Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Cancer ; 14(9): 1648-1659, 2023.
Article in English | MEDLINE | ID: mdl-37325054

ABSTRACT

Cervical cancer (CC) is one of the leading cancers among the female reproductive system. The piwi-interacting RNA (piRNA) function and biogenesis has been studied in various cancers, including CC. But the precise mechanism of piRNA in CC is still unknown. In our study, we found that piRNA-17458 was overexpressed in CC tissues and cells. piRNA-17458 mimic and inhibitor promoted and suppressed proliferation, migration and invasion ability of CC cells, respectively. We also demonstrated that piRNA-17458 mimic could contribute to tumor growth in mice xenograft models. Besides, we also found that the piRNA-17458 mimic could enhance mRNA N6-methyladenosine(m6A) levels and increase WTAP stability in CC cells, while the effects of the mimic was reversed by the WTAP knockdown. The results of dual luciferase reporter assay showed that WTAP was a direct target of piRNA-17458. Knockdown of WTAP attenuated proliferation, migration and invasion of CC cells in piRNA-17458 mimic group. Our finding not only demonstrates for the first time that piRNA-17458 is overexpressed in CC tissues and cells, but also shows that piRNA-17458 promotes tumorigenesis of CC in a WTAP-mediated m6A methylation manner.

2.
Ecotoxicol Environ Saf ; 259: 115030, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37216864

ABSTRACT

As an emerging endocrine-disrupting component with a chemical structure related to Bisphenol A (BPA), Bisphenol AF (BPAF) has become widely distributed in the environment and human surroundings. Although numerous studies have focused on its reproductive toxicity, the impact of prenatal BPAF exposure on the reproductive system of adult male offspring, particularly testicular morphology and function, as well as the underlying mechanisms, remains largely understudied. This study found prenatal BPAF exposure at a dose of 300 µg/kg b.w. induced a 32% loss of seminal vesicle weight, a 12% reduction in the anogenital distance index (AGI), and impairments to testicular morphology, such as a reduced diameter of seminiferous tubules and thickness of the seminiferous epithelium, as well as a more than 2 - fold decrease in testosterone level, and 41% and 19% reduction of sperm count and vitality, respectively, in the 10 week-old male offsprings. Testicular RNA-Seq data showed that 334 differential expressed genes (DEGs) were primarily involved in several immunological processes, including host defense response, innate and adaptive immune response, cellular response to interferon (IFN)-ß and γ, antigen processing and presentation, regulation of T cell activation, etc. Importantly, our results revealed a pattern recognition receptor - absent in melanoma-2 (Aim2) was significantly increased in the testes of exposed males, thus triggering a testicular innate antiviral immunological response, leading to an increase of F4/80+ and CD11b+ macrophage. Subsequently, Aim2 activated the downstream signaling nuclear factor kappa-B (NF-κB), stimulated the transcription of IFN-ß and -γ, and then induced cytokine production while upregulating MHC class II molecules to activate CD4+ and CD8+ Tcells, suggesting that an adaptive immune response was also elicited. The results demonstrated that prenatal BPAF exposure could provoke innate and adaptive immunological responses in the testes of adult males through the Aim2-NF-κB-IFNs signaling pathway. Our work provided insights into understanding the reproductive toxicity caused by BPAF and clarified the possible mechanisms, which offered a potential therapeutic target and treatment strategy for BPAF exposure-induced reproductive dysfunction.


Subject(s)
Prenatal Exposure Delayed Effects , Testis , Pregnancy , Female , Male , Humans , Testis/metabolism , Prenatal Exposure Delayed Effects/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , CD8-Positive T-Lymphocytes/metabolism , Semen , Benzhydryl Compounds/toxicity , Benzhydryl Compounds/metabolism , Immunity
3.
Biomater Sci ; 10(17): 4822-4836, 2022 Aug 24.
Article in English | MEDLINE | ID: mdl-35858469

