1
|
Wu Y, Wang G, Yang R, Zhou D, Chen Q, Wu Q, Chen B, Yuan L, Qu N, Wang H, Hassan M, Zhao Y, Liu M, Shen Z, Zhou W. Activation of PERK/eIF2α/ATF4 signaling inhibits ERα expression in breast cancer. Neoplasia 2025; 65:101165. [PMID: 40252311 PMCID: PMC12023901 DOI: 10.1016/j.neo.2025.101165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 04/09/2025] [Accepted: 04/09/2025] [Indexed: 04/21/2025]
Abstract
Approximately 70-80% of breast cancers rely on estrogen receptor alpha (ERα) for growth. The unfolded protein response (UPR), a cellular response to endoplasmic reticulum stress (ERS), is an important process crucial for oncogenic transformation. The effect of ERS on ERα expression and signaling remains incompletely elucidated. Here, we focused on the regulatory mechanisms of ERS on ERα expression in ER-positive breast cancer (ER+ BC). Our results demonstrate that ERα protein and mRNA levels in ER+ BC cells are considerably reduced by the ERS inducers thapsigargin (TG) and brefeldin A (BFA) via the PERK/eIF2α/ATF4 signaling pathway. ChIP-qPCR and luciferase reporter gene analysis revealed that ERS induction facilitated ATF4 binding to the ESR1 (the gene encoding ERα) promoter region, thereby suppressing ESR1 promoter activity and inhibiting ERα expression. Furthermore, selective activation of PERK signaling or ATF4 overexpression attenuated ERα expression and tumor cell growth both in vitro and in vivo. In conclusion, our results demonstrate that ERS suppresses ERα expression transcriptionally via the PERK/eIF2α/ATF4 signaling. Our study provides insights into the treatment of ER+ BC by targeting ERα signaling through selective activation of the PERK branch of the UPR.
Collapse
Affiliation(s)
- Yuanli Wu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, PR China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, PR China
| | - Gang Wang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, PR China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, PR China
| | - Ruixue Yang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, PR China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, PR China
| | - Duanfang Zhou
- Department of Pharmacy, Women and Children's Hospital of Chongqing Medical University/Chongqing Health Center for Women and Children, Chongqing, 401147, PR China
| | - Qingjuan Chen
- Department of Oncology, 3201 Hospital of Xi'an Jiaotong University Health Science Center, Hanzhong, 723000, Shaanxi, PR China
| | - Qiuya Wu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, PR China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, PR China
| | - Bo Chen
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, PR China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, PR China
| | - Lie Yuan
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, PR China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, PR China
| | - Na Qu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, PR China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, PR China
| | - Hongmei Wang
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, PR China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, PR China; Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, PR China
| | - Moustapha Hassan
- Experimental Cancer Medicine, Division of Biomolecular and Cellular Medicine (BCM), Department of Laboratory Medicine, Karolinska Institutet, Huddinge, 14186, Sweden
| | - Ying Zhao
- Experimental Cancer Medicine, Division of Biomolecular and Cellular Medicine (BCM), Department of Laboratory Medicine, Karolinska Institutet, Huddinge, 14186, Sweden
| | - Mingpu Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, PR China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, PR China
| | - Zhengze Shen
- Department of Pharmacy, Yongchuan Hospital of Chongqing Medical University, Chongqing, 402160, PR China.
| | - Weiying Zhou
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, PR China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing, 400016, PR China; Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing Medical University, Chongqing, 400016, PR China.
| |
Collapse
|
2
|
Cáceres E, Salazar P, Torbey MT, Wilson CM, Bragin DE, Selwyn RG, Divani AA. Tauroursodeoxycholic acid reduces brain lesion volume and cortical edema in a rat model of Traumatic brain injury. Brain Res 2025:149773. [PMID: 40490087 DOI: 10.1016/j.brainres.2025.149773] [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: 03/12/2025] [Revised: 05/27/2025] [Accepted: 06/06/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND Traumatic brain injury (TBI) continues to be a major cause of death and disability worldwide. Effective treatment options are limited, and randomized clinical data has been futile. OBJECTIVE In this study, we assessed the efficacy of tauroursodeoxycholic acid (TUDCA) in treating TBI using a controlled cortical impact injury model in male Wistar rats. METHODS Animals were randomized into two experimental groups: 1) TBI with sham treatment (Control, n = 12) and 2) TBI treated with intravenous infusion of 300 mg/kg TUDCA (TUDCA-treated group, n = 10) 30 min post-injury. Anxiety levels and magnetic resonance imaging studies were performed on days 1 and 7 post-injury. Animals were euthanized on day 7 for histopathological assessments. RESULTS On days 1 and 7, we observed a smaller brain lesion volume in the TUDCA-treated group compared with the Control group and greater cytotoxic edema on day 1 in the Control group. Elevated plus maze revealed a lower anxiety index for the TUDCA-treated group on day 7. CONCLUSION Our study showed that TBI rats treated with TUDCA at a hyperacute stage had a statistically significant reduction in lesion volume and improved levels of anxiety. However, a dose-response relationship and ideal therapeutic window still need to be determined. Future studies should consider a multiday therapy paradigm to identify the optimum intervention frequency in a mixed-gender.
Collapse
Affiliation(s)
- Eder Cáceres
- Unisabana Center for Translational Science, School of Medicine, Universidad de La Sabana, Chía, Colombia; Bioscience PhD. School of Engineering, Universidad de La Sabana, Chía, Colombia; Department of Critical Care, Clínica Universidad de La Sabana, Chía, Colombia
| | | | - Michel T Torbey
- Department of Neurology, University of Oklahoma, Oklahoma City, OH, USA
| | - Colin M Wilson
- Department of Radiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Denis E Bragin
- Lovelace Biomedical Research Institute, 2425 Ridgecrest Dr. SE, Albuquerque, NM, USA; Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Reed G Selwyn
- Department of Radiology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Afshin A Divani
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM, USA.
| |
Collapse
|
3
|
Yang L, Xue R, Yang C, Lv Y, Li S, Xiang W, Guo X, Zhou J. Endoplasmic reticulum stress on glioblastoma: Tumor growth promotion and immunosuppression. Int Immunopharmacol 2025; 157:114806. [PMID: 40339490 DOI: 10.1016/j.intimp.2025.114806] [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: 09/22/2024] [Revised: 04/10/2025] [Accepted: 05/03/2025] [Indexed: 05/10/2025]
Abstract
Exogenous or endogenous factors such as hypoxia, nutritional deficiencies, acidic microenvironments and their own high metabolic demands usually lead to tumor endoplasmic reticulum dysfunction and trigger endoplasmic reticulum stress (ERS). ERS sensors intercept such stress signals, which subsequently initiate the unfolded protein response (UPR), enabling tumor cells to adapt robustly in the hostile environment. Many studies have found that the ERS response affects a variety of tumor-infiltrating immune cells and suppresses their anti-tumor responses through different mechanisms. Given that glioblastoma (GBM) are immunosuppressive "cold tumors" with a poor prognosis. This paper not only discusses the promotion of GBM growth by ERS response, but also reviews the mechanisms by which ERS response promotes an immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Luxia Yang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Ruifeng Xue
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Chaoge Yang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Yancheng Lv
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Shenjie Li
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Wei Xiang
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China
| | - Xiyuan Guo
- Public Center of Experimental Technology, The School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, China; Division of Clinical Chemistry, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand.
| | - Jie Zhou
- Department of Neurosurgery, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China; School of Clinical Medicine, Sichuan Clinical Research Center for Neurosurgery, Luzhou, Sichuan, China.
| |
Collapse
|
4
|
Yang J, Wang Y, Liu F, Zhang Y, Han F. Crosstalk between ferroptosis and endoplasmic reticulum stress: A potential target for ovarian cancer therapy (Review). Int J Mol Med 2025; 55:97. [PMID: 40314096 PMCID: PMC12045474 DOI: 10.3892/ijmm.2025.5538] [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: 01/27/2025] [Accepted: 04/08/2025] [Indexed: 05/03/2025] Open
Abstract
Ferroptosis is a unique mode of cell death driven by iron‑dependent phospholipid peroxidation, and its mechanism primarily involves disturbances in iron metabolism, imbalances in the lipid antioxidant system and accumulation of lipid peroxides. Protein processing, modification and folding in the endoplasmic reticulum (ER) are closely related regulatory processes that determine cell function, fate and survival. The uncontrolled proliferative capacity of malignant cells generates an unfavorable microenvironment characterized by high metabolic demand, hypoxia, nutrient deprivation and acidosis, which promotes the accumulation of misfolded or unfolded proteins in the ER, leading to ER stress (ERS). Ferroptosis and ERS share common pathways in several diseases, and the two interact to affect cell survival and death. Additionally, cell death pathways are not linear signaling cascades, and different pathways of cell death may be interrelated at multiple levels. Ferroptosis and ERS in ovarian cancer (OC) have attracted increasing research interest; however, both are discussed separately regarding OC. The present review aims to summarize the associations and potential links between ferroptosis and ERS, aiming to provide research references for the development of therapeutic approaches for the management of OC.
Collapse
Affiliation(s)
- Jiaqi Yang
- Postgraduate School of Traditional Chinese Gynecology, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Yu Wang
- Postgraduate School of Traditional Chinese Gynecology, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Fangyuan Liu
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Yizhong Zhang
- Postgraduate School of Traditional Chinese Gynecology, Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| | - Fengjuan Han
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, Heilongjiang 150040, P.R. China
| |
Collapse
|
5
|
Rhine K, Li R, Kopalle HM, Rothamel K, Ge X, Epstein E, Mizrahi O, Madrigal AA, Her HL, Gomberg TA, Hermann A, Schwartz JL, Daniels AJ, Manor U, Ravits J, Signer RAJ, Bennett EJ, Yeo GW. Neuronal aging causes mislocalization of splicing proteins and unchecked cellular stress. Nat Neurosci 2025; 28:1174-1184. [PMID: 40456907 DOI: 10.1038/s41593-025-01952-z] [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] [Received: 04/02/2024] [Accepted: 03/14/2025] [Indexed: 06/11/2025]
Abstract
Aging is one of the most prominent risk factors for neurodegeneration, yet the molecular mechanisms underlying the deterioration of old neurons are mostly unknown. To efficiently study neurodegeneration in the context of aging, we transdifferentiated primary human fibroblasts from aged healthy donors directly into neurons, which retained their aging hallmarks, and we verified key findings in aged human and mouse brain tissue. Here we show that aged neurons are broadly depleted of RNA-binding proteins, especially spliceosome components. Intriguingly, splicing proteins-like the dementia- and ALS-associated protein TDP-43-mislocalize to the cytoplasm in aged neurons, which leads to widespread alternative splicing. Cytoplasmic spliceosome components are typically recruited to stress granules, but aged neurons suffer from chronic cellular stress that prevents this sequestration. We link chronic stress to the malfunctioning ubiquitylation machinery, poor HSP90α chaperone activity and the failure to respond to new stress events. Together, our data demonstrate that aging-linked deterioration of RNA biology is a key driver of poor resiliency in aged neurons.
Collapse
Affiliation(s)
- Kevin Rhine
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Rachel Li
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Hema M Kopalle
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Katherine Rothamel
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
- Center for RNA Technologies & Therapeutics, University of California San Diego, La Jolla, CA, USA
| | - Xuezhen Ge
- Department of Cell & Developmental Biology, University of California San Diego, La Jolla, CA, USA
| | - Elle Epstein
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Orel Mizrahi
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Assael A Madrigal
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Hsuan-Lin Her
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
- Bioinformatics & Systems Biology Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Trent A Gomberg
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Anita Hermann
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Joshua L Schwartz
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Amanda J Daniels
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Cell & Developmental Biology, University of California San Diego, La Jolla, CA, USA
- Division of Regenerative Medicine, Department of Medicine, Sanford Stem Cell Institute, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Uri Manor
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, USA
- Department of Cell & Developmental Biology, University of California San Diego, La Jolla, CA, USA
| | - John Ravits
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Robert A J Signer
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA
- Division of Regenerative Medicine, Department of Medicine, Sanford Stem Cell Institute, Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Eric J Bennett
- Department of Cell & Developmental Biology, University of California San Diego, La Jolla, CA, USA
| | - Gene W Yeo
- Department of Cellular & Molecular Medicine, University of California San Diego, La Jolla, CA, USA.
- Sanford Stem Cell Institute Innovation Center and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, USA.
- Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA.
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA.
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA.
- Center for RNA Technologies & Therapeutics, University of California San Diego, La Jolla, CA, USA.
- Bioinformatics & Systems Biology Graduate Program, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Shukla M, Narayan M. Proteostasis and Its Role in Disease Development. Cell Biochem Biophys 2025; 83:1725-1741. [PMID: 39422790 PMCID: PMC12123047 DOI: 10.1007/s12013-024-01581-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
Proteostasis (protein homeostasis) refers to the general biological process that maintains the proper balance between the synthesis of proteins, their folding, trafficking, and degradation. It ensures proteins are functional, locally distributed, and appropriately folded inside cells. Genetic information enclosed in mRNA is translated into proteins. To ensure newly synthesized proteins take on the exact three-dimensional conformation, molecular chaperones assist in proper folding. Misfolded proteins can be refolded or targeted for elimination to stop aggregation. Cells utilize different degradation pathways, for instance, the ubiquitin-proteasome system, the autophagy-lysosome pathway, and the unfolded protein response, to degrade unwanted or damaged proteins. Quality control systems of the cell monitor the folding of proteins. These checkpoint mechanisms are aimed at degrading or refolding misfolded or damaged proteins. Under stress response pathways, such as heat shock response and unfolded protein response, which are triggered under conditions that perturb proteostasis, the capacity for folding is increased, and degradation pathways are activated to help cells handle stressful conditions. The deregulation of proteostasis is implicated in a variety of illnesses, comprising cancer, metabolic diseases, cardiovascular diseases, and neurological disorders. Therapeutic strategies with a deeper insight into the mechanism of proteostasis are crucial for the treatment of illnesses linked with proteostasis and to support cellular health. Thus, proteostasis is required not only for the maintenance of cellular homeostasis and function but also for proper protein function and prevention of injurious protein aggregation. In this review, we have covered the concept of proteostasis, its mechanism, and how disruptions to it can result in a number of disorders.
Collapse
Affiliation(s)
- Manisha Shukla
- Department of Biotechnology, Pandit S.N. Shukla University, Shahdol, Madhya Pradesh, India
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, University of Texas, El Paso, TX, USA.
| |
Collapse
|
7
|
Hendershot LM. A BiP-centric View of Endoplasmic Reticulum Functions and of My Career. J Mol Biol 2025; 437:169052. [PMID: 40024435 DOI: 10.1016/j.jmb.2025.169052] [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: 12/31/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025]
Abstract
After completing my post-doctoral training at the University of Alabama, Birmingham and a brief period on the faculty there, I joined the Department of Tumor Cell Biology at St. Jude Children's Research Hospital in 1987 as an Assistant Member and started my independent research program. For the following 37 years, I led a relatively small basic research group comprised at various times of post-doctoral fellows, graduate students, undergraduate students, and research technicians; many of whom I am still in contact. Last year I closed the lab and transitioned to an emeritus position at St. Jude. I continue to maintain several research collaborations covering areas of research that have long been dear to my heart. My post-doctoral studies on BiP revealed that it controlled immunoglobulin assembly and transport, and as such, played a critical role in the fidelity of the immune response. My lab continued to define BiP's functions in protein folding and subunit assembly, as well as, in degradation of proteins that failed to mature properly using biochemical, cell-based, and biophysical analyses. Several ER localized co-factors that regulate the activity of BiP and allow it to contribute to its multiple ER functions were identified by our group. These include DnaJ family members and nucleotide change factors. Through a variety of collaborative studies, we pursued BiP's functions in maintaining the permeability barrier of the translocon, contributing to ER calcium stores, and regulating the up-stream transducers of the UPR, a stress response that is activated by the accumulation of unfolded proteins in the ER.
Collapse
Affiliation(s)
- Linda M Hendershot
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States.
| |
Collapse
|
8
|
Hermanson JN, Barny LA, Plate L. Development of an Adaptive, Economical, and Easy-to-Use SP3-TMT Automated Sample Preparation Workflow for Quantitative Proteomics. J Proteome Res 2025. [PMID: 40423998 DOI: 10.1021/acs.jproteome.5c00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Liquid handling robots have been developed to automate various steps of the bottom-up proteomics workflow, however, protocols for the generation of isobarically labeled peptides remain limited. Existing methods often require costly specialty devices and are constrained by fixed workflows. To address this, we developed a cost-effective, flexible, automated sample preparation protocol for TMT-labeled peptides using the Biomek i5 liquid handler (Beckman Coulter). Our approach leverages single-pot solid-phase-enhanced sample preparation with paramagnetic beads to streamline protein cleanup and digestion. The protocol also allows for adjustment of trypsin concentration and peptide-to-TMT ratio to increase throughput and reduce costs, respectively. We compared our automated and manual 18-plex TMT-Pro labeling workflows by monitoring select protein markers of the unfolded protein response in pharmacologically activatable, engineered cell lines. Overall, the automated protocol demonstrated equivalent performance in peptide and protein identifications, digestion and labeling efficiency, and an enhancement in the dynamic range of TMT quantifications. Compared to the manual method, the Biomek protocol significantly reduces hands-on time and minimizes sample handling errors. The 96-well format additionally allows for the number of TMT reactions to be scaled up quickly without a significant increase in user interaction. Our optimized automated workflow enhances throughput, reproducibility, and cost-effectiveness, making it a valuable tool for high-throughput proteomics studies.
Collapse
Affiliation(s)
- Jake N Hermanson
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37240-0002, United States
| | - Lea A Barny
- Program in Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee 37240-0002, United States
| | - Lars Plate
- Department of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37240-0002, United States
- Program in Chemical and Physical Biology, Vanderbilt University, Nashville, Tennessee 37240-0002, United States
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37240-0002, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| |
Collapse
|
9
|
Boyack I, Berlied A, Peterson C. A Potential Role for c-MYC in the Regulation of Meibocyte Cell Stress. Cells 2025; 14:709. [PMID: 40422212 PMCID: PMC12109776 DOI: 10.3390/cells14100709] [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: 03/29/2025] [Revised: 04/29/2025] [Accepted: 05/12/2025] [Indexed: 05/28/2025] Open
Abstract
The integrated stress response (ISR) is a key regulator of cell survival, promoting apoptosis through the effector protein CHOP in instances of prolonged or severe stress. The ISR's role in the initiation and progression of epithelial malignancies has been investigated; however, the ISR has not been evaluated in ocular adnexal sebaceous carcinoma (SebCA). Though uncommon, mortality rates of up to 40% have been reported, and the mechanisms underlying SebCA tumorigenesis remain unresolved; however, c-MYC upregulation has been documented. Our objective was to determine the role of MYC in modulating the ISR in the Meibomian gland. Human Meibomian gland epithelial cells (HMGECs) were subject to both pharmacologic and genetic manipulations of MYC expression. Cytotoxicity, proliferation, and changes in protein and gene expression were assessed. Conditionally MYC-overexpressing mice were subject to topical 4-hydroxytamoxifen (4-OHT) induction of the eyelids prior to tissue harvest for histology, immunohistochemistry, immunoblotting, and qPCR. MYC-inhibited HMGECs exhibited dose-dependent decreased proliferation, increased CHOP expression, and increased apoptosis. Conversely, MYC-overexpressing HMGECs and Meibomian glands from 4-OHT-induced mice demonstrated suppressed CHOP expression, reduced apoptosis, and upregulated fatty acid synthase expression. These results suggest that MYC inhibition induces the ISR and promotes apoptosis, while MYC induction suppresses CHOP expression. High MYC expression may, therefore, serve as a mechanism for SebCA to elude cell death by promoting lipogenesis.
Collapse
Affiliation(s)
| | | | - Cornelia Peterson
- Department of Comparative Pathobiology, Tufts University, North Grafton, MA 01536, USA
| |
Collapse
|
10
|
Weissenstein U, Tschumi S, Leonhard B, Baumgartner S. A fermented Mistletoe (Viscum album L.) extract elicits markers characteristic for immunogenic cell death driven by endoplasmic reticulum stress in vitro. BMC Complement Med Ther 2025; 25:175. [PMID: 40369535 PMCID: PMC12076857 DOI: 10.1186/s12906-025-04909-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 05/01/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Immune evasion is a characteristic hallmark of cancer. Immunotherapies aim to activate and support the body's immune system to recognize and fight tumor cells. Induction of immunogenic cell death (ICD) and the associated activation of danger signaling pathways can increase the immunogenicity of tumor cells. Therapeutic ICD stimuli activate endoplasmic reticulum stress pathways and apoptosis leading to the cellular expression of damage-associated molecular patterns (DAMPs). The aim of our in vitro study was to investigate whether mistletoe extracts induce characteristics of immunogenic tumor cell death in cancer cell lines. METHODS Three human breast cancer cell lines and one murine melanoma cell line (SKBR3, MDA-MB-231, MCF-7, and B16F10) were treated with aqueous, fermented Viscum album extract (VAE: Iscador Qu spec.) and taxol or tunicamycin as positive controls, respectively. To investigate whether VAE induces ribotoxic stress, we measured the ER stress regulators p-eIF2a, ATF4, and CHOP by Western blot. Cell surface exposure of DAMPs (calreticulin, heat shock proteins hsp70 and hsp90), apoptosis and induction of mitochondrial reactive oxygen species (ROS) were assessed by flow cytometry. HMGB1 and ATP were quantified by ELISA and chemiluminescence assay, respectively. RESULTS Treatment with VAE resulted in phosphorylation of eIF2α in all cancer cell lines tested and increased calreticulin (CRT) exposure on the surface of pre-apoptotic SKBR3 breast cancer and B16F10 mouse melanoma cells. VAE exerted a concentration-dependent effect in all cell lines, resulting in a significantly increased exposure of three DAMPs (CRT, hsp70 and hsp90) on the surface of early apoptotic cells. Furthermore, VAE elevated mitochondrial ROS production and the release of ATP. HMGB1 release was not induced by VAE. CONCLUSIONS In this in vitro study, we demonstrated for the first time the potential of a mistletoe extract to induce surrogate markers of immunogenic cancer cell death. This is a primary step in investigating the potential of VAEs to contribute to ICD-induced tumor-specific immune activation.
Collapse
Affiliation(s)
| | | | | | - Stephan Baumgartner
- Society for Cancer Research, Arlesheim, Switzerland
- Institute of Integrative Medicine, Witten/Herdecke University, Herdecke, Germany
| |
Collapse
|
11
|
Chen H, LaFlamme CW, Wang YD, Blan AW, Koehler N, Mendonca Moraes R, Olszewski AR, Almanza Fuerte EP, Bonkowski ES, Bajpai R, Lavado A, Pruett-Miller SM, Mefford HC. Patient-derived models of UBA5-associated encephalopathy identify defects in neurodevelopment and highlight potential therapeutic avenues. Sci Transl Med 2025; 17:eadn8417. [PMID: 40333994 DOI: 10.1126/scitranslmed.adn8417] [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: 01/03/2024] [Revised: 01/17/2025] [Accepted: 04/16/2025] [Indexed: 05/09/2025]
Abstract
UBA5 encodes for the E1 enzyme of the UFMylation cascade, which plays an essential role in endoplasmic reticulum (ER) homeostasis. The clinical phenotypes of UBA5-associated encephalopathy include developmental delays, epilepsy, and intellectual disability. To date, there is no humanized neuronal model to study the cellular and molecular consequences of UBA5 pathogenic variants. We developed and characterized patient-derived cortical organoid cultures from two patients with compound heterozygous variants in UBA5. Both shared the same missense variant, which encodes a hypomorphic allele (p.A371T), along with a nonsense variant (p.G267* or p.A123fs*4). Single-cell RNA sequencing of 100-day organoids identified defects in GABAergic interneuron development. We demonstrated aberrant neuronal firing and reduction in size of patient-derived organoids. Mechanistically, we showed that ER homeostasis is perturbed along with an exacerbated unfolded protein response pathway in engineered U87-MG cells and patient-derived organoids expressing UBA5 pathogenic variants. We also assessed two potential therapeutic modalities that augmented UBA5 protein abundance to rescue aberrant molecular and cellular phenotypes. We assessed SINEUP, a long noncoding RNA that augments translation efficiency, and CRISPRa, a modified CRISPR-Cas9 approach to augment transcription efficiency to increase UBA5 protein production. Our study provides a humanized model that allows further investigations of UBA5 variants in the brain and highlights promising approaches to alleviate cellular aberrations for this rare, developmental disorder.
Collapse
Affiliation(s)
- Helen Chen
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christy W LaFlamme
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yong-Dong Wang
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Aidan W Blan
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Nikki Koehler
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Renata Mendonca Moraes
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Athena R Olszewski
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Edith P Almanza Fuerte
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Emily S Bonkowski
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Richa Bajpai
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alfonso Lavado
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Shondra M Pruett-Miller
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Heather C Mefford
- Center for Pediatric Neurological Disease Research, Pediatric Translational Neuroscience Initiative, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
- St. Jude Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
12
|
Anisimova AS, Karagöz GE. HaloPROTAC3 treatment activates the unfolded protein response of the endoplasmic reticulum in nonengineered mammalian cell lines. Mol Biol Cell 2025; 36:mr3. [PMID: 40105918 DOI: 10.1091/mbc.e24-08-0342] [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: 03/21/2025] Open
Abstract
Proteins fused to HaloTag, an engineered haloalkane dehalogenase, can be depleted by a heterobifunctional degrader compound HaloPROTAC3. The binding of HaloPROTAC3 to both the HaloTag and the E3 ligase von Hippel-Lindau (VHL) brings them into proximity and mediates the degradation of the HaloTag fusion proteins. Here, we generated a colon cancer cell line HCT116 expressing HaloTag fused to the RNA-binding protein IGF2BP3 to study its function. HaloPROTAC3 treatment depleted 75% of HaloTag-IGF2BP3 in 5 h. Transcriptomics revealed that HaloPROTAC3 treatment resulted in the destabilization of IGF2BP3 target mRNAs and activated the unfolded protein response (UPR). Surprisingly, we found that HaloPROTAC3 results in UPR activation in nonengineered mammalian cells. Our data demonstrate that HaloPROTAC3 causes mild endoplasmic reticulum stress independent of IGF2BP3 function and shall guide future studies using the HaloPROTAC3 protein depletion strategy.
Collapse
Affiliation(s)
- Aleksandra S Anisimova
- Max Perutz Labs Vienna, Vienna BioCenter, Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - G Elif Karagöz
- Max Perutz Labs Vienna, Vienna BioCenter, Vienna, Austria
- Medical University of Vienna, Vienna, Austria
| |
Collapse
|
13
|
Bosakova M, Abraham SP, Wachtell D, Zieba JT, Kot A, Nita A, Czyrek AA, Koudelka A, Ursachi VC, Feketova Z, Rico-Llanos G, Svozilova K, Kocerova P, Fafilek B, Gregor T, Kotaskova J, Duran I, Vanhara P, Doubek M, Mayer J, Soucek K, Krakow D, Krejci P. Endoplasmic reticulum stress disrupts signaling via altered processing of transmembrane receptors. Cell Commun Signal 2025; 23:209. [PMID: 40307870 PMCID: PMC12044870 DOI: 10.1186/s12964-025-02208-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/17/2025] [Indexed: 05/02/2025] Open
Abstract
Cell communication systems based on polypeptide ligands use transmembrane receptors to transmit signals across the plasma membrane. In their biogenesis, receptors depend on the endoplasmic reticulum (ER)-Golgi system for folding, maturation, transport and localization to the cell surface. ER stress, caused by protein overproduction and misfolding, is a well-known pathology in neurodegeneration, cancer and numerous other diseases. How ER stress affects cell communication via transmembrane receptors is largely unknown. In disease models of multiple myeloma, chronic lymphocytic leukemia and osteogenesis imperfecta, we show that ER stress leads to loss of the mature transmembrane receptors FGFR3, ROR1, FGFR1, LRP6, FZD5 and PTH1R at the cell surface, resulting in impaired downstream signaling. This is caused by downregulation of receptor production and increased intracellular retention of immature receptor forms. Reduction of ER stress by treatment of cells with the chemical chaperone tauroursodeoxycholic acid or by expression of the chaperone protein BiP resulted in restoration of receptor maturation and signaling. We show a previously unappreciated pathological effect of ER stress; impaired cellular communication due to altered receptor processing. Our findings have implications for disease mechanisms related to ER stress and are particularly important when receptor-based pharmacological approaches are used for treatment.
Collapse
Affiliation(s)
- Michaela Bosakova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
| | - Sara P Abraham
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
| | - Davis Wachtell
- Department of Orthopaedic Surgery, Human Genetics, and Obstetrics and Gynecology, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Jennifer T Zieba
- Department of Orthopaedic Surgery, Human Genetics, and Obstetrics and Gynecology, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Alexander Kot
- Department of Orthopaedic Surgery, Human Genetics, and Obstetrics and Gynecology, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Alexandru Nita
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200, Brno, Czech Republic
| | - Aleksandra Anna Czyrek
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
| | - Adolf Koudelka
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Vlad-Constantin Ursachi
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
| | - Zuzana Feketova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
| | - Gustavo Rico-Llanos
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
| | - Katerina Svozilova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200, Brno, Czech Republic
| | - Petra Kocerova
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200, Brno, Czech Republic
| | - Bohumil Fafilek
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
| | - Tomas Gregor
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
| | - Jana Kotaskova
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, 62500, Brno, Czech Republic
| | - Ivan Duran
- Department of Orthopaedic Surgery, Human Genetics, and Obstetrics and Gynecology, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Petr Vanhara
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic
| | - Michael Doubek
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, 62500, Brno, Czech Republic
| | - Jiri Mayer
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, 62500, Brno, Czech Republic
| | - Karel Soucek
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic
- Department of Experimental Biology, Faculty of Science, Masaryk University, 62500, Brno, Czech Republic
- Department of Cytokinetics, Institute of Biophysics, Czech Academy of Sciences, 61265, Brno, Czech Republic
| | - Deborah Krakow
- Department of Orthopaedic Surgery, Human Genetics, and Obstetrics and Gynecology, University of California at Los Angeles, Los Angeles, CA, 90095, USA.
| | - Pavel Krejci
- Department of Biology, Faculty of Medicine, Masaryk University, 62500, Brno, Czech Republic.
- Institute of Animal Physiology and Genetics, Czech Academy of Sciences, 60200, Brno, Czech Republic.
- International Clinical Research Center, St. Anne's University Hospital, 65691, Brno, Czech Republic.
| |
Collapse
|
14
|
Volloch V, Rits-Volloch S. Alzheimer's Is a Multiform Disease of Sustained Neuronal Integrated Stress Response Driven by the C99 Fragment Generated Independently of AβPP; Proteolytic Production of Aβ Is Suppressed in AD-Affected Neurons: Evolution of a Theory. Int J Mol Sci 2025; 26:4252. [PMID: 40362488 PMCID: PMC12073115 DOI: 10.3390/ijms26094252] [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: 03/18/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The present Perspective analyzes the remarkable evolution of the Amyloid Cascade Hypothesis 2.0 (ACH2.0) theory of Alzheimer's disease (AD) since its inception a few years ago, as reflected in the diminishing role of amyloid-beta (Aβ) in the disease. In the initial iteration of the ACH2.0, Aβ-protein-precursor (AβPP)-derived intraneuronal Aβ (iAβ), accumulated to neuronal integrated stress response (ISR)-eliciting levels, triggers AD. The neuronal ISR, in turn, activates the AβPP-independent production of its C99 fragment that is processed into iAβ, which drives the disease. The second iteration of the ACH2.0 stemmed from the realization that AD is, in fact, a disease of the sustained neuronal ISR. It introduced two categories of AD-conventional and unconventional-differing mainly in the manner of their causation. The former is caused by the neuronal ISR triggered by AβPP-derived iAβ, whereas in the latter, the neuronal ISR is elicited by stressors distinct from AβPP-derived iAβ and arising from brain trauma, viral and bacterial infections, and various types of inflammation. Moreover, conventional AD always contains an unconventional component, and in both forms, the disease is driven by iAβ generated independently of AβPP. In its third, the current, iteration, the ACH2.0 posits that proteolytic production of Aβ is suppressed in AD-affected neurons and that the disease is driven by C99 generated independently of AβPP. Suppression of Aβ production in AD seems an oxymoron: Aβ is equated with AD, and the later is inconceivable without the former in an ingrained Amyloid Cascade Hypothesis (ACH)-based notion. But suppression of Aβ production in AD-affected neurons is where the logic leads, and to follow it we only need to overcome the inertia of the preexisting assumptions. Moreover, not only is the generation of Aβ suppressed, so is the production of all components of the AβPP proteolytic pathway. This assertion is not a quantum leap (unless overcoming the inertia counts as such): the global cellular protein synthesis is severely suppressed under the neuronal ISR conditions, and there is no reason for constituents of the AβPP proteolytic pathway to be exempted, and they, apparently, are not, as indicated by the empirical data. In contrast, tau protein translation persists in AD-affected neurons under ISR conditions because the human tau mRNA contains an internal ribosomal entry site in its 5'UTR. In current mouse models, iAβ derived from AβPP expressed exogenously from human transgenes elicits the neuronal ISR and thus suppresses its own production. Its levels cannot principally reach AD pathology-causing levels regardless of the number of transgenes or the types of FAD mutations that they (or additional transgenes) carry. Since the AβPP-independent C99 production pathway is inoperative in mice, the current transgenic models have no potential for developing the full spectrum of AD pathology. What they display are only effects of the AβPP-derived iAβ-elicited neuronal ISR. The paper describes strategies to construct adequate transgenic AD models. It also details the utilization of human neuronal cells as the only adequate model system currently available for conventional and unconventional AD. The final alteration of the ACH2.0, introduced in the present Perspective, is that AβPP, which supports neuronal functionality and viability, is, after all, potentially produced in AD-affected neurons, albeit not conventionally but in an ISR-driven and -compatible process. Thus, the present narrative begins with the "omnipotent" Aβ capable of both triggering and driving the disease and ends up with this peptide largely dislodged from its pedestal and retaining its central role in triggering the disease in only one, although prevalent (conventional), category of AD (and driving it in none). Among interesting inferences of the present Perspective is the determination that "sporadic AD" is not sporadic at all ("non-familial" would be a much better designation). The term has fatalistic connotations, implying that the disease can strike at random. This is patently not the case: The conventional disease affects a distinct subpopulation, and the basis for unconventional AD is well understood. Another conclusion is that, unless prevented, the occurrence of conventional AD is inevitable given a sufficiently long lifespan. This Perspective also defines therapeutic directions not to be taken as well as auspicious ways forward. The former category includes ACH-based drugs (those interfering with the proteolytic production of Aβ and/or depleting extracellular Aβ). They are legitimate (albeit inefficient) preventive agents for conventional AD. There is, however, a proverbial snowball's chance in hell of them being effective in symptomatic AD, lecanemab, donanemab, and any other "…mab" or "…stat" notwithstanding. They comprise Aβ-specific antibodies, inhibitors of beta- and gamma-secretase, and modulators of the latter. In the latter category, among ways to go are the following: (1) Depletion of iAβ, which, if sufficiently "deep", opens up a tantalizing possibility of once-in-a-lifetime preventive transient treatment for conventional AD and aging-associated cognitive decline, AACD. (2) Composite therapy comprising the degradation of C99/iAβ and concurrent inhibition of the neuronal ISR. A single transient treatment could be sufficient to arrest the progression of conventional AD and prevent its recurrence for life. Multiple recurrent treatments would achieve the same outcome in unconventional AD. Alternatively, the sustained reduction/removal of unconventional neuronal ISR-eliciting stressors through the elimination of their source would convert unconventional AD into conventional one, preventable/treatable by a single transient administration of the composite C99/iAβ depletion/ISR suppression therapy. Efficient and suitable ISR inhibitors are available, and it is explicitly clear where to look for C99/iAβ-specific targeted degradation agents-activators of BACE1 and, especially, BACE2. Directly acting C99/iAβ-specific degradation agents such as proteolysis-targeting chimeras (PROTACs) and molecular-glue degraders (MGDs) are also viable options. (3) A circumscribed shift (either upstream or downstream) of the position of transcription start site (TSS) of the human AβPP gene, or, alternatively, a gene editing-mediated excision or replacement of a small, defined segment of its portion encoding 5'-untranslated region of AβPP mRNA; targeting AβPP RNA with anti-antisense oligonucleotides is another possibility. If properly executed, these RNA-based strategies would not interfere with the protein-coding potential of AβPP mRNA, and each would be capable of both preventing and stopping the AβPP-independent generation of C99 and thus of either preventing AD or arresting the progression of the disease in its conventional and unconventional forms. The paper is interspersed with "validation" sections: every conceptually significant notion is either validated by the existing data or an experimental procedure validating it is proposed.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sophia Rits-Volloch
- Division of Molecular Medicine, Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
15
|
Ezer S, Ronin N, Yanovsky-Dagan S, Rotem-Bamberger S, Halstuk O, Wexler Y, Ben-Moshe Z, Plaschkes I, Benyamini H, Saada A, Inbal A, Harel T. Transcriptome analysis of atad3-null zebrafish embryos elucidates possible disease mechanisms. Orphanet J Rare Dis 2025; 20:181. [PMID: 40234890 PMCID: PMC12001410 DOI: 10.1186/s13023-025-03709-0] [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: 07/05/2024] [Accepted: 04/02/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND ATAD3A, a nuclear gene encoding the ATAD3A protein, has diverse roles in mitochondrial processes, encompassing mitochondrial dynamics, mitochondrial DNA maintenance, metabolic pathways and inter-organellar interactions. Pathogenic variants in this gene cause neurological diseases in humans with recognizable genotype-phenotype correlations. Yet, gaps in knowledge remain regarding the underlying pathogenesis. METHODS To further investigate the gene function and its implication in health and disease, we utilized CRISPR/Cas9 genome editing to generate a knockout model of the zebrafish ortholog gene, atad3. We characterized the phenotype of the null model, performed mitochondrial and functional tests, and compared the transcriptome of null embryos to their healthy siblings. RESULTS Analysis of atad3-null zebrafish embryos revealed microcephaly, small eyes, pericardial edema and musculature thinning, closely mirroring the human rare disease phenotype. Larvae exhibited delayed hatching and embryonic lethality by 13 days post-fertilization (dpf). Locomotor activity, ATP content, mitochondrial content, and mitochondrial activity were all reduced in the mutant embryos. Transcriptome analysis at 3 dpf via RNA-sequencing indicated decline in most mitochondrial pathways, accompanied by a global upregulation of cytosolic tRNA synthetases, presumably secondary to mitochondrial stress and possibly endoplasmic reticulum (ER)-stress. Differential expression of select genes was corroborated in fibroblasts from an affected individual. CONCLUSIONS The atad3-null zebrafish model emerges as a reliable representation of human ATAD3A-associated disorders, with similarities in differentially expressed pathways and processes. Furthermore, our study underscores mitochondrial dysfunction as the primary underlying pathogenic mechanism in ATAD3A-associated disorders and identifies potential readouts for therapeutic studies.
Collapse
Affiliation(s)
- Shlomit Ezer
- Department of Genetics, Hadassah Medical Organization, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nathan Ronin
- Department of Medical Neurobiology, Institute for Medical Research-Israel-Canada, Jerusalem, Israel
- The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | | | - Shahar Rotem-Bamberger
- Department of Medical Neurobiology, Institute for Medical Research-Israel-Canada, Jerusalem, Israel
- The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Orli Halstuk
- Department of Genetics, Hadassah Medical Organization, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yair Wexler
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Zohar Ben-Moshe
- Department of Neurobiology, The George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel Aviv, Israel
| | - Inbar Plaschkes
- Info-CORE, Bioinformatics Unit of the I-CORE, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Hadar Benyamini
- Info-CORE, Bioinformatics Unit of the I-CORE, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ann Saada
- Department of Genetics, Hadassah Medical Organization, Jerusalem, Israel
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Laboratory Sciences, Hadassah Academic College , Jerusalem, Israel
| | - Adi Inbal
- Department of Medical Neurobiology, Institute for Medical Research-Israel-Canada, Jerusalem, Israel
- The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Tamar Harel
- Department of Genetics, Hadassah Medical Organization, Jerusalem, Israel.
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
- Department of Genetics, Hadassah-Hebrew University Medical Center, POB 12000, Jerusalem, Israel.
| |
Collapse
|
16
|
Zhou H, Zhang J, Wang R, Huang J, Xin C, Song Z. The unfolded protein response is a potential therapeutic target in pathogenic fungi. FEBS J 2025. [PMID: 40227882 DOI: 10.1111/febs.70100] [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: 11/03/2024] [Revised: 01/15/2025] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
Pathogenic fungal infections cause significant morbidity and mortality, particularly in immunocompromised patients. The frequent emergence of multidrug-resistant strains challenges existing antifungal therapies, driving the need to investigate novel antifungal agents that target new molecular moieties. Pathogenic fungi are subjected to various environmental stressors, including pH, temperature, and pharmacological agents, both in natural habitats and the host body. These stressors elevate the risk of misfolded or unfolded protein production within the endoplasmic reticulum (ER) which, if not promptly mitigated, can lead to the accumulation of these proteins in the ER lumen. This accumulation triggers an ER stress response, potentially jeopardizing fungal survival. The unfolded protein response (UPR) is a critical cellular defense mechanism activated by ER stress to restore the homeostasis of protein folding. In recent years, the regulatory role of the UPR in pathogenic fungi has garnered significant attention, particularly for its involvement in fungal adaptation, regulation of virulence, and drug resistance. In this review, we comparatively analyze the UPRs of fungi and mammals and examine the potential utility of the UPR as a molecular antifungal target in pathogenic fungi. By clarifying the specificity and regulatory functions of the UPR in pathogenic fungi, we highlight new avenues for identifying potential therapeutic targets for antifungal treatments.
Collapse
Affiliation(s)
- Hao Zhou
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Jinping Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
| | - Rong Wang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Ju Huang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Caiyan Xin
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Zhangyong Song
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
- Public Center of Experimental Technology, Southwest Medical University, Luzhou, China
- Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, China
| |
Collapse
|
17
|
Dabsan S, Zur G, Abu-Freha N, Sofer S, Grossman-Haham I, Gilad A, Igbaria A. Cytosolic and endoplasmic reticulum chaperones inhibit wt-p53 to increase cancer cells' survival by refluxing ER-proteins to the cytosol. eLife 2025; 14:e102658. [PMID: 40202782 PMCID: PMC11981610 DOI: 10.7554/elife.102658] [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: 08/26/2024] [Accepted: 02/25/2025] [Indexed: 04/10/2025] Open
Abstract
The endoplasmic reticulum (ER) is an essential sensing organelle responsible for the folding and secretion of almost one-third of eukaryotic cells' total proteins. However, environmental, chemical, and genetic insults often lead to protein misfolding in the ER, accumulating misfolded proteins, and causing ER stress. To solve this, several mechanisms were reported to relieve ER stress by decreasing the ER protein load. Recently, we reported a novel ER surveillance mechanism by which proteins from the secretory pathway are refluxed to the cytosol to relieve the ER of its content. The refluxed proteins gain new prosurvival functions in cancer cells, thereby increasing cancer cell fitness. We termed this phenomenon ER to CYtosol Signaling (or 'ERCYS'). Here, we found that in mammalian cells, ERCYS is regulated by DNAJB12, DNAJB14, and the HSC70 cochaperone SGTA. Mechanistically, DNAJB12 and DNAJB14 bind HSC70 and SGTA - through their cytosolically localized J-domains to facilitate ER-protein reflux. DNAJB12 is necessary and sufficient to drive this phenomenon to increase AGR2 reflux and inhibit wt-p53 during ER stress. Mutations in DNAJB12/14 J-domain prevent the inhibitory interaction between AGR2-wt-p53. Thus, targeting the DNAJB12/14-HSC70/SGTA axis is a promising strategy to inhibit ERCYS and impair cancer cell fitness.
Collapse
Affiliation(s)
- Salam Dabsan
- Department of Life Sciences, Faculty of Natural Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Gali Zur
- Department of Life Sciences, Faculty of Natural Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Naim Abu-Freha
- Institute of Gastroenterology and Liver Diseases, Soroka Medical Center, Faculty of Health Sciences, Ben Gurion University of the NegevBeer ShevaIsrael
| | - Shahar Sofer
- Department of Life Sciences, Faculty of Natural Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Iris Grossman-Haham
- Department of Life Sciences, Faculty of Natural Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
- The Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Ayelet Gilad
- Department of Life Sciences, Faculty of Natural Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| | - Aeid Igbaria
- Department of Life Sciences, Faculty of Natural Sciences, Ben-Gurion University of the NegevBeer ShevaIsrael
| |
Collapse
|
18
|
Zandhuis ND, Bradarić A, van der Zwaan C, Hoogendijk AJ, Popović B, Wolkers MC. Combined Deletion of ZFP36L1 and ZFP36L2 Drives Superior Cytokine Production in T Cells at the Cost of Cell Fitness. Eur J Immunol 2025; 55:e202451641. [PMID: 40249077 PMCID: PMC12007392 DOI: 10.1002/eji.202451641] [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] [Received: 11/30/2024] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/19/2025]
Abstract
A key feature of cytotoxic CD8+ T cells for eliminating pathogens and malignant cells is their capacity to produce proinflammatory cytokines, which include TNF and IFNγ. Provided that these cytokines are highly toxic, a tight control of their production is imperative. RNA-binding proteins (RBPs) are essential for the fine-tuning of cytokine production. The role of the RBP ZFP36L1 and its sister protein ZFP36L2 herein has been established, but their relative contribution to cytokine production is not well understood. We here compared the effect of ZFP36L1 and ZFP36L2 single and double deficiency in murine effector CD8+ T cells. Whereas single deficient T cells significantly increased cytokine production, double deficiency completely unleashed the cytokine production. Not only the TNF production was substantially prolonged in double-deficient T cells. Also, the production of IFNγ reached unprecedented levels with >90% IFNγ-producing T cells compared with 3% in WT T cells after 3 days of continuous activation. This continuous cytokine production by double-deficient T cells was also observed in tumor-infiltrating lymphocytes in vivo, however, with no effect on tumor growth. ZFP36L1 and ZFP36L2 double deficiency resulted in decreased cell viability, impaired STAT5 signaling, and dysregulated cell cycle progression. In conclusion, while combined deletion in ZFP36L1 and ZFP36L2 can drive continuous cytokine production even upon chronic activation, safeguards are in place to counteract such super-cytokine producers.
Collapse
Affiliation(s)
- Nordin D. Zandhuis
- Department of ResearchT Cell Differentiation LabSanquin Blood Supply FoundationAmsterdamThe Netherlands
- Amsterdam UMC Landsteiner Laboratory University of Amsterdam Cancer Center Amsterdam Cancer Immunologyand Amsterdam Institute for Infection & ImmunityAmsterdamThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Antonia Bradarić
- Department of ResearchT Cell Differentiation LabSanquin Blood Supply FoundationAmsterdamThe Netherlands
- Amsterdam UMC Landsteiner Laboratory University of Amsterdam Cancer Center Amsterdam Cancer Immunologyand Amsterdam Institute for Infection & ImmunityAmsterdamThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Carmen van der Zwaan
- Amsterdam UMC Landsteiner Laboratory University of Amsterdam Cancer Center Amsterdam Cancer Immunologyand Amsterdam Institute for Infection & ImmunityAmsterdamThe Netherlands
- Department of ResearchBleeding & HemostasisSanquin Blood Supply FoundationAmsterdamThe Netherlands
| | - Arie J. Hoogendijk
- Amsterdam UMC Landsteiner Laboratory University of Amsterdam Cancer Center Amsterdam Cancer Immunologyand Amsterdam Institute for Infection & ImmunityAmsterdamThe Netherlands
- Department of ResearchBleeding & HemostasisSanquin Blood Supply FoundationAmsterdamThe Netherlands
| | - Branka Popović
- Department of ResearchT Cell Differentiation LabSanquin Blood Supply FoundationAmsterdamThe Netherlands
- Amsterdam UMC Landsteiner Laboratory University of Amsterdam Cancer Center Amsterdam Cancer Immunologyand Amsterdam Institute for Infection & ImmunityAmsterdamThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| | - Monika C. Wolkers
- Department of ResearchT Cell Differentiation LabSanquin Blood Supply FoundationAmsterdamThe Netherlands
- Amsterdam UMC Landsteiner Laboratory University of Amsterdam Cancer Center Amsterdam Cancer Immunologyand Amsterdam Institute for Infection & ImmunityAmsterdamThe Netherlands
- Oncode InstituteUtrechtThe Netherlands
| |
Collapse
|
19
|
Sastre J, Pérez S, Sabater L, Rius-Pérez S. Redox signaling in the pancreas in health and disease. Physiol Rev 2025; 105:593-650. [PMID: 39324871 DOI: 10.1152/physrev.00044.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024] Open
Abstract
This review addresses oxidative stress and redox signaling in the pancreas under healthy physiological conditions as well as in acute pancreatitis, chronic pancreatitis, pancreatic cancer, and diabetes. Physiological redox homeodynamics is maintained mainly by NRF2/KEAP1, NF-κB, protein tyrosine phosphatases, peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α), and normal autophagy. Depletion of reduced glutathione (GSH) in the pancreas is a hallmark of acute pancreatitis and is initially accompanied by disulfide stress, which is characterized by protein cysteinylation without increased glutathione oxidation. A cross talk between oxidative stress, MAPKs, and NF-κB amplifies the inflammatory cascade, with PP2A and PGC1α as key redox regulatory nodes. In acute pancreatitis, nitration of cystathionine-β synthase causes blockade of the transsulfuration pathway leading to increased homocysteine levels, whereas p53 triggers necroptosis in the pancreas through downregulation of sulfiredoxin, PGC1α, and peroxiredoxin 3. Chronic pancreatitis exhibits oxidative distress mediated by NADPH oxidase 1 and/or CYP2E1, which promotes cell death, fibrosis, and inflammation. Oxidative stress cooperates with mutant KRAS to initiate and promote pancreatic adenocarcinoma. Mutant KRAS increases mitochondrial reactive oxygen species (ROS), which trigger acinar-to-ductal metaplasia and progression to pancreatic intraepithelial neoplasia (PanIN). ROS are maintained at a sufficient level to promote cell proliferation, while avoiding cell death or senescence through formation of NADPH and GSH and activation of NRF2, HIF-1/2α, and CREB. Redox signaling also plays a fundamental role in differentiation, proliferation, and insulin secretion of β-cells. However, ROS overproduction promotes β-cell dysfunction and apoptosis in type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Luis Sabater
- Liver, Biliary and Pancreatic Unit, Hospital Clínico, Department of Surgery, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biology, University of Valencia, Valencia, Spain
| |
Collapse
|
20
|
Dong Z, Wang X, Wang P, Bai M, Wang T, Chu Y, Qin Y. Idiopathic Pulmonary Fibrosis Caused by Damaged Mitochondria and Imbalanced Protein Homeostasis in Alveolar Epithelial Type II Cell. Adv Biol (Weinh) 2025; 9:e2400297. [PMID: 39390651 PMCID: PMC12001015 DOI: 10.1002/adbi.202400297] [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: 05/30/2024] [Revised: 08/21/2024] [Indexed: 10/12/2024]
Abstract
Alveolar epithelial Type II (ATII) cells are closely associated with early events of Idiopathic pulmonary fibrosis (IPF). Proteostasis dysfunction, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction are known causes of decreased proliferation of alveolar epithelial cells and the secretion of pro-fibrotic mediators. Here, a large body of evidence is systematized and a cascade relationship between protein homeostasis, endoplasmic reticulum stress, mitochondrial dysfunction, and fibrotropic cytokines is proposed, providing a theoretical basis for ATII cells dysfunction as a possible pathophysiological initiating event for idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
- Zhaoxiong Dong
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- Institute of BiophysicsChinese Academy of Sciences 15 Datun RoadChaoyang DistrictBeijing100101China
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Xiaolong Wang
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
| | - Peiwen Wang
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Mingjian Bai
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijing100101China
| | - Tianyu Wang
- School of Chemistry and Biological EngineeringUniversity of Science and Technology BeijingBeijing100101China
| | - Yanhui Chu
- College of Life ScienceMudanjiang Medical UniversityMudanjiang157000China
| | - Yan Qin
- Affiliated Cancer Hospital & Institute of Guangzhou Medical UniversityState Key Laboratory of Respiratory DiseaseSchool of Biomedical EngineeringGuangzhou Medical UniversityGuangzhou510260China
- Institute of BiophysicsChinese Academy of Sciences 15 Datun RoadChaoyang DistrictBeijing100101China
| |
Collapse
|
21
|
Kuzminsky I, Ghanim M. Immunity responses as checkpoints for efficient transmission of begomoviruses by whiteflies. Virology 2025; 605:110462. [PMID: 40020542 DOI: 10.1016/j.virol.2025.110462] [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: 12/12/2024] [Revised: 02/05/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025]
Abstract
Begomoviruses are a group of single stranded DNA plant viruses exclusively transmitted by the sweet potato whitefly Bemisia tabaci in a persistent, circulative manner. After acquisition from plant phloem, this group of viruses circulate and are retained within the whitefly, interacting with tissues, cells and molecular pathways for maintaining the safety of the infective intact virions, by exploiting cellular mechanisms and avoiding degradation by the insect immune responses. During retention, the virions are internalized in the midgut cells, exit and spend hours-days in the hemolymph and cross into salivary gland cells, before transmission. Destroying this group of viruses by the insect immune system seems inefficient for the most part, by examining their very efficient transmission. Thus, within the various sites along the transmission pathway especially in the midgut, it is thought that the immune system with its various layers is activated for avoiding the damage caused by the viruses on one hand, and for ensuring their safe circulation and transmission on the other hand. Begomoviruses have evolved mechanisms for counteracting and exploiting the activated immune system for their safe translocation within the whitefly. In this review, we discuss the various levels of immunity activated against begomoviruses in B. tabaci, taking other pathogen-vector systems as examples and reflecting relevant components on the interactions between B. tabaci and Begomoviruses.
Collapse
Affiliation(s)
- Ilana Kuzminsky
- Department of Entomology, Volcani Center, Rishon LeZion, 7505101, Israel; Department of Agroecology and Plant Health, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, 7610001, Israel
| | - Murad Ghanim
- Department of Entomology, Volcani Center, Rishon LeZion, 7505101, Israel.
| |
Collapse
|
22
|
Zhao L, Wu X, Li Q, Shen Y, Zeng S, Wang J, Liu Q. Inhibition of CCN5 Protects Against Apoptosis and Endoplasmic Reticulum Stress in Bisphenol A-Induced Sertoli Cells via p38/JNK MAPK Signaling Pathway. DNA Cell Biol 2025; 44:174-185. [PMID: 40184555 DOI: 10.1089/dna.2024.0247] [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] [Indexed: 04/06/2025] Open
Abstract
Bisphenol A (BPA) is the most common endocrine disruptor that has toxicity to the reproductive system and male infertility. However, the underlying mechanisms of BPA's toxicity to Sertoli cells remain poorly understood. Cellular communication network factor 5 (CCN5) is reported to regulate cell proliferation, apoptosis, and differentiation. Our study demonstrated a significant elevation of CCN5 expression in the testis of nonobstructive azoospermia patients and TM4 Sertoli cells exposed to BPA. Knockdown of CCN5 reduced apoptotic cells after BPA treatment, as determined by flow cytometry and terminal deoxynucleotidyl transferase dUTP nick end labeling assays. Cells exposed to BPA showed increased expressions of Bax and cleaved poly(ADP-ribose) polymerase, decreased expression of Bcl-2, as well as elevated activities of caspase-3 and caspase-9 in BPA-induced TM4 cells, which were reversed by CCN5 inhibition. Loss of CCN5 declined phosphorylation of protein kinase R-like endoplasmic reticulum kinase and eukaryotic translation initiation factor 2A and decreased activating transcription factor 4 and C/EBP-homologous protein in BPA-treated cells. Furthermore, silencing CCN5 blocked BPA-induced phosphorylation of p38 and c-Jun N-terminal kinase (JNK). Administration of anisomycin, a mitogen-activated protein kinase (MAPK) activator, reversed the effects of CCN5 knockdown on BPA-induced endoplasmic reticulum (ER) stress and apoptosis. Taken together, CCN5 promotes apoptosis and ER stress in Sertoli cells exposed to BPA by activating the p38/JNK MAPK signaling pathway.
Collapse
Affiliation(s)
- Lijiang Zhao
- Tianjin Medical University, Tianjin, China
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Xueqing Wu
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Qiang Li
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Yan Shen
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Sheng Zeng
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
| | - Jinbao Wang
- Reproductive Medicine Center, Shanxi Children's Hospital, Shanxi Women and Children Hospital, Taiyuan, China
| | - Qian Liu
- Department of Urology, Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
23
|
Gong J, Li J, Li J, He A, Ren B, Zhao M, Li K, Zhang Y, He M, Liu Y, Wang Z. Impact of Microglia-Derived Extracellular Vesicles on Resident Central Nervous System Cell Populations After Acute Brain Injury Under Various External Stimuli Conditions. Mol Neurobiol 2025:10.1007/s12035-025-04858-w. [PMID: 40126599 DOI: 10.1007/s12035-025-04858-w] [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: 11/19/2024] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Acute brain injuries (ABI) caused by various emergencies can lead to structural and functional damage to brain tissue. Common causes include traumatic brain injury, cerebral hemorrhage, ischemic stroke, and heat stroke. Globally, ABI represent a significant portion of neurosurgical cases. Previous studies have emphasized the significant therapeutic potential of stem cell-derived extracellular vesicles (EVs). Recent research indicates that EVs extracted from resident cells in the central nervous system (CNS) also show therapeutic potential following brain injury. Microglia, as innate immune cells of the CNS, respond to changes in the internal environment by altering their phenotype and secreting EVs that impact various CNS cells, including neurons, astrocytes, oligodendrocytes, endothelial cells, neural stem cells (NSCs), and microglia themselves. Notably, under different external stimuli, microglia can either promote neuronal survival, angiogenesis, and myelin regeneration while reducing glial scarring and inflammation, or they can exert opposite effects. This review summarizes and evaluates the current research findings on how microglia-derived EVs influence various CNS cells after ABI under different external stimuli. It analyzes the interaction mechanisms between EVs and resident CNS cells and discusses potential future research directions and clinical applications.
Collapse
Affiliation(s)
- Junjie Gong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Jing Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Jian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Anqi He
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Bingcheng Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Mingyu Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Kexin Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Yuchi Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Mengyao He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Yuheng Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China.
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China.
| |
Collapse
|
24
|
Barny LA, Hermanson JN, Garcia SK, Stauffer PE, Plate L. Dissecting Branch-Specific Unfolded Protein Response Activation in Drug-Tolerant BRAF-Mutant Melanoma using Data-Independent Acquisition Mass Spectrometry. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644425. [PMID: 40196682 PMCID: PMC11974750 DOI: 10.1101/2025.03.20.644425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Cells rely on the Unfolded Protein Response (UPR) to maintain ER protein homeostasis (proteostasis) when faced with elevated levels of misfolded and aggregated proteins. The UPR is comprised of three main branches-ATF6, IRE1, and PERK-that coordinate the synthesis of proteins involved in folding, trafficking, and degradation of nascent proteins to restore ER function. Dysregulation of the UPR is linked to numerous diseases, including neurodegenerative disorders, cancer, and diabetes. Despite its importance, identifying UPR targets has been challenging due to their heterogeneous induction, which varies by cell type and tissue. Additionally, defining the magnitude and range of UPR-regulated genes is difficult because of intricate temporal regulation, feedback between UPR branches, and extensive cross-talk with other stress-signaling pathways. To comprehensively identify UPR-regulated proteins and determine their branch specificity, we developed a data-independent acquisition (DIA) liquid-chromatography mass spectrometry (LC-MS) pipeline. Our optimized workflow improved identifications of low-abundant UPR proteins and leveraged an automated SP3-based protocol on the Biomek i5 liquid handler for label-free peptide preparation. Using engineered stable cell lines that enable selective pharmacological activation of each UPR branch without triggering global UPR activation, we identified branch-specific UPR proteomic targets. These targets were subsequently applied to investigate proteomic changes in multiple patient-derived BRAF-mutant melanoma cell lines treated with a BRAF inhibitor (PLX4720, i.e., vemurafenib). Our findings revealed differential regulation of the XBP1s branch of the UPR in the BRAF-mutant melanoma cell lines after PLX4720 treatment, likely due to calcium activation, suggesting that the UPR plays a role as a non-genetic mechanism of drug tolerance in melanoma. In conclusion, the validated branch-specific UPR proteomic targets identified in this study provide a robust framework for investigating this pathway across different cell types, drug treatments, and disease conditions in a high-throughput manner.
Collapse
Affiliation(s)
- Lea A Barny
- Chemical and Physical Biology Program, Vanderbilt University Medical Center, Nashville, TN, 37235
| | - Jake N Hermanson
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235
| | - Sarah K Garcia
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235
| | - Philip E Stauffer
- Chemical and Physical Biology Program, Vanderbilt University Medical Center, Nashville, TN, 37235
| | - Lars Plate
- Chemical and Physical Biology Program, Vanderbilt University Medical Center, Nashville, TN, 37235
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, 37235
- Department of Chemistry, Vanderbilt University, Nashville, TN, 37235
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, 37232
| |
Collapse
|
25
|
Zavarzadeh PG, Panchal K, Bishop D, Gilbert E, Trivedi M, Kee T, Ranganathan S, Arunagiri A. Exploring proinsulin proteostasis: insights into beta cell health and diabetes. Front Mol Biosci 2025; 12:1554717. [PMID: 40109403 PMCID: PMC11919908 DOI: 10.3389/fmolb.2025.1554717] [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: 01/02/2025] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Proinsulin misfolding is central to diabetes. This review examines the cellular mechanisms regulating proinsulin proteostasis in pancreatic β-cells, encompassing genetic factors such as insulin gene mutations, and exploring the roles of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR), ER redox balance, mitochondrial function, and the influence of extrinsic factors. Mutations in the INS gene, particularly those affecting cysteine residues, impair folding and disulfide bond formation, often exhibiting dominant-negative effects on the wild-type proinsulin. The importance of ER quality control mechanisms, including chaperones and oxidoreductases, in facilitating proper folding and degradation of misfolded proinsulin is emphasized. Disruptions in these systems, due to genetic mutations, ER stress, or impaired ER-to-Golgi trafficking, lead to proinsulin accumulation and β-cell dysfunction. The unfolded protein response (UPR), especially the PERK and IRE1α-XBP1 pathways, emerges as a central regulator of protein synthesis and ER stress management. The review also discusses the role of mitochondrial health, ER redox state, and extrinsic factors such as diet and medications in influencing proinsulin proteostasis. Finally, the structural insights from NMR and molecular dynamics simulations are discussedhighlighting the dynamics of misfolding and underscoring the importance of disulfide bonds. These mechanistic insights suggest innovative strategies targeting thiol/disulfide redox systems in cells to mitigate protein misfolding diseases including diabetes.
Collapse
Affiliation(s)
| | - Kathigna Panchal
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, United States
| | - Dylan Bishop
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, United States
| | - Elizabeth Gilbert
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, United States
| | - Mahi Trivedi
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, United States
| | - Tovaria Kee
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, United States
| | | | - Anoop Arunagiri
- Department of Biological Sciences, East Tennessee State University, Johnson City, TN, United States
| |
Collapse
|
26
|
Nghiem THT, Nguyen KA, Kusuma F, Park S, Park J, Joe Y, Han J, Chung HT. The PERK-eIF2α-ATF4 Axis Is Involved in Mediating ER-Stress-Induced Ferroptosis via DDIT4-mTORC1 Inhibition and Acetaminophen-Induced Hepatotoxicity. Antioxidants (Basel) 2025; 14:307. [PMID: 40227255 PMCID: PMC11939615 DOI: 10.3390/antiox14030307] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 04/15/2025] Open
Abstract
Ferroptosis, a regulated form of cell death characterized by lipid peroxidation and iron accumulation, is increasingly recognized for its role in disease pathogenesis. The unfolded protein response (UPR) has been implicated in both endoplasmic reticulum (ER) stress and ferroptosis-mediated cell fate decisions; yet, the specific mechanism remains poorly understood. In this study, we demonstrated that ER stress induced by tunicamycin and ferroptosis triggered by erastin both activate the UPR, leading to the induction of ferroptotic cell death. This cell death was mitigated by the application of chemical chaperones and a ferroptosis inhibitor. Among the three arms of the UPR, the PERK-eIF2α-ATF4 signaling axis was identified as a crucial mediator in this process. Mechanistically, the ATF4-driven induction of DDIT4 plays a pivotal role, facilitating ferroptosis via the inhibition of the mTORC1 pathway. Furthermore, acetaminophen (APAP)-induced hepatotoxicity was investigated as a model of eIF2α-ATF4-mediated ferroptosis. Our findings reveal that the inhibition of eIF2α-ATF4 or ferroptosis protects against APAP-induced liver damage, underscoring the therapeutic potential of targeting these pathways. Overall, this study not only clarifies the intricate role of the PERK-eIF2α-ATF4 axis in ER-stress-and erastin-induced ferroptosis but also extends these findings to a clinically relevant model, providing a foundation for potential therapeutic interventions in conditions characterized by dysregulated ferroptosis and ER stress.
Collapse
Affiliation(s)
- Thu-Hang Thi Nghiem
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea;
| | - Kim Anh Nguyen
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea; (K.A.N.); (F.K.); (S.P.)
| | - Fedho Kusuma
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea; (K.A.N.); (F.K.); (S.P.)
| | - Soyoung Park
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea; (K.A.N.); (F.K.); (S.P.)
| | - Jeongmin Park
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea; (J.P.); (Y.J.)
| | - Yeonsoo Joe
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea; (J.P.); (Y.J.)
| | - Jaeseok Han
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea; (K.A.N.); (F.K.); (S.P.)
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Hun Taeg Chung
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea; (J.P.); (Y.J.)
| |
Collapse
|
27
|
Shishova A, Ivin Y, Gladneva E, Fominykh K, Dyugay I, Gmyl A. Modulation of Ire1-Xbp1 Defense Pathway in Encephalomyocarditis Virus-Infected HeLa Cells. Viruses 2025; 17:360. [PMID: 40143290 PMCID: PMC11946305 DOI: 10.3390/v17030360] [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: 01/24/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
A key contributor to the pathogenicity of viruses is their interaction with cellular defense mechanisms, including UPR (unfolded protein response) that counteracts the accumulation of misfolded proteins in the endoplasmic reticulum (known as ER stress). One of the UPR branches is mediated by the IRE1 (inositol-requiring enzyme 1) protein, which possesses protein kinase and RNase activities that facilitate the unconventional cytoplasmic splicing of XBP1 mRNA, leading to the upregulation of the XBP1 transcription factor. In this study, we demonstrate that Encephalomyocarditis Virus (Cardiovirus rueckerti) is able to suppress IRE1-dependent XBP1 activation. HeLa cells infection with EMCV resulted in the modulation of phosphorylated IRE1 levels throughout the infection cycle. Viral infection did not result in the accumulation of spliced XBP1 mRNA. Moreover, the addition of a chemical inducer of ER stress (dithiothreitol) to infected cells led to a markedly lower accumulation of spliced XBP1 mRNA as compared to the level of this mRNA in inducer-treated mock-infected cells. Thus, our results demonstrate the ability of picornaviruses to modulate another defensive activity of the host cell.
Collapse
Affiliation(s)
- Anna Shishova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia; (Y.I.); (E.G.); (K.F.); (I.D.)
- Institute for Translational Medicine and Biotechnology, First Moscow State Medical University (Sechenov University), 117418 Moscow, Russia
| | - Yury Ivin
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia; (Y.I.); (E.G.); (K.F.); (I.D.)
| | - Ekaterina Gladneva
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia; (Y.I.); (E.G.); (K.F.); (I.D.)
| | - Ksenia Fominykh
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia; (Y.I.); (E.G.); (K.F.); (I.D.)
| | - Ilya Dyugay
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia; (Y.I.); (E.G.); (K.F.); (I.D.)
| | - Anatoly Gmyl
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), 108819 Moscow, Russia; (Y.I.); (E.G.); (K.F.); (I.D.)
| |
Collapse
|
28
|
Dabsan S, Twito G, Biadsy S, Igbaria A. Less is better: various means to reduce protein load in the endoplasmic reticulum. FEBS J 2025; 292:976-989. [PMID: 38865586 PMCID: PMC11880973 DOI: 10.1111/febs.17201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/08/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
The endoplasmic reticulum (ER) is an important organelle that controls the intracellular and extracellular environments. The ER is responsible for folding almost one-third of the total protein population in the eukaryotic cell. Disruption of ER-protein folding is associated with numerous human diseases, including metabolic disorders, neurodegenerative diseases, and cancer. During ER perturbations, the cells deploy various mechanisms to increase the ER-folding capacity and reduce ER-protein load by minimizing the number of substrates entering the ER to regain homeostasis. These mechanisms include signaling pathways, degradation mechanisms, and other processes that mediate the reflux of ER content to the cytosol. In this review, we will discuss the recent discoveries of five different ER quality control mechanisms, including the unfolded protein response (UPR), ER-associated-degradation (ERAD), pre-emptive quality control, ER-phagy and ER to cytosol signaling (ERCYS). We will discuss the roles of these processes in decreasing ER-protein load and inter-mechanism crosstalk.
Collapse
Affiliation(s)
- Salam Dabsan
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Gal Twito
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Suma Biadsy
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Aeid Igbaria
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael
| |
Collapse
|
29
|
Acosta-Alvear D, Harnoss JM, Walter P, Ashkenazi A. Homeostasis control in health and disease by the unfolded protein response. Nat Rev Mol Cell Biol 2025; 26:193-212. [PMID: 39501044 DOI: 10.1038/s41580-024-00794-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 02/27/2025]
Abstract
Cells rely on the endoplasmic reticulum (ER) to fold and assemble newly synthesized transmembrane and secretory proteins - essential for cellular structure-function and for both intracellular and intercellular communication. To ensure the operative fidelity of the ER, eukaryotic cells leverage the unfolded protein response (UPR) - a stress-sensing and signalling network that maintains homeostasis by rebalancing the biosynthetic capacity of the ER according to need. The metazoan UPR can also redirect signalling from cytoprotective adaptation to programmed cell death if homeostasis restoration fails. As such, the UPR benefits multicellular organisms by preserving optimally functioning cells while removing damaged ones. Nevertheless, dysregulation of the UPR can be harmful. In this Review, we discuss the UPR and its regulatory processes as a paradigm in health and disease. We highlight important recent advances in molecular and mechanistic understanding of the UPR that enable greater precision in designing and developing innovative strategies to harness its potential for therapeutic gain. We underscore the rheostatic character of the UPR, its contextual nature and critical open questions for its further elucidation.
Collapse
Affiliation(s)
| | - Jonathan M Harnoss
- Department of General, Visceral, Thoracic and Transplant Surgery, University Hospital Giessen, Giessen, Germany
| | - Peter Walter
- Altos Labs, Inc., Bay Area Institute of Science, Redwood City, CA, USA.
| | - Avi Ashkenazi
- Research Oncology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
30
|
Hermanson JN, Barny LA, Plate L. Development of an Adaptive, Economical, and Easy-to-Use SP3-TMT Automated Sample Preparation Workflow for Quantitative Proteomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639731. [PMID: 40060590 PMCID: PMC11888279 DOI: 10.1101/2025.02.23.639731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Liquid handling robots have been developed to automate various steps of the bottom-up proteomics workflow, however, protocols for the generation of isobarically labeled peptides remain limited. Existing methods often require costly specialty devices and are constrained by fixed workflows. To address this, we developed a cost-effective, flexible, automated sample preparation protocol for TMT-labeled peptides using the Biomek i5 liquid handler. Our approach leverages Single-Pot Solid-Phase-Enhanced Sample Preparation (SP3) with paramagnetic beads to streamline protein cleanup and digestion. The protocol also allows for adjustment of trypsin concentration and peptide-to-TMT ratio to increase throughput and reduce costs, respectively. We compared our automated and manual 18-plex TMT-Pro labeling workflows by monitoring select protein markers of the Unfolded Protein Response (UPR) in pharmacologically activatable, engineered cell lines. Overall, the automated protocol demonstrated equivalent performance in peptide and protein identifications, digestion and labeling efficiency, and an enhancement in the dynamic range of TMT quantifications. Compared to the manual method, the Biomek protocol significantly reduces hands-on time and minimizes sample handling errors. The 96-well format additionally allows for the number of TMT reactions to be scaled up quickly without a significant increase in user interaction. Our optimized automated workflow enhances throughput, reproducibility, and cost-effectiveness, making it a valuable tool for high-throughput proteomics studies.
Collapse
Affiliation(s)
- Jake N Hermanson
- Department of Biological Sciences, Vanderbilt University Nashville, Tennessee
| | - Lea A Barny
- Program in Chemical and Physical Biology, Vanderbilt University Nashville, Tennessee
| | - Lars Plate
- Department of Biological Sciences, Vanderbilt University Nashville, Tennessee
- Program in Chemical and Physical Biology, Vanderbilt University Nashville, Tennessee
- Department of Chemistry, Vanderbilt University Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
31
|
Wang Y, Wei W, Zhang Y, Miao J, Bao X, Lu C. MLKL as an emerging machinery for modulating organelle dynamics: regulatory mechanisms, pathophysiological significance, and targeted therapeutics. Front Pharmacol 2025; 16:1512968. [PMID: 40070567 PMCID: PMC11893596 DOI: 10.3389/fphar.2025.1512968] [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: 10/17/2024] [Accepted: 02/04/2025] [Indexed: 03/14/2025] Open
Abstract
Mixed lineage kinase domain-like protein (MLKL) is a pseudokinase featured by a protein kinase-like domain without catalytic activity. MLKL was originally discovered to be phosphorylated by receptor-interacting protein kinase 1/3, typically increase plasma membrane permeabilization, and disrupt the membrane integrity, ultimately executing necroptosis. Recent evidence uncovers the association of MLKL with diverse cellular organelles, including the mitochondrion, lysosome, endosome, endoplasmic reticulum, and nucleus. Thus, this review mainly focuses on the regulatory functions, mechanisms, and targets of MLKL in organelles rather than necroptosis and summarize the medical significance in multiple diseases. On this basis, we conclude and analyze the current progress and prospect for the development of MLKL-related drugs, from natural products, small-molecule chemical compounds, to proteolysis-targeting chimera. This review is aimed to propel the development of MLKL as a valid drug target and the discovery of novel MLKL-related drugs, and promote their further applications.
Collapse
Affiliation(s)
| | | | | | | | - Xiaofeng Bao
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Chunfeng Lu
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
32
|
Zobeydi AM, Mousavi Namavar SN, Sadeghi Shahdani M, Choobineh S, Kordi MR, Rakhshan K. Mitigating doxorubicin-induced hepatotoxicity in male rats: The role of aerobic interval training and curcumin supplementation in reducing oxidative stress, endoplasmic reticulum stress and apoptosis. Sci Rep 2025; 15:6604. [PMID: 39994295 PMCID: PMC11850886 DOI: 10.1038/s41598-025-91133-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
Doxorubicin (DOXO) is a powerful anthracycline chemotherapeutic drug, but its clinical usage has been limited by its deleterious effects on different organs, particularly hepatotoxicity. The aim of this study was to establish the combined effects of aerobic interval training (AIT) and curcumin supplementation on mitigating oxidative damage and endoplasmic reticulum (ER) stress-mediated apoptosis in a rat model of DOXO-induced hepatotoxicity. Fifty-six male Sprague-Dawley rats were randomly split into six groups: control (CON), vehicle, doxorubicin (Dox), doxorubicin + curcumin (Dox-C), doxorubicin + AIT (Dox-A), and doxorubicin + curcumin + AIT (Dox-AC). DOXO was intraperitoneally injected weekly (4 mg/kg/week) for five weeks. Curcumin supplementation (100 mg/kg/day) and AIT (4 min at 80-90% of VO2max intermitted by 3 min of active rest at 65-75% of VO2max) were conducted five times a week for six weeks. Finally, the hepatic tissue and blood samples were collected to assess histopathological changes, liver damage biomarkers, and the protein expression of oxidative stress, ER stress, and apoptosis markers. Tissue sections revealed that AIT and curcumin supplementation significantly improved hepatotoxicity induced by DOXO, as evidenced by the positive effects on histopathological alterations and serum markers of hepatic damage (P < 0.05). Both curcumin and AIT significantly reduced DOXO-triggered oxidative damage, ER stress, and apoptosis (P < 0.05), with the latter showing slightly higher effectiveness. Consequently, the combination of AIT with curcumin supplementation exhibits protective effects against chronic hepatotoxicity induced by DOXO, with AIT demonstrating relatively greater efficacy in increasing antioxidant capacity and reducing ER stress and apoptosis.
Collapse
Affiliation(s)
- Amir Mohammad Zobeydi
- Department of Exercise Physiology, Faculty of Sport Sciences and Health, University of Tehran, Tehran, Iran.
| | | | - Maryam Sadeghi Shahdani
- Department of Exercise Physiology, Faculty of Sport Sciences and Health, University of Tehran, Tehran, Iran
| | - Siroos Choobineh
- Department of Exercise Physiology, Faculty of Sport Sciences and Health, University of Tehran, Tehran, Iran.
| | - Mohammad Reza Kordi
- Department of Exercise Physiology, Faculty of Sport Sciences and Health, University of Tehran, Tehran, Iran
| | - Kamran Rakhshan
- Department of Medical Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Pullen KM, Finethy R, Ko SHB, Reames CJ, Sassetti CM, Lauffenburger DA. Cross-species transcriptomics translation reveals a role for the unfolded protein response in Mycobacterium tuberculosis infection. NPJ Syst Biol Appl 2025; 11:19. [PMID: 39955299 PMCID: PMC11830044 DOI: 10.1038/s41540-024-00487-6] [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: 09/07/2024] [Accepted: 12/25/2024] [Indexed: 02/17/2025] Open
Abstract
Numerous studies have identified similarities in blood transcriptomic signatures of tuberculosis (TB) phenotypes between mice and humans, including type 1 interferon production and innate immune cell activation. However, murine infection pathophysiology is distinct from human disease. We hypothesized that this is partly due to differences in the relative importance of biological pathways across species. To address this animal-to-human gap, we applied a systems modeling framework, Translatable Components Regression, to identify the axes of variation in the preclinical data most relevant to human TB disease state. Among the pathways our cross-species model pinpointed as highly predictive of human TB phenotype was the infection-induced unfolded protein response. To validate this mechanism, we confirmed that this cellular stress pathway modulates immune functions in Mycobacterium tuberculosis-infected mouse macrophages. Our work demonstrates how systems-level computational models enhance the value of animal studies for elucidating complex human pathophysiology.
Collapse
Affiliation(s)
- Krista M Pullen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ryan Finethy
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
| | - Seung-Hyun B Ko
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Charlotte J Reames
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
| | - Christopher M Sassetti
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA.
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
34
|
Varlamova EG, Gudkov SV, Turovsky EA. Differential effect of cerium nanoparticles on the viability, redox-status and Ca 2+-signaling system of cancer cells of various origins. Arch Biochem Biophys 2025; 764:110261. [PMID: 39645139 DOI: 10.1016/j.abb.2024.110261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/12/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The present study aims to understand the molecular mechanism underlying the therapeutic effect of cerium nanoparticles (CeNPs) in oncology. Cancer cells were treated with different concentrations of pure nanocerium of different sizes synthesized by laser ablation. Due to the not insignificant influence of surface defects and oxygen species on the ROS-modulating properties of cerium nanoparticles, the nanoparticles were not coated with surfactants or organic molecules during synthesis, which could potentially inhibit a number of pro-oxidative effects. Reactive oxygen species (ROS) production, expression of genes encoding redox-status proteins, selenoproteins and proteins regulating cell death and endoplasmic reticulum stress (ER-stress) were investigated as indicators of the molecular mechanism of cancer cell death. Studies were conducted on the effects of cerium nanoparticles on the Ca2+ signaling system of cancer cells of different origins. Mouse fibroblasts (L-929 cell line) were used as non-cancerous ("normal") cells for which a whole series of experiments were performed, and a comparative analysis of the effects of nanoceria. It was found that 75 nm-sized cerium nanoparticles did not affect the redox-status and ROS production of cancer cells. In fibroblast cells, however, this nanoparticle diameter led to a deterioration of the cellular redox status and ROS production in a wide range of nanoparticle concentrations. Larger nanoparticles (100 nm-sized and 160 nm-sized), on the other hand, showed a different effect on cancer cells of different origins. In mouse fibroblast L-929 cells, however, 100 nm-sized or 160 nm-sized CeNPs acted in a high concentration range to disrupt mitochondrial membrane potential and activate early apoptosis. High concentrations of CeNPs were required to increase ROS production, reduce redox-status and induce apoptosis in human A-172 glioblastoma cells compared to the hepatocellular carcinoma cell line HepG2 and the breast cancer cell line MCF-7. In the A-172 glioblastoma cells, ER-stress was also not activated and their Ca2+ signaling system was activated by a significantly higher concentration of CeNPs, which could also contribute to the formation of tolerance of this cancer cell line to nanoceria. The Ca2+ signaling system of mouse fibroblasts was found to be highly sensitive to activation by nanoceria and the cells produced Ca2+ signals with higher amplitude compared to A-172 and MCF-7 cells.
Collapse
Affiliation(s)
- Elena G Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia
| | - Sergey V Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilove st., 119991, Moscow, Russia
| | - Egor A Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", 142290, Pushchino, Russia.
| |
Collapse
|
35
|
Cerqua M, Foiani M, Boccaccio C, Comoglio PM, Altintas DM. The integrated stress response drives MET oncogene overexpression in cancers. EMBO J 2025; 44:1107-1130. [PMID: 39774381 PMCID: PMC11832788 DOI: 10.1038/s44318-024-00338-4] [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: 05/17/2024] [Revised: 11/09/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer cells rely on invasive growth to survive in a hostile microenvironment; this growth is characterised by interconnected processes such as epithelial-to-mesenchymal transition and migration. A master regulator of these events is the MET oncogene, which is overexpressed in the majority of cancers; however, since mutations in the MET oncogene are seen only rarely in cancers and are relatively infrequent, the mechanisms that cause this widespread MET overexpression remain obscure. Here, we show that the 5' untranslated region (5'UTR) of MET mRNA harbours two functional stress-responsive elements, conferring translational regulation by the integrated stress response (ISR), regulated by phosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) at serine 52. ISR activation by serum starvation, leucine deprivation, hypoxia, irradiation, thapsigargin or gemcitabine is followed by MET protein overexpression. We mechanistically link MET translation to the ISR by (i) mutation of the two uORFs within the MET 5'UTR, (ii) CRISPR/Cas9-mediated mutation of eIF2α (S52A), or (iii) the application of ISR pathway inhibitors. All of these interventions reduce stress-induced MET overexpression. Finally, we show that blocking stress-induced MET translation blunts MET-dependent invasive growth. These findings indicate that upregulation of the MET oncogene is a functional requirement linking integrated stress response to cancer progression.
Collapse
Affiliation(s)
- Marina Cerqua
- IFOM ETS-The AIRC Institute of Molecular Oncology, 20139, Milano, Italy
| | - Marco Foiani
- IFOM ETS-The AIRC Institute of Molecular Oncology, 20139, Milano, Italy
| | - Carla Boccaccio
- Candiolo Cancer Institute, 10060 Candiolo, Torino, Italy
- Department of Oncology, University of Torino, 10100, Torino, Italy
| | - Paolo M Comoglio
- IFOM ETS-The AIRC Institute of Molecular Oncology, 20139, Milano, Italy.
| | - Dogus M Altintas
- IFOM ETS-The AIRC Institute of Molecular Oncology, 20139, Milano, Italy.
| |
Collapse
|
36
|
Xu S, Xiao S, Qi J, Yao M, He P, Wang R, Wei E, Wang Q, Zhang Y, Tang X, Shen Z. Glucose-regulated protein 78 regulates the subunit-folding of the CCT complex by modulating gene expression and protein interaction in the microsporidian Nosema bombycis. Int J Biol Macromol 2025; 290:138971. [PMID: 39708871 DOI: 10.1016/j.ijbiomac.2024.138971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024]
Abstract
Chaperonin containing tailless complex polypeptide 1 (CCT) functions as a molecular chaperone and is essential for ensuring proper protein folding. Glucose-regulated protein 78 (GRP78/Bip), also a type of chaperone, not only assists in folding of proteins, but also facilitates the transportation of proteins into the endoplasmic reticulum (ER) via the Sec protein complex. In this study, we identified the CCTη of N. bombycis (NbCCTη) for the first time. Immunoprecipitations and mass spectrometry (IP-MS) of NbCCTη analysis showed that NbBip may interact with CCT subunits. Yeast two-hybrid assays validated that NbBip interacts with NbCCTη, as well as NbCCTα and NbCCTε. Furthermore, RNA interference on NbBip brought about radical expression of NbCCTα, NbCCTε, and NbCCTη, while RNAi on NbCCT subunits resulted in abnormal expression of NbBip. Immunofluorescence assay results showed that NbBip colocalized with NbCCTα and NbCCTη, and CCTη colocalized with Nbβ-tubulin and Nbactin in the parasite. Collectively, these findings suggest that NbBip may act as a crucial factor in the subunit-folding and assembly of CCT complex in N. bombycis.
Collapse
Affiliation(s)
- Sheng Xu
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Shengyan Xiao
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Jingru Qi
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Mingshuai Yao
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Ping He
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Runpeng Wang
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Erjun Wei
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China
| | - Qiang Wang
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China; Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, People's Republic of China
| | - Yiling Zhang
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China; Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, People's Republic of China
| | - Xudong Tang
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China; Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, People's Republic of China
| | - Zhongyuan Shen
- Jiangsu University of Science and Technology, Zhenjiang, People's Republic of China; Sericulture Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, People's Republic of China.
| |
Collapse
|
37
|
Koike S, Kimura H, Ogasawara Y. Polysulfide and persulfide-mediated activation of the PERK-eIF2α-ATF4 pathway increases Sestrin2 expression and reduces methylglyoxal toxicity. Redox Biol 2025; 79:103450. [PMID: 39667306 PMCID: PMC11697784 DOI: 10.1016/j.redox.2024.103450] [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: 10/26/2024] [Accepted: 11/29/2024] [Indexed: 12/14/2024] Open
Abstract
Unfolded protein response (UPR) is activated in cells under endoplasmic reticulum (ER) stress. One sensor protein involved in this response is PERK, which is activated through its redox-dependent oligomerization. Prolonged UPR activation is associated with the development and progression of various diseases, making it essential to understanding the redox regulation of PERK. Sulfane sulfur, such as polysulfides and persulfides, can modify the cysteine residues and regulate the function of various proteins. However, the regulatory mechanism and physiological effects of sulfane sulfur on the PERK-eIF2α-ATF4 pathway remain poorly understood. This study focuses on the persulfidation of PERK to elucidate the effects of polysulfides on the PERK-eIF2α-ATF4 pathway and investigate its cytoprotective mechanism. Here, we demonstrated that polysulfide treatment promoted the oligomerization of PERK and PTP1B in neuronal cells using western blotting under nonreducing conditions. We also observed that l-cysteine, a biological source of sulfane sulfur, promoted the oligomerization of PERK and the knockdown of CBS and 3-MST, two sulfane sulfur-producing enzymes, and reduced PERK oligomerization induced by l-cysteine treatment. Furthermore, the band shift assay and LC-MS/MS studies revealed that polysulfides and persulfides induce PTP1B and PERK persulfidation. Additionally, polysulfides promoted eIF2α phosphorylation and ATF4 accumulation in the nucleus, suggesting that polysulfides activate the PERK-eIF2α-ATF4 pathway in neuronal cells. Moreover, polysulfides protected neuronal cells from methylglyoxal-induced toxicity, and this protective effect was reduced when the expression of Sestrin2, regulated by ATF4 activity, was suppressed. This study identified a novel mechanism for the activation of the PERK-eIF2α-ATF4 pathway through persulfidation by polysulfides and persulfides. Interestingly, activation of this pathway overcame the toxicity of methylglyoxal in dependence on Sestrin2 expression. These findings deepen our understanding of neuronal diseases involving ER stress and UPR disturbance and may inspire new therapeutic strategies.
Collapse
Affiliation(s)
- Shin Koike
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Hideo Kimura
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, 1-1-1 Daigaku-Dori, Sanyo-Onoda 756-0884, Yamaguchi, Japan
| | - Yuki Ogasawara
- Department of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan.
| |
Collapse
|
38
|
Yamazaki A, Omura I, Kamikawa Y, Hide M, Tanaka A, Kaneko M, Imaizumi K, Saito A. Unfolded protein response modulates Tyrosinase levels and melanin production during melanogenesis. J Dermatol Sci 2025; 117:36-44. [PMID: 39818444 DOI: 10.1016/j.jdermsci.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/09/2024] [Accepted: 01/05/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND Melanocytes protect the body from ultraviolet radiation by synthesizing melanin. Tyrosinase, a key enzyme in melanin production, accumulates in the endoplasmic reticulum (ER) during melanin synthesis, potentially causing ER stress. However, regulating ER function for melanin synthesis has been less studied than controlling Tyrosinase activity. OBJECTIVE This study investigates the regulatory mechanisms of melanin production, focusing on ER stress and the ER stress-induced response. METHODS B16 mouse melanoma cells induced to undergo melanogenesis were treated with unfolded protein response (UPR) inhibitors or chemical chaperones, and their effects on melanogenesis were analyzed. RESULTS During melanogenesis in B16 cells stimulated by alpha-melanocyte-stimulating hormone (α-MSH), ER stress and UPR activation occurred, accompanied by increased Tyrosinase protein. Reducing IRE1 and ATF6 branch activity lowered melanin levels, while chemical chaperone treatment restored melanin production and increased Tyrosinase levels. CONCLUSION UPR activation, linked to elevated Tyrosinase levels, influences melanin production during melanogenesis. Modulating UPR can regulate melanin synthesis and provides a potential new approach for treating pigmentation disorders.
Collapse
Affiliation(s)
- Akari Yamazaki
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Issei Omura
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Frontier Science and Interdisciplinary Research, Faculty of Medicine, Kanazawa University, Ishikawa, Japan
| | - Yasunao Kamikawa
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Michihiro Hide
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Dermatology, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Akio Tanaka
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masayuki Kaneko
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Kazunori Imaizumi
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Atsushi Saito
- Department of Biochemistry, Institute of Biomedical & Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Frontier Science and Interdisciplinary Research, Faculty of Medicine, Kanazawa University, Ishikawa, Japan.
| |
Collapse
|
39
|
Li Y, Sun S. RNA dysregulation in neurodegenerative diseases. EMBO J 2025; 44:613-638. [PMID: 39789319 PMCID: PMC11790913 DOI: 10.1038/s44318-024-00352-6] [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: 06/19/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Dysregulation of RNA processing has in recent years emerged as a significant contributor to neurodegeneration. The diverse mechanisms and molecular functions underlying RNA processing underscore the essential role of RNA regulation in maintaining neuronal health and function. RNA molecules are bound by RNA-binding proteins (RBPs), and interactions between RNAs and RBPs are commonly affected in neurodegeneration. In this review, we highlight recent progress in understanding dysregulated RNA-processing pathways and the causes of RBP dysfunction across various neurodegenerative diseases. We discuss both established and emerging mechanisms of RNA-mediated neuropathogenesis in this rapidly evolving field. Furthermore, we explore the development of potential RNA-targeting therapeutic approaches for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yini Li
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Shuying Sun
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Departments of Neuroscience, Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
40
|
Giltrap AM, Morris N, Dong YY, Cochrane SA, Krulle T, Hoekman S, Semmelroth M, Wollnik C, Palmai-Pallag T, Carpenter EP, Hollick J, Parkes A, Rudhard Y, Davis BG. Lipid-Modulated, Graduated Inhibition of N-Glycosylation Pathway Priming Suggests Wide Tolerance of ER Proteostasis to Stress. ACS CENTRAL SCIENCE 2025; 11:107-115. [PMID: 39866694 PMCID: PMC11758266 DOI: 10.1021/acscentsci.4c01506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/28/2025]
Abstract
Protein N-glycosylation is a cotranslational modification that takes place in the endoplasmic reticulum (ER). Disruption of this process can result in accumulation of misfolded proteins, known as ER stress. In response, the unfolded protein response (UPR) restores proteostasis or responds by controlling cellular fate, including increased expression of activating transcription factor 4 (ATF4) that can lead to apoptosis. The ability to control and manipulate such a stress pathway could find use in relevant therapeutic areas, such as in treating cancerous states in which the native ER stress response is often already perturbed. The first committed step in the N-glycosylation pathway is therefore a target for potential ER stress modulation. Here, using structure-based design, the scaffold of the natural product tunicamycin allows construction of a panel capable of graduated inhibition of DPAGT1 through lipid-substituent-modulated interaction. The development of a quantitative, high-content, cellular immunofluorescence assay allowed precise determination of downstream mechanistic consequences (through the nuclear localization of key proxy transcription factor ATF4 as a readout of resulting ER stress). Only the most potent inhibition of DPAGT1 generates an ER stress response. This suggests that even low-level "background" biosynthetic flux toward protein glycosylation is sufficient to prevent response to ER stress. "Tuned" inhibitors of DPAGT1 also now seemingly successfully decouple protein glycosylation from apoptotic response to ER stress, thereby potentially allowing access to cellular states that operate at the extremes of normal ER stress.
Collapse
Affiliation(s)
- Andrew M. Giltrap
- The
Rosalind Franklin Institute, Harwell Science & Innovation Campus, Harwell OX11 0FA, U.K.
- Department
of Chemistry, University of Technology Sydney, 15 Broadway, Ultimo, Sydney, NSW 2007, Australia
| | - Niamh Morris
- The
Rosalind Franklin Institute, Harwell Science & Innovation Campus, Harwell OX11 0FA, U.K.
| | - Yin Yao Dong
- MRC
Weatherall Institute of Molecular Medicine, John
Radcliffe Hospital, Oxford OX3 9DS, U.K.
| | | | - Thomas Krulle
- Evotec, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K.
| | - Steven Hoekman
- Evotec, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K.
| | - Martin Semmelroth
- Evotec SE, Manfred Eigen Campus, Essener Bogen
7, 22419 Hamburg, Germany
| | - Carina Wollnik
- Evotec SE, Manfred Eigen Campus, Essener Bogen
7, 22419 Hamburg, Germany
| | - Timea Palmai-Pallag
- The
Rosalind Franklin Institute, Harwell Science & Innovation Campus, Harwell OX11 0FA, U.K.
| | | | - Jonathan Hollick
- Evotec, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K.
| | - Alastair Parkes
- Evotec, 114 Innovation Drive, Milton Park, Abingdon, Oxfordshire OX14 4RZ, U.K.
| | - York Rudhard
- Evotec SE, Manfred Eigen Campus, Essener Bogen
7, 22419 Hamburg, Germany
| | - Benjamin G. Davis
- The
Rosalind Franklin Institute, Harwell Science & Innovation Campus, Harwell OX11 0FA, U.K.
- Department
of Chemistry, University of Oxford, Oxford OX1 3TA, U.K.
- Department
of Pharmacology, University of Oxford, Oxford OX1 3QT, U.K.
| |
Collapse
|
41
|
Buttari B, Tramutola A, Rojo AI, Chondrogianni N, Saha S, Berry A, Giona L, Miranda JP, Profumo E, Davinelli S, Daiber A, Cuadrado A, Di Domenico F. Proteostasis Decline and Redox Imbalance in Age-Related Diseases: The Therapeutic Potential of NRF2. Biomolecules 2025; 15:113. [PMID: 39858508 PMCID: PMC11764413 DOI: 10.3390/biom15010113] [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: 11/26/2024] [Revised: 12/20/2024] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a master regulator of cellular homeostasis, overseeing the expression of a wide array of genes involved in cytoprotective processes such as antioxidant and proteostasis control, mitochondrial function, inflammation, and the metabolism of lipids and glucose. The accumulation of misfolded proteins triggers the release, stabilization, and nuclear translocation of NRF2, which in turn enhances the expression of critical components of both the proteasomal and lysosomal degradation pathways. This process facilitates the clearance of toxic protein aggregates, thereby actively maintaining cellular proteostasis. As we age, the efficiency of the NRF2 pathway declines due to several factors including increased activity of its repressors, impaired NRF2-mediated antioxidant and cytoprotective gene expression, and potential epigenetic changes, though the precise mechanisms remain unclear. This leads to diminished antioxidant defenses, increased oxidative damage, and exacerbated metabolic dysregulation and inflammation-key contributors to age-related diseases. Given NRF2's role in mitigating proteotoxic stress, the pharmacological modulation of NRF2 has emerged as a promising therapeutic strategy, even in aged preclinical models. By inducing NRF2, it is possible to mitigate the damaging effects of oxidative stress, metabolic dysfunction, and inflammation, thus reducing protein misfolding. The review highlights NRF2's therapeutic implications for neurodegenerative diseases and cardiovascular conditions, emphasizing its role in improving proteostasis and redox homeostasis Additionally, it summarizes current research into NRF2 as a therapeutic target, offering hope for innovative treatments to counteract the effects of aging and associated diseases.
Collapse
Affiliation(s)
- Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.B.); (E.P.)
| | - Antonella Tramutola
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University, 00185 Rome, Italy;
| | - Ana I. Rojo
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), National Institute of Health Carlos III (ISCIII), Instituto de Investigación Sanitaria La Paz (IdiPaz), 28049 Madrid, Spain; (A.I.R.); (A.C.)
| | - Niki Chondrogianni
- Institute of Chemical Biology, National Hellenic Research Foundation, 116 35 Athens, Greece;
| | - Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura 00185, Uttar Pradesh, India;
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (L.G.)
| | - Letizia Giona
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.B.); (L.G.)
- PhD Program in Science of Nutrition, Metabolism, Aging and Gender-Related Diseases, Faculty of Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Joana P. Miranda
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisboa, Portugal;
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy; (B.B.); (E.P.)
| | - Sergio Davinelli
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, 86100 Campobasso, Italy;
| | - Andreas Daiber
- Department for Cardiology 1, University Medical Center Mainz, Molecular Cardiology, Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Antonio Cuadrado
- Department of Biochemistry, Faculty of Medicine, Autonomous University of Madrid, Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), National Institute of Health Carlos III (ISCIII), Instituto de Investigación Sanitaria La Paz (IdiPaz), 28049 Madrid, Spain; (A.I.R.); (A.C.)
| | - Fabio Di Domenico
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University, 00185 Rome, Italy;
| |
Collapse
|
42
|
Volloch V, Rits-Volloch S. Production of Amyloid-β in the Aβ-Protein-Precursor Proteolytic Pathway Is Discontinued or Severely Suppressed in Alzheimer's Disease-Affected Neurons: Contesting the 'Obvious'. Genes (Basel) 2025; 16:46. [PMID: 39858593 PMCID: PMC11764795 DOI: 10.3390/genes16010046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
A notion of the continuous production of amyloid-β (Aβ) via the proteolysis of Aβ-protein-precursor (AβPP) in Alzheimer's disease (AD)-affected neurons constitutes both a cornerstone and an article of faith in the Alzheimer's research field. The present Perspective challenges this assumption. It analyses the relevant empirical data and reaches an unexpected conclusion, namely that in AD-afflicted neurons, the production of AβPP-derived Aβ is either discontinued or severely suppressed, a concept that, if proven, would fundamentally change our understanding of the disease. This suppression, effectively self-suppression, occurs in the context of the global inhibition of the cellular cap-dependent protein synthesis as a consequence of the neuronal integrated stress response (ISR) elicited by AβPP-derived intraneuronal Aβ (iAβ; hence self-suppression) upon reaching certain levels. Concurrently with the suppression of the AβPP proteolytic pathway, the neuronal ISR activates in human neurons, but not in mouse neurons, the powerful AD-driving pathway generating the C99 fragment of AβPP independently of AβPP. The present study describes molecular mechanisms potentially involved in these phenomena, propounds novel approaches to generate transgenic animal models of AD, advocates for the utilization of human neuronal cells-based models of the disease, makes verifiable predictions, suggests experiments designed to validate the proposed concept, and considers its potential research and therapeutic implications. Remarkably, it opens up the possibility that the conventional production of AβPP, BACE enzymes, and γ-secretase components is also suppressed under the neuronal ISR conditions in AD-affected neurons, resulting in the dyshomeostasis of AβPP. It follows that whereas conventional AD is triggered by AβPP-derived iAβ accumulated to the ISR-eliciting levels, the disease, in its both conventional and unconventional (triggered by the neuronal ISR-eliciting stressors distinct from iAβ) forms, is driven not (or not only) by iAβ produced in the AβPP-independent pathway, as we proposed previously, but mainly, possibly exclusively, by the C99 fragment generated independently of AβPP and not cleaved at the γ-site due to the neuronal ISR-caused deficiency of γ-secretase (apparently, the AD-driving "substance X" predicted in our previous study), a paradigm consistent with a dictum by George Perry that Aβ is "central but not causative" in AD. The proposed therapeutic strategies would not only deplete the driver of the disease and abrogate the AβPP-independent production of C99 but also reverse the neuronal ISR and ameliorate the AβPP dyshomeostasis, a potentially significant contributor to AD pathology.
Collapse
Affiliation(s)
- Vladimir Volloch
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115, USA
| | - Sophia Rits-Volloch
- Division of Molecular Medicine, Children’s Hospital, Boston, MA 02115, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
43
|
Ji F, Zhang J, Mao L, Tan Y, Ye M, He X, Zhao Y, Liu J, Zhang Y, Zhang N, Shi J, Yan J, Cai X, Zhao B, Jin J, Xu P, Roessler S, Zheng X, Ji J. Liver-specific gene PGRMC1 blocks c-Myc-induced hepatocarcinogenesis through ER stress-independent PERK activation. Nat Commun 2025; 16:50. [PMID: 39747098 PMCID: PMC11696091 DOI: 10.1038/s41467-024-55745-2] [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: 03/07/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025] Open
Abstract
Roles of liver-specific genes (LSGs) in tumor initiation and progression are rarely explored in hepatocellular carcinoma (HCC). Here we show that LSGs are generally downregulated in HCC tumor tissues compared to non-HCC liver tissues, and low-LSG HCCs show poor prognosis and the activated c-Myc pathway. Among the c-Myc- and patient prognosis-associated LSGs, PGRMC1 significantly blocks c-Myc-induced orthotopic HCC formation. The role of PGRMC1 depends on its localization to the endoplasmic reticulum (ER) membrane, where PGRMC1 interacts with PERK through their ER luminal domains. This interaction in turn activates PERK in an ER stress-independent manner, which phosphorylates eIF2α and consequently inhibits c-Myc protein translation. In HCC patients, PGRMC1 level is significantly reduced in tumor tissues and negatively associated with the c-Myc signature. Patients with low-PGRMC1 in their tumors have poor prognosis. Collectively, deregulated LSGs in HCC are associated with the c-Myc pathway activation and PGRMC1 blocks c-Myc-induced hepatic carcinogenesis through promoting ER stress-independent PERK activation.
Collapse
Affiliation(s)
- Fubo Ji
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jianjuan Zhang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Liping Mao
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yaqi Tan
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Meihua Ye
- Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310000, China
| | - Xianglei He
- Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, 310000, China
| | - Yongzhi Zhao
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jiaxin Liu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yan Zhang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Nachuan Zhang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jiong Shi
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, 210008, China
| | - Jianing Yan
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Xiujun Cai
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China
| | - Bin Zhao
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jianping Jin
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Pinglong Xu
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, 69120, Germany
| | - Xin Zheng
- Taoharmony Biotech L.L.C., Hangzhou, Zhejiang, 310018, China
| | - Junfang Ji
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, Zhejiang, 321000, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310016, China.
| |
Collapse
|
44
|
Clarke JPWE, Messmer ML, Pilon J, Reding J, Thibault PA, Salapa HE, Levin MC. Dysfunctional RNA binding protein induced neurodegeneration is attenuated by inhibition of the integrated stress response. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167562. [PMID: 39521193 DOI: 10.1016/j.bbadis.2024.167562] [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: 06/17/2024] [Revised: 10/14/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
Dysfunction of the RNA binding protein heterogeneous nuclear ribonucleoprotein A1 (hnRNP A1) contributes to neurodegeneration, the primary cause of permanent disability in multiple sclerosis (MS). To better understand the role of hnRNP A1 dysfunction in the pathogenesis of neurodegeneration, we utilized optogenetics-driven hnRNP A1 clustering to model its dysfunction in neuron-like differentiated Neuro-2A cells. hnRNP A1 clustering activates the integrated stress response (ISR) and results in a neurodegenerative phenotype marked by decreased neuronal protein translation and neurite loss. Small molecule inhibition of the ISR with either PERKi (GSK2606414) or ISRIB (integrated stress response inhibitor) attenuated both the decrease in neuronal translation and neurite loss, without affecting hnRNP A1 clustering. We then confirmed a strong association between hnRNP A1 clustering and ISR activation in neurons from MS brains. These data illustrate that hnRNP A1 dysfunction promotes neurodegeneration by activation of the ISR in vitro and in vivo, thus revealing a novel therapeutic target to reduce neurodegeneration and subsequent disability in MS.
Collapse
Affiliation(s)
- Joseph-Patrick W E Clarke
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada.
| | - Miranda L Messmer
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | - Jacob Pilon
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Health Sciences, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | - Jenna Reding
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | - Patricia A Thibault
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | - Hannah E Salapa
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada
| | - Michael C Levin
- Office of the Saskatchewan Multiple Sclerosis Clinical Research Chair, University of Saskatchewan, Saskatoon, SK S7K-0M7, Canada; Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK S7K 0M7, Canada; Department of Medicine, Neurology Division, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada; Department of Health Sciences, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada; Department of Anatomy, Physiology and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N-5E5, Canada.
| |
Collapse
|
45
|
Majerciak V, Zheng ZM. Induction of translation-suppressive G3BP1 + stress granules and interferon-signaling cGAS condensates by transfected plasmid DNA. HLIFE 2025; 3:21-37. [PMID: 40078969 PMCID: PMC11902918 DOI: 10.1016/j.hlife.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Plasmid DNA transfection is one of the fundamental tools of biomedical research. Here, we found that plasmid DNA transfection mediated by liposomes activates multiple innate immune responses in several widely used cell lines. Their activations were visible by detection of stress granules (SG) and cGAS-DNA condensates (cGC) in the transfected cells in a plasmid DNA dose-dependent manner. The elevated levels of phosphorylated eukaryotic translation initiation factor 2 subunit alpha (eIF2α), interferon regulatory factor 3 (IRF3), and signal transducer and activator of transcription 1 (STAT1) were induced in plasmid DNA-transfected cells. The formation of SG but not cGC required active transcription and formation of dsRNA in transfected cells. Plasmid DNA-induced SG or cGC were mutually exclusive because of triggering two distinct pathways. Knockdown (KD) of PKR before plasmid DNA transfection led to abolish SG without affecting cGC formation. Conversely, cGAS KD could prevent cGC without affecting SG formation. In addition, plasmid DNA-induced SG and cGC formation could be prevented, respectively, by co-expression of KSHV proteins ORF57 (PKR inhibitor) and ORF52 (cGAS inhibitor). Inhibition of SG formation mediated by PKR KD, but not cGC KD, also led to increased expression of transgenes, indicating that PKR activation represents a major roadblock to gene expression. Together, these data indicate that plasmid DNA triggers innate immune responses in the transfected cells and causes a significant cellular perturbation that should be considered during experiment design and data interpretation.
Collapse
Affiliation(s)
- Vladimir Majerciak
- Tumor Virus RNA Biology Section, HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Maryland, USA
| | | |
Collapse
|
46
|
Ali A, Matveyenka M, Pickett DN, Rodriguez A, Kurouski D. Tubulin-Binding Region Modulates Cholesterol-Triggered Aggregation of Tau Proteins. J Neurochem 2025; 169:e16294. [PMID: 39777699 PMCID: PMC11731895 DOI: 10.1111/jnc.16294] [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: 09/19/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 01/11/2025]
Abstract
A hallmark of Alzheimer disease (AD) and tauopathies, severe neurodegenerative diseases, is the progressive aggregation of Tau, also known as microtubule-associated Tau protein. Full-length Tau1-441, also known as 2N4R, contains two N-terminal inserts that bind to tubulin. This facilitates the self-assembly of tubulin simultaneously enhancing stability of cell microtubules. Other Tau isoforms have one (1N4R) or zero (0N4R) N-terminal inserts, which makes 2N4R Tau more and 0N4R less effective in promoting microtubule self-assembly. A growing body of evidence indicates that lipids can alter the aggregation rate of Tau isoforms. However, the role of N-terminal inserts in Tau-lipid interactions remains unclear. In this study, we utilized a set of biophysical methods to determine the extent to which N-terminal inserts alter interactions of Tau isoforms with cholesterol, one of the most important lipids in plasma membranes. Our results showed that 2 N insert prevents amyloid-driven aggregation of Tau at the physiological concentration of cholesterol, while the absence of this N-terminal repeat (1N4R and 0N4R Tau) resulted in the self-assembly of Tau into toxic amyloid fibrils. We also found that the presence of cholesterol in the lipid bilayers caused a significant increase in the cytotoxicity of 1N4R and 0N4R Tau to neurons. This effect was not observed for 2N4R Tau fibrils formed in the presence of lipid membranes with low, physiological, and elevated concentrations of cholesterol. Using molecular assays, we found that Tau aggregates primarily exert cytotoxicity by damaging cell endosomes, endoplasmic reticulum, and mitochondria.
Collapse
Affiliation(s)
- Abid Ali
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Mikhail Matveyenka
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Davis N Pickett
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Axell Rodriguez
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Dmitry Kurouski
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
47
|
Tak J, Kim YS, Kim SG. Roles of X-box binding protein 1 in liver pathogenesis. Clin Mol Hepatol 2025; 31:1-31. [PMID: 39355873 PMCID: PMC11791611 DOI: 10.3350/cmh.2024.0441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/06/2024] [Accepted: 09/27/2024] [Indexed: 10/03/2024] Open
Abstract
The prevalence of drug-induced liver injury (DILI) and viral liver infections presents significant challenges in modern healthcare and contributes to considerable morbidity and mortality worldwide. Concurrently, metabolic dysfunctionassociated steatotic liver disease (MASLD) has emerged as a major public health concern, reflecting the increasing rates of obesity and leading to more severe complications such as fibrosis and hepatocellular carcinoma. X-box binding protein 1 (XBP1) is a distinct transcription factor with a basic-region leucine zipper structure, whose activity is regulated by alternative splicing in response to disruptions in endoplasmic reticulum (ER) homeostasis and the unfolded protein response (UPR) activation. XBP1 interacts with a key signaling component of the highly conserved UPR and is critical in determining cell fate when responding to ER stress in liver diseases. This review aims to elucidate the emerging roles and molecular mechanisms of XBP1 in liver pathogenesis, focusing on its involvement in DILI, viral liver infections, MASLD, fibrosis/cirrhosis, and liver cancer. Understanding the multifaceted functions of XBP1 in these liver diseases offers insights into potential therapeutic strategies to restore ER homeostasis and mitigate liver damage.
Collapse
Affiliation(s)
- Jihoon Tak
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Korea
| | - Yun Seok Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Korea
| |
Collapse
|
48
|
Cerrato G, Liu P, Zhao L, Petrazzuolo A, Humeau J, Schmid ST, Abdellatif M, Sauvat A, Kroemer G. AI-based classification of anticancer drugs reveals nucleolar condensation as a predictor of immunogenicity. Mol Cancer 2024; 23:275. [PMID: 39702289 DOI: 10.1186/s12943-024-02189-3] [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: 10/04/2024] [Accepted: 11/28/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND Immunogenic cell death (ICD) inducers are often identified in phenotypic screening campaigns by the release or surface exposure of various danger-associated molecular patterns (DAMPs) from malignant cells. This study aimed to streamline the identification of ICD inducers by leveraging cellular morphological correlates of ICD, specifically the condensation of nucleoli (CON). METHODS We applied artificial intelligence (AI)-based imaging analyses to Cell Paint-stained cells exposed to drug libraries, identifying CON as a marker for ICD. CON was characterized using SYTO 14 fluorescent staining and holotomographic microscopy, and visualized by AI-deconvoluted transmitted light microscopy. A neural network-based quantitative structure-activity relationship (QSAR) model was trained to link molecular descriptors of compounds to the CON phenotype, and the classifier was validated using an independent dataset from the NCI-curated mechanistic collection of anticancer agents. RESULTS CON strongly correlated with the inhibition of DNA-to-RNA transcription. Cytotoxic drugs that inhibit RNA synthesis without causing DNA damage were as effective as conventional cytotoxicants in inducing ICD, as demonstrated by DAMPs release/exposure and vaccination efficacy in mice. The QSAR classifier successfully predicted drugs with a high likelihood of inducing CON. CONCLUSIONS We developed AI-based algorithms for predicting CON-inducing drugs based on molecular descriptors and their validation using automated micrographs analysis, offering a new approach for screening ICD inducers with minimized adverse effects in cancer therapy.
Collapse
Affiliation(s)
- Giulia Cerrato
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France.
- Onco-Pheno-Screen Platform, Centre de Recherche des Cordeliers, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
| | - Peng Liu
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France
- Onco-Pheno-Screen Platform, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Liwei Zhao
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France
- Onco-Pheno-Screen Platform, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Adriana Petrazzuolo
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- International Centre for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Sciences, Università Degli Studi di Milano, Milan, Italy
| | - Juliette Humeau
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche en Cancérologie de Lyon (CRCL), Equipe Oncopharmacologie, Faculté Rockfeller, Lyon, France
| | - Sophie Theresa Schmid
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Mahmoud Abdellatif
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Allan Sauvat
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France.
- Onco-Pheno-Screen Platform, Centre de Recherche des Cordeliers, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Institut Universitaire de France, Sorbonne Université, Inserm U1138, Paris, France.
- Onco-Pheno-Screen Platform, Centre de Recherche des Cordeliers, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Department of Biology, Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
- Centre de Recherche des Cordeliers, 15 Rue de l'École de Médecine, Paris, 75006, France.
| |
Collapse
|
49
|
Chowdhury D, Jang CE, Lajoie P, Renaud SJ. A stress paradox: the dual role of the unfolded protein response in the placenta. Front Endocrinol (Lausanne) 2024; 15:1525189. [PMID: 39758342 PMCID: PMC11695235 DOI: 10.3389/fendo.2024.1525189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 12/03/2024] [Indexed: 01/07/2025] Open
Abstract
The placenta is a temporary organ that forms during pregnancy and is essential for fetal development and maternal health. As an endocrine organ, proper placental function requires continual production, folding, and transport of proteins and lipids. Central to these processes is the endoplasmic reticulum (ER), a dynamic organelle responsible for maintaining cellular protein and lipid synthesis and processing. ER stress occurs when there is an accumulation of unfolded or misfolded proteins, which triggers the activation of cellular pathways collectively called the unfolded protein response. Unfolded protein response pathways act to alleviate the misfolded protein burden and restore ER homeostasis, or if unresolved, initiate cell death. While prolonged ER stress has been linked to deficient placental function and adverse pregnancy outcomes, basal activation of unfolded protein response pathways is required for placental development and function. This review explores the importance of ER homeostasis in placental development and function, examining how disruptions in ER stress responses may contribute to adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Diba Chowdhury
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Chloe E. Jang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Children’s Health Research Institute, Lawson Health Research Institute, London, ON, Canada
| | - Patrick Lajoie
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Children’s Health Research Institute, Lawson Health Research Institute, London, ON, Canada
| | - Stephen J. Renaud
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Children’s Health Research Institute, Lawson Health Research Institute, London, ON, Canada
| |
Collapse
|
50
|
Ill CR, Marikar NC, Nguyen V, Nangia V, Darnell AM, Vander Heiden MG, Reigan P, Spencer SL. BRAF V600 and ErbB inhibitors directly activate GCN2 in an off-target manner to limit cancer cell proliferation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.19.629301. [PMID: 39763857 PMCID: PMC11702603 DOI: 10.1101/2024.12.19.629301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Targeted kinase inhibitors are well known for their promiscuity and off-target effects. Herein, we define an off-target effect in which several clinical BRAFV600 inhibitors, including the widely used dabrafenib and encorafenib, interact directly with GCN2 to activate the Integrated Stress Response and ATF4. Blocking this off-target effect by co-drugging with a GCN2 inhibitor in A375 melanoma cells causes enhancement rather than suppression of cancer cell outgrowth, suggesting that the off-target activation of GCN2 is detrimental to these cells. This result is mirrored in PC9 lung cancer cells treated with erlotinib, an EGFR inhibitor, that shares the same off-target activation of GCN2. Using an in silico kinase inhibitor screen, we identified dozens of FDA-approved drugs that appear to share this off-target activation of GCN2 and ATF4. Thus, GCN2 activation may modulate the therapeutic efficacy of some kinase inhibitors, depending on the cancer context.
Collapse
Affiliation(s)
- C Ryland Ill
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Nasreen C Marikar
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Vu Nguyen
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz, Aurora, CO, USA
| | - Varuna Nangia
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
- University of Colorado School of Medicine, University of Colorado Anschutz, Aurora, CO, USA
| | - Alicia M Darnell
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, MA, USA
- Current address: Department of Pharmacology and Cancer Biology, Duke University School of Medicine, NC, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, MA, USA
- Dana-Farber Cancer Institute, MA, USA
| | - Philip Reigan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz, Aurora, CO, USA
| | - Sabrina L Spencer
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|