Skip to main content

Main menu

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics

User menu

  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart

Search

  • Advanced search
Anticancer Research
  • Other Publications
    • Anticancer Research
    • In Vivo
    • Cancer Genomics & Proteomics
  • Register
  • Subscribe
  • My alerts
  • Log in
  • My Cart
Anticancer Research

Advanced Search

  • Home
  • Current Issue
  • Archive
  • Info for
    • Authors
    • Editorial Policies
    • Subscribers
    • Advertisers
    • Editorial Board
    • Special Issues 2025
  • Journal Metrics
  • Other Publications
    • In Vivo
    • Cancer Genomics & Proteomics
    • Cancer Diagnosis & Prognosis
  • More
    • IIAR
    • Conferences
    • 2008 Nobel Laureates
  • About Us
    • General Policy
    • Contact
  • Visit us on Facebook
  • Follow us on Linkedin
Research ArticleExperimental Studies
Open Access

PDIA6 and Maspin in Prostate Cancer

TORA K. SMULDERS-SRINIVASAN, SARAH E. JENKINSON, LOUISE J. BROWN, VASILEIOS P. LENIS and ROSEMARY BASS
Anticancer Research December 2023, 43 (12) 5331-5340; DOI: https://doi.org/10.21873/anticanres.16736
TORA K. SMULDERS-SRINIVASAN
1Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, U.K.;
2School of Health & Life Sciences, Teesside University, Middlesbrough, U.K.;
3National Horizons Centre, Teesside University, Darlington, U.K.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • For correspondence: t.smulders@tees.ac.uk
SARAH E. JENKINSON
1Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, U.K.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
LOUISE J. BROWN
1Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, U.K.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
VASILEIOS P. LENIS
2School of Health & Life Sciences, Teesside University, Middlesbrough, U.K.;
3National Horizons Centre, Teesside University, Darlington, U.K.
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
ROSEMARY BASS
1Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, U.K.;
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

Background/Aim: PDIA6 is a disulphide isomerase of the PDI family, known to mediate disulphide bond formation in the endoplasmic reticulum. However, PDI-related proteins also function in other parts of the cell and PDIA6 has been shown to be involved in many types of cancers. We previously identified PDIA6 as a putative Maspin interactor. Maspin has itself been implicated in prostate cancer progression. Our aim was to further explore the roles of Maspin in prostate cancer and establish whether PDIA6 is also involved in prostate cancer. Materials and Methods: RNA levels of PDIA6 and Maspin in prostate cell lines were measured using RT-PCR. Bioinformatics analysis of the TCGA database was used to find RNA levels of PDIA6 and Maspin in prostate cancer. siRNAs were used to knock-down PDIA6, and proliferation and migration assays were conducted on those cells. Results: PDIA6 and Maspin RNA were shown to be expressed at varying levels in prostate cell lines. RNAseq data showed that PDIA6 expression was significantly increased in prostate adenocarcinoma samples, while Maspin RNA expression was decreased. When PDIA6 expression was knocked-down using siRNA in prostate cell lines, proliferation was decreased substantially in the two prostate cancer cell lines (DU145 and PC3) and also decreased in the normal prostate cell line (PNT1a), though less strongly. Conclusion: PDIA6 expression is higher in prostate cancer cells compared to normal prostate cells. Decreasing PDIA6 expression decreases proliferation. Thus, PDIA6 is a promising target for prostate cancer therapeutics.

Key Words:
  • Prostate cancer
  • PDIA6
  • Maspin
  • tumor
  • cancer therapeutics
  • serpin family
  • PDI-P5
  • SERPIN B5
  • ERP5
  • TXNDC7

Disulphide isomerases of the protein disulphide isomerase (PDI) family have been widely studied as molecular chaperones, mediating correct protein folding and disulphide bond formation in the endoplasmic reticulum (ER) (1-4). However, PDI-related proteins also function outside the ER. Recently, PDIA6 (also known as P5, PDI-P5, ERP5, TXNDC7) has been identified as an unconventional RNA-binding protein, with its tumorigenic properties in cutaneous melanoma requiring RNA binding (5). PDIA6 has been shown to be highly expressed in many types of cancer including non-small lung cancer (NSLC or NSCLC; 6, 7), pulmonary squamous cell carcinoma (SCC; 8), nonfibrotic hepatocellular carcinoma (nfHCC; 9), primary ductal breast cancer (10), bladder cancer (11), ovarian cancer (12), leukemia, glioblastoma, breast, colon, ovarian, and uterine cancer cells (13), gastric cancer (14), oral squamous cell carcinoma (OSCC; 15), pancreatic cancer (16), renal cell carcinoma (17), and multiple myeloma (18), but has not been previously identified in prostate cancer.

PDIA6 has been associated with gradually increasing levels during cancer progression. In breast cancer, lymph node metastasis showed higher levels of PDIA6 expression (10). Expression of PDIA6 (and all PDI family members tested) increased during ovarian cancer progression (12). In multiple myeloma, PDIA6 progressively increased in expression at both RNA and protein levels with more severity (18). In NSCLC, PDIA6 expression showed a gradual increase as tumor stage progressed (7). Malignant clinicopathologic features in OSCC were found to be associated with higher PDIA6 expression (15). In pancreatic cancer, higher levels of PDIA6 were associated with poorer patient prognosis, when compared with lower levels of PDIA6 (16).

In addition, PDIA6 expression can be used for predicting cancer prognosis and survival (7, 15, 16). Knocking down PDIA6 expression enhanced cell death and chemosensitivity in cisplatin-resistant cancer cells in NSCLC (6, 7) and gastric cancer (14) as well as chemosensitivity to DDP in pancreatic cancer (16). Knockdown of PDIA6 expression also caused a decrease in cancer cell proliferation (7, 11, 13-17), migration (13, 15, 16), and invasion (11, 15, 16). Knocking down PDIA6 expression reduced clonogenicity, anoikis resistance, and 3D-growth in a number of melanoma cell lines and substantially decreased the ability of melanoma cells to metastasize to the lung (5). Accordingly, when PDIA6 was overexpressed, there was an increase in cancer cell proliferation (7, 14-16, 19), migration (15, 16), and invasion (15, 16). PDIA6 was expressed more highly in a subset of hepatocellular carcinoma that includes portal vein tumor thrombosis (20). PDIA6 appears to be signaling through the Wnt/Beta-catenin pathway (11, 14, 16, 19), as well as the JNK/c-Jun pathway through MAP4K1 (7). Knockdown of PDIA6 expression inhibited tumorigenesis (7, 11, 16) while overexpression of PDIA6 promoted tumorigenesis (15, 16). Thus, inhibition of PDIA6 is a strong candidate for cancer therapy as PDIs appear to be druggable targets (21). Indeed, PDIA6 is one of the proteins inhibited by zafirlukast, a pan-thiol isomerase inhibitor (22). Zafirlukast has been used in a pilot clinical trial for women with relapsed ovarian cancer (23).

We previously identified a putative interaction between PDIA6 and Maspin (24). Maspin is a non-inhibitory member of the serpin family that has been shown to have effects on a range of cell types consistent with anti-metastatic activity (25). It was originally identified as a tumor metastasis suppressor in breast cancer (26), but Maspin has since been implicated in the progression of cancers from other origins, including those of the prostate (27-29). Maspin has been shown to influence a range of cell functions that impact on cell migration and survival; it decreased cell migration, invasion, proliferation, and angiogenesis while it increased cell adhesion and sensitivity to apoptotic stimuli (29). In this study, we have aimed to further explore the roles of Maspin in prostate cancer.

Though PDIA6 has not previously been identified in prostate cancer, other PDI family members have been, including PDIA2, PDIA4, and PDIA3. PDIA2 levels were shown to be a good prognostic marker: higher levels were correlated with poorer prostate cancer survival (30). PDIA4 was overexpressed in prostate cancer cells that were docetaxel resistant (PC-3 and C4-2B), while upregulation of PDIA4 increased the docetaxel resistance in those same cell lines (31). PDIA3 was more highly expressed in more severe prostate cancers (32). PDIA3 was also identified as a potential membrane receptor for Vitamin D or metabolites – Vitamin D3 being a potential treatment for prostate cancer (33, 34). PDIA3 RNA was found in LNCaP, PC3, and PNT-2 prostate cancer cell lines and PDIA3 protein in LNCaP cells (33). In prostate tumor tissues, PDIA3 and LEDGF/p75 proteins were found to be elevated and LEDGF/p75 bound to and transactivated the PDIA3 promoter, regulating PDIA3 expression (35). A new transcript isoform of PDIA3 called PDIA3N was detected in five prostate cancer cell lines and the higher levels of PDIA3N compared to PDIA3 corresponded to more severe cancer (36). Zafirlukast, the pan-thiol isomerase inhibitor mentioned above, inhibits PDIA1, PDIA2, PDIA3, PDIA4, and PDIA6 (22), which might make it very effective for prostate cancer treatment. Indeed, zafirlukast was found to inhibit growth of multiple cancer cell lines, including the PC3 prostate cancer cell line (23). Though PDIA2, PDIA3, and PDIA4 have been identified in the context of prostate cancer, PDIA6 has not. Thus, in this study, we have aimed to find out whether PDIA6 is also involved in prostate cancer, as it has been previously found in other cancers.

Materials and Methods

Cell lines. DU145, PC3 and LNCaP cell lines were obtained from ATCC and cultured in RPMI, supplemented with 10% (v/v) fetal calf serum (FCS) and penicillin/streptomycin. PNT1a were a kind gift from Prof. Edwards (University of East Anglia, UK); they were cultured in the same way as the other prostate derived lines, except that the medium was also supplemented with 10mM HEPES. Cell lines were authenticated by short tandem repeat profiling (DDC, DNA Diagnostics Centre, London, UK). All cell culture reagents were from ThermoFisher Scientific (Paisley, UK).

Reverse transcription (RT)-PCR. Specific oligonucleotide primers and fluorogenic probes (Table I) were designed for Maspin, PDIA6, and 18S by Primer Design Ltd, (Southampton, UK) using probes labeled with a FAM fluorophore reporter (emission 520 nm) at the 5′ end and a TAMRA (quencher emission 583 nm) at the 3′ end to allow measurement by quantitative real-time RT-PCR using a BioRad CFX96.

View this table:
  • View inline
  • View popup
  • Download powerpoint
Table I.

Primers and probes for RT-PCR.

Bioinformatics analyses. Transcriptomics data was obtained by The Cancer Genome Atlas (TCGA) (37), the largest data pool of cancer genetic data that is publicly available, and it was accessed via the Xena UCSC portal (38). RNAseq analysis was performed on 595 prostate tissues comprising 100 normal and 495 prostate adenocarcinoma (primary tumor) samples. The sequencing reads were obtained by the Cancer Genomics Project (37) using the Illumina platform and normalized by RSEM (RNA-Seq by Expectation Maximization) (39) and log transformed (log2). A Welch Two Sample t-test was applied on the two groups (Normal tissue/Primary Tumor) by using R version 3.5.1. The results were presented as box and whiskers plots by using ggplot2 version 3.3.5.

siRNA knock-down. Predesigned siRNAs directed against human PDIA6 and AllStars Negative Control siRNA (referred to as control or ctrl) were obtained from Qiagen (Manchester, UK). siRNAs for PDIA6 NM_005742 were referred to as #1 (Hs_TXNDC7_1), #2 (Hs_TXNDC7_2), #3 (Hs_PDIA6_1) and #4 (Hs_PDIA6_2), with the full sequences of the siRNAs here: #1- Hs_TXNDC7_1 SI00753683 223133 AAGATGAAATTTGCTCTGCTA; 2-Hs_TXNDC7_2 SI00753690 223135 ACGGGATTAGAGGATTTC CTA; 3- Hs_PDIA6_1 SI03097871 223138 CTGGCAGTGA ATGGTCTGTAT; 4- Hs_PDIA6_2 SI03101812 223139 GACGA CAGCTTTGATAAGAAT.

Cells were transfected with 100 nM +100 nM siRNA (of 2 different siRNAs or 200 nM control) using oligofectamine, according to manufacturer’s instructions (Invitrogen). After 4 h, complete medium was added. 72 h after transfection with siRNA, cells were used. Some cells were lysed to perform Western blots. The Western blots were carried out as reported previously (40). Commercially available mouse monoclonal antibodies were used to detect PDIA6 (PDI-P5; PA3-008, Cambridge Bioscience, Cambridge, UK). Secondary antibodies were from Dako (Ely, UK). For proliferation assays, cells were then seeded as below and analyzed on the Xcelligence immediately. For migration assays, cells were then seeded as below and analyzed the next day.

Proliferation assay. The proliferation assay was performed as described previously (41), except that cells were seeded at 10,000 cells/well 3 days after siRNA treatment. To account for differences in batches, control cell index values for the three control triplicates per experiment were averaged and that mean was subtracted from the controls and the experimental wells to present in the graph and for statistical analyses.

Migration assay. The migration assay was performed as described previously (41), except that cells were seeded at 7,500 cells/well (in a 24-well-plate) 3 days after siRNA treatment, and then imaged starting 24 h later. Ten cells were counted per well in 3 wells per condition. To account for differences in batches, control migration values in microns/min for the three control triplicates per experiment were averaged and that mean was subtracted from the control and the experimental wells to present in the graph and for statistical analyses.

Statistical analyses. Data are presented as mean±standard error of the mean (SEM). Significance was judged using Generalized Linear Models in SPSS, unless otherwise stated. A statistically significant difference was defined as p<0.05.

Results

PDIA6 and Maspin were expressed in normal and cancerous prostate cell lines. To begin investigating the possible roles of Maspin and PDIA6 in prostate cancer, their expression was examined in prostate cell lines (Figure 1). LNCaP, PC3, and DU145 were prostate cancer cell lines (42-44), while PNT1a cells were immortalized prostate luminal epithelial cells (45).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

Study design. (a) Experimental design for studies involving prostate cell lines. (b) Experimental design for bioinformatics analysis of prostate RNAseq data.

PDIA6 RNA levels were decreased in LNCaP cells compared to the other cell lines (Figure 2a). Maspin, on the other hand, did not appear to be expressed at all in DU145 cells (Figure 2b), consistent with previous research (46). PNT1a prostate cells had much lower Maspin RNA expression compared to LNCaP and PC3 cells. Thus, both PDIA6 and Maspin were found to be expressed at varying RNA levels in normal and cancerous prostate cell lines (Figure 2a-b).

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

PDIA6 and Maspin are expressed in normal and cancerous prostate cells. (a) RNA expression of PDIA6 in prostate cancer cell lines DU145, PC3, and LNCaP, as well as normal prostate cell line PNT1a as analyzed by RT-PCR. (b) RNA expression of Maspin in prostate cancer cell lines DU145, LNCaP, and PC3, as well as normal prostate cell line PNT1a as analyzed by RT-PCR. (c) RNAseq analysis of PDIA6 RNA levels in normal prostate and primary prostate tumors. n=595 prostate tissues comprising 100 normal and 495 prostate adenocarcinoma (primary tumor) samples. ***p<2.2e-16. (d) RNAseq analysis of Maspin RNA levels in normal prostate and primary prostate tumors. n=595 prostate tissues comprising 100 normal and 495 prostate adenocarcinoma (primary tumor) samples. ***p<1.818e-05.

To further explore the roles of PDIA6 and Maspin in prostate cancer, we analyzed the expression of PDIA6 and Maspin in 100 normal and 495 prostate adenocarcinoma (primary tumor) samples using publicly available transcriptomics data from The Cancer Genome Atlas (TCGA) (37) (Figure 1). PDIA6 expression was significantly and substantially increased in the tumor samples [p<2.2e-16; t(593)=−30.316; 95% CI=−1.675 to −1.470; Figure 2c]. In contrast, the expression of Maspin was slightly, but significantly, reduced in the tumor samples [p<1.818e-05; t(593)=4.436; 95% CI=0.516 to 1.346; Figure 2d]. Thus, Maspin decreased, consistent with its previously identified role as a tumor suppressor in prostate cancer (Figure 2d), in contrast, PDIA6 increased (Figure 2c).

Knock-down of PDIA6 RNA levels decreased prostate cell proliferation. Cancer cells show higher levels of proliferation and PDIA6 showed increased expression in prostate cancer (Figure 2c). Thus, PDIA6 could be involved in causing increased levels of proliferation in cancer. To investigate the role of PDIA6 in proliferation, siRNA treatment was used. Combinations of two siRNAs targeted to the PDIA6 sequence (1+3 or 2+4, see Materials and Methods section; Figure 1) were able to largely decrease protein expression of PDIA6 in DU145, PC3, and PNT1a cells compared to the scrambled siRNA control, though none of the siRNA treatments showed a complete loss of PDIA6 protein (Figure 3a). Proliferation of these PDIA6 knock-down cells was then examined and significant differences were seen based on cell type and specific siRNA treatment [cell type overall effect: χ2(2)=26.97, p<0.001, treatment overall effect: χ2(2)=80.9, p<0.001, Figure 3b]. The different siRNA treatments had different effects on the different cell types [interaction effect: χ2(4)=18.1, p=0.001, Figure 3b]. The strongest decreases in proliferation were seen in DU145 cells, where both sets of siRNA combinations caused substantially and significantly decreased proliferation when compared to the control siRNA-treated cells (control vs. both siRNA treatments: p<0.001; Figure 3b). The other cancerous cell line, PC3, also showed significant decreases in proliferation, though the 2+4 showed a smaller, though still significant, decrease when compared to controls (control vs. 1+3: p<0.001; control vs. 2+4: p=0.044; Figure 3b). Interestingly the PNT1a cells that are derived from normal prostate tissue showed the least decrease in proliferation upon PDIA6 knockdown (Figure 3b). The 2+4 siRNA treatment actually showed no significant decrease in proliferation at all, while the 1+3 siRNA treatment only showed a modest, though significant decrease in proliferation (p=0.001; Figure 3b). Thus, PDIA6 knockdown in prostate cell lines resulted in significant decreases in proliferation, with more substantial decreases in the prostate cancer cell lines than the noncancerous PNT1a prostate cells (Figure 3).

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

Effect of PDIA6 knockdown on the proliferation of prostate cells. (a) Western blots with anti-PDIA6 showing the extent of the siRNA knockdown of PDIA6 protein in the two cancerous prostate cell lines DU145 and PC3 as well as the ‘normal’ prostate cell line PNT1a. Individual siRNAs were not effective, so combinations of 1+3 and 2+4 were used. (b) Proliferation of cells normalized to control siRNA-treated cells for DU145, PC3, and PNT1a prostate cells. n=9 wells for each cell line and siRNA treatment. *p<0.05, **p<0.01, ***p<0.001.

Knock-down of PDIA6 RNA levels had no consistent effect on prostate cell migration. As cell migration is an important characteristic of metastatic cancer cells and Maspin is thought to be involved in preventing cancer metastasis, particularly affecting epithelial-to-mesenchymal transition (47), the effects of PDIA6 knock-down were also assessed on prostate cell migration (Figure 1). The 1+3 and 2+4 siRNA combinations were used again to knock-down PDIA6 RNA leading to decreased PDIA6 protein levels (examples in Figure 3a). In these experiments, prostate cell migration over time was analyzed. Significant differences were seen between treatments and their effects on the different cell types, but no significant differences were seen based on cell type [cell type overall effect: χ2(2)=2.7, n.s., p=0.255, treatment overall effect: χ2(2)=12.2, p=0.002, interaction effect: χ2(4)=17.98, p=0.001; Figure 4]. For DU145 and PNT1a cells, neither PDIA6-targeted siRNA combination had any significant effect on cell migration when compared with controls (Figure 4). For PC3 cells, the knock-down in PDIA6 protein showed significant, but inconsistent effects on PC3 cell migration (Figure 4). Treatment with the PDIA6-targeted 1+3 siRNA showed a small but significant decrease in PC3 cell migration (p=0.004), while treatment with the PDIA6-targeted 2+4 siRNA showed a small but significant increase in PC3 cell migration (p=0.018; Figure 4). Thus, knock-down of PDIA6 protein did not consistently affect the migration capacity of either cancerous or normal prostate cell lines (Figure 4).

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Effect of PDIA6 knockdown on the migration of prostate cells. Migration of 10 cells per well (microns/min) was averaged and normalized to the control siRNA treated cells for each cell line. n=90 cells for DU145 and PC3 and n=60 cells for PNT1a (for each siRNA treatment for each cell line), *p<0.05, **p<0.01.

Discussion

We previously reported an in vitro interaction between Maspin and PDIA6 (24). In the current work, we showed that PDIA6 and Maspin are both expressed at the RNA level in normal prostate and prostate cancer cell lines and tumors (Figure 2). Consistent with previous studies investigating prostate cancer (48), Maspin in our RNAseq analysis showed decreased levels in prostate adenocarcinoma primary tumors (Figure 2d). However, the first-line treatment for prostate cancer is androgen ablation, which is known to increase maspin expression (49), thus patient cancers will likely have an increased level of Maspin. After treatment, in hormone-refractory prostate cancer, higher levels of Maspin allowed the prostate cancer cells PC3 to respond better to the anti-tumor abilities of curcumin (50).

Though PDIA6 has been extensively studied in a number of different cancer types (21, 51), it has never previously been analyzed in prostate cancer. In this current RNAseq analysis, we have found that PDIA6 RNA expression is significantly increased in prostate adenocarcinoma primary tumors (Figure 2c). This is consistent with the PDIA6 expression increase in many different cancer types (6-15). In addition, PDIA2, PDIA3, and PDIA4 all are increased in prostate cancer (30, 31, 33, 35, 36).

We also showed in this current study that siRNA knock-down of PDIA6 causes a decrease in prostate cancer cell proliferation (DU145 and PC3; Figure 3). The same knock-down of PDIA6 in the normal prostate cell line, PNT1a, decreases proliferation, but much less (Figure 3). This is consistent with the literature showing decreases in proliferation when PDIA6 is knocked down in other cancer types (7, 11, 13-17). In NSCLC (7), OSCC (15), renal (17), and pancreatic cancer (16), PDIA6 is shown to increase proliferation by inhibiting apoptosis and in NSCLC, there appears to be an interaction with MAP4K1 causing a decrease in JNK/c-Jun signaling (7). In bladder (11), gastric (14), renal (17), and pancreatic (16) cancers, PDIA6 appears to increase proliferation by promoting the Wnt/β-catenin pathway. In our current study, we did not find any consistent effect of PDIA6 knockdown on migration in prostate cancer or normal prostate cell lines (Figure 4). In other cancer types or cell lines, PDIA6 knockdown has shown a decrease in cell migration (13, 15, 16). Interestingly, when a human malignant glioma cell line (U87MG) was treated with siRNA to PDIA6, migration and invasion actually increased (52). It may be that PDIA6 knockdown has different effects on migration depending on the cellular and cancer context.

Intriguingly, as mentioned above, zafirlukast, an FDA-approved medication for asthma, is a pan-thiol isomerase inhibitor, shown to inhibit PDIA1, 2, 3, 4, and 6 (22). As thiol isomerases are known to be involved in cancer signaling, progression, and metastasis, zafirlukast has been used in a pilot clinical trial for women with relapsed ovarian cancer (23). The outcome marker was only the tumor marker CA-125 and none of the four women showed a decrease in CA-125, but there was a decreased rate of rise of CA-125 and there were few side effects (23). In addition, zafirlukast inhibited the growth of the PC3 prostate cancer cell line (23). Seeing that zafirlukast is a promising treatment for relapsed ovarian cancer, a specific inhibitor of PDIA6 or a more general thiol isomerase inhibitor like zafirlukast are likely good candidates for prostate cancer treatment as well–particularly since we have shown knocking down PDIA6 expression decreases prostate cancer cell line proliferation.

Conclusion

In summary, our findings revealed PDIA6 could be involved in prostate cancer and that Maspin expression was decreased in prostate adenocarcinoma, consistent with its role as a tumor suppressor. We showed that PDIA6 expression was highly increased in prostate adenocarcinoma and drove proliferation in prostate cancer cell lines. PDIA6 has not previously been shown to be involved in prostate cancer, so our research has shown a novel role for PDIA6 in prostate cancer. Our research identifies Maspin and particularly PDIA6 as targets for further investigation in prostate cancer. PDIA6 is more highly expressed in many cancer types and lowering PDIA6 expression decreases cancer proliferation. Since it is a druggable target, PDIA6 is a strong candidate for prostate cancer therapeutics.

Footnotes

  • ↵* Current address: School of Life Sciences, University of Warwick, Coventry, U.K.

  • Authors’ Contributions

    Tora K. Smulders-Srinivasan: Conceptualization, Investigation, Writing - Original Draft, Writing - Review & Editing, Visualization. Sarah E. Jenkinson: Investigation, Writing - Review & Editing. Louise J. Brown: Investigation, Writing - Review & Editing. Vasileios P. Lenis: Conceptualization, Investigation, Writing - Original Draft, Writing - Review & Editing, Visualization. Rosemary Bass: Conceptualization, Investigation, Writing - Original Draft, Writing - Review & Editing, Visualization, Supervision, Project administration, Funding acquisition.

  • Conflicts of Interest

    There are no conflicts of interest for the Authors of this research article.

  • Funding

    This work was supported by The JGW Patterson Foundation. The funders had no roles in in study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the article for publication.

  • Received September 8, 2023.
  • Revision received October 11, 2023.
  • Accepted October 30, 2023.
  • Copyright © 2023 International Institute of Anticancer Research (Dr. George J. Delinasios), All rights reserved.

This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY-NC-ND) 4.0 international license (https://creativecommons.org/licenses/by-nc-nd/4.0).

References

  1. ↵
    1. Fu J,
    2. Gao J,
    3. Liang Z,
    4. Yang D
    : PDI-regulated disulfide bond formation in protein folding and biomolecular assembly. Molecules 26(1): 171, 2020. DOI: 10.3390/molecules26010171
    OpenUrlCrossRefPubMed
    1. Matsusaki M,
    2. Kanemura S,
    3. Kinoshita M,
    4. Lee Y-H,
    5. Inaba K,
    6. Okumura M
    : The Protein Disulfide Isomerase Family: from proteostasis to pathogenesis. Biochim Biophys Acta Gen Subj 1864(2): 129338, 2020. DOI: 10.1016/j.bbagen.2019.04.003
    OpenUrlCrossRef
    1. Appenzeller-Herzog C,
    2. Ellgaard L
    : The human PDI family: Versatility packed into a single fold. Biochim Biophys Acta Mol Cell Res 1783(4): 535-548, 2008. DOI: 10.1016/j.bbamcr.2007.11.010
    OpenUrlCrossRefPubMed
  2. ↵
    1. Kanemura S,
    2. Matsusaki M,
    3. Inaba K,
    4. Okumura M
    : PDI family members as guides for client folding and assembly. Int J Mol Sci 21(24): 9351, 2020. DOI: 10.3390/ijms21249351
    OpenUrlCrossRefPubMed
  3. ↵
    1. Mestre-Farràs N,
    2. Guerrero S,
    3. Bley N,
    4. Rivero E,
    5. Coll O,
    6. Borràs E,
    7. Sabidó E,
    8. Indacochea A,
    9. Casillas-Serra C,
    10. Järvelin AI,
    11. Oliva B,
    12. Castello A,
    13. Hüttelmaier S,
    14. Gebauer F
    : Melanoma RBPome identification reveals PDIA6 as an unconventional RNA-binding protein involved in metastasis. Nucleic Acids Res 50(14): 8207-8225, 2022. DOI: 10.1093/nar/gkac605
    OpenUrlCrossRefPubMed
  4. ↵
    1. Tufo G,
    2. Jones AW,
    3. Wang Z,
    4. Hamelin J,
    5. Tajeddine N,
    6. Esposti DD,
    7. Martel C,
    8. Boursier C,
    9. Gallerne C,
    10. Migdal C,
    11. Lemaire C,
    12. Szabadkai G,
    13. Lemoine A,
    14. Kroemer G,
    15. Brenner C
    : The protein disulfide isomerases PDIA4 and PDIA6 mediate resistance to cisplatin-induced cell death in lung adenocarcinoma. Cell Death Differ 21(5): 685-695, 2014. DOI: 10.1038/cdd.2013.193
    OpenUrlCrossRefPubMed
  5. ↵
    1. Bai Y,
    2. Liu X,
    3. Qi X,
    4. Liu X,
    5. Peng F,
    6. Li H,
    7. Fu H,
    8. Pei S,
    9. Chen L,
    10. Chi X,
    11. Zhang L,
    12. Zhu X,
    13. Song Y,
    14. Wang Y,
    15. Meng S,
    16. Jiang T,
    17. Shao S
    : PDIA6 modulates apoptosis and autophagy of non-small cell lung cancer cells via the MAP4K1/JNK signaling pathway. EBioMedicine 42: 311-325, 2019. DOI: 10.1016/j.ebiom.2019.03.045
    OpenUrlCrossRefPubMed
    1. Lihong H,
    2. Linlin G,
    3. Yiping G,
    4. Yang S,
    5. Xiaoyu Q,
    6. Zhuzhu G,
    7. Xiaohan Y,
    8. Xin Z,
    9. Liyan X,
    10. Shujuan S
    : Proteomics approaches for identification of tumor relevant protein targets in pulmonary squamous cell carcinoma by 2D-DIGE-MS. PLoS One 9(4): e95121, 2014. DOI: 10.1371/journal.pone.0095121
    OpenUrlCrossRefPubMed
    1. Negroni L,
    2. Taouji S,
    3. Arma D,
    4. Pallares-Lupon N,
    5. Leong K,
    6. Beausang LA,
    7. Latterich M,
    8. Bossé R,
    9. Balabaud C,
    10. Schmitter JM,
    11. Bioulac-Sage P,
    12. Zucman-Rossi J,
    13. Rosenbaum J,
    14. Chevet E
    : Integrative quantitative proteomics unveils proteostasis imbalance in human hepatocellular carcinoma developed on nonfibrotic livers. Mol Cell Proteomics 13(12): 3473-3483, 2014. DOI: 10.1074/mcp.M114.043174
    OpenUrlAbstract/FREE Full Text
  6. ↵
    1. Ramos FS,
    2. Serino LT,
    3. Carvalho CM,
    4. Lima RS,
    5. Urban CA,
    6. Cavalli IJ,
    7. Ribeiro EM
    : PDIA3 and PDIA6 gene expression as an aggressiveness marker in primary ductal breast cancer. Genet Mol Res 14(2): 6960-6967, 2015. DOI: 10.4238/2015.June.26.4
    OpenUrlCrossRefPubMed
  7. ↵
    1. Cheng HP,
    2. Liu Q,
    3. Li Y,
    4. Li XD,
    5. Zhu CY
    : The inhibitory effect of PDIA6 downregulation on bladder cancer cell proliferation and invasion. Oncol Res 25(4): 587-593, 2017. DOI: 10.3727/096504016X14761811155298
    OpenUrlCrossRefPubMed
  8. ↵
    1. Samanta S,
    2. Tamura S,
    3. Dubeau L,
    4. Mhawech-Fauceglia P,
    5. Miyagi Y,
    6. Kato H,
    7. Lieberman R,
    8. Buckanovich RJ,
    9. Lin YG,
    10. Neamati N
    : Expression of protein disulfide isomerase family members correlates with tumor progression and patient survival in ovarian cancer. Oncotarget 8(61): 103543-103556, 2017. DOI: 10.18632/oncotarget.21569
    OpenUrlCrossRefPubMed
  9. ↵
    1. Horibe T,
    2. Torisawa A,
    3. Masuda Y,
    4. Kawakami K
    : Functional analysis of protein disulfide isomerase P5 in glioblastoma cells as a novel anticancer target. Oncol Rep 41(2):961-972, 2018. DOI: 10.3892/or.2018.6868
    OpenUrlCrossRefPubMed
  10. ↵
    1. Yan C,
    2. Song X,
    3. Wang S,
    4. Wang J,
    5. Li L
    : Knockdown of PDIA6 inhibits cell proliferation and enhances the chemosensitivity in gastric cancer cells. Cancer Manag Res 12: 11051-11062, 2020. DOI: 10.2147/CMAR.S267711
    OpenUrlCrossRefPubMed
  11. ↵
    1. Mao L,
    2. Wu X,
    3. Gong Z,
    4. Yu M,
    5. Huang Z
    : PDIA6 contributes to aerobic glycolysis and cancer progression in oral squamous cell carcinoma. World J Surg Oncol 19(1): 88, 2021. DOI: 10.1186/s12957-021-02190-w
    OpenUrlCrossRefPubMed
  12. ↵
    1. Ma Y,
    2. Xia P,
    3. Wang Z,
    4. Xu J,
    5. Zhang L,
    6. Jiang Y
    : PDIA6 promotes pancreatic cancer progression and immune escape through CSN5-mediated deubiquitination of β-catenin and PD-L1. Neoplasia 23(9): 912-928, 2021. DOI: 10.1016/j.neo.2021.07.004
    OpenUrlCrossRefPubMed
  13. ↵
    1. Huang Y,
    2. He P,
    3. Ding J
    : Protein disulfide isomerase family 6 promotes the imatinib-resistance of renal cell carcinoma by regulation of Wnt3a-Frizzled1 axis. Bioengineered 12(2): 12157-12166, 2021. DOI: 10.1080/21655979.2021.2005218
    OpenUrlCrossRefPubMed
  14. ↵
    1. Zhong L,
    2. Yang X,
    3. Zhou Y,
    4. Xiao J,
    5. Li H,
    6. Tao J,
    7. Xi Q,
    8. Chu C,
    9. Li C,
    10. Yang X,
    11. Yang C,
    12. Zhang Y,
    13. Shuai P,
    14. Liu Y,
    15. Yu M,
    16. Shi Y,
    17. Hu J,
    18. Zhang W,
    19. Gong B,
    20. Yang Z
    : Exploring the R-ISS stage-specific regular networks in the progression of multiple myeloma at single-cell resolution. Sci China Life Sci 65(9): 1811-1823, 2022. DOI: 10.1007/s11427-021-2097-1
    OpenUrlCrossRefPubMed
  15. ↵
    1. Gao H,
    2. Sun B,
    3. Fu H,
    4. Chi X,
    5. Wang F,
    6. Qi X,
    7. Hu J,
    8. Shao S
    : PDIA6 promotes the proliferation of HeLa cells through activating the Wnt/β-catenin signaling pathway. Oncotarget 7(33): 53289-53298, 2016. DOI: 10.18632/oncotarget.10795
    OpenUrlCrossRefPubMed
  16. ↵
    1. Xu X,
    2. Wei X,
    3. Ling Q,
    4. Cheng J,
    5. Zhou B,
    6. Xie H,
    7. Zhou L,
    8. Zheng S
    : Identification of two portal vein tumor thrombosis associated proteins in hepatocellular carcinoma: Protein disulfide-isomerase A6 and apolipoprotein A-I. J Gastroenterol Hepatol 26(12): 1787-1794, 2011. DOI: 10.1111/j.1440-1746.2011.06796.x
    OpenUrlCrossRefPubMed
  17. ↵
    1. Powell LE,
    2. Foster PA
    : Protein disulphide isomerase inhibition as a potential cancer therapeutic strategy. Cancer Med 10(8): 2812-2825, 2021. DOI: 10.1002/cam4.3836
    OpenUrlCrossRefPubMed
  18. ↵
    1. Holbrook LM,
    2. Keeton SJ,
    3. Sasikumar P,
    4. Nock S,
    5. Gelzinis J,
    6. Brunt E,
    7. Ryan S,
    8. Pantos MM,
    9. Verbetsky CA,
    10. Gibbins JM,
    11. Kennedy DR
    : Zafirlukast is a broad-spectrum thiol isomerase inhibitor that inhibits thrombosis without altering bleeding times. Br J Pharmacol 178(3): 550-563, 2021. DOI: 10.1111/bph.15291
    OpenUrlCrossRefPubMed
  19. ↵
    1. Gelzinis JA,
    2. Szahaj MK,
    3. Bekendam RH,
    4. Wurl SE,
    5. Pantos MM,
    6. Verbetsky CA,
    7. Dufresne A,
    8. Shea M,
    9. Howard KC,
    10. Tsodikov OV,
    11. Garneau-Tsodikova S,
    12. Zwicker JI,
    13. Kennedy DR
    : Targeting thiol isomerase activity with zafirlukast to treat ovarian cancer from the bench to clinic. FASEB J 37(5): e22914, 2023. DOI: 10.1096/fj.202201952R
    OpenUrlCrossRefPubMed
  20. ↵
    1. Al-Mamun M,
    2. Ravenhill L,
    3. Srisukkham W,
    4. Hossain A,
    5. Fall C,
    6. Ellis V,
    7. Bass R
    : Effects of noninhibitory serpin maspin on the actin cytoskeleton: a quantitative image modeling approach. Microsc Microanal 22(2): 394-409, 2016. DOI: 10.1017/S1431927616000520
    OpenUrlCrossRefPubMed
  21. ↵
    1. Bodenstine TM,
    2. Seftor REB,
    3. Khalkhali-Ellis Z,
    4. Seftor EA,
    5. Pemberton PA,
    6. Hendrix MJC
    : Maspin: molecular mechanisms and therapeutic implications. Cancer Metastasis Rev 31(3-4): 529-551, 2012. DOI: 10.1007/s10555-012-9361-0
    OpenUrlCrossRefPubMed
  22. ↵
    1. Zou Z,
    2. Anisowicz A,
    3. Hendrix MJC,
    4. Thor A,
    5. Neveu M,
    6. Sheng S,
    7. Rafidi K,
    8. Seftor E,
    9. Sager R
    : Maspin, a serpin with tumor-suppressing activity in human mammary epithelial cells. Science 263(5146): 526-529, 1994. DOI: 10.1126/science.8290962
    OpenUrlAbstract/FREE Full Text
  23. ↵
    1. Riddick AC,
    2. Shukla CJ,
    3. Pennington CJ,
    4. Bass R,
    5. Nuttall RK,
    6. Hogan A,
    7. Sethia KK,
    8. Ellis V,
    9. Collins AT,
    10. Maitland NJ,
    11. Ball RY,
    12. Edwards DR
    : Identification of degradome components associated with prostate cancer progression by expression analysis of human prostatic tissues. Br J Cancer 92(12): 2171-2180, 2005. DOI: 10.1038/sj.bjc.6602630
    OpenUrlCrossRefPubMed
    1. Luo JL,
    2. Tan W,
    3. Ricono JM,
    4. Korchynskyi O,
    5. Zhang M,
    6. Gonias SL,
    7. Cheresh DA,
    8. Karin M
    : Nuclear cytokine-activated IKKα controls prostate cancer metastasis by repressing Maspin. Nature 446(7136): 690-694, 2007. DOI: 10.1038/nature05656
    OpenUrlCrossRefPubMed
  24. ↵
    1. Berardi R,
    2. Morgese F,
    3. Onofri A,
    4. Mazzanti P,
    5. Pistelli M,
    6. Ballatore Z,
    7. Savini A,
    8. De Lisa M,
    9. Caramanti M,
    10. Rinaldi S,
    11. Pagliaretta S,
    12. Santoni M,
    13. Pierantoni C,
    14. Cascinu S
    : Role of maspin in cancer. Clin Transl Med 2(1): 8, 2013. DOI: 10.1186/2001-1326-2-8
    OpenUrlCrossRefPubMed
  25. ↵
    1. Lyu P,
    2. Zhang SD,
    3. Yuen HF,
    4. McCrudden CM,
    5. Wen Q,
    6. Chan KW,
    7. Kwok HF
    : Identification of TWIST-interacting genes in prostate cancer. Sci China Life Sci 60(4): 386-396, 2017. DOI: 10.1007/s11427-016-0262-6
    OpenUrlCrossRefPubMed
  26. ↵
    1. Qian S,
    2. Zhang S,
    3. Wu Y,
    4. Ding Y,
    5. Shen, Li X
    : Protein disulfide isomerase 4 drives docetaxel resistance in prostate cancer. Chemotherapy 65(5-6): 125-133, 2020. DOI: 10.1159/000511505
    OpenUrlCrossRefPubMed
  27. ↵
    1. Pressinotti NC,
    2. Klocker H,
    3. Schäfer G,
    4. Luu VD,
    5. Ruschhaupt M,
    6. Kuner R,
    7. Steiner E,
    8. Poustka A,
    9. Bartsch G,
    10. Sültmann H
    : Differential expression of apoptotic genes PDIA3 and MAP3K5 distinguishes between low- and high-risk prostate cancer. Mol Cancer 8: 130, 2009. DOI: 10.1186/1476-4598-8-130
    OpenUrlCrossRefPubMed
  28. ↵
    1. Karlsson S,
    2. Olausson J,
    3. Lundh D,
    4. Sögård P,
    5. Mandal A,
    6. Holmström KO,
    7. Stahel A,
    8. Bengtsson J,
    9. Larsson D
    : Vitamin D and prostate cancer: The role of membrane initiated signaling pathways in prostate cancer progression. J Steroid Biochem Mol Biol 121(1-2): 413-416, 2010. DOI: 10.1016/j.jsbmb.2010.03.083
    OpenUrlCrossRefPubMed
  29. ↵
    1. Karlsson S,
    2. Diaz Cruz MA,
    3. Faresjö M,
    4. Khamou AP,
    5. Larsson D
    : Inhibition of CYP27B1 and CYP24 increases the anti-proliferative effects of 25-hydroxyvitamin D3 in LNCaP cells. Anticancer Res 41(10): 4733-4740, 2021. DOI: 10.21873/anticanres.15288
    OpenUrlAbstract/FREE Full Text
  30. ↵
    1. Basu A,
    2. Cajigas-Du Ross CK,
    3. Rios-Colon L,
    4. Mediavilla-Varela M,
    5. Daniels-Wells TR,
    6. Leoh LS,
    7. Rojas H,
    8. Banerjee H,
    9. Martinez SR,
    10. Acevedo-Martinez S,
    11. Casiano CA
    : LEDGF/p75 overexpression attenuates oxidative stress-induced necrosis and upregulates the oxidoreductase ERP57/PDIA3/GRP58 in prostate cancer. PLoS One 11(1): e0146549, 2016. DOI: 10.1371/journal.pone.0146549
    OpenUrlCrossRefPubMed
  31. ↵
    1. Diaz Cruz MA,
    2. Karlsson S,
    3. Szekeres F,
    4. Faresjö M,
    5. Lund D,
    6. 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 48(3): 2429-2436, 2021. DOI: 10.1007/s11033-021-06277-1
    OpenUrlCrossRefPubMed
  32. ↵
    1. Cancer Genome Atlas Research Network,
    2. Weinstein JN,
    3. Collisson EA,
    4. Mills GB,
    5. Shaw KR,
    6. Ozenberger BA,
    7. Ellrott K,
    8. Shmulevich I,
    9. Sander C,
    10. Stuart JM
    : The Cancer Genome Atlas Pan-Cancer analysis project. Nat Genet 45(10): 1113-1120, 2013. DOI: 10.1038/ng.2764
    OpenUrlCrossRefPubMed
  33. ↵
    1. Goldman MJ,
    2. Craft B,
    3. Hastie M,
    4. Repečka K,
    5. McDade F,
    6. Kamath A,
    7. Banerjee A,
    8. Luo Y,
    9. Rogers D,
    10. Brooks AN,
    11. Zhu J,
    12. Haussler D
    : Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol 38(6): 675-678, 2020. DOI: 10.1038/s41587-020-0546-8
    OpenUrlCrossRefPubMed
  34. ↵
    1. Li B,
    2. Dewey CN
    : RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinformatics 12: 323, 2011. DOI: 10.1186/1471-2105-12-323
    OpenUrlCrossRefPubMed
  35. ↵
    1. Bass R,
    2. Wagstaff L,
    3. Ravenhill L,
    4. Ellis V
    : Binding of extracellular maspin to beta1 integrins inhibits vascular smooth muscle cell migration. J Biol Chem 284(40): 27712-27720, 2009. DOI: 10.1074/jbc.M109.038919
    OpenUrlAbstract/FREE Full Text
  36. ↵
    1. Keane KM,
    2. Bell PG,
    3. Lodge JK,
    4. Constantinou CL,
    5. Jenkinson SE,
    6. Bass R,
    7. Howatson G
    : Phytochemical uptake following human consumption of Montmorency tart cherry (L. Prunus cerasus) and influence of phenolic acids on vascular smooth muscle cells in vitro. Eur J Nutr 55(4): 1695-1705, 2016. DOI: 10.1007/s00394-015-0988-9
    OpenUrlCrossRefPubMed
  37. ↵
    1. Stone KR,
    2. Mickey DD,
    3. Wunderli H,
    4. Mickey GH,
    5. Paulson DF
    : Isolation of a human prostate carcinoma cell line (DU 145). Int J Cancer 21(3): 274-281, 1978. DOI: 10.1002/ijc.2910210305
    OpenUrlCrossRefPubMed
    1. Kaighn ME,
    2. Narayan KS,
    3. Ohnuki Y,
    4. Lechner JF,
    5. Jones LW
    : Establishment and characterization of a human prostatic carcinoma cell line (pc-3). Investig Urol 17(1): 16-23, 1979.
    OpenUrlPubMed
  38. ↵
    1. Gibas Z,
    2. Becher R,
    3. Kawinski E,
    4. Horoszewicz J,
    5. Sandberg AA
    : A high-resolution study of chromosome changes in a human prostatic carcinoma cell line (LNCaP). Cancer Genet Cytogenet 11(4): 399-404, 1984. DOI: 10.1016/0165-4608(84)90020-7
    OpenUrlCrossRefPubMed
  39. ↵
    1. Cussenot O,
    2. Berthon P,
    3. Berger R,
    4. Mowszowicz I,
    5. Faille A,
    6. Hojman F,
    7. Teillac P,
    8. Le Duc A,
    9. Calvo F
    : Immortalization of human adult normal prostatic epithelial cells by liposomes containing large T-SV40 gene. J Urol 146(3): 881-886, 1991. DOI: 10.1016/s0022-5347(17)37953-3
    OpenUrlCrossRefPubMed
  40. ↵
    1. Biliran H Jr.,
    2. Sheng S
    : Pleiotrophic inhibition of pericellular urokinase-type plasminogen activator system by endogenous tumor suppressive maspin1. Cancer Res 61(24): 8676-8682, 2001.
    OpenUrlAbstract/FREE Full Text
  41. ↵
    1. Cai Z,
    2. Zhou Y,
    3. Lei T,
    4. Chiu J,
    5. He Q
    : Mammary serine protease inhibitor inhibits epithelial growth factor-induced epithelial-mesenchymal transition of esophageal carcinoma cells. Cancer 115(1): 36-48, 2009. DOI: 10.1002/cncr.23991
    OpenUrlCrossRefPubMed
  42. ↵
    1. Machtens S,
    2. Serth J,
    3. Bokemeyer C,
    4. Bathke W,
    5. Minssen A,
    6. Kollmannsberger C,
    7. Hartmann J,
    8. Knüchel R,
    9. Kondo M,
    10. Jonas U,
    11. Kuczyk M
    : Expression of the p53 and maspin protein in primary prostate cancer: Correlation with clinical features. Int J Cancer 95(5): 337-342, 2001. DOI: 10.1002/1097-0215(20010920)95:5<337::AID-IJC1059>3.0.CO;2-1
    OpenUrlCrossRefPubMed
  43. ↵
    1. Zou Z,
    2. Zhang W,
    3. Young D,
    4. Gleave MG,
    5. Rennie P,
    6. Connell T,
    7. Connelly R,
    8. Moul J,
    9. Srivastava S,
    10. Sesterhenn I
    : Maspin expression profile in human prostate cancer (cap) and in vitro induction of maspin expression by androgen ablation. Clin Cancer Res 8(5): 1172-1177, 2002.
    OpenUrlAbstract/FREE Full Text
  44. ↵
    1. Cheng WL,
    2. Huang CY,
    3. Tai CJ,
    4. Chang YJ,
    5. Hung CS
    : Maspin enhances the anticancer activity of curcumin in hormone-refractory prostate cancer cells. Anticancer Res 38(2): 863-870, 2018. DOI: 10.21873/anticanres.12295
    OpenUrlAbstract/FREE Full Text
  45. ↵
    1. Lee E,
    2. Lee DH
    : Emerging roles of protein disulfide isomerase in cancer. BMB Rep 50(8): 401-410, 2017. DOI: 10.5483/bmbrep.2017.50.8.107
    OpenUrlCrossRefPubMed
  46. ↵
    1. Kim T-W,
    2. Ryu H-H,
    3. Li S-Y,
    4. Li C-H,
    5. Lim S-H,
    6. Jang W-Y,
    7. Jung S
    : PDIA6 regulation of ADAM17 shedding activity and EGFR-mediated migration and invasion of glioblastoma cells. J Neurosurg 126(6): 1829-1838, 2016. DOI: 10.3171/2016.5.JNS152831
    OpenUrlCrossRefPubMed
PreviousNext
Back to top

In this issue

Anticancer Research: 43 (12)
Anticancer Research
Vol. 43, Issue 12
December 2023
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover
  • Index by author
  • Back Matter (PDF)
  • Ed Board (PDF)
  • Front Matter (PDF)
Print
Download PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for your interest in spreading the word on Anticancer Research.

NOTE: We only request your email address so that the person you are recommending the page to knows that you wanted them to see it, and that it is not junk mail. We do not capture any email address.

Enter multiple addresses on separate lines or separate them with commas.
PDIA6 and Maspin in Prostate Cancer
(Your Name) has sent you a message from Anticancer Research
(Your Name) thought you would like to see the Anticancer Research web site.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
6 + 11 =
Solve this simple math problem and enter the result. E.g. for 1+3, enter 4.
Citation Tools
PDIA6 and Maspin in Prostate Cancer
TORA K. SMULDERS-SRINIVASAN, SARAH E. JENKINSON, LOUISE J. BROWN, VASILEIOS P. LENIS, ROSEMARY BASS
Anticancer Research Dec 2023, 43 (12) 5331-5340; DOI: 10.21873/anticanres.16736

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Reprints and Permissions
Share
PDIA6 and Maspin in Prostate Cancer
TORA K. SMULDERS-SRINIVASAN, SARAH E. JENKINSON, LOUISE J. BROWN, VASILEIOS P. LENIS, ROSEMARY BASS
Anticancer Research Dec 2023, 43 (12) 5331-5340; DOI: 10.21873/anticanres.16736
Twitter logo Facebook logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Materials and Methods
    • Results
    • Discussion
    • Conclusion
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF

Related Articles

  • No related articles found.
  • PubMed
  • Google Scholar

Cited By...

  • Bladder Volume <200 ml During a Course of Moderate Hypofractionated Irradiation in Patients With Localized Prostate Cancer
  • Changes in Bladder Volume During Radiotherapy for Prostate Cancer
  • Google Scholar

More in this TOC Section

  • Inhibition of Plasminogen Activator Inhibitor-1 (PAI-1) by Tiplaxtinin Reduces Aggressiveness of Cervical Carcinoma Cells
  • High Expression of OR1F1 Protein as a Potential Prognostic Biomarker for Esophageal Squamous Cell Carcinoma
  • Epigallocatechin Gallate Induces miR-192/215 Suppression of EGR1 in Gastric Cancer
Show more Experimental Studies

Similar Articles

Keywords

  • Prostate cancer
  • PDIA6
  • Maspin
  • tumor
  • cancer therapeutics
  • serpin family
  • PDI-P5
  • SERPIN B5
  • ERP5
  • TXNDC7
Anticancer Research

© 2025 Anticancer Research

Powered by HighWire