Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
1.
Int J Antimicrob Agents ; 63(1): 107002, 2024 Jan.
Article En | MEDLINE | ID: mdl-37838150

Antibiotic resistance has become a major threat, contributing significantly to morbidity and mortality globally. Administering non-antibiotic therapy, such as antimicrobial peptides, is one potential strategy for effective treatment of multi-drug-resistant Gram-negative bacterial infections. Bactericidal/permeability-increasing protein (BPI) derived from neutrophils has bactericidal and endotoxin-neutralizing activity. However, the protective roles and mechanisms of BPI in multi-drug-resistant bacterial infections have not been fully elucidated. In this study, a chimeric BPI23-Fcγ recombined protein comprising the functional N terminus of BPI and Fcγ was constructed and expressed by adenovirus vector 5 (Ad5). Ad5-BPI23-Fcγ or recombinant BPI23-Fcγ protein significantly improved the survival of mice with pneumonia induced by a minimal lethal dose of multi-drug-resistant Acinetobacter baumannii or Klebsiella pneumoniae by ameliorating lung pathology and reducing pro-inflammatory cytokines. Transfection with Ad5-BPI23-Fcγ significantly decreased the bacterial load and endotoxaemia, which was associated with enhanced bactericidal ability and elevated the phagocytic activity of neutrophils in vitro and in vivo. In addition, Ad5-BPI23-Fcγ transfection significantly increased the recruitment of neutrophils to lung, increased the proportion and number of neutrophils in peripheral blood, and promoted the maturation of bone marrow (BM) neutrophils after drug-resistant A. baumannii infection. BPI23-Fcγ and neutrophils synergistically enhanced bactericidal activity and decreased pro-inflammatory cytokines. These results demonstrated that the chimeric BPI23-Fcγ protein protected mice from pneumonia induced by multi-drug-resistant A. baumannii infection by direct bactericidal effects and promotion of neutrophil recruitment, phagocytosis and maturation. Chimeric BPI23-Fcγ may be a promising candidate as a non-antibiotic biological agent for multi-drug-resistant A. baumannii infection.


Acinetobacter baumannii , Pneumonia , Animals , Mice , Neutrophils , Receptors, IgG , Membrane Proteins , Cytokines , Pneumonia/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use
2.
Cancer Med ; 12(19): 19491-19499, 2023 10.
Article En | MEDLINE | ID: mdl-37724594

OBJECTIVE: To evaluate the risk of myasthenia gravis (MG) associated with immune checkpoint inhibitors (ICI). METHODS: Adverse event (AE) reports related to MG, myasthenic syndrome, and MG crisis for durvalumab, atezolizumab, pembrolizumab, nivolumab, avelumab, and ipilimumab in the US FDA Adverse Event Reporting System (FAERS) from Q1 2004 to Q3 2022 were collected. The proportional reporting odds ratio (PRR) method was used to evaluate the correlation between the six drugs and the three AEs. Statistical significance was defined as having reports ≥3, PRR ≥ 2, and chi-square (χ2 ) ≥ 4. RESULTS: A total of 36, 78, 276, 380, 5, and 53 AE reports were collected for durvalumab, atezolizumab, pembrolizumab, nivolumab, avelumab, and ipilimumab, respectively. For myasthenic syndrome, the PRR values reflecting the correlation with the drugs were 27.83 (χ2 = 102.66), 26.20 (χ2 = 235.67), 44.17 (χ2 = 1313.98), 32.09 (χ2 = 1229.54), 21.31 (χ2 = 151.15), and 0 for durvalumab, atezolizumab, pembrolizumab, nivolumab, avelumab, and ipilimumab, respectively. For MG, the PRR values reflecting the correlation with the drugs were 24.21 (χ2 = 682.04), 18.34 (χ2 = 900.27), 39.32 (χ2 = 7945.15), 26.93 (χ2 = 6636.45), 14.73 (χ2 = 566.47), and 15.69 (χ2 = 54.77) for durvalumab, atezolizumab, pembrolizumab, nivolumab, avelumab, and ipilimumab, respectively. For MG crisis, there were no data for durvalumab, atezolizumab, avelumab, and ipilimumab; the PRR values reflecting the correlation with the drugs were 16.54 (χ2 = 225.23) and 9.20 (χ2 = 119.14) for pembrolizumab and nivolumab, respectively. All six drugs were statistically correlated with their corresponding AEs. CONCLUSIONS: ICI may lead to ICIs-associated MG during therapy. Analysis of FAERS data identified signals for AEs of MG with ICI regimens. Practitioners should consider the factors that may increase the likelihood of MG. The findings support a continued surveillance and risk factor identification.


