1
|
Lin C, Xie S, Wang M, Shen J. PRDX1 knockdown promotes erastin-induced ferroptosis and impedes diffuse large B-cell lymphoma development by inhibiting the MAPK/ERK pathway. BMC Cancer 2025; 25:806. [PMID: 40307771 PMCID: PMC12042503 DOI: 10.1186/s12885-025-14173-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 04/16/2025] [Indexed: 05/02/2025] Open
Abstract
AIM Diffuse large B-cell lymphoma (DLBCL) is an aggressive lymphoma and DLBCL cells are highly sensitive to ferroptosis. The purpose of this research was to evaluate the role and molecular mechanism of peroxiredoxin 1 (PRDX1) on ferroptosis in DLBCL. METHODS The expression of PRDX1 in DLBCL tissues and cells was detected using bioinformatics analysis and reverse transcription quantitative PCR. The impacts of PRDX1 on DLBCL cell proliferation, apoptosis, migration, invasion, and ferroptosis were assessed through a series of in vitro experiments. A xenograft tumor model was constructed to verify the roles of PRDX1 in vivo. Transcriptome sequencing was conducted to identify PRDX1-mediated signaling pathways. Anisomycin, an agonist of mitogen-activated protein kinase (MAPK), was used to explore the modulation of PRDX1 on the MAPK pathway. RESULTS PRDX1 expression was upregulated in DLBCL. PRDX1 knockdown inhibited DLBCL cell proliferation, migration, and invasion, promoted apoptosis, and suppressed xenograft tumor growth. PRDX1 knockdown boosted erastin-induced ferroptosis by increasing the levels of iron and MDA, while decreasing the levels of GSH. It also promoted COX2 protein expression and inhibited GPX4 and SLC7A11 protein levels. PRDX1 knockdown reduced the phosphorylation levels of MEK and ERK both under conditions with or without erastin induction. The MAPK/ERK pathway agonist anisomycin, significantly reversed the inhibitory effects of PRDX1 knockdown on the malignant behaviors of DLBCL cells and the promotion of ferroptosis. CONCLUSION PRDX1 knockdown facilitates erastin-induced ferroptosis and obstacles DLBCL progression by inhibiting the MAPK/ERK pathway, offering a potential treatment strategy for DLBCL treatment.
Collapse
MESH Headings
- Ferroptosis/drug effects
- Ferroptosis/genetics
- Humans
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Animals
- Mice
- Peroxiredoxins/genetics
- Peroxiredoxins/metabolism
- MAP Kinase Signaling System/drug effects
- MAP Kinase Signaling System/genetics
- Piperazines/pharmacology
- Cell Proliferation/drug effects
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Cell Movement
- Gene Knockdown Techniques
- Gene Expression Regulation, Neoplastic
- Apoptosis/drug effects
- Male
- Female
Collapse
Affiliation(s)
- Chuanming Lin
- Department of Hematology, First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Economic Development District, Ganzhou City, Jiangxi Province, 341000, China
- Fujian Medical University Union Hospital, No. 29, Xinquan Road, Gulou District, Fuzhou City, Fujian Province, 350001, China
| | - Shuiling Xie
- Department of Hematology, First Affiliated Hospital of Gannan Medical University, No. 128, Jinling Road, Economic Development District, Ganzhou City, Jiangxi Province, 341000, China
| | - Menger Wang
- Gannan Medical University, Rongjiang New District University Park, Ganzhou City, Jiangxi Province, 341099, China
| | - Jianzhen Shen
- Fujian Medical University Union Hospital, No. 29, Xinquan Road, Gulou District, Fuzhou City, Fujian Province, 350001, China.
| |
Collapse
|
2
|
Li J, Li W, Li L, Wang W, Zhang M, Tang X. Induction of Peroxiredoxin 1 by Hypoxia Promotes Cellular Autophagy and Cell Proliferation in Oral Leukoplakia via HIF-1α/BNIP3 Pathway. J Mol Histol 2024; 55:403-413. [PMID: 38758520 DOI: 10.1007/s10735-024-10197-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/23/2024] [Indexed: 05/18/2024]
Abstract
Hypoxia is a key trigger in the transformation of oral leukoplakia into oral cancer. However, it is still too early to determine the role of hypoxia in the development of oral leukoplakia. Prx1, an antioxidant protein, upregulated by hypoxia, regulates cellular autophagy in leukoplakia. This study aimed to understand the mechanisms by which hypoxia induces Prx1 expression during autophagy in oral leukoplakia. We used an experimental model of tongue epithelial hyperplasia induced by 4-nitroquinoline-1-oxide (4NQO) and dysplastic oral keratinocytes. Prx1 knockdown DOK cells, Leuk-1 cells and control cells were harvested, and cell proliferation was assayed using the Cell Counting Kit-8. Several hypoxia and autophagy-related proteins were examined using quantitative real-time polymerase chain reaction, immunohistochemistry, immunofluorescence, and western blotting in cells and mouse tongue tissues. In addition, the ultrastructure of the cells was observed by transmission electron microscopy. Hypoxia induces cell proliferation, autophagic vesicles and the expression of Prx1, BNIP3, LC3II/I and Beclin-1 in DOK and Leuk-1 cells. However, these effects were all attenuated by Prx1 knockdown. Histologically, 4NQO induced epithelial hyperplasia in the tongue mucosa. The expression of proliferation marker PCNA, autophagy-related proteins LC3B and Beclin-1, as well as HIF-1α/BNIP3 was significantly lower in the tongue tissues of Prx1flox/flox:Cre+ mice compared with Prx1flox/flox mice. In Prx1flox/flox:Cre+ mice, an increased expression of HIF-1α/BNIP3, LC3B and Beclin-1 was detected in epithelial hyperplasia tongue tissues compared to normal tissues. The current study suggests that Prx1 may promotes cell proliferation and autophagy in oral leukoplakia cells via the HIF-1α/BNIP3 pathway.
Collapse
Affiliation(s)
- Jing Li
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China
| | - Wenjing Li
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China
| | - Lingyu Li
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China
| | - Wenchao Wang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China
| | - Min Zhang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China.
| | - Xiaofei Tang
- Beijing Institute of Dental Research, Beijing Stomatological Hospital and School of Stomatology, Capital Medical University, Dongcheng District, Beijing, China.
| |
Collapse
|
3
|
Borović Šunjić S, Jaganjac M, Vlainić J, Halasz M, Žarković N. Lipid Peroxidation-Related Redox Signaling in Osteosarcoma. Int J Mol Sci 2024; 25:4559. [PMID: 38674143 PMCID: PMC11050283 DOI: 10.3390/ijms25084559] [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: 03/29/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Oxidative stress and lipid peroxidation play important roles in numerous physiological and pathological processes, while the bioactive products of lipid peroxidation, lipid hydroperoxides and reactive aldehydes, act as important mediators of redox signaling in normal and malignant cells. Many types of cancer, including osteosarcoma, express altered redox signaling pathways. Such redox signaling pathways protect cancer cells from the cytotoxic effects of oxidative stress, thus supporting malignant transformation, and eventually from cytotoxic anticancer therapies associated with oxidative stress. In this review, we aim to explore the status of lipid peroxidation in osteosarcoma and highlight the involvement of lipid peroxidation products in redox signaling pathways, including the involvement of lipid peroxidation in osteosarcoma therapies.
Collapse
Affiliation(s)
- Suzana Borović Šunjić
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia; (M.J.); (J.V.); (M.H.)
| | | | | | | | - Neven Žarković
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia; (M.J.); (J.V.); (M.H.)
| |
Collapse
|
4
|
Effects of Antioxidant Gene Overexpression on Stress Resistance and Malignization In Vitro and In Vivo: A Review. Antioxidants (Basel) 2022; 11:antiox11122316. [PMID: 36552527 PMCID: PMC9774954 DOI: 10.3390/antiox11122316] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Reactive oxygen species (ROS) are normal products of a number of biochemical reactions and are important signaling molecules. However, at the same time, they are toxic to cells and have to be strictly regulated by their antioxidant systems. The etiology and pathogenesis of many diseases are associated with increased ROS levels, and many external stress factors directly or indirectly cause oxidative stress in cells. Within this context, the overexpression of genes encoding the proteins in antioxidant systems seems to have become a viable approach to decrease the oxidative stress caused by pathological conditions and to increase cellular stress resistance. However, such manipulations unavoidably lead to side effects, the most dangerous of which is an increased probability of healthy tissue malignization or increased tumor aggression. The aims of the present review were to collect and systematize the results of studies devoted to the effects resulting from the overexpression of antioxidant system genes on stress resistance and carcinogenesis in vitro and in vivo. In most cases, the overexpression of these genes was shown to increase cell and organism resistances to factors that induce oxidative and genotoxic stress but to also have different effects on cancer initiation and promotion. The last fact greatly limits perspectives of such manipulations in practice. The overexpression of GPX3 and SOD3 encoding secreted proteins seems to be the "safest" among the genes that can increase cell resistance to oxidative stress. High efficiency and safety potential can also be found for SOD2 overexpression in combinations with GPX1 or CAT and for similar combinations that lead to no significant changes in H2O2 levels. Accumulation, systematization, and the integral analysis of data on antioxidant gene overexpression effects can help to develop approaches for practical uses in biomedical and agricultural areas. Additionally, a number of factors such as genetic and functional context, cell and tissue type, differences in the function of transcripts of one and the same gene, regulatory interactions, and additional functions should be taken into account.
Collapse
|
5
|
Kılıç N, Boyacıoğlu Ö, Saltoğlu GT, Bulduk EB, Kurt G, Korkusuz P. Thioredoxin System and miR-21, miR-23a/b and let-7a as Potential Biomarkers for Brain Tumor Progression: Preliminary Case Data. World Neurosurg 2022; 167:e1299-e1309. [PMID: 36096386 DOI: 10.1016/j.wneu.2022.09.024] [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: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND The thioredoxin system and microRNAs (miRNAs) are potential targets for both cancer progression and treatment. However, the role of miRNAs and their relation with the expression profile of thioredoxin system in brain tumor progression remains unclear. METHODS In this study, we aimed to determine the expression profiles of redox components Trx-1, TrxR-1 and PRDX-1, and oncogenic miR-21, miR-23a/b and let-7a and oncosuppressor miR-125 in different brain tumor tissues and their association with increasing tumor grade. We studied Trx-1, TrxR-1, and PRDX-1 messenger RNA expression levels by quantitative real-time polymerase chain reaction and protein levels by Western blot and miR-23a, miR-23b, miR-125a, miR-21, and let-7a miRNA expression levels by quantitative real-time polymerase chain reaction in 16 glioma, 15 meningioma, 5 metastatic, and 2 benign tumor samples. We also examined Trx-1, TrxR-1, and PRDX-1 protein levels in serum samples of 36 patients with brain tumor and 37 healthy volunteers by enzyme-linked immunosorbent assay. RESULTS We found that Trx-1, TrxR-1, and PRDX-1 presented high messenger RNA expression but low protein expression in low-grade brain tumor tissues, whereas they showed higher protein expression in sera of patients with low-grade brain tumors. miR-23b, miR-21, miR-23a, and let-7a were highly expressed in low-grade brain tumor tissues and positively correlated with the increase in thioredoxin system activity. CONCLUSIONS Our findings showed that Trx-1, TrxR-1, miR-21, miR-23a/b, and let-7a might be used for brain tumor diagnosis in the clinic. Further prospective studies including molecular pathway analyses are required to validate the miRNA/Trx system regulatory axis in brain tumor progression.
Collapse
Affiliation(s)
- Nedret Kılıç
- Department of Medical Biochemistry, Faculty of Medicine, Atılım University, Gölbaşı, Ankara, Turkey.
| | - Özge Boyacıoğlu
- Department of Medical Biochemistry, Faculty of Medicine, Atılım University, Gölbaşı, Ankara, Turkey; Department of Bioengineering, Graduate School of Science and Engineering, Hacettepe University, Beytepe, Ankara, Turkey
| | - Gamze Turna Saltoğlu
- Department of Biochemistry, Faculty of Medicine, Kırşehir Ahi Evran University, Bağbaşı, Kırşehir, Turkey
| | - Erkut Baha Bulduk
- Department of Neurosurgery, Faculty of Medicine, Atılım University, Gölbaşı, Ankara, Turkey
| | - Gökhan Kurt
- Department of Neurosurgery, Faculty of Medicine, Gazi University, Beşevler, Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Faculty of Medicine, Hacettepe University, Sıhhiye, Ankara, Turkey
| |
Collapse
|
6
|
OTSUKA N, ISHIMARU K, MURAKAMI M, GOTO M, HIRATA A, SAKAI H. The immunohistochemical detection of peroxiredoxin 1 and 2 in canine spontaneous vascular endothelial tumors. J Vet Med Sci 2022; 84:914-923. [PMID: 35584951 PMCID: PMC9353087 DOI: 10.1292/jvms.22-0102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 05/01/2022] [Indexed: 11/22/2022] Open
Abstract
Peroxiredoxin (PRDX) is an antioxidant enzyme family with six isoforms (PRDX1-6). The main function of PRDXs is to decrease cellular oxidative stress by reducing reactive oxygen species, such as hydrogen peroxide, to H2O. Recently, it has been reported that PRDXs are overexpressed in various malignant tumors in humans, and are involved in the development, proliferation, and metastasis of tumors. However, studies on the expression of PRDXs in tumors of animals are limited. Therefore, in the present study, we immunohistochemically investigated the expression of PRDX1 and 2 in spontaneous canine hemangiosarcoma (HSA) and hemangioma (HA), as well as in selected normal tissue and granulation tissue, including newly formed blood vessels. Although there were some exceptions, immunolocalization of PRDX1 and 2 in normal canine tissues was similar to those in humans, rats, or mice. In granulation tissue, angiogenic endothelial cells were strongly positive for PRDX1 and 2, whereas quiescent endothelial cells in mature vessels were negative. Both PRDX1 and 2 were significantly highly expressed in HSA compared to HA. There were no significant differences in the expression of PRDX1 and 2 among the subtypes and primary sites of HSA. These results suggest that PRDX1 and 2 may be involved in the angiogenic phenotypes of endothelial cells in granulation tissue as well as in the behavior in the malignant endothelial tumors.
Collapse
Affiliation(s)
- Narumi OTSUKA
- Laboratory of Veterinary Pathology, Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Kairi ISHIMARU
- Laboratory of Veterinary Pathology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Mami MURAKAMI
- Laboratory of Veterinary Clinical Oncology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Minami GOTO
- Laboratory of Veterinary Pathology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Akihiro HIRATA
- Laboratory of Veterinary Pathology, Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
- Laboratory of Veterinary Pathology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| | - Hiroki SAKAI
- Laboratory of Veterinary Pathology, Joint Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
- Laboratory of Veterinary Pathology, Faculty of Applied Biological Sciences, Gifu University, Gifu, Japan
| |
Collapse
|
7
|
Shen Y, Xu H, Li L, Lu Y, Zhang M, Huang X, Tang X. Assessment of Potential Prognostic Value of Peroxiredoxin 1 in Oral Squamous Cell Carcinoma. Cancer Manag Res 2021; 13:5725-5737. [PMID: 34290530 PMCID: PMC8289334 DOI: 10.2147/cmar.s319048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 06/28/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose The role of the peroxiredoxin (PRDX) family in oral squamous cell carcinoma (OSCC) remains unclear. This study aimed to investigate the expression of PRDXs and their effects on the prognosis in OSCC. Methods The expression of PRDXs and their effects on prognosis were analysed in 216 OSCC samples from The Cancer Genome Atlas (TCGA) database. OSCC tissues and adjacent noncancerous tissues (ANTs) were obtained from 68 clinical patients. Quantitative real-time (qRT)-PCR, Western blot, and immunohistochemical (IHC) staining were used to verify the relationship between the expression level of PRDX1 and different clinical features. Gene set enrichment analysis (GSEA) was used to examine the molecular mechanism of PRDX1 in OSCC. Results PRDX1 was found to be the only gene in PRDX family that highly expressed in OSCC samples and affected the prognosis of patients with OSCC. PRDX1 expression was significantly related to tumor stage, lymphatic metastasis, and pathological grade. A nomogram consisting of tumor stage, N stage, and PRDX1 level was constructed. GSEA showed that high expression of PRDX1 involved many cancer-related molecular functions and signaling pathways. Conclusion PRDX1 may play an important role in the occurrence and development of OSCC, and may be a potential new target for OSCC treatment.
Collapse
Affiliation(s)
- Yajun Shen
- Beijing Institute of Dental Research, Beijing Key Laboratory, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Haoyue Xu
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Lingyu Li
- Beijing Institute of Dental Research, Beijing Key Laboratory, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Yunping Lu
- Beijing Institute of Dental Research, Beijing Key Laboratory, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Min Zhang
- Beijing Institute of Dental Research, Beijing Key Laboratory, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Xin Huang
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Xiaofei Tang
- Beijing Institute of Dental Research, Beijing Key Laboratory, Beijing Stomatological Hospital & School of Stomatology, Capital Medical University, Beijing, 100050, People's Republic of China
| |
Collapse
|
8
|
Song Z, Pearce MC, Jiang Y, Yang L, Goodall C, Miranda CL, Milovancev M, Bracha S, Kolluri SK, Maier CS. Delineation of hypoxia-induced proteome shifts in osteosarcoma cells with different metastatic propensities. Sci Rep 2020; 10:727. [PMID: 31959767 PMCID: PMC6971036 DOI: 10.1038/s41598-019-56878-x] [Citation(s) in RCA: 15] [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: 12/21/2018] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma (OS) is the most common bone cancer in children and young adults. Solid tumors are characterized by intratumoral hypoxia, and hypoxic cells are associated with the transformation to aggressive phenotype and metastasis. The proteome needed to support an aggressive osteosarcoma cell phenotype remains largely undefined. To link metastatic propensity to a hypoxia-induced proteotype, we compared the protein profiles of two isogenic canine OS cell lines, POS (low metastatic) and HMPOS (highly metastatic), under normoxia and hypoxia. Label-free shotgun proteomics was applied to comprehensively characterize the hypoxia-responsive proteome profiles in the OS cell phenotypes. Hypothesis-driven parallel reaction monitoring was used to validate the differential proteins observed in the shotgun data and to monitor proteins of which we expected to exhibit hypoxia responsiveness, but which were absent in the label-free shotgun data. We established a "distance" score (|zHMPOS - zPOS|), and "sensitivity" score (|zHypoxia - zNormoxia) to quantitatively evaluate the proteome shifts exhibited by OS cells in response to hypoxia. Evaluation of the sensitivity scores for the proteome shifts observed and principal component analysis of the hypoxia-responsive proteins indicated that both cell types acquire a proteome that supports a Warburg phenotype with enhanced cell migration and proliferation characteristics. Cell migration and glucose uptake assays combined with protein function inhibitor studies provided further support that hypoxia-driven adaption of pathways associated with glycolytic metabolism, collagen biosynthesis and remodeling, redox regulation and immunomodulatory proteins typify a proteotype associated with an aggressive cancer cell phenotype. Our findings further suggest that proteins involved in collagen remodeling and immune editing may warrant further evaluation as potential targets for anti-metastatic treatment strategies in osteosarcoma.
Collapse
Affiliation(s)
- Zifeng Song
- Department of Chemistry, Oregon State University, Oregon, USA
| | - Martin C Pearce
- Department of Environmental & Molecular Toxicology, Oregon State University, Oregon, USA
| | - Yuan Jiang
- Department of Statistics, Oregon State University, Oregon, USA
| | - Liping Yang
- Department of Chemistry, Oregon State University, Oregon, USA
| | - Cheri Goodall
- College of Veterinary Medicine, Oregon State University, Oregon, USA
| | | | - Milan Milovancev
- College of Veterinary Medicine, Oregon State University, Oregon, USA
| | - Shay Bracha
- College of Veterinary Medicine, Oregon State University, Oregon, USA
| | - Siva K Kolluri
- Department of Environmental & Molecular Toxicology, Oregon State University, Oregon, USA
- Linus Pauling Institute, Oregon State University, Oregon, USA
| | - Claudia S Maier
- Department of Chemistry, Oregon State University, Oregon, USA.
- Linus Pauling Institute, Oregon State University, Oregon, USA.
| |
Collapse
|
9
|
Lu E, Hu X, Pan C, Chen J, Xu Y, Zhu X. Up-regulation of peroxiredoxin-1 promotes cell proliferation and metastasis and inhibits apoptosis in cervical cancer. J Cancer 2020; 11:1170-1181. [PMID: 31956363 PMCID: PMC6959069 DOI: 10.7150/jca.37147] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/26/2019] [Indexed: 02/07/2023] Open
Abstract
Objective: To investigate the effect of peroxiredoxin 1 (PRDX1) on the biological behavior of cervical cancer cells and the possible mechanism. Materials and methods: The expression of PRDX1 in human cervical cancer tissues and adjacent non-tumor tissues were detected by immunohistochemistry (IHC). Lentivirus containing PRDX1-cDNA or shRNA against PRDX1 was constructed to overexpress or knockdown PRDX1 in SiHa cervical cancer cells. Cell proliferation was tested by CCK-8 and BrdU incorporation assay and cell apoptosis was evaluated by AnnexinV-PE /7AAD assay. Scratch wound and transwell invasion assay were used to test migration and invasion activity after PRDX1 was overexpressed or suppressed. Furthermore, the effect of PRDX1 on cell proliferation and apoptosis was also studied using a xenograft model of nude mice. Results: The expression of PRDX1 protein was significantly up-regulated in the tumor tissues compared with the paired adjacent non-tumor tissues. Meanwhile, PRDX1 overexpression was associated with tumor stage, lymphatic metastasis and differentiation. Overexpression of PRDX1 significantly promoted proliferation and inhibited apoptosis by increasing the expression of Nanog, proliferating cell nuclear antigen (PCNA), B-cell lymphoma-2 (Bcl-2) and downregulating the expression of Bcl2-associated X protein (BAX) in SiHa cervical cancer cells. Moreover, PRDX1 overexpression increased invasion and migration of SiHa cervical cancer cells via up-regulating the expression of Snail and matrix metalloprotein 9 (MMP-9) and down-regulating the expression of E-cadherin. Knockdown of PRDX1 resulted in the opposite results. The role of PRDX1 in promoting SiHa cervical cancer cell proliferation and inhibiting apoptosis has also been confirmed in vivo in a mouse xenograft model. Conclusions: PRDX1 promoted cell proliferation, migration, and invasion and suppressed apoptosis of cervical cancer possibly via regulating the expression of related protein.
Collapse
Affiliation(s)
| | | | | | | | | | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou 325027, China
| |
Collapse
|
10
|
Song Y, Liu H, Cui C, Peng X, Wang C, Tian X, Li W. Silencing of Peroxiredoxin 1 Inhibits the Proliferation of Esophageal Cancer Cells and Promotes Apoptosis by Inhibiting the Activity of the PI3K/AKT Pathway. Cancer Manag Res 2019; 11:10883-10890. [PMID: 31920397 PMCID: PMC6941600 DOI: 10.2147/cmar.s235317] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 12/12/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE To study the effect of peroxiredoxin 1 (PRDX1) on esophageal squamous carcinoma cells and determine whether it plays a role in regulating the PI3K/AKT signaling pathway. METHODS Three esophageal squamous cell carcinoma cell lines (Eca-109, EC9706, and KYSE150) and one normal cell line (human esophageal epithelial cells) were selected. The protein expression of peroxiredoxin 1 (PRDX1) and the activity of the PI3K/AKT pathway were detected via Western blotting. The proliferation ability of cells was detected through the MTT assay and cell clone formation. Apoptosis was detected using flow cytometry. Subsequently, cells were treated with a PI3K/AKT pathway inhibitor and activator, alone or in combination with silencing of PRDX1, and the above indicators were re-tested. RESULTS The expression of PRDX1 and activity of PI3K/AKT pathway-associated proteins were higher in esophageal cancer cells than in normal esophageal epithelial cells. Compared with normal human esophageal epithelial cells, the proliferation of the three types of esophageal cancer cells was increased, whereas their level of apoptosis was decreased (p<0.05). In Eca-109 cells (cell line with silenced expression of PRDX1), the expression of PRDX1 was significantly decreased. In contrast to the control group, the proliferation and clonality of cells in the silencing PRDX1 group was decreased, the proportion of apoptotic cells was increased, and the phosphorylation levels of PI3K and AKT were decreased (p<0.05). Compared with the control group, treatment with the inhibitor LY294002 alone significantly inhibited cell proliferation and promoted apoptosis (p<0.05); this effect was similar to that observed in the silencing PRDX1 group. CONCLUSION PRDX1 was highly expressed in esophageal cancer cells. Silencing of PRDX1 can inhibit the proliferation of esophageal cancer cells and promote apoptosis. The mechanism involved in this process may be related to the inhibition of the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Yingjian Song
- Department of Thoracic Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai264000, Shandong, People’s Republic of China
| | - Huimin Liu
- Department of Gastroenterology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai264000, Shandong, People’s Republic of China
| | - Chunling Cui
- Library, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai264000, Shandong, People’s Republic of China
| | - Xiaonu Peng
- Department of Thoracic Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai264000, Shandong, People’s Republic of China
| | - Chaoyang Wang
- Department of Thoracic Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai264000, Shandong, People’s Republic of China
| | - Xudong Tian
- Department of Thoracic Surgery, Liaocheng People’s Hospital, Liaocheng252000, Shandong, People’s Republic of China
| | - Wenjun Li
- Department of Thoracic Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai264000, Shandong, People’s Republic of China
| |
Collapse
|