1
|
Chen F, Chen R, Yang L, Shen B, Wang Y, Gao Y, Tan R, Zhao X. Magnesium-assisted hydrogen improves isoproterenol-induced heart failure. Med Gas Res 2025; 15:459-470. [PMID: 40300881 PMCID: PMC12124708 DOI: 10.4103/mgr.medgasres-d-24-00135] [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: 12/06/2024] [Revised: 02/07/2025] [Accepted: 03/19/2025] [Indexed: 05/01/2025] Open
Abstract
Heart failure (HF) is a leading cause of mortality among patients with cardiovascular disease and is often associated with myocardial apoptosis and endoplasmic reticulum stress (ERS). While hydrogen has demonstrated potential in reducing oxidative stress and ERS, recent evidence suggests that magnesium may aid in hydrogen release within the body, further enhancing these protective effects. This study aimed to investigate the cardioprotective effects of magnesium in reducing apoptosis and ERS through hydrogen release in a rat model of isoproterenol (ISO)-induced HF. Magnesium was administered orally to ISO-induced HF rats, which improved cardiac function, reduced myocardial fibrosis and cardiac hypertrophy, and lowered the plasma levels of creatine kinase-MB, cardiac troponin-I, and N-terminal B-type natriuretic peptide precursor in ISO-induced HF rats. It also inhibited cardiomyocyte apoptosis by upregulating B-cell lymphoma-2, downregulating Bcl-2-associated X protein, and suppressing ERS markers (glucose-related protein 78, activating transcription factor 4, and C/EBP-homologous protein). Magnesium also elevated hydrogen levels in blood, plasma, and cardiac tissue, as well as in artificial gastric juice and pure water, where hydrogen release lasted for at least four hours. Additionally, complementary in vitro experiments were conducted using H9C2 cardiomyocyte injury models, with hydrogen-rich culture medium as the intervention. Hydrogen-rich culture medium improved the survival and proliferation of ISO-treated H9C2 cells, reduced the cell surface area, inhibited apoptosis, and downregulated ERS pathway proteins. However, the protective effects of hydrogen were negated by tunicamycin (an inducer of ERS) in H9C2 cells. In conclusion, magnesium exerts significant cardioprotection by mitigating ERS and apoptosis through hydrogen release effects in ISO-induced HF.
Collapse
Affiliation(s)
- Fengbao Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Ruimin Chen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Lili Yang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
- New Drug Evaluation Center of Shandong Academy of Pharmaceutical Sciences, Shandong Academy of Pharmaceutical Sciences, Ji’nan, Shandong Province, China
| | - Bowen Shen
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
- School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Yunting Wang
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Yongfeng Gao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Rui Tan
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| | - Xiaomin Zhao
- Institute of Pharmacology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, Shandong Province, China
| |
Collapse
|
2
|
Xu W, Li W, Kuai D, Li Y, Sun W, Liu X, Xu B. Identification of endoplasmic reticulum stress-related genes as prognostic markers in colon cancer. Cancer Biol Ther 2025; 26:2458820. [PMID: 40169935 PMCID: PMC11970746 DOI: 10.1080/15384047.2025.2458820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 12/20/2024] [Accepted: 01/22/2025] [Indexed: 04/03/2025] Open
Abstract
Endoplasmic reticulum stress (ERS) has been implicated in the pathogenesis of various cancers, including colon cancer, by regulating tumor cell survival, growth, and immune response. However, the specific genes involved in ERS that could serve as prognostic markers in colon cancer remain underexplored. This study aims to identify and validate endoplasmic reticulum stress related genes (ERSRGs) in colon cancer that correlate with patient prognosis, thereby enhancing the understanding of ERS in oncological outcomes and potential therapeutic targeting. We utilized bioinformatics analyses to identify ERSRGs from publicly available colon cancer datasets. Differential expression analysis and survival analysis were performed to assess the prognostic significance of these genes. Validation was conducted through quantitative real-time PCR (RT-qPCR) on selected colon cancer cell lines. Our study identified nine ERS related genes (ASNS, ATF4, ATF6B, BOK, CLU, DDIT3, MANF, SLC39A14, TRAF2) involved in critical pathways including IL-12, PI3K-AKT, IL-7, and IL-23 signaling, and linked to 1-, 3-, and 5-year survival of patients with colon cancer. A multivariate Cox model based on these ERS related genes demonstrated significant prognostic power. Further, TRAF2 strong correlated with immune cells infiltration, suggesting its potential roles in modulating immune responses in the tumor microenvironment. The RT-qPCR validation confirmed the differential expression of these genes in human colon cancer cell lines versus human normal colonic epithelial cell line. The identified ERSRGs could serve as valuable prognostic markers and may offer new insights into the therapeutic targeting of ERS in colon cancer.
Collapse
Affiliation(s)
- Wenjing Xu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Wei Li
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Dayu Kuai
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Yaqiang Li
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Wei Sun
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xian Liu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| | - Baohong Xu
- Department of Gastroenterology, Beijing Luhe Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Zhang W, Ma F, Su X, Zhu M, Wang X. Antimicrobial peptide WK-13-3D promotes apoptosis, autophagy, and ubiquitination in triple-negative breast cancer via binding immunoglobulin protein (BiP). Chem Biol Interact 2025; 415:111530. [PMID: 40294882 DOI: 10.1016/j.cbi.2025.111530] [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: 01/30/2025] [Revised: 04/08/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE To elucidate the inhibitory mechanism of antimicrobial peptide WK-13-3D on triple-negative breast cancer (TNBC) by targeting the binding immunoglobulin protein (BiP), a key endoplasmic reticulum (ER) chaperone regulating unfolded protein response and tumor survival. METHODS TNBC cell lines (MDA-MB-231 and MDA-MB-468) were treated with WK-13-3D to assess proliferation, migration, invasion, and apoptosis. Pull-down assays identified interacting proteins, and Western blotting (WB) analyzed alterations in BiP, PERK, eIF2α, p-eIF2α, Caspase3, Cleaved-Caspase3, Bax, LC3, P62, AKT, p-AKT, mTOR, and p-mTOR. Transmission electron microscopy examined intracellular structures, while qPCR measured BiP mRNA levels. The effects of WK-13-3D on BiP ubiquitination were explored via co-immunoprecipitation (Co-IP). Animal tumor models were used to confirm the inhibitory effects, with BiP and Ki67 (a nuclear proliferation marker indicating actively dividing tumor cells) expression analyzed by immunohistochemistry (IHC). RESULTS WK-13-3D inhibited TNBC cell proliferation, migration, and invasion, while promoting apoptosis. Pull-down experiments identified 268 interacting proteins, with BiP being the most frequent. Databases (TIMER and TCGA) showed high BiP expression in breast cancer, associated with poor prognosis. WB assays revealed that WK-13-3D activated ER stress-induced apoptosis and autophagy via BiP. Co-IP demonstrated that WK-13-3D mediated BiP ubiquitination at sites 352 and 547 through K6 and K29 chains. IHC analysis further confirmed decreased Ki67 levels in WK-13-3D-treated tumors, reflecting suppressed proliferative activity. Animal experiments confirmed tumor growth inhibition. CONCLUSION WK-13-3D promotes apoptosis, autophagy and Ubiquitination in TNBC by modulating BiP.
Collapse
Affiliation(s)
- Wenjing Zhang
- College of Laboratory Medicine, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Fei Ma
- College of Laboratory Medicine, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Xuhong Su
- College of Laboratory Medicine, Ningxia Medical University, Yinchuan, 750004, PR China
| | - Mingxing Zhu
- College of Basic Medical Sciences, Ningxia Medical University, Yinchuan, 750004, PR China.
| | - Xiuqing Wang
- College of Laboratory Medicine, Ningxia Medical University, Yinchuan, 750004, PR China; Ningxia Key Laboratory of Clinical Pathogenic Microorganisms, Ningxia Medical University, Yinchuan, 750004, PR China.
| |
Collapse
|
4
|
Wang J, Bai Y, Liu XY, Li S, Wang Y, Zhang YM, Hong ZY. NFAT2 promotes sorafenib resistance in hepatocellular carcinoma cells by modulating calcium ion signalling. Arch Biochem Biophys 2025; 769:110440. [PMID: 40288600 DOI: 10.1016/j.abb.2025.110440] [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: 01/05/2025] [Revised: 04/01/2025] [Accepted: 04/24/2025] [Indexed: 04/29/2025]
Abstract
Treating hepatocellular carcinoma (HCC) remains challenging due to the drug resistance of HCC cells, which limits the clinical efficacy of sorafenib. This study elucidates the role of nuclear factor activated T cells 2 (NFAT2) in sorafenib resistance in HCC cells and reveals the underlying mechanism. Sorafenib-resistant cell lines were constructed, with NFAT2 overexpressed and knocked down via genetic engineering. Fura-2 detected intracellular calcium ion concentration; transmission electron microscopy (TEM) assessed organelle damage; wound healing and transwell assays evaluated cell migration and invasion; clone formation and CCK8 assays measured cell proliferation. Flow cytometry detected apoptosis; Western blot analyzed protein expressions. Tumorigenesis was evaluated using a sorafenib-resistant HCC orthotopic xenograft mouse model. We found that NFAT2 was upregulated in MHCC97H-SR and HepG2-SR cells. Overexpression of NFAT2 inhibited Ca2+ influx in MHCC97H-SR, reduced the expression of Ca2+ regulation-related proteins (p-PLCγ, p-IP3R, p-CaMKII), ER-related proteins (CPR94, CPR78), and oxidative stress-related proteins (NOX2, NOX4). NFAT2 overexpression inhibited apoptosis and enhanced cell migration, invasion, and proliferation. NFAT2 knockdown reduced tumorigenesis. Our study uncovered a mechanism by which NFAT2 increases HCC cell resistance to sorafenib by altering intracellular calcium ion signals, highlighting NFAT2 as a promising target for HCC drug therapy.
Collapse
Affiliation(s)
- Jian Wang
- Department of Comprehensive Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Yi Bai
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Xian-Yi Liu
- Department of Comprehensive Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Shuang Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, China
| | - Ying Wang
- Department of Comprehensive Surgery, Tianjin First Central Hospital, Tianjin, China
| | - Ya-Min Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Tianjin First Central Hospital, Tianjin, China.
| | - Zhang-Yong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
5
|
Yousef EH, El Gayar AM, El-Magd NFA. Insights into Sorafenib resistance in hepatocellular carcinoma: Mechanisms and therapeutic aspects. Crit Rev Oncol Hematol 2025; 212:104765. [PMID: 40389183 DOI: 10.1016/j.critrevonc.2025.104765] [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: 02/05/2025] [Revised: 05/07/2025] [Accepted: 05/11/2025] [Indexed: 05/21/2025] Open
Abstract
The most prevalent primary hepatic cancer, hepatocellular carcinoma (HCC), has a bad prognosis. HCC prevalence and related deaths have increased in recent decades. Food and Drug Administration (FDA) has licensed Sorafenib as a first-line treatment for individuals with advanced HCC. Despite this, some clinical studies indicate that a significant percentage of liver cancer patients exhibit insensitivity to sorafenib. Furthermore, the overall effectiveness of sorafenib is far from adequate, and the number of patients who benefit from therapy is low. In recent years, many researchers have focused on the mechanisms underlying sorafenib resistance. Acquired resistance to sorafenib in HCC cells has been reported to be facilitated by dysregulation of signal transducer and activator of transcription 3 (STAT3) activation, angiogenesis, autophagy, hypoxia-induced pathways, epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs), ferroptosis, and non-coding RNAs (ncRNAs). Recent clinical trials, including comparisons of sorafenib with immune checkpoint inhibitors like tislelizumab, have shown promise in improving patient outcomes. Additionally, combination therapies targeting complementary pathways are under investigation to overcome resistance and enhance treatment efficacy. The limitation of Sorafenib's effectiveness has been partially but not completely clarified. Furthermore, while certain regimens have demonstrated positive results, more clinical trials are required to confirm them. Future research should focus on identifying predictive biomarkers for therapy response, targeting the tumor microenvironment, and exploring novel therapeutic agents and personalized medicine strategies. A deeper understanding of these mechanisms will be essential for developing more effective therapeutic approaches and improving the prognosis of patients with advanced HCC. This article discusses strategies that may be employed to enhance the success of treatment and summarizes new research on the possible pathways that lead to sorafenib resistance.
Collapse
Affiliation(s)
- Eman H Yousef
- Biochemistry department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Pharmacology and Biochemistry department, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34511, Egypt.
| | - Amal M El Gayar
- Biochemistry department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Nada F Abo El-Magd
- Biochemistry department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
6
|
Wang L, Gao X, Zuo X, Wang T, Shi X. Prognostic value of circadian rhythm-associated genes in breast cancer. World J Surg Oncol 2025; 23:186. [PMID: 40369575 PMCID: PMC12077051 DOI: 10.1186/s12957-025-03829-8] [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: 01/07/2025] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
OBJECTIVE Breast cancer (BC) remains the most prevalent malignancy among women. Clinical evidence indicates that genetic variations related to circadian rhythms, as well as the timing of therapeutic interventions, influence the response to radiation therapy and the toxicity of pharmacological treatments in women with BC. This study aimed to identify key circadian rhythm-related genes (CRGs) using bioinformatics and machine learning, and construct a prognostic model to predict clinical outcomes. METHODS Transcriptome data for BC were retrieved from The Cancer Genome Atlas database. Univariate Cox regression and least absolute shrinkage and selection operator regression analyses were used to develop a prognostic model based on CRGs. The predictive performance of the risk score model was evaluated. Univariate and multivariate Cox regression analyses were applied to construct the prognostic model and stratify patients into high-risk and low-risk groups. Additionally, differences in immune microenvironment, immunotherapy efficacy, and tumor mutation burden were assessed between risk groups. RESULTS A prognostic risk score model comprising 17 CRGs was developed. The areas under the receiver operating characteristic curve for overall survival at 1, 3, 5, and 7 years exceeded 0.6, indicating acceptable predictive performance. Calibration plots and decision curve analyses demonstrated the use of the model in prognostic prediction. Significant differences in immune microenvironment, immunotherapy efficacy, and tumor mutation burden were identified between the low-risk and high-risk groups. CONCLUSION The circadian rhythm-based gene model, effectively predicted the prognosis of individuals with BC, highlighting its potential to inform personalized therapeutic strategies and improve patient outcomes.
Collapse
Affiliation(s)
- Ling Wang
- Department of Breast Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang 5th, Dongcheng District, Beijing, 100700, China
| | - Xiang Gao
- Department of Breast Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang 5th, Dongcheng District, Beijing, 100700, China
| | - Ximeng Zuo
- Department of Breast Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang 5th, Dongcheng District, Beijing, 100700, China
| | - Tangshun Wang
- Department of Breast Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang 5th, Dongcheng District, Beijing, 100700, China
| | - Xiaoguang Shi
- Department of Breast Surgery, Dongzhimen Hospital, Beijing University of Chinese Medicine, Haiyuncang 5th, Dongcheng District, Beijing, 100700, China.
| |
Collapse
|
7
|
Xiao W, Gao M, Mo B, Huang X, Du Z, Wang S, Chen J, Luo S, Xing H. Endoplasmic Reticulum-Targeted Phototherapy Remodels the Tumor Immunopeptidome to Enhance Immunogenic Cell Death and Adaptive Anti-Tumor Immunity. Pharmaceuticals (Basel) 2025; 18:491. [PMID: 40283929 PMCID: PMC12030737 DOI: 10.3390/ph18040491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Endoplasmic reticulum (ER)-targeted phototherapy has emerged as a promising approach to amplify ER stress, induce immunogenic cell death (ICD), and enhance anti-tumor immunity. However, its impact on the antigenicity of dying tumor cells remains poorly understood. Methods: Laser activation of the ER-targeted photosensitizer ER-Cy-poNO2 was performed to investigate its effects on tumor cell antigenicity. Transcriptomic analysis was carried out to assess gene expression changes. Immunopeptidomics profiling was used to identify high-affinity major histocompatibility complex class I (MHC-I) ligands. In vitro functional studies were conducted to evaluate dendritic cell maturation and T lymphocyte activation, while in vivo experiments were performed by combining the identified peptide with poly IC to evaluate anti-tumor immunity. Results: Laser activation of ER-Cy-poNO2 significantly remodeled the antigenic landscape of 4T-1 tumor cells, enhancing their immunogenicity. Transcriptomic analysis revealed upregulation of antigen processing and presentation pathways. Immunopeptidomics profiling identified multiple high-affinity MHC-I ligands, with IF4G3986-994 (QGPKTIEQI) showing exceptional immunogenicity. In vitro, IF4G3986-994 promoted dendritic cell maturation and enhanced T lymphocytes activation. In vivo, the combination of IF4G3986-994 with poly IC elicited robust anti-tumor immunity, characterized by increased CD8+ T lymphocytes infiltration, reduced regulatory T cells (Tregs) in the tumor microenvironment, elevated systemic Interferon-gamma (IFN-γ) levels, and significant tumor growth inhibition without systemic toxicity. Conclusions: These findings establish a mechanistic link between ER stress-driven ICD, immunopeptidome remodeling, and adaptive immune activation, highlighting the potential of ER-targeted phototherapy as a platform for identifying immunogenic peptides and advancing peptide-based cancer vaccines.
