1
|
Ruiz CF, Ge X, McDonnell R, Agabiti SS, McQuaid DC, Tang A, Kharwa M, Goodell J, Saavedra-Peña RDM, Wing A, Li G, Medici NP, Robert ME, Varshney RR, Rudolph MC, Gorelick FS, Wysolmerski J, Canals D, Haley JD, Rodeheffer MS, Muzumdar MD. Diet-induced phospholipid remodeling dictates ferroptosis sensitivity and tumorigenesis in the pancreas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.645324. [PMID: 40235976 PMCID: PMC11996499 DOI: 10.1101/2025.04.04.645324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
High-fat diet (HFD) intake has been linked to an increased risk of pancreatic ductal adenocarcinoma (PDAC), a lethal and therapy-resistant cancer. However, whether and how specific dietary fats drive cancer development remains unresolved. Leveraging an oncogenic Kras -driven mouse model that closely mimics human PDAC progression, we screened a dozen isocaloric HFDs differing solely in fat source and representing the diversity of human fat consumption. Unexpectedly, diets rich in oleic acid - a monounsaturated fatty acid (MUFA) typically associated with good health - markedly enhanced tumorigenesis. Conversely, diets high in polyunsaturated fatty acids (PUFAs) suppressed tumor progression. Relative dietary fatty acid saturation levels (PUFA/MUFA) governed pancreatic membrane phospholipid composition, lipid peroxidation, and ferroptosis sensitivity in mice, concordant with circulating PUFA/MUFA levels being linked to altered PDAC risk in humans. These findings directly implicate dietary unsaturated fatty acids in controlling ferroptosis susceptibility and tumorigenesis, supporting potential "precision nutrition" strategies for PDAC prevention.
Collapse
|
2
|
Zhang DD. Thirty years of NRF2: advances and therapeutic challenges. Nat Rev Drug Discov 2025:10.1038/s41573-025-01145-0. [PMID: 40038406 DOI: 10.1038/s41573-025-01145-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2025] [Indexed: 03/06/2025]
Abstract
Over the last 30 years, NRF2 has evolved from being recognized as a transcription factor primarily involved in redox balance and detoxification to a well-appreciated master regulator of cellular proteostasis, metabolism and iron homeostasis. NRF2 plays a pivotal role in diverse pathologies, including cancer, and metabolic, inflammatory and neurodegenerative disorders. It exhibits a Janus-faced duality, safeguarding cellular integrity in normal cells against environmental insults to prevent disease onset, whereas in certain cancers, constitutively elevated NRF2 levels provide a tumour survival advantage, promoting progression, therapy resistance and metastasis. Advances in understanding the mechanistic regulation of NRF2 and its roles in human pathology have propelled the investigation of NRF2-targeted therapeutic strategies. This Review dissects the mechanistic intricacies of NRF2 signalling, its cross-talk with biological processes and its far-reaching implications for health and disease, highlighting key discoveries that have shaped innovative therapeutic approaches targeting NRF2.
Collapse
Affiliation(s)
- Donna D Zhang
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, FL, USA.
- University of Florida Health Cancer Center, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
3
|
Chaudhary S, Siddiqui JA, Pothuraju R, Bhatia R. Ribosome biogenesis, altered metabolism and ribotoxic stress response in pancreatic ductal adenocarcinoma tumor microenvironment. Cancer Lett 2025; 612:217484. [PMID: 39842499 DOI: 10.1016/j.canlet.2025.217484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy with a poor overall survival rate. Cellular stress response pathways promoting cancer cell fitness in harsh tumor microenvironment (TME) play a critical role in cancer growth and survival. The influence of oncogenic Kras, multi-functional heterogeneous cancer-associated fibroblasts (CAFs), and immunosuppressive TME on cancer cells makes the disease more complex and difficult to treat. The desmoplastic reaction by CAFs comprises approximately 90 % of the tumor, with only 10 % of cancer cells making things even more complicated, resulting in therapy resistance. Consistently increasing fibrosis creates a hypoxic environment and elevated interstitial fluid pressure inside the tumor constraining vascular supply. Stress conditions in TME alter translation efficiency and metabolism to fulfill the energy requirements of rapidly growing cancer cells. Extensive research has been conducted on multiple molecular and metabolic regulators in PDAC TME. However, the role of TME in influencing translation programs, a prerequisite for cell cycle progression and functional/growth requirements for cancer cells, remains elusive. This review highlights the recent advancements in understanding altered translational programs in PDAC TME. We emphasize the role of ribosome biogenesis, ribosome-induced stress response, and the concept of specialized ribosomes and their probable role in mutationally rewiring the pancreatic TME.
Collapse
Affiliation(s)
- Sanjib Chaudhary
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, 781039, Guwahati, Assam, India
| | - Jawed Akhtar Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA; Cancer Center Research Institute, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ramesh Pothuraju
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thiruvananthapuram, Kerala, India.
| | - Rakesh Bhatia
- Amity School of Biological Sciences, Amity University Punjab, 82A, Mohali, Punjab, 140306, India.
| |
Collapse
|
4
|
McGirr T, Onar O, Jafarnejad SM. Dysregulated ribosome quality control in human diseases. FEBS J 2025; 292:936-959. [PMID: 38949989 PMCID: PMC11880988 DOI: 10.1111/febs.17217] [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: 02/12/2024] [Revised: 05/31/2024] [Accepted: 06/20/2024] [Indexed: 07/03/2024]
Abstract
Precise regulation of mRNA translation is of fundamental importance for maintaining homeostasis. Conversely, dysregulated general or transcript-specific translation, as well as abnormal translation events, have been linked to a multitude of diseases. However, driven by the misconception that the transient nature of mRNAs renders their abnormalities inconsequential, the importance of mechanisms that monitor the quality and fidelity of the translation process has been largely overlooked. In recent years, there has been a dramatic shift in this paradigm, evidenced by several seminal discoveries on the role of a key mechanism in monitoring the quality of mRNA translation - namely, Ribosome Quality Control (RQC) - in the maintenance of homeostasis and the prevention of diseases. Here, we will review recent advances in the field and emphasize the biological significance of the RQC mechanism, particularly its implications in human diseases.
Collapse
Affiliation(s)
- Tom McGirr
- Patrick G. Johnston Centre for Cancer ResearchQueen's University BelfastUK
| | - Okan Onar
- Patrick G. Johnston Centre for Cancer ResearchQueen's University BelfastUK
- Department of Biology, Faculty of ScienceAnkara UniversityTurkey
| | | |
Collapse
|
5
|
Yang Z, Han T, Yang R, Zhang Y, Qin Y, Hou J, Huo F, Feng Z, Ding Y, Yang J, Zhou G, Wang S, Xie X, Lin P, Chen ZN, Wu J. Dicoumarol sensitizes hepatocellular carcinoma cells to ferroptosis induced by imidazole ketone erastin. Front Immunol 2025; 16:1531874. [PMID: 40007539 PMCID: PMC11852437 DOI: 10.3389/fimmu.2025.1531874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 01/13/2025] [Indexed: 02/27/2025] Open
Abstract
Introduction Ferroptosis, an iron-dependent form of regulated cell death, is characterized by the lethal accumulation of lipid peroxides on cellular membranes. It not only inhibits tumor growth but also enhances immunotherapy responses and overcomes drug resistance in cancer therapy. The inhibition of the cystine-glutamate antiporter, system Xc-, induces ferroptosis. Imidazole ketone erastin (IKE), an inhibitor of the system Xc- functional subunit solute carrier family 7 member 11 (SLC7A11), is an effective and metabolically stable inducer of ferroptosis with potential in vivo applications. However, tumor cells exhibited differential sensitivity to IKE-induced ferroptosis. The intrinsic factors determining sensitivity to IKE-induced ferroptosis remain to be explored to improve its efficacy. Methods Bulk RNA-sequencing data from hepatocellular carcinoma (HCC) and normal liver tissues were collected from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases. Differentially expressed genes were identified and intersected with the ferroptosis-related genes (FRGs) listed in the FerrDb database, yielding the identification of 13 distinct FRGs. Results A ferroptosis signature index model (Risk Score) was developed to predict HCC prognosis. And SLC7A11 and NAD(P)H quinone dehydrogenase 1 (NQO1) were identified as candidate FRGs indicating poor prognosis of HCC. Dicoumarol (DIC), an inhibitor of NQO1, was subsequently employed to assess its sensitizing effects on IKE in HCC treatment. In HCC cell lines and the subcutaneous xenograft model, the combined suppression of SLC7A11 and NQO1 significantly enhanced the inhibitory effect on tumor growth by inducing ferroptosis. Discussion In conclusion, our findings demonstrate that DIC sensitized HCC cells to IKE-induced ferroptosis in HCC. Moreover, the identification of potential drugs that enhance the susceptibility of HCC cells to ferroptosis could provide novel therapeutic strategies for the treatment of HCC.
Collapse
Affiliation(s)
- Ziwei Yang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Tixin Han
- Shaanxi Key Laboratory of Bio-electromagnetic Detection and Intelligent Sensing, Military Biomedical Engineering School, Fourth Military Medical University, Xi'an, China
| | - Ruibin Yang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yinuo Zhang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yifei Qin
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
- Institutes of Biomedicine and Department of Cell Biology, Jinan University, Guangzhou, China
| | - Jialu Hou
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Fei Huo
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Zhuan Feng
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yaxin Ding
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Jiali Yang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Gang Zhou
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Shijie Wang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Xiaohang Xie
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Peng Lin
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Zhi-Nan Chen
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Jiao Wu
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
6
|
Xing X, Li XQ, Yin SQ, Ma HT, Xiao SY, Tulamaiti A, Yang Y, Jiang SH, Hu LP, Zhang ZG, Huo YM, Li DX, Yang XM, Zhang XL. OASL promotes immune evasion in pancreatic ductal adenocarcinoma by enhancing autolysosome-mediated degradation of MHC-I. Theranostics 2025; 15:2104-2120. [PMID: 39990208 PMCID: PMC11840728 DOI: 10.7150/thno.103494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/28/2024] [Indexed: 02/25/2025] Open
Abstract
Rationale: Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with a global prevalence and poor prognosis, largely due to immune escape mechanisms. However, the potential reasons for the decreased infiltration of cytotoxic T lymphocytes (CTLs) in PDAC remain inadequately understood. In this study, we aimed to elucidate the molecular mechanisms contributing to the low-CTLs infiltration in patients with PDAC. Methods: Bioinformatic analyses were used to identify key factors associated with low-CTLs infiltration in PDAC and the role of oligoadenylate synthetase-like (OASL) was mainly focused in our study. Immunohistochemistry (IHC) was used to assess the relationship between the expression of OASL and the prognosis of patients. Western blotting, Flow cytometry, Co-immunoprecipitation and Immunofluorescence were applied to elucidate the molecular mechanism by which OASL mediates immune escape in PDAC. The orthotopic PDAC models were constructed to evaluate the effects of OASL-knockdown on CD8+ T cells infiltration and tumor growth in vivo. Results: OASL was found to be significantly upregulated in PDAC and negatively correlated with the major histocompatibility complex class I (MHC-I) expression, which is associated with worse patient prognosis. Notably, OASL-knockdown leads to a significant increase in CD8+ T cell infiltration and slows tumor growth in vivo. Mechanistic studies revealed that OASL -knockdown restored the total and surface MHC-I level through impairing neighbor of BRCA1 gene 1 (NBR1)-mediated autophagy-lysosomal degradation of MHC-I. Conclusions: Targeting OASL enhances the immune response in PDAC, providing a novel therapeutic strategy to improve outcomes in PDAC patients.
Collapse
Affiliation(s)
- Xin Xing
- Shanghai University of Medicine & Health Sciences affiliated Sixth People's Hospital South Campus, Shanghai, China
- Shanghai Fengxian District Central Hospital, School of Medicine, Anhui University of Science and Technology, Shanghai, China
| | - Xia-Qing Li
- Shanghai Fengxian District Central Hospital, School of Medicine, Anhui University of Science and Technology, Shanghai, China
| | - Shi-Qi Yin
- Shanghai Fengxian District Central Hospital, School of Medicine, Anhui University of Science and Technology, Shanghai, China
| | - Hong-Tai Ma
- Shanghai Fengxian District Central Hospital, School of Medicine, Anhui University of Science and Technology, Shanghai, China
| | - Shu-Yu Xiao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Aziguli Tulamaiti
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Shu-Heng Jiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Peng Hu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yan-Miao Huo
- Department of Biliary-Pancreatic Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Dong-Xue Li
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Mei Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xue-Li Zhang
- Shanghai University of Medicine & Health Sciences affiliated Sixth People's Hospital South Campus, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
7
|
Fan L, Guo D, Zhu C, Gao C, Wang Y, Yin F, Liu M, Zhou Y, Wei T, Xiong X, Yu K, Le A. LRRC45 accelerates bladder cancer development and ferroptosis inhibition via stabilizing NRF2 by competitively KEAP1 interaction. Free Radic Biol Med 2025; 226:29-42. [PMID: 39522565 DOI: 10.1016/j.freeradbiomed.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Centrosomal dysregulation is closely linked to the genesis and progression of tumors. A comprehensive analysis of single-cell RNA sequencing (scRNA-seq) data has revealed that leucine-rich repeat-containing protein 45 (LRRC45), a centrosome linker protein crucial for maintaining centrosome cohesion and a member of the leucine-rich repeat-containing proteins (LRRCs) family, is significantly upregulated in bladder cancer. Notably, the elevated expression levels of LRRC45 were strongly correlated with a poor prognosis in patients. Furthermore, the depletion of LRRC45 in bladder cancer cells markedly inhibited tumorigenic proliferation and increased intracellular iron and reactive oxygen species (ROS) levels. It ultimately triggered ferroptosis, an iron-dependent form of programmed cell death characterized by lipid peroxidation. Mechanistic studies revealed that LRRC45 exerts its oncogenic effects through competitive interaction with Kelch-like ECH-associated protein 1 (KEAP1), which inhibits the ubiquitin-proteasome-mediated degradation of nuclear factor erythroid 2-related factor 2 (NRF2). This interaction enhances the nuclear translocation of NRF2 and its subsequent anti-ferroptotic activity. In conclusion, our studies highlight the critical role of LRRC45 in enhancing the stability of NRF2, thereby promoting the tumorigenic potential of bladder cancer. These insights suggest that targeting LRRC45 could serve as a promising molecular target for developing novel therapeutic interventions for bladder cancer.
Collapse
Affiliation(s)
- Linwei Fan
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Dingfan Guo
- The First Clinical Medical School of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chao Zhu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Chenqi Gao
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yu Wang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Fang Yin
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Mengwei Liu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yanyu Zhou
- The First Clinical Medical School of Nanchang University, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Tiancheng Wei
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Xinxin Xiong
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| | - Kuai Yu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| | - Aiping Le
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
8
|
Zhang Y, Qi F, Chen P, Liu BF, Li Y. Spatially defined microenvironment for engineering organoids. BIOPHYSICS REVIEWS 2024; 5:041302. [PMID: 39679203 PMCID: PMC11646138 DOI: 10.1063/5.0198848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/01/2024] [Indexed: 12/17/2024]
Abstract
In the intricately defined spatial microenvironment, a single fertilized egg remarkably develops into a conserved and well-organized multicellular organism. This observation leads us to hypothesize that stem cells or other seed cell types have the potential to construct fully structured and functional tissues or organs, provided the spatial cues are appropriately configured. Current organoid technology, however, largely depends on spontaneous growth and self-organization, lacking systematic guided intervention. As a result, the structures replicated in vitro often emerge in a disordered and sparse manner during growth phases. Although existing organoids have made significant contributions in many aspects, such as advancing our understanding of development and pathogenesis, aiding personalized drug selection, as well as expediting drug development, their potential in creating large-scale implantable tissue or organ constructs, and constructing multicomponent microphysiological systems, together with functioning at metabolic levels remains underutilized. Recent discoveries have demonstrated that the spatial definition of growth factors not only induces directional growth and migration of organoids but also leads to the formation of assembloids with multiple regional identities. This opens new avenues for the innovative engineering of higher-order organoids. Concurrently, the spatial organization of other microenvironmental cues, such as physical stresses, mechanical loads, and material composition, has been minimally explored. This review delves into the burgeoning field of organoid engineering with a focus on potential spatial microenvironmental control. It offers insight into the molecular principles, expected outcomes, and potential applications, envisioning a future perspective in this domain.
Collapse
Affiliation(s)
- Yilan Zhang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fukang Qi
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
9
|
Wang T, Liu M, Li X, Zhang S, Gu H, Wei X, Wang X, Xu Z, Shen T. Naturally-derived modulators of the Nrf2 pathway and their roles in the intervention of diseases. Free Radic Biol Med 2024; 225:560-580. [PMID: 39368519 DOI: 10.1016/j.freeradbiomed.2024.09.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/07/2024]
Abstract
Cumulative evidence has verified that persistent oxidative stress is involved in the development of various chronic diseases, including pulmonary, neurodegenerative, kidney, cardiovascular, and liver diseases, as well as cancers. Nuclear factor erythroid 2-related factor 2 (Nrf2) plays a pivotal role in regulating cellular oxidative stress and inflammatory reactions, making it a focal point for disease prevention and treatment strategies. Natural products are essential resources for discovering leading molecules for new drug research and development. In this review, we comprehensively outlined the progression of the knowledge on the Nrf2 pathway, Nrf2 activators in clinical trials, the naturally-derived Nrf2 modulators (particularly from 2014-present), as well as their effects on the pathogenesis of chronic diseases.
Collapse
Affiliation(s)
- Tian Wang
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Mingjie Liu
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xinyu Li
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Sen Zhang
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Haoran Gu
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Xuan Wei
- Shandong Center for Food and Drug Evaluation and Inspection, Jinan, Shandong, PR China
| | - Xiaoning Wang
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China
| | - Zhenpeng Xu
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China.
| | - Tao Shen
- Key Lab of Chemical Biology (MOE), Shandong Engineering Research Center for Traditional Chinese Medicine Standard, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, PR China.
| |
Collapse
|
10
|
Hayashi M, Okazaki K, Papgiannakopoulos T, Motohashi H. The Complex Roles of Redox and Antioxidant Biology in Cancer. Cold Spring Harb Perspect Med 2024; 14:a041546. [PMID: 38772703 PMCID: PMC11529857 DOI: 10.1101/cshperspect.a041546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Redox reactions control fundamental biochemical processes, including energy production, metabolism, respiration, detoxification, and signal transduction. Cancer cells, due to their generally active metabolism for sustained proliferation, produce high levels of reactive oxygen species (ROS) compared to normal cells and are equipped with antioxidant defense systems to counteract the detrimental effects of ROS to maintain redox homeostasis. The KEAP1-NRF2 system plays a major role in sensing and regulating endogenous antioxidant defenses in both normal and cancer cells, creating a bivalent contribution of NRF2 to cancer prevention and therapy. Cancer cells hijack the NRF2-dependent antioxidant program and exploit a very unique metabolism as a trade-off for enhanced antioxidant capacity. This work provides an overview of redox metabolism in cancer cells, highlighting the role of the KEAP1-NRF2 system, selenoproteins, sulfur metabolism, heme/iron metabolism, and antioxidants. Finally, we describe therapeutic approaches that can be leveraged to target redox metabolism in cancer.
Collapse
Affiliation(s)
- Makiko Hayashi
- Department of Pathology, New York University School of Medicine, New York, New York 10016, USA
| | - Keito Okazaki
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | | | - Hozumi Motohashi
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
11
|
Swamynathan MM, Kuang S, Watrud KE, Doherty MR, Gineste C, Mathew G, Gong GQ, Cox H, Cheng E, Reiss D, Kendall J, Ghosh D, Reczek CR, Zhao X, Herzka T, Špokaitė S, Dessus AN, Kim ST, Klingbeil O, Liu J, Nowak DG, Alsudani H, Wee TL, Park Y, Minicozzi F, Rivera K, Almeida AS, Chang K, Chakrabarty RP, Wilkinson JE, Gimotty PA, Diermeier SD, Egeblad M, Vakoc CR, Locasale JW, Chandel NS, Janowitz T, Hicks JB, Wigler M, Pappin DJ, Williams RL, Cifani P, Tuveson DA, Laporte J, Trotman LC. Dietary pro-oxidant therapy by a vitamin K precursor targets PI 3-kinase VPS34 function. Science 2024; 386:eadk9167. [PMID: 39446948 PMCID: PMC11975464 DOI: 10.1126/science.adk9167] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 08/27/2024] [Indexed: 10/26/2024]
Abstract
Men taking antioxidant vitamin E supplements have increased prostate cancer (PC) risk. However, whether pro-oxidants protect from PC remained unclear. In this work, we show that a pro-oxidant vitamin K precursor [menadione sodium bisulfite (MSB)] suppresses PC progression in mice, killing cells through an oxidative cell death: MSB antagonizes the essential class III phosphatidylinositol (PI) 3-kinase VPS34-the regulator of endosome identity and sorting-through oxidation of key cysteines, pointing to a redox checkpoint in sorting. Testing MSB in a myotubular myopathy model that is driven by loss of MTM1-the phosphatase antagonist of VPS34-we show that dietary MSB improved muscle histology and function and extended life span. These findings enhance our understanding of pro-oxidant selectivity and show how definition of the pathways they impinge on can give rise to unexpected therapeutic opportunities.
Collapse
Affiliation(s)
- Manojit Mosur Swamynathan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Shan Kuang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | | | - Mary R. Doherty
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Charlotte Gineste
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, Inserm U1258, Strasbourg University, Illkirch CEDEX 67404, France
| | - Grinu Mathew
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- Eppley Institute, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Grace Q. Gong
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Hilary Cox
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Eileen Cheng
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - David Reiss
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, Inserm U1258, Strasbourg University, Illkirch CEDEX 67404, France
| | - Jude Kendall
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Diya Ghosh
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Colleen R. Reczek
- Department of Medicine, Biochemistry & Molecular Genetics, Northwestern University, Chicago, IL 60611, USA
| | - Xiang Zhao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Tali Herzka
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Saulė Špokaitė
- MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | | | - Seung Tea Kim
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Olaf Klingbeil
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Juan Liu
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh NC 27695
| | - Dawid G. Nowak
- Department of Medicine, Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, New York, NY 10065, USA
- Division of Hematology and Medical Oncology, Department of Medicine, New York Presbyterian Hospital, Weill Cornell Medicine, New York, New York, NY 10065, USA
| | - Habeeb Alsudani
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Tse-Luen Wee
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | | | - Keith Rivera
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Ana S. Almeida
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- APC Microbiome Ireland and School of Microbiology, University College Cork, Cork T12 K8AF, Ireland
| | - Kenneth Chang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Ram P. Chakrabarty
- Department of Medicine, Biochemistry & Molecular Genetics, Northwestern University, Chicago, IL 60611, USA
| | - John E. Wilkinson
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Phyllis A. Gimotty
- Perelman School of Medicine, Division of Biostatistics, University of Pennsylvania, PA 19104, USA
| | - Sarah D. Diermeier
- University of Otago, Department of Biochemistry, Dunedin 9016, New Zealand
| | - Mikala Egeblad
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- School of Medicine, Johns Hopkins University, Baltimore, MD 21205 USA
| | | | - Jason W. Locasale
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh NC 27695
| | - Navdeep S. Chandel
- Department of Medicine, Biochemistry & Molecular Genetics, Northwestern University, Chicago, IL 60611, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - James B. Hicks
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
- Michelson Center for Convergent Biosciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Michael Wigler
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Darryl J. Pappin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | | | - Paolo Cifani
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - David A. Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| | - Jocelyn Laporte
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, CNRS UMR7104, Inserm U1258, Strasbourg University, Illkirch CEDEX 67404, France
| | - Lloyd C. Trotman
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11771, USA
| |
Collapse
|
12
|
Chen L, Guo P, Li Z, Hu X, Wang D, Yu L, Zhu D, Tang H, Luo H. Transcription factor Nrf2 regulating the interaction of p16 facilitates hydroquinone-induced malignant transformation of TK6 cells by accelerating cell proliferation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 285:117142. [PMID: 39357381 DOI: 10.1016/j.ecoenv.2024.117142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/20/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
The nuclear factor erythroid 2-related factor 2 (Nrf2) is overexpressed in multiple tumor cells. Nevertheless, the role of Nrf2 in malignant transformation induced by hydroquinone (HQ) is unknown. Here, we hypothesized that Nrf2 might participate in HQ-induced malignant transformation of TK6 cells, a line of normal human lymphoblastoid cells, by accelerating cell proliferation and regulating cell cycle progression. The data indicated that TK6 cells chronically exposed to HQ continuously activated Nrf2-Keap1 signaling pathway. Furthermore, we found that defects in Nrf2 inhibited cell proliferation and prevented cells from entering S phase from G1 phase. Mechanistically, Nrf2 is involved in cell cycle abnormalities induced by prolonged exposure to HQ by binding to p16, thereby activating the p16/Rb signaling pathway. Taken together, Nrf2 might be a potential driver of carcinogenesis that promotes malignant cell proliferation and affects cell cycle distribution.
Collapse
Affiliation(s)
- Lin Chen
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Pu Guo
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Zhuanzhuan Li
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Xiaoyi Hu
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Dewang Wang
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Lingxue Yu
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Delong Zhu
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Huanwen Tang
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| | - Hao Luo
- Dongguan Key Laboratory of Environmental Medicine, The First Dongguan Affiliated Hospital, School of Public Health, Guangdong Medical University, Dongguan 523808, China.
| |
Collapse
|
13
|
Cascio G, Aguirre KN, Church KP, Hughes RO, Nease LA, Delclaux I, Davis HJ, Piskounova E. Transcriptional Isoforms of NAD + kinase regulate oxidative stress resistance and melanoma metastasis. Redox Biol 2024; 76:103289. [PMID: 39167913 PMCID: PMC11381905 DOI: 10.1016/j.redox.2024.103289] [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: 06/12/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Metastasizing cancer cells encounter a multitude of stresses throughout the metastatic cascade. Oxidative stress is known to be a major barrier for metastatic colonization, such that metastasizing cancer cells must rewire their metabolic pathways to increase their antioxidant capacity. NADPH is essential for regeneration of cellular antioxidants and several NADPH-regenerating pathways have been shown to play a role in metastasis. We have found that metastatic melanoma cells have increased levels of both NADPH and NADP+ suggesting increased de novo biosynthesis of NADP+. De novo biosynthesis of NADP+ occurs through a single enzymatic reaction catalyzed by NAD+ kinase (NADK). Here we show that different NADK isoforms are differentially expressed in metastatic melanoma cells, with Isoform 3 being specifically upregulated in metastasis. We find that Isoform 3 is more potent in expanding the NADP(H) pools, increasing oxidative stress resistance and promoting metastatic colonization compared to Isoform 1. We have found that Isoform 3 is transcriptionally upregulated by oxidative stress through the action of NRF2. Together, our work presents a previously uncharacterized role of NADK isoforms in oxidative stress resistance and metastasis and suggests that NADK Isoform 3 is a potential therapeutic target in metastatic disease.
Collapse
Affiliation(s)
- Graciela Cascio
- Sandra and Edward Meyer Cancer Center, 413 East 69th Street, Belfer Research Building, Weill Cornell Medicine, 10021, New York, NY, USA
| | - Kelsey N Aguirre
- Sandra and Edward Meyer Cancer Center, 413 East 69th Street, Belfer Research Building, Weill Cornell Medicine, 10021, New York, NY, USA
| | - Kellsey P Church
- Sandra and Edward Meyer Cancer Center, 413 East 69th Street, Belfer Research Building, Weill Cornell Medicine, 10021, New York, NY, USA
| | - Riley O Hughes
- Sandra and Edward Meyer Cancer Center, 413 East 69th Street, Belfer Research Building, Weill Cornell Medicine, 10021, New York, NY, USA; Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Leona A Nease
- Sandra and Edward Meyer Cancer Center, 413 East 69th Street, Belfer Research Building, Weill Cornell Medicine, 10021, New York, NY, USA
| | - Ines Delclaux
- Sandra and Edward Meyer Cancer Center, 413 East 69th Street, Belfer Research Building, Weill Cornell Medicine, 10021, New York, NY, USA
| | - Hannah J Davis
- Sandra and Edward Meyer Cancer Center, 413 East 69th Street, Belfer Research Building, Weill Cornell Medicine, 10021, New York, NY, USA
| | - Elena Piskounova
- Sandra and Edward Meyer Cancer Center, 413 East 69th Street, Belfer Research Building, Weill Cornell Medicine, 10021, New York, NY, USA; Department of Dermatology, Weill Cornell Medicine, New York, NY, USA; Department of Biochemistry, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Jin X, Lou X, Qi H, Zheng C, Li B, Siwu X, Liu R, Lv Q, Zhao A, Ruan J, Jiang M. NRF2 signaling plays an essential role in cancer progression through the NRF2-GPX2-NOTCH3 axis in head and neck squamous cell carcinoma. Oncogenesis 2024; 13:35. [PMID: 39333079 PMCID: PMC11437035 DOI: 10.1038/s41389-024-00536-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
The activation of nuclear factor erythroid 2-related factor 2 (NRF2) has been observed in various cancers. Yet its exact contribution to the development of head and neck squamous cell carcinoma (HNSCC) remains undetermined. We previously found that NRF2 signaling is critical for the differentiation of squamous basal progenitor cells, while disruption of NRF2 causes basal cell hyperplasia. In this study, we revealed a correlation between elevated NRF2 activity and poor outcomes in HNSCC patients. We demonstrated that NRF2 facilitates tumor proliferation, migration, and invasion, as evidenced by both in vitro and in vivo studies. Significantly, NRF2 augments the expression of the antioxidant enzyme GPX2, thereby enhancing the proliferative, migratory, and invasive properties of HNSCC cells. Activation of GPX2 is critical for sustaining cancer stem cells (CSCs) by up-regulating NOTCH3, a key driver of cancer progression. These results elucidate that NRF2 regulates HNSCC progression through the NRF2-GPX2-NOTCH3 axis. Our findings proposed that pharmacological targeting of the NRF2-GPX2-NOTCH3 axis could be a potential therapeutic approach against HNSCC.
Collapse
Affiliation(s)
- Xiaoye Jin
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Xiayuan Lou
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Haoxiang Qi
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, China
| | - Chao Zheng
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Bo Li
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Xuerong Siwu
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Ren Liu
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China
| | - Qiaoli Lv
- Institute of Cancer Research, Jiangxi Cancer Hospital, Nanchang, China
| | - An Zhao
- Institute of Cancer Research, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jian Ruan
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University and Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou, China
| | - Ming Jiang
- Center for Genetic Medicine, the Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Genetics, Zhejiang University International School of Medicine, Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Hangzhou, China.
| |
Collapse
|
15
|
Zhang X, He Y, Shen J, Zhou B, Qin H, Zhang S, Huang Z. Study on the Mechanism of FOXA2 Activation on Glutathione Metabolic Reprogramming Mediated by ETV4 Transcription to Facilitate Colorectal Cancer Malignant Progression. Biochem Genet 2024:10.1007/s10528-024-10918-y. [PMID: 39316306 DOI: 10.1007/s10528-024-10918-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 09/13/2024] [Indexed: 09/25/2024]
Abstract
The metabolic imbalance of glutathione (GSH) has been widely recognized in most cancers, but the specific molecular mechanism of GSH metabolic regulation in the malignant progression of colorectal cancer (CRC) is unexplored. The objective of our project is to elucidate whether ETV4 affects the malignant progression of CRC through GSH metabolic reprogramming. Bioinformatics and molecular experiments measured the expression of ETV4 in CRC, and in vitro experiments explored the impact of ETV4 on CRC malignant progression. The Kyoto Encyclopedia of Genes and Genomes (KEGG) identified the pathway of ETV4 enrichment. The bioinformatics approach identified FOXA2 as an upstream regulatory factor of ETV4. The dual-luciferase assay, chromatin immunoprecipitation (ChIP) and co-immunoprecipitation (Co-IP) experiment verified the binding relationship between ETV4 and FOXA2. Cell viability, migration, and invasion abilities were determined by conducting CCK-8, wound healing, and Transwell assays, respectively. The expression levels of N-cadherin, E-cadherin, and vimentin were determined by utilizing immunofluorescence (IF). Metabolism-related enzymes GCLM, GCLC, and GSTP1 levels were detected to evaluate the GSH metabolism level by analyzing the GSH/GSSG ratio. In vivo experiments were performed to explore the effect of FOXA2/ETV4 on CRC progression, and the expression of related proteins was detected by western blot. ETV4 was highly expressed in CRC. Knocking down ETV4 suppressed CRC cell viability, migration, invasion, and epithelial-mesenchymal transition (EMT) progression in vitro. ETV4 was abundant in the GSH metabolic pathway, and overexpression of ETV4 facilitated CRC malignant progression through activation of the GSH metabolism. In addition, in vitro cellular experiments and in vivo experiments in nude mice confirmed that FOXA2 transcriptionally activated ETV4. Knocking down FOXA2 repressed the malignant phenotype of CRC cells by suppressing GSH metabolism. These effects were reversed by overexpressing ETV4. Our results indicated that FOXA2 transcriptionally activates ETV4 to facilitate CRC malignant progression by modulating the GSH metabolic pathway. Targeting the FOXA2/ETV4 axis or GSH metabolism may be an effective approach for CRC treatment.
Collapse
Affiliation(s)
- Xiangcheng Zhang
- Department of General Surgery, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, 3 Foziling Road, Qingxiu District, Nanning, 530021, China.
| | - Yali He
- Department of Critical Care Medicine, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, Nanning, China
| | - Jiayue Shen
- Department of General Surgery, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, 3 Foziling Road, Qingxiu District, Nanning, 530021, China
| | - Bingchuan Zhou
- Department of General Surgery, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, 3 Foziling Road, Qingxiu District, Nanning, 530021, China
| | - Huabo Qin
- Department of General Surgery, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, 3 Foziling Road, Qingxiu District, Nanning, 530021, China
| | - Shuai Zhang
- Department of General Surgery, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, 3 Foziling Road, Qingxiu District, Nanning, 530021, China
| | - Zixiang Huang
- Department of General Surgery, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-Sen University, 3 Foziling Road, Qingxiu District, Nanning, 530021, China
| |
Collapse
|
16
|
Leal AS, Liby KT. The BRD4 Inhibitor I-BET-762 Reduces HO-1 Expression in Macrophages and the Pancreas of Mice. Int J Mol Sci 2024; 25:9985. [PMID: 39337472 PMCID: PMC11432103 DOI: 10.3390/ijms25189985] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
In pancreatic cancer, the tumor microenvironment (TME) accounts for up to 90% of the tumor mass. Pancreatitis, characterized by the increased infiltration of macrophages into the pancreas, is a known risk factor for pancreatic cancer. The NRF2 (nuclear factor erythroid 2-related factor 2) transcription factor regulates responses to oxidative stress and can promote cancer and chemoresistance. NRF2 also attenuates inflammation through the regulation of macrophage-specific genes. Heme oxygenase 1 (HO-1) is expressed by anti-inflammatory macrophages to degrade heme, and its expression is dependent on NRF2 translocation to the nucleus. In macrophages stimulated with conditioned media from pancreatic cancer cells, HO-1 protein levels increased, which correlated with higher NRF2 expression in the nuclear fraction. Significant differences in macrophage infiltration and HO-1 expression were detected in LSL-KrasG12D/+; Pdx-1-Cre (KC) mice, Nrf2 whole-body knockout (KO) mice and wildtype mice with pancreatitis. Since epigenetic modulation is a mechanism used by tumors to regulate the TME, using small molecules as epigenetic modulators to activate immune recognition is therapeutically desirable. When the bromodomain inhibitor I-BET-762 was used to treat macrophages or mice with pancreatitis, high levels of HO-1 were reduced. This study shows that bromodomain inhibitors can be used to prevent physiological responses to inflammation that promote tumorigenesis.
Collapse
Affiliation(s)
- Ana S. Leal
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Karen T. Liby
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
17
|
Harary PM, Hori YS, Persad ARL, Tayag A, Ustrzynski L, Emrich SC, Rahimy E, Park DJ, Li G, Chang SD. KEAP1-mutant atypical meningioma: illustrative case. JOURNAL OF NEUROSURGERY. CASE LESSONS 2024; 8:CASE24387. [PMID: 39250830 PMCID: PMC11404106 DOI: 10.3171/case24387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/19/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND While genetic testing of tumors is commonly used to inform the selection of systemic therapies, there is limited evidence for the application of radiotherapy for brain cancer. Recent studies have shown that Kelch-like ECH-associated protein 1 (KEAP1), a key regulator of cellular responses to oxidative and electrophilic stress, is associated with radioresistance in multiple cancer types. Several studies have reported the clinical significance of KEAP1 mutation in brain metastasis; however, the effect of KEAP1 mutations on radioresponse in meningioma has never been reported. OBSERVATIONS The authors present the case of a 40-year-old female with a KEAP1 mutation-positive atypical meningioma that was initially treated with resection followed by intensity-modulated radiation therapy (IMRT). Recurrence was observed at 15 months, requiring reoperation and adjuvant stereotactic radiosurgery (SRS). An excellent treatment response was observed at 7 months post-SRS with an improvement in reported symptoms, although bevacizumab was required for the resolution of radiation necrosis observed 2 months post-SRS. LESSONS To the authors' knowledge, this is the first report of KEAP1-mutant meningioma, including its clinical course after comprehensive management. Notably, treatment included multimodal radiotherapy with IMRT followed by SRS. SRS led to an excellent treatment response at the 7-month follow-up. However, radiation necrosis developed after both radiotherapy treatments, suggesting that radiological modification can be beneficial in patients with KEAP1 mutations. https://thejns.org/doi/10.3171/CASE24387.
Collapse
Affiliation(s)
- Paul M Harary
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Yusuke S Hori
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Amit R L Persad
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Armine Tayag
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Louisa Ustrzynski
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Sara C Emrich
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Elham Rahimy
- Radiation Oncology, Stanford University School of Medicine, Stanford, California
| | - David J Park
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Gordon Li
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| | - Steven D Chang
- Departments of Neurosurgery, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
18
|
Gan L, Wang W, Jiang J, Tian K, Liu W, Cao Z. Dual role of Nrf2 signaling in hepatocellular carcinoma: promoting development, immune evasion, and therapeutic challenges. Front Immunol 2024; 15:1429836. [PMID: 39286246 PMCID: PMC11402828 DOI: 10.3389/fimmu.2024.1429836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the predominant form of liver cancer and ranks as the third leading cause of cancer-related mortality globally. The liver performs a wide range of tasks and is the primary organ responsible for metabolizing harmful substances and foreign compounds. Oxidative stress has a crucial role in growth and improvement of hepatocellular carcinoma (HCC). Nuclear factor erythroid 2 (1)-related factor 2 (Nrf2) is an element that regulates transcription located in the cytoplasm. It controls the balance of redox reactions by stimulating the expression of many genes that depend on antioxidant response elements. Nrf2 has contrasting functions in the normal, healthy liver and HCC. In the normal liver, Nrf2 provides advantageous benefits, while in HCC it promotes harmful effects that support the growth and survival of HCC. Continuous activation of Nrf2 has been detected in HCC and promotes its advancement and aggressiveness. In addition, Activation of Nrf2 may lead to immune evasion, weakening the immune cells' ability to attack tumors and thereby promoting tumor development. Furthermore, chemoresistance in HCC, which is considered a form of stress response to chemotherapy medications, significantly impedes the effectiveness of HCC treatment. Stress management is typically accomplished by activating specific signal pathways and chemical variables. One important element in the creation of chemoresistance in HCC is nuclear factor-E2-related factor 2 (Nrf2). Nrf2 is a transcription factor that regulates the activation and production of a group of genes that encode proteins responsible for protecting cells from damage. This occurs through the Nrf2/ARE pathway, which is a crucial mechanism for combating oxidative stress within cells.
Collapse
Affiliation(s)
- Lin Gan
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Wei Wang
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Jinxiu Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Ke Tian
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Wei Liu
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Zhumin Cao
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| |
Collapse
|
19
|
Riazi-Tabrizi N, Khalaj-Kondori M, Safaei S, Amini M, Hassanian H, Maghsoudi M, Hasani S, Baradaran B. NRF2 Suppression Enhances the Susceptibility of Pancreatic Cancer Cells, Miapaca-2 to Paclitaxel. Mol Biotechnol 2024; 66:2441-2454. [PMID: 37740817 DOI: 10.1007/s12033-023-00872-2] [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: 02/09/2023] [Accepted: 09/04/2023] [Indexed: 09/25/2023]
Abstract
Pancreatic cancer is one of the most deadly diseases, with a very high metastasis and low survival rate. High levels of NRF2 have been detected in numerous malignancies, including head, neck, lung, and colon cancers, promoting the expansion and survival of cancer cells and chemical resistance to stressful conditions and affecting the response to treatment. To evaluate the possibility that modulation of NRF2 expression could be effective in treating pancreatic cancer cells, we explored the effect of knockdown of the NRF2 gene by NRF2-specific siRNA and its influence in combination with paclitaxel on pancreatic cancer cells. Miapaca-2 cell line, due to the high expression of the NRF2 gene, was selected for this study. Then, Miapaca-2 cells in different groups were treated with NRF2 siRNA and paclitaxel separately and in combination. After that, cell viability was measured by MTT assay and apoptosis induction by Annexin V-FITC/PI staining test. Cell cycle and autophagy were examined by flow cytometry, and cell migration was assessed by wound-healing assay. Finally, the expression of genes involved in apoptosis, Bax, Caspase-3, Caspase-9, and genes related to migration pathway, MMP-2, and MMP-9 in different groups were measured using qRT-PCR. Combined use of NRF2-specific siRNA with paclitaxel significantly reduced NRF2 gene expression in pancreatic cancer cells. NRF2 siRNA transfection significantly reduced cell viability. In addition, paclitaxel combination therapy with NRF2 siRNA strengthens the anti-tumor effects, such as inhibiting cell migration and provoking apoptosis, and autophagy and the cell cycle arrest in the G2 phase. NRF2 suppression augmented the expression of Bax, Caspase-3, and Caspase-9 genes and lowered the expression of Bcl-2, MMP-2, and MMP-9 genes, which play crucial roles in the pathways of apoptosis and cell migration, respectively. NRF2 siRNA enhances the susceptibility of Miapaca-2 cells to paclitaxel in pancreatic cancer cells. Thereby, suppressing NRF2 in combination with paclitaxel can be a new and efficacious treatment approach in treating pancreatic cancer.
Collapse
Affiliation(s)
- Negin Riazi-Tabrizi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamidreza Hassanian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohadeseh Maghsoudi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Shima Hasani
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
20
|
Martinez S, Wu S, Geuenich M, Malik A, Weber R, Woo T, Zhang A, Jang GH, Dervovic D, Al-Zahrani KN, Tsai R, Fodil N, Gros P, Gallinger S, Neely GG, Notta F, Sendoel A, Campbell K, Elling U, Schramek D. In vivo CRISPR screens reveal SCAF1 and USP15 as drivers of pancreatic cancer. Nat Commun 2024; 15:5266. [PMID: 38902237 PMCID: PMC11189927 DOI: 10.1038/s41467-024-49450-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 06/05/2024] [Indexed: 06/22/2024] Open
Abstract
Functionally characterizing the genetic alterations that drive pancreatic cancer is a prerequisite for precision medicine. Here, we perform somatic CRISPR/Cas9 mutagenesis screens to assess the transforming potential of 125 recurrently mutated pancreatic cancer genes, which revealed USP15 and SCAF1 as pancreatic tumor suppressors. Mechanistically, we find that USP15 functions in a haploinsufficient manner and that loss of USP15 or SCAF1 leads to reduced inflammatory TNFα, TGF-β and IL6 responses and increased sensitivity to PARP inhibition and Gemcitabine. Furthermore, we find that loss of SCAF1 leads to the formation of a truncated, inactive USP15 isoform at the expense of full-length USP15, functionally coupling SCAF1 and USP15. Notably, USP15 and SCAF1 alterations are observed in 31% of pancreatic cancer patients. Our results highlight the utility of in vivo CRISPR screens to integrate human cancer genomics and mouse modeling for the discovery of cancer driver genes with potential prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Sebastien Martinez
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Shifei Wu
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Michael Geuenich
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ahmad Malik
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ramona Weber
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Tristan Woo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Amy Zhang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Gun Ho Jang
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Dzana Dervovic
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Khalid N Al-Zahrani
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Ricky Tsai
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Nassima Fodil
- Department of Biochemistry, Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Philippe Gros
- Department of Biochemistry, Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Steven Gallinger
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - G Gregory Neely
- Dr. John and Anne Chong Lab for Functional Genomics, Charles Perkins Centre, and School of Life and Environmental Sciences, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Faiyaz Notta
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- PanCuRx Translational Research Initiative, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Ataman Sendoel
- Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Kieran Campbell
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Ulrich Elling
- Institute of Molecular Biotechnology of the Austrian Academy of Science (IMBA), Dr. Bohr-Gasse 3, Vienna BioCenter (VBC), 1030, Vienna, Austria
| | - Daniel Schramek
- Centre for Molecular and Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
21
|
Galletti G, Halima A, Gjyrezi A, Zhang J, Zimmerman B, Worroll D, Kallergi G, Barreja R, Ocean A, Saxena A, McGraw TE, Nanus DM, Elemento O, Altorki NK, Tagawa ST, Giannakakou P. Transferrin receptor-based circulating tumor cell enrichment provides a snapshot of the molecular landscape of solid tumors and correlates with clinical outcomes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.16.24309003. [PMID: 38947080 PMCID: PMC11213041 DOI: 10.1101/2024.06.16.24309003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Circulating tumor cells (CTCs) captured from the bloodstream of patients with solid tumors have the potential to accelerate precision oncology by providing insight into tumor biology, disease progression and response to treatment. However, their potential is hampered by the lack of standardized CTC enrichment platforms across tumor types. EpCAM-based CTC enrichment, the most commonly used platform, is limited by EpCAM downregulation during metastasis and the low EpCAM expression in certain tumor types, including the highly prevalent and lethal NSCLC. In this study we demonstrate that Transferrin Receptor (TfR) is a selective, efficient biomarker for CTC identification and capture in patients with prostate, pancreatic and NSCLC. TfR identifies significantly higher CTC counts than EpCAM, and TfR + -CTC enumeration correlates with disease progression in metastatic prostate and pancreatic cancers, and overall survival and osimetrinib-resistance in non-small cell lung cancer (NSCLC). Profiling of TfR + -CTCs provides a snapshot of the molecular landscape of each respective tumor type and identifies potential mechanisms underlying treatment response to EGFR TKi and immune checkpoint inhibitors in NSCLC. One sentence summary Transferrin Receptor identifies circulating tumor cells in solid tumors.
Collapse
|
22
|
Baiskhanova D, Schäfer H. The Role of Nrf2 in the Regulation of Mitochondrial Function and Ferroptosis in Pancreatic Cancer. Antioxidants (Basel) 2024; 13:696. [PMID: 38929135 PMCID: PMC11201043 DOI: 10.3390/antiox13060696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) represents the master regulator of the cellular antioxidant response and plays a critical role in tumorigenesis. This includes a preventive effect of Nrf2 on cell death through ferroptosis, which represents an essential mechanism of therapy resistance in malignant tumors, such as pancreatic ductal adenocarcinoma (PDAC) as one of the most aggressive and still incurable tumors. Addressing this issue, we provide an overview on Nrf2 mediated antioxidant response with particular emphasis on its effect on mitochondria as the organelle responsible for the execution of ferroptosis. We further outline how deregulated Nrf2 adds to the progression and therapy resistance of PDAC, especially with respect to the role of ferroptosis in anti-cancer drug mediated cell killing and how this is impaired by Nrf2 as an essential mechanism of drug resistance. Our review further discusses recent approaches for Nrf2 inhibition by natural and synthetic compounds to overcome drug resistance based on enhanced ferroptosis. Finally, we provide an outlook on therapeutic strategies based on Nrf2 inhibition combined with ferroptosis inducing drugs.
Collapse
Affiliation(s)
- Dinara Baiskhanova
- Laboratory of Molecular Gastroenterology and Tumor Biology, Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany;
| | | |
Collapse
|
23
|
Ross AB, Gorhe D, Kim JK, Hodapp S, DeVine L, Chan KM, Chio IIC, Jovanovic M, Ayres Pereira M. Systematic analysis of proteome turnover in an organoid model of pancreatic cancer by dSILO. CELL REPORTS METHODS 2024; 4:100760. [PMID: 38677284 PMCID: PMC11133751 DOI: 10.1016/j.crmeth.2024.100760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/26/2024] [Accepted: 03/25/2024] [Indexed: 04/29/2024]
Abstract
The role of protein turnover in pancreatic ductal adenocarcinoma (PDA) metastasis has not been previously investigated. We introduce dynamic stable-isotope labeling of organoids (dSILO): a dynamic SILAC derivative that combines a pulse of isotopically labeled amino acids with isobaric tandem mass-tag (TMT) labeling to measure proteome-wide protein turnover rates in organoids. We applied it to a PDA model and discovered that metastatic organoids exhibit an accelerated global proteome turnover compared to primary tumor organoids. Globally, most turnover changes are not reflected at the level of protein abundance. Interestingly, the group of proteins that show the highest turnover increase in metastatic PDA compared to tumor is involved in mitochondrial respiration. This indicates that metastatic PDA may adopt alternative respiratory chain functionality that is controlled by the rate at which proteins are turned over. Collectively, our analysis of proteome turnover in PDA organoids offers insights into the mechanisms underlying PDA metastasis.
Collapse
Affiliation(s)
- Alison B Ross
- Department of Biological Sciences, Columbia University, New York City, NY 10027, USA
| | - Darvesh Gorhe
- Department of Biological Sciences, Columbia University, New York City, NY 10027, USA
| | - Jenny Kim Kim
- Department of Biological Sciences, Columbia University, New York City, NY 10027, USA
| | - Stefanie Hodapp
- Department of Biological Sciences, Columbia University, New York City, NY 10027, USA
| | - Lela DeVine
- Department of Biology, Barnard College, New York, NY 10027, USA; Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Karina M Chan
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Iok In Christine Chio
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA.
| | - Marko Jovanovic
- Department of Biological Sciences, Columbia University, New York City, NY 10027, USA.
| | - Marina Ayres Pereira
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
24
|
Hu S, Yuan Y, Yan R, Xie H, Cai R, Xu J. NRF2 regulates EGF stability through OTUD4 in lung adenocarcinoma. Biochem Biophys Res Commun 2024; 708:149798. [PMID: 38518721 DOI: 10.1016/j.bbrc.2024.149798] [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: 02/29/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024]
Abstract
NRF2 (NFE2L2) is a transcription factor mainly for regulating cellular antioxidant response and therefore promotes tumor progression. The target genes of NRF2 also play important roles in cellular processes including glucose metabolism, de novo serine synthesis, iron metabolism, etc. Here, by modulating NRF2 expression in lung adenocarcinoma (LUAD) cells, we showed that NRF2 regulated EGF expression at protein level. Furthermore, EGF was identified as a ubiquitinated protein. We predicted three deubiquitinases of EGF, and OTUD4 had the highest correlation with NRF2 in LUAD among the three. OTUD4 expression was reduced upon NRF2 knocking-down and recovered upon NRF2 rescuing in A549 cells. Then a potential binding site for NRF2 in OTUD4 promoter was searched out. By binding with OTUD4 promoter, NRF2 transcriptionally activated OTUD4, thus promoted EGF deubiquitination and enhanced its stability. More importantly, OTUD4 and NRF2 expression was found being correlated in LUAD patients. The data collectively revealed a novel mechanism of NRF2 regulating on EGF stability through OTUD4 in LUAD.
Collapse
Affiliation(s)
- Shuming Hu
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Yuan
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Ruihan Yan
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Rong Cai
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jiaqian Xu
- Department of Biochemistry & Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai Institute of Immunology, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.
| |
Collapse
|
25
|
Gu M, Liu Y, Xin P, Guo W, Zhao Z, Yang X, Ma R, Jiao T, Zheng W. Fundamental insights and molecular interactions in pancreatic cancer: Pathways to therapeutic approaches. Cancer Lett 2024; 588:216738. [PMID: 38401887 DOI: 10.1016/j.canlet.2024.216738] [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: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/18/2024] [Indexed: 02/26/2024]
Abstract
The gastrointestinal tract can be affected by a number of diseases that pancreatic cancer (PC) is a malignant manifestation of them. The prognosis of PC patients is unfavorable and because of their diagnosis at advanced stage, the treatment of this tumor is problematic. Owing to low survival rate, there is much interest towards understanding the molecular profile of PC in an attempt in developing more effective therapeutics. The conventional therapeutics for PC include surgery, chemotherapy and radiotherapy as well as emerging immunotherapy. However, PC is still incurable and more effort should be performed. The molecular landscape of PC is an underlying factor involved in increase in progression of tumor cells. In the presence review, the newest advances in understanding the molecular and biological events in PC are discussed. The dysregulation of molecular pathways including AMPK, MAPK, STAT3, Wnt/β-catenin and non-coding RNA transcripts has been suggested as a factor in development of tumorigenesis in PC. Moreover, cell death mechanisms such as apoptosis, autophagy, ferroptosis and necroptosis demonstrate abnormal levels. The EMT and glycolysis in PC cells enhance to ensure their metastasis and proliferation. Furthermore, such abnormal changes have been used to develop corresponding pharmacological and nanotechnological therapeutics for PC.
Collapse
Affiliation(s)
- Ming Gu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Yang Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Peng Xin
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Zimo Zhao
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Xu Yang
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Ruiyang Ma
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Taiwei Jiao
- Department of Gastroenterology and Endoscopy, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Wenhui Zheng
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
26
|
Pillai R, LeBoeuf SE, Hao Y, New C, Blum JLE, Rashidfarrokhi A, Huang SM, Bahamon C, Wu WL, Karadal-Ferrena B, Herrera A, Ivanova E, Cross M, Bossowski JP, Ding H, Hayashi M, Rajalingam S, Karakousi T, Sayin VI, Khanna KM, Wong KK, Wild R, Tsirigos A, Poirier JT, Rudin CM, Davidson SM, Koralov SB, Papagiannakopoulos T. Glutamine antagonist DRP-104 suppresses tumor growth and enhances response to checkpoint blockade in KEAP1 mutant lung cancer. SCIENCE ADVANCES 2024; 10:eadm9859. [PMID: 38536921 PMCID: PMC10971495 DOI: 10.1126/sciadv.adm9859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024]
Abstract
Loss-of-function mutations in KEAP1 frequently occur in lung cancer and are associated with poor prognosis and resistance to standard of care treatment, highlighting the need for the development of targeted therapies. We previously showed that KEAP1 mutant tumors consume glutamine to support the metabolic rewiring associated with NRF2-dependent antioxidant production. Here, using preclinical patient-derived xenograft models and antigenic orthotopic lung cancer models, we show that the glutamine antagonist prodrug DRP-104 impairs the growth of KEAP1 mutant tumors. We find that DRP-104 suppresses KEAP1 mutant tumors by inhibiting glutamine-dependent nucleotide synthesis and promoting antitumor T cell responses. Using multimodal single-cell sequencing and ex vivo functional assays, we demonstrate that DRP-104 reverses T cell exhaustion, decreases Tregs, and enhances the function of CD4 and CD8 T cells, culminating in an improved response to anti-PD1 therapy. Our preclinical findings provide compelling evidence that DRP-104, currently in clinical trials, offers a promising therapeutic approach for treating patients with KEAP1 mutant lung cancer.
Collapse
Affiliation(s)
- Ray Pillai
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, VA New York Harbor Healthcare System, New York, NY 10016, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sarah E. LeBoeuf
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Yuan Hao
- Applied Bioinformatics Laboratories, New York University Langone Health, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Connie New
- Departments of Biological Engineering and Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jenna L. E. Blum
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ali Rashidfarrokhi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Shih Ming Huang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Christian Bahamon
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Warren L. Wu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Burcu Karadal-Ferrena
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Alberto Herrera
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Ellie Ivanova
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Michael Cross
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jozef P. Bossowski
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Hongyu Ding
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Makiko Hayashi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Sahith Rajalingam
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Triantafyllia Karakousi
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Volkan I. Sayin
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Center for Cancer Research, University of Gothenburg, 41345 Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Kamal M. Khanna
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
- Department of Microbiology, New York University Langone Health, New York, NY 10016, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Robert Wild
- Dracen Pharmaceuticals Inc., San Diego, CA 92121, USA
| | - Aristotelis Tsirigos
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - John T. Poirier
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Charles M. Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY 10655, USA
| | - Shawn M. Davidson
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ 08901, USA
| | - Sergei B. Koralov
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY 10016, USA
| |
Collapse
|
27
|
Ali MY, Bar-Peled L. Chemical proteomics to study metabolism, a reductionist approach applied at the systems level. Cell Chem Biol 2024; 31:446-451. [PMID: 38518745 DOI: 10.1016/j.chembiol.2024.02.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/02/2023] [Accepted: 02/28/2024] [Indexed: 03/24/2024]
Abstract
Cellular metabolism encompasses a complex array of interconnected biochemical pathways that are required for cellular homeostasis. When dysregulated, metabolism underlies multiple human pathologies. At the heart of metabolic networks are enzymes that have been historically studied through a reductionist lens, and more recently, using high throughput approaches including genomics and proteomics. Merging these two divergent viewpoints are chemical proteomic technologies, including activity-based protein profiling, which combines chemical probes specific to distinct enzyme families or amino acid residues with proteomic analysis. This enables the study of metabolism at the network level with the precision of powerful biochemical approaches. Herein, we provide a primer on how chemical proteomic technologies custom-built for studying metabolism have unearthed fundamental principles in metabolic control. In parallel, these technologies have leap-frogged drug discovery through identification of novel targets and drug specificity. Collectively, chemical proteomics technologies appear to do the impossible: uniting systematic analysis with a reductionist approach.
Collapse
Affiliation(s)
- Md Yousuf Ali
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Liron Bar-Peled
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
28
|
Döppler HR, Storz P. Macrophage-induced reactive oxygen species in the initiation of pancreatic cancer: a mini-review. Front Immunol 2024; 15:1278807. [PMID: 38576613 PMCID: PMC10991718 DOI: 10.3389/fimmu.2024.1278807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 03/12/2024] [Indexed: 04/06/2024] Open
Abstract
Pancreatic inflammation is a risk factor for the development of pancreatic cancer. Increased presence of inflammatory macrophages can be found in response to a KRAS mutation in acinar cells or in response to experimentally-induced pancreatitis. Inflammatory macrophages induce pancreatic acinar cells to undergo dedifferentiation to a duct-like progenitor stage, a process called acinar-to-ductal metaplasia (ADM). Occurrence of ADM lesions are believed to be the initiating event in tumorigenesis. Here we will discuss how macrophage-induced oxidative stress contributes to ADM and how ADM cells shape the fibrotic stroma needed for further progression.
Collapse
Affiliation(s)
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
29
|
Trifylli EM, Kriebardis AG, Koustas E, Papadopoulos N, Fortis SP, Tzounakas VL, Anastasiadi AT, Sarantis P, Vasileiadi S, Tsagarakis A, Aloizos G, Manolakopoulos S, Deutsch M. A Current Synopsis of the Emerging Role of Extracellular Vesicles and Micro-RNAs in Pancreatic Cancer: A Forward-Looking Plan for Diagnosis and Treatment. Int J Mol Sci 2024; 25:3406. [PMID: 38542378 PMCID: PMC10969997 DOI: 10.3390/ijms25063406] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 12/25/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest malignancies worldwide, while it persists as the fourth most prevalent cause of cancer-related death in the United States of America. Although there are several novel therapeutic strategies for the approach of this intensely aggressive tumor, it remains a clinical challenge, as it is hard to identify in early stages, due to its asymptomatic course. A diagnosis is usually established when the disease is already in its late stages, while its chemoresistance constitutes an obstacle to the optimal management of this malignancy. The discovery of novel diagnostic and therapeutic tools is considered a necessity for this tumor, due to its low survival rates and treatment failures. One of the most extensively investigated potential diagnostic and therapeutic modalities is extracellular vesicles (EVs). These vesicles constitute nanosized double-lipid membraned particles that are characterized by a high heterogeneity that emerges from their distinct biogenesis route, their multi-variable sizes, and the particular cargoes that are embedded into these particles. Their pivotal role in cell-to-cell communication via their cargo and their implication in the pathophysiology of several diseases, including pancreatic cancer, opens new horizons in the management of this malignancy. Meanwhile, the interplay between pancreatic carcinogenesis and short non-coding RNA molecules (micro-RNAs or miRs) is in the spotlight of current studies, as they can have either a role as tumor suppressors or promoters. The deregulation of both of the aforementioned molecules leads to several aberrations in the function of pancreatic cells, leading to carcinogenesis. In this review, we will explore the role of extracellular vesicles and miRNAs in pancreatic cancer, as well as their potent utilization as diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Eleni Myrto Trifylli
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, Section of Medical Laboratories, School of Health & Caring Sciences, University of West Attica (UniWA), Ag. Spyridonos Str., 12243 Egaleo, Greece; (E.M.T.); (S.P.F.)
- First Department of Internal Medicine, 417 Army Share Fund Hospital, 11521 Athens, Greece;
- GI-Liver Unit, 2nd Department of Internal Medicine National and Kapodistrian University of Athens, General Hospital of Athens “Hippocratio”, 114 Vas Sofias, 11527 Athens, Greece; (S.V.); (S.M.); (M.D.)
| | - Anastasios G. Kriebardis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, Section of Medical Laboratories, School of Health & Caring Sciences, University of West Attica (UniWA), Ag. Spyridonos Str., 12243 Egaleo, Greece; (E.M.T.); (S.P.F.)
| | - Evangelos Koustas
- Oncology Department, General Hospital Evangelismos, 10676 Athens, Greece;
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Nikolaos Papadopoulos
- Second Department of Internal Medicine, 401 General Military Hospital, 11527 Athens, Greece;
| | - Sotirios P. Fortis
- Laboratory of Reliability and Quality Control in Laboratory Hematology (HemQcR), Department of Biomedical Sciences, Section of Medical Laboratories, School of Health & Caring Sciences, University of West Attica (UniWA), Ag. Spyridonos Str., 12243 Egaleo, Greece; (E.M.T.); (S.P.F.)
| | - Vassilis L. Tzounakas
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece; (A.T.A.); (V.L.T.)
| | - Alkmini T. Anastasiadi
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece; (A.T.A.); (V.L.T.)
| | - Panagiotis Sarantis
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Sofia Vasileiadi
- GI-Liver Unit, 2nd Department of Internal Medicine National and Kapodistrian University of Athens, General Hospital of Athens “Hippocratio”, 114 Vas Sofias, 11527 Athens, Greece; (S.V.); (S.M.); (M.D.)
| | - Ariadne Tsagarakis
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA;
| | - Georgios Aloizos
- First Department of Internal Medicine, 417 Army Share Fund Hospital, 11521 Athens, Greece;
| | - Spilios Manolakopoulos
- GI-Liver Unit, 2nd Department of Internal Medicine National and Kapodistrian University of Athens, General Hospital of Athens “Hippocratio”, 114 Vas Sofias, 11527 Athens, Greece; (S.V.); (S.M.); (M.D.)
| | - Melanie Deutsch
- GI-Liver Unit, 2nd Department of Internal Medicine National and Kapodistrian University of Athens, General Hospital of Athens “Hippocratio”, 114 Vas Sofias, 11527 Athens, Greece; (S.V.); (S.M.); (M.D.)
| |
Collapse
|
30
|
Zhou Q, Meng Y, Li D, Yao L, Le J, Liu Y, Sun Y, Zeng F, Chen X, Deng G. Ferroptosis in cancer: From molecular mechanisms to therapeutic strategies. Signal Transduct Target Ther 2024; 9:55. [PMID: 38453898 PMCID: PMC10920854 DOI: 10.1038/s41392-024-01769-5] [Citation(s) in RCA: 159] [Impact Index Per Article: 159.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/21/2024] [Accepted: 02/03/2024] [Indexed: 03/09/2024] Open
Abstract
Ferroptosis is a non-apoptotic form of regulated cell death characterized by the lethal accumulation of iron-dependent membrane-localized lipid peroxides. It acts as an innate tumor suppressor mechanism and participates in the biological processes of tumors. Intriguingly, mesenchymal and dedifferentiated cancer cells, which are usually resistant to apoptosis and traditional therapies, are exquisitely vulnerable to ferroptosis, further underscoring its potential as a treatment approach for cancers, especially for refractory cancers. However, the impact of ferroptosis on cancer extends beyond its direct cytotoxic effect on tumor cells. Ferroptosis induction not only inhibits cancer but also promotes cancer development due to its potential negative impact on anticancer immunity. Thus, a comprehensive understanding of the role of ferroptosis in cancer is crucial for the successful translation of ferroptosis therapy from the laboratory to clinical applications. In this review, we provide an overview of the recent advancements in understanding ferroptosis in cancer, covering molecular mechanisms, biological functions, regulatory pathways, and interactions with the tumor microenvironment. We also summarize the potential applications of ferroptosis induction in immunotherapy, radiotherapy, and systemic therapy, as well as ferroptosis inhibition for cancer treatment in various conditions. We finally discuss ferroptosis markers, the current challenges and future directions of ferroptosis in the treatment of cancer.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yu Meng
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Lei Yao
- Department of General Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Jiayuan Le
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yihuang Liu
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- Furong Laboratory, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, 87 Xiangya Road, Changsha, 410008, Hunan Province, China.
| |
Collapse
|
31
|
Wu K, El Zowalaty AE, Sayin VI, Papagiannakopoulos T. The pleiotropic functions of reactive oxygen species in cancer. NATURE CANCER 2024; 5:384-399. [PMID: 38531982 DOI: 10.1038/s43018-024-00738-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 01/19/2024] [Indexed: 03/28/2024]
Abstract
Cellular redox homeostasis is an essential, dynamic process that ensures the balance between reducing and oxidizing reactions within cells and thus has implications across all areas of biology. Changes in levels of reactive oxygen species can disrupt redox homeostasis, leading to oxidative or reductive stress that contributes to the pathogenesis of many malignancies, including cancer. From transformation and tumor initiation to metastatic dissemination, increasing reactive oxygen species in cancer cells can paradoxically promote or suppress the tumorigenic process, depending on the extent of redox stress, its spatiotemporal characteristics and the tumor microenvironment. Here we review how redox regulation influences tumorigenesis, highlighting therapeutic opportunities enabled by redox-related alterations in cancer cells.
Collapse
Affiliation(s)
- Katherine Wu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Ahmed Ezat El Zowalaty
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Volkan I Sayin
- Institute of Clinical Sciences, Department of Surgery, Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden.
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Thales Papagiannakopoulos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA.
- Perlmutter NYU Cancer Center, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
32
|
Wu D, Zhang K, Khan FA, Pandupuspitasari NS, Guan K, Sun F, Huang C. A comprehensive review on signaling attributes of serine and serine metabolism in health and disease. Int J Biol Macromol 2024; 260:129607. [PMID: 38253153 DOI: 10.1016/j.ijbiomac.2024.129607] [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: 09/24/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 01/24/2024]
Abstract
Serine is a metabolite with ever-expanding metabolic and non-metabolic signaling attributes. By providing one‑carbon units for macromolecule biosynthesis and functional modifications, serine and serine metabolism largely impinge on cellular survival and function. Cancer cells frequently have a preference for serine metabolic reprogramming to create a conducive metabolic state for survival and aggressiveness, making intervention of cancer-associated rewiring of serine metabolism a promising therapeutic strategy for cancer treatment. Beyond providing methyl donors for methylation in modulation of innate immunity, serine metabolism generates formyl donors for mitochondrial tRNA formylation which is required for mitochondrial function. Interestingly, fully developed neurons lack the machinery for serine biosynthesis and rely heavily on astrocytic l-serine for production of d-serine to shape synaptic plasticity. Here, we recapitulate recent discoveries that address the medical significance of serine and serine metabolism in malignancies, mitochondrial-associated disorders, and neurodegenerative pathologies. Metabolic control and epigenetic- and posttranslational regulation of serine metabolism are also discussed. Given the metabolic similarities between cancer cells, neurons and germ cells, we further propose the relevance of serine metabolism in testicular homeostasis. Our work provides valuable hints for future investigations that will lead to a deeper understanding of serine and serine metabolism in cellular physiology and pathology.
Collapse
Affiliation(s)
- Di Wu
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Kejia Zhang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China
| | - Faheem Ahmed Khan
- Research Center for Animal Husbandry, National Research and Innovation Agency, Jakarta Pusat 10340, Indonesia
| | | | - Kaifeng Guan
- School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China.
| | - Fei Sun
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| | - Chunjie Huang
- Institute of Reproductive Medicine, School of Medicine, Nantong University, Nantong 226001, China.
| |
Collapse
|
33
|
Yaylim İ, Aru M, Farooqi AA, Hakan MT, Buttari B, Arese M, Saso L. Regulation of Nrf2/Keap1 signaling pathway in cancer drug resistance by galectin-1: cellular and molecular implications. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:8. [PMID: 38434765 PMCID: PMC10905161 DOI: 10.20517/cdr.2023.79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Oxidative stress is characterized by the deregulation of the redox state in the cells, which plays a role in the initiation of various types of cancers. The activity of galectin-1 (Gal-1) depends on the cell redox state and the redox state of the microenvironment. Gal-1 expression has been related to many different tumor types, as it plays important roles in several processes involved in cancer progression, such as apoptosis, cell migration, adhesion, and immune response. The erythroid-2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1) signaling pathway is a crucial mechanism involved in both cell survival and cell defense against oxidative stress. In this review, we delve into the cellular and molecular roles played by Gal-1 in the context of oxidative stress onset in cancer cells, particularly focusing on its involvement in activating the Nrf2/Keap1 signaling pathway. The emerging evidence concerning the anti-apoptotic effect of Gal-1, together with its ability to sustain the activation of the Nrf2 pathway in counteracting oxidative stress, supports the role of Gal-1 in the promotion of tumor cells proliferation, immuno-suppression, and anti-tumor drug resistance, thus highlighting that the inhibition of Gal-1 emerges as a potential strategy for the restraint and regression of tumor progression. Overall, a deeper understanding of the multi-functionality and disease-specific expression profiling of Gal-1 will be crucial for the design and development of novel Gal-1 inhibitors as anticancer agents. Excitingly, although it is still understudied, the ever-growing knowledge of the sophisticated interplay between Gal-1 and Nrf2/Keap1 will enable researchers to gain valuable insights into the underlying causes of carcinogenesis and metastasis.
Collapse
Affiliation(s)
- İlhan Yaylim
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul 34280, Turkiye
| | - Melek Aru
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul 34280, Turkiye
- Department of Medical Education, Istinye University Faculty of Medicine, Istanbul 34396, Turkiye
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 54000, Pakistan
| | - Mehmet Tolgahan Hakan
- Department of Molecular Medicine, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul 34280, Turkiye
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, Rome 00161, Italy
| | - Marzia Arese
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University, Rome 00185, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University, Rome 00185, Italy
| |
Collapse
|
34
|
Ray SK, Mukherjee S. Emerging Role of Ferroptosis in Breast Cancer: Characteristics, Therapy, and Translational Implications for the Present and Future. Curr Mol Med 2024; 24:1470-1482. [PMID: 37711099 DOI: 10.2174/1566524023666230913105735] [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: 05/29/2023] [Revised: 07/09/2023] [Accepted: 07/27/2023] [Indexed: 09/16/2023]
Abstract
Ferroptosis is a nonapoptotic, iron-dependent form of cell death that can be actuated in disease cells by expected improvements and manufactured specialists. Different studies have recently resurrected the role of this newly discovered cell death pathway and demonstrated its efficacy in treating breast cancer. Breast cancer is the most well-known type of cancer among women worldwide. Despite many years of research focusing on cell death in breast cancer, counting apoptosis, clinical treatment leftovers are difficult due to the high likelihood of recurrence. Ferroptosis is defined by a lack of lipid peroxide repair capacity by phospholipid hydroperoxides GPX4, accessibility of redox-active iron, and followed oxidation of polyunsaturated fatty acids acid-containing phospholipids signalling, amino acid and iron metabolism, ferritinophagy, epithelial-tomesenchymal transition, cell adhesion, and mevalonate and phospholipid biosynthesis can all be factors that influence ferroptosis susceptibility. Ferroptosis, an iron-dependent controlled cell death caused by excessive lipid peroxidation, has been entwined in breast cancer development and therapeutic response for the past decade. Advances in enhancing clinical drugs targeting ferroptosis are developing silver linings to treat breast cancer. Ferroptosis is influenced by metabolism and the expression of certain genes, making it a prospective therapeutic target for monitoring malignant growth and an appealing target for precision cancer medication disclosure. In the coming years, research into biomarkers to follow ferroptosis in patients with breast cancer and the course of events and the subsequent use of novel ferroptosis-based treatments will be captious. We present a fundamental analysis of the actual understanding of molecular mechanisms along with regulatory networks associated with ferroptosis, expected physiological functions in growth concealment, ferroptosis-associated differentially expressed genes, treatment targeting potential, and recent advances in the development of therapeutic strategies in this review.
Collapse
Affiliation(s)
- Suman Kumar Ray
- Independent researcher, Bhopal, Madhya Pradesh, 462020, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh, 462020, India
| |
Collapse
|
35
|
Ngo HKC, Le H, Surh YJ. Nrf2, A Target for Precision Oncology in Cancer Prognosis and Treatment. J Cancer Prev 2023; 28:131-142. [PMID: 38205365 PMCID: PMC10774478 DOI: 10.15430/jcp.2023.28.4.131] [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: 12/15/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Activating nuclear factor-erythroid 2-related factor (Nrf2), a master regulator of redox homeostasis, has been shown to suppress initiation of carcinogenesis in normal cells. However, this transcription factor has recently been reported to promote proliferation of some transformed or cancerous cells. In tumor cells, Nrf2 is prone to mutations that result in stabilization and concurrent accumulation of its protein product. A hyperactivated mutant form of Nrf2 could support the cancer cells for enhanced proliferation, invasiveness, and resistance to chemotherapeutic agents and radiotherapy, which are associated with a poor clinical outcome. Hence understanding mutations in Nrf2 would have a significant impact on the prognosis and treatment of cancer in the era of precision medicine. This perspective would provide an insight into the genetic alterations in Nrf2 and suggest the application of small molecules, RNAi, and genome editing technologies, particularly CRISR-Cas9, in therapeutic intervention of cancer in the context of the involvement of Nrf2 mutations.
Collapse
Affiliation(s)
- Hoang Kieu Chi Ngo
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Hoang Le
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, USA
| | - Young-Joon Surh
- Tumor Microenvironment Global Core Research Center, College of Pharmacy, Seoul National University, Seoul, Korea
- Cancer Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
36
|
Di Giorgio E, Choudhary H, Ferino A, Cortolezzis Y, Dalla E, D’Este F, Comelli M, Rapozzi V, Xodo LE. Suppression of the KRAS- NRF2 axis shifts arginine into the phosphocreatine energy system in pancreatic cancer cells. iScience 2023; 26:108566. [PMID: 38144458 PMCID: PMC10746371 DOI: 10.1016/j.isci.2023.108566] [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: 05/24/2023] [Revised: 10/21/2023] [Accepted: 11/21/2023] [Indexed: 12/26/2023] Open
Abstract
In pancreatic ductal adenocarcinomas (PDAC), the KRASG12D-NRF2 axis controls cellular functions such as redox homeostasis and metabolism. Disruption of this axis through suppression of NRF2 leads to profound reprogramming of metabolism. Unbiased transcriptome and metabolome analyses showed that PDAC cells with disrupted KRASG12D-NRF2 signaling (NRF2-/- cells) shift from aerobic glycolysis to metabolic pathways fed by amino acids. Metabolome, RNA-seq and qRT-PCR analyses revealed a blockade of the urea cycle, making NRF2-/- cells dependent on exogenous arginine for survival. Arginine is channeled into anabolic pathways, including the synthesis of phosphocreatine, which generates an energy buffer essential for cell growth. A similar switch was observed in tumor clones that had survived FOLFIRINOX therapy or blockade of KRAS signaling. Inhibition of the creatine pathway with cyclocreatine reduced both ATP and invasion rate in 3D spheroids from NRF2-deficient PDAC cells. Our study provides basis for the rational development of combination therapies for pancreatic cancer.
Collapse
Affiliation(s)
- Eros Di Giorgio
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Himanshi Choudhary
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Annalisa Ferino
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Ylenia Cortolezzis
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Emiliano Dalla
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Francesca D’Este
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Marina Comelli
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Valentina Rapozzi
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| | - Luigi E. Xodo
- Department of Medicine, Laboratory of Biochemistry, P.le Kolbe 4, 33100 Udine, Italy
| |
Collapse
|
37
|
Boutin R, Lee HF, Guan TL, Nguyen TT, Huang XF, Waller DD, Lu J, Christine Chio II, Michel RP, Sebag M, Tsantrizos YS. Discovery and Evaluation of C6-Substituted Pyrazolopyrimidine-Based Bisphosphonate Inhibitors of the Human Geranylgeranyl Pyrophosphate Synthase and Evaluation of Their Antitumor Efficacy in Multiple Myeloma, Pancreatic Ductal Adenocarcinoma, and Colorectal Cancer. J Med Chem 2023; 66:15776-15800. [PMID: 37982711 PMCID: PMC10832233 DOI: 10.1021/acs.jmedchem.3c01271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Novel C6-substituted pyrazolo[3,4-d]pyrimidine- and C2-substituted purine-based bisphosphonate (C6-PyraP-BP and C2-Pur-BP, respectively) inhibitors of the human geranylgeranyl pyrophosphate synthase (hGGPPS) were designed and evaluated for their ability to block the proliferation of multiple myeloma (MM), pancreatic ductal adenocarcinoma (PDAC), and colorectal cancer (CRC) cells. Pyrazolo[3,4-d]pyrimidine analogs were identified that induce selective intracellular target engagement leading to apoptosis and downregulate the prenylation of Rap-1A in MM, PDAC, and CRC cells. The C6-PyraP-BP inhibitor RB-07-16 was found to exhibit antitumor efficacy in xenograft mouse models of MM and PDAC, significantly reducing tumor growth without substantially increasing liver enzymes or causing significant histopathologic damage, usually associated with hepatotoxicity. RB-07-16 is a metabolically stable compound in cross-species liver microsomes, does not inhibit key CYP 450 enzymes, and exhibits good systemic circulation in rat. Collectively, the current studies provide encouraging support for further optimization of the pyrazolo[3,4-d]pyrimidine-based GGPPS inhibitors as potential human therapeutics for various cancers.
Collapse
Affiliation(s)
- Rebecca Boutin
- Department of Chemistry, McGill University, Montreal, Québec H3A 0B8, Canada
| | - Hiu-Fung Lee
- Department of Chemistry, McGill University, Montreal, Québec H3A 0B8, Canada
| | - Tian Lai Guan
- Department of Chemistry, McGill University, Montreal, Québec H3A 0B8, Canada
- Department of Biochemistry, McGill University, Montreal, Québec H3G 1Y6, Canada
| | - Tan Trieu Nguyen
- Department of Medicine, McGill University, Montreal, Québec H3A 1A1, Canada
| | - Xian Fang Huang
- Department of Medicine, McGill University, Montreal, Québec H3A 1A1, Canada
| | - Daniel D Waller
- Terry Fox Laboratory, BC Cancer Research Institute, Vancouver, British Columbia V5Z 1L3, Canada
| | - Jordan Lu
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Iok In Christine Chio
- Institute for Cancer Genetics, Department of Genetics and Development, Columbia University Irving Medical Center, New York, New York 10032, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - René P Michel
- Department of Pathology, McGill University, Montréal, Québec H3A 1A1, Canada
| | - Michael Sebag
- Department of Medicine, McGill University, Montreal, Québec H3A 1A1, Canada
- Division of Hematology, McGill University Health Center, Montreal, Québec H4A 3J1, Canada
| | - Youla S Tsantrizos
- Department of Chemistry, McGill University, Montreal, Québec H3A 0B8, Canada
- Department of Biochemistry, McGill University, Montreal, Québec H3G 1Y6, Canada
| |
Collapse
|
38
|
Jia W, Gong X, Ye Z, Li N, Zhan X. Nitroproteomics is instrumental for stratification and targeted treatments of astrocytoma patients: expert recommendations for advanced 3PM approach with improved individual outcomes. EPMA J 2023; 14:673-696. [PMID: 38094577 PMCID: PMC10713973 DOI: 10.1007/s13167-023-00348-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/11/2023] [Indexed: 12/05/2024]
Abstract
Protein tyrosine nitration is a selectively and reversible important post-translational modification, which is closely related to oxidative stress. Astrocytoma is the most common neuroepithelial tumor with heterogeneity and complexity. In the past, the diagnosis of astrocytoma was based on the histological and clinical features, and the treatment methods were nothing more than surgery-assisted radiotherapy and chemotherapy. Obviously, traditional methods short falls an effective treatment for astrocytoma. In late 2021, the World Health Organization (WHO) adopted molecular biomarkers in the comprehensive diagnosis of astrocytoma, such as IDH-mutant and DNA methylation, which enabled the risk stratification, classification, and clinical prognosis prediction of astrocytoma to be more correct. Protein tyrosine nitration is closely related to the pathogenesis of astrocytoma. We hypothesize that nitroproteome is significantly different in astrocytoma relative to controls, which leads to establishment of nitroprotein biomarkers for patient stratification, diagnostics, and prediction of disease stages and severity grade, targeted prevention in secondary care, treatment algorithms tailored to individualized patient profile in the framework of predictive, preventive, and personalized medicine (PPPM; 3P medicine). Nitroproteomics based on gel electrophoresis and tandem mass spectrometry is an effective tool to identify the nitroproteins and effective biomarkers in human astrocytomas, clarifying the biological roles of oxidative/nitrative stress in the pathophysiology of astrocytomas, functional characteristics of nitroproteins in astrocytomas, nitration-mediated signal pathway network, and early diagnosis and treatment of astrocytomas. The results finds that these nitroproteins are enriched in mitotic cell components, which are related to transcription regulation, signal transduction, controlling subcellular organelle events, cell perception, maintaining cell homeostasis, and immune activity. Eleven statistically significant signal pathways are identified in astrocytoma, including remodeling of epithelial adherens junctions, germ cell-sertoli cell junction signaling, 14-3-3-mediated signaling, phagosome maturation, gap junction signaling, axonal guidance signaling, assembly of RNA polymerase III complex, and TREM1 signaling. Furthermore, protein tyrosine nitration is closely associated with the therapeutic effects of protein drugs, and molecular mechanism and drug targets of cancer. It provides valuable data for studying the protein nitration biomarkers, molecular mechanisms, and therapeutic targets of astrocytoma towards PPPM (3P medicine) practice. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00348-y.
Collapse
Affiliation(s)
- Wenshuang Jia
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xiaoxia Gong
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Zhen Ye
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Na Li
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Medical Science and Technology Innovation Center, Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
39
|
Noè R, Inglese N, Romani P, Serafini T, Paoli C, Calciolari B, Fantuz M, Zamborlin A, Surdo NC, Spada V, Spacci M, Volta S, Ermini ML, Di Benedetto G, Frusca V, Santi C, Lefkimmiatis K, Dupont S, Voliani V, Sancineto L, Carrer A. Organic Selenium induces ferroptosis in pancreatic cancer cells. Redox Biol 2023; 68:102962. [PMID: 38029455 PMCID: PMC10698006 DOI: 10.1016/j.redox.2023.102962] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 11/10/2023] [Indexed: 12/01/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDA) cells reprogram both mitochondrial and lysosomal functions to support growth. At the same time, this causes significant dishomeostasis of free radicals. While this is compensated by the upregulation of detoxification mechanisms, it also represents a potential vulnerability. Here we demonstrate that PDA cells are sensitive to the inhibition of the mevalonate pathway (MVP), which supports the biosynthesis of critical antioxidant intermediates and protect from ferroptosis. We attacked the susceptibility of PDA cells to ferroptotic death with selenorganic compounds, including dibenzyl diselenide (DBDS) that exhibits potent pro-oxidant properties and inhibits tumor growth in vitro and in vivo. DBDS treatment induces the mobilization of iron from mitochondria enabling uncontrolled lipid peroxidation. Finally, we showed that DBDS and statins act synergistically to promote ferroptosis and provide evidence that combined treatment is a viable strategy to combat PDA.
Collapse
Affiliation(s)
- Roberta Noè
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Noemi Inglese
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Patrizia Romani
- Department of Molecular Medicine, University of Padova, 35126, Padova, Italy
| | - Thauan Serafini
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy
| | - Carlotta Paoli
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Beatrice Calciolari
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Marco Fantuz
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Agata Zamborlin
- NEST-Scuola Normale Superiore, 56127, Pisa, Italy; Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, 56127, Pisa, Italy
| | - Nicoletta C Surdo
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy
| | - Vittoria Spada
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy
| | - Martina Spacci
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy
| | - Sara Volta
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy
| | - Maria Laura Ermini
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, 56127, Pisa, Italy
| | - Giulietta Di Benedetto
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy
| | - Valentina Frusca
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, 56127, Pisa, Italy; Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127, Pisa, Italy
| | - Claudio Santi
- Group of Catalysis and Green Organic Chemistry, Department of Pharmaceutical Sciences, University of Perugia, 06122, Perugia, PG, Italy
| | - Konstantinos Lefkimmiatis
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Sirio Dupont
- Department of Molecular Medicine, University of Padova, 35126, Padova, Italy
| | - Valerio Voliani
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, 56127, Pisa, Italy; Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genova, 16148, Genoa, Italy.
| | - Luca Sancineto
- Group of Catalysis and Green Organic Chemistry, Department of Pharmaceutical Sciences, University of Perugia, 06122, Perugia, PG, Italy.
| | - Alessandro Carrer
- Veneto Institute of Molecular Medicine (VIMM), 35129, Padova, Italy; Department of Biology, University of Padova, 35126, Padova, Italy.
| |
Collapse
|
40
|
Huang J, Zhang L, Yang R, Yao L, Gou J, Cao D, Pan Z, Li D, Pan Y, Zhang W. Eukaryotic translation initiation factor 4A1 in the pathogenesis and treatment of cancers. Front Mol Biosci 2023; 10:1289650. [PMID: 38028556 PMCID: PMC10666758 DOI: 10.3389/fmolb.2023.1289650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Abnormal translate regulation is an important phenomenon in cancer initiation and progression. Eukaryotic translation initiation factor 4A1 (eIF4A1) protein is an ATP-dependent Ribonucleic Acid (RNA) helicase, which is essential for translation and has bidirectional RNA unwinders function. In this review, we discuss the levels of expression, regulatory mechanisms and protein functions of eIF4A1 in different human tumors. eIF4A1 is often involved as a target of microRNAs or long non-coding RNAs during the epithelial-mesenchymal transition, associating with the proliferation and metastasis of tumor cells. eIF4A1 protein exhibits the promising biomarker for rapid diagnosis of pre-cancer lesions, histological phenotypes, clinical staging diagnosis and outcome prediction, which provides a novel strategy for precise medical care and target therapy for patients with tumors at the same time, relevant small molecule inhibitors have also been applied in clinical practice, providing reliable theoretical support and clinical basis for the development of this gene target.
Collapse
Affiliation(s)
- Jinghong Huang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Lei Zhang
- Clinical Laboratory, First Affiliated Hospital of Shihezi University, Shihezi, Xinjiang, China
| | - Rui Yang
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Lixia Yao
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Jinming Gou
- Troops of the People’s Liberation Army, Urumqi, Xinjiang, China
| | - Dongdong Cao
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Zeming Pan
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Dongmei Li
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, School of Medicine, Shihezi University, Shihezi, Xinjiang, China
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Beijing Tuberculosis and Thoracic Tumor Research Institute, Capital Medical University, Beijing, China
| | - Wei Zhang
- Shihezi People’s Hospital, Shihezi, Xinjiang, China
| |
Collapse
|
41
|
Radyk MD, Nelson BS, Halbrook CJ, Wood A, Lavoie B, Salvatore L, Corfas G, Colacino JA, Shah YM, Crawford HC, Lyssiotis CA. Glucose-6-phosphate dehydrogenase deficiency accelerates pancreatic acinar-to-ductal metaplasia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.06.565895. [PMID: 37986898 PMCID: PMC10659312 DOI: 10.1101/2023.11.06.565895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Activating mutations in KRAS extensively reprogram cellular metabolism to support the continuous growth, proliferation, and survival of pancreatic tumors. Targeting these metabolic dependencies are promising approaches for the treatment of established tumors. However, metabolic reprogramming is required early during tumorigenesis to provide transformed cells selective advantage towards malignancy. Acinar cells can give rise to pancreatic tumors through acinar-to-ductal metaplasia (ADM). Dysregulation of pathways that maintain acinar homeostasis accelerate tumorigenesis. During ADM, acinar cells transdifferentiate to duct-like cells, a process driven by oncogenic KRAS. The metabolic reprogramming that is required for the transdifferentiation in ADM is unclear. We performed transcriptomic analysis on mouse acinar cells undergoing ADM and found metabolic programs are globally enhanced, consistent with the transition of a specialized cell to a less differentiated phenotype with proliferative potential. Indeed, we and others have demonstrated how inhibiting metabolic pathways necessary for ADM can prevent transdifferentiation and tumorigenesis. Here, we also find NRF2-target genes are differentially expressed during ADM. Among these, we focused on the increase in the gene coding for NADPH-producing enzyme, Glucose-6-phosphate dehydrogenase (G6PD). Using established mouse models of KrasG12D-driven pancreatic tumorigenesis and G6PD-deficiency, we find that mutant G6pd accelerates ADM and pancreatic intraepithelial neoplasia. Acceleration of cancer initiation with G6PD-deficiency is dependent on its NADPH-generating function in reactive oxygen species (ROS) management, as opposed to other outputs of the pentose phosphate pathway. Together, this work provides new insights into the function of metabolic pathways during early tumorigenesis.
Collapse
Affiliation(s)
- Megan D. Radyk
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Barbara S. Nelson
- Doctoral Program in Cancer Biology, University of Michigan, Ann Arbor, USA
| | - Christopher J. Halbrook
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, CA, USA
- University of California Irvine Chao Family Comprehensive Cancer Center, Orange, CA, USA
| | - Alexander Wood
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Brooke Lavoie
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Lucie Salvatore
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Gabriel Corfas
- Kresge Hearing Research Institute and Dept. of Otolaryngology, Head and Neck Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Justin A. Colacino
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Department of Nutritional Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Program in the Environment, College of Literature, Sciences, and the Arts, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Yatrik M. Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
| | - Howard C. Crawford
- Department of Surgery, Henry Ford Pancreatic Cancer Center, Henry Ford Health System, Detroit, MI, USA
| | - Costas A. Lyssiotis
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- University of Michigan Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
42
|
Amin KN, Rajaguru P, Suzuki T, Sarkar K, Ganesan K, Ramkumar KM. Quantitative proteomic analyses uncover regulatory roles of Nrf2 in human endothelial cells. Cell Stress Chaperones 2023; 28:731-747. [PMID: 37488350 PMCID: PMC10746666 DOI: 10.1007/s12192-023-01366-5] [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: 04/05/2023] [Revised: 06/13/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2), a transcriptional regulator, is the predominant factor in modulating oxidative stress and other cellular signaling responses. Studies from our lab and others highlighted that activation of the Nrf2 pathway by small molecules improves endothelial function by suppressing oxidative and endoplasmic reticulum (ER) stress. However, the exact mechanisms by which Nrf2 elicits these effects are unknown. In the present study, we developed CRISPR/Cas9-mediated Nrf2 knocked-out human endothelial cells, and proteomic signature was studied using LC-MS/MS. We identified 723 unique proteins, of which 361 proteins were found to be differentially regulated and further screened in the Nrf2ome online database, where we identified a highly interconnected signaling network in which 70 proteins directly interact with Nrf2. These proteins were found to regulate some key cellular and metabolic processes in the regulation actin cytoskeleton, ER stress, angiogenesis, inflammation, Hippo signaling pathway, and epidermal growth factor/fibroblast growth factor (EGF/FGF) signaling pathway. Our findings suggest the role of Nrf2 in maintaining endothelium integrity and its relationship with the crucial cellular processes which help develop novel therapeutics against endothelial dysfunction and its associated complications.
Collapse
Affiliation(s)
- Karan Naresh Amin
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Palanichamy Rajaguru
- Department of Biotechnology, Central University of Tamil Nadu, Tiruvarur, 610005, India
| | - Takayoshi Suzuki
- Division Cellular and Gene Therapy Products, National Institute of Health Sciences, Setagaya-Ku, Tokyo, 158-8501, Japan
| | - Koustav Sarkar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Kumar Ganesan
- School of Chinese Medicine, LKS Faculty of Medicine, University of Hong Kong, 10 Sassoon Road, Hong Kong, 999077, China
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India.
| |
Collapse
|
43
|
Xia L, Ma W, Afrashteh A, Sajadi MA, Fakheri H, Valilo M. The nuclear factor erythroid 2-related factor 2/p53 axis in breast cancer. Biochem Med (Zagreb) 2023; 33:030504. [PMID: 37841775 PMCID: PMC10564154 DOI: 10.11613/bm.2023.030504] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
One of the most important factors involved in the response to oxidative stress (OS) is the nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates the expression of components such as antioxidative stress proteins and enzymes. Under normal conditions, Kelch-like ECH-associated protein 1 (Keap1) keeps Nrf2 in the cytoplasm, thus preventing its translocation to the nucleus and inhibiting its role. It has been established that Nrf2 has a dual function; on the one hand, it promotes angiogenesis and cancer cell metastasis while causing resistance to drugs and chemotherapy. On the other hand, Nrf2 increases expression and proliferation of glutathione to protect cells against OS. p53 is a tumour suppressor that activates the apoptosis pathway in aging and cancer cells in addition to stimulating the glutaminolysis and antioxidant pathways. Cancer cells use the antioxidant ability of p53 against OS. Therefore, in the present study, we discussed function of Nrf2 and p53 in breast cancer (BC) cells to elucidate their role in protection or destruction of cancer cells as well as their drug resistance or antioxidant properties.
Collapse
Affiliation(s)
- Lei Xia
- Surgical oncology ward 2, Qinghai Provincial People’s Hospital, Xining Qinghai, China
| | - Wenbiao Ma
- Surgical oncology ward 2, Qinghai Provincial People’s Hospital, Xining Qinghai, China
| | - Ahmad Afrashteh
- Department of Periodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hadi Fakheri
- Paramedical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
44
|
Praharaj PP, Singh A, Patra S, Bhutia SK. Co-targeting autophagy and NRF2 signaling triggers mitochondrial superoxide to sensitize oral cancer stem cells for cisplatin-induced apoptosis. Free Radic Biol Med 2023; 207:72-88. [PMID: 37423560 DOI: 10.1016/j.freeradbiomed.2023.07.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/09/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023]
Abstract
Cancer stem cell (CSC) populations are regulated by autophagy, which in turn modulates tumorigenicity and malignancy. In this study, we demonstrated that cisplatin treatment enriches the CSCs population by increasing autophagosome formation and speeding up autophagosome-lysosome fusion by recruiting RAB7 to autolysosomes. Further, cisplatin treatment stimulates lysosomal activity and increases autophagic flux in oral CD44+ cells. Interestingly, both ATG5- and BECN1-dependent autophagy are essential for maintaining cancer stemness, self-renewal, and resistance to cisplatin-induced cytotoxicity in oral CD44+ cells. Moreover, we discovered that autophagy-deficient (shATG5 and/or shBECN1) CD44+ cells activates nuclear factor, erythroid 2 like 2 (NRF2) signaling, which in turn reduces the elevated reactive oxygen species (ROS) level enhancing cancer stemness. Genetic inhibition of NRF2 (siNRF2) in autophagy-deficient CD44+ cells increases mitochondrial ROS (mtROS) level, reducing cisplatin-resistance CSCs, and pre-treatment with mitoTEMPO [a mitochondria-targeted superoxide dismutase (SOD) mimetic] lessened the cytotoxic effect enhancing cancer stemness. We also found that inhibiting autophagy (with CQ) and NRF2 signaling (with ML-385) combinedly increases cisplatin cytotoxicity, thereby suppressing the expansion of oral CD44+ cells; this finding has the potential to be clinically applicable in resolving CSC-associated chemoresistance and tumor relapse in oral cancer.
Collapse
Affiliation(s)
- Prakash P Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India
| | - Amruta Singh
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India
| | - Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India
| | - Sujit K Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha, 769008, India.
| |
Collapse
|
45
|
Jomova K, Raptova R, Alomar SY, Alwasel SH, Nepovimova E, Kuca K, Valko M. Reactive oxygen species, toxicity, oxidative stress, and antioxidants: chronic diseases and aging. Arch Toxicol 2023; 97:2499-2574. [PMID: 37597078 PMCID: PMC10475008 DOI: 10.1007/s00204-023-03562-9] [Citation(s) in RCA: 665] [Impact Index Per Article: 332.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 07/24/2023] [Indexed: 08/21/2023]
Abstract
A physiological level of oxygen/nitrogen free radicals and non-radical reactive species (collectively known as ROS/RNS) is termed oxidative eustress or "good stress" and is characterized by low to mild levels of oxidants involved in the regulation of various biochemical transformations such as carboxylation, hydroxylation, peroxidation, or modulation of signal transduction pathways such as Nuclear factor-κB (NF-κB), Mitogen-activated protein kinase (MAPK) cascade, phosphoinositide-3-kinase, nuclear factor erythroid 2-related factor 2 (Nrf2) and other processes. Increased levels of ROS/RNS, generated from both endogenous (mitochondria, NADPH oxidases) and/or exogenous sources (radiation, certain drugs, foods, cigarette smoking, pollution) result in a harmful condition termed oxidative stress ("bad stress"). Although it is widely accepted, that many chronic diseases are multifactorial in origin, they share oxidative stress as a common denominator. Here we review the importance of oxidative stress and the mechanisms through which oxidative stress contributes to the pathological states of an organism. Attention is focused on the chemistry of ROS and RNS (e.g. superoxide radical, hydrogen peroxide, hydroxyl radicals, peroxyl radicals, nitric oxide, peroxynitrite), and their role in oxidative damage of DNA, proteins, and membrane lipids. Quantitative and qualitative assessment of oxidative stress biomarkers is also discussed. Oxidative stress contributes to the pathology of cancer, cardiovascular diseases, diabetes, neurological disorders (Alzheimer's and Parkinson's diseases, Down syndrome), psychiatric diseases (depression, schizophrenia, bipolar disorder), renal disease, lung disease (chronic pulmonary obstruction, lung cancer), and aging. The concerted action of antioxidants to ameliorate the harmful effect of oxidative stress is achieved by antioxidant enzymes (Superoxide dismutases-SODs, catalase, glutathione peroxidase-GPx), and small molecular weight antioxidants (vitamins C and E, flavonoids, carotenoids, melatonin, ergothioneine, and others). Perhaps one of the most effective low molecular weight antioxidants is vitamin E, the first line of defense against the peroxidation of lipids. A promising approach appears to be the use of certain antioxidants (e.g. flavonoids), showing weak prooxidant properties that may boost cellular antioxidant systems and thus act as preventive anticancer agents. Redox metal-based enzyme mimetic compounds as potential pharmaceutical interventions and sirtuins as promising therapeutic targets for age-related diseases and anti-aging strategies are discussed.
Collapse
Affiliation(s)
- Klaudia Jomova
- Department of Chemistry, Faculty of Natural Sciences, Constantine the Philosopher University in Nitra, Nitra, 949 74, Slovakia
| | - Renata Raptova
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, 812 37, Slovakia
| | - Suliman Y Alomar
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Saleh H Alwasel
- Zoology Department, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Department of Chemistry, Faculty of Sciences, University of Hradec Kralove, 50005, Hradec Kralove, Czech Republic
| | - Marian Valko
- Faculty of Chemical and Food Technology, Slovak University of Technology, Bratislava, 812 37, Slovakia.
| |
Collapse
|
46
|
Lin L, Wu Q, Lu F, Lei J, Zhou Y, Liu Y, Zhu N, Yu Y, Ning Z, She T, Hu M. Nrf2 signaling pathway: current status and potential therapeutic targetable role in human cancers. Front Oncol 2023; 13:1184079. [PMID: 37810967 PMCID: PMC10559910 DOI: 10.3389/fonc.2023.1184079] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 08/18/2023] [Indexed: 10/10/2023] Open
Abstract
Cancer is a borderless global health challenge that continues to threaten human health. Studies have found that oxidative stress (OS) is often associated with the etiology of many diseases, especially the aging process and cancer. Involved in the OS reaction as a key transcription factor, Nrf2 is a pivotal regulator of cellular redox state and detoxification. Nrf2 can prevent oxidative damage by regulating gene expression with antioxidant response elements (ARE) to promote the antioxidant response process. OS is generated with an imbalance in the redox state and promotes the accumulation of mutations and genome instability, thus associated with the establishment and development of different cancers. Nrf2 activation regulates a plethora of processes inducing cellular proliferation, differentiation and death, and is strongly associated with OS-mediated cancer. What's more, Nrf2 activation is also involved in anti-inflammatory effects and metabolic disorders, neurodegenerative diseases, and multidrug resistance. Nrf2 is highly expressed in multiple human body parts of digestive system, respiratory system, reproductive system and nervous system. In oncology research, Nrf2 has emerged as a promising therapeutic target. Therefore, certain natural compounds and drugs can exert anti-cancer effects through the Nrf2 signaling pathway, and blocking the Nrf2 signaling pathway can reduce some types of tumor recurrence rates and increase sensitivity to chemotherapy. However, Nrf2's dual role and controversial impact in cancer are inevitable consideration factors when treating Nrf2 as a therapeutic target. In this review, we summarized the current state of biological characteristics of Nrf2 and its dual role and development mechanism in different tumor cells, discussed Keap1/Nrf2/ARE signaling pathway and its downstream genes, elaborated the expression of related signaling pathways such as AMPK/mTOR and NF-κB. Besides, the main mechanism of Nrf2 as a cancer therapeutic target and the therapeutic strategies using Nrf2 inhibitors or activators, as well as the possible positive and negative effects of Nrf2 activation were also reviewed. It can be concluded that Nrf2 is related to OS and serves as an important factor in cancer formation and development, thus provides a basis for targeted therapy in human cancers.
Collapse
Affiliation(s)
- Li Lin
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Qing Wu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Feifei Lu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Jiaming Lei
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yanhong Zhou
- Department of Medical School of Facial Features, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Yifei Liu
- School of Biomedical Engineering, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Ni Zhu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - You Yu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Zhifeng Ning
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Tonghui She
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| | - Meichun Hu
- Key Laboratory of Environmental Related Diseases and One Health, School of Basic Medical Sciences, Xianning Medical College, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
47
|
Antal CE, Oh TG, Aigner S, Luo EC, Yee BA, Campos T, Tiriac H, Rothamel KL, Cheng Z, Jiao H, Wang A, Hah N, Lenkiewicz E, Lumibao JC, Truitt ML, Estepa G, Banayo E, Bashi S, Esparza E, Munoz RM, Diedrich JK, Sodir NM, Mueller JR, Fraser CR, Borazanci E, Propper D, Von Hoff DD, Liddle C, Yu RT, Atkins AR, Han H, Lowy AM, Barrett MT, Engle DD, Evan GI, Yeo GW, Downes M, Evans RM. A super-enhancer-regulated RNA-binding protein cascade drives pancreatic cancer. Nat Commun 2023; 14:5195. [PMID: 37673892 PMCID: PMC10482938 DOI: 10.1038/s41467-023-40798-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 08/10/2023] [Indexed: 09/08/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy in need of new therapeutic options. Using unbiased analyses of super-enhancers (SEs) as sentinels of core genes involved in cell-specific function, here we uncover a druggable SE-mediated RNA-binding protein (RBP) cascade that supports PDAC growth through enhanced mRNA translation. This cascade is driven by a SE associated with the RBP heterogeneous nuclear ribonucleoprotein F, which stabilizes protein arginine methyltransferase 1 (PRMT1) to, in turn, control the translational mediator ubiquitin-associated protein 2-like. All three of these genes and the regulatory SE are essential for PDAC growth and coordinately regulated by the Myc oncogene. In line with this, modulation of the RBP network by PRMT1 inhibition reveals a unique vulnerability in Myc-high PDAC patient organoids and markedly reduces tumor growth in male mice. Our study highlights a functional link between epigenetic regulation and mRNA translation and identifies components that comprise unexpected therapeutic targets for PDAC.
Collapse
Affiliation(s)
- Corina E Antal
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tae Gyu Oh
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73117, USA
| | - Stefan Aigner
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - En-Ching Luo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Brian A Yee
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tania Campos
- The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK
| | - Hervé Tiriac
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA
- Department of Surgery, Division of Surgical Oncology, University of California San Diego, La Jolla, CA, 92037, USA
| | - Katherine L Rothamel
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Zhang Cheng
- Center for Epigenomics, University of California San Diego, La Jolla, CA, 92037, USA
| | - Henry Jiao
- Center for Epigenomics, University of California San Diego, La Jolla, CA, 92037, USA
| | - Allen Wang
- Center for Epigenomics, University of California San Diego, La Jolla, CA, 92037, USA
| | - Nasun Hah
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | | | - Jan C Lumibao
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Morgan L Truitt
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Gabriela Estepa
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Ester Banayo
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Senada Bashi
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Edgar Esparza
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA
- Department of Surgery, Division of Surgical Oncology, University of California San Diego, La Jolla, CA, 92037, USA
| | - Ruben M Munoz
- Molecular Medicine Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - Jolene K Diedrich
- Mass Spectrometry Core for Proteomics and Metabolomics, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Nicole M Sodir
- The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK
- Genentech, Department of Translational Oncology, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Jasmine R Mueller
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| | - Cory R Fraser
- HonorHealth Research Institute, Scottsdale, AZ, 85258, USA
- Scottsdale Pathology Associates, Scottsdale, AZ, 85260, USA
| | - Erkut Borazanci
- Molecular Medicine Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
- HonorHealth Research Institute, Scottsdale, AZ, 85258, USA
| | - David Propper
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, USA
| | - Daniel D Von Hoff
- Molecular Medicine Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
- HonorHealth Research Institute, Scottsdale, AZ, 85258, USA
| | - Christopher Liddle
- Storr Liver Centre, Westmead Institute for Medical Research and Sydney Medical School, University of Sydney, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Ruth T Yu
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Annette R Atkins
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Haiyong Han
- Molecular Medicine Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - Andrew M Lowy
- The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK
- Department of Surgery, Division of Surgical Oncology, University of California San Diego, La Jolla, CA, 92037, USA
| | - Michael T Barrett
- Molecular Medicine Division, Translational Genomics Research Institute, Phoenix, AZ, 85004, USA
| | - Dannielle D Engle
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Gerard I Evan
- The Francis Crick Institute, 1 Midland Rd, London, NW1 1AT, UK
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA, 92037, USA
| | - Michael Downes
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| | - Ronald M Evans
- Gene Expression Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| |
Collapse
|
48
|
Shakya A, Liu P, Godek J, McKee NW, Dodson M, Anandhan A, Ooi A, Garcia JGN, Costa M, Chapman E, Zhang DD. The NRF2-p97-NRF2 negative feedback loop. Redox Biol 2023; 65:102839. [PMID: 37573837 PMCID: PMC10428046 DOI: 10.1016/j.redox.2023.102839] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 07/15/2023] [Accepted: 08/04/2023] [Indexed: 08/15/2023] Open
Abstract
p97 is a ubiquitin-targeted ATP-dependent segregase that regulates proteostasis, in addition to a variety of other cellular functions. Previously, we demonstrated that p97 negatively regulates NRF2 by extracting ubiquitylated NRF2 from the KEAP1-CUL3-RBX1 E3 ubiquitin ligase complex, facilitating proteasomal destruction. In the current study, we identified p97 as an NRF2-target gene that contains a functional ARE, indicating the presence of an NRF2-p97-NRF2 negative feedback loop that maintains redox homeostasis. Using CRISPR/Cas9 genome editing, we generated endogenous p97 ARE-mutated BEAS-2B cell lines. These p97 ARE-mutated cell lines exhibit altered expression of p97 and NRF2, as well as a compromised response to NRF2 inducers. Importantly, we also found a positive correlation between NRF2 activation and p97 expression in human cancer patients. Finally, using chronic arsenic-transformed cell lines, we demonstrated a synergistic effect of NRF2 and p97 inhibition in killing cancer cells with high NRF2 and p97 expression. Our study suggests dual upregulation of NRF2 and p97 occurs in certain types of cancers, suggesting that inhibition of both NRF2 and p97 could be a promising treatment strategy for stratified cancer patients.
Collapse
Affiliation(s)
- Aryatara Shakya
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Pengfei Liu
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA; National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; International Joint Research Center on Cell Stress and Disease Diagnosis and Therapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jack Godek
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Nicholas W McKee
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Matthew Dodson
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Annadurai Anandhan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Aikseng Ooi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Joe G N Garcia
- The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, FL, 33458, USA
| | - Max Costa
- Departments of Environmental Medicine, and Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10010, USA
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA.
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
49
|
Wei YC, Zhu JY, Wu J, Yu S, Li W, Zhu MX, Liu TS, Cui YH, Li Q. Nestin overexpression reduces the sensitivity of gastric cancer cells to trastuzumab. J Gastrointest Oncol 2023; 14:1694-1706. [PMID: 37720426 PMCID: PMC10502550 DOI: 10.21037/jgo-22-1048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 07/21/2023] [Indexed: 09/19/2023] Open
Abstract
Background Trastuzumab (TRA) shows significant efficacy in patients with human epidermal growth factor receptor 2 (HER2)-positive gastric cancer (GC). While TRA can help treat HER2-positive breast cancer, TRA resistance is a key clinical challenge. Nestin reportedly regulates the cellular redox homeostasis in lung cancer. This study aimed at identifying the functions of Nestin on the TRA sensitivity of HER2-positive GC cells. Methods Real-time polymerase chain reaction (PCR) and Western blotting (WB) were performed to explore the association between the mRNA and protein expression profiles, respectively, of Nestin and the Keap1-Nrf2 pathway. The influence of Nestin overexpression on the in vitro sensitivity of GC cells to TRA was explored by Cell Counting Kit-8 (CCK-8) assay, colony formation assay, reactive oxygen species (ROS) detection, and flow cytometry. Results TRA treatment caused Nestin downregulation in two HER2-positive GC cell lines (MKN45 and NCI-N87). Nestin overexpression reduced the sensitivity of GC cells to TRA. The expression and activity of Nrf2 and relevant downstream antioxidant genes were increased by Nestin overexpression. Nestin overexpression also significantly suppressed TRA-induced apoptosis and ROS generation. In vivo tumor growth experiment with female BALB/c nude mice indicated that Nestin upregulation restored the tumor growth rate which was inhibited by TRA treatment. Conclusions Collectively, the inhibitory effect of Nestin on the TRA sensitivity of cells to TRA was confirmed in this study. These results imply that the antioxidant Nestin-Nrf2 axis may play a role in the mechanism underlying the resistance of GC cells to TRA.
Collapse
Affiliation(s)
- Yi-Chou Wei
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiang-Yi Zhu
- Department of Radiotherapy, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Wu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meng-Xuan Zhu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tian-Shu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue-Hong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qian Li
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
50
|
Lintern N, Smith AM, Jayne DG, Khaled YS. Photodynamic Stromal Depletion in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:4135. [PMID: 37627163 PMCID: PMC10453210 DOI: 10.3390/cancers15164135] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/13/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest solid malignancies, with a five-year survival of less than 10%. The resistance of the disease and the associated lack of therapeutic response is attributed primarily to its dense, fibrotic stroma, which acts as a barrier to drug perfusion and permits tumour survival and invasion. As clinical trials of chemotherapy (CT), radiotherapy (RT), and targeted agents have not been successful, improving the survival rate in unresectable PDAC remains an urgent clinical need. Photodynamic stromal depletion (PSD) is a recent approach that uses visible or near-infrared light to destroy the desmoplastic tissue. Preclinical evidence suggests this can resensitise tumour cells to subsequent therapies whilst averting the tumorigenic effects of tumour-stromal cell interactions. So far, the pre-clinical studies have suggested that PDT can successfully mediate the destruction of various stromal elements without increasing the aggressiveness of the tumour. However, the complexity of this interplay, including the combined tumour promoting and suppressing effects, poses unknowns for the clinical application of photodynamic stromal depletion in PDAC.
Collapse
Affiliation(s)
- Nicole Lintern
- School of Biomedical Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Andrew M. Smith
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds LS9 7TF, UK
| | - David G. Jayne
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds LS9 7TF, UK
| | - Yazan S. Khaled
- Leeds Institute of Medical Research, St James’s University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|