1
|
Ansari AS, Kucharski C, Kc R, Nisakar D, Rahim R, Jiang X, Brandwein J, Uludağ H. Lipopolymer/siRNA complexes engineered for optimal molecular and functional response with chemotherapy in FLT3-mutated acute myeloid leukemia. Acta Biomater 2024; 188:297-314. [PMID: 39236794 DOI: 10.1016/j.actbio.2024.08.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Approximately 25% of newly diagnosed AML patients display an internal tandem duplication (ITD) in the fms-like tyrosine kinase 3 (FLT3) gene. Although both multi-targeted and FLT3 specific tyrosine kinase inhibitors (TKIs) are being utilized for clinical therapy, drug resistance, short remission periods, and high relapse rates are challenges that still need to be tackled. RNA interference (RNAi), mediated by short interfering RNA (siRNA), presents a mechanistically distinct therapeutic platform with the potential of personalization due to its gene sequence-driven mechanism of action. This study explored the use of a non-viral approach for delivery of FLT3 siRNA (siFLT3) in FLT3-ITD positive AML cell lines and primary cells as well as the feasibility of combining this treatment with drugs currently used in the clinic. Treatment of AML cell lines with FLT3 siRNA nanocomplexes resulted in prominent reduction in cell proliferation rates and induction of apoptosis. Quantitative analysis of relative mRNA transcript levels revealed downregulation of the FLT3 gene, which was accompanied by a similar decline in FLT3 protein levels. Moreover, an impact on leukemic stem cells was observed in a small pool of primary AML samples through significantly reduced colony numbers. An absence of a molecular response post-treatment with lipopolymer/siFLT3 complexes in peripheral blood mononuclear cells, obtained from healthy individuals, denoted a passive selectivity of the complexes towards malignant cells. The effect of combining lipopolymer/siFLT3 complexes with daunorubucin and FLT3 targeting TKI gilteritinib led to a significant augmentation of anti-leukemic activity. These findings demonstrate the promising potential of RNAi implemented with lipopolymer complexes for AML molecular therapy. The study prospectively supports the addition of RNAi therapy to current treatment modalities available to target the heterogeneity prevalent in AML. STATEMENT OF SIGNIFICANCE: We show that a clinically validated target, the FLT3 gene, can be eradicated in leukemia cells using non-viral RNAi. We validated these lipopolymers as effective vehicles to deliver nucleic acids to leukemic cells. The potency of the lipopolymers was superior to that of the 'gold-standard' delivery agent, lipid nanoparticles (LNPs), which are not effective in leukemia cells at clinically relevant doses. Mechanistic studies were undertaken to probe structure-function relationships for effective biomaterial formulations. Cellular and molecular responses to siRNA treatment have been characterized in cell models, including leukemia patient-derived cells. The use of the siRNA therapy with clinically used chemotherapy was demonstrated.
Collapse
MESH Headings
- Humans
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- RNA, Small Interfering/pharmacology
- Cell Line, Tumor
- Mutation/genetics
- Apoptosis/drug effects
- Cell Proliferation/drug effects
- Polymers/chemistry
- Polymers/pharmacology
- Aniline Compounds
- Pyrazines
Collapse
Affiliation(s)
- Aysha S Ansari
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Cezary Kucharski
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Remant Kc
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Daniel Nisakar
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Ramea Rahim
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Xiaoyan Jiang
- Terry Fox Laboratory, BC Cancer Research Institute and Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joseph Brandwein
- Division of Hematology, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Hasan Uludağ
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada; Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
2
|
Kermpatsou D, Olsson F, Wåhlén E, Söderberg O, Lennartsson J, Norlin M. Cellular responses to silencing of PDIA3 (protein disulphide-isomerase A3): Effects on proliferation, migration, and genes in control of active vitamin D. J Steroid Biochem Mol Biol 2024; 240:106497. [PMID: 38460707 DOI: 10.1016/j.jsbmb.2024.106497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/15/2024] [Accepted: 03/03/2024] [Indexed: 03/11/2024]
Abstract
The active form of vitamin D, 1,25-dihydroxyvitamin D3, is known to act via VDR (vitamin D receptor), affecting several physiological processes. In addition, PDIA3 (protein disulphide-isomerase A3) has been associated with some of the functions of 1,25-dihydroxyvitamin D3. In the present study we used siRNA-mediated silencing of PDIA3 in osteosarcoma and prostate carcinoma cell lines to examine the role(s) of PDIA3 for 1,25-dihydroxyvitamin D3-dependent responses. PDIA3 silencing affected VDR target genes and significantly altered the 1,25-dihydroxyvitamin D3-dependent induction of CYP24A1, essential for elimination of excess 1,25-dihydroxyvitamin D3. Also, PDIA3 silencing significantly altered migration and proliferation in prostate PC3 cells, independently of 1,25-dihydroxyvitamin D3. 1,25-Dihydroxyvitamin D3 increased thermostability of PDIA3 in cellular thermal shift assay, supporting functional interaction between PDIA3 and 1,25-dihydroxyvitamin D3-dependent pathways. In summary, our data link PDIA3 to 1,25-dihydroxyvitamin D3-mediated signalling, underline and extend its role in proliferation and reveal a novel function in maintenance of 1,25-dihydroxyvitamin D3 levels.
Collapse
Affiliation(s)
- Despoina Kermpatsou
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala Biomedical Centre, Box 591, Uppsala S-751 24, Sweden
| | - Frida Olsson
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala Biomedical Centre, Box 591, Uppsala S-751 24, Sweden
| | - Erik Wåhlén
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala Biomedical Centre, Box 591, Uppsala S-751 24, Sweden
| | - Ola Söderberg
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala Biomedical Centre, Box 591, Uppsala S-751 24, Sweden
| | - Johan Lennartsson
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala Biomedical Centre, Box 591, Uppsala S-751 24, Sweden
| | - Maria Norlin
- Department of Pharmaceutical Biosciences, Science for Life Laboratory, Uppsala University, Uppsala Biomedical Centre, Box 591, Uppsala S-751 24, Sweden.
| |
Collapse
|
3
|
Yang M, Li Q, Yang H, Li Y, Lu L, Wu X, Liu Y, Li W, Shen J, Xiao Z, Zhao Y, Du F, Chen Y, Deng S, Cho CH, Li X, Li M. Downregulation of PDIA3 inhibits gastric cancer cell growth through cell cycle regulation. Biomed Pharmacother 2024; 173:116336. [PMID: 38412717 DOI: 10.1016/j.biopha.2024.116336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 02/16/2024] [Accepted: 02/23/2024] [Indexed: 02/29/2024] Open
Abstract
OBJECTIVE Protein disulfide isomerase A3 (PDIA3) promotes the correct folding of newly synthesized glycoproteins in the endoplasmic reticulum. PDIA3 is overexpressed in most tumors, and it may become a biomarker of cancer prognosis and immunotherapy. Our study aims to detect the expression level of PDIA3 in gastric cancer (GC) and its association with GC development as wells as the underlying mechanisms. METHODS GC cell lines with PDIA3 knockdown by siRNA, CRISPR-cas9 sgRNAs or a pharmacological inhibitor of LOC14 were prepared and used. PDIA3 knockout GC cells were established by CRISPR-cas9-PDIA3 system. The proliferation, migration, invasion and cell cycle of GC cells were analyzed by cell counting kit-8 assay, wound healing assay, transwell assay and flow cytometry, respectively. Immunodeficient nude mice was used to evaluate the role of PDIA3 in tumor formation. Quantitative PCR and western blot were used for examining gene and protein expressions. RNA sequencing was performed to see the altered gene expression. RESULTS The expressions of PDIA3 in GC tissues and cells were increased significantly, and its expression was negatively correlated with the three-year survival rate of GC patients. Down-regulation of PDIA3 by siRNA, LOC14 or CRISPR-cas9 significantly inhibited proliferation, invasion and migration of GC cells TMK1 and AGS, with cell cycle arrested at G2/M phase. Meanwhile, decreased PDIA3 significantly inhibited growth of tumor xenograft in vivo. It was found that cyclin G1 (encoded by CCNG1 gene) expression was decreased by downregulation of PDIA3 in GC cells both in vitro and in vivo. In addition, protein levels of other cell cycle related factors including cyclin D1, CDK2, and CDK6 were also significantly decreased. Further study showed that STAT3 was associated with PDIA3-mediated cyclin G1 regulation. CONCLUSION PDIA3 plays an oncogenic role in GC. Our findings unfolded the functional role of PDIA3 in GC development and highlighted a novel target for cancer therapeutic strategy.
Collapse
Affiliation(s)
- Min Yang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Nanbu people's Hospital, Ministry of Pharmacy, Nanchong, Sichuan, China
| | - Qianxiu Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Huan Yang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Yifan Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Lan Lu
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yubin Liu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China
| | - Wanping Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Fukuan Du
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Shuai Deng
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaobing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China.
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China; Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, Sichuan, China; South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China.
| |
Collapse
|
4
|
Bourebaba L, Kępska M, Qasem B, Zyzak M, Łyczko J, Klemens M, Mularczyk M, Marycz K. Sex hormone-binding globulin improves lipid metabolism and reduces inflammation in subcutaneous adipose tissue of metabolic syndrome-affected horses. Front Mol Biosci 2023; 10:1214961. [PMID: 38146533 PMCID: PMC10749534 DOI: 10.3389/fmolb.2023.1214961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 11/14/2023] [Indexed: 12/27/2023] Open
Abstract
Equine metabolic syndrome (EMS) is a steadily growing endocrine disorder representing a real challenge in veterinary practice. As a multifactorial condition, EMS is characterized by three main metabolic abnormalities including insulin resistance, increased adiposity or obesity and hoof laminitis. Adipose tissue dysfunction is recognized as a core pathophysiological determinant of EMS, as it strongly participates to lipotoxicity and systemic metaflammation, both of which have been closely linked to the development of generalized insulin resistance. Besides, sex hormone binding globulin (SHBG) is an important sex steroids transporters that has been recently proposed as an important metabolic mediator. Therefore, the aim of this study was to verify whether SHBG treatment may ameliorate subcutaneous adipose tissue metabolic failure under EMS condition in terms of lipidome homeostasis, lipid metabolism programs, insulin signalling and local inflammation. Subcutaneous adipose tissue (SAT) biopsies were collected post-mortem from healthy (n = 3) and EMS (n = 3) slaughtered horses. SHBG protein has been applied to SAT samples from EMS horses for 24 h at a final concentration of 50 nM, while control groups (healthy and untreated EMS) were cultured in the presence of SHBG-vehicle only. Tissues from all groups were afterwards secured for downstream analysis of gene expression using RT-qPCR, protein levels by Western blot and ELISA assay and lipidomics through GC-MS technique. Obtained results showcased that SHBG intervention efficiently normalized the altered fatty acids (FAs) profiles by lowering the accumulation of saturated and trans FAs, as well as the pro-inflammatory arachidonic and linoleic acids. Moreover, SHBG showed promising value for the regulation of adipocyte lipolysis and engorgement by lowering the levels of perilipin-1. SHBG exerted moderated effect toward SCD1 and FASN enzymes expression, but increased the LPL abundance. Interestingly, SHBG exhibited a negative regulatory effect on pro-adipogenic stimulators and induced higher expression of KLF3, IRF3 and β-catenin, known as strong adipogenesis repressors. Finally, SHBG protein showed remarkable ability in restoring the insulin signal transduction, IR/IRS/Pi3K/AKT phosphorylation events and GLUT4 transporter abundance, and further attenuate pro-inflammatory response by lowering IL-6 tissue levels and targeting the PDIA3/ERK axis. Overall, the obtained data clearly demonstrate the benefice of SHBG treatment in the regulation of adipose tissue metabolism in the course of EMS and provide new insights for the development of molecular therapies with potential translational application to human metabolic disorders.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Martyna Kępska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Badr Qasem
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Magdalena Zyzak
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Jacek Łyczko
- Department of Food Chemistry and Biocatalysis, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Marta Klemens
- Department of Food Chemistry and Biocatalysis, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | - Malwina Mularczyk
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
- International Institute of Translational Medicine, Wisznia Mała, Poland
| | - Krzysztof Marycz
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wrocław University of Environmental and Life Sciences, Wrocław, Poland
- International Institute of Translational Medicine, Wisznia Mała, Poland
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
5
|
Wang L, Wang X, Zhang J, Duan J, Tang C, Zhang L, Zeng H, Li H, Li Y, Zhou Y. The role of PDIA3 in oral squamous cell carcinoma and its value as A diagnostic and prognostic biomarker. Heliyon 2023; 9:e22596. [PMID: 38213579 PMCID: PMC10782160 DOI: 10.1016/j.heliyon.2023.e22596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 01/13/2024] Open
Abstract
Background This study aimed to investigate the role of protein disulfide isomerase A3 (PDIA3) in oral squamous cell carcinoma (OSCC) and evaluate its significance as a diagnostic and prognostic biomarker. Methods Comprehensive bioinformatics analysis of the OSCC dataset from The Cancer Genome Atlas (TCGA) was performed. PDIA3 was depleted in CAL27 and SCC25 OSCC cells by transfection with PDIA3-specific siRNA oligos. The effects of PDIA3 downregulation on cell viability, apoptosis, and cell migration were evaluated using CCK8, ELISA, and wound healing assays, respectively. Results The mRNA and protein expression of PDIA3 was significantly up-regulated in OSCC tissues compared to adjacent normal tissues. Knockdown of PDIA3 led to significantly decreased cell viability, increased apoptosis, and suppressed migratory ability in OSCC cells. The Kaplan-Meier survival curve showed that patients with higher PDIA3 expression levels had shorter survival than those with low PDIA3 levels. The receiver operating characteristic (ROC) curve indicated that PDIA3 had high sensitivity and accuracy for detecting OSCC (area under the curve (AUC): 0.917, CI: 0.879-0.955). Univariate and multivariate Cox regression analyses identified PDIA3 as an independent prognostic factor of OSCC. Furthermore, the depletion of PDIA3 inhibited AKT activity in OSCC cells. Gene set enrichment analysis (GSEA) indicated that PDIA3 is involved in various important biological functions and signaling pathways closely related to cancer development. Conclusion PDIA3 plays an oncogenic role in OSCC and represents a good candidate as a diagnostic and prognostic biomarker for OSCC.
Collapse
Affiliation(s)
- Lin Wang
- College of Stomatology, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Xinxin Wang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Laboratory Center of Stomatology, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jia Zhang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jiafeng Duan
- Department of Implant Dentistry, Xi'an Nobel Dental Hospital, Xi'an, Shaanxi, 710021, China
| | - Chengfang Tang
- College of Stomatology, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Linmei Zhang
- College of Stomatology, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Hui Zeng
- College of Stomatology, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Hantong Li
- College of Stomatology, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Yuefan Li
- College of Stomatology, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| | - Yan Zhou
- College of Stomatology, Xi'an Medical University, Xi'an, Shaanxi, 710021, China
| |
Collapse
|
6
|
Expression and Prognostic Significance of PDIA3 in Cervical Cancer. Int J Genomics 2022; 2022:4382645. [PMID: 36406049 PMCID: PMC9674421 DOI: 10.1155/2022/4382645] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 08/22/2022] [Accepted: 10/19/2022] [Indexed: 11/13/2022] Open
Abstract
To investigate the expression of protein disulfide isomerase A3 (PDIA3/ERP57) in cervical cancer and its clinical prognostic significance as well as its function and possible action mechanism in the progression of cervical cancer. Based on TIMER2.0 database, the human protein map (Human Protein Atlas) was used to determine the expression level of PDIA3 protein for the analysis of PDIA3 expression in 39 The Cancer Genome Atlas (TCGA) tumors. The PDIA3 expression in cervical cancer tissues in the TCGA and Genotype-Tissue Expression databases was further verified based on the GEPIA2 database to analyze the relationship between the PDIA3 expression and the pathological stage of cervical cancer patients. Immunohistochemistry was used to detect the PDIA3 expression in cervical cancer tissue microarray, including 111 cancer tissue samples and 24 adjacent cancer tissue samples, and the relationship between PDIA3 protein expression and clinical characteristics of patients with cervical cancer was analyzed. The Kaplan–Meier method and log-rank test were used for survival analysis. Based on the cBioPortal database, the Spearman's and Pearson's methods were used to analyze the correlation between PDIA3 expression and DNA methylation. The correlation between PDIA3 expression and the infiltration levels of each immune cell in cervical cancer was evaluated. The STRING was used to construct protein interaction network. Based on LinkedOmics database, the Spearman's method was used to analyze the co-expressed genes of PDIA3 in TCGA cervical cancer. The gene ontology functional enrichment analysis was performed on Top 50 differentially co-expressed genes based on DAVID database. The PDIA3 expression in cervical cancer tissues was significantly higher than that in normal tissues, which (F = 2.74, PR (>F) = 0.0436) was significantly increased with the progression of tumor stage, and PDIA3 showed strong immunoreactivity in cervical cancer tissues. In cervical cancer patients, overall survival (P = 0.014), disease-specific survival (P = 0.013), disease-free interval (P = 0.023), and progression-free interval (P = 0.001) in those with high expression of PDIA3 were significantly lower than those with low expression, suggesting that high expression of PDIA3 was associated with poor prognosis. In cervical cancer, high expression of PDIA3 was associated with DNA methylation and negatively correlated with B cell memory (r = −0.132, P = 0.021), T cell regulatory (r = −0.127, P = 0.026), monocytes (r = −0.204, P = 0), and macrophages M2 (r = −0.142, P = 0.013), whereas positively correlated with levels of NK cell activated (r = 0.162, P = 0.005) and mast cells activated (r = 0.119, P = 0.037). The genes positively correlated with PDIA3 expression included HSPA5 and PPIB, which were mainly enriched in biological processes, such as endoplasmic reticulum (ER) protein folding and ER stress response. PDIA3 can be used as a marker of poor prognosis of cervical cancer. The expression level of PDIA3 is closely related to the survival and prognosis of cervical cancer patients, DNA methylation, and immune cell infiltration.
Collapse
|
7
|
Pan-Cancer Analysis of PDIA3: Identifying It as a Potential Biomarker for Tumor Prognosis and Immunotherapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9614819. [PMID: 36046686 PMCID: PMC9423987 DOI: 10.1155/2022/9614819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022]
Abstract
Protein disulfide isomerase A3 (PDIA3) is a kind of thiol oxidoreductase with a wide range of functions, and its expression is elevated in a variety of tumors, which is closely related to the invasion and metastasis of tumor cells, and has a significant impact on the immunogenicity of tumor cells. Although more and more studies have shown that PDIA3 plays an important role in the occurrence and development of many tumors, there is no systematic pan-cancer study on PDIA3. Therefore, in this study, the differential expression of PDIA3 in 33 kinds of tumors was analyzed to explore its ability to regulate tumor immunity as a biomarker and evaluate its role in different cancer onset stages or clinical prognosis. In this paper, by analyzing the multilevel data including 33 kinds of cancers in the databases of Cancer Genome Atlas (TCGA), UCSC Xena, Cancer Cell Encyclopedia (CCLE), Genotypic Tissue Expression (GTEx), Human Protein Atlas (HPA), cBioPortal, and GDC; the differential expression level of PDIA3 in different types of malignant tumors and its relationship with prognosis and the potential correlation between PDIA3 expression and microsatellite instability (MSI), tumor mutation load (TMB), mismatch repair gene (MMR), DNA methylation level, and immune infiltration level were analyzed with bioinformatics. The results showed that PDIA3 was highly expressed in 19 types of cancers, but downregulated only in THCA. Next, PDIA3 in different tumors was positively or negatively correlated with patient outcome, Kaplan-Meier survival analysis showed that PDIA3 plays an important role in the prognosis of patients with KIRP, KICH, and CESC and may be used as a prognostic biomarker, and the methylation level of PDIA3 promoter region was closely related to patient outcome in eight tumors. The expression level of PDIA3 was correlated with TMB in 13 tumors and MSI in 9 tumors. Among them, the expression level of PDIA3 in THYM has the strongest correlation with TMB, and the expression level of PDIA3 in READ has the strongest correlation with MSI. In addition, the expression of PDIA3 in eight kinds of tumors, including BRCA, HNSC, THYM, LGG, LUAD, LUSC, PRAD, and THCA, had the highest correlation with the infiltration degree of immune cells, and the expression of PDIA3 had the highest correlation with the infiltration degree of 11 kinds of immune cells, including regulatory T cell and macrophages. And LGG is the tumor most likely to be affected by the tumor microenvironment to affect its development and prognosis. To sum up, this study suggests that PDIA3 plays an important role in the occurrence and development of KIRP, KICH, and CESC and in the immunotherapeutic response of THYM, READ, and LGG and can be used as a prognostic biomarker for these tumors.
Collapse
|
8
|
Fang B, Li Q, Wan Z, OuYang Z, Zhang Q. Exploring the Association Between Cervical Microbiota and HR-HPV Infection Based on 16S rRNA Gene and Metagenomic Sequencing. Front Cell Infect Microbiol 2022; 12:922554. [PMID: 35800388 PMCID: PMC9253761 DOI: 10.3389/fcimb.2022.922554] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
The relationship between the cervico-vaginal microbiome and high-risk human papillomavirus (HR-HPV) is well observed. However, there is a lack of adequate research regarding the cervical microbiota in HR-HPV infection. Most published research results have used 16S rRNA gene sequencing technology; this technology only focuses on marker sequences, resulting in incomplete gene information acquisition. Metagenomic sequencing technology can effectively compensate for the deficiency of 16S rRNA gene sequencing, thus improving the analysis of microbiota function. Cervical swab samples from 20 females with HR-HPV infection and 20 uninfected (Control) women were analyzed through 16S rRNA gene and metagenomic sequencing. Our results indicated that the composition and function of the cervical microbiota of HR-HPV infection differed notably from that of control women. Compared with control women, Firmicutes was decreased during HR-HPV infection, whereas Actinobacteria was increased. At the genus level, Lactobacillus was enriched in control women, while levels of Gardnerella and Bifidobacterium were lower. At the species level, Lactobacillus crispatus, L. jensenii, and L. helveticus were enriched in control women; these were the top three species with biomarker significance between the two groups. Eight pathways and four KEGG orthologies of the cervical microbiota of statistical differences were identified between the HR-HPV infection and control women. Collectively, our study described the cervical microbiota and its potential function during HR-HPV infection. Biomarkers of cervical microbiota and the changed bacterial metabolic pathways and metabolites can help clarify the pathogenic mechanism of HR-HPV infection, making them promising targets for clinical treatment and intervention for HR-HPV infection and cervical carcinoma.
Collapse
Affiliation(s)
- Bingyan Fang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Gynecology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Qun Li
- Graduate Collaborative Training Base of Guangdong Second Provincial General Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Zixian Wan
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Gynecology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Zhenbo OuYang
- Department of Gynecology, Guangdong Second Provincial General Hospital, Guangzhou, China
| | - Qiushi Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Gynecology, Guangdong Second Provincial General Hospital, Guangzhou, China
- *Correspondence: Qiushi Zhang,
| |
Collapse
|
9
|
Gu X, Li X, Zhang X, Feng R, Zheng M, Liu L, Yang J, Sun H, Zhang Q, Bian T, Zhao H, Liu Y. MicroRNA-mediated high expression of PDIA3 was correlated with poor prognosis of patients with LUAD. Genomics 2022; 114:110417. [PMID: 35724731 DOI: 10.1016/j.ygeno.2022.110417] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 11/15/2022]
Abstract
Lung cancer, especially lung adenocarcinoma (LUAD) as the most common subtype has threatened the health of people. Even though more and more patients diagnosed as LUAD could be treated efficiently or even cured, a spilt of patients still suffer from disease. Here, on the basis of previous research, we firstly performed the mRNA expression of PDIA3 in pan-cancer, and differential expression between tumor and normal groups was followed. We further analyzed the survival difference and ultimately the expression of PDIA3 in LUAD was selected as our current study. Next, we investigated the mRNA and protein expression of PDIA3 from online databases and performed qRT-PCR and western blotting to verify the outcomes. We still analyzed the correlation between the expression of PDIA3 and clinicopathologic parameters and predicted the potential signal pathways as well as the possible upstream molecular of PDIA3. Considering the correlation of PDIA3 and immune infiltration, related analysis of PDIA3 and immune biomarkers along with PD-1/PD-L1, CTLA-4 were made. We clarified the expression of PDIA3 was upregulated in LUAD and its oncogenic role may be played through tumor infiltration. Thus targeting PDIA3 and immune checkpoint could enhance the efficacy of immunotherapy on patients with LUAD.
Collapse
Affiliation(s)
- Xue Gu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Xiaoli Li
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Xue Zhang
- Department of Oncology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Ran Feng
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Miaosen Zheng
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Lei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Juanjuan Yang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Hui Sun
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Qing Zhang
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Tingting Bian
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Hongyu Zhao
- Department of Radiotherapy, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, Nantong, China; Medical school of Nantong University, Nantong, China.
| |
Collapse
|
10
|
Fabarius A, Samra V, Drews O, Mörz H, Bierbaum M, Darwich A, Weiss C, Brendel S, Kleiner H, Seifarth W, Greffrath W, Hofmann WK, Schmitt CA, Popp HD. Evidence for Recombinant GRP78, CALR, PDIA3 and GPI as Mediators of Genetic Instability in Human CD34+ Cells. Cancers (Basel) 2022; 14:2883. [PMID: 35740549 PMCID: PMC9221337 DOI: 10.3390/cancers14122883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/03/2022] [Accepted: 06/08/2022] [Indexed: 12/10/2022] Open
Abstract
Soluble factors released from irradiated human mesenchymal stromal cells (MSC) may induce genetic instability in human CD34+ cells, potentially mediating hematologic disorders. Recently, we identified four key proteins in the secretome of X-ray-irradiated MSC, among them three endoplasmic reticulum proteins, the 78 kDa glucose-related protein (GRP78), calreticulin (CALR), and protein disulfide-isomerase A3 (PDIA3), as well as the glycolytic enzyme glucose-6-phosphate isomerase (GPI). Here, we demonstrate that exposition of CD34+ cells to recombinant GRP78, CALR, PDIA3 and GPI induces substantial genetic instability. Increased numbers of γH2AX foci (p < 0.0001), centrosome anomalies (p = 0.1000) and aberrant metaphases (p = 0.0022) were detected in CD34+ cells upon incubation with these factors. Specifically, γH2AX foci were found to be induced 4−5-fold in response to any individual of the four factors, and centrosome anomalies by 3−4 fold compared to control medium, which contained none of the recombinant proteins. Aberrant metaphases, not seen in the context of control medium, were detected to a similar extent than centrosome anomalies across the four factors. Notably, the strongest effects were observed when all four factors were collectively provided. In summary, our data suggest that specific components of the secretome from irradiated MSC act as mediators of genetic instability in CD34+ cells, thereby possibly contributing to the pathogenesis of radiation-induced hematologic disorders beyond direct radiation-evoked DNA strand breaks.
Collapse
Affiliation(s)
- Alice Fabarius
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Vanessa Samra
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Oliver Drews
- Biomedical Mass Spectrometry, Center for Medical Research, Johannes Kepler University, 4020 Linz, Austria;
| | - Handan Mörz
- Department of Neurophysiology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (H.M.); (W.G.)
| | - Miriam Bierbaum
- Department of Radiation Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Ali Darwich
- Department of Orthopedics and Trauma Surgery, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Christel Weiss
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany;
| | - Susanne Brendel
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Helga Kleiner
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Wolfgang Seifarth
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Wolfgang Greffrath
- Department of Neurophysiology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (H.M.); (W.G.)
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany; (A.F.); (V.S.); (S.B.); (H.K.); (W.S.); (W.-K.H.)
| | - Clemens A. Schmitt
- Department of Hematology and Oncology, Kepler University Hospital, Johannes Kepler University, 4020 Linz, Austria;
- Medical Department, Division of Hematology, Oncology and Tumor Immunology, Campus Virchow-Klinikum, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine, Helmholtz Association, 13125 Berlin, Germany
| | - Henning D. Popp
- Department of Hematology and Oncology, Kepler University Hospital, Johannes Kepler University, 4020 Linz, Austria;
| |
Collapse
|
11
|
Tu Z, Ouyang Q, Long X, Wu L, Li J, Zhu X, Huang K. Protein Disulfide-Isomerase A3 Is a Robust Prognostic Biomarker for Cancers and Predicts the Immunotherapy Response Effectively. Front Immunol 2022; 13:837512. [PMID: 35401558 PMCID: PMC8989738 DOI: 10.3389/fimmu.2022.837512] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 03/01/2022] [Indexed: 12/26/2022] Open
Abstract
Background Protein disulfide isomerase A3 (PDIA3) is a member of the protein disulfide isomerase (PDI) family that participates in protein folding through its protein disulfide isomerase function. It has been reported to regulate the progression of several cancers, but its function in cancer immunotherapy is unknown. Methods The RNA-seq data of cancer and normal tissues were downloaded from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases. The Cbioportal dataset was used to explore the genomic alteration information of PDIA3 in pan-cancer. Human Protein Atlas (HPA) and ComPPI websites were employed to mine the protein information of PDIA3, and western blot assay was performed to monitor the upregulated PDIA3 expression in clinical GBM samples. The univariate Cox regression and the Kaplan–Meier method were utilized to appraise the prognostic role of PDIA3 in pan-cancer. Gene Set Enrichment Analysis (GSEA) was applied to search the associated cancer hallmarks with PDIA3 expression. TIMER2.0 was the main platform to investigate the immune cell infiltrations related to PDIA3 in pan-cancer. The associations between PDIA3 and immunotherapy biomarkers were performed by Spearman correlation analysis. The immunoblot was used to quantify the PDIA3 expression levels, and the proliferative and invasive ability of glioma cells was determined by colony formation and transwell assays. Findings PDIA3 is overexpressed in most cancer types and exhibits prognosis predictive ability in various cancers, and it is especially expressed in the malignant cells and monocytes/macrophages. In addition, PDIA3 is significantly correlated with immune-activated hallmarks, cancer immune cell infiltrations, and immunoregulators, and the most interesting finding is that PDIA3 could significantly predict anti-PDL1 therapy response. Besides, specific inhibitors that correlated with PDIA3 expression in different cancer types were also screened by using Connectivity Map (CMap). Finally, knockdown of PDIA3 significantly weakened the proliferative and invasive ability of glioma cells. Interpretation The results revealed that PDIA3 acts as a robust tumor biomarker. Its function in protein disulfide linkage regulation could influence protein synthesis, degradation, and secretion, and then shapes the tumor microenvironment, which might be further applied to develop novel anticancer inhibitors.
Collapse
Affiliation(s)
- Zewei Tu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China.,Institute of Neuroscience, Nanchang University, Nanchang, China.,JXHC Key Laboratory of Neurological Medicine, Nanchang, China
| | - Qin Ouyang
- The Second Clinical Medical College of Nanchang University, Nanchang, China
| | - Xiaoyan Long
- East China Institute of Digital Medical Engineering, Shangrao, China
| | - Lei Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China.,Institute of Neuroscience, Nanchang University, Nanchang, China.,JXHC Key Laboratory of Neurological Medicine, Nanchang, China
| | - Jingying Li
- Department of Comprehensive Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China.,Institute of Neuroscience, Nanchang University, Nanchang, China.,JXHC Key Laboratory of Neurological Medicine, Nanchang, China
| | - Kai Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China.,Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, China.,Institute of Neuroscience, Nanchang University, Nanchang, China.,JXHC Key Laboratory of Neurological Medicine, Nanchang, China
| |
Collapse
|
12
|
Samieyan Dehkordi S, Mousavi SH, Ebrahimi M, Alizadeh S, Hedayati Asl AA, Mohammad M, Aliabedi B. Upregulation of hsa-miR-625-5p Inhibits Invasion of Acute Myeloid Leukemia Cancer Cells through ILK/AKT Pathway. CELL JOURNAL 2022; 24:76-84. [PMID: 35279963 PMCID: PMC8918266 DOI: 10.22074/cellj.2022.7658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/08/2020] [Indexed: 11/04/2022]
Abstract
Objective Acute myeloid leukemia (AML) is characterized by abnormalities of differentiation and growth of primary hematopoietic stem cells (HSCs) in the blood and bone marrow. In many studies, miR-625-5p has been shown to inhibit downstream pathways from affecting the metastasis and invasion of the integrin-linked kinase (ILK) signaling pathway. It has been proved that the expression of miR-625-5p decreases in AML cell lines. This study aimed to investigate the effect of miR-625-5p upregulation on the invasion of KG1 ell line in vitro. Materials and Methods In this experimental study, we investigated the impact of upregulation of miR-625-5p on invasion via the ILK/AKT pathway in the KG1 cell line. After transfection using the viral method, the cellular invasion was assessed by invasion assay and the levels of miR-625-5p genes and protein were evaluated by quantitative polymerase chain reaction (qPCR) and western blotting. Moreover, CXCR4 level was assessed by flow cytometry. Results The invasion significantly reduced in MiR-625-5p-transfected KG1 cells (P<0.01) that was concomitant with remarkably decreasing in the expression levels of ILK, NF-κB, and COX2 genes compare with the control group (P<0.01). Incontrast, MMP9, AP1, and AKT significantly increased (P<0.01, P<0.001 and P<0.01, respectively) and GSK3β did not change significantly in MiR-625-5p-transfected KG1 cells. The protein level of NF-κB decreased (P<0.01) and MMP9 increased, however it was not significant. Moreoever, the expression of CXCR4 was significantly lower (P<0.01) in comparison with the control group. Conclusion miR-625-5p leads to a reduction in cell invasion in the AML cell line through ILK pathway. Therefore, it could be a breakthrough in future AML-related research. However, further studies are needed to support this argument.
Collapse
Affiliation(s)
- Sahar Samieyan Dehkordi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Hadi Mousavi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran,P.O.Box: 14177-44361Department of HematologySchool of Allied Medical SciencesTehran University of Medical
SciencesTehranIranP.O.Box: 16635-148Department of Stem Cells and Developmental BiologyRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
Emails:,
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran,Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology,
ACECR, Tehran, Iran,P.O.Box: 14177-44361Department of HematologySchool of Allied Medical SciencesTehran University of Medical
SciencesTehranIranP.O.Box: 16635-148Department of Stem Cells and Developmental BiologyRoyan Institute for Stem Cell Biology and TechnologyACECRTehranIran
Emails:,
| | - Shaban Alizadeh
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Abbas Hedayati Asl
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran,Department of Regenerative Biomedicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology,
ACECR, Tehran, Iran
| | - Monireh Mohammad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and
Technology, ACECR, Tehran, Iran
| | - Bahareh Aliabedi
- Department of Hematology, School of Allied Medical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Jia J, Wang Y, Huang R, Du F, Shen X, Yang Q, Li J. Protein disulfide-isomerase A3 knockdown attenuates oxidized low-density lipoprotein-induced oxidative stress, inflammation and endothelial dysfunction in human umbilical vein endothelial cells by downregulating activating transcription factor 2. Bioengineered 2022; 13:1436-1446. [PMID: 34983301 PMCID: PMC8805980 DOI: 10.1080/21655979.2021.2018980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease implicated in oxidative stress and endothelial dysfunction. Protein disulfide-isomerase A3 (PDIA3) has been reported to regulate oxidative stress and suppress inflammation. This study aimed to explore the function of PDIA3 in atherosclerosis and the underlying mechanisms. PDIA3 expression in oxidized low-density lipoprotein (ox-LDL)-induced human umbilical vein endothelial cells (HUVECs) was detected using RT-qPCR and Western blotting. Following PDIA3 knockdown through transfection with small interfering RNA targeting PDIA3, cell viability, oxidative stress and inflammation in ox-LDL-induced HUVECs was examined using a Cell Counting Kit-8, corresponding kits and ELISA, respectively. The levels of CD31, α-smooth muscle, iNOS, p-eNOS, eNOS and NO were assessed using RT-qPCR, Western blotting and an NO kit to reflect endothelial dysfunction in ox-LDL-induced HUVECs. The relationship between PDIA3 and the activating transcription factor 2 (ATF2) was confirmed using co-immunoprecipitation. In addition, ATF2 expression was examined following PDIA3 silencing. The results indicated that PDIA3 was highly expressed in ox-LDL-induced HUVECs. PDIA3 silencing increased cell viability, and reduced oxidative stress and inflammation, as evidenced by the decreased levels of reactive oxygen species, malondialdehyde, TNF-α, IL-1β and IL-6, and increased superoxide dismutase and glutathione peroxidase activity. In addition, PDIA3 deletion improved endothelial dysfunction. PDIA3 interacted with ATF2, and PDIA3 deletion downregulated ATF2 expression. Furthermore, ATF2 overexpression reversed the effects of PDIA3 knockdown on ox-LDL-induced damage of HUVECs. Collectively, PDIA3 knockdown was found to attenuate ox-LDL-induced oxidative stress, inflammation and endothelial dysfunction in HUVECs by downregulating ATF2 expression, showing promise for the future treatment of atherosclerosis.
Collapse
Affiliation(s)
- Jing Jia
- Department of Anesthetic Surgery, Baotou Steel Hospital, Baotou, China
| | - Yueping Wang
- Department of Cardiology, Baotou Steel Hospital, Baotou, China
| | - Ruijuan Huang
- Laser Treatment Center, Baotou Steel Hospital, Baotou, China
| | - Fengxia Du
- Department of Intensive Medicine, Baotou Steel Hospital, Baotou, China
| | - Xiaozhu Shen
- Department of Geriatrics, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Qiurong Yang
- Nursing Department, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| | - Juan Li
- Nursing Department, The Second People's Hospital of Lianyungang, Lianyungang, Jiangsu, China
| |
Collapse
|
14
|
Li T, Xu J, Liu Y. A Novel Circular RNA CircRFX3 Serves as a Sponge for MicroRNA-587 in Promoting Glioblastoma Progression via Regulating PDIA3. Front Cell Dev Biol 2021; 9:757260. [PMID: 34950658 PMCID: PMC8691731 DOI: 10.3389/fcell.2021.757260] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/20/2021] [Indexed: 02/05/2023] Open
Abstract
An increasing number of studies have indicated that circular RNAs (circRNAs) participate in the progression of numerous tumors. However, the functions of circRNAs in glioblastoma (GBM) remain largely unknown. In this study, we focused on a novel circRNA (hsa_circRFX3_003) that was spliced from RFX3, which we named circRFX3. We confirmed that the expression of circRFX3 was substantially increased in GBM cell lines and clinical GBM tissues. The results of a series of overexpression and knockdown assays indicated that circRFX3 could boost the proliferation, invasion, and migration of GBM cells. By performing dual-luciferase reporter gene and RNA pull-down assays, we verified that circRFX3 could sponge microRNA-587 (miR-587) to exercise its function as a competing endogenous RNA (ceRNA) in the development of GBM. In addition, PDIA3 was proven to be a downstream target of miR-587 and to regulate the Wnt/β-catenin pathway. In conclusion, circRFX3 could act as a cancer-promoting circRNA to boost the development of GBM and regulate the miR-587/PDIA3/β-catenin axis. This study might provide a novel target for the treatment of GBM with molecular therapy.
Collapse
Affiliation(s)
- Tong Li
- The Department of Neurosurgery of West China Hospital of Sichuan University, Chengdu, China
| | - Jianguo Xu
- The Department of Neurosurgery of West China Hospital of Sichuan University, Chengdu, China
| | - Yi Liu
- The Department of Neurosurgery of West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Paglia G, Antonini L, Cervoni L, Ragno R, Sabatino M, Minacori M, Rubini E, Altieri F. A Comparative Analysis of Punicalagin Interaction with PDIA1 and PDIA3 by Biochemical and Computational Approaches. Biomedicines 2021; 9:biomedicines9111533. [PMID: 34829762 PMCID: PMC8614999 DOI: 10.3390/biomedicines9111533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022] Open
Abstract
In a previous work, it was shown that punicalagin, an active ingredient of pomegranate, is able to bind to PDIA3 and inhibit its disulfide reductase activity. Here we provide evidence that punicalagin can also bind to PDIA1, the main expressed form of protein disulfide isomerase (PDI). In this comparative study, the affinity and the effect of punicalagin binding on each protein were evaluated, and a computational approach was used to identify putative binding sites. Punicalagin binds to either PDIA1 or PDIA3 with a similar affinity, but the inhibition efficacy on protein reductase activity is higher for PDIA3. Additionally, punicalagin differently affects the thermal denaturation profile of both proteins. Molecular docking and molecular dynamics simulations led to propose a punicalagin binding mode on PDIA1 and PDIA3, identifying the binding sites at the redox domains a’ in two different pockets, suggesting different effects of punicalagin on proteins’ structure. This study provides insights to develop punicalagin-based ligands, to set up a rational design for PDIA3 selective inhibitors, and to dissect the molecular determinant to modulate the protein activity.
Collapse
Affiliation(s)
- Giuliano Paglia
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (G.P.); (L.C.); (M.M.); (E.R.)
| | - Lorenzo Antonini
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.A.); (R.R.); (M.S.)
| | - Laura Cervoni
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (G.P.); (L.C.); (M.M.); (E.R.)
| | - Rino Ragno
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.A.); (R.R.); (M.S.)
| | - Manuela Sabatino
- Rome Center for Molecular Design, Department of Drug Chemistry and Technology, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (L.A.); (R.R.); (M.S.)
| | - Marco Minacori
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (G.P.); (L.C.); (M.M.); (E.R.)
| | - Elisabetta Rubini
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (G.P.); (L.C.); (M.M.); (E.R.)
- Enrico ed Enrica Sovena Foundation, 00199 Rome, Italy
| | - Fabio Altieri
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (G.P.); (L.C.); (M.M.); (E.R.)
- Correspondence:
| |
Collapse
|
16
|
Diaz Cruz MA, Karlsson S, Szekeres F, Faresjö M, Lund D, Larsson D. Differential expression of protein disulfide-isomerase A3 isoforms, PDIA3 and PDIA3N, in human prostate cancer cell lines representing different stages of prostate cancer. Mol Biol Rep 2021; 48:2429-2436. [PMID: 33761087 PMCID: PMC8060222 DOI: 10.1007/s11033-021-06277-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 03/11/2021] [Indexed: 12/24/2022]
Abstract
Prostate cancer (PCa) is a highly heterogeneous and unpredictable progressive disease. Sensitivity of PCa cells to androgens play a central role in tumor aggressiveness but biomarkers with high sensitivity and specificity that follow the progression of the disease has not yet been verified. The vitamin D endocrine system and its receptors, the Vitamin D Receptor (VDR) and the Protein Disulfide-Isomerase A3 (PDIA3), are related to anti-tumoral effects as well as carcinogenesis and have therefore been suggested as potential candidates for the prevention and therapy of several cancer forms, including PCa. In this study, we evaluated the mRNA expression of VDR and PDIA3 involved in vitamin D signaling in cell lines representing different stages of PCa (PNT2, P4E6, LNCaP, DU145 and PC3). This study further aimed to evaluate vitamin D receptors and their isoforms as potential markers for clinical diagnosis of PCa. A novel transcript isoform of PDIA3 (PDIA3N) was identified and found to be expressed in all PCa cell lines analyzed. Androgen-independent cell lines showed a higher mRNA expression ratio between PDIA3N/PDIA3 contrary to androgen-dependent cell lines that showed a lower mRNA expression ratio between PDIA3N/PDIA3. The structure of PDIA3N differed from PDIA3. PDIA3N was found to be a N-truncated isoform of PDIA3 and differences in protein structure suggests an altered protein function i.e. cell location, thioredoxin activity and affinity for 1,25(OH)2D3. Collectively, PDIA3 transcript isoforms, the ratio between PDIA3N/PDIA3 and especially PDIA3N, are proposed as candidate markers for future studies with different stages of PCa progression.
Collapse
Affiliation(s)
- Maria Araceli Diaz Cruz
- Research School of Health and Welfare, School of Health and Welfare, University of Jönköping, Jönköping, Sweden.
| | - Sandra Karlsson
- Department of Natural Science and Biomedicine, School of Health and Welfare, University of Jönköping, Jönköping, Sweden
| | - Ferenc Szekeres
- Department of Biomedicine, School of Health Sciences, University of Skövde, Skövde, Sweden
| | - Maria Faresjö
- Department of Natural Science and Biomedicine, School of Health and Welfare, University of Jönköping, Jönköping, Sweden
| | - Dan Lund
- Department of Natural Science and Biomedicine, School of Health and Welfare, University of Jönköping, Jönköping, Sweden
| | - Dennis Larsson
- Sahlgrenska University Hospital, Gothia Forum - for Clinical Research, Gothenburg, Sweden
| |
Collapse
|
17
|
Song D, Liu H, Wu J, Gao X, Hao J, Fan D. Insights into the role of ERp57 in cancer. J Cancer 2021; 12:2456-2464. [PMID: 33758622 PMCID: PMC7974888 DOI: 10.7150/jca.48707] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 02/04/2021] [Indexed: 12/28/2022] Open
Abstract
Endoplasmic reticulum resident protein 57 (ERp57) has a molecular weight of 57 kDa, belongs to the protein disulfide-isomerase (PDI) family, and is primarily located in the endoplasmic reticulum (ER). ERp57 functions in the quality control of nascent synthesized glycoproteins, participates in major histocompatibility complex (MHC) class I molecule assembly, regulates immune responses, maintains immunogenic cell death (ICD), regulates the unfolded protein response (UPR), functions as a 1,25-dihydroxy vitamin D3 (1,25(OH)2D3) receptor, regulates the NF-κB and STAT3 pathways, and participates in DNA repair processes and cytoskeletal remodeling. Recent studies have reported ERp57 overexpression in various human cancers, and altered expression and aberrant functionality of ERp57 are associated with cancer growth and progression and changes in the chemosensitivity of cancers. ERp57 may become a potential biomarker and therapeutic target to combat cancer development and chemoresistance. Here, we summarize the available knowledge of the role of ERp57 in cancer and the underlying mechanisms.
Collapse
Affiliation(s)
- Danyang Song
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Hao Liu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an 710032, China
| | - Jian Wu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an 710032, China
| | - Xiaoliang Gao
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an 710032, China
| | - Jianyu Hao
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Daiming Fan
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an 710032, China
| |
Collapse
|
18
|
Holbrook L, Keeton SJ, Sasikumar P, Nock S, Gelzinis J, Brunt E, Ryan S, Pantos MM, Verbetsky CA, Gibbins JM, Kennedy DR. Zafirlukast is a broad-spectrum thiol isomerase inhibitor that inhibits thrombosis without altering bleeding times. Br J Pharmacol 2021; 178:550-563. [PMID: 33080041 PMCID: PMC9328650 DOI: 10.1111/bph.15291] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 09/14/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Multiple members of the thiol isomerase (TI) family of enzymes are present in and released by platelets. Inhibition of these enzymes results in diminished platelet responses, aggregation, adhesion and thrombus formation. Recently, the therapeutic potential of TI inhibition has been recognised and drug-development technologies were used to identify selective small molecule inhibitors. To date, few pan-TI inhibitors have been characterised and the most studied, bacitracin, is known to be nephrotoxic, which prohibits its systemic therapeutic usage. EXPERIMENTAL APPROACH We therefore sought to identify novel broad-spectrum inhibitors of these enzymes and test their effects in vivo. A total of 3,641 compounds were screened for inhibitory effects on the redox activity of ERp5, protein disulphide isomerase (PDI), ERp57, ERp72 and thioredoxin in an insulin turbidity assay. Of the lead compounds identified, zafirlukast was selected for further investigation. KEY RESULTS When applied to platelets, zafirlukast diminished platelet responses in vitro. Zafirlukast was antithrombotic in murine models of thrombosis but did not impair responses in a model of haemostasis. Since TIs are known to modulate adhesion receptor function, we explored the effects of zafirlukast on cell migration. This was inhibited independently of cysteinyl LT receptor expression and was associated with modulation of cell-surface free thiol levels consistent with alterations in redox activity on the cell surface. CONCLUSION AND IMPLICATIONS We identify zafirlukast to be a novel, potent, broad-spectrum TI inhibitor, with wide-ranging effects on platelet function, thrombosis and integrin-mediated cell migration. Zafirlukast is antithrombotic but does not cause bleeding.
Collapse
Affiliation(s)
- Lisa‐Marie Holbrook
- Institute for Cardiovascular and Metabolic Research, School of Biological SciencesUniversity of ReadingReadingUK
- School of Cardiovascular Medicine and SciencesKing's College LondonLondonUK
| | - Shirley J. Keeton
- Institute for Cardiovascular and Metabolic Research, School of Biological SciencesUniversity of ReadingReadingUK
| | - Parvathy Sasikumar
- Institute for Cardiovascular and Metabolic Research, School of Biological SciencesUniversity of ReadingReadingUK
- Centre for HaematologyImperial College LondonLondonUK
| | - Sophie Nock
- Institute for Cardiovascular and Metabolic Research, School of Biological SciencesUniversity of ReadingReadingUK
| | - Justine Gelzinis
- College of Pharmacy and Health SciencesWestern New England UniversitySpringfieldMassachusettsUSA
| | - Elizabeth Brunt
- Institute for Cardiovascular and Metabolic Research, School of Biological SciencesUniversity of ReadingReadingUK
- College of Pharmacy and Health SciencesWestern New England UniversitySpringfieldMassachusettsUSA
| | - Sarah Ryan
- Institute for Cardiovascular and Metabolic Research, School of Biological SciencesUniversity of ReadingReadingUK
| | - Megan M. Pantos
- College of Pharmacy and Health SciencesWestern New England UniversitySpringfieldMassachusettsUSA
| | - Christina A. Verbetsky
- College of Pharmacy and Health SciencesWestern New England UniversitySpringfieldMassachusettsUSA
| | - Jonathan M. Gibbins
- Institute for Cardiovascular and Metabolic Research, School of Biological SciencesUniversity of ReadingReadingUK
| | - Daniel R. Kennedy
- College of Pharmacy and Health SciencesWestern New England UniversitySpringfieldMassachusettsUSA
| |
Collapse
|
19
|
Lam STT, Lim CJ. Cancer Biology of the Endoplasmic Reticulum Lectin Chaperones Calreticulin, Calnexin and PDIA3/ERp57. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021; 59:181-196. [PMID: 34050867 DOI: 10.1007/978-3-030-67696-4_9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The lectin chaperones calreticulin (CALR) and calnexin (CANX), together with their co-chaperone PDIA3, are increasingly implicated in studies of human cancers in roles that extend beyond their primary function as quality control facilitators of protein folding within the endoplasmic reticulum (ER). Led by the discovery that cell surface CALR functions as an immunogen that promotes anti-tumour immunity, studies have now expanded to include their potential uses as prognostic markers for cancers, and in regulation of oncogenic signaling that regulate such diverse processes including integrin-dependent cell adhesion and migration, proliferation, cell death and chemotherapeutic resistance. The diversity stems from the increasing recognition that these proteins have an equally diverse spectrum of subcellular and extracellular localization, and which are aberrantly expressed in tumour cells. This review describes key foundational discoveries and highlight recent findings that further our understanding of the plethora of activities mediated by CALR, CANX and PDIA3.
Collapse
Affiliation(s)
- Shing Tat Theodore Lam
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada.,Michael Cuccione Childhood Cancer Research Program, B.C. Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Chinten James Lim
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada. .,Michael Cuccione Childhood Cancer Research Program, B.C. Children's Hospital Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
20
|
Gisbert-Ferrándiz L, Cosin-Roger J, Hernández C, Macias-Ceja DC, Ortiz-Masiá D, Salvador P, Wildenberg ME, Esplugues JV, Alós R, Navarro F, Calatayud S, Barrachina MD. The vitamin D receptor Taq I polymorphism is associated with reduced VDR and increased PDIA3 protein levels in human intestinal fibroblasts. J Steroid Biochem Mol Biol 2020; 202:105720. [PMID: 32565249 DOI: 10.1016/j.jsbmb.2020.105720] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/12/2020] [Accepted: 06/14/2020] [Indexed: 12/19/2022]
Abstract
The synonymous single nucleotide polymorphism (SNP) rs731236, located in the vitamin D receptor (VDR) gene (Taq I) has been associated with both decreased levels of the protein in peripheral blood mononuclear cells and a fibrosis-related complication in Crohn´s disease (CD). Interactions between VDR and a protein-disulfide isomerase-associated 3 (PDIA3) in the regulation of extracellular matrix have been reported and we aim to analyze the relevance of the VDR genotypes and the effects of Vitamin D (VD) in the expression of VDR, PDIA3 and proliferation of intestinal fibroblasts. Human intestinal fibroblasts were isolated from the non-affected surgical resections of colorectal patients and classified according to the VDR genotype. In some cases, cells were transfected with specific PDIA3 siRNA. Basal and VD-stimulated expression of VDR, PDIA3 and Collagen 1A1 (COL1A1) as well as fibroblast migration/proliferation were analyzed. Our data show that intestinal fibroblasts homozygous for the C allele in the VDR gene exhibited lower VDR protein levels and higher proliferation than cells homozygous for the T allele. VD increased VDR and attenuated the accelerated proliferation of CC fibroblasts. The diminished VDR level detected in CC cells was associated with increased levels of both PDIA3 and COL1A1 expression and the transient silencing of PDIA3 significantly reduced COL1A1 expression. We conclude that intestinal fibroblasts homozygous for the C allele in the VDR gene exhibited: reduced VDR protein levels, increased proliferation and increased PDIA3/COL1A1 expression. Treatment with VD increased VDR and attenuated proliferation of these cells.
Collapse
Affiliation(s)
- Laura Gisbert-Ferrándiz
- Departamento De Farmacología and CIBER, Facultad De Medicina, Universidad De Valencia, Valencia, Spain
| | | | | | | | - Dolores Ortiz-Masiá
- Departamento De Medicina, Facultad De Medicina, Universidad De Valencia, Valencia, Spain
| | - Pedro Salvador
- Departamento De Farmacología and CIBER, Facultad De Medicina, Universidad De Valencia, Valencia, Spain
| | - M E Wildenberg
- Tytgat Institute for Liver and Intestinal Research, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands, Department of Gastroenterology and Hepatology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Juan V Esplugues
- Departamento De Farmacología and CIBER, Facultad De Medicina, Universidad De Valencia, Valencia, Spain; FISABIO, Valencia, Spain
| | | | | | - Sara Calatayud
- Departamento De Farmacología and CIBER, Facultad De Medicina, Universidad De Valencia, Valencia, Spain
| | - María D Barrachina
- Departamento De Farmacología and CIBER, Facultad De Medicina, Universidad De Valencia, Valencia, Spain.
| |
Collapse
|
21
|
Zhou H, Zhang C, Li H, Chen L, Cheng X. A novel risk score system of immune genes associated with prognosis in endometrial cancer. Cancer Cell Int 2020; 20:240. [PMID: 32549787 PMCID: PMC7294624 DOI: 10.1186/s12935-020-01317-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 06/02/2020] [Indexed: 12/24/2022] Open
Abstract
Background Endometrial cancer was the commonest gynecological malignancy in developed countries. Despite striking advances in multimodality management, however, for patients in advanced stage, targeted therapy still remained a challenge. Our study aimed to investigate new biomarkers for endometrial cancer and establish a novel risk score system of immune genes in endometrial cancer. Methods The clinicopathological characteristics and gene expression data were downloaded from The Cancer Genome Atlas (TCGA) database. Differentially expressed genes (DEGs) of immune genes between tumors and normal tissues were identified. Protein–protein interaction (PPI) network of immune genes and transcriptional factors was integrated and visualized in Cytoscape. Univariate and multivariate analysis were employed for key genes to establish a new risk score system. Receiver operating characteristic (ROC) curve and survival analysis were performed to investigate the prognostic value of the model. Association between clinical characteristics and the model was analyzed by logistic regression. For validation, we identified 34 patients with endometrial cancer from Fudan University Shanghai Cancer Center (FUSCC). We detected 14-genes mRNA expression and calculated the risk scores of each patients and we performed survival analysis between the high-risk group and the low-risk group. Results 23 normal tissues and 552 tumor tissues were obtained from TCGA database. 410 immune-related DEGs was identified by difference analysis and correlation analysis. KEGG and GO analysis revealed these DEGs were enriched in cell adhesion, chemotaxis, MAPK pathways and PI3K-Akt signaling pathway, which might regulate tumor progression and migration. All genes were screened for risk model construction and 14 hub immune-related genes (HTR3E, CBLC, TNF, PSMC4, TRAV30, PDIA3, FGF8, PDGFRA, ESRRA, SBDS, CRHR1, LTA, NR2F1, TNFRSF18) were prognostic in endometrial cancer. The area under the curve (AUC) was 0.787 and the high-risk group estimated by the model possessed worse outcome (P < 0.001). Multivariate analysis suggested that the model was indeed an independent prognostic factor (high-risk vs. low-risk, HR = 1.14, P < 0.001). Meanwhile, the high-risk group was prone to have higher grade (P = 0.002) and advanced clinical stage (P = 0.018). In FUSCC validation set, the high-risk group had worse survival than the low-risk group (P < 0.001). Conclusions In conclusion, the novel risk model of immune genes had some merits in predicting the prognosis of endometrial cancer and had strong correlation with clinical outcomes. Furthermore, it might provide new biomarkers for targeted therapy in endometrial cancer.
Collapse
Affiliation(s)
- Hongyu Zhou
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chufan Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Haoran Li
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lihua Chen
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xi Cheng
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai, 200032 China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Wang Z, Zhang H, Cheng Q. PDIA4: The basic characteristics, functions and its potential connection with cancer. Biomed Pharmacother 2019; 122:109688. [PMID: 31794946 DOI: 10.1016/j.biopha.2019.109688] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 11/07/2019] [Accepted: 11/16/2019] [Indexed: 12/19/2022] Open
Abstract
Disulfide bond formation is catalyzed by the protein disulfide Isomerases (PDI) family. This is a critical step in protein folding which occurs within the endoplasmic reticulum. PDIA4, as a member of the PDI family, can cause the adjustment of αIIβ 3 affinities which activate platelet and promote thrombosis formation. Endoplasmic reticulum response is triggered by accumulation of abnormal folding proteins concomitant with increasing PDIA4 expression. Besides, current researches indicate that activated platelets and ERS response affect tumor progression. And PDIA4, as previous reported, also participates in tumor progression by affecting cell apoptosis and DNA repair machinery without specific mechanisms revealed.Therefore, PDI inhibitor might possess great potential value in against tumor progression. In this review, we summarize information on PDIA4 including its the basic characteristics and its implication on tumor.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, PR China; Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, PR China; National Clinical Research Center for Geriatric Disorders, Changsha 410008, PR China.
| |
Collapse
|
23
|
Li G, Zheng P, Wang H, Ai Y, Mao X. Long Non-Coding RNA TUG1 Modulates Proliferation, Migration, And Invasion Of Acute Myeloid Leukemia Cells Via Regulating miR-370-3p/MAPK1/ERK. Onco Targets Ther 2019; 12:10375-10388. [PMID: 31819520 PMCID: PMC6890183 DOI: 10.2147/ott.s217795] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/04/2019] [Indexed: 12/20/2022] Open
Abstract
Background Acute myeloid leukemia (AML) is the most common form of acute leukemia in adults. Long non-coding RNA taurine-upregulated gene 1 (lncRNA TUG1) has been discovered to participate in multiple cancers including AML. However, the detailed mechanism of TUG1 in AML remains obscure. Materials and methods AML cell lines HL-60 and Kasumi-1 were taken as cell models. TUG1 knockdown or overexpression cell lines were generated. Then, the biological influence of TUG1 on cancer cells was studied using CCK-8 assay, transwell assay and Western blot in vitro. Interaction between TUG1 and miR-370-3p was determined by bioinformatics analysis, RT-PCR, and luciferase assay. Western blot, RT-PCR, and luciferase assay were carried out to validate the interaction between miR-370-3p and its target gene Mitogen-Activated Protein Kinase 1 (MAPK1). Results Knockdown of TUG1 markedly reduced viability and metastasis of AML cells, while its overexpression had the opposite effect. MAPK1 was verified as a target gene of miR-370-3p. TUG1 could reduce the level of functional miR-370-3p, facilitate MAPK1 expression, and in turn activate ERK1/2 signaling. Conclusion TUG1 could modulate malignant phenotypes of AML cells via miR-370-3p/MAPK1/ERK signaling. Our study would help to clarify the mechanism of AML tumorigenesis and progression.
Collapse
Affiliation(s)
- Gang Li
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, People's Republic of China
| | - Peiming Zheng
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, People's Republic of China
| | - Huiling Wang
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, People's Republic of China
| | - Yushu Ai
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, People's Republic of China
| | - Xiaohuan Mao
- Department of Clinical Laboratory, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou 450003, Henan, People's Republic of China
| |
Collapse
|