed that XIST acts as a molecular sponge for miR-21 and miR-21 directly targets with 3'-UTR of PI3KR1. Furthermore, XIST knockdown inhibited PI3KRI and AKT expression, and promoted Bcl-2 and Bax expression. In short, the current study showed that XIST was a crucial regulator in progression and oxaliplatin resistance of MM, providing a novel insight into the pathogenesis and underlying therapeutic target for MM.


Assuntos
Biomarcadores Tumorais/genética , Resistencia a Medicamentos Antineoplásicos/genética , Melanoma/patologia , Oxaliplatina/farmacologia , RNA Longo não Codificante/genética , Neoplasias Cutâneas/patologia , Idoso , Antineoplásicos/farmacologia , Proliferação de Células , Estudos de Coortes , Progressão da Doença , Feminino , Seguimentos , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Melanoma/tratamento farmacológico , Melanoma/genética , Prognóstico , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Taxa de Sobrevida , Células Tumorais Cultivadas
14.
Oncol Rep ; 39(4): 1892-1900, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29393444

RESUMO

Drug-resistance is the key reason for the ineffectiveness of chemotherapy in pancreatic cancer. Thus, it is very important to explore the molecular mechanisms of drug­resistance and the methods of effective intervention. In the present study, we investigated the effect of tyrphostin B42, also called AG490, on histone deacetylase (HDAC) inhibitor trichostatin A (TSA)­induced resistance in pancreatic cancer cells (PCCs). Evidence from phase contrast microscope revealed that TSA­resistant cells (PANC­1­TSA) had higher proliferative activity than non­resistant cells (PANC­1). This over­proliferative activity induced by TSA may be associated with abnormal activation of JAK2/STAT3 signaling, which can be strengthened by interleukin­6 (IL­6), a STAT3­upstream inducer, resulting in enhanced expression of STAT3-downstream target genes including c­Myc, c­Src, HIF­1α, and CCND1. In addition, increased expression of Bcl­2 mRNA and decreased expression of Bax mRNA in PANC­1­TSA cells indicated that TSA induced the inhibition of mitochondrial-dependent apoptosis in PCCs. Tyrphostin B42 treatment evidently antagonized the activation of IL­6/JAK2/STAT3 in a dose­dependent manner. As a result, tyrphostin B42 inhibited the over­proliferative activity of PANC­1­TSA cells, and downregulated the expression of IL­6/JAK2/STAT3­downstream target genes. Moreover, tyrphostin B42 induced the apoptosis of PCCs by regulating the expression of mitochondrial­related genes. Therefore, these findings demonstrated that tyrphostin B42 attenuated TSA­mediated resistance in PCCs by antagonizing the IL­6/JAK2/STAT3 signaling.


Assuntos
Interleucina-6/genética , Janus Quinase 2/genética , Neoplasias Pancreáticas/tratamento farmacológico , Fator de Transcrição STAT3/genética , Tirfostinas/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Ácidos Hidroxâmicos/efeitos adversos , Ácidos Hidroxâmicos/farmacologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Transdução de Sinais/efeitos dos fármacos
15.
Mol Med Rep ; 16(1): 737-745, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28560403

RESUMO

Sedum sarmentosum Bunge (SSBE) is a perennial plant widely distributed in Asian countries, and its extract is traditionally used for the treatment of certain inflammatory diseases. Our previous studies demonstrated that SSBE has marked renal anti­fibrotic effects. However, the underlying molecular mechanisms remain to be fully elucidated. The present study identified that SSBE exerts its inhibitory effect on the myofibroblast phenotype and renal fibrosis via the hedgehog signaling pathway in vivo and in vitro. In rats with unilateral ureteral obstruction (UUO), SSBE administration reduced kidney injury and alleviated interstitial fibrosis by decreasing the levels of transforming growth factor (TGF)­ß1 and its receptor, and inhibiting excessive accumulation of extracellular matrix (ECM) components, including type I and III collagens. In addition, SSBE suppressed the expression of proliferating cell nuclear antigen, and this anti­proliferative activity was associated with downregulation of hedgehog signaling activity in SSBE­treated UUO kidneys. In cultured renal tubular epithelial cells (RTECs), recombinant TGF­ß1 activated hedgehog signaling, and resulted in induction of the myofibroblast phenotype. SSBE treatment inhibited the activation of hedgehog signaling and partially reversed the fibrotic phenotype in TGF­ß1­treated RTECs. Similarly, aristolochic acid­mediated upregulated activity of hedgehog signaling was reduced by SSBE treatment, and thereby led to the abolishment of excessive ECM accumulation. Therefore, these findings suggested that SSBE attenuates the myofibroblast phenotype and renal fibrosis via suppressing the hedgehog signaling pathway, and may facilitate the development of treatments for kidney fibrosis.


Assuntos
Proteínas Hedgehog/metabolismo , Rim/metabolismo , Extratos Vegetais/farmacologia , Sedum/química , Transdução de Sinais/efeitos dos fármacos , Animais , Biomarcadores , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Matriz Extracelular/metabolismo , Fibrose , Regulação da Expressão Gênica , Imuno-Histoquímica , Rim/patologia , Nefropatias/tratamento farmacológico , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Ratos , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
16.
Mediators Inflamm ; 2016: 2174682, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27313397

RESUMO

Background. Macrophage migration inhibitory factor (MIF) is an important immunoregulatory cytokine involved in inflammation, which may be one important reason resulting in matrix deposition in renal tissues after injury. However, the underlying mechanisms have not yet been elucidated. Methods and Results. We uncovered a crucial role of MIF in inflammation and collagen deposition in vivo and in vitro. In rats, ureteral obstruction induced tubular injury, matrix accumulation, and inflammatory cell infiltration. Additionally, enhanced MIF levels in the obstructed kidneys were closely related to the increasing numbers of CD68-positive macrophages. These obstruction-induced injuries can be relieved by recanalization, consequently resulting in downregulated expression of MIF and its receptor CD74. Similarly, ischemia reperfusion induced renal injury, and it was accompanied by elevated MIF levels and macrophages infiltration. In cultured tubular epithelial cells (TECs), aristolochic acid (AA) promoted matrix production and increased MIF expression, as well as the release of macrophage-related factors. Inhibition of MIF with an antagonist ISO-1 resulted in the abolishment of these genotypes in AA-treated TECs. Conclusion. MIF plays an important role in macrophage-related inflammation and matrix deposition in kidney tissues following injury. MIF as a specific inhibitor may have therapeutic potential for patients with inflammatory and fibrotic kidney diseases.


Assuntos
Inflamação/metabolismo , Oxirredutases Intramoleculares/metabolismo , Rim/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Ácidos Aristolóquicos/farmacologia , Western Blotting , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Imunofluorescência , Imuno-Histoquímica , Oxirredutases Intramoleculares/antagonistas & inibidores , Isoxazóis/farmacologia , Rim/imunologia , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Obstrução Ureteral/metabolismo
17.
Oncol Rep ; 35(5): 2775-84, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26987050

RESUMO

Sedum sarmentosum Bunge, a traditional Chinese herbal medicine, has a wide range of clinical applications including antibiosis, anti-inflammation and anti-oxidation. In the present study, we identified that its extract (SSBE) exerts pancreatic anticancer activity in vitro and in vivo. In the cultured pancreatic cancer PANC-1 cell line, SSBE inhibited cell growth in a concentration-dependent manner, and it was accompanied by the downregulated expression of proliferating cell nuclear antigen (PCNA). In addition, SSBE treatment also increased cellular apoptosis in a mitochondrial-dependent manner. Moreover, SSBE induced p53 expression, reduced c-Myc expression, and inhibited epithelial-mesenchymal transition (EMT). The antiproliferative activity of SSBE in the pancreatic cancer cells was found to be closely related to cell cycle arrest at the G2/M phase by upregulating p21(Waf1/CIP1) expression. Further study showed that this inhibitory effect of SSBE was through downregulation of the activity of the proliferation-related Hedgehog signaling pathway. Exogenous recombinant protein Shh was used to activate Hedgehog signaling, thereby resulting in the abolishment of the SSBE-mediated inhibition of pancreatic cancer cell growth. In animal xenograft models of pancreatic cancer, activated Hedgehog signaling was also observed compared with the vehicle controls, but was reduced by SSBE administration. As a result, SSBE suppressed the growth of pancreatic tumors. Thus, these findings demonstrate that SSBE has therapeutic potential for pancreatic cancer, and this anticancer effect in pancreatic cancer cells is associated with inhibition of the Hedgehog signaling pathway.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Extratos Vegetais/farmacologia , Animais , Pontos de Checagem do Ciclo Celular , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Expressão Gênica/efeitos dos fármacos , Proteínas Hedgehog/metabolismo , Humanos , Concentração Inibidora 50 , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Sedum/química , Transdução de Sinais , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Exp Ther Med ; 10(5): 1759-1767, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26640547

RESUMO

BK polyomavirus (BKV) is important pathogen for kidney transplant recipients, as it is frequently re-activated, leading to nephropathy. The aim of this study was to investigate the phylogenetic reconstruction and polymorphism of the VP2 gene in BKV isolated from Chinese kidney transplant recipients. Phylogenetic analysis was carried out in the VP2 region from 135 BKV-positive samples and 28 reference strains retrieved from GenBank. The unweighted pair-group method with arithmetic mean (UPGMA) grouped all strains into subtypes, but failed to subdivide strains into subgroups. Among the plasma and urine samples, all plasma (23/23) and 82 urine samples (82/95) were identified to contain subtype I; the other 10 urine samples contained subtype IV. A 86-bp fragment was identified as a highly conserved sequence. Following alignment with 36 published BKV sequences from China, 92 sites of polymorphism were identified, including 11 single nucleotide polymorphisms (SNPs) prevalent in Chinese individuals and 30 SNPs that were specific to the two predominant subtypes I and IV. The limitations of the VP2 gene segment in subgrouping were confirmed by phylogenetic analysis. The conserved sequence and polymorphism identified in this study may be helpful in the detection and genotyping of BKV.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA