Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Qiou Wei is active.

Publication


Featured researches published by Qiou Wei.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Sulfiredoxin–Peroxiredoxin IV axis promotes human lung cancer progression through modulation of specific phosphokinase signaling

Qiou Wei; Hong Jiang; Zhen Xiao; Alyson R. Baker; Matthew R. Young; Timothy D. Veenstra; Nancy H. Colburn

Oxidative stress is known to cause tumorigenesis through induction of DNA and lipid damage. It also promotes cancer progression through a largely unknown mechanism. Sulfiredoxin (Srx) is a novel oxidative stress-induced antioxidant protein whose function in tumorigenesis and cancer progression has not been well studied. We report that Srx is highly expressed in human lung cancer. Knockdown of Srx reduces anchorage-independent colony formation, cell migration, and invasion of human lung cancer cells. Srx preferentially interacts with Peroxiredoxin (Prx) IV relative to other Prxs due to its intrinsic higher binding affinity. Knockdown of Prx IV recapitulates the phenotypic changes of depleting Srx. Disruption or enhancement of the Srx–Prx IV axis leads respectively to reduction or acceleration of tumor growth and metastasis formation in vivo. Through identification and validation of the downstream mediators we unraveled the Srx-mediated signaling network that traverses AP-1–activating and other phosphokinase signaling cascades. Our work reveals that the Srx–Prx IV axis is critical for lung cancer maintenance and metastasis, suggesting that targeting the Srx–Prx IV axis may provide unique effective strategies for cancer prevention and treatment.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Sulfiredoxin is an AP-1 target gene that is required for transformation and shows elevated expression in human skin malignancies

Qiou Wei; Hong Jiang; Connie P. Matthews; Nancy H. Colburn

Previous studies have shown that a dominant negative form of c-Jun (TAM67) suppresses mouse skin carcinogenesis both in vitro and in vivo. The current study identifies Sulfiredoxin (Srx) as a unique target of activator protein-1 (AP-1) activation and TAM67 inhibition. Manipulation of Srx levels by ShRNA or over-expression demonstrates that Srx is critical for redox homeostasis through reducing hyperoxidized peroxiredoxins. In JB6 cells, knockdown of Srx abolishes tumor promoter-induced transformation and enhances cell sensitivity to oxidative stress. Knockdown of Srx also impairs c-Jun phosphorylation, implicating a role for Srx in the feedback regulation of AP-1 activity. Screening of patient tissues by tissue microarray reveals elevated Srx expression in several types of human skin cancers. Our study indicates that Srx is a functionally significant target of AP-1 blockade that may have value in cancer prevention or treatment.


Cancer Letters | 2015

The sulfiredoxin-peroxiredoxin (Srx-Prx) axis in cell signal transduction and cancer development.

Murli Mishra; Hong Jiang; Lisha Wu; Hedy A. Chawsheen; Qiou Wei

Redox signaling is a critical component of cell signaling pathways that are involved in the regulation of cell growth, metabolism, hormone signaling, immune regulation and variety of other physiological functions. Peroxiredoxin (Prx) is a family of thiol-based peroxidase that acts as a regulator of redox signaling. Members of Prx family can act as antioxidants and chaperones. Sulfiredoxin (Srx) is an antioxidant protein that exclusively reduces over-oxidized typical 2-Cys Prx. Srx has different affinities for individual Prx and it also catalyzes the deglutathionylation of variety of substrates. Individual component of the Srx-Prx system plays critical role in carcinogenesis by modulating cell signaling pathways involved in cell proliferation, migration and metastasis. Expression levels of individual component of the Srx-Prx axis have been correlated with patient survival outcome in multiple cancer types. This review will summarize the molecular basis of differences in the affinity of Srx for individual Prx and the role of individual component of the Srx-Prx system in tumor progression and metastasis. This enhanced understanding of molecular aspects of Srx-Prx interaction and its role in cell signal transduction will help define the Srx-Prx system as a future therapeutic target in human cancer.


Carcinogenesis | 2014

Tumor promoter-induced sulfiredoxin is required for mouse skin tumorigenesis

Lisha Wu; Hong Jiang; Hedy A. Chawsheen; Murli Mishra; Matthew R. Young; Matthieu Gerard; Michel B. Toledano; Nancy H. Colburn; Qiou Wei

Sulfiredoxin (Srx), the exclusive enzyme that reduces the hyperoxidized inactive form of peroxiredoxins (Prxs), has been found highly expressed in several types of human skin cancer. To determine whether Srx contributed to skin tumorigenesis in vivo, Srx null mice were generated on an FVB background. Mouse skin tumorigenesis was induced by a 7,12-dimethylbenz[α]anthracene/12-O-tetradecanoylphorbol-13-acetate (DMBA/TPA) protocol. We found that the number, volume and size of papillomas in Srx(-/-) mice were significantly fewer compared with either wild-type (Wt) or heterozygous (Het) siblings. Histopathological analysis revealed more apoptotic cells in tumors from Srx(-/-) mice. Mechanistic studies in cell culture revealed that Srx was stimulated by TPA in a redox-independent manner. This effect was mediated transcriptionally through the activation of mitogen-activated protein kinase and Jun-N-terminal kinase. We also demonstrated that Srx was capable of reducing hyperoxidized Prxs to facilitate cell survival under oxidative stress conditions. These findings suggested that loss of Srx protected mice, at least partially, from DMBA/TPA-induced skin tumorigenesis. Therefore, Srx has an oncogenic role in skin tumorigenesis and targeting Srx may provide novel strategies for skin cancer prevention or treatment.


Molecular Cancer Research | 2015

Sulfiredoxin Promotes Colorectal Cancer Cell Invasion and Metastasis through a Novel Mechanism of Enhancing EGFR Signaling

Hong Jiang; Lisha Wu; Jing Chen; Murli Mishra; Hedy A. Chawsheen; Haining Zhu; Qiou Wei

Sulfiredoxin (SRXN1/Srx) is a multifunction enzyme with a primary antioxidant role of reducing the overoxidized inactive form of peroxiredoxins (Prxs). The function and mechanisms of Srx in cancer development are not well understood. Here, Srx is preferentially expressed in human colorectal cancer cells but not in normal colon epithelial cells. Loss-of-function studies demonstrate that knockdown of Srx in poorly differentiated colorectal cancer cells not only leads to the inhibition of colony formation and cell invasion in vitro, but also reduces tumor xenograft growth and represses metastasis to distal organs in a mouse orthotopic implantation model. Notably, exactly opposite effects were observed in gain-of-function experiments when Srx was ectopically expressed in well-differentiated colorectal cancer cells. Mechanistically, expression of Srx enhances the activation of MAPK signaling through increasing the C-terminal tyrosine phosphorylation levels of EGFR. This function of Srx is mediated through its inhibition of EGFR acetylation at K1037, a novel posttranslational modification of EGFR in human colorectal cancer cells identified by liquid chromatography-electrospray ionization-tandem mass spectrometry (LC/ESI/MS-MS) proteomic analysis. Furthermore, abolishment of K1037 acetylation in human colorectal cancer cells by site-specific mutagenesis leads to sustained activation of EGFR–MAPK signaling. Combined, these data reveal that Srx promotes colorectal cancer cell invasion and metastasis through a novel mechanism of enhancing EGFR signaling. Implications: Sulfiredoxin is a critical oncogenic protein that can be used as a molecular target to develop therapeutics for patients with metastatic colorectal cancer. Mol Cancer Res; 13(12); 1554–66. ©2015 AACR.


Genes and Diseases | 2018

A critical role of the thioredoxin domain containing protein 5 (TXNDC5) in redox homeostasis and cancer development

Hedy A. Chawsheen; Qi Ying; Hong Jiang; Qiou Wei

Correct folding of nascent peptides occurs in the endoplasmic reticulum (ER). It is a complicate process primarily accomplished by the coordination of multiple redox proteins including members of the protein disulfide isomerase (PDI) family. As a critical member of the PDI family, thioredoxin domain containing protein 5 (TXNDC5) assists the folding of newly synthesized peptides to their mature form through series of disulfide bond exchange reactions. Interestingly, TXNDC5 is frequently found overexpressed in specimens of many human diseases including various types of cancer. In this review, we summarized the biochemical function of TXNDC5 in mammalian cells and the recent progress on the understanding of its role and molecular mechanisms in cancer development. Findings of TXNDC5 in the activation of intracellular signaling pathways, stimulation of cell growth & proliferation, facilitation of cell survival and modulation of extracellular matrix to affect cancer cell invasion and metastasis are reviewed. These published studies suggest that strategies of targeting TXNDC5 can be developed as potentially valuable methods for the treatment of certain types of cancer in patients.


Cancer Letters | 2018

Nrf2-activated expression of sulfiredoxin contributes to urethane-induced lung tumorigenesis

Murli Mishra; Hong Jiang; Hedy A. Chawsheen; Matthieu Gerard; Michel B. Toledano; Qiou Wei

Lung cancer is the leading cause of cancer death worldwide. Cigarette smoking and exposure to chemical carcinogens are among the risk factors of lung tumorigenesis. In this study, we found that cigarette smoke condensate and urethane significantly stimulated the expression of sulfiredoxin (Srx) at the transcript and protein levels in cultured normal lung epithelial cells, and such stimulation was mediated through the activation of nuclear related factor 2 (Nrf2). To study the role of Srx in lung cancer development in vivo, mice with Srx wildtype, heterozygous or knockout genotype were subjected to the same protocol of urethane treatment to induce lung tumors. By comparing tumor multiplicity and volume between groups of mice with different genotype, we found that Srx knockout mice had a significantly lower number and smaller size of lung tumors. Mechanistically, we demonstrated that loss of Srx led to a decrease of tumor cell proliferation as well as an increase of tumor cell apoptosis. These data suggest that Srx may have an oncogenic role that contributes to the development of lung cancer in smokers or urethane-exposed human subjects.


Cancer Research | 2013

Abstract 2727: Sulfiredoxin is required for AOM/DSS induced mouse colon carcinogenesis and invasiveness of human colon cancer cells.

Qiou Wei; Hong Jiang; Alyson R. Baker; Lisa K. Dodge; Matthieu Gerard; Matthew R. Young; Michel B. Toledano; Nancy H. Colburn

Proceedings: AACR 104th Annual Meeting 2013; Apr 6-10, 2013; Washington, DC Sulfiredoxin (Srx) is the enzyme that reduces the overoxidized inactive form of peroxiredoxins (Prxs). To study the function of Srx in carcinogenesis in vivo, we tested whether loss of Srx protects mice from cancer development. Srx null mice were generated and colon carcinogenesis was induced by an azoxymethane (AOM) and dextran sulfate sodium (DSS) protocol. Compared to either wildtype or heterozygotes, Srx-/- mice had significantly reduced rates in both tumor multiplicity and volume. Mechanistic studies reveal that loss of Srx did not alter tumor cell proliferation; however, increased apoptosis and decreased inflammatory cell infiltration were obvious in tumors from Srx null mice compared to those from wildtype control. In addition to the AOM/DSS model, examination of Srx expression in human reveals a tissue-specific expression pattern. Srx expression was also demonstrated in tumors from colorectal cancer patients and the levels of expression were associated with patients’ clinic stage. These data provide the first in vivo evidence that loss of Srx renders mice resistance to AOM/DSS-induced colon carcinogenesis, suggesting that Srx has a critical oncogenic role in cancer development, and Srx may be used as a marker for human colon cancer pathogenicity. Citation Format: Qiou Wei, Hong Jiang, Alyson Baker, Lisa K. Dodge, Matthieu Gerard, Matthew R. Young, Michel B. Toledano, Nancy H. Colburn. Sulfiredoxin is required for AOM/DSS induced mouse colon carcinogenesis and invasiveness of human colon cancer cells. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 2727. doi:10.1158/1538-7445.AM2013-2727 Note: This abstract was not presented at the AACR Annual Meeting 2013 because the presenter was unable to attend.


Cancer Research | 2011

Abstract 3079: Identification of the translational targets of tumor suppressor Pdcd4

Noriko Yoshikawa; Matthew R. Young; Glenn Hegamyer; Alyson R. Baker; Arti N. Santhanam; Qiou Wei; Nancy H. Colburn

Proceedings: AACR 102nd Annual Meeting 2011‐‐ Apr 2‐6, 2011; Orlando, FL Programmed-cell-death-4 (Pdcd4) is a novel protein that functions as a transformation suppressor. Pdcd4 deficiency renders mice susceptible to increased tumor multiplicity in initiation-promotion skin carcinogenesis. On the contrary transgenic expression of Pdcd4 in mice inhibits DMBA-TPA induced skin tumorigenesis and tumor progression. Pdcd4 interacts with translation initiation factors eIF4A and eIF4G to inhibit translation in an mRNA-specific fashion and consequently blocks pro-oncogenic events such as activation of activator protein-1 (AP-1)-dependent transcription, anchorage-independent transformation and invasion. Pdcd4 expression decreases during carcinogenesis in several human cancer sites including colon, esophagus, lung, pancreas and brain and its activation appears to be important in the mechanism by which some cancer therapeutic drugs work. While Pdcd4 is not mutationally inactivated, its expression is post translationally regulated. Pdcd4 expression is decreased during carcinogenesis by mechanisms involving microRNA-21 (miR21) inhibition of translation and by S6 kinase-dependent proteasome degradation. Loss of Pdcd4 has been shown to affect transcription of TIMP-2, MAP4K1, LOX and u-PAR. Although the tumor suppressor Pdcd4 acts directly on the translation initiation complex, translational targets of Pdcd4 have yet to be identified. In order to identify and characterize functionally significant translational targets of tumor suppressor Pdcd4 when it inhibits translation during tumor promotion, tumorigenesis and invasion of malignant tumors, we knocked down Pdcd4 in T47D breast cancer cells. Loss of Pdcd4 increases cell migration and invasion as well as activity of luciferase reporters of AP-1-dependent transcription and translation initiation. To identify translational targets of Pdcd4, translationally active mRNA species were fractionated by sucrose gradient. Polyribosome bound mRNA fractions were pooled and analyzed for mRNA levels by microarray. Knockdown of Pdcd4 produced more than 2 fold increases in 14 mRNAs in the translationally active polyribosomal fractions compared to the control. The polysome shifts in Pdcd4 translational targets are being confirmed by quantitative RT-PCR and Western blot analysis of protein. Pdcd4 targets recently confirmed by quantitative RT-PCR are PPARGC1B and delta p73, both known to be oncogenic, as well as zinc finger protein ZNF281. The functional significance of these targets is being determined. Discovery of the mRNAs selectively targeted by Pdcd4 will facilitate the elucidation of the mechanism by which Pdcd4 suppresses tumorigenesis and may provide additional targets for intervention. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 3079. doi:10.1158/1538-7445.AM2011-3079


Cancer Research | 2010

Abstract 1481: Srx-Prx IV axis is required for human lung cancer cell maintenance, migration, and invasion

Qiou Wei; Hong Jiang; Zhen Xiao; Andrew G. Stephen; Alyson R. Baker; Matthew R. Young; Timothy D. Veenstra; Nancy H. Colburn

Sulfiredoxin (Srx) is a novel antioxidant protein that has been shown to reduce the hyper oxidized Peroxiredoxins (Prxs). Our previous study has demonstrated that Srx is required for tumor promoter induced cell transformation and is highly expressed in certain types of human skin cancers (Wei et al PNAS 2008). In this study, our goal is to further examine the expression of Srx in a broad range of human tumors and to explore its functional significance and molecular mechanisms in human cancer development. We found that Srx is highly expressed in several types of human lung cancers and its level is correlated with cancer malignancy and metastasis. Using human lung cancer A549 cell in culture, we found that knockdown of Srx significantly reduces anchorage independent growth in soft agar, cell migration in a wound-healing assay and cell invasiveness in a matrigel invasion assay. In an effort to discover downstream mediators of Srx-signaling pathway, we used affinity-based pull down and mass spectrometry methods to identify proteins that interact with Srx. To our surprise, Prx IV was found to be the most abundant protein that interacts with Srx in at least three different cell lines and the preferential interaction occurs independently of its expression levels. To characterize the Srx-Prx IV interaction, bacterial expressed recombinant Srx, Prx I, II, III and IV were individually purified. In an in vitro binding assay using Surface Plasmon Resonance technique, we confirmed that purified Srx has a much higher binding affinity to Prx IV than to Prx I, II or III. With a lentiviral based shRNA technology, each individual Prx was specifically knocked down in A549 cells. The phenotypic changes of these cells were compared to the Srx knockdown cells. We found that knockdown of Prx IV, but not other Prxs, was sufficient to recapitulate the attenuation of tumor phenotype and progression of Srx knockdown cells. In addition, we also confirmed that Prx IV undergoes rapid hyperoxidation in the presence of oxidative stress and is preferentially reduced by Srx in A549 cells. Overall our data indicate that Srx is highly expressed in human lung cancers and Srx-Prx IV axis is critical for human lung cancer cell maintenance, migration and invasion. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1481.

Collaboration


Dive into the Qiou Wei's collaboration.

Top Co-Authors

Avatar

Hong Jiang

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar

Nancy H. Colburn

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Matthew R. Young

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alyson R. Baker

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Lisha Wu

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Timothy D. Veenstra

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Zhen Xiao

Science Applications International Corporation

View shared research outputs
Researchain Logo
Decentralizing Knowledge