ABSTRACT

Inflammatory bowel disease (IBD) is associated with chronic gut immune dysregulation and altered microbiome and metabolite composition. Bile acids and their receptors such as the farnesoid X receptor (FXR) form a crucial component of the chemical communications between the intestinal microbiota and the host immune system; thus, alterations in the bile acid pool affect intestinal homeostasis and exacerbate IBD. Considering the promising therapeutic effect of mesenchymal stem cell-derived exosomes (MSC-Ex) on IBD, this study assessed the regulatory effect of MSC-Ex on the gut bacteria composition and diversity, metabolites, and their related functions and pathways, as well as key inflammatory and anti-inflammatory cytokines during the mitigation of IBD. The dextran sulfate sodium (DSS)-induced IBD model of BABL/C mice was established, consisting of three groups: control, DSS, and MSC-Ex groups. Post administration of MSC-Ex, the effect was evaluated via hematoxylin and eosin (H&E) staining, immunohistochemistry (IHC), qRT-PCR, and western blotting. Mice fecal samples were obtained for metagenomics and metabolomics analysis via 16S rRNA gene sequencing and UHPLC/Q-TOF-MS respectively. Results showed that MSC-Ex mitigated colitis by significantly relieving the macroscopic and microscopic features of inflammation, modulating the gut metagenomics and metabolomics profile, and increasing colonic FXR. MSC-Ex improved the gut microbiota composition by significantly restoring the structure of OTUs and colitis-induced reduction in α-diversity, increasing the abundance of 'healthy' bacteria, decreasing disease-associated bacteria, decreasing detrimental functions, and enhancing other vital cellular functions. For the first time, we demonstrate that MSC-Ex mitigates colitis in mice by modulating the gut metagenomics-metabolomics-FXR axis, thus providing potential therapeutic targets.


Subject(s)
Colitis , Exosomes , Inflammatory Bowel Diseases , Mesenchymal Stem Cells , Animals , Bacteria/genetics , Bile Acids and Salts , Colitis/chemically induced , Dextran Sulfate/toxicity , Disease Models, Animal , Exosomes/metabolism , Inflammatory Bowel Diseases/chemically induced , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred C57BL , RNA, Ribosomal, 16S
4.
Gut Pathog ; 14(1): 26, 2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35729658

ABSTRACT

Inflammatory bowel disease (IBD), a chronic gut immune dysregulation and dysbiosis condition is rapidly increasing in global incidence. Regardless, there is a lack of ideal diagnostic markers, while conventional treatment provides scarce desired results, thus, the exploration for better options. Changes in the gut microbial composition and metabolites either lead to or are caused by the immune dysregulation that characterizes IBD. This study examined the fecal metagenomics and metabolomic changes in IBD patients. A total of 30 fecal samples were collected from 15 IBD patients and 15 healthy controls for 16S rDNA gene sequencing and UHPLC/Q-TOF-MS detection of metabolomics. Results showed that there was a severe perturbation of gut bacteria community composition, diversity, metabolites, and associated functions and metabolic pathways in IBD. This included a significantly decreased abundance of Bacteroidetes and Firmicutes, increased disease-associated phyla such as Proteobacteria and Actinobacteria, and increased Escherichia coli and Klebsiella pneumoniae in IBD. A total of 3146 metabolites were detected out of which 135 were differentially expressed between IBD and controls. Metabolites with high sensitivity and specificity in differentiating IBD from healthy individuals included 6,7,4'-trihydroxyisoflavone and thyroxine 4'-o-.beta.-d-glucuronide (AUC = 0.92), normorphine and salvinorin a (AUC = 0.90), and trichostachine (AUC = 0.91). Moreover, the IBD group had significantly affected pathways including primary bile acid biosynthesis, vitamin digestion and absorption, and carbohydrate metabolism. This study reveals that the combined evaluation of metabolites and fecal microbiome can be useful to discriminate between healthy subjects and IBD patients and consequently serve as therapeutic and diagnostic targets.

5.
Biomed Pharmacother ; 140: 111752, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34044275

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by intense immune dysregulation, gut microbiota imbalance, and intestinal epithelium destruction. Among the factors that contribute to the pathogenesis of IBD, lymphatics have received less attention, hence less studied, characterized, and explored. However, in recent years, the role of the lymphatic system in gastrointestinal pathophysiology continues to be highlighted. This paper examines the implications of lymphatic changes in IBD pathogenesis related to immune cells, gut microbiota, intestinal and mesenteric epithelial barrier integrity, and progression to colorectal cancer (CRC). Therapeutic targets of lymphatics in IBD studies are also presented. Available studies indicate that lymph nodes and other secondary lymphatic tissues, provide highly specialized microenvironments for mounting effective immune responses and that lymphatic integrity plays a significant role in small intestine homeostasis, where the lymphatic vasculature effectively controls tissue edema, leukocyte exit, bacterial antigen, and inflammatory chemokine clearance. In IBD, there are functional and morphological alterations in intestinal and mesenteric lymphatic vessels (more profoundly in Crohn's disease [CD] compared to ulcerative colitis [UC]), including lymphangiogenesis, lymphangiectasia, lymphadenopathy, and lymphatic vasculature blockade, affecting not only immunity but gut microbiota and epithelial barrier integrity. While increased lymphangiogenesis is primarily associated with a good prognosis of IBD, increased lymphangiectasia, lymphadenopathy, and lymphatic vessel occlusion correlate with poor prognosis. IBD therapies that target the lymphatic system seek to increase lymphangiogenesis via induction of lymphangiogenic factors and inhibition of its antagonists. The resultant increased lymphatic flow coupled with other anti-inflammatory activities restores gut homeostasis.


Subject(s)
Inflammatory Bowel Diseases/immunology , Lymphatic Vessels/immunology , Animals , Colorectal Neoplasms/etiology , Colorectal Neoplasms/immunology , Gastrointestinal Microbiome , Humans , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/microbiology
6.
Mol Med Rep ; 19(1): 499-507, 2019 01.
Article in English | MEDLINE | ID: mdl-30483785

ABSTRACT

Diabetic cardiomyopathy is one of the major complications of diabetes, and due to the increasing number of patients with diabetes it is a growing concern. Diabetes­induced cardiomyopathy has a complex pathogenesis and histone deacetylase­mediated epigenetic processes are of prominent importance. The olfactory bromodomain­containing protein 4 (BRD4) is a protein that recognizes and binds acetylated lysine. It has been reported that the high expression of BRD4 is involved in the process of cardiac hypertrophy. The aim of the present study was to investigate the function of BRD4 in the process of high glucose (HG)­induced cardiac hypertrophy, and to clarify whether epigenetic regulation involving BRD4 is an important mechanism. It was revealed that BRD4 expression levels were increased in H9C2 cells following 48 h of HG stimulation. This result was also observed in a diabetic rat model. Furthermore, HG stimulation resulted in the upregulation of the myocardial hypertrophy marker, atrial natriuretic peptide, the cytoskeletal protein α­actin and fibrosis­associated genes including transforming growth factor­ß, SMAD family member 3, connective tissue growth factor and collagen, type 1, α1. However, administration of the specific BRD4 inhibitor JQ1 (250 nM) for 48 h reversed this phenomenon. Furthermore, protein kinase B (AKT) phosphorylation was activated by HG stimulation and suppressed by JQ1. In conclusion, BRD4 serves an important role in the pathogenesis of HG­induced cardiomyocyte hypertrophy through the AKT pathway.


Subject(s)
Cardiomegaly/metabolism , Cardiomegaly/pathology , Diabetic Cardiomyopathies/metabolism , Glucose/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Biomarkers/metabolism , Cell Line , Epigenesis, Genetic/physiology , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats , Rats, Wistar , Up-Regulation/physiology
7.
Cancer Sci ; 109(12): 3762-3773, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30290033

ABSTRACT

Urea Transporter B (UT-B) is a membrane channel protein that mediates the rapid transmembrane transport of urea and participates in urine concentration. Urea Transporter B is expressed in skin, but we found that there is little expression in human melanoma tissue. In this study, we examined the effects of UT-B overexpression in melanoma. The results indicated that there is no UT-B mRNA expression in B16 cells, and UT-B overexpression repressed B16 cell proliferation and induced apoptosis in vitro. We show that UT-B overexpression causes increased reactive oxygen species production, which may be caused by mitochondria dysfunction. The mitochondrial membrane potential (ΨΔm) was lower in UT-B-overexpressing B16 cells. The proteins involved in complexes I, III, IV and V of the respiratory chain were clearly downregulated in UT-B-overexpressing B16 cells, which would strongly reduce the activity of the electron transport chain. We found that mitochondrial release of cytochrome C into the cytoplasm also increased, indicating that apoptosis had been activated. In addition, UT-B overexpression reduced AKT phosphorylation and MDM2 expression and increased p53 expression; p53 activation may be involved in the anticancer effects of UT-B overexpression. Urea Transporter B overexpression also inhibited tumor growth in vivo. In conclusion, we demonstrated that UT-B may be related to the occurrence of melanoma and play a role in tumor development.


Subject(s)
Melanoma, Experimental/metabolism , Melanoma/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mitochondria/physiology , Skin Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Aged , Animals , Cell Death , Cell Line, Tumor , Cytochromes c/metabolism , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/genetics , Melanoma, Experimental/genetics , Membrane Potential, Mitochondrial , Mice , Middle Aged , Signal Transduction , Skin Neoplasms/genetics , Urea Transporters
8.
Mol Med Rep ; 16(6): 9137-9142, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29039579

ABSTRACT

The therapeutic options available for the treatment of advanced non-small cell lung cancer have increased over the past decade. Small molecule gene therapy has emerged as an effective therapy for the treatment of lung cancer in vitro and in vivo although it has not been tested in a clinical setting. In particular, therapies that target the negative feedback loop between p53 and murine double minute 2 (MDM2) provide a favorable outcome by maintaining activation of the tumor suppressor gene p53. The present study used transfection to simultaneously knockdown MDM2 expression using small interfering (si)RNA, and overexpress wild­type p53 in H1299 cells. The effects of transfection on cell proliferation and cell cycle progression were determined using an MTT assay and flow cytometry, and the effects on mRNA and protein expression were determined by western blotting and reverse transcription polymerase chain reaction. The results indicated that simultaneously knocking down MDM2 and overexpressing p53 was able to inhibit proliferation and induce G1 cell cycle arrest in H1299 cells, compared with either alone. These findings indicated that the si­MDM2­p53 co­expression plasmid may induce cell cycle arrest, and may be considered a novel therapeutic option for the treatment of lung cancer.


Subject(s)
Carrier Proteins/genetics , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression , RNA Interference , RNA, Small Interfering/genetics , Tumor Suppressor Protein p53/genetics , Animals , Cell Cycle Checkpoints/genetics , Cell Cycle Proteins/genetics , Cell Line, Tumor , Humans , Mice , Plasmids/genetics , RNA, Messenger/genetics , Transfection
9.
Cancer Immunol Immunother ; 65(6): 677-88, 2016 06.
Article in English | MEDLINE | ID: mdl-27034233

ABSTRACT

The phenotypic diversity of tumor-associated macrophages (TAMs) increases with tumor development. One of the hallmarks of malignancy is the polarization of TAMs from a pro-immune (M1) phenotype to an immunosuppressive (M2) phenotype. However, the molecular basis of this process is still unclear. Endostatin is a powerful inhibitor of angiogenesis capable of suppressing tumor growth and metastasis. Here, we demonstrate that endostatin induces RAW264.7 cell polarization toward the M1 phenotype in vitro. Endostatin has no effect on TAM numbers in vivo, but results in an increased proportion of F4/80(+)Nos2(+) cells and a decreased proportion of F4/80(+)CD206(+) cells. Overexpression of endostatin in RAW264.7 cells resulted in a decrease in the phosphorylation of STAT3, an increase in expression of vascular endothelial growth factor A and placental growth factor, and an increase in the phosphorylation of STAT1, IκBα and p65 proteins compared with controls. These results indicate that endostatin regulates macrophage polarization, promoting the M1 phenotype by targeting NF-κB and STAT signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/immunology , Endostatins/pharmacology , Macrophages/drug effects , Macrophages/physiology , Phenotype , Animals , Biomarkers , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Endostatins/genetics , Female , Gene Expression , Mice , Tumor Burden/drug effects
10.
Mol Med Rep ; 13(1): 461-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26573068

ABSTRACT

Breast cancer urgently requires improved therapeutic strategies. In the current study, a Pvp53 plasmid that co­expressed p53 and short­interfering RNA against vascular endothelial growth factor (si­VEGF) was developed to replace single plasmid transfections. Whether Pvp53 exhibited improved anti­tumor effects in breast cancer MDA­MB­231 cells was investigated in the present study. Pvp53 significantly reduced the Bcl­2/Bax ratio and increased the expression of cleaved caspase­3 and 8. Compared with p53 and si­VEGF single transfections, the Pvp53 co­expression plasmid significantly increased the proportion of apoptotic cells and inhibited cell motility and proliferation. These results indicated that the Pvp53 co­expression plasmid has greater inhibitory effects on breast cancer MDA­MB­231 cells than single plasmids.


Subject(s)
Plasmids/metabolism , RNA, Small Interfering/metabolism , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/metabolism , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Shape , Cell Survival , Gene Expression Regulation, Neoplastic , Humans , Transfection , Tumor Suppressor Protein p53/genetics , Vascular Endothelial Growth Factor A/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...