1
|
Choi JC. Perinuclear organelle trauma at the nexus of cardiomyopathy pathogenesis arising from loss of function LMNA mutation. Nucleus 2025; 16:2449500. [PMID: 39789731 PMCID: PMC11730615 DOI: 10.1080/19491034.2024.2449500] [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: 10/31/2024] [Revised: 12/22/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Over the past 25 years, nuclear envelope (NE) perturbations have been reported in various experimental models with mutations in the LMNA gene. Although the hypothesis that NE perturbations from LMNA mutations are a fundamental feature of striated muscle damage has garnered wide acceptance, the molecular sequalae provoked by the NE damage and how they underlie disease pathogenesis such as cardiomyopathy (LMNA cardiomyopathy) remain poorly understood. We recently shed light on one such consequence, by employing a cardiomyocyte-specific Lmna deletion in vivo in the adult heart. We observed extensive NE perturbations prior to cardiac function deterioration with collateral damage in the perinuclear space. The Golgi is particularly affected, leading to cytoprotective stress responses that are likely disrupted by the progressive deterioration of the Golgi itself. In this review, we discuss the etiology of LMNA cardiomyopathy with perinuclear 'organelle trauma' as the nexus between NE damage and disease pathogenesis.
Collapse
Affiliation(s)
- Jason C. Choi
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
2
|
Armin F, Panahpour H, Salimnejad R, Saadati H, Abedi A, Aslani MR. Altered lung inflammation and expression of endoplasmic reticulum stress markers in male mice aged-asthma model. Tissue Cell 2025; 95:102938. [PMID: 40319712 DOI: 10.1016/j.tice.2025.102938] [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: 11/15/2024] [Revised: 03/27/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Although there is a strong correlation between aging and asthma pathophysiology, the underlying mechanism has not been fully elucidated. The endoplasmic reticulum (ER) has been demonstrated to be a crucial intracellular organelle involved in the pathogenesis of numerous diseases. The aim of the current study was to investigate ER stress markers in the lung tissue of aged ovalbumin (OVA)-sensitized mice. Male BALB/C mice were randomly divided into the following four groups (10 in each): 1) control, 2) OVA-sensitized (OVA), 3) D-galactose-induced aging (D-gal), and 4) OVA-sensitized associated with D-galactose-induced aging (OVA+D-gal). Total WBCs and differential cells were counted using the bronchoalveolar lavage (BAL) fluid. Lung tissue was analyzed for ER stress markers (ATF4, ATF6, GRP78, CHOP, and XBP1) through Real Time-PCR technique as well as histopathological assessment. The data indicated a significant increase in both total WBCs and differential cells within the OVA, D-gal, and OVA+D-gal groups when compared to the control group, particularly evident in the OVA+D-gal group. Also, the results showed that the increased expression of ER stress markers (ATF4, ATF6, GRP78, CHOP, and XBP1) was significantly higher in the OVA, D-gal, and OVA+D-gal groups than in the control group. Interestingly, the increased expression of CHOP, ATF4, and XBP1 was observed in the OVA+D-gal group more than in other groups. In aged OVA-sensitized mice, the findings revealed a maladaptive ER stress response in their lung tissue, characterized by elevated levels of CHOP, ATF4, and XBP1 expression.