Antineoplastic Agents, Immunological , Myasthenia Gravis , Neoplasms , Humans , Nivolumab/therapeutic use , Ipilimumab/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Neoplasms/drug therapy , Myasthenia Gravis/chemically induced , Myasthenia Gravis/epidemiology , Myasthenia Gravis/complications
3.
Hand Surg Rehabil ; 42(6): 530-535, 2023 Dec.
Article En | MEDLINE | ID: mdl-37776975

PURPOSE: This study aimed to introduce a technique of external fixation using a combination of bone cement and K-wires, to treat pathological fractures related to solitary digital enchondroma close to the finger joints. METHODS: From October 2015 to January 2021, 21 patients (8 males and 13 females) with acute pathological fracture due to solitary digital enchondroma close to the finger joints were treated with cemented K-wire external fixators. Mean age was 32 (19-51) years. The digits involved were the index (n = 4), long (n = 4), ring (n = 6), and little (n = 7) fingers. Time to bone healing and complications were assessed. At final follow-up, active range of motion, grip strength and key pinch strength of the tumor-involved and contralateral healthy digits were measured and compared. Functional outcomes were evaluated on Takigawa criteria. Pain was measured on a 10-cm visual analog scale. We assessed the affected upper extremity on the Musculoskeletal Tumor Society score questionnaire. RESULTS: Mean bone healing time was 5.5 (4-8) weeks. Pin site infection was observed in 1 patient and cured with dressing changes. Mean follow-up was 34 months, with no recurrences or refractures. Mean active range of motion of the proximal interphalangeal joint, grip and key pinch strength of the involved digits reached 92%, 97%, and 99% of the contralateral digits, respectively. On Takigawa criteria, 20 functional results were excellent and 1 good. Mean pain score was 1 (0-3) cm. Mean Musculoskeletal Tumor Society score was 95 (80-100). CONCLUSION: The combination of bone cement and K-wires is a reliable technique for pathological fracture related to solitary enchondroma close to the joints of the digits, leading to good functional outcomes. LEVEL OF EVIDENCE: Therapeutic study, Level IVa.


Chondroma , Fractures, Bone , Fractures, Spontaneous , Male , Female , Humans , Adult , External Fixators , Bone Cements , Fracture Fixation , Fractures, Bone/surgery , Chondroma/diagnostic imaging , Chondroma/surgery , Pain
4.
Ann Transl Med ; 9(12): 1024, 2021 Jun.
Article En | MEDLINE | ID: mdl-34277824

BACKGROUND: Percutaneous kyphoplasty (PKP) is a highly practical technology to treat osteoporotic vertebral compression fractures (OVCFs). However, the operation time and radiation exposure remain problematic. This study explored the differences in surgical effects and safety between a novel steerable percutaneous kyphoplasty (S-PKP) and traditional PKP in order to achieve better clinical outcomes for OVCF patients. It is also exploring whether the new technology could reduce the radiation exposure. METHODS: This study recruited 72 patients (between March 2019 and January 2020) with OVCFs (single vertebra). The patients were semi-randomly divided these patients into two groups according to ID numbers: a S-PKP group (33 cases) and a PKP group (39 cases). We evaluated the clinical efficacy using the kyphotic Cobb angle, Oswestry disability index (ODI), visual analogue scale (VAS) score, injected cement volume, operation time, intraoperative radiation times, bone cement leakage, and postoperative complications. Patients were followed up once preoperatively, and at 1 day, 6 months, and 1 year postoperatively. RESULTS: There were no cases of cement leakage or postoperative complications. There were no significant differences in gender, age, Bone mineral density T-score (BMD T) value, Cobb angle between the two groups (P>0.05). Intraoperative bone cement injection was approximately 5.25±1.37 and 5.32±1.29 mL in the PKP and S-PKP groups respectively. The postoperative VAS score and ODI of the two groups at 1 day, 6 months, and 1 year were markedly lower than before (P<0.05). There was a considerable improvement in the Cobb angle postoperatively (P<0.05). However, as the follow-up time extended, the Cobb Angle increased. The operation time and X-ray exposure times of patients in the PKP group were notably higher than those in the S-PKP group. The operation time was 51.59±9.14 min in the PKP group and 30.76±4.82 min in the S-PKP group. The frequency of intraoperative radiation was 105.9±31.93 times in the PKP group and 47.42±11.88 times in the S-PKP group. CONCLUSIONS: Early results showed that S-PKP is a safe and efficient method for the treatment of OVCFs. S-PKP can reduce the operation time and radiation exposure. TRIAL REGISTRATION: Chinese Clinical Trial Registry ChiCTR2100046727.

5.
Anim Cells Syst (Seoul) ; 24(5): 267-274, 2020 Sep 10.
Article En | MEDLINE | ID: mdl-33209200

Gastric cancer is a leading cause of cancer death worldwide. Endoplasmic reticulum (ER) stress-induced apoptosis has been confirmed to be important in the treatment of gastric cancer. MiR-637 has recently been found to exert inhibitory effects on gastric cancer, and this study aimed to investigate whether miR-637 could regulate apoptosis through ER stress. The results showed that tunicamycin (TM) induced downregulation of miR-637 in gastric cancer cells (AGS) and increase of apoptosis and ER stress. Overexpression of miR-637 promoted TM-induced apoptosis and expression of ER stress associated proteins (GRP78 and CHOP), but inhibited expression of Calreticulin. MiR-637 could bind with the 3'-UTR of CALR, and negatively regulated the expression of CALR. The co-transfection of miR-637 and CALR in AGS cells show that, CALR overexpression could reverse the pro-apoptosis effects of miR-637 in TM-treated cells. In conclusion, the present study suggests that miR-637 participates in ER stress-induced apoptosis in gastric cancer cells by suppressing CALR expression. miR-637 or CALR may be a future potential target for gastric cancer treatment.

6.
Front Immunol ; 11: 614169, 2020.
Article En | MEDLINE | ID: mdl-33552078

Objective: The objective of this study was to understand the role of bactericidal permeability increasing protein (BPI) in the pathogenesis of experimental murine colitis. Methods: We used the Cre-LoxP system to generate BPI knockout (BPI KO) mice. Acute colitis was induced in BPI KO mice and wild-type (WT) mice by subjecting the mice to 5% dextran sulfate sodium (DSS). Mice were observed for symptoms of experimental colitis. The survival of BPI KO mice to infection with Acinetobacter baumannii, a gram-negative bacterium, was also assessed. Results: Southern blot, RT-PCR, and western blot results showed that the 2nd and 3rd exons of the murine Bpi gene were knocked out systemically, confirming successful construction of the BPI KO mouse. BPI KO mice subjected to DSS showed increased symptoms of experimental colitis, increased colonic mucosal damage, increased epithelial permeability, elevated levels of serum LPS, and a disrupted fecal microbiome as compared with WT mice. Furthermore, BPI KO mice challenged intraperitoneally with A. baumannii died sooner than WT mice, and the total number of bacteria in the abdominal cavity, spleen, and liver was increased in BPI KO mice as compared to WT mice. Conclusions: We successfully generated BPI KO mice. The BPI KO mice developed worse colitis than WT mice by increased colitis symptoms and colonic mucosal damage, elevated levels of serum LPS, and a disrupted microbiome. BPI could be a potential target for treatment of ulcerative colitis in humans.


Acinetobacter Infections/immunology , Acinetobacter baumannii/immunology , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Colitis/metabolism , Lipopolysaccharides/blood , Abdomen/microbiology , Acinetobacter Infections/mortality , Animals , Antimicrobial Cationic Peptides/genetics , Blood Proteins/genetics , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Feces/microbiology , Female , Gene Knockout Techniques , Heterozygote , Liver/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microbiota , Spleen/microbiology
7.
Biol Res ; 52(1): 61, 2019 Dec 23.
Article En | MEDLINE | ID: mdl-31870440

BACKGROUND: Papillary thyroid cancer (PTC) is the most common malignancy of all thyroid cancers. LncRNA LINC00460 has been proved to play roles in the oncogenesis and progression of various tumors, including papillary thyroid cancer. However, the potential molecular mechanism of LINC00460 in PTC is poorly investigated. RESULTS: LINC00460 was upregulated in PTC tissues and cells. Raf1 was upregulated in PTC tissues, but miR-485-5p was down-regulated. High LINC00460 expression was associated with poor prognosis. LINC00460 knockdown suppressed proliferation, migration, invation and EMT of PTC cells. Bioinformatics prediction revealed that LINC00460 had binding sites with miR-485-5p, which was validated by luciferase reporter assay. In addition, miR-485-5p was confirmed to directly target Raf1 3'-UTR. Moreover, LINC00460 promoted PTC progression by sponging miR-485-5p to elevate the expression of Raf1. Knockdown of LINC00460 restrained tumor growth in vivo. CONCLUSION: LINC00460 induced proliferation, migration, invation and EMT of PTC cells by regulating the LINC00460/miR-485-5p/Raf1 axis, which indicated that LINC00460 may be a potential biomarker and therapeutic target for PTC.


MicroRNAs/genetics , RNA, Long Noncoding/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology , Apoptosis , Cell Proliferation , Cell Transformation, Neoplastic , Disease Progression , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Neoplasm Staging , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Up-Regulation
8.
Biol. Res ; 52: 61, 2019. tab, graf
Article En | LILACS | ID: biblio-1100913

BACKGROUND: Papillary thyroid cancer (PTC) is the most common malignancy of all thyroid cancers. LncRNA LINC00460 has been proved to play roles in the oncogenesis and progression of various tumors, including papillary thyroid cancer. However, the potential molecular mechanism of LINC00460 in PTC is poorly investigated. RESULTS: LINC00460 was upregulated in PTC tissues and cells. Raf1 was upregulated in PTC tissues, but miR-485-5p was down-regulated. High LINC00460 expression was associated with poor prognosis. LINC00460 knockdown suppressed proliferation, migration, invation and EMT of PTC cells. Bioinformatics prediction revealed that LINC00460 had binding sites with miR-485-5p, which was validated by luciferase reporter assay. In addition, miR-485-5p was confirmed to directly target Raf1 3'-UTR. Moreover, LINC00460 promoted PTC progression by sponging miR-485-5p to elevate the expression of Raf1. Knockdown of LINC00460 restrained tumor growth in vivo. CONCLUSION: LINC00460 induced proliferation, migration, invation and EMT of PTC cells by regulating the LINC00460/miR-485-5p/Raf1 axis, which indicated that LINC00460 may be a potential biomarker and therapeutic target for PTC.


Humans , Male , Female , Middle Aged , Thyroid Neoplasms/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Up-Regulation , Cell Transformation, Neoplastic , Apoptosis , Disease Progression , Cell Proliferation , Thyroid Cancer, Papillary/genetics , Neoplasm Staging
9.
Biochimie ; 147: 98-104, 2018 Apr.
Article En | MEDLINE | ID: mdl-29360495

Micro-RNAs have been reported to play crucial roles in a diversity of cellular processes such as cell proliferation, differentiation and development by regulating the expression of specific genes. They have also been shown to play vital roles in several diseases such as cancer. In the present study, we investigated the role of miR-143-3p in breast cancer. Our results showed that the expression of miR-143-3p is significantly downregulated in breast cancer cells. Upregulation of miR-143-3p inhibited the proliferation and migration of breast cancer cells. Conversely, inhibition of miR-143-3p promoted the proliferation of cancer cells. Bioinformatics analysis and other several experiments revealed MAPK7 as the potential target of miR-143-3p. Quantitative RT-PCR showed that the expression of MAPK7 correlated well with the expression of miR-143. Moreover, the inhibition of MAPK 7 in breast cancer cells abrogated the effects of miR-143 indicating that miR-143-3p-exerted effects on breast cancer are mediated by MAPK7. Takentogether, these results provide strong clues about the therapeutic potential of miR-143-3p in the treatment of breast cancer.


Breast Neoplasms/pathology , Cell Movement/genetics , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 7/genetics , Base Sequence , Cell Line, Tumor , Cell Proliferation/genetics , Humans , Neoplasm Invasiveness
10.
Biochem Biophys Res Commun ; 495(2): 1594-1600, 2018 01 08.
Article En | MEDLINE | ID: mdl-29217194

Long non-coding RNAs (lncRNA) have been demonstrated to act as essential regulators in the development and progression of breast cancer. In our study, we found that long noncoding RNA SNHG15 was highly expressed in breast cancer tissues and cell lines. And the expression of SNHG15 was correlated with TNM stage, lymphnode metastasis and survival in breast cancer patients. SNHG15 knockdown significantly inhibited the proliferation and induced apoptosis in breast cancer cells in vitro and in vivo. Besides, SNHG15 downregulation suppressed cell migration and invasion in MCF-7 and BT-20 cells, and inhibited epithelial-mesenchymal transition (EMT). In mechanism, we found that SNHG15 acted as a competing endogenous RNA to sponge miR-211-3p, which was downregulated in breast cancers and inhibited cell proliferation and migration. Our results showed that there was a negative correlation between SNHG15 and miR-211-3p expression in breast cancer patients. Collectively, we, for the first time, revealed the functions of SNHG15 and miR-211-3p in breast cancer.


Breast Neoplasms/genetics , Breast Neoplasms/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Animals , Apoptosis/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Heterografts , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , RNA, Neoplasm/antagonists & inhibitors , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
11.
Mol Med Rep ; 16(6): 9685-9691, 2017 Dec.
Article En | MEDLINE | ID: mdl-29039535

In previous years, progranulin (PGRN) has attracted increasing attention due to its oncogenic roles in several types of tumor. However, the clinical relevance of PGRN in gastric cancer remains to be elucidated. In the present study, 120 retrospective tissue samples were obtained from patients with primary gastric cancer, and the expression of PGRN was detected using immunohistochemistry. The results showed that 71 cases exhibited a high expression of PGRN, which was markedly higher than the 49 cases with a low expression of PGRN. Subsequent χ2 analysis confirmed for the first time, to the best of our knowledge, that a high level of PGRN was positively correlated with lymph node metastasis (P=0.048), lymphatic invasion (P=0.018) and advanced clinical stage (P=0.027). Survival analysis showed that PGRN was positively correlated with poorer overall survival (OS; P=0.0043) and progression­free survival (PFS; P=0.0022). Univariate and multivariate Cox regression analysis showed that PGRN and clinical stage had a significant effect on the OS and PFS of the patients with gastric cancer. In addition, cell experiments confirmed that extracellular PGRN promoted the intracellular expression of PGRN in a concentration­dependent manner in gastric cancer cells. The AKT and extracellular signal­regulated kinase signaling pathways were involved in the upregulation of intracellular PGRN induced by extracellular PGRN in MKN­45 and MGC­803 gastric cancer cells. Taken together, the results of the present study suggested that PGRN may be important in the progression and prognosis of gastric cancer, and that the expression of PGRN was regulated in a positive feedback loop. These findings enhance current knowledge regarding PGRN in tumors.


Extracellular Signal-Regulated MAP Kinases/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Stomach Neoplasms/metabolism , Adult , Aged , Cell Line, Tumor , Female , Gene Expression , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Progranulins , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology
12.
APMIS ; 124(10): 888-95, 2016 Oct.
Article En | MEDLINE | ID: mdl-27501062

Bactericidal/permeability increasing (BPI) is an antibiotic protein which kills Gram-negative bacteria and neutralizes endotoxin. We have previously developed a recombinant adeno-associated virus which contains human BPI amino acid residues 1-199 and Fc fragment of human IgG1 gene (AAV-hBPI-Fc) and shown that the recombinant virus can protect mice from lethal endotoxemia. However, whether AAV-hBPI-Fc can be used in vivo for the long term remains unclear. To address this, we established an adeno-associated virus-containing mouse BPI and Fc fragment genes (muBPI-Fc) and compared antigenicity of these recombinant proteins in murine models. Immunohistochemistry showed the expression of both fusion proteins at injected sites. ELISA and Western blotting showed that the muBPI-Fc protein was detected in serum up to 8 weeks after injection, without generation of autoantibodies against muBPI-Fc. In contrast, expressed hBPI-Fc protein was only detected on the 2nd week, whereas the autoantibody against hBPI-Fc protein occurred in serum from the 4th week to the end of study. muBPI-Fc also reduced production of proinflammatory cytokines and protected mice from endotoxemia and bacteremia. Our data showed that AAV-muBPI-Fc has potential long-term efficacy as an anti-endotoxin and has anti-bacterial activity in mice, suggesting the potential clinical application of AAV-hBPI-Fc, such as in endotoxin shock.


Biological Therapy/methods , Endotoxemia/prevention & control , Endotoxemia/therapy , Escherichia coli Infections/prevention & control , Escherichia coli Infections/therapy , Adenoviridae/genetics , Animals , Anti-Bacterial Agents/metabolism , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/genetics , Blood Proteins/metabolism , Blotting, Western , Disease Models, Animal , Drug Carriers , Enzyme-Linked Immunosorbent Assay , Female , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Mice, Inbred BALB C , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serum/chemistry , Time Factors , Treatment Outcome
13.
Biochem Biophys Res Commun ; 430(1): 364-9, 2013 Jan 04.
Article En | MEDLINE | ID: mdl-23154182

Interleukin (IL)-35 is a novel heterodimeric cytokine in the IL-12 family and is composed of two subunits: Epstein-Barr virus-induced gene 3 (EBI3) and IL-12p35. IL-35 is expressed in T regulatory (Treg) cells and contributes to the immune suppression function of these cells. In contrast, we found that both IL-35 subunits were expressed concurrently in most human cancer cell lines compared to normal cell lines. In addition, we found that TNF-α and IFN-γ stimulation led to increased IL-35 expression in human cancer cells. Furthermore, over-expression of IL-35 in human cancer cells suppressed cell growth in vitro, induced cell cycle arrest at the G1 phase, and mediated robust apoptosis induced by serum starvation, TNF-α, and IFN-γ stimulation through the up-regulation of Fas and concurrent down-regulation of cyclinD1, survivin, and Bcl-2 expression. In conclusion, our results reveal a novel functional role for IL-35 in suppressing cancer activity, inhibiting cancer cell growth, and increasing the apoptosis sensitivity of human cancer cells through the regulation of genes related to the cell cycle and apoptosis. Thus, this research provides new insights into IL-35 function and presents a possible target for the development of novel cancer therapies.


Apoptosis/physiology , Cell Proliferation , Interleukin-12 Subunit p35/biosynthesis , Interleukins/biosynthesis , Neoplasms/metabolism , Neoplasms/pathology , Tumor Suppressor Proteins/biosynthesis , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cyclin D1/biosynthesis , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Interferon-gamma/pharmacology , Minor Histocompatibility Antigens , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Survivin , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation , fas Receptor/biosynthesis
14.
Int J Oncol ; 41(4): 1419-24, 2012 Oct.
Article En | MEDLINE | ID: mdl-22797702

Cytokine-like 1 (CYTL1) was first identified in CD34+ cells derived from bone marrow and cord blood. The biological functions of CYTL1 remain largely unknown. Here, we reveal a relationship between CYTL1 expression and the biological characteristics of neuroblastoma (NB). The expression of CYTL1 was detected in 10 human tumor cell lines and human NB tissues by RT-PCR and real-time PCR. The inhibitory effect of CYTL1 knockdown on the proliferation, migration and invasion of SH-SY5Y human neuroblastoma cells was studied using the CCK-8 assay and Transwell chamber assays. Among the 10 human tumor cell lines that we examined, CYTL1 was expressed only in SH-SY5Y human neuroblastoma cells. Furthermore, we also observed high levels of CYTL1 expression in human NB tissues. When CYTL1 expression was blocked by siRNA, SH-SY5Y cells showed decreased proliferation, migration and invasion activities. Taken together, our results showed the first evidence of CYTL1 expression in SH-SY5Y neuroblastoma cells and human NB tissues, revealed a possible link between CYTL1 and NB development, and suggested CYTL1 as a potential therapeutic target and diagnosis biomarker for NB.


Cell Proliferation , Neoplasm Metastasis/genetics , Neuroblastoma/genetics , Receptors, Cytokine/genetics , Cell Line, Tumor , Cell Movement/genetics , Child , Child, Preschool , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Male , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/pathology , Neuroblastoma/pathology , RNA, Small Interfering/genetics , Receptors, Cytokine/antagonists & inhibitors , Receptors, Cytokine/biosynthesis
15.
J Med Microbiol ; 61(Pt 9): 1262-1269, 2012 Sep.
Article En | MEDLINE | ID: mdl-22683658

Infections caused by Gram-negative bacteria (GNB) are increasingly common and can result in significant mortality rates due to the development of sepsis. To examine the potential usage of a recombinant Ad5-BPI(23)-Fcγ1 virus as a biological treatment against systemic infection by GNB, a construct containing the human bactericidal/permeability increasing protein (BPI) gene, encoding the functional N terminus (amino acid residues 1-199) of human BPI, and the Fcγ1 gene, encoding the Fc segment of human immunoglobulin G1, was inserted into an adenovirus serotype 5 (Ad5) chromosome to produce a recombinant Ad5-BPI(23)-Fcγ1 virus. Human A549 cells in culture and BALB/c mice were infected with the recombinant Ad5-BPI(23)-Fcγ1 virus and BPI(23)-Fcγ1 expression was confirmed by Western blot analysis and ELISA. The concentrations of BPI(23)-Fcγ1 protein were 5.59 µg ml(-1) in vitro and 21.37 ng ml(-1) in vivo and it was observed that these concentrations were sufficient to decrease endotoxin concentrations while enhancing bactericidal activity. In addition, mice treated with the recombinant Ad5-BPI(23)-Fcγ1 virus had decreased levels of IL-1ß and TNF-α and were protected from an E. coli O111 : B4 challenge. Our data support the concept that Ad5-mediated BPI(23)-Fcγ1 gene delivery could be used as an ancillary biological treatment in the management of infection caused by GNB.


Adenoviridae/genetics , Antimicrobial Cationic Peptides/genetics , Blood Proteins/genetics , Escherichia coli Infections/prevention & control , Genetic Therapy/methods , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Recombinant Fusion Proteins/genetics , Adenoviridae/pathogenicity , Animals , Antimicrobial Cationic Peptides/metabolism , Blood Proteins/metabolism , Cell Line , Disease Models, Animal , Endotoxemia/prevention & control , Escherichia coli/pathogenicity , Genetic Vectors , Humans , Immunoglobulin Fc Fragments/metabolism , Mice , Mice, Inbred BALB C , Recombinant Fusion Proteins/metabolism , Transfection , Treatment Outcome
16.
Virulence ; 3(3): 309-18, 2012 May 01.
Article En | MEDLINE | ID: mdl-22546898

A new hypervirulent (hypermucoviscous) clinical variant of Klebsiella pneumoniae (hvKP) has emerged over the last decade. Our goal is to identify new mechanisms, which increase the virulence hvKP. It has been shown that hvKP strains produce more biofilm than "classical" stains of K. pneumoniae, therefore we hypothesized that biofilm formation may contribute to the pathogenesis of systemic infection. To test this hypothesis, transposon mutants of the model pathogen hvKP1 were generated and screened for decreased production of biofilm. Three mutant constructs with disruptions in glnA [putatively encodes glutamine synthetase, hvKP1 glnA:: EZ::TN < KAN-2 > (glnA::Tn)], sucD [putatively encodes succinyl-CoA synthase α subunit, hvKP1 sucD:: EZ::TN < KAN-2 > (sucD::Tn)], and tag [putatively encodes transcriptional antiterminator of glycerol uptake operon, hvKP1 tag:: EZ::TN < KAN-2 > (tag::Tn)] were chosen for further characterization and use in biologic studies. Quantitative assays performed in rich laboratory medium and human ascites confirmed the phenotype and a hypermucoviscosity assay established that capsule production was not affected. However, compared with its wild-type parent, neither planktonic cells nor biofilms of glnA::Tn, sucD::Tn and tag::Tn displayed a change to the bactericidal activity of 90% human serum. Likewise, when assessed in a rat subcutaneous abscess model, the growth and survival of glnA::Tn, sucD::Tn and tag::Tn in abscess fluid was similar to hvKP1. In this report we identify three new genes that contribute to biofilm formation in hvKP1. However, decreased biofilm production due to disruption of these genes does not affect the sensitivity of these mutant constructs to 90% human serum when in planktonic form or within a biofilm. Further, their virulence in an in vivo abscess model was unaffected.


Abscess/microbiology , Biofilms/growth & development , Klebsiella Infections/microbiology , Klebsiella pneumoniae/physiology , Klebsiella pneumoniae/pathogenicity , Microbial Viability , Polysaccharides, Bacterial/metabolism , Virulence Factors/metabolism , Animals , Blood Bactericidal Activity , DNA Transposable Elements , Disease Models, Animal , Gene Knockout Techniques , Humans , Klebsiella pneumoniae/genetics , Klebsiella pneumoniae/immunology , Male , Mutagenesis, Insertional , Rats , Rats, Long-Evans , Serum/immunology , Virulence Factors/genetics , Young Adult
17.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 26(11): 1143-5, 2010 Nov.
Article Zh | MEDLINE | ID: mdl-21055353

AIM: To study the affinity of endotoxin for three antibacterial peptides derived from N-terminal domain of bactericidal/permeability-increasing protein(BPI). METHODS: To design and synthesize three peptides from N-terminal domain of BPI, BPI22-36, BPI85-99 and BPI147-161.Surface plasmon resonance(SPR) was used to monitor the interaction between LPS/lipid A and the peptides and polymyxin B(PMB) immobilized on CM5 sensor chip. RESULTS: In three peptides, BPI147-161 was proved to have a best binding capacity with LPS/lipid A, followed by BPI85-99, but BPI22-36 could not interact with LPS/lipid A.The affinity constant K(A) of BPI147-161 with lipid A was 1.12 x 106 L/mol. In contrast, the K(A) of PMB was 5.58 x 106 L/mol. CONCLUSION: The results suggest that the peptide BPI147-161 from BPI effectively neutralize endotoxin and probably provide a novel treatment method for septic shock.


Antimicrobial Cationic Peptides/chemistry , Blood Proteins/chemistry , Endotoxins/chemistry , Surface Plasmon Resonance/methods , Lipid A/chemistry , Lipopolysaccharides/chemistry
18.
BMC Cancer ; 10: 467, 2010 Sep 01.
Article En | MEDLINE | ID: mdl-20809956

BACKGROUND: Overexpression of Bmi-1 has been observed in a variety of cancers, and it has been suggested to be an independent prognostic marker for the patients. The objective of this study was to determine the level of Bmi-1 expression or its autoantibodies in human esophageal squamous cell carcinoma (ESCC) and to correlate it with clinicopathologic data. METHODS: We first examined Bmi-1 expression in ESCC cell lines and tumor samples by RT-PCR and Western blot analysis. We then analyzed Bmi-1 protein expression in 171 clinicopathologically characterized ESCC cases by immunohistochemistry. In addition, we detected its autoantibodies in sera of patients with ESCC by ELISA. RESULTS: We found that Bmi-1 expression was higher in the immortalized cells, cancer cell lines and most cancer tissue than in non-tumorous control tissue at both mRNA and protein level. In addition, Bmi-1 expression was observed in 64.3% (110 of 171) archive ESCC specimen by immunohistochemistry analysis, and the location of Bmi-1 in ESCC was in the nuclei instead of cytoplasm of tumor cells. There was a significant difference of Bmi-1 expression in patients categorized according to stage (P = 0.003) and pN classification (P = 0.047). Multivariate analysis suggested that Bmi-1 expression was an independent prognostic marker for ESCC patients. A prognostic significance of Bmi-1 was also found in the subgroup of T3~T4 and N1 tumor classification. Bmi-1 autoantibodies were detected in sera of 39.0% (62 of 159) ESCC patients. The correlations between anti-Bmi-1 antibodies and tumor stage (P = 0.040), or lymph node status (P < 0.001) were significant. CONCLUSIONS: Our results suggest that Bmi-1 protein is a valuable marker of ESCC progression. The presence of Bmi-1 autoantibodies in sera from patients with ESCC may have clinical utility in esophageal cancer diagnosis.


Autoantibodies/blood , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Nuclear Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Adult , Aged , Aged, 80 and over , Blotting, Western , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/secondary , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Staging , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Polycomb Repressive Complex 1 , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/immunology , RNA, Messenger/genetics , Repressor Proteins/genetics , Repressor Proteins/immunology , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Tumor Cells, Cultured
19.
PLoS Pathog ; 6(6): e1000940, 2010 Jun 03.
Article En | MEDLINE | ID: mdl-20532215

It has been recently reported that a side population of cells in nasopharyngeal carcinoma (NPC) displayed characteristics of stem-like cancer cells. However, the molecular mechanisms underlying the modulation of such stem-like cell populations in NPC remain unclear. Epstein-Barr virus was the first identified human tumor virus to be associated with various malignancies, most notably NPC. LMP2A, the Epstein-Barr virus encoded latent protein, has been reported to play roles in oncogenic processes. We report by immunostaining in our current study that LMP2A is overexpressed in 57.6% of the nasopharyngeal carcinoma tumors sampled and is mainly localized at the tumor invasive front. We found also in NPC cells that the exogenous expression of LMP2A greatly increases their invasive/migratory ability, induces epithelial-mesenchymal transition (EMT)-like cellular marker alterations, and stimulates stem cell side populations and the expression of stem cell markers. In addition, LMP2A enhances the transforming ability of cancer cells in both colony formation and soft agar assays, as well as the self-renewal ability of stem-like cancer cells in a spherical culture assay. Additionally, LMP2A increases the number of cancer initiating cells in a xenograft tumor formation assay. More importantly, the endogenous expression of LMP2A positively correlates with the expression of ABCG2 in NPC samples. Finally, we demonstrate that Akt inhibitor (V) greatly decreases the size of the stem cell side populations in LMP2A-expressing cells. Taken together, our data indicate that LMP2A induces EMT and stem-like cell self-renewal in NPC, suggesting a novel mechanism by which Epstein-Barr virus induces the initiation, metastasis and recurrence of NPC.


Herpesvirus 4, Human/genetics , Mesoderm/pathology , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/virology , Viral Matrix Proteins/metabolism , Animals , Biomarkers, Tumor , Blotting, Western , Case-Control Studies , Cell Adhesion , Cell Movement , Cell Transformation, Neoplastic , Colony-Forming Units Assay , Epithelial Cells/pathology , Epithelial Cells/virology , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Flow Cytometry , Fluorescent Antibody Technique , Herpesvirus 4, Human/isolation & purification , Humans , Mesoderm/virology , Mice , Mice, Nude , Nasopharyngeal Neoplasms/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Viral Matrix Proteins/genetics
20.
J Clin Invest ; 119(12): 3626-36, 2009 Dec.
Article En | MEDLINE | ID: mdl-19884659

The polycomb group protein B lymphoma Mo-MLV insertion region 1 homolog (Bmi-1) is dysregulated in various cancers, and its upregulation strongly correlates with an invasive phenotype and poor prognosis in patients with nasopharyngeal carcinomas. However, the underlying mechanism of Bmi-1-mediated invasiveness remains unknown. In the current study, we found that upregulation of Bmi-1 induced epithelial-mesenchymal transition (EMT) and enhanced the motility and invasiveness of human nasopharyngeal epithelial cells, whereas silencing endogenous Bmi-1 expression reversed EMT and reduced motility. Furthermore, upregulation of Bmi-1 led to the stabilization of Snail, a transcriptional repressor associated with EMT, via modulation of PI3K/Akt/GSK-3beta signaling. Chromatin immunoprecipitation assays revealed that Bmi-1 transcriptionally downregulated expression of the tumor suppressor PTEN in tumor cells through direct association with the PTEN locus. This in vitro analysis was consistent with the statistical inverse correlation detected between Bmi-1 and PTEN expression in a cohort of human nasopharyngeal carcinoma biopsies. Moreover, ablation of PTEN expression partially rescued the migratory/invasive phenotype of Bmi-1-silenced cells, indicating that PTEN might be a major mediator of Bmi-1-induced EMT. Our results provide functional and mechanistic links between the oncoprotein Bmi-1 and the tumor suppressor PTEN in the development and progression of cancer.


Nasopharynx/cytology , Nasopharynx/metabolism , Nuclear Proteins/metabolism , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Down-Regulation , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Silencing , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Mesoderm/cytology , Mesoderm/metabolism , Mice , Mice, Nude , Nasopharyngeal Neoplasms/etiology , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Transplantation , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/antagonists & inhibitors , Repressor Proteins/genetics , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/metabolism , Transplantation, Heterologous
...