1
|
Jadhav A, Menon A, Gupta K, Singh N. Molecular and therapeutic insight into ER stress signalling in NSCLC. J Drug Target 2025; 33:877-886. [PMID: 39883064 DOI: 10.1080/1061186x.2025.2461105] [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: 01/21/2024] [Revised: 01/15/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
Endoplasmic Reticulum (ER) stress is intricately involved in cancer development, progression and response to chemotherapy. ER stress related genes might play an important role in predicting the prognosis in lung adenocarcinoma patients and may be manipulated to improve the treatment outcome and overall survival rate. In this review, we analysed the contribution of the three major ER stress pathways-IRE1, ATF6, and PERK-in lung cancer pathogenesis via modulation of tumour microenvironment (TME) and processes as metastasis, angiogenesis, apoptosis and N-glycosylation. Furthermore, we discuss the regulatory role of microRNAs in fine-tuning ER stress pathways in Non-Small Cell Lung Cancer (NSCLC). Our review also highlights various promising strategies to overcome chemoresistance by targeting ER stress pathways, offering new therapeutic opportunities.
Collapse
Affiliation(s)
- Aastha Jadhav
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Arjun Menon
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Kush Gupta
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar, Gujarat, India
| | - Neeru Singh
- Department of Biotechnology and Bioengineering, Institute of Advanced Research, Gandhinagar, Gujarat, India
| |
Collapse
|
2
|
Varone E, Retini M, Cherubini A, Chernorudskiy A, Marrazza A, Guidarelli A, Cagnotto A, Beeg M, Gobbi M, Fumagalli S, Bolis M, Guarrera L, Barbera MC, Grasselli C, Bleve A, Generali D, Milani M, Mari M, Salmona M, Piersanti G, Bottegoni G, Broggini M, Janssen-Heininger YMW, Cho J, Cantoni O, Zito E. Small molecule-mediated inhibition of the oxidoreductase ERO1A restrains aggressive breast cancer by impairing VEGF and PD-L1 in the tumor microenvironment. Cell Death Dis 2025; 16:105. [PMID: 39962052 PMCID: PMC11833095 DOI: 10.1038/s41419-025-07426-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/09/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
Cancer cells adapt to harsh environmental conditions by inducing the Unfolded Protein Response (UPR), of which ERO1A is a mediator. ERO1A aids protein folding by acting as a protein disulfide oxidase, and under cancer-related hypoxia conditions, it favors the folding of angiogenic VEGFA, leading tumor cells to thrive and spread. The upregulation of ERO1A in cancer cells, oppositely to the dispensability of ERO1A activity in healthy cells, renders ERO1A a perfect target for cancer therapy. Here, we report the upregulation of ERO1A in a cohort of aggressive triple-negative breast cancer (TNBC) patients in which ERO1A levels correlate with a higher risk of breast tumor recurrence and metastatic spread. For ERO1A target validation and therapy in TNBC, we designed new ERO1A inhibitors in a structure-activity campaign of the prototype EN460. Cell-based screenings showed that the presence of the Micheal acceptor in the compound is necessary to engage the cysteine 397 of ERO1A but not sufficient to set out the inhibitory effect on ERO1A. Indeed, the ERO1 inhibitor must adopt a non-coplanar rearrangement within the ERO1A binding site. I2 and I3, two new EN460 analogs with different phenyl-substituted moieties, efficiently inhibited ERO1A, blunting VEGFA secretion. Accordingly, in vitro assays to measure ERO1A engagement and inhibition confirmed that I2 and I3 bind ERO1A and restrain its activity with a IC50 in a low micromolar range. EN460, I2 and I3 triggered breast cancer cytotoxicity while specifically inhibiting ERO1A in a dose-dependent manner. I2 more efficiently impaired cancer-relevant features such as VEGFA secretion and related cell migration. I2 also acted on the tumor microenvironment and viability of xenografts and syngeneic TNBC. Thus, small molecule-mediated ERO1A pharmacological inhibition is feasible and promises to lead to effective therapy for the still incurable TNBC.
Collapse
Affiliation(s)
- Ersilia Varone
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Michele Retini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alessandro Cherubini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alexander Chernorudskiy
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang, China
| | - Alice Marrazza
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alfredo Cagnotto
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marten Beeg
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Marco Bolis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Bioinformatics Core Unit, Institute of Oncology Research (IOR), Bellinzona, Switzerland
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Chiara Grasselli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Augusto Bleve
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Daniele Generali
- U.O. Patologia Mammaria e Tumori Cerebrali, Azienda Socio-Sanitaria Territoriale, Cremona, Italia
| | - Manuela Milani
- U.O. Patologia Mammaria e Tumori Cerebrali, Azienda Socio-Sanitaria Territoriale, Cremona, Italia
| | - Michele Mari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mario Salmona
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giovanni Piersanti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Giovanni Bottegoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | - Massimo Broggini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Yvonne M W Janssen-Heininger
- Departments of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
3
|
Zhao WB, Sheng R. The correlation between mitochondria-associated endoplasmic reticulum membranes (MAMs) and Ca 2+ transport in the pathogenesis of diseases. Acta Pharmacol Sin 2025; 46:271-291. [PMID: 39117969 PMCID: PMC11756407 DOI: 10.1038/s41401-024-01359-9] [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: 03/18/2024] [Accepted: 07/16/2024] [Indexed: 08/10/2024]
Abstract
Mitochondria and the endoplasmic reticulum (ER) are vital organelles that influence various cellular physiological and pathological processes. Recent evidence shows that about 5%-20% of the mitochondrial outer membrane is capable of forming a highly dynamic physical connection with the ER, maintained at a distance of 10-30 nm. These interconnections, known as MAMs, represent a relatively conserved structure in eukaryotic cells, acting as a critical platform for material exchange between mitochondria and the ER to maintain various aspects of cellular homeostasis. Particularly, ER-mediated Ca2+ release and recycling are intricately associated with the structure and functionality of MAMs. Thus, MAMs are integral in intracellular Ca2+ transport and the maintenance of Ca2+ homeostasis, playing an essential role in various cellular activities including metabolic regulation, signal transduction, autophagy, and apoptosis. The disruption of MAMs observed in certain pathologies such as cardiovascular and neurodegenerative diseases as well as cancers leads to a disturbance in Ca2+ homeostasis. This imbalance potentially aggravates pathological alterations and disease progression. Consequently, a thorough understanding of the link between MAM-mediated Ca2+ transport and these diseases could unveil new perspectives and therapeutic strategies. This review focuses on the changes in MAMs function during disease progression and their implications in relation to MAM-associated Ca2+ transport.
Collapse
Affiliation(s)
- Wen-Bin Zhao
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
4
|
Liu S, Zhang X, Wang W, Li X, Sun X, Zhao Y, Wang Q, Li Y, Hu F, Ren H. Metabolic reprogramming and therapeutic resistance in primary and metastatic breast cancer. Mol Cancer 2024; 23:261. [PMID: 39574178 PMCID: PMC11580516 DOI: 10.1186/s12943-024-02165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024] Open
Abstract
Metabolic alterations, a hallmark of cancer, enable tumor cells to adapt to their environment by modulating glucose, lipid, and amino acid metabolism, which fuels rapid growth and contributes to treatment resistance. In primary breast cancer, metabolic shifts such as the Warburg effect and enhanced lipid synthesis are closely linked to chemotherapy failure. Similarly, metastatic lesions often display distinct metabolic profiles that not only sustain tumor growth but also confer resistance to targeted therapies and immunotherapies. The review emphasizes two major aspects: the mechanisms driving metabolic resistance in both primary and metastatic breast cancer, and how the unique metabolic environments in metastatic sites further complicate treatment. By targeting distinct metabolic vulnerabilities at both the primary and metastatic stages, new strategies could improve the efficacy of existing therapies and provide better outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Shan Liu
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xingda Zhang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenzheng Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Sun
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yuqian Zhao
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qi Wang
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yingpu Li
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Fangjie Hu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| | - He Ren
- Department of oncological surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
5
|
Voronkova MA, Johnson B, Gandhi N, Koomen JM, Patrick M, Shanthi Bhupathi S, Wu VM, Elliott A, Vanderwalde A, Halmos B, Hazlehurst LA. ERO1A levels are a prognostic indicator in EGFR mutated non small cell lung cancer. NPJ Precis Oncol 2024; 8:250. [PMID: 39496753 PMCID: PMC11535241 DOI: 10.1038/s41698-024-00736-1] [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: 04/09/2024] [Accepted: 10/14/2024] [Indexed: 11/06/2024] Open
Abstract
We have identified endoplasmic reticulum oxidoreductase 1 alpha (ERO1A) as a poor prognostic indicator in epidermal growth factor receptor (EGFR)-mutated non-small cell lung cancer (EGFRMUT-NSCLC). In addition, comparison of high versus low ERO1A expression among cohorts of EGFRMUT-NSCLC primary samples revealed that ERO1A expression correlated with increased expression of proteins that regulate secretion. Using the CPTAC proteomic data set in lung adenocarcinoma we found that high ERO1A protein expression correlated with both extracellular matrix and matrix modifying enzymes. In this report, we found that ablating ERO1A expression was a determinant of clonogenicity, tumor sphere formation, spheroid growth and growth in vivo, as well as response to Osimertinib. We validated that ERO1A-knockout EGFRMUT-LUAD cell lines demonstrated a reduction in secretion of both laminin gamma 2 (LAMC2) and the collagen modifying enzyme lysyl oxidase-like 2 (LOXL2). Our work supports the role of ERO1A in modulating the tumor microenvironment that is likely to contribute to tumor progression.
Collapse
Affiliation(s)
- M A Voronkova
- West Virginia University Cancer Institute, Morgantown, WV, USA
| | - B Johnson
- West Virginia University Cancer Institute, Morgantown, WV, USA
| | - N Gandhi
- Caris Life Sciences, Phoenix, AZ, USA
| | - J M Koomen
- Molecular Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - M Patrick
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA
| | | | - V M Wu
- West Virginia University Cancer Institute, Morgantown, WV, USA
| | - A Elliott
- Caris Life Sciences, Phoenix, AZ, USA
| | | | - B Halmos
- Montefiore Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - L A Hazlehurst
- West Virginia University Cancer Institute, Morgantown, WV, USA.
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA.
- Department of Medical Oncology, West Virginia University School of Medicine, Morgantown, WV, USA.
| |
Collapse
|
6
|
Guo H, Li Y, Wang S, Yang Y, Xu T, Zhao J, Wang J, Zuo W, Wang P, Zhao G, Wang H, Hou W, Dong H, Cai Y. Dysfunction of astrocytic glycophagy exacerbates reperfusion injury in ischemic stroke. Redox Biol 2024; 74:103234. [PMID: 38861834 PMCID: PMC11215420 DOI: 10.1016/j.redox.2024.103234] [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: 05/05/2024] [Revised: 05/27/2024] [Accepted: 06/06/2024] [Indexed: 06/13/2024] Open
Abstract
Glycophagy has evolved from an alternative glycogen degradation pathway into a multifaceted pivot to regulate cellular metabolic hemostasis in peripheral tissues. However, the pattern of glycophagy in the brain and its potential therapeutic impact on ischemic stroke remain unknown. Here, we observed that the dysfunction of astrocytic glycophagy was caused by the downregulation of the GABA type A receptor-associated protein like 1 (GABARAPL1) during reperfusion in ischemic stroke patients and mice. PI3K-Akt pathway activation is involved in driving GABARAPL1 downregulation during cerebral reperfusion. Moreover, glycophagy dysfunction-induced glucosamine deficiency suppresses the nuclear translocation of specificity protein 1 and TATA binding protein, the transcription factors for GABARAPL1, by decreasing their O-GlcNAcylation levels, and accordingly feedback inhibits GABARAPL1 in astrocytes during reperfusion. Restoring astrocytic glycophagy by overexpressing GABARAPL1 decreases DNA damage and oxidative injury in astrocytes and improves the survival of surrounding neurons during reperfusion. In addition, a hypocaloric diet in the acute phase after cerebral reperfusion can enhance astrocytic glycophagic flux and accelerate neurological recovery. In summary, glycophagy in the brain links autophagy, metabolism, and epigenetics together, and glycophagy dysfunction exacerbates reperfusion injury after ischemic stroke.
Collapse
Affiliation(s)
- Haiyun Guo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yumeng Li
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shiquan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yongheng Yang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tiantian Xu
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jianshuai Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jin Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wenqiang Zuo
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Pengju Wang
- The State Key Laboratory of Cancer Biology, Department of Immunology, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Guangchao Zhao
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huaning Wang
- Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Yanhui Cai
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China; Department of Psychiatry, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
7
|
Wang Z, Zong H, Liu W, Lin W, Sun A, Ding Z, Chen X, Wan X, Liu Y, Hu Z, Zhang H, Li H, Liu Y, Li D, Zhang S, Zha X. Augmented ERO1α upon mTORC1 activation induces ferroptosis resistance and tumor progression via upregulation of SLC7A11. J Exp Clin Cancer Res 2024; 43:112. [PMID: 38610018 PMCID: PMC11015652 DOI: 10.1186/s13046-024-03039-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND The dysregulated mechanistic target of rapamycin complex 1 (mTORC1) signaling plays a critical role in ferroptosis resistance and tumorigenesis. However, the precise underlying mechanisms still need to be fully understood. METHODS Endoplasmic reticulum oxidoreductase 1 alpha (ERO1α) expression in mTORC1-activated mouse embryonic fibroblasts, cancer cells, and laryngeal squamous cell carcinoma (LSCC) clinical samples was examined by quantitative real-time PCR (qRT-PCR), western blotting, immunofluorescence (IF), and immunohistochemistry. Extensive in vitro and in vivo experiments were carried out to determine the role of ERO1α and its downstream target, member 11 of the solute carrier family 7 (SLC7A11), in mTORC1-mediated cell proliferation, angiogenesis, ferroptosis resistance, and tumor growth. The regulatory mechanism of ERO1α on SLC7A11 was investigated via RNA-sequencing, a cytokine array, an enzyme-linked immunosorbent assay, qRT-PCR, western blotting, IF, a luciferase reporter assay, and a chromatin immunoprecipitation assay. The combined therapeutic effect of ERO1α inhibition and the ferroptosis inducer imidazole ketone erastin (IKE) on mTORC1-activated cells was evaluated using cell line-derived xenografts, LSCC organoids, and LSCC patient-derived xenograft models. RESULTS ERO1α is a functional downstream target of mTORC1. Elevated ERO1α induced ferroptosis resistance and exerted pro-oncogenic roles in mTORC1-activated cells via upregulation of SLC7A11. Mechanically, ERO1α stimulated the transcription of SLC7A11 by activating the interleukin-6 (IL-6)/signal transducer and activator of transcription 3 (STAT3) pathway. Moreover, ERO1α inhibition combined with treatment using the ferroptosis inducer IKE exhibited synergistic antitumor effects on mTORC1-activated tumors. CONCLUSIONS The ERO1α/IL-6/STAT3/SLC7A11 pathway is crucial for mTORC1-mediated ferroptosis resistance and tumor growth, and combining ERO1α inhibition with ferroptosis inducers is a novel and effective treatment for mTORC1-related tumors.
Collapse
Affiliation(s)
- Zixi Wang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China
- Children's Hospital of Fudan University, National Children's Medical Center, And Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Huaiyuan Zong
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China
| | - Weiwei Liu
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Wei Lin
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Anjiang Sun
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China
| | - Zhao Ding
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xu Chen
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China
| | - Xiaofeng Wan
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, China
| | - Yanyan Liu
- Department of Thyroid and Breast Surgery, Hefei First People's Hospital, Hefei, 230061, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences and School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Hongwu Li
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Anhui Public Health Clinical Center, Hefei, 230011, China
| | - Yehai Liu
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Dapeng Li
- Department of Otorhinolaryngology, Head & Neck Surgery, The Affiliated Bozhou Hospital of Anhui Medical University, No. 616 Duzhong Road, Bozhou, 236800, Anhui Province, China.
| | - Sumei Zhang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China.
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, No. 81 Meishan Road, Hefei, 230032, Anhui Province, China.
- Department of Otorhinolaryngology, Head & Neck Surgery, The Affiliated Bozhou Hospital of Anhui Medical University, No. 616 Duzhong Road, Bozhou, 236800, Anhui Province, China.
| |
Collapse
|
8
|
Germani S, Van Ho AT, Cherubini A, Varone E, Chernorudskiy A, Renna GM, Fumagalli S, Gobbi M, Lucchetti J, Bolis M, Guarrera L, Craparotta I, Rastelli G, Piccoli G, de Napoli C, Nogara L, Poggio E, Brini M, Cattaneo A, Bachi A, Simmen T, Calì T, Quijano-Roy S, Boncompagni S, Blaauw B, Ferreiro A, Zito E. SEPN1-related myopathy depends on the oxidoreductase ERO1A and is druggable with the chemical chaperone TUDCA. Cell Rep Med 2024; 5:101439. [PMID: 38402623 PMCID: PMC10982971 DOI: 10.1016/j.xcrm.2024.101439] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 12/06/2023] [Accepted: 01/31/2024] [Indexed: 02/27/2024]
Abstract
Selenoprotein N (SEPN1) is a protein of the endoplasmic reticulum (ER) whose inherited defects originate SEPN1-related myopathy (SEPN1-RM). Here, we identify an interaction between SEPN1 and the ER-stress-induced oxidoreductase ERO1A. SEPN1 and ERO1A, both enriched in mitochondria-associated membranes (MAMs), are involved in the redox regulation of proteins. ERO1A depletion in SEPN1 knockout cells restores ER redox, re-equilibrates short-range MAMs, and rescues mitochondrial bioenergetics. ERO1A knockout in a mouse background of SEPN1 loss blunts ER stress and improves multiple MAM functions, including Ca2+ levels and bioenergetics, thus reversing diaphragmatic weakness. The treatment of SEPN1 knockout mice with the ER stress inhibitor tauroursodeoxycholic acid (TUDCA) mirrors the results of ERO1A loss. Importantly, muscle biopsies from patients with SEPN1-RM exhibit ERO1A overexpression, and TUDCA-treated SEPN1-RM patient-derived primary myoblasts show improvement in bioenergetics. These findings point to ERO1A as a biomarker and a viable target for intervention and to TUDCA as a pharmacological treatment for SEPN1-RM.
Collapse
Affiliation(s)
- Serena Germani
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Andrew Tri Van Ho
- Basic and Translational Myology Laboratory, Université Paris, BFA, UMR 8251, CNRS, 75013 Paris, France
| | | | - Ersilia Varone
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | | | | | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Jacopo Lucchetti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Bolis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Bioinformatics Core Unit, Institute of Oncology Research (IOR), 6500 Bellinzona, Switzerland
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Giorgia Rastelli
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. D'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Giorgia Piccoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Cosimo de Napoli
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Leonardo Nogara
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Department of Pharmaceutical Sciences, University of Padova, Padova, Italy
| | - Elena Poggio
- Department of Biology, University of Padova, Padova, Italy
| | - Marisa Brini
- Department of Pharmaceutical Sciences, University of Padova, Padova, Italy; Department of Biology, University of Padova, Padova, Italy; Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy
| | | | - Angela Bachi
- IFOM-ETS AIRC Institute of Molecular Oncology, Milan, Italy
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Tito Calì
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy; Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Susana Quijano-Roy
- APHP-Université Paris-Saclay, Reference Center for Neuromuscular Disorders Nord-Est-Ile de France, FILNEMUS, ERN-Euro-NMD, Creteil, France; Pediatric Neurology and ICU Department, DMU Santé Enfant Adolescent (SEA), Raymond Poincaré University Hospital, Garches, France
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. D'Annunzio of Chieti-Pescara, 66100 Chieti, Italy
| | - Bert Blaauw
- Department of Biomedical Sciences, University of Padua, Padua, Italy; Venetian Institute of Molecular Medicine, Padova, Italy.
| | - Ana Ferreiro
- Basic and Translational Myology Laboratory, Université Paris, BFA, UMR 8251, CNRS, 75013 Paris, France; APHP, Reference Center for Neuromuscular Disorders Nord-Est-Ile de France, Neuromyology Department, Groupe Hospitalier Pitié-Salpêtrière, Paris, France.
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
9
|
Chen P, Sharma A, Weiher H, Schmidt-Wolf IGH. Biological mechanisms and clinical significance of endoplasmic reticulum oxidoreductase 1 alpha (ERO1α) in human cancer. J Exp Clin Cancer Res 2024; 43:71. [PMID: 38454454 PMCID: PMC10921667 DOI: 10.1186/s13046-024-02990-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 02/21/2024] [Indexed: 03/09/2024] Open
Abstract
A firm link between endoplasmic reticulum (ER) stress and tumors has been wildly reported. Endoplasmic reticulum oxidoreductase 1 alpha (ERO1α), an ER-resident thiol oxidoreductase, is confirmed to be highly upregulated in various cancer types and associated with a significantly worse prognosis. Of importance, under ER stress, the functional interplay of ERO1α/PDI axis plays a pivotal role to orchestrate proper protein folding and other key processes. Multiple lines of evidence propose ERO1α as an attractive potential target for cancer treatment. However, the unavailability of specific inhibitor for ERO1α, its molecular inter-relatedness with closely related paralog ERO1β and the tightly regulated processes with other members of flavoenzyme family of enzymes, raises several concerns about its clinical translation. Herein, we have provided a detailed description of ERO1α in human cancers and its vulnerability towards the aforementioned concerns. Besides, we have discussed a few key considerations that may improve our understanding about ERO1α in tumors.
Collapse
Affiliation(s)
- Peng Chen
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, 3127, Bonn, Germany
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, 3127, Bonn, Germany
- Department of Neurosurgery, University Hospital Bonn, 53127, Bonn, Germany
| | - Hans Weiher
- Department of Applied Natural Sciences, Bonn-Rhein-Sieg University of Applied Sciences, 53359, Rheinbach, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn, 3127, Bonn, Germany.
| |
Collapse
|
10
|
Zito E, Guarrera L, Janssen-Heininger YMW. Fingerprint of the oxido-reductase ERO1: A protein disulfide bond producer and supporter of cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189027. [PMID: 38007054 PMCID: PMC11046445 DOI: 10.1016/j.bbcan.2023.189027] [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: 09/26/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/27/2023]
Abstract
Endoplasmic reticulum oxidoreductin 1 (ERO1) alpha (ERO1A) is an endoplasmic reticulum (ER)-localized protein disulfide oxidoreductase, involved in the disulfide bond formation of proteins. ERO1's activity in oxidative protein folding is redundant in higher eukaryotes and its loss is well compensated. Although it is dispensable in non-cancer cells, high ERO1 levels are seen with different cancers and predict their malignant phenotype. ERO1 fosters tumor aggressiveness and the response to drug therapy in hypoxic and highly metastatic tumors. It regulates vascular endothelial growth factor (VEGF) levels, oxidative folding and N-glycosylation in hypoxic conditions, boosting tumor fitness and angiogenesis on multiple levels. In addition, ERO1 regulates protein death ligand-1 (PD-L1) on tumors, interfering with the related immune surveillance mechanism, hence acting on the tumors' response to immune check-point inhibitors (ICI). This all points to inhibition of ERO1 as an effective pharmacological tool, selectively targeting tumors while sparing non-cancer cells from cytotoxicity. The critical discussion here closely examines the molecular basis for ERO1's involvement in tumors and ERO1 inhibition strategies for their treatment.
Collapse
Affiliation(s)
- Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Yvonne M W Janssen-Heininger
- Departments of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA.
| |
Collapse
|
11
|
Bassot A, Chen J, Takahashi-Yamashiro K, Yap MC, Gibhardt CS, Le GNT, Hario S, Nasu Y, Moore J, Gutiérrez T, Mina L, Mast H, Moses A, Bhat R, Ballanyi K, Lemieux H, Sitia R, Zito E, Bogeski I, Campbell RE, Simmen T. The endoplasmic reticulum kinase PERK interacts with the oxidoreductase ERO1 to metabolically adapt mitochondria. Cell Rep 2023; 42:111899. [PMID: 36586409 DOI: 10.1016/j.celrep.2022.111899] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 10/04/2022] [Accepted: 12/08/2022] [Indexed: 12/31/2022] Open
Abstract
Endoplasmic reticulum (ER) homeostasis requires molecular regulators that tailor mitochondrial bioenergetics to the needs of protein folding. For instance, calnexin maintains mitochondria metabolism and mitochondria-ER contacts (MERCs) through reactive oxygen species (ROS) from NADPH oxidase 4 (NOX4). However, induction of ER stress requires a quick molecular rewiring of mitochondria to adapt to new energy needs. This machinery is not characterized. We now show that the oxidoreductase ERO1⍺ covalently interacts with protein kinase RNA-like ER kinase (PERK) upon treatment with tunicamycin. The PERK-ERO1⍺ interaction requires the C-terminal active site of ERO1⍺ and cysteine 216 of PERK. Moreover, we show that the PERK-ERO1⍺ complex promotes oxidization of MERC proteins and controls mitochondrial dynamics. Using proteinaceous probes, we determined that these functions improve ER-mitochondria Ca2+ flux to maintain bioenergetics in both organelles, while limiting oxidative stress. Therefore, the PERK-ERO1⍺ complex is a key molecular machinery that allows quick metabolic adaptation to ER stress.
Collapse
Affiliation(s)
- Arthur Bassot
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada
| | - Junsheng Chen
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada
| | | | - Megan C Yap
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada
| | - Christine Silvia Gibhardt
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Giang N T Le
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Saaya Hario
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yusuke Nasu
- Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Jack Moore
- Alberta Proteomics and Mass Spectrometry Facility, University of Alberta, 4096 Katz Research Building, Edmonton AB T6G2E1, Canada
| | - Tomas Gutiérrez
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada
| | - Lucas Mina
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada
| | - Heather Mast
- Faculty Saint-Jean, Department of Medicine, Faculty of Medicine and Dentistry, Edmonton, AB T6G2H7, Canada
| | - Audric Moses
- Department of Pediatrics, Edmonton, AB T6G2H7, Canada
| | - Rakesh Bhat
- Precision Biolaboratories, St. Albert, AB T8N 5A7, Canada
| | - Klaus Ballanyi
- Department of Physiology, University of Alberta, Edmonton, AB T6G2H7, Canada
| | - Hélène Lemieux
- Faculty Saint-Jean, Department of Medicine, Faculty of Medicine and Dentistry, Edmonton, AB T6G2H7, Canada
| | - Roberto Sitia
- Division of Genetics and Cell Biology, Università Vita-Salute IRCCS Ospedale San Raffaele, 20132 Milano, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri, 20156 Milano, Italy; Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino PU, Italy
| | - Ivan Bogeski
- Molecular Physiology, Institute of Cardiovascular Physiology, University Medical Center, Georg-August-University, Göttingen, Germany
| | - Robert E Campbell
- Department of Chemistry, University of Alberta, Edmonton, AB T6G 2G2, Canada; Department of Chemistry, Graduate School of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, Edmonton, AB T6G 2G2, Canada.
| |
Collapse
|
12
|
Cherubini A, Zito E. ER stress as a trigger of UPR and ER-phagy in cancer growth and spread. Front Oncol 2022; 12:997235. [PMID: 36408145 PMCID: PMC9667062 DOI: 10.3389/fonc.2022.997235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/12/2022] [Indexed: 01/25/2023] Open
Abstract
Tumors can survive environmental and metabolic stress by triggering homeostatic responses that re-establish the pre-stress status and permit them to grow and thrive. The endoplasmic reticulum (ER) is the organelle where proteins undergo post-translational modifications and are folded and exported to the secretory pathway. Its environment and activity are therefore fundamental for proteostasis, i.e., the plethora of mechanisms controlling protein formation, folding, degradation, and secretion, needed to assure protein balance and cellular health. In different tumor-related conditions, such as after the activation of oncogenes or under hypoxia and nutrient deprivation, the ER experiences stress, triggered by a high load of proteins to be folded compared to the limited folding capacity of the organelle. As a consequence, three ER membrane sensors and the related unfolded protein response (UPR) are activated. The UPR comprises a complex interconnection between signal transduction pathways that promote a homeostatic response that acts by increasing the amount of protein chaperones and of proteins involved in ER-associated protein degradation (ERAD) on one hand and attenuating protein translation on the other. ER-phagy, literally "eating" the ER, is part of another homeostatic response consisting of the clearance of non-functional ER portions including misfolded proteins. This response is also activated by a set of dedicated ER-phagy receptors after ER stimuli, which overlap the stimuli generating ER stress. Thus, the UPR and ER-phagy are two closely related homeostatic mechanisms that cooperate in re-establishing ER homeostasis. However, while the role of the UPR in favoring cancer growth and thriving by promoting angiogenesis, metastasis, chemotherapy resistance, and epithelial-to-mesenchymal transition is consolidated, that of ER-phagy is still in its infancy. This essay provides an overview of emerging concepts on ER stress, the UPR, and ER-phagy and their crosstalk in tumorigenesis. We also critically review new findings on their pharmacological targeting in cancer.
Collapse
Affiliation(s)
- Alessandro Cherubini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy,Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy,Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy,*Correspondence: Ester Zito,
| |
Collapse
|