Collapse
Affiliation(s)
- Farshad Armin
- Department of Physiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Hamdollah Panahpour
- Department of Physiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ramin Salimnejad
- Department of Anatomical Sciences, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Hakimeh Saadati
- Department of Physiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Ali Abedi
- Department of Physiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mohammad Reza Aslani
- Department of Physiology, Faculty of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran; Lung Diseases Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
3
|
Kusakari S, Suzuki H, Nawa M, Sudo K, Yamazaki R, Miyagi T, Ohara T, Matsuoka M, Kanekura K. Excessive expression of progranulin leads to neurotoxicity rather than neuroprotection. Neurobiol Dis 2025; 209:106895. [PMID: 40180225 DOI: 10.1016/j.nbd.2025.106895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025] Open
Abstract
Frontotemporal dementia (FTD) is an early onset form of dementia characterized by frontotemporal lobar atrophy accompanied by behavioral, personality, language, and motor deficits. Heterozygous mutations in GRN gene encoding progranulin (PGRN) are the genetic causes of FTD. Since PGRN is a neurotrophic and anti-inflammatory factor, most FTD-related PGRN mutations are thought to cause FTD due to haploinsufficiency. Therefore, therapies that increase PGRN levels by the administration of recombinant PGRN or viral vectors are attracting attention as an approach to the treatment of FTD. However, the mechanisms underlying the neuroprotective effects of PGRN remain unclear. To investigate the neuroprotective mechanisms of PGRN in vivo, we generated human PGRN transgenic (Tg) mice using the CAG promoter. Unexpectedly, mice overexpressing wild-type human PGRN showed a shortened lifespan and cerebellar dysfunction, including the loss of Purkinje cells. Furthermore, PGRN Tg mice developed cognitive impairment, gliosis, and lysosomal abnormalities. FTD-causative R432C-PGRN mutant Tg mice also showed FTD-like phenotypes, such as neuronal loss, gliosis, and behavioral deficits. In cultured cells, overexpression of PGRN induced endoplasmic reticulum (ER) stress and apoptotic cell death, suggesting that continuous increases in PGRN expression through viral vectors or genetic manipulation are neurotoxic and that PGRN-replacement therapy may be required to maintain optimal PGRN levels for each neuron type and brain region.
Collapse
Affiliation(s)
- Shinya Kusakari
- Department of Pharmacology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan.
| | - Hiroaki Suzuki
- Department of Pharmacology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Mikiro Nawa
- Department of Pharmacology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Katsuko Sudo
- Pre-Clinical Research Center, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Rio Yamazaki
- Department of Pharmacology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Tamami Miyagi
- Department of Pharmacology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Tomoko Ohara
- Department of Pharmacology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Masaaki Matsuoka
- Department of Pharmacology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Kohsuke Kanekura
- Department of Pharmacology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan.
| |
Collapse
|
4
|
Kwon IJ, Lee EJ, Park JH, Kim JY, Park S, Bae YJ, Hwang S, Na HW, Cha N, Jang G, Kim HJ, Lee HK, Oh SH. Independent and Combined Effects of Particulate Matter and Sleep Deprivation on Human Skin Barrier. Ann Dermatol 2025; 37:131-139. [PMID: 40432361 PMCID: PMC12117546 DOI: 10.5021/ad.25.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND The exposome encompasses all factors people encounter through life, with the skin constantly exposed. While particulate matter (PM) and sleep deprivation are known to contribute to barrier dysfunction, their combined effects remain unclear. OBJECTIVE To evaluate the independent and combined effects of PM exposure and short-term sleep deprivation on skin barrier function. METHODS Forty healthy Korean women (aged 24-58 years) were enrolled in this study. Forearms were divided into 4 sites: control, PM exposure, sleep deprivation, and PM plus sleep deprivation. Parameters such as trans-epidermal water loss (TEWL), hydration, elasticity, roughness, and redness were measured at baseline and post-exposure. RNA sequencing and reverse transcription-polymerase chain reaction were conducted on tape-stripped skin samples. RESULTS PM exposure significantly increased TEWL (+25.59%, p<0.01), roughness (+21.9%, p<0.01), and redness (+13.7%, p<0.0001) while reducing elasticity (-3.98%, p<0.01). Sleep deprivation modestly reduced elasticity (-1.39%, p<0.05) without affecting other parameters. Combined PM and sleep deprivation did not further exacerbate barrier dysfunction compared to PM alone. RNA sequencing revealed reduced FLG and LORICRIN expression and upregulated endoplasmic reticulum (ER) stress markers (HSP90B1, CANX) in both PM and sleep deprivation conditions. CONCLUSION PM exposure impaired skin barrier function, while short-term sleep deprivation alone did not significantly affect the barrier, either independently or in combination with PM. However, it was observed that the sleep deprivation-only, while not directly causing barrier damage, induced changes in ER stress-related gene expression in tape-stripped skin samples, like the PM exposure-only. This suggests that such signaling pathways could potentially exacerbate skin barrier deterioration.
Collapse
Affiliation(s)
- Il Joo Kwon
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Eun Jung Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | | | - Ji Young Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Seohyun Park
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Yu Jeong Bae
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Shinwon Hwang
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Hye-Won Na
- Research and Innovation Center, Amorepacific, Yongin, Korea
| | - Nari Cha
- Research and Innovation Center, Amorepacific, Yongin, Korea
| | - Geunhyuk Jang
- Research and Innovation Center, Amorepacific, Yongin, Korea
| | | | | | - Sang Ho Oh
- Department of Dermatology and Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
5
|
Xu W, Li X, He F, Zhao H, Wu J, Li M, Dai X, Li Y, Hu X, Li X, Cen J, Guo P, Duan S. Folate receptor-targeted pH-sensitive liposomes loaded with TGX-221 against prostate cancer by inhibiting PI3K/110β signaling. NANOSCALE ADVANCES 2025; 7:3267-3280. [PMID: 40212450 PMCID: PMC11979785 DOI: 10.1039/d5na00009b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/05/2025] [Indexed: 05/29/2025]
Abstract
Prostate cancer (PCa) is the most common cancer in men and the leading cause of cancer death worldwide. Overactivation of PI3K signaling has been reported to be associated with PCa. TGX221 is an effective specific inhibitor of PI3K, but its clinical application is greatly limited due to its poor solubility. Herein, by using folic acid-PEG-cholesterol semi-succinate (FA-PEG-CHEMS) as the targeting component, we developed a folate receptor-targeted pH-sensitive liposomal delivery system loaded with TGX221 (FA-Lip-TGX221) that could realize effective delivery and controlled release of drugs in the tumor. The prepared liposomes exhibited a uniform particle size and high stability. In addition, FA-Lip-TGX221 could be effectively internalized by PC-3 cells due to its ability to target folate receptors, thereby accumulating in tumor tissues. Meanwhile, in vitro and in vivo experiments suggested that FA-Lip-TGX221 could activate the PERK-ATF4-CHOP signaling pathway by inhibiting PI3K/110β signaling in PCa, thus significantly promoting endoplasmic reticulum (ER) stress-mediated cancer cell death. In conclusion, FA-Lip-TGX221 is a promising nano-delivery vehicle for the treatment of PCa, and also provide valuable references for all tumors overexpressing folate receptors.
Collapse
Affiliation(s)
- Weibo Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
- Medical School, Henan University Kaifeng 475004 China
| | - Xiaohan Li
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Fujin He
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Han Zhao
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Jing Wu
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Mengyu Li
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Xiaoying Dai
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Yanmin Li
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Xiaojiao Hu
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
| | - Xiaodong Li
- Medical School, Henan University Kaifeng 475004 China
| | - Juan Cen
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
- The First Affiliated Hospital of Henan University Kaifeng Henan 475004 PR China
| | - Peng Guo
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi 710061 China
| | - Shaofeng Duan
- Key Laboratory of Natural Medicine and Immune Engineering, School of Pharmacy, Henan University Kaifeng Henan 475004 PR China
- The First Affiliated Hospital of Henan University Kaifeng Henan 475004 PR China
| |
Collapse
|
6
|
Zhang J, Wang W, Zhang X, Wang F, Geng S, Wang X, Wang T. Using endoplasmic reticulum engineering to improve recombinant protein production in CHO cells. Int J Biol Macromol 2025; 315:144695. [PMID: 40424905 DOI: 10.1016/j.ijbiomac.2025.144695] [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: 12/08/2024] [Revised: 05/22/2025] [Accepted: 05/25/2025] [Indexed: 05/29/2025]
Abstract
Chinese hamster ovary (CHO) cells are commonly used to produce recombinant therapeutic proteins (RTPs). While recent strategies have significantly improved the expression levels of RTPs in CHO cells, insufficient secretion and endoplasmic reticulum (ER) stress remain major bottlenecks. Therefore, further understanding of the mechanism of the ER stress response, optimization of ER-related folding and degradation pathways, and development of more efficient ER engineering tools are expected to overcome this issue and maximize RTP production. In this review, we summarize the role of ER in recombinant proteins production and explore ER engineering strategies to improve the yield of recombinant proteins in CHO cells. We further discuss ER-related strategies that can improve recombinant protein production, future research directions, and prospective applications.
Collapse
Affiliation(s)
- Junhe Zhang
- Institutes of Health Central Plains, Xinxiang Key Laboratory for Tumor Drug Screening and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, China; International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China.
| | - Weifeng Wang
- Institutes of Health Central Plains, Xinxiang Key Laboratory for Tumor Drug Screening and Targeted Therapy, Xinxiang Medical University, Xinxiang 453003, China; International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Xi Zhang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Fang Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Shaolei Geng
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Xiaoyin Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China
| | - Tianyun Wang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, Xinxiang 453003, China.
| |
Collapse
|
7
|
Liu Y, Li S, Wang K, Wang Y, Wang Y, Zhang C, Wu H, Wang G, Qin F, Song Z, Tao Y. Unveiling the HSP90 inhibitor mediated effects on endoplasmic reticulum stress and redox signaling:from a cancer inhibitor to retinal degeneration catalyst. Free Radic Biol Med 2025:S0891-5849(25)00697-5. [PMID: 40414464 DOI: 10.1016/j.freeradbiomed.2025.05.414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Revised: 05/20/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Retinal degeneration (RD) is a class of polygenic blind eye disease characterized by photoreceptors loss and dysfunction of retinal pigment epithelium. Thus far, there is no effective treatment to save the declining vision in RD patients. Animal models are highly precious tools for studying the pathological mechanisms of RD, and for screening potential therapeutics. AUY922 is a heat shock protein 90 inhibitor that exhibits potent anti-cancer effects. However, it causes adverse ocular reactions such as reduced visual acuity and night blindness. This study intends to explore the pathological mechanism underlying the AUY922 induced RD. In vitro study, AUY922 induced cytotoxic effects on the 661W cells, which are ascribed to endoplasmic reticulum (ER) stress and oxidative damages. ER stress inhibitor 4-PBA alleviated 661W cells apoptosis and oxidative stress. Subsequently, AUY922 was delivered into the vitreous cavity of mouse and induced selective photoreceptor death and visual impairments. Overactivation of neuroglial and retinal remodeling occurred during the degenerative process. Moreover, enhanced CHOP expression was tied to profound disturbances in redox homeostasis, which readied photoreceptors for apoptosis. The underlying mechanism should be attributed to the activation of the PERK-eIF2α-ATF4-CHOP pathway. AUY922 can compensate for the high toxicity and instability of traditional inducers in RD modeling. These results not only enrich our understanding of the toxicology of AUY922 but also provide clues for establishing reliable RD models.
Collapse
Affiliation(s)
- Yashuang Liu
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Siyu Li
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Kexin Wang
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Yiwen Wang
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Yange Wang
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Chenxu Zhang
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Hao Wu
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Gang Wang
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Fangyuan Qin
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China
| | - Zongming Song
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China.
| | - Ye Tao
- Department of Ophthalmology, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Henan Eye Institute, Henan Eye Hospital, Zhengzhou 450003, China.
| |
Collapse
|
8
|
Zhang X, Fang Y, Rong D, Li J, Li Z, Qiu H, Chen Q, Yang J, Wang C, Huang J, Zhao Q, Yang S, Wang H. A novel taxane SB-T-101141 triggers a noncanonical ferroptosis to overcome Paclitaxel resistance of breast cancer via iron homeostasis-related KHSRP. Cell Death Dis 2025; 16:403. [PMID: 40389408 PMCID: PMC12089390 DOI: 10.1038/s41419-025-07710-0] [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: 01/10/2025] [Revised: 04/16/2025] [Accepted: 05/01/2025] [Indexed: 05/21/2025]
Abstract
Acquired multidrug resistance impedes the clinical application of paclitaxel. Here, we disclosed that the taxane SB-T-101141 efficiently contributed to a novel ferroptosis-like cell death of Paclitaxel-resistant and parental breast cancer cells. Functionally, SB-T-101141 facilitated the production of iron and ferrous ions along with reactive oxygen species (ROS), composed of lipid ROS and lipid peroxidation-derived aldehydes, including malonaldehyde (MDA), and glutathione (GSH) depletion. Iron chelators and ROS scavengers significantly attenuated cell death, and the inorganic ROS rendered by SB-T-101141. However, the ferroptosis-associated lipid oxide inhibitors could not block the lipid ROS and cell death triggered by SB-T-101141. Meanwhile, via genome-scale CRISPR-Cas9 screening, we uncovered that SB-T-101141 bound to the KH-type splicing regulatory protein (KHSRP) to inhibit the iron-dependent expression of CDGSH iron sulfur domain 1 (CISD1) associated with iron homeostasis, which consequently led to a novel type of ferroptosis of breast tumors. Moreover, RNA deep sequencing indicated that SB-T-101141 synergistically enhanced the iron-dependent activation of JNK and PERK pathways via KHSRP. Altogether, our results here demonstrate the potential clinical application of SB-T-101141 as a novel ferroptosis inducer in Paclitaxel-resistant breast cancer treatment.
Collapse
Affiliation(s)
- Xiaomei Zhang
- Centre for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ying Fang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dade Rong
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Jie Li
- Engineering Research Center of Tibetan Medicine Detection Technology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Zhe Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Huidan Qiu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qiuxia Chen
- Centre for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jing Yang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Changwei Wang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, China
| | - Junxiu Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qin Zhao
- Engineering Research Center of Tibetan Medicine Detection Technology, School of Medicine, Xizang Minzu University, Xianyang, China
| | - Shulan Yang
- Centre for Translational Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Haihe Wang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, China.
- Clinical Medical Research Centre for Plateau Gastroenterological Diseases of Xizang Autonomous Region, Xizang Minzu University, Xianyang, China.
| |
Collapse
|
9
|
Trivedi A, Saxena V, Banaee M, Bakhasha J, Arya N, Yadav KK, Rizzo MG, Faggio C. Unveiling the crosstalk between unfolded protein response and apoptosis in triclosan induced hepatotoxicity in Labeo rohita. Sci Rep 2025; 15:17089. [PMID: 40379834 DOI: 10.1038/s41598-025-93997-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 03/11/2025] [Indexed: 05/19/2025] Open
Abstract
Triclosan (TCS), a widely used antimicrobial, has emerged as a concerning aquatic pollutant, especially post-COVID-19 due to increased disinfectant use. The study aimed to investigate the toxic impacts of TCS on Labeo rohita, revealing a 96-h LC50 of 0.742 mg/L. Well-acclimatized fish were categorised into three groups: one control and two treatment (T1 and T2) that were exposed to 1/10th and 1/5th of 96-h LC50 i.e., 0.0742 mg/L and 0.148 mg/L, respectively for 6 weeks under semi-static condition. Significantly (p < 0.05) elevated extents of reactive oxygen species (ROS) in the liver indicated enhanced oxidative stress. The activities of enzymatic antioxidants viz. superoxide dismutase (SOD) and catalase (CAT) were significantly (p < 0.05) increased while the reduced glutathione (GSH) levels were significantly (p < 0.05) decreased in a dose- and duration-dependent manner. A significant (p < 0.05) increase was observed in the lipid peroxidation (LPO) rate, which coincided with disruptions in the histological structure of the liver. Significant (p < 0.05) induction in Micronuclei frequency validated genomic instability. Furthermore, Endoplasmic Reticulum (ER) stress was marked by the increased intracellular calcium levels and elevated expression of grp78, chop, atf4, perk, eIF2α and gadd34 in TCS-exposed groups. Moreover, the consequent activation of bax, caspase-3, caspase-9, apaf-1 and the regulation of bcl-2 due to ER-stress, ultimately resulted in apoptosis of hepatocytes. Pearson correlation matrix and Principal Component Analysis (PCA) further underscored key interactions among parameters. Our findings demonstrate that the TCS, at its sub-lethal concentrations, is causing severe anomalies and even cell-death via triggering ER-stress-mediated apoptotic pathway in the fish liver.
Collapse
Affiliation(s)
- Abha Trivedi
- Toxicogenomics Laboratory, Department of Animal Science, M.J.P. Rohilkhand University, Bareilly, 243006, India.
| | - Vaishnavi Saxena
- Toxicogenomics Laboratory, Department of Animal Science, M.J.P. Rohilkhand University, Bareilly, 243006, India
| | - Mahdi Banaee
- Department of Environmental Sciences, Faculty of Natural Resources, Behbahan Khatam Alanbia University of Technology, Behbahan, Iran
| | - Jumman Bakhasha
- Toxicogenomics Laboratory, Department of Animal Science, M.J.P. Rohilkhand University, Bareilly, 243006, India
| | - Neeti Arya
- Toxicogenomics Laboratory, Department of Animal Science, M.J.P. Rohilkhand University, Bareilly, 243006, India
| | - Kamlesh K Yadav
- Department of Zoology, Government Degree College, Bakkha Kheda, Unnao, 209801, India
| | - Maria Giovanna Rizzo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Caterina Faggio
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
- Department of Eco-Sustainable Marine Biotechnology, Stazione Zoologica Anton Dohrn, Naples, Italy
| |
Collapse
|
10
|
Muneeb M, Abdallah DM, El-Abhar HS, Wadie W, Ahmed KA, Abul Fadl YS. Antiplatelet therapy as a novel approach in Parkinson's disease: Repositioning Ticagrelor to alleviate rotenone-induced parkinsonism via modulation of ER stress, apoptosis, and autophagy. Neuropharmacology 2025; 269:110346. [PMID: 39914616 DOI: 10.1016/j.neuropharm.2025.110346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/02/2025] [Accepted: 02/03/2025] [Indexed: 02/18/2025]
Abstract
Cardiovascular diseases, such as myocardial infarction, ischemic stroke, and coronary heart ailments have been closely associated with Parkinson's disease (PD). Despite this established link, the potential neuroprotective impact of the potent antiplatelet agent ticagrelor (Tica) remains unexplored against PD. Thus, we hypothesized that Tica could be repurposed as a therapeutic agent against PD. Rotenone experimental model was adopted in Wistar male rats by administering rotenone subcutaneously on alternate days during a 21-day experimental period and treating a subset of rats with Tica orally for the last 11 consecutive days. The administration of Tica improved motor function (open field test, hanging wire test) and restored striatal histological features. Additionally, Tica opposed the rotenone effect and markedly obliterated the striatal α-synuclein content but enhanced the protein expression of tyrosine hydroxylase and dopamine content. On the molecular level, Tica inhibited striatal endoplasmic reticulum stress (ERS) as evidenced by the downregulation of the ER-resident transmembrane sensor inositol-requiring enzyme 1 alpha and its downstream molecular targets, TNF receptor-associated factor 2 and c-Jun N-terminal kinase, along with a reduction in caspase-3 activity. On the other hand, Tica augmented the autophagy machinery by upregulating the autophagosome markers Beclin-1 and light chain 3-II, while inhibiting the content of cathepsin D. Therefore, the current study is the first to accentuate the neuroprotective potential of Tica in a rat model of PD via modulating the crosstalk between ERS, apoptosis, and autophagy to represent a potential novel therapeutic candidate for managing PD, particularly in patients with or prone to cardiovascular diseases.
Collapse
Affiliation(s)
- Muhammad Muneeb
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt (FUE), 90th Str., 11835, Cairo, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., 11562 Cairo, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt (FUE), 90th Str., 11835, Cairo, Egypt.
| | - Walaa Wadie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Κasr El-Aini Str., 11562 Cairo, Egypt
| | - Kawkab A Ahmed
- Pathology Department, Faculty of Veterinary Medicine, Cairo University, Giza, 12211, Egypt
| | - Yasmine S Abul Fadl
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt (FUE), 90th Str., 11835, Cairo, Egypt
| |
Collapse
|
11
|
Liao X, Li X, Mou A, Zhang Q, Dong Y, Li Y, Zhang X, Xu Q. Thermal acclimatization mechanisms in the cold-water Ophiuroid Ophiura sarsii vadicola: Regulation of protein homeostasis and metabolic pathways. J Therm Biol 2025; 130:104137. [PMID: 40413918 DOI: 10.1016/j.jtherbio.2025.104137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 04/22/2025] [Accepted: 05/06/2025] [Indexed: 05/27/2025]
Abstract
Global climate change is driving rapid warming in the Pacific Arctic cold-water regions. As dominant species in these areas, the Ophiura sarsii complex suffer from ocean warming stress, resulting in altered distribution patterns, physiological behaviors, and thermotolerance. However, their molecular response mechanisms to ocean warming remain unclear. We conducted transcriptomic analyses of O. sarsii vadicola under various temperature treatments: a control group at in-situ temperature (7 °C), two warming groups at the sub-lethal temperature 19 °C for one week (short-term) and two months (long-term), and a low-temperature reference group of O. sarsii at 0 °C. The results showed that a total of 66 core differentially expressed genes (58 up-regulated, 8 downregulated) were identified, with expression patterns varying by warming duration. Ophiura sarsii vadicola exhibited a more pronounced response to long-term warming than short-term ones. Three transcription factors HMGB3, METTL19, and HEAT were identified as key regulators within the co-expression networks. Furthermore, changes in folding enzymes activity and an upregulation of protein processing and chaperone genes were observed, indicating a potential activation of the unfolded protein response in brittle stars. Elevated temperatures also induced changes in multiple energy metabolism-related pathways. These findings suggest that O. sarsii vadicola may mitigate the negative effects of ocean warming by regulating key gene expression, enhancing protein homeostasis control and energy redistribution. This study provides valuable insights into the evolution and adaptability mechanisms of cold-water species under global climate change.