Collapse
Affiliation(s)
- Weidong Xiao
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Mingquan Gao
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Banghui Mo
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Xie Huang
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zaizhi Du
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shufeng Wang
- Department of Immunology, College of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Jianhong Chen
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| | - Shenglin Luo
- State Key Laboratory of Trauma and Chemical Poisoning, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Haiyan Xing
- Department of Pharmacy, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing 400042, China
| |
Collapse
|
8
|
Zhang R, Zhang Z, Xie L, Yu Z, Gao R, Zhang ZR, Zhang Y, Wei X, Chen Y, Jiao S, Gao Y, Guo JP. In vitro analysis of the molecular mechanisms of ursolic acid against ovarian cancer. BMC Complement Med Ther 2025; 25:65. [PMID: 39984915 PMCID: PMC11846399 DOI: 10.1186/s12906-025-04808-y] [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: 12/20/2023] [Accepted: 02/03/2025] [Indexed: 02/23/2025] Open
Abstract
Ovarian cancer is one of most common gynaecologic malignancy and ranks third in cancer-related deaths among women. Ursolic acid (UA) is a pharmacologically active pentacyclic triterpenoid isolated from a large variety of vegetables, fruits and many traditional medicinal plants. However, the mechanism of action of UA in inhibiting the proliferation of ovarian cancer cells remains unclear. Consequently, this experiment was designed to elucidate the mechanism of action of UA in inhibiting the proliferation of ovarian cancer cells in greater detail.The results indicated that UA was capable of effectively inhibiting the proliferation, migration, and colony formation of ovarian cancer cells.UA was observed to up-regulate Bcl-2-associated X protein(BAX)and cysteinyl aspartate specific proteinase 3 (Caspase3) expression and down-regulating B-cell lymphoma-2(Bcl-2) expression.Meanwhile, UA up-regulated Sequestosome 1(p62)expression and down-regulated coiled-coil, moesin-like BCL2-interacting protein(Becline1), microtubule-associated proteins light chain 3(LC3), Phosphoinositide 3-Kinase(PI3K), andProtein Kinase B( AKT) expression, thus effectively inhibiting autophagy in ovarian cancer cells.Furthermore, UA upregulated pancreatic ER kinase (PKR)-like ER kinase (PERK), eukaryotic translation initiation factor 2 A(eIF2α), and The C/EBP Homologous Protein(CHOP) expression.In addition UA upregulates PERK, eIF2α, and CHOP expression and effectively promotes endoplasmic reticulum stress(ERS).In conclusion, UA can inhibit ovarian cancer cell proliferation, migration, colony formation, and may inhibit tumor cell autophagy by promoting tumor cell ERS, and ultimately promote ovarian cancer cell apoptosis.
Collapse
Affiliation(s)
- Ru Zhang
- School of Clinical Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Zhaopeng Zhang
- School of Pharmacy, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Lulu Xie
- Affiliated Hospital, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Ziqing Yu
- School of Clinical Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Rui Gao
- School of Clinical Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Zhi-Run Zhang
- School of Clinical Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Ying Zhang
- School of Clinical Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Xuyang Wei
- School of Clinical Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Yang Chen
- School of Clinical Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Sue Jiao
- School of Clinical Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Yiren Gao
- Affiliated Hospital, Changchun University of Traditional Chinese Medicine, Changchun, China.
| | - Jun-Peng Guo
- School of Clinical Medicine, Changchun University of Traditional Chinese Medicine, Changchun, China.
| |
Collapse
|
9
|
Liu Z, Liu Q, Zeng A, Song L. Regulatory function of endoplasmic reticulum stress in colorectal cancer: Mechanism, facts, and perspectives. Int Immunopharmacol 2025; 147:114024. [PMID: 39764998 DOI: 10.1016/j.intimp.2025.114024] [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: 10/08/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/29/2025]
Abstract
Colorectal cancer (CRC) is an exceedingly common and profoundly impactful malignancy of the digestive system, posing a grave threat to human health. Endoplasmic reticulum stress (ERS) is an intracellular biological reaction that mobilizes the unfolded protein response (UPR) to tackling dysregulation in protein homeostasis. This process subtly modulates the cell to either restore normal cellular function or steer it towards apoptosis. The high metabolic demands of CRC cells sculpt a rigorous tumor microenvironment (TME), compelling CRC cells to experience ERS. Adaptive responses induced by mild ERS furnish the necessary conditions for the survival of CRC cells, whereas the cell death mechanisms triggered by sustained ERS could be considered a prospective strategy for cancer therapy. Considering the complex regulation of ERS in cancer development, this article offers a comprehensive review of the molecular mechanisms through which ERS influences CRC fate. It provides crucial insights for exploring the role of ERS in the occurrence and progression of CRC, laying a new theoretical foundation for devising precise therapeutic strategies targeting ERS. Furthermore, by synthesizing extensive clinical and preclinical studies, we delve into therapeutic strategies targeting ERS, including the potential of targeting ERS in immunotherapy, the utilization of native compounds, advancements in proteasome inhibitors, and the potential synergies of these strategies with traditional chemotherapy agents and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Zihan Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan 610041, China.
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
10
|
Hu H, Wen F, Zhen T, Zhang M, Qin J, Huang J, Chen Z, Yu M, Hu S, Fang M, Zeng JZ. Design, synthesis, and biological evaluation of N 1-(2-(adamantan-1-yl)-1H-indol-5-yl)-N 2-(substituent)-1,2-dicarboxamides as anticancer agents targeting Nur77-mediated endoplasmic reticulum stress. Bioorg Chem 2025; 155:108113. [PMID: 39787915 DOI: 10.1016/j.bioorg.2024.108113] [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: 10/31/2024] [Revised: 12/17/2024] [Accepted: 12/28/2024] [Indexed: 01/12/2025]
Abstract
Targeting endoplasmic reticulum (ER) stress-induced apoptosis has attracted considerable research interest in anti-cancer drug development. Nur77 is a potential therapeutic target in many cancers and several Nur77 modulators have recently been identified as effective anticancer agents by activating ER stress. As an ongoing work, this study reports a new series of novel N1-(2-(adamantan-1-yl)-1H-indol-5-yl)-N2-(substituent)-1,2-dicarboxamides as potent Nur77 modulators that cause ER stress-induced apoptosis. Among this new series, most compounds show improved cytotoxicity against liver cancer (HepG2 and Huh7) and breast cancer (MCF-7 and MDA-MB-231) cell lines. The representative analog 15h dramatically induces Nur77 expression and cell apoptosis, showing excellent growth inhibition of HepG2 and MCF-7 cells (IC50 < 5.0 μM). Mechanistically, 15h binds (KD = 0.477 μM) and activates Nur77-mediated ER stress through the PERK-ATF4 and IRE1 signaling pathways, thereby inducing cell apoptosis. In vivo, 15h treatment strongly suppresses HepG2 xenograft tumor growth (tumor shrink by 54.06 %). In summary, we synthesize a series of novel indole derivatives, among which 15h has significantly improved pharmacological activity against various cancer cells. We further identify 15h as a novel ligand of Nur77, which may serve a therapeutic lead for developing new cancer therapy.
Collapse
Affiliation(s)
- Hongyu Hu
- Xingzhi College, Zhejiang Normal University, Lanxi 321004, China
| | - Fangfang Wen
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China; Community Health Service Center of Dashi Panyu, Guangzhou 511430, China
| | - Tidong Zhen
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Minda Zhang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jingbo Qin
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jiangang Huang
- Xingzhi College, Zhejiang Normal University, Lanxi 321004, China
| | - Zhirong Chen
- The Second People's Hospital of Panyu Guangzhou, Guangzhou 511430, China
| | - Mingyue Yu
- Xingzhi College, Zhejiang Normal University, Lanxi 321004, China; College of Chemsitry and Bioengineering, Yichun 336000, China
| | - Shengwei Hu
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Meijuan Fang
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China.
| | - Jin-Zhang Zeng
- State Key Laboratory of Cellular Stress Biology, Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
11
|
Liu G, Shi Y, Wang J, Gao H, Liu J, Wang H, Wang T, Wei Y. The construction of a breast cancer prognostic model by combining genes related to hypoxia and endoplasmic reticulum stress. Comput Methods Biomech Biomed Engin 2025:1-14. [PMID: 39868728 DOI: 10.1080/10255842.2025.2453941] [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: 06/04/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025]
Abstract
Breast cancer (BC) is a malignant tumor that occurs in breast tissue. This project aims to predict the prognosis of BC patients using genes related to hypoxia and endoplasmic reticulum stress (ERS). RNA-seq and clinical data for BC were downloaded from TCGA and GEO databases. Hypoxia and ERS-related genes were collected from the Genecards database. Univariate/multivariate Cox regression and Lasso regression analyses were used to screen genes and construct prognostic models. Patients were divided into high-risk (HR) and low-risk (LR) groups based on risk scores. The CIBERSORT algorithm was used to analyze differences in immune infiltration between the two groups. The mutations of the two groups were analyzed statistically. The CellMiner database was used for drug prediction and the TISCH database for single-cell sequencing analysis. We screened 8 feature genes to construct a prognostic model. Patients in the HR group had a remarkably worse prognosis. TP53 exhibited a higher mutation frequency in the HR group. CIBERSORT analysis uncovered a remarkable increase in the infiltration levels of Macrophages M0 and Tregs in cancer patients and HR patients. Drug sensitivity prediction demonstrated that the expression of IVL was greatly negatively linked with the sensitivity of COLCHICINE. PTGS2 had a remarkably negative correlation with the Vincristine sensitivity. The prognostic model based on 8 hypoxia and ERS-related genes can predict the survival, immune status, and potential drugs of BC patients, bringing a new perspective on individualized treatment.
Collapse
Affiliation(s)
- Guohua Liu
- Department of the Third General Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| | - Yuan Shi
- Department of the Third General Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| | - Jing Wang
- Department of the Third General Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| | - Haitao Gao
- Department of the Third General Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| | - Jiacai Liu
- Department of the Third General Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| | - Huihua Wang
- Department of the Third General Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| | - Tiantian Wang
- Department of the Third General Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| | - Ya Wei
- Department of the Third General Surgery, Anyang Tumor Hospital, Anyang, Henan, China
| |
Collapse
|
12
|
Aidiel M, Abdul Mutalib M, Ramasamy R, Nik Ramli NN, Tang SGH, Adam SH. Mechanistic Insights into the Anticancer Potential of Methoxyflavones Analogs: A Review. Molecules 2025; 30:346. [PMID: 39860214 PMCID: PMC11768088 DOI: 10.3390/molecules30020346] [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: 10/10/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/27/2025] Open
Abstract
2-phenylchromen-4-one, commonly known as flavone, plays multifaceted roles in biological response that can be abundantly present in natural sources. The methoxy group in naturally occurring flavones promotes cytotoxic activity in various cancer cell lines by targeting protein markers, in facilitating ligand-protein binding mechanisms and activating cascading downstream signaling pathways leading to cell death. However, the lipophilic nature of these analogs is a key concern as it impacts drug membrane transfer. While lipophilicity is crucial for drug efficacy, the excessive lipophilic effects in flavonoids can reduce water solubility and hinder drug transport to target sites. Recent in vitro studies suggest that the incorporation of polar hydroxyl groups which can form hydrogen bonds and stabilize free radicals may help overcome the challenges associated with methoxy groups while maintaining their essential lipophilic properties. Naturally coexisting with methoxyflavones, this review explores the synergistic role of hydroxy and methoxy moieties through hydrogen bonding capacity in maximizing cytotoxicity against cancer cell lines. The physicochemical analysis revealed the potential intramolecular interaction and favorable electron delocalization region between both moieties to improve cytotoxicity levels. Together, the analysis provides a useful strategy for the structure-activity relationship (SAR) of flavonoid analogs in distinct protein markers, suggesting optimal functional group positioning to achieve balanced lipophilicity, effective hydrogen bonding, and simultaneously minimized steric hindrance in targeting specific cancer cell types.
Collapse
Affiliation(s)
- Mohammad Aidiel
- School of Graduate Studies, Management & Science University, University Drive, Off Persiaran Olahraga, Section 13, Shah Alam 40100, Malaysia; (M.A.); (N.N.N.R.)
| | - Maisarah Abdul Mutalib
- School of Graduate Studies, Management & Science University, University Drive, Off Persiaran Olahraga, Section 13, Shah Alam 40100, Malaysia; (M.A.); (N.N.N.R.)
| | - Rajesh Ramasamy
- Department of Pathology, Faculty of Medicine and Health Science, Universiti Putra Malaysia, Serdang 43400, Malaysia;
| | - Nik Nasihah Nik Ramli
- School of Graduate Studies, Management & Science University, University Drive, Off Persiaran Olahraga, Section 13, Shah Alam 40100, Malaysia; (M.A.); (N.N.N.R.)
| | - Shirley Gee Hoon Tang
- Center for Toxicology and Health Risk Studies (CORE), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia;
| | - Siti Hajar Adam
- Preclinical Department, Faculty of Medicine & Defence Health, Universiti Pertahanan Nasional Malaysia, Kuala Lumpur 57000, Malaysia;
| |
Collapse
|
13
|
Dai SY, Xiao Z, Shen F, Lim I, Rao J. Light-Controlled Intracellular Synthesis of Poly(luciferin) Polymers Induces Cell Paraptosis. J Am Chem Soc 2025; 147:2037-2048. [PMID: 39757486 DOI: 10.1021/jacs.4c15644] [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: 01/07/2025]
Abstract
Accumulation of misfolded proteins challenges cellular proteostasis and is implicated in aging and chronic disorders. Cancer cells, moreover, face an elevated level of basal proteotoxic stress; hence, exacerbating endoplasmic reticulum (ER) stress has been shown to induce programmed cell death while enhancing anticancer immunogenicity. We hypothesize that hydrophobic abiotic macromolecules can trigger a similar stress response. Most polymers and nanoparticles, however, are sequestered in endo/lysosomes after endocytosis, which prevents their interaction with the proteostasis machinery. We adopted an in situ polymerization approach to synthesize polymers in cells with cell-permeable monomers. Specifically, we developed a biocompatible polycondensation between l-cysteine and 2-cyanobenzothiazole (CBT) with photochemical control to form insoluble poly(luciferin) aggregates. We identified that in situ polymerization activates the BiP-PERK-CHOP pathway of the unfolded protein response and that the unresolved ER stress initiates a form of regulated cell death consistent with paraptosis. In addition, the dying cells emit damage-associated molecular patterns (DAMPs), indicating an immunogenic cell death that could potentiate antitumor immunity. Our results show that in situ polymerization mimics misfolded protein aggregates to induce proteotoxic stress and cancer cell death, offering a novel therapeutic strategy to exploit cancer vulnerability.
Collapse
Affiliation(s)
- Sheng-Yao Dai
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Zhen Xiao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Fangfang Shen
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Irene Lim
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Jianghong Rao
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford, California 94305, United States
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
14
|
Kim TW, Ko SG. A Novel PPARγ Modulator Falcarindiol Mediates ER Stress-Mediated Apoptosis by Regulating NOX4 and Overcomes Radioresistance in Breast Cancer. Antioxidants (Basel) 2024; 13:1533. [PMID: 39765861 PMCID: PMC11727077 DOI: 10.3390/antiox13121533] [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: 10/28/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 01/15/2025] Open
Abstract
The extract of the rhizome of Cnidium officinale Makino has potential anti-cancer and anti-inflammatory effects in many diseases, such as cancer. However, the biological functions of falcarindiol (FAD) in breast cancer are not fully understood. This study proved the anti-inflammatory and anti-cancer effects of FAD in breast cancer. Breast cancer models confirmed that FAD reduces cell viability and decreases the tumor volume of xenograft mouse models in a dose-dependent manner. FAD mediated caspase-3-dependent apoptosis in MDA-MB-231 and MCF-7 cells, whereas Z-VAD-FMK in combination with FAD inhibited caspase-3-induced apoptosis. FAD mediates apoptosis through cytosolic reactive oxygen species (ROS) and calcium (Ca2+) production and ER stress signaling pathways. In addition, FAD combined with thapsigargin (TG) exerts a synergistic apoptotic cell death effect. In the loss-of-function experiments, PERK or CHOP ablation suppressed intracellular ROS and Ca2+ release and ER stress-induced apoptosis in FAD-treated breast cancer models. Since there is a relationship between ROS and NADPH Oxidase 4 (NOX4), Nox4 ablation blocked ER stress-mediated apoptotic cell death by inhibiting ROS release in FAD-induced breast cancer models. Radioresistant models, such as MCF-7R and MDA-MB-231R, were developed to address the cellular radioresistance in clinical radiotherapy. FAD combined with radiation (2 Gy) overcame radioresistance via the inhibition of the epithelial-mesenchymal transition (EMT) phenomenon, such as the upregulation of PPARγ, VIM, and CDH2 and the downregulation of CDH1. Consequently, these results show that FAD may be a novel treatment as a breast cancer therapy.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Biopharmaceutical Engineering, Dongguk University-WISE, Gyeongju 38066, Republic of Korea
| | - Seong-Gyu Ko
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
15
|
Bhamidipati P, Nagaraju GP, Malla R. Immunoglobulin-binding protein and Toll-like receptors in immune landscape of breast cancer. Life Sci 2024; 358:123196. [PMID: 39481836 DOI: 10.1016/j.lfs.2024.123196] [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: 06/02/2024] [Revised: 07/29/2024] [Accepted: 10/27/2024] [Indexed: 11/03/2024]
Abstract
Breast cancer (BC) is a complex disease exhibiting significant heterogeneity and encompassing various molecular subtypes. Among these, triple-negative breast cancer (TNBC) stands out as one of the most challenging types, characterized by its aggressive nature and poor prognosis. This review embarks on a comprehensive exploration of the immune landscape of BC, with a primary focus on the functional and structural characterization of immunoglobulin-binding protein (BiP) and its pivotal role in regulating the unfolded response (UPR) pathway of proteins. Moreover, we unravel the multifaceted functions of BiP in BC, with a special emphasis on the involvement of cell surface BiP in TNBC metastasis, drug resistance, and its contribution to the formation of the tumor microenvironment (TME). We also provide mechanistic insights into how ER-resident BiP mediates the sensitization of drug-resistant BC to different treatment strategies, thereby offering promising avenues for therapeutic intervention. We also delve into the role of Toll-like receptors (TLRs), shedding light on their diverse expression patterns across BC and their influence on modulating the tumor immune response. Understanding the interplay between BiP, TLRs, and the immune response, especially in TNBC, opens avenues for novel immunotherapies. Future research should focus on developing targeted therapies that activate ER-resident BiP or inhibit cell surface BiP, and modulate TLR signaling. Moreover, exploring BiP as a biomarker for TNBC diagnosis, prognosis, and treatment response will be crucial for personalized medicine.
Collapse
Affiliation(s)
- Priyamvada Bhamidipati
- Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - RamaRao Malla
- Cancer Biology Laboratory, Department of Life Sciences, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh 530045, India.
| |
Collapse
|
16
|
Sun J, Ren H, Wang J, Xiao X, Zhu L, Wang Y, Yang L. CHAC1: a master regulator of oxidative stress and ferroptosis in human diseases and cancers. Front Cell Dev Biol 2024; 12:1458716. [PMID: 39534397 PMCID: PMC11554486 DOI: 10.3389/fcell.2024.1458716] [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: 07/02/2024] [Accepted: 09/10/2024] [Indexed: 11/16/2024] Open
Abstract
CHAC1, an essential regulator of oxidative stress and ferroptosis, is increasingly recognized for its significant roles in these cellular processes and its impact on various human diseases and cancers. This review aims to provide a comprehensive overview of CHAC1's molecular functions, regulatory mechanisms, and effects in different pathological contexts. Specifically, the study objectives are to elucidate the biochemical pathways involving CHAC1, explore its regulatory network, and discuss its implications in disease progression and potential therapeutic strategies. As a γ-glutamyl cyclotransferase, CHAC1 degrades glutathione, affecting calcium signaling and mitochondrial function. Its regulation involves transcription factors like ATF4 and ATF3, which control CHAC1 mRNA expression. CHAC1 is crucial for maintaining redox balance and regulating cell death pathways in cancer. Its elevated levels are associated with poor prognosis in many cancers, indicating its potential as a biomarker and therapeutic target. Additionally, CHAC1 influences non-cancerous diseases such as neurodegenerative and cardiovascular disorders. Therapeutically, targeting CHAC1 could increase cancer cell sensitivity to ferroptosis, aiding in overcoming resistance to standard treatments. This review compiles current knowledge and recent discoveries, emphasizing CHAC1's vital role in human diseases and its potential in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Jiasen Sun
- Department of Gastroenterology, Ankang Central Hospital, Ankang, Shaanxi, China
| | - Hui Ren
- Department of Cardiovascular Disease, Ankang Central Hospital, Ankang, Shaanxi, China
| | - Jiawen Wang
- Department of Cardiovascular Disease, Ankang Central Hospital, Ankang, Shaanxi, China
| | - Xiang Xiao
- Department of Gastroenterology, Ankang Central Hospital, Ankang, Shaanxi, China
| | - Lin Zhu
- Department of Cardiovascular Disease, Ankang Central Hospital, Ankang, Shaanxi, China
| | - Yanyan Wang
- Department of Cardiovascular Disease, Ankang Central Hospital, Ankang, Shaanxi, China
| | - Lili Yang
- Department of Cardiovascular Disease, Ankang Central Hospital, Ankang, Shaanxi, China
| |
Collapse
|
17
|
Xue T, Wang X, Pan X, Liu M, Xu F. PTX promotes breast cancer migration and invasion by recruiting ATF4 to upregulate FGF19. Cell Signal 2024; 122:111309. [PMID: 39053672 DOI: 10.1016/j.cellsig.2024.111309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 07/03/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Widely-spread among women, breast cancer is a malignancy with fatalities, and chemotherapy is a vital treatment option for it. Recent studies have underscored the potential of chemotherapeutic agents such as paclitaxel, adriamycin, cyclophosphamide, and gemcitabine, among others, in facilitating tumor metastasis, with paclitaxel being extensively researched in this context. The molecular mechanism of these genes and their potential relevance to breast cancer is noteworthy. METHOD Clinical tissue specimens were used to analyze the expression and clinical significance of FGF19 or P-FGFR4 in patients with breast cancer before and after chemotherapy. qRT-PCR, ELISA, immunofluorescence and Western blotting were used to detect the expression level of FGF19 in breast cancer cells. The biological impacts of paclitaxel, FGF19, and ATF4 on breast cancer cells were assessed through CCK8, Transwell, and Western blot assays. The expression of ATF4 in breast cancer cells was determined through database analysis, Western blot analysis, qRT-PCR, and immunofluorescence. The direct interaction between FGF19 and ATF4 was confirmed by a luciferase assay, and Western blotting was used to assess the levels of key proteins in the stress response pathway. To confirm the effects of PTX and FGF19 in vivo, we established a lung metastasis model in nude mice. RESULTS FGF19 expression was increased in breast cancer patients after chemotherapy. Paclitaxel can boost the migration and invasion of breast cancer cells, accompanied by an increase in FGF19 expression. ATF4 might be involved in facilitating the enhancing effect of FGF19 on breast cancer cell migration. Finally, stimulation during paclitaxel treatment could trigger a stress response, influencing the expression of FGF19 and the migration of breast cancer cells. CONCLUSION These data suggest that paclitaxel regulates FGF19 expression through ATF4 and thus promotes breast cancer cell migration and invasion.
Collapse
Affiliation(s)
- Ting Xue
- School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| | - Xuezhen Wang
- School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China
| | - Xianjun Pan
- Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Mei Liu
- Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China
| | - Faliang Xu
- School of Medicine, Chongqing University, Chongqing 400044, People's Republic of China; Chongqing University Cancer Hospital, Chongqing 400030, People's Republic of China.
| |
Collapse
|
18
|
Wu M, Zhang L, Pi L, Liu L, Wang S, Wu Y, Pan H, Liu M, Yi Z. IRE1α inhibitor enhances paclitaxel sensitivity of triple-negative breast cancer cells. Cell Oncol (Dordr) 2024; 47:1797-1809. [PMID: 38888849 DOI: 10.1007/s13402-024-00961-7] [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] [Accepted: 05/18/2024] [Indexed: 06/20/2024] Open
Abstract
PURPOSE Breast cancer is the most commonly diagnosed cancer in women, and triple-negative breast cancer (TNBC) accounts for approximately 15%-20% of all breast cancers. TNBC is highly invasive and malignant. Due to the lack of relevant receptor markers, the prognosis of TNBC is poor and the five-year survival rate is low. Paclitaxel is the first-line drug for the treatment of TNBC, which can inhibit cell mitosis. However, many patients develop drug resistance during treatment, leading to chemotherapy failure. Therefore, finding new therapeutic combinations to overcome TNBC drug resistance can provide new strategies for improving the survival rate of TNBC patients. METHODS Cell viability assay, RT-qPCR, Colony formation assay, Western blot, and Xenogeneic transplantation methods were used to investigate roles and mechanisms of IRE1α/XBP1s pathway in the paclitaxel-resistant TNBC cells, and combined paclitaxel and IRE1α inhibitor in the treatment of TNBC was examined in vitro and in vivo. RESULTS We found activation of UPR in paclitaxel-resistant cells, confirming that IRE1α/XBP1 promotes paclitaxel resistance in TNBC. In addition, we demonstrated that the combination of paclitaxel and IRE1α inhibitors can synergistically inhibit the proliferation of TNBC tumors both in vitro and in vivo,suggesting that IRE1α inhibitors combined with paclitaxel may be a new treatment option for TNBC. CONCLUSIONS In this study, we demonstrated the important role of IRE1α signaling in mediating paclitaxel resistance and identified that combination therapies targeting IRE1α signaling could overcome paclitaxel resistance and enhance chemotherapy efficacy.
Collapse
Affiliation(s)
- Min Wu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dong Chuan Rd, Shanghai, 200241, China
| | - Lin Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dong Chuan Rd, Shanghai, 200241, China
| | - Lifu Pi
- Shanghai World Foreign Language Academy, Shanghai, 200030, China
| | - Layang Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dong Chuan Rd, Shanghai, 200241, China
| | - Siyu Wang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dong Chuan Rd, Shanghai, 200241, China
| | - Yujie Wu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dong Chuan Rd, Shanghai, 200241, China
| | - Hongli Pan
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dong Chuan Rd, Shanghai, 200241, China
- Department of Reproductive Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, 272000, China
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dong Chuan Rd, Shanghai, 200241, China
| | - Zhengfang Yi
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, 500 Dong Chuan Rd, Shanghai, 200241, China.
| |
Collapse
|
19
|
Suwannalert P, Panpinyaporn P, Wantanachaisaeng P, Teeppaibul T, Worawichitchaikun T, Koomsang T, Naktubtim C, Payuhakrit W. 17-AAG Induces Endoplasmic Reticulum Stress-mediated Apoptosis in Breast Cancer Cells, Possibly Through PERK/eIF2α Up-regulation. In Vivo 2024; 38:2228-2238. [PMID: 39187325 PMCID: PMC11363749 DOI: 10.21873/invivo.13687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/22/2024] [Accepted: 05/27/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND/AIM Breast cancer is the most predominant type of cancer affecting women worldwide and the current therapeutic treatment for breast cancer patients is not adequately effective. This study aimed to investigate the mechanism of 17-AAG, a heat shock protein (HSP90) inhibitor, as a treatment for inducing breast cancer cell apoptosis. MATERIALS AND METHODS The pharmacology network was employed to examine the correlation of 17-AAG with the gene expression profiles of breast cancer, obtained by Gene Expression Profiling Interactive Analysis (GEPIA). MTT and flow cytometry were utilized to investigate cell proliferation and cell apoptosis, respectively. Dichloro-dihydro-fluorescein diacetate (DCFH-DA) assay and western blot analysis were employed to examine the correlation between cellular oxidant levels and protein expression. Immunofluorescence staining was utilized to confirm the protein localization and assess DNA damage. RESULTS The pharmacological network analysis revealed that HSP90 serves as the common target connecting 17-AAG and breast cancer genes. Treatment with 17-AAG significantly increased cell apoptosis. Moreover, the treatment resulted in up-regulation of cellular oxidant levels and PERK/eIF2α expression. In line with these, protein localization after treatment revealed an increase in DNA damage, correlating with higher ER stress levels. Furthermore, GEPIA demonstrated that PERK and eIF2α expression were significantly higher in breast invasive carcinoma compared to other tumor types. CONCLUSION HSP90 emerges as a potential target for inducing apoptosis in breast cancer cells by disrupting protein homeostasis in the endoplasmic reticulum, possibly through PERK/eIF2α up-regulation. 17-AAG, an HSP90 inhibitor, may therefore potentially hold an alternative therapeutic strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Prasit Suwannalert
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Pathobiology Information and Learning Center, Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | | | - Teerapat Teeppaibul
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Thidarat Koomsang
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Pathobiology Information and Learning Center, Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Chonnapat Naktubtim
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Pathobiology Information and Learning Center, Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Witchuda Payuhakrit
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand;
- Pathobiology Information and Learning Center, Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
20
|
Mu W, Zhi Y, Zhou J, Wang C, Chai K, Fan Z, Lv G. Endoplasmic reticulum stress and quality control in relation to cisplatin resistance in tumor cells. Front Pharmacol 2024; 15:1419468. [PMID: 38948460 PMCID: PMC11211601 DOI: 10.3389/fphar.2024.1419468] [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: 04/18/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024] Open
Abstract
The endoplasmic reticulum (ER) is a crucial organelle that orchestrates key cellular functions like protein folding and lipid biosynthesis. However, it is highly sensitive to disturbances that lead to ER stress. In response, the unfolded protein response (UPR) activates to restore ER homeostasis, primarily through three sensors: IRE1, ATF6, and PERK. ERAD and autophagy are crucial in mitigating ER stress, yet their dysregulation can lead to the accumulation of misfolded proteins. Cisplatin, a commonly used chemotherapy drug, induces ER stress in tumor cells, activating complex signaling pathways. Resistance to cisplatin stems from reduced drug accumulation, activation of DNA repair, and anti-apoptotic mechanisms. Notably, cisplatin-induced ER stress can dualistically affect tumor cells, promoting either survival or apoptosis, depending on the context. ERAD is crucial for degrading misfolded proteins, whereas autophagy can protect cells from apoptosis or enhance ER stress-induced apoptosis. The complex interaction between ER stress, cisplatin resistance, ERAD, and autophagy opens new avenues for cancer treatment. Understanding these processes could lead to innovative strategies that overcome chemoresistance, potentially improving outcomes of cisplatin-based cancer treatments. This comprehensive review provides a multifaceted perspective on the complex mechanisms of ER stress, cisplatin resistance, and their implications in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhongqi Fan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
21
|
Zhang Z, Sun B, Lu J, Bai P, Su Y, Li Y. Norcantharidin inhibits the malignant progression of cervical cancer by inducing endoplasmic reticulum stress. Mol Med Rep 2024; 29:71. [PMID: 38456480 PMCID: PMC10955518 DOI: 10.3892/mmr.2024.13195] [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: 11/26/2023] [Accepted: 01/30/2024] [Indexed: 03/09/2024] Open
Abstract
The antitumor effect of norcantharidin (NCTD) has been widely reported. However, whether NCTD can inhibit cervical cancer remains unknown. In the present study, it was shown that NCTD inhibited the viability of cervical cancer cells and caused cell cycle arrest in a concentration‑dependent manner. Further analysis revealed that the NCTD‑induced reduction in cell viability could be reversed by the inhibitor of apoptosis z‑VAD‑FMK and by the inhibitor of endoplasmic reticulum (ER) stress, 4‑phenylbutyric acid (4‑PBA). Additionally, NCTD led to the accumulation of reactive oxygen species as well as a decrease in the mitochondrial membrane potential in cervical cancer cells, whereas 4‑PBA pre‑treatment attenuated these alterations. In addition, NCTD increased the expression of the apoptosis‑related proteins Bip, activating transcription factor (ATF) 4 and C/EBP homologous protein in a concentration‑dependent manner. Moreover, NCTD significantly increased the expression of the ER stress‑related signaling molecules protein kinase R‑like ER kinase, inositol‑requiring enzyme 1 and ATF6, but 4‑PBA abolished these effects. In vivo experiments showed that NCTD significantly inhibited the growth of subcutaneous tumors in mice. Additionally, the expression of ER stress‑related molecules and apoptosis‑related proteins increased significantly after NCTD treatment. In conclusion, NCTD induces apoptosis by activating ER stress and ultimately curtails the progression of cervical cancer.
Collapse
Affiliation(s)
- Zhongbao Zhang
- Department of Gynecology, Tongliao City Hospital, Tongliao, Inner Mongolia Autonomous Region, P.R. China
| | - Beibei Sun
- Department of Gynecology, Tongliao City Hospital, Tongliao, Inner Mongolia Autonomous Region, P.R. China
| | - Jinqiu Lu
- Department of Gynecology, Tongliao City Hospital, Tongliao, Inner Mongolia Autonomous Region, P.R. China
| | - Penglai Bai
- Department of Gynecology, Tongliao City Hospital, Tongliao, Inner Mongolia Autonomous Region, P.R. China
| | - Yu Su
- Department of Gynecology, Tongliao City Hospital, Tongliao, Inner Mongolia Autonomous Region, P.R. China
| | - Yanchun Li
- Department of Gynecology, Tongliao City Hospital, Tongliao, Inner Mongolia Autonomous Region, P.R. China
| |
Collapse
|
22
|
Kim R, Kin T, Beck WT. Impact of Complex Apoptotic Signaling Pathways on Cancer Cell Sensitivity to Therapy. Cancers (Basel) 2024; 16:984. [PMID: 38473345 DOI: 10.3390/cancers16050984] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Anticancer drugs induce apoptotic and non-apoptotic cell death in various cancer types. The signaling pathways for anticancer drug-induced apoptotic cell death have been shown to differ between drug-sensitive and drug-resistant cells. In atypical multidrug-resistant leukemia cells, the c-Jun/activator protein 1 (AP-1)/p53 signaling pathway leading to apoptotic death is altered. Cancer cells treated with anticancer drugs undergo c-Jun/AP-1-mediated apoptotic death and are involved in c-Jun N-terminal kinase activation and growth arrest- and DNA damage-inducible gene 153 (Gadd153)/CCAAT/enhancer-binding protein homologous protein pathway induction, regardless of the p53 genotype. Gadd153 induction is associated with mitochondrial membrane permeabilization after anticancer drug treatment and involves a coupled endoplasmic reticulum stress response. The induction of apoptosis by anticancer drugs is mediated by the intrinsic pathway (cytochrome c, Cyt c) and subsequent activation of the caspase cascade via proapoptotic genes (e.g., Bax and Bcl-xS) and their interactions. Anticancer drug-induced apoptosis involves caspase-dependent and caspase-independent pathways and occurs via intrinsic and extrinsic pathways. The targeting of antiapoptotic genes such as Bcl-2 enhances anticancer drug efficacy. The modulation of apoptotic signaling by Bcl-xS transduction increases the sensitivity of multidrug resistance-related protein-overexpressing epidermoid carcinoma cells to anticancer drugs. The significance of autophagy in cancer therapy remains to be elucidated. In this review, we summarize current knowledge of cancer cell death-related signaling pathways and their alterations during anticancer drug treatment and discuss potential strategies to enhance treatment efficacy.
Collapse
Affiliation(s)
- Ryungsa Kim
- Department of Breast Surgery, Hiroshima Mark Clinic, 1-4-3F, 2-Chome Ohte-machi, Naka-ku, Hiroshima 730-0051, Japan
| | - Takanori Kin
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, Suita, Osaka 565-0871, Japan
| | - William T Beck
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
23
|
Lim CH, Fang XQ, Kang H, Oh T, Lee S, Kim YS, Lim JH. ER Stress-Activated HSF1 Governs Cancer Cell Resistance to USP7 Inhibitor-Based Chemotherapy through the PERK Pathway. Int J Mol Sci 2024; 25:2768. [PMID: 38474017 DOI: 10.3390/ijms25052768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 02/20/2024] [Accepted: 02/25/2024] [Indexed: 03/14/2024] Open
Abstract
Ubiquitin-specific protease 7 inhibitors (USP7i) are considered a novel class of anticancer drugs. Cancer cells occasionally become insensitive to anticancer drugs, known as chemoresistance, by acquiring multidrug resistance, resulting in poor clinical outcomes in patients with cancer. However, the chemoresistance of cancer cells to USP7i (P22077 and P5091) and mechanisms to overcome it have not yet been investigated. In the present study, we generated human cancer cells with acquired resistance to USP7i-induced cell death. Gene expression profiling showed that heat stress response (HSR)- and unfolded protein response (UPR)-related genes were largely upregulated in USP7i-resistant cancer cells. Biochemical studies showed that USP7i induced the phosphorylation and activation of heat shock transcription factor 1 (HSF1), mediated by the endoplasmic reticulum (ER) stress protein kinase R-like ER kinase (PERK) signaling pathway. Inhibition of HSF1 and PERK significantly sensitized cancer cells to USP7i-induced cytotoxicity. Our study demonstrated that the ER stress-PERK axis is responsible for chemoresistance to USP7i, and inhibiting PERK is a potential strategy for improving the anticancer efficacy of USP7i.
Collapse
Affiliation(s)
- Chang-Hoon Lim
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
- BK21 Program, Department of Applied Life Science, Graduate School, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
| | - Xue-Quan Fang
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
- BK21 Program, Department of Applied Life Science, Graduate School, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
| | - Hyeji Kang
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
| | - Taerim Oh
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
| | - Seonghoon Lee
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
- BK21 Program, Department of Applied Life Science, Graduate School, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
| | - Young-Seon Kim
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
| | - Ji-Hong Lim
- Department of Medicinal Biosciences, College of Biomedical & Health Science, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
- BK21 Program, Department of Applied Life Science, Graduate School, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
- Center for Metabolic Diseases, Konkuk University, 268, Chungwon-daero, Chungju 27478, Chungbuk, Republic of Korea
| |
Collapse
|
24
|
Yang B, Wang S, Yang Y, Li X, Yu F, Wang T. Endoplasmic reticulum stress in breast cancer: a predictive model for prognosis and therapy selection. Front Immunol 2024; 15:1332942. [PMID: 38440732 PMCID: PMC10910050 DOI: 10.3389/fimmu.2024.1332942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/05/2024] [Indexed: 03/06/2024] Open
Abstract
Background Breast cancer (BC) is a leading cause of mortality among women, underscoring the urgent need for improved therapeutic predictio. Developing a precise prognostic model is crucial. The role of Endoplasmic Reticulum Stress (ERS) in cancer suggests its potential as a critical factor in BC development and progression, highlighting the importance of precise prognostic models for tailored treatment strategies. Methods Through comprehensive analysis of ERS-related gene expression in BC, utilizing both single-cell and bulk sequencing data from varied BC subtypes, we identified eight key ERS-related genes. LASSO regression and machine learning techniques were employed to construct a prognostic model, validated across multiple datasets and compared with existing models for its predictive accuracy. Results The developed ERS-model categorizes BC patients into distinct risk groups with significant differences in clinical prognosis, confirmed by robust ROC, DCA, and KM analyses. The model forecasts survival rates with high precision, revealing distinct immune infiltration patterns and treatment responsiveness between risk groups. Notably, we discovered six druggable targets and validated Methotrexate and Gemcitabine as effective agents for high-risk BC treatment, based on their sensitivity profiles and potential for addressing the lack of active targets in BC. Conclusion Our study advances BC research by establishing a significant link between ERS and BC prognosis at both the molecular and cellular levels. By stratifying patients into risk-defined groups, we unveil disparities in immune cell infiltration and drug response, guiding personalized treatment. The identification of potential drug targets and therapeutic agents opens new avenues for targeted interventions, promising to enhance outcomes for high-risk BC patients and paving the way for personalized cancer therapy.
Collapse
Affiliation(s)
- Bin Yang
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- NHC Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, Guizhou, China
| | - Shu Wang
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yanfang Yang
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- NHC Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, Guizhou, China
| | - Xukui Li
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- NHC Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, Guizhou, China
| | - Fuxun Yu
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- NHC Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, Guizhou, China
| | - Tao Wang
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
- NHC Key Laboratory of Pulmonary Immune-Related Diseases, Guizhou Provincial People's Hospital, Guizhou University, Guiyang, Guizhou, China
| |
Collapse
|
25
|
Yang T, Li W, Zhou J, Xu M, Huang Z, Ming J, Huang T. A novel bystander effect in tamoxifen treatment: PPIB derived from ER+ cells attenuates ER- cells via endoplasmic reticulum stress-induced apoptosis. Cell Death Dis 2024; 15:147. [PMID: 38360722 PMCID: PMC10869711 DOI: 10.1038/s41419-024-06539-3] [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: 08/22/2023] [Revised: 02/01/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024]
Abstract
Tamoxifen (TAM) is the frontline therapy for estrogen receptor-positive (ER+) breast cancer in premenopausal women that interrupts ER signaling. As tumors with elevated heterogeneity, amounts of ER-negative (ER-) cells are present in ER+ breast cancer that cannot be directly killed by TAM. Despite complete remissions have been achieved in clinical practice, the mechanism underlying the elimination of ER- cells during TAM treatment remains an open issue. Herein, we deciphered the elimination of ER- cells in TAM treatment from the perspective of the bystander effect. Markable reductions were observed in tumorigenesis of ER- breast cancer cells by applying both supernatants from TAM-treated ER+ cells and a transwell co-culture system, validating the presence of a TAM-induced bystander effect. The major antitumor protein derived from ER+ cells, peptidyl-prolyl cis-trans isomerase B (PPIB), is the mediator of the TAM-induced bystander effect identified by quantitative proteomics. The attenuation of ER- cells was attributed to activated BiP/eIF2α/CHOP axis and promoted endoplasmic reticulum stress (ERS)-induced apoptosis, which can also be triggered by PPIB independently. Altogether, our study revealed a novel TAM-induced bystander effect in TAM treatment of ER+ breast cancer, raising the possibility of developing PPIB as a synergistic antitumor agent or even substitute endocrine therapy.
Collapse
Affiliation(s)
- Tinglin Yang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenhui Li
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jun Zhou
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ming Xu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ziwei Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jie Ming
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
26
|
He J, Zhou Y, Sun L. Emerging mechanisms of the unfolded protein response in therapeutic resistance: from chemotherapy to Immunotherapy. Cell Commun Signal 2024; 22:89. [PMID: 38297380 PMCID: PMC10832166 DOI: 10.1186/s12964-023-01438-0] [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: 10/23/2023] [Accepted: 12/12/2023] [Indexed: 02/02/2024] Open
Abstract
The accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER) causes ER stress and activates the unfolded protein response (UPR). As an adaptive cellular response to hostile microenvironments, such as hypoxia, nutrient deprivation, oxidative stress, and chemotherapeutic drugs, the UPR is activated in diverse cancer types and functions as a dynamic tumour promoter in cancer development; this role of the UPR indicates that regulation of the UPR can be utilized as a target for tumour treatment. T-cell exhaustion mainly refers to effector T cells losing their effector functions and expressing inhibitory receptors, leading to tumour immune evasion and the loss of tumour control. Emerging evidence suggests that the UPR plays a crucial role in T-cell exhaustion, immune evasion, and resistance to immunotherapy. In this review, we summarize the molecular basis of UPR activation, the effect of the UPR on immune evasion, the emerging mechanisms of the UPR in chemotherapy and immunotherapy resistance, and agents that target the UPR for tumour therapeutics. An understanding of the role of the UPR in immune evasion and therapeutic resistance will be helpful to identify new therapeutic modalities for cancer treatment. Video Abstract.
Collapse
Affiliation(s)
- Jiang He
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Huan, China.
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, 410008, China.
- Center for Molecular Imaging of Central, South University, Xiangya Hospital, Changsha, 410008, China.
| | - You Zhou
- Department of Pathology, Tongji Medical College Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lunquan Sun
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, 410008, Huan, China.
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, 410008, China.
- Center for Molecular Imaging of Central, South University, Xiangya Hospital, Changsha, 410008, China.
| |
Collapse
|
27
|
Ghosh MK, Roy S. Chromosomal instability (CIN) triggers immune evasion and metastatic potential in cancer through rewired STING signalling. MOLECULAR BIOMEDICINE 2024; 5:4. [PMID: 38253764 PMCID: PMC10803705 DOI: 10.1186/s43556-023-00166-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Affiliation(s)
- Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India.
| | - Srija Roy
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata-700091 & 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700032, India
| |
Collapse
|
28
|
Joshi P, Verma K, Kumar Semwal D, Dwivedi J, Sharma S. Mechanism insights of curcumin and its analogues in cancer: An update. Phytother Res 2023; 37:5435-5463. [PMID: 37649266 DOI: 10.1002/ptr.7983] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 07/05/2023] [Accepted: 07/30/2023] [Indexed: 09/01/2023]
Abstract
Cancer is the world's second leading cause of mortality and one of the major public health problems. Cancer incidence and mortality rates remain high despite the great advancements in existing therapeutic, diagnostic, and preventive approaches. Therefore, a quest for less toxic and more efficient anti-cancer strategies is still at the forefront of the current research. Traditionally important, curcumin commonly known as a wonder molecule has received considerable attention as an anti-cancer, anti-inflammatory, and antioxidant candidate. However, limited water solubility and low bioavailability restrict its extensive utility in different pathological states. The investigators are making consistent efforts to develop newer strategies to overcome its limitations by designing different analogues with better pharmacokinetic and pharmacodynamic properties. The present review highlights the recent updates on curcumin and its analogues with special emphasis on various mechanistic pathways involved in anti-cancer activity. In addition, the structure-activity relationship of curcumin analogues has also been precisely discussed. This article will also provide key information for the design and development of newer curcumin analogues with desired pharmacokinetic and pharmacodynamic profiles and will provide in depth understanding of molecular pathways involved in the anti-cancer activities.
Collapse
Affiliation(s)
- Priyanka Joshi
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Kanika Verma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Deepak Kumar Semwal
- Faculty of Biomedical Sciences, Uttarakhand Ayurved University, Dehradun, Uttarakhand, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali, Rajasthan, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan, India
| |
Collapse
|
29
|
Li Y, Zhang Q, Yang J, He W, Jiang Y, Chen Y, Wang Y. Metformin combined with glucose starvation synergistically suppress triple-negative breast cancer by enhanced unfolded protein response. Biochem Biophys Res Commun 2023; 675:146-154. [PMID: 37473529 DOI: 10.1016/j.bbrc.2023.07.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/13/2023] [Indexed: 07/22/2023]
Abstract
Metformin (MET) is a well-documented drug used in the treatment of type II diabetes. Recent studies have revealed its potential anti-tumor effects in various types of cancer. However, the dosage of MET required to exhibit anti-tumor activity is considerably higher than the clinically recommended dosage. In this study, we investigated the synergistical anti-tumor effect of glucose deprivation and MET in MDA-MB-231 cells, which represents a triple-negative breast cancer subtype (TNBC). Our findings demonstrate that glucose deprivation significantly enhances the anti-tumor activity of MET by reducing cell proliferation and increasing cell apoptosis. RNA-seq was performed to identify the key molecules involved in this process. Our results indicate that unfolded protein response of endoplasmic reticulum (UPRER) was significantly activated upon glucose starvation combining with MET compared to glucose starvation alone. Notably, the combined treatment significantly activated UPRER signaling pathway through ATF4/ATF3/CHOP axis, due to enhanced UPRER stress. In conclusion, our study suggests that the synergistic effects of MET and glucose deprivation suppress cell proliferation in TNBC by activating pro-apoptotic molecules through UPRER stress. These findings have potential implications for the anti-tumor application of MET in TNBC.
Collapse
Affiliation(s)
- Ying Li
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, PR China; Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, PR China
| | - Qingqian Zhang
- Department of Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, PR China
| | - Jintao Yang
- Key Laboratory of Digital Technology in Medical Diagnostics of Zhejiang Province, Dian Diagnostics Group Co., Ltd., Hangzhou, Zhejiang Province, Hangzhou, PR China
| | - Weiping He
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou, PR China
| | - Yulan Jiang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, PR China
| | - Yu Chen
- School of Pharmacy, Hangzhou Normal University, Hangzhou, PR China
| | - Yifan Wang
- Cancer Institute of Integrative Medicine, Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, PR China; Key Laboratory of Cancer Prevention and Therapy Combining Traditional Chinese and Western Medicine of Zhejiang Province, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, PR China.
| |
Collapse
|
30
|
Sun X, Yuan Z, Zhang L, Ren M, Yang J, Xu Y, Hao J. Comprehensive Analysis of SLC35A2 in Pan-Cancer and Validation of Its Role in Breast Cancer. J Inflamm Res 2023; 16:3381-3398. [PMID: 37593196 PMCID: PMC10427759 DOI: 10.2147/jir.s419994] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023] Open
Abstract
Purpose Elucidation of the oncogenic role of SLC35A2 in human tumors and the potential function and clinical significance in breast cancer. Methods Pan-cancer analysis was performed via various bioinformatics tools to explain the pathogenic role of SLC35A2. A prognostic nomogram was also developed based on the SLC35A2 expression and clinicopathological characteristics in breast cancer patients. In addition, the role of SLC35A2 was validated in breast cancer by in vivo and in vitro experiments. Results SLC35A2 expression is increased in 27 tumor types, and its high expression is substantially correlated with poor prognosis in patients with a variety of cancers. Receiver operating characteristic (ROC) curves showed that SLC35A2 expression levels could accurately distinguish most tumor tissues from normal tissues. High SLC35A2 expression was linked to increased immune infiltration in myeloid-derived suppressor cells (MDSC), as well as immune checkpoints, ferroptosis-related genes, tumor mutational burden (TMB), and microsatellite instability (MSI). SLC35A2 may be involved in tumorigenesis by regulating the glycosylation process. Furthermore, multivariate Cox analysis showed that SLC35A2 was an independent prognostic factor for breast cancer. And the nomogram model had good predictive accuracy for the prognosis of breast cancer patients. Meanwhile, cellular experiments demonstrated that knockdown of SLC35A2 could significantly inhibit the proliferation, migration and invasion of breast cancer cells, while increasing the protein level of E-cadherin and decreasing N-cadherin. A nude mouse xenograft model showed that inhibition of SLA35A2 expression could significantly inhibit tumor growth. Conclusion SLC35A2 has good diagnostic and prognostic values in multiple cancers and is closely related to tumor immune infiltration. In addition, SLA35A2 as an oncogene in breast cancer may be involved in the progression of epithelial mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Xiaonan Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Zhichao Yuan
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Lu Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Min Ren
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Jing Yang
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Yidan Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| | - Jiqing Hao
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, People’s Republic of China
| |
Collapse
|
31
|
Bian X, Fan N, Li M, Han D, Li J, Fan L, Li X, Kong L, Tang H, Ding S, Song F, Li S, Cheng W. An ER-Horse Detonating Stress Cascade for Hepatocellular Carcinoma Nanotherapy. ACS NANO 2023; 17:4896-4912. [PMID: 36811530 DOI: 10.1021/acsnano.2c11922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Persisting and excessive endoplasmic reticulum stress (ERS) can evoke rapid cell apoptosis. Therapeutic interference of ERS signaling holds enormous potential for cancer nanotherapy. Herein, a hepatocellular carcinoma (HCC) cell-derived ER vesicle (ERV) encapsulating siGRP94, denoted as ER-horse, has been developed for precise HCC nanotherapy. Briefly, ER-horse, like the Trojan horse, was recognized via homotypic camouflage, imitated the physiological function of ER, and exogenously opened the Ca2+ channel. Consequently, the mandatory pouring-in of extracellular Ca2+ triggered the aggravated stress cascade (ERS and oxidative stress) and apoptosis pathway with the inhibition of unfolded protein response by siGRP94. Collectively, our findings provide a paradigm for potent HCC nanotherapy via ERS signaling interference and exploring therapeutic interference of physiological signal transduction pathways for precision cancer therapy.
Collapse
Affiliation(s)
- Xintong Bian
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Ningke Fan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Meng Li
- The Second Affliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Daobin Han
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jia Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lu Fan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xinyu Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Liangsheng Kong
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Hua Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Shijia Ding
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), College of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Fangzhou Song
- Molecular Medicine and Cancer Research Center, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Siqiao Li
- Department of Forensic Medicine, Faculty of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Wei Cheng
- The Center for Clinical Molecular Medical Detection, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| |
Collapse
|
32
|
Genovese I, Fornetti E, Ruocco G. Mitochondria inter-organelle relationships in cancer protein aggregation. Front Cell Dev Biol 2022; 10:1062993. [PMID: 36601538 PMCID: PMC9806238 DOI: 10.3389/fcell.2022.1062993] [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: 10/06/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Mitochondria are physically associated with other organelles, such as ER and lysosomes, forming a complex network that is crucial for cell homeostasis regulation. Inter-organelle relationships are finely regulated by both tether systems, which maintain physical proximity, and by signaling cues that induce the exchange of molecular information to regulate metabolism, Ca2+ homeostasis, redox state, nutrient availability, and proteostasis. The coordinated action of the organelles is engaged in the cellular integrated stress response. In any case, pathological conditions alter functional communication and efficient rescue pathway activation, leading to cell distress exacerbation and eventually cell death. Among these detrimental signals, misfolded protein accumulation and aggregation cause major damage to the cells, since defects in protein clearance systems worsen cell toxicity. A cause for protein aggregation is often a defective mitochondrial redox balance, and the ER freshly translated misfolded proteins and/or a deficient lysosome-mediated clearance system. All these features aggravate mitochondrial damage and enhance proteotoxic stress. This review aims to gather the current knowledge about the complex liaison between mitochondria, ER, and lysosomes in facing proteotoxic stress and protein aggregation, highlighting both causes and consequences. Particularly, specific focus will be pointed to cancer, a pathology in which inter-organelle relations in protein aggregation have been poorly investigated.
Collapse
Affiliation(s)
- Ilaria Genovese
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia (IIT), Rome, Italy,*Correspondence: Ilaria Genovese,
| | - Ersilia Fornetti
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Giancarlo Ruocco
- Center for Life Nano and Neuro Science, Istituto Italiano di Tecnologia (IIT), Rome, Italy,Department of Physics, Sapienza University of Rome, Rome, Italy
| |
Collapse
|