Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 14226, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38902401

ABSTRACT

This study aimed to investigate impacts of Omicron infection on cancer patients in China. A retrospective study was conducted, including 347 cancer patients undergoing radiotherapy or chemoradiotherapy between July 2022 and March 2023. Three groups involved: 108 patients without SARS-CoV-2 infection (non-COVID-19 group), 102 patients beginning treatment 10 days after first SARS-CoV-2 infection (≥ 10 days COVID-19 group), and 137 patients beginning treatment less than 10 days after first SARS-CoV-2 infection (< 10 days COVID-19 group). SAA, hsCRP, ALT, etc., were used to assess COVID-19 infection. Serum levels of SAA, hsCRP and IL-6 were all raised in two COVID-19-infected groups (SAA < 0.01, hsCRP < 0.01, IL-6 < 0.05), but PCT, ALT, LDH and HBDH levels were only elevated in ≥ 10 days COVID-19 group (PCT = 0.0478, ALT = 0.0022, LDH = 0.0313, HBDH = 0.0077). Moreover, moderate and severe infected cases were higher in ≥ 10 days COVID-19 group than < 10 days COVID-19 group (12/102 vs 5/137, p = 0.0211), but no significance in myelosuppression and completion rates among three groups. Omicron infection led to inflammation, liver and cardiovascular injury on cancer patients, but delay duration of radiotherapy or chemoradiotherapy after infection did not affect the completion rates and myelosuppression of current therapy. Besides, severity of Omicron infection was even worse among cancer patients who received delayed treatment.


Subject(s)
COVID-19 , Chemoradiotherapy , Neoplasms , SARS-CoV-2 , Humans , COVID-19/therapy , Female , Male , Middle Aged , Neoplasms/radiotherapy , Neoplasms/therapy , Neoplasms/drug therapy , Chemoradiotherapy/adverse effects , Retrospective Studies , Aged , SARS-CoV-2/isolation & purification , Adult , China/epidemiology
2.
Front Oncol ; 12: 986343, 2022.
Article in English | MEDLINE | ID: mdl-36591520

ABSTRACT

Background: Exosomal miRNA had been proved as the promising biomarkers for multiple cancers including epithelial ovarian cancer (EOC). This study aimed to validate the diagnostic accuracy of exosomal miR-320d, miR-4479, and miR-6763-5p for EOC. Materials and methods: Exosomes isolated from the plasma by ultracentrifugation were verified using TEM, qNano and western blot. MiRNAs sequencing was used to screen out the differential exosomal miRNAs and miR-320d, miR-4479, and miR-6763-5p were selected as candidates, which were further verified by RT-qPCR in 168 healthy donors and 161 primary EOC patients. Besides, the diagnostic accuracy of these three exosomal miRNAs were evaluated using the receiver operating characteristic curve (ROC). Results: MiRNAs sequencing revealed 95 differential exosomal miRNAs between EOC patients and healthy donors. Subsequently, exosomal miR-320d, miR-4479, and miR-6763-5p were significantly down regulated in EOC patients compared with healthy controls and benign patients. More importantly, these three miRNAs could serve as circulating diagnostics biomarkers for EOC, possessing areas under the curve (AUC) of 0.6549, 0.7781, and 0.6834, respectively. Moreover, these three exosomal miRNAs levels were closely associated with lymph node metastasis, meanwhile exosomal miR-320d and miR-4479 expression was related to tumor stage. Conclusion: Exosomal miR-320d, miR-4479, and miR-6763-5p might serve as potential biomarkers for EOC.

4.
Exp Ther Med ; 22(1): 723, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34007332

ABSTRACT

Histone lysine demethylation modification is a critical epigenetic modification. Lysine demethylase 2A (KDM2A), a Jumonji C domain-containing demethylase, demethylates the dimethylated H3 lysine 36 (H3K36) residue and exerts little or no activity on monomethylated and trimethylated H3K36 residues. KDM2A expression is regulated by several factors, such as microRNAs, and the phosphorylation of KDM2A also plays a vital role in its function. KDM2A mainly recognizes the unmethylated region of CpG islands and subsequently demethylates histone H3K36 residues. In addition, KDM2A recognizes and binds to phosphorylated proteins, and promotes their ubiquitination and degradation. KDM2A plays an important role in chromosome remodeling and gene transcription, and is involved in cell proliferation and differentiation, cell metabolism, heterochromosomal homeostasis and gene stability. Notably, KDM2A is crucial for tumorigenesis and progression. In the present review, the documented biological functions of KDM2A in physiological and pathological processes are comprehensively summarized.

5.
J Pathol ; 252(4): 358-370, 2020 12.
Article in English | MEDLINE | ID: mdl-32770671

ABSTRACT

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related death worldwide. Lipogenesis has been considered as a critical player in HCC initiation and progression. However, the underlying mechanism is still not fully understood. Here, we identified zinc fingers and homeoboxes 2 (ZHX2), an HCC-associated tumor suppressor, as an important repressor of de novo lipogenesis. Ectopic expression of ZHX2 significantly inhibited de novo lipogenesis in HCC cells and decreased expression of FASN, ACL, ACC1, and SCD1. In accordance with this, ZHX2 was negatively associated with SREBP1c, the master regulator of de novo lipogenesis, in HCC cell lines and human specimens. Results from silencing and overexpression demonstrated that ZHX2 inhibited de novo lipogenesis and consequent HCC progression via repression of SREBP1c. Furthermore, treatment with the SREBP1c inhibitor fatostatin dampened the spontaneous formation of tumors in liver-specific Zhx2 knockout mice. Mechanistically, ZHX2 increased expression of miR-24-3p transcriptionally, which targeted SREBP1c and led to its degradation. In conclusion, our data suggest a novel mechanism through which ZHX2 suppresses HCC progression, which may provide a new strategy for the treatment of HCC. © 2020 The Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Homeodomain Proteins/metabolism , Lipogenesis/genetics , Liver Neoplasms/metabolism , MicroRNAs/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Transcription Factors/metabolism , Adult , Aged , Animals , Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease Models, Animal , Fatty Acids, Nonesterified/metabolism , Female , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Homeodomain Proteins/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Knockout , MicroRNAs/genetics , Middle Aged , Pyridines/pharmacology , Sterol Regulatory Element Binding Protein 1/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 1/genetics , Thiazoles/pharmacology , Transcription Factors/genetics , Triglycerides/metabolism
6.
EBioMedicine ; 53: 102676, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32114388

ABSTRACT

BACKGROUND: Liver cancer stem cells (CSCs) are critical determinants of HCC relapse and therapeutic resistance, but the mechanisms underlying the maintenance of CSCs are poorly understood. We aimed to explore the role of tumor repressor Zinc-fingers and homeoboxes 2 (ZHX2) in liver CSCs. METHODS: CD133+ or EPCAM+ stem-like liver cancer cells were sorted from tumor tissues of HCC patients and HCC cell lines by flow cytometry. In addition, sorafenib-resistant cells, tumor-sphere forming cells and side population (SP) cells were respectively cultured and isolated as hepatic CSCs. The tumor-initiating and chemoresistance properties of ZHX2-overexpressing and ZHX2-knockdown cells were analyzed in vivo and in vitro. Microarray, luciferase reporter assay, chromatin immunoprecipitation (ChIP) and ChIP-on-chip analyses were performed to explore ZHX2 target genes. The expression of ZHX2 and its target gene were determined by quantitative RT-PCR, western blot, immunofluorescence and immunohistochemical staining in hepatoma cells and tumor and adjacent tissues from HCC patients. RESULTS: ZHX2 expression was significantly reduced in liver CSCs from different origins. ZHX2 deficiency led to enhanced liver tumor progression and expansion of CSC populations in vitro and in vivo. Re-expression of ZHX2 restricted capabilities of hepatic CSCs in supporting tumor initiation, self-renewal and sorafenib-resistance. Mechanically, ZHX2 suppressed liver CSCs via inhibiting KDM2A-mediated demethylation of histone H3 lysine 36 (H3K36) at the promoter regions of stemness-associated transcription factors, such as NANOG, SOX4 and OCT4. Moreover, patients with lower expression of ZHX2 and higher expression of KDM2A in tumor tissues showed significantly poorer survival. CONCLUSION: ZHX2 counteracts stem cell traits through transcriptionally repressing KDM2A in HCC. Our data will aid in a better understanding of molecular mechanisms underlying HCC relapse and drug resistance.


Subject(s)
Carcinoma, Hepatocellular/genetics , F-Box Proteins/metabolism , Histone Code , Homeodomain Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Liver Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Transcription Factors/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , F-Box Proteins/genetics , Female , Hep G2 Cells , Homeodomain Proteins/genetics , Humans , Jumonji Domain-Containing Histone Demethylases/genetics , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Methylation , Mice , Mice, Inbred C57BL , Middle Aged , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , SOXC Transcription Factors/genetics , SOXC Transcription Factors/metabolism , Transcription Factors/genetics
7.
J Immunol ; 204(8): 2232-2241, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32179636

ABSTRACT

Sepsis is a life-threatening condition with limited therapeutic options, characterized as excessive systemic inflammation and multiple organ failure. Macrophages play critical roles in sepsis pathogenesis. Metabolism orchestrates homeostasis of macrophages. However, the precise mechanism of macrophage metabolism during sepsis remains poorly elucidated. In this study, we identified the key role of zinc fingers and homeoboxes (Zhx2), a ubiquitous transcription factor, in macrophage glycolysis and sepsis by enhancing 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (Pfkfb3) expression. Mice with myeloid Zhx2-specific deletion (abbreviated as MKO) showed more resistance to cecal ligation and puncture and LPS-induced sepsis, exhibiting as prolonged survival, attenuated pulmonary injury, and reduced level of proinflammatory cytokines, such as TNF-α, IL-6, and IL-1ß. Interestingly, Zhx2 deletion conferred macrophage tolerance to LPS-induced glycolysis, accompanied by reduced proinflammatory cytokines and lactate. Consistently, treatment of glycolytic inhibitor 2-deoxyglucose almost completely abrogated the protection of mice from LPS-induced sepsis initiated by Zhx2 deletion in macrophages. RNA sequencing and chromatin immunoprecipitation assays confirmed that Zhx2 enhanced transcription of Pfkfb3, the glycolysis rate-limiting enzyme, via binding with Pfkfb3 promoter. Furthermore, Pfkfb3 overexpression not only rescued the reduction of macrophage glycolysis caused by Zhx2 deficiency, displaying as extracellular acidification rates and lactate production but also destroyed the resistance of mice to LPS-induced sepsis initiated by transfer of bone marrow-derived macrophages from MKO mice. These findings highlight the novel role of transcription factor Zhx2 in sepsis via regulating Pfkfb3 expression and reprogramming macrophage metabolism, which would shed new insights into the potential strategy to intervene sepsis.


Subject(s)
Glycolysis , Homeodomain Proteins/metabolism , Macrophages/immunology , Phosphofructokinase-2/metabolism , Shock, Septic/immunology , Shock, Septic/metabolism , Animals , Ligation , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Punctures , Shock, Septic/chemically induced
8.
Cancer Biother Radiopharm ; 33(2): 60-64, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29634414

ABSTRACT

OBJECTIVE: Nasopharyngeal carcinoma (NPC) shows the leading morbidity in otorhinolaryngological malignant tumor. It is a common malignancy in China with obvious reginal distribution. NPC is a polygenic disease that is affected by numerous factors. Protein tyrosine phosphatase nonreceptor type 12 (PTPN12) regulates multiple tumor proliferation and development, including breast cancer and colon cancer. However, the role of PTPN12 in NPC occurrence and development has not been elucidated. PATIENTS AND METHODS: NPC cell line CNE2 was cultured in vitro and divided into three groups, including control, empty plasmid, and PTPN12 groups. PTPN12 mRNA and protein expressions were tested by real-time polymerase chain reaction and Western blot. CNE2 cell proliferation was detected by MTT assay. Cell migration was determined by wound healing assay. Cell apoptosis was evaluated by caspase 3 activity detection. Epidermal growth factor receptor (EGFR) expression was assessed by Western blot. RESULTS: PTPN12 plasmid transfection increased PTPN12 mRNA and protein expressions, suppressed cell proliferation and migration, reduced EGFR level, and enhanced caspase 3 activity compared with control and empty plasmid groups (p < 0.05). CONCLUSIONS: PTPN12 regulates NPC proliferation and migration through negative regulating EGFR. It could be treated as a molecular target for NPC diagnosis and prognosis analysis.


Subject(s)
Carcinoma/metabolism , ErbB Receptors/metabolism , Nasopharyngeal Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Carcinoma/genetics , Carcinoma/pathology , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , ErbB Receptors/genetics , Humans , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Protein Tyrosine Phosphatase, Non-Receptor Type 12/biosynthesis , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transfection
9.
Antiviral Res ; 153: 114-123, 2018 05.
Article in English | MEDLINE | ID: mdl-29580980

ABSTRACT

Hepatitis B virus (HBV) covalently closed circular DNA (cccDNA), the stable genomic form as the template for viral transcription, plays a crucial role in viral persistence which remains a major global health problem. While accumulating evidence suggests the involvement of transcription factors and epigenetic machinery in cccDNA transcription, the roles of host transcription factors which contribute to epigenetic modification of cccDNA remain largely unknown. Zinc finger and homeoboxes 2 (ZHX2) is abundantly expressed in adult hepatocytes, where it acts as a transcriptional repressor and tumor suppressor by directly inhibiting the promoter activities of target genes. However, whether ZHX2 influences HBV replication or is involved in cccDNA epigenetic regulation remain unknown. In this study, we investigated the role of ZHX2 in cccDNA transcription. Analysis of immunohistochemistry showed that ZHX2 nuclear expression negatively correlated with serum HBV DNA and HBeAg. Remarkably, ZHX2 significantly decreased HBV antigens expression, pregenomic RNA (pgRNA) and HBV core particle DNA production both in vitro and in mouse livers supporting HBV antigens expression and cccDNA transcription. Dual luciferase and cccDNA ChIP assays confirmed that ZHX2 could bind to cccDNA and transcriptionally inhibit HBV promoter activities. In addition, ZHX2 suppressed the expression of histone regulator genes, such as cccDNA bound p300/CBP, and led to epigenetic repression of cccDNA. These findings highlight the roles of a novel restriction factor, ZHX2, in modulating HBV replication via regulating HBV promoter activities and cccDNA modifications. This study furthers our understanding of HBV transcription from cccDNA and offers new insights on potential HBV therapy.


Subject(s)
Gene Expression Regulation, Viral/drug effects , Hepatitis B virus/growth & development , Hepatitis B virus/immunology , Homeodomain Proteins/metabolism , Host-Pathogen Interactions , Transcription Factors/metabolism , Transcription, Genetic/drug effects , Virus Replication , Animals , Cell Line , Chromatin Immunoprecipitation , Epigenesis, Genetic/drug effects , Genes, Reporter , Hepatitis B e Antigens/blood , Humans , Immunohistochemistry , Liver/pathology , Luciferases/analysis , Mice
10.
J Leukoc Biol ; 102(6): 1313-1322, 2017 12.
Article in English | MEDLINE | ID: mdl-28754800

ABSTRACT

T cell Ig and mucin domain-containing molecule 3 (Tim-3) has been found to play important roles in autoimmune diseases, but whether Tim-3-mediated engulfment of apoptotic cells is involved in systemic lupus erythematosus (SLE) remains to be elucidated. In this study, we verified the role of human Tim-3 (hTim-3) as the receptor of phosphatidylserine (PS) in human embryonic kidney (HEK)293 cells, which initiated the engulfment of apoptotic cells. Both IgV and the mucin domain of Tim-3 were crucial in the phagocytosis of apoptotic cells, and there existed the key cytoplasmic domain for signal transduction. Alanine at 111, locating around the FG-CC' loop of hTim-3, was necessary for its engulfment of apoptotic cells. In accordance, Tim-3 on CD14+ cells negatively correlated with the percentage of peripheral apoptotic cells in control subjects. However, although Tim-3 was significantly increased on CD14+ cells in SLE patients, peripheral apoptotic cells remained much higher than those in control subjects. Tim-3 on CD14+ cells showed positive correlation with percentage of apoptotic cells and level of dsDNA, indicating the involvement of Tim-3 in SLE. Accordingly, soluble Tim-3 (sTim-3) was significantly increased in plasma of SLE patients, which might contribute to higher expression of a disintegrin and metalloproteinase (ADAM)-10. Pretreatment with both plasma from SLE patients and recombinant sTim-3 greatly inhibited hTim-3-initiated phagocytosis of apoptotic cells. Furthermore, anti-Tim-3 antibody depletion of plasma from SLE patients reversed the decreased phagocytosis of apoptotic cells. Collectively, our data suggest that sTim-3 might play inhibitory roles in impaired Tim-3-mediated clearance of apoptotic cells in SLE.


Subject(s)
Apoptosis , Hepatitis A Virus Cellular Receptor 2/metabolism , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/pathology , Phagocytosis , Adult , Annexin A5/metabolism , Case-Control Studies , Cell Count , Cell Line , Female , Hepatitis A Virus Cellular Receptor 2/chemistry , Humans , Lipopolysaccharide Receptors/metabolism , Lupus Erythematosus, Systemic/blood , Male , Monocytes/metabolism , Protein Domains , Recombinant Proteins/pharmacology , Solubility
11.
Article in Chinese | MEDLINE | ID: mdl-26281059

ABSTRACT

OBJECTIVE: To explore the clinical application of gastric pharyngeal anastomosis assisted by laparoscope. METHOD: Apply laparoscope in the gastric pharyngeal anastomosis for 4 cases of advanced hypopharyngeal carcinoma and cervical esophageal carcinoma patients. RESULT: Gastric pharyngeal anastomosis assisted by laparoscope were successfully completed in all 4 patients, all patients avoided thoracotomy or laparotomy, one patient occurred pharyngeal fistula, and died six months later. One patient had cervical lymph node metastasis a year and a half later, without treatment again because of economicissue. The remaining two patients were still alive, one patient had survived 3 years and a half after operation, the other had survived 2 years and a half after operation. CONCLUSION: Gastric pharyngeal anastomosis assisted by laparoscope is feasible. It can reduce the operation wound, improve the safety of operation and patients' life quality.


Subject(s)
Esophageal Neoplasms/surgery , Hypopharyngeal Neoplasms/surgery , Laparoscopy , Pharynx/surgery , Anastomosis, Surgical , Fistula/pathology , Humans , Lymphatic Metastasis , Neck , Pharynx/pathology , Survival Rate
12.
Cancer Sci ; 106(10): 1463-73, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26177443

ABSTRACT

In a previous study, we found that ERGIC3 was a novel lung cancer-related gene by screening libraries of differentially expressed genes. In this study, we developed a new murine monoclonal antibody (mAb) against ERGIC3. This avid antibody (6-C4) is well suited for immunohistochemistry, immunoblotting and solid-phase immunoassays. Furthermore, we systematically investigated expressions of ERGIC3 in a broad variety of normal human tissues and various types of tumors by immunohistochemistry. In normal human tissues, 6-C4 reacted only in some epithelial cells, such as hepatocytes, gastrointestinal epithelium, ducts and acini of the pancreas, proximal and distal tubules of the kidney, and mammary epithelial cells; however, most normal human tissues were not stained. Moreover, almost all carcinomas that originated from the epithelial cells were positive for 6-C4, whereas all sarcomas were negative. Notably, 6-C4 strongly stained non-small cell lung cancer (NSCLC) cells but did not react with normal lung tissues. Hence, ERGIC3 mAb could be used in histopathological diagnosis and cytopathological testing to detect early-stage NSCLC. We also studied the mechanisms of ERGIC3 regulation in vitro and in vivo by means of bioinformatics analysis, luciferase reporter assay, miRNA expression profiling and miRNA transfection. Results showed that miR-203a downregulation induced ERGIC3 overexpression in NSCLC cells.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , Antibodies, Monoclonal/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Down-Regulation/genetics , Gene Expression/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Membrane Proteins/genetics , Membrane Proteins/immunology , MicroRNAs/genetics
13.
Article in Chinese | MEDLINE | ID: mdl-25464581

ABSTRACT

OBJECTIVE: To investigate the feasibility and clinical characteristics of small partial laryngectomy without tracheotomy for T1-2 stage glottic carcinoma. METHOD: Forty-five patients with laryngeal squamaous cell carcinoma in T1-2 stage received small partial laryngectomy without tracheotomy. RESULT: All patients were primarily healed and were hospitalized for an average of 11.5 days post-operatively. In all patients, the function of respiration and the reflection of cough were normal, and laryngeal obstruction did not happen. The only postoperative complication was subcutaneous emphysema noted in 29 patients. Among them, subcutaneous emphysema extincted after 4-6 days in 26 patiens, only 3 patiens suffered from delayed healing because the subcutaneous emphysema extincted after 2 weeks. Mild subcutaneous emphysema did not affect the function of respiration and deglutition, healing of wound, and psychology of patients. All patients had been followed-up for 1-13 years. Only 2 patients died of tumor recurrence or metastasis. The function of respiration and deglutition were normal in the living patients, and no implanting metastasis on surface of trachea were found. CONCLUSION: The theoretical foundation of small partial laryngectomy without tracheotomy for T1-2 stage glottic carcinoma has been well established. This surgical technique is feasible, safe and effective. It can significantly improve clinical outcome of T1-2 stage glottic carcinoma with minimal invasiveness. Furthermore, it can obviously abate the surgical, physiological and psychological trauma on patients.


Subject(s)
Glottis , Laryngeal Neoplasms/surgery , Laryngectomy/methods , Adult , Aged , Aged, 80 and over , Humans , Male , Middle Aged , Tracheotomy
SELECTION OF CITATIONS
SEARCH DETAIL
...