Collapse
Affiliation(s)
- Xiaomei Liao
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China; Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, 266237, China; School of Ocean Sciences, China University of Geosciences (Beijing), Beijing, 100083, China.
| | - Xinlong Li
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China; Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Anning Mou
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China; Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Qian Zhang
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China; Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Yue Dong
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China; Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Yixuan Li
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China.
| | - Xuelei Zhang
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China; Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| | - Qinzeng Xu
- Key Laboratory of Marine Eco-Environmental Science and Technology, First Institute of Oceanography, Ministry of Natural Resources, Qingdao, 266061, China; Laboratory for Marine Ecology and Environmental Science, Qingdao Marine Science and Technology Center, Qingdao, 266237, China.
| |
Collapse
|
12
|
Park YJ, Lu TC, Jackson T, Goodman LD, Ran L, Chen J, Liang CY, Harrison E, Ko C, Chen X, Wang B, Hsu AL, Ochoa E, Bieniek KF, Yamamoto S, Zhu Y, Zheng H, Qi Y, Bellen HJ, Li H. Distinct systemic impacts of Aβ42 and Tau revealed by whole-organism snRNA-seq. Neuron 2025:S0896-6273(25)00299-5. [PMID: 40381615 DOI: 10.1016/j.neuron.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/27/2025] [Accepted: 04/17/2025] [Indexed: 05/20/2025]
Abstract
Both neuronal and peripheral tissues become disrupted in Alzheimer's disease (AD). However, a comprehensive understanding of how AD impacts different tissues across the whole organism is lacking. Using Drosophila, we generated an AD Fly Cell Atlas (AD-FCA) based on whole-organism single-nucleus transcriptomes of 219 cell types from flies expressing AD-associated proteins, either human amyloid-β 42 peptide (Aβ42) or Tau, in neurons. We found that Aβ42 primarily affects the nervous system, including sensory neurons, while Tau induces accelerated aging in peripheral tissues. We identified a neuronal cluster enriched in Aβ42 flies, which has high lactate dehydrogenase (LDH) expression. This LDH-high cluster is conserved in 5XFAD mouse and human AD datasets. We found a conserved defect in fat metabolism from both fly and mouse tauopathy models. The AD-FCA offers new insights into how Aβ42 or Tau systemically and differentially affects a whole organism and provides a valuable resource for understanding brain-body communication in neurodegeneration.
Collapse
Affiliation(s)
- Ye-Jin Park
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tzu-Chiao Lu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tyler Jackson
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Program in Cancer Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lindsey D Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Lindsey Ran
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jiaye Chen
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chung-Yi Liang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Erin Harrison
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christina Ko
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xi Chen
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Baiping Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ao-Lin Hsu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Department of Internal Medicine, Division of Geriatric and Palliative Medicine, University of Michigan, Ann Arbor, MI 28109, USA
| | - Elizabeth Ochoa
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Kevin F Bieniek
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229, USA; Department of Pathology & Laboratory Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yi Zhu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yanyan Qi
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Hongjie Li
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Panda SK, Sanchez-Pajares IR, Rehman A, Del Vecchio V, Mele L, Chipurupalli S, Robinson N, Desiderio V. ER stress and/or ER-phagy in drug resistance? Three coincidences are proof. Cell Commun Signal 2025; 23:223. [PMID: 40361118 PMCID: PMC12070796 DOI: 10.1186/s12964-025-02232-w] [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: 02/10/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025] Open
Abstract
Cancer is influenced by the tumor microenvironment (TME), which includes factors such as pH, hypoxia, immune cells, and blood vessels. These factors affect cancer cell growth and behavior. The tumor microenvironment triggers adaptive responses such as endoplasmic reticulum (ER) stress, unfolded protein response (UPR), and autophagy, posing a challenge to cancer treatment. