Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biochem Genet ; 62(1): 112-124, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37270714

ABSTRACT

Ovarian cancer (OV) is a highly heterogeneous gynecological tumor that makes the prognostic prediction challenging. Resistance to platinum-based chemotherapy is associated with a poor prognosis in OV. There seems to be an overlap between molecular mechanisms responsible for platinum resistance and immunogenicity in OV. However, the predictive role of platinum resistance-related immune genes for OV prognosis needs to be further explored. In our study, the mRNA expression data of OV patients with corresponding clinical information were collected from The Cancer Genome Atlas (TCGA) cohort and International Cancer Genome Consortium (ICGC) cohort. A multigene signature was constructed for OV patients in the TCGA cohort using the least absolute shrinkage and selection operator (LASSO) Cox regression model according to the optimal value of λ and was validated in the ICGC cohort. Furthermore, we performed functional analysis to explore the immune status between low- and high-risk groups based on the median value of the risk score for the multigene signature. Our data showed that there were 41.1% of the platinum resistance-related genes which differentially expressed between immune score low- and high-OV patients in the TCGA cohort. Univariate Cox regression analysis identified 30 differentially expressed genes (DEGs) associated with overall survival (OS) (P < 0.05). 14 genes were identified to construct a novel platinum resistance-related immune model for classifying OV patients into the low- and high- risk groups. Patients in the low-risk group showed significantly higher OS than those in the high-risk group (P < 0.0001 in the both TCGA and ICGC cohort), which was associated with different immune status for the two risk groups. A novel platinum resistance-related immune model can be used for prognostic prediction in OV. Targeting tumor immunity may be a therapeutic alternative for OV with platinum resistance.


Subject(s)
Ovarian Neoplasms , Humans , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Gene Expression , Risk Factors
2.
Mol Med ; 29(1): 134, 2023 10 06.
Article in English | MEDLINE | ID: mdl-37803271

ABSTRACT

Positive human epidermal growth factor receptor 2 (HER2) expression is associated with an increased risk of metastases especially those to the brain in patients with advanced breast cancer (BC). Neratinib as a tyrosine kinase inhibitor can prevent the transduction of HER1, HER2 and HER4 signaling pathways thus playing an anticancer effect. Moreover, neratinib has a certain efficacy to reverse drug resistance in patients with BC with previous HER2 monoclonal antibody or targeted drug resistance. Neratinib, as monotherapy and in combination with other therapies, has been tested in the neoadjuvant, adjuvant, and metastatic settings. Neratinib with high anticancer activity is indicated for the prolonged adjuvant treatment of HER2-positive early BC, or in combination with other drugs including trastuzumab, capecitabine, and paclitaxel for the treatment of advanced HER2-positive BC especially cancers with central nervous system (CNS) metastasis to reduce the risk of BC recurrence. This article reviewed the pharmacological profiles, efficacy, safety, tolerability, and current clinical trials pertaining to neratinib, with a particular focus on the use of neratinib in patients with metastatic breast cancer (MBC) involving the CNS. We further discussed the use of neratinib for HER2-negative and HER2-mutant breast cancers, and mechanisms of resistance to neratinib. The current evidence suggests that neratinib has promising efficacy in patients with BC which is at least non-inferior compared to previous therapeutic regimens. The most common AE was diarrhea, and the incidence, severity and duration of neratinib-related grade 3 diarrhea can be reduced with loperamide. Of note, neratinib has the potential to effectively control and prevent brain metastasis in patients with advanced BC, providing a therapeutic strategy for HER2-positive BC.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Neoplasm Recurrence, Local , Trastuzumab/adverse effects , Receptor, ErbB-2/metabolism , Diarrhea/chemically induced , Diarrhea/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects
3.
J Transl Med ; 20(1): 365, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35962408

ABSTRACT

BACKGROUND: Efficacy of conventional sequential chemotherapy paradigm for advanced gastric cancer (AGC) patients has largely plateaued. Dynamic molecular changes during and after sequential chemotherapy have not been fully delineated. We aimed to profile the molecular evolutionary process of AGC patients during sequential chemotherapy by next generation sequencing (NGS) of plasma circulating tumor DNA (ctDNA). METHODS: A total of 30 chemo-naïve patients who were diagnosed with unresectable advanced or metastatic stomach adenocarcinoma were enrolled. All patients received sequential chemotherapy regimens following the clinical guideline. One hundred and eight serial peripheral blood samples were collected at baseline, radiographical assessment and disease progression. Plasma ctDNA was isolated and a customized NGS panel was used to detect the genomic features of ctDNA including single nucleotide variants (SNVs) and gene-level copy number variations (CNVs). KEGG pathway enrichment analysis was performed. RESULTS: Platinum-based combination chemotherapy was administrated as first-line regimen. Objective response rate was 50% (15/30). Patients with higher baseline values of copy number instability (CNI), CNVs and variant allel frequency (VAF) were more sensitive to platinum-based first-line regimens. Tumor mutation burden (TMB), CNI and CNV burden at partial response and stable disease were significantly lower than those at baseline, where at progressive disease they recovered to baseline levels. Dynamic change of TMB (ΔTMB) was correlated with progression-free survival of first-line treatment. Fluctuating changes of SNVs and gene-level CNVs could be observed during sequential chemotherapy. Under the pressure of conventional chemotherapy, the number of novel gene-level CNVs were found to be higher than that of novel SNVs. Such novel molecular alterations could be enriched into multiple common oncologic signaling pathways, including EGFR tyrosine kinase inhibitor resistance and platinum drug resistance pathways, where their distributions were found to be highly heterogenous among patients. The impact of subsequent regimens, including paclitaxel-based and irinotecan-based regimens, on the molecular changes driven by first-line therapy was subtle. CONCLUSION: Baseline and dynamic changes of genomic features of ctDNA could be biomarkers for predicting response of platinum-based first-line chemotherapy in AGC patients. After treatment with standard chemotherapy regimens, convergent oncologic pathway enrichment was identified, which is yet characterized by inter-patient heterogenous gene-level CNVs.


Subject(s)
Circulating Tumor DNA , Lung Neoplasms , Stomach Neoplasms , Biomarkers, Tumor/genetics , Circulating Tumor DNA/genetics , DNA Copy Number Variations/genetics , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/pathology , Mutation/genetics , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics
4.
Future Oncol ; 18(23): 2495-2503, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35703115

ABSTRACT

This is an open-label, single-center, multi-cohort phase Ib trial, which consists of three cohorts, including cohort 1 (HER2 negative gastric or gastric esophageal junction adenocarcinoma), cohort 2 (esophageal squamous cell carcinoma and head and neck squamous cell carcinoma) and cohort 3 (hepato-biliary-pancreatic and non-stomach non-esophagi gastrointestinal carcinoma). All eligible patients will be treated by camrelizumab (200 mg, every 2 weeks) and capecitabine (500 mg, twice a day, per os). The primary end point is the safety profiles of camrelizumab plus metronomic capecitabine according to CTCAE v5.0. The secondary end points are progression free survival, overall survival, objective response rate, disease control rate and duration of response. Planned enrollment is 20 subjects for each cohort. Total duration of this trial is expected to be 2 years.


Immune checkpoint inhibitors (ICIs) such as PD-1 inhibitors have been used to treat gastrointestinal cancer patients in clinical practices. Combination with other drugs can improve the efficacy of ICIs. Metronomic chemotherapy using low dose and high frequency of cytotoxic drugs has multi-targeted anti-tumor effects and can be a potential partner of ICIs. In this study, the authors assess the safety and efficacy of a combination of a PD-1 inhibitor (camrelizumab) and an oral chemotherapy drug (capecitabine with metronomic dose) in patients with metastatic treatment-refractory solid tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Neoplasms , Adenocarcinoma/drug therapy , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Capecitabine/therapeutic use , Clinical Trials, Phase I as Topic , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Humans , Neoplasms/drug therapy , Stomach Neoplasms/drug therapy
5.
Mol Ther ; 29(4): 1512-1528, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33388421

ABSTRACT

Cancer-associated lymphatic endothelial cells (LECs) are an active barrier to the effector arm of the anti-tumor immune response; however, it remains unclear how LECs become immunosuppressive in the tumor microenvironment (TME). Exosomal microRNAs (miRNAs) have recently been implicated in intercellular crosstalk within the TME. Here, we report a mechanistic model via which cervical cancer-secreted, exosome-encapsulated microRNA (miR)-1468-5p promotes lymphatic PD-L1 upregulation and lymphangiogenesis to impair T cell immunity. Subsequently, exosomal miR-1468-5p epigenetically activates the JAK2/STAT3 pathway in LECs by directly targeting homeobox containing 1 (HMBOX1) in the SOCS1 promoter, activating an immunosuppressive program that allows cancer cells to escape anti-cancer immunity. Furthermore, clinical data reveal that high serum exosomal miR-1468-5p levels correlate with TME immunosuppressive status and poor prognosis in cervical cancer (CCa) patients. Taken together, our results suggest that cancer-secreted exosomal miR-1468-5p instructs LECs to form an integrated immunosuppressive TME component and may be a prognostic biomarker and therapeutic target for CCa.


Subject(s)
Endothelial Cells/immunology , Homeodomain Proteins/genetics , MicroRNAs/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Uterine Cervical Neoplasms/therapy , B7-H1 Antigen/genetics , Cellular Reprogramming/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Exosomes/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunity/genetics , Immunity/immunology , Immunosuppression Therapy/methods , Janus Kinase 2/genetics , Lymphangiogenesis/genetics , Lymphatic Vessels/immunology , Lymphatic Vessels/pathology , Promoter Regions, Genetic/genetics , STAT3 Transcription Factor/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Escape/genetics , Tumor Escape/immunology , Tumor Microenvironment/immunology , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/immunology
6.
Angiogenesis ; 24(3): 549-565, 2021 08.
Article in English | MEDLINE | ID: mdl-33484377

ABSTRACT

Lymphatic remodelling in the hypoxic tumour microenvironment (TME) is critically involved in the metastasis of cervical squamous cell carcinoma (CSCC); however, its underlying mechanisms remain unclear. Here, we uncovered a novel lymphatic pattern in the hypoxic TME, wherein lymphatic vessels (LVs) are encapsulated by tumour-associated macrophages (TAMs) to form an interconnected network. We describe these aggregates as LVEM (LVs encapsulated by TAMs) considering their advantageous metastatic capacity and active involvement in early lymph node metastasis (LNM). Mechanistic investigations revealed that interleukin-10 (IL-10) derived from hypoxic TAMs adjacent to LVs was a prerequisite for lymphangiogenesis and LVEM formation through its induction of Sp1 upregulation in lymphatic endothelial cells (LECs). Interestingly, Sp1high LECs promoted the transactivation of C-C motif chemokine ligand 1 (CCL1) to facilitate TAM and tumour cell recruitment, thereby forming a positive feedback loop to strengthen the LVEM formation. Knockdown of Sp1 or blockage of CCL1 abrogated LVEM and consequently attenuated LNM. Notably, CSCCnon-LNM is largely devoid of hypoxic TAMs and the resultant LVEM, which might explain its metastatic delay. These findings identify a novel and efficient metastasis-promoting lymphatic pattern in the hypoxic TME, which might provide new targets for anti-metastasis therapy and prognostic assessment.


Subject(s)
Lymphangiogenesis , Lymphatic Vessels/metabolism , Tumor-Associated Macrophages/metabolism , Uterine Cervical Neoplasms/metabolism , Adult , Animals , Cell Hypoxia , Female , Humans , Lymphatic Vessels/pathology , Mice , Neoplasm Metastasis , RAW 264.7 Cells , THP-1 Cells , Tumor-Associated Macrophages/pathology , Uterine Cervical Neoplasms/pathology
7.
Int J Med Sci ; 17(15): 2276-2284, 2020.
Article in English | MEDLINE | ID: mdl-32922192

ABSTRACT

Purpose: The present study focused on the long-term prognostic value of dynamic body mass index (BMI) change in gastric cancer patients who underwent gastrectomy. Methods: Clinical data from a total of 576 gastric cancer patients who underwent radical gastrectomy were collected. Univariate and multivariate analyses were performed to demonstrate the association between dynamic BMI variables (BMI before surgery, 1 month, 6 months or 12 months after surgery) and prognosis (DFS and OS). The correlation between BMI loss after surgery and survival outcomes was also evaluated. Results: Post-operative BMI, especially BMI at one year after surgery (p<0.001), was an independent risk factor of recurrence and mortality, wherein patients with high-BMI (≥23) showed significantly better outcomes than patients with normal-BMI (18.5-23) (DFS, HR:0.49; 95% CI:0.31-0.78; OS, HR:0.30; 95% CI: 0.15-0.59). On the contrary, low-BMI (<18.5) patients presented with worse outcomes (DFS, HR: 1.34; 95% CI: 1.00-1.80; OS, HR: 1.68; 95% CI: 1.20-2.34). In addition, compared with moderate BMI loss (≤10%), severe postoperative BMI loss (>10%) at one year was independently associated with substantially worse prognosis for DFS (HR: 1.54; 95% CI: 1.15-2.08) and OS (HR: 1.45; 95% CI: 1.02-2.06). Subgroup analysis indicated that gender (p=0.03), extent of resection (p<0.001), tumor site (p=0.001) and perineural invasion (p=0.007) were associated with postoperative BMI loss at one year. The prognostic value of postoperative BMI loss at one year was consistent among most clinicopathological subgroups, except for tumor site (interaction p=0.025 for OS). Conclusion: In Chinese gastric cancer patients who underwent gastrectomy, higher postoperative BMI (≥ 23) was significantly associated with longer survival time, whereas severe BMI loss (>10%) at one year after surgery was associated with worse outcomes. Thus, body weight maintenance after treatment is important, and dynamic monitoring of body weight and nutritional status should be emphasized in clinical practice.


Subject(s)
Gastrectomy , Neoplasm Recurrence, Local/epidemiology , Stomach Neoplasms/surgery , Weight Loss , Adult , Aged , Aged, 80 and over , Body Mass Index , China/epidemiology , Disease-Free Survival , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Nutritional Status , Postoperative Period , Preoperative Period , Prognosis , Retrospective Studies , Stomach Neoplasms/mortality , Young Adult
8.
Int J Med Sci ; 17(13): 1946-1955, 2020.
Article in English | MEDLINE | ID: mdl-32788873

ABSTRACT

Gastric cancer is a heterogeneous tumor that underlying molecular mechanisms are largely unclear. This study aimed to elucidate the expression level of HGF-c-MET in gastric cancer patients and to investigate the prognostic and diagnostic value of HGF-c-MET. In silico analysis of the TCGA and GEO database found that HGF and c-MET mRNA expression are significantly higher in gastric cancer tissues than those in peritumor tissues. Both higher mRNA expression of HGF and c-MET were associated with a poorer prognosis. c-MET expression was modulated by methylation in the promoter regions. HGF was positively correlated with CD8+ T cell, CD4+ T cell, macrophage, neutrophil and dendritic cell. Furthermore, functional enrichment analysis and protein-protein interaction networks further shown that HGF-c-MET and related proteins mainly participated in growth factor receptor binding, protein tyrosine kinase activity and signaling receptor binding. Finally, outcome of GSEA analysis showed 13 shared KEGG pathways enriched in high expressed group of HGF and c-MET.


Subject(s)
Cell Proliferation/genetics , Hepatocyte Growth Factor/genetics , Proto-Oncogene Proteins c-met/genetics , Stomach Neoplasms/genetics , Adult , Aged , Cell Movement/genetics , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Phosphorylation , Signal Transduction/genetics , Stomach Neoplasms/epidemiology , Stomach Neoplasms/pathology
9.
Angiogenesis ; 22(3): 397-410, 2019 08.
Article in English | MEDLINE | ID: mdl-30993566

ABSTRACT

AIMS: Recently, cancer-derived exosomes were shown to have pro-metastasis function in cancer, but the mechanism remains unclear. Angiogenesis is essential for tumor progression and is a great promising therapeutic target for advanced cervical cancer. Here, we investigated the role of cervical cancer cell-secreted exosomal miR-221-3p in tumor angiogenesis. METHODS AND RESULTS: miR-221-3p was found to be closely correlated with microvascular density in cervical squamous cell carcinoma (CSCC) by evaluating the microvascular density with immunohistochemistry and miR-221-3p expression with in situ hybridization in clinical specimens. Using the groups of CSCC cell lines (SiHa and C33A) with miR-221-3p overexpression and silencing, the CSCC exosomes were characterized by electron microscopy, western blotting, and fluorescence microscopy. The enrichment of miR-221-3p in CSCC exosomes and its transfer into human umbilical vein endothelial cells (HUVECs) were confirmed by qRT-PCR. CSCC exosomal miR-221-3p promoted angiogenesis in vitro in Matrigel tube formation assay, spheroid sprouting assay, migration assay, and wound healing assay. Then, exosome intratumoral injection indicated that CSCC exosomal miR-221-3p promoted tumor growth in vivo. Thrombospondin-2 (THBS2) was bioinformatically predicted to be a direct target of miR-221-3p, and this was verified by using the in vitro and in vivo experiments described above. Additionally, overexpression of THBS2 in HUVECs rescued the angiogenic function of miR-221-3p. CONCLUSIONS: Our results suggest that CSCC exosomes transport miR-221-3p from cancer cells to vessel endothelial cells and promote angiogenesis by downregulating THBS2. Therefore, CSCC-derived exosomal miR-221-3p could be a possible novel diagnostic biomarker and therapeutic target for CSCC progression.


Subject(s)
Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/genetics , Exosomes/metabolism , MicroRNAs/metabolism , Neovascularization, Pathologic/genetics , Thrombospondins/metabolism , Uterine Cervical Neoplasms/blood supply , Uterine Cervical Neoplasms/genetics , Adult , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Exosomes/ultrastructure , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MicroRNAs/genetics , Microvessels/pathology , Middle Aged , Neovascularization, Pathologic/pathology , RNA Transport
10.
Mol Carcinog ; 58(3): 388-397, 2019 03.
Article in English | MEDLINE | ID: mdl-30362630

ABSTRACT

To explore the mechanisms through which hypoxic tumor microenvironment (TME) modulates the transition of tumor-associated macrophages (TAMs). The migration ability of RAW264.7 macrophages was determined by transwell assay. Flow cytometric, western blot and immunofluorescence analyses of CD206 further validated the M2 polarization of macrophages. Immunofluorescence, western blot and qRT-PCR were performed to detect the expression of neuropilin-1 (Nrp-1) and carbonic anhydrase IX (CAIX). An intermittent hypobaric hypoxia (IH) animal model was established to evaluate the role of hypoxia in activating M2-like TAMs in vivo. We also used immunohistochemistry to analyze the association between CAIX, CD163+ macrophages and Nrp-1 in a series of 72 human cervical cancer specimens. We found that the hypoxic cervical TME educated the recruited macrophages to transform into the M2 phenotype. Nrp-1 expression was significantly increased in hypoxia-primed cervical cancer cells. Blocking Nrp-1 expression prevented hypoxic cells from recruiting and polarizing macrophages towards the M2 phenotype. Hypoxia exposure significantly increased the expression of Nrp-1 as well as the infiltration of macrophages in vivo. Consistently, immunochemical staining in serial tissue sections of cervical cancer revealed upregulated levels of Nrp-1 in CAIX-positive hypoxic regions along with a concurrent significant elevation of M2 macrophages. Nrp-1 and M2-like TAMs were related to the malignant properties of cervical cancer, such as the FIGO stage and lymph node metastasis. Nrp-1 plays critical roles in hypoxic TME-induced activation and pro-tumoral effects of TAMs in cervical cancer. Interfering with Nrp-1 may be a potential therapeutic strategy in treating cervical cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Hypoxia/physiopathology , Macrophages/pathology , Neuropilin-1/metabolism , Tumor Microenvironment , Uterine Cervical Neoplasms/pathology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Carbonic Anhydrase IX/genetics , Carbonic Anhydrase IX/metabolism , Cell Movement , Cell Proliferation , Female , Humans , Lymphatic Metastasis , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Middle Aged , Neuropilin-1/genetics , Prognosis , Tumor Cells, Cultured , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
11.
Int J Med Sci ; 16(8): 1142-1148, 2019.
Article in English | MEDLINE | ID: mdl-31523177

ABSTRACT

Background: The aims of this study were to investigate the expression pattern of CDK12 protein in gastric cancer, and to analyze the correlations of CDK12 expression between CD8+ cell density and CCL12 expression. Methods: Eighty-six paired tumor and non-tumor samples were collected from patients who underwent radical surgery and had pathological confirmed gastric adenocarcinoma. Immunohistochemistry was used to assess CDK12 expression and CD8+ cell density. Expression of CDK12 and CCL21 mRNA was detected by quantitative reverse transcription-polymerase chain reaction. Results: CDK12 expression in gastric tumor tissues was significantly higher than it in paired non-tumor tissues (P<0.001). High expression of CDK12 was identified in 43 cases (50%), and it was significantly correlated with Lauren's classification (diffuse type) and number of metastatic lymph nodes (≥15). High CDK12 protein level indicated a relative poorer overall survival than patients with CKD12 low expression, while it was not identified as an independent prognostic factor. Median number of CD8+ cells in tumor tissues was 51 (range: 0-292). Number of CD8+ cells was positively correlated with CDK12 expression score in tumor tissues (r=0.243, P=0.024). Positive correlation was also found between CDK12 and CCL21 mRNA expression (r=0.419, P=0.017). Conclusion: High CDK12 expression was detected in gastric cancer which was correlated with malignant phenotypes and worse outcome. Positive correlations of CD8+ cell number and CCL21 mRNA expression with CDK12 level were identified.


Subject(s)
Adenocarcinoma/pathology , CD8-Positive T-Lymphocytes/pathology , Chemokine CCL21/metabolism , Cyclin-Dependent Kinases/metabolism , Stomach Neoplasms/pathology , Adenocarcinoma/metabolism , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/metabolism , Cell Count , Chemokine CCL21/genetics , Cyclin-Dependent Kinases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Stomach Neoplasms/metabolism
15.
Am J Nephrol ; 46(5): 371-379, 2017.
Article in English | MEDLINE | ID: mdl-29069649

ABSTRACT

BACKGROUND: Systemic lupus erythematosus (SLE) is characterized by abnormal activated T cells, autoreactive B cells, and massive cytokines. The CD4+ T cells determined B-cells differentiation and cytokines production. The programmed death 1 (PD-1) is the checkpoint immunoinhibitory receptor of activated T cells, and its engagement could exhaust T cells. In this study, we investigated the role of PD-1 systemic engagement with PD-L1-Ig in lupus-like nephritis in SLE mice. METHODS: The murine PD-L1-Ig was injected into SLE-prone mice. The proteinuria and survival ratio were monitored. The production of anti-dsDNA autoantibodies and cytokines in serum were measured by enzyme-linked immunosorbent assay. The cytokine-producing T cells (interferon-γ, IFN-γ and IL-17α) in kidney and spleen were detected with flowcytometry. The pathological evaluation of the Ig deposition in the glomeruliand was determined with immunofluorescence. Lymphocytes in 24-h urine were detected with flowcytometry. RESULTS: The systemic administration of PD-L1-Ig activated PD-1-PD-L1 axis of CD4+ T lymphocytes, suppressed Th17 formation in many organs, including the spleen and the kidney, demolished abnormal production of cytokines (IFN-γ, IL-17, and IL-10) and anti-dsDNA autoantibodies in serum, inhibited immunoglobulin G deposition in the glomeruli with the decrease of proteinuria, and activated T cells in urine. Accordingly, the systemic conjugation of PD-L1-PD-1 impaired renal autoimmune injure and prolonged survival time. CONCLUSION: Our research demonstrated that the protective function of systemic activation of PD-1-PD-L1 axis with PD-L1-Ig attenuates the nephritis in SLE-prone mice, which facilitates us to understand the suppressive function of PD-1-PD-L1 axis in the pathogenesis and progress of the lupus nephritis, and to explore a possible effective therapeutic strategy to SLE.


Subject(s)
Immunoglobulin G/immunology , Kidney/pathology , Lupus Nephritis/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Antibodies, Antinuclear/blood , Antibodies, Antinuclear/immunology , Autoantibodies/blood , Autoantibodies/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cytokines/metabolism , Disease Models, Animal , Female , Flow Cytometry , Humans , Kidney/cytology , Kidney/immunology , Lupus Nephritis/blood , Lupus Nephritis/mortality , Lupus Nephritis/pathology , Lymphocyte Activation , Mice , Mice, Inbred NZB , Programmed Cell Death 1 Receptor/metabolism , Spleen/cytology , Spleen/immunology , Spleen/pathology , Survival Rate
16.
BMC Cancer ; 15: 343, 2015 May 02.
Article in English | MEDLINE | ID: mdl-25929737

ABSTRACT

BACKGROUND: We have preliminarily reported MTA2 expression in gastric cancer and its biological functions by using knockdown cell models, while the molecular mechanisms of MTA2 in regulating malignant behaviors are still unclear. METHODS: MTA2 overexpression models were established by transfection assay in gastric cancer cells BGC-823 and MKN28. Cell proliferation assay, colony formation in soft agar, wound-healing assay and transwell migration assay were performed with MTA2 overexpression and negative control (NC) cells. Subcutaneous xenografts and pulmonary metastasis models by BGC-823/MTA2 and BGC-823/NC cells were used to observe the capacity of growth and metastasis in vivo. Differential gene expression in MTA2 knockdown and overexpression cells was analyzed by microarrays. IL-11, which demonstrated as differential expression in microarray, was detected by real-time PCR, western blot, ELISA and immunohistochemistry staining. Recombinant human IL-11 (rhIL-11) was administrated in cell proliferation and colony formation as rescue assay. RESULTS: The numbers of colonies in soft agar were significantly more in BGC-823/MTA2 and MKN28/MTA2 cells, comparing with those in their NC cells. Capabilities of cell proliferation, wound-healing and cell migration were not significantly changed in MTA2 overexpression cells. The sizes of subcutaneous xenografts and pulmonary metastases of BGC-832/MTA2 cells were significantly larger than those in BGC-823/NC group. Differential expression of IL-11 was identified by genome expression microarray both in MTA2 knockdown and overexpression cells. IL-11 expression was elevated in BGC-823/MTA2 cells, whereas reduced in SGC-7901/shMTA2 cells. Administration of rhIL-11 recovered colony formation capacity of SGC-7901/shMTA2 cells. CONCLUSIONS: MTA2 overexpression enhances colony formation and tumor growth of gastric cancer cells, but not plays important role in cancer cell migration and metastasis. IL-11 is one of the downstream effectors of MTA2 in regulating gastric cancer cells growth.


Subject(s)
Cell Transformation, Neoplastic/genetics , Histone Deacetylases/biosynthesis , Interleukin-11/genetics , Repressor Proteins/biosynthesis , Stomach Neoplasms/genetics , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Histone Deacetylases/genetics , Humans , Interleukin-11/metabolism , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Repressor Proteins/genetics , Stomach Neoplasms/pathology
17.
Cancers (Basel) ; 16(5)2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38473224

ABSTRACT

The concept and policies of multicancer early detection (MCED) have gained significant attention from governments worldwide in recent years. In the era of burgeoning artificial intelligence (AI) technology, the integration of MCED with AI has become a prevailing trend, giving rise to a plethora of MCED AI products. However, due to the heterogeneity of both the detection targets and the AI technologies, the overall diversity of MCED AI products remains considerable. The types of detection targets encompass protein biomarkers, cell-free DNA, or combinations of these biomarkers. In the development of AI models, different model training approaches are employed, including datasets of case-control studies or real-world cancer screening datasets. Various validation techniques, such as cross-validation, location-wise validation, and time-wise validation, are used. All of the factors show significant impacts on the predictive efficacy of MCED AIs. After the completion of AI model development, deploying the MCED AIs in clinical practice presents numerous challenges, including presenting the predictive reports, identifying the potential locations and types of tumors, and addressing cancer-related information, such as clinical follow-up and treatment. This study reviews several mature MCED AI products currently available in the market, detecting their composing factors from serum biomarker detection, MCED AI training/validation, and the clinical application. This review illuminates the challenges encountered by existing MCED AI products across these stages, offering insights into the continued development and obstacles within the field of MCED AI.

18.
Oncogene ; 43(2): 106-122, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37950040

ABSTRACT

VPS35 is a key subunit of the retromer complex responsible for recognising cytosolic retrieval signals in cargo and is involved in neurodegenerative disease and tumour progression. However, the function and molecular mechanism of VPS35 in gastric cancer (GC) remains largely unknown. Here, we demonstrated that VPS35 was significantly upregulated in GC, which was associated with poor survival. VPS35 promoted GC cell proliferation and metastasis both in vitro and in vivo. Mechanistically, VPS35 activated FAK-SRC kinases through integrin-mediated outside-in signalling, leading to the activation of YAP and subsequent IL-6 expression induction in tumour cells. What's more, combined mass spectrometry analysis of MGC-803 cell and bioinformatic analysis, we found that phosphorylation of VPS35 was enhanced in GC cells, and phosphorylated VPS35 has enhanced interaction with ITGB3. VPS35 interacted with ITGB3 and affected the recycling of ITGB3 in GC cells. Gain- and loss-of-function experiments revealed that VPS35 promoted tumour proliferation and metastasis via the IL-6/STAT3 pathway. Interestingly, we also found that STAT3 directly bound to the VPS35 promoter and increased VPS35 transcription, thereby establishing a positive regulatory feedback loop. In addition, we demonstrated that VPS35 knockdown sensitised GC cells to 5-FU and cisplatin. These findings provide evidence that VPS35 promotes tumour proliferation and metastasis, and highlight the potential of targeting VPS35- and IL-6/STAT3-mediated tumour interactions as promising therapeutic strategies for GC.


Subject(s)
Neurodegenerative Diseases , Stomach Neoplasms , Humans , Cell Line, Tumor , Cell Proliferation , Integrins/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Signal Transduction , STAT3 Transcription Factor/metabolism , Stomach Neoplasms/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , YAP-Signaling Proteins , src-Family Kinases
19.
Oncogene ; 43(3): 171-188, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37989866

ABSTRACT

A proportion of gastric cancer (GC) patients suffer from peritoneal metastasis (PM) in the late stage of tumor and these patients have a poor prognosis. To provide more care for GC patient with PM, a deeper exploration of the molecular characteristics of GC-PM is needed. Here we performed the in vitro and in vivo study to illustrate the effect of HOXA11 over-expressed GC cells on peritoneal mesothelial cells (HMrSV5), transcriptomics analyses of HMrSV5 cells co-cultured with HOXA11 over-expressed GC cells, counterparts or alone, cytokine array analyses of serum-free culture medium of HOXA11 over-expressed GC cells, we validated our findings through genetic manipulation of HMrSV5 cells and neutralizing antibodies targeting cytokines secreted by HOXA11 over-expressed GC cells in vitro, as well as utilized human peritoneal metastatic lesions to validate expression of potential targets. We identified that HOXA11 over-expressed GC cells strongly propelled mesothelial fibrosis in vivo and in vitro, and HOXA11 regulated paracrine and autocrine of PDGF BB and TGF ß1 in GC cells to propel mesothelial fibrosis. Meanwhile, HOXA11 over-expressed GC cells drove PDGF BB and TGF ß1 secretion to activate developmental-process related genes in HMrSV5 cells, including Egr1, which processes dependent on miR-181a-5p. Then, Egr1 could mediate peritoneal mesothelial fibrosis. Correspondingly, Egr1 over-expressed HMrSV5 cells supported migration and peritoneal dissemination of GC cells. Together our results suggest that a feedforward amplifier circuity governing GC cells and mesothelial cells in peritoneum contribute to peritoneal metastasis of GC cells.


Subject(s)
MicroRNAs , Peritoneal Neoplasms , Stomach Neoplasms , Humans , Becaplermin/metabolism , Cell Line, Tumor , Epithelium/metabolism , Fibrosis , Homeodomain Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Peritoneal Neoplasms/metabolism , Peritoneum/metabolism , Stomach Neoplasms/pathology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
20.
Mol Cancer ; 12(1): 102, 2013 Sep 08.
Article in English | MEDLINE | ID: mdl-24010737

ABSTRACT

BACKGROUND: MTA2 gene belongs to metastasis associated family, and is highly expressed in some solid tumors, including gastric cancer. Its biological function in gastric cancer is currently undefined. METHODS: Metastasis-associated tumor gene family 2 (MTA2) and transcription factor specificity protein 1 (Sp1) expression were detected in 127 gastric cancer samples by immunohistochemistry staining. SGC-7901 and AGS gastric cancer cell lines transfected by MTA2 shRNA was used for biological function investigation. Binding and regulation activities of Sp1 on MTA2 promoter were investigated by chromatin immunoprecipitation and luciferase reporter gene. RESULTS: The expression rate of MTA2 in gastric cancer tissues was 55.9% (71/127), and its expression was closely related to the depth of tumor invasion, lymph nodes metastasis, and TNM staging. MTA2 knockdown in human SGC-7901 and AGS gastric cancer cells significantly inhibited migration and invasion in vitro, and disrupted structure of cytoskeleton. MTA2 knockdown also attenuated xenografts growth and lung metastasis in nude mice model. MTA2 expression was positively correlated with transcription factor Sp1 in gastric cancer tissues (r = 0.326, P < 0.001). Sp1 bound to human MTA2 gene promoter at region from -1043 bp to -843 bp. Transcriptional activity of MTA2 promoter could be enhanced by Sp1 overexpression. CONCLUSIONS: MTA2 knockdown impairs invasion and metastasis of gastric cancer cells, and attenuates xenografts growth in vivo. Sp1 regulates MTA2 expression at transcriptional level.


Subject(s)
Adenocarcinoma/metabolism , Histone Deacetylases/genetics , Lung Neoplasms/genetics , Repressor Proteins/genetics , Sp1 Transcription Factor/physiology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Actin Cytoskeleton/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/secondary , Animals , Binding Sites , Cell Line, Tumor , Cell Movement , Gene Expression Regulation, Neoplastic , Histone Deacetylases/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Promoter Regions, Genetic , Repressor Proteins/metabolism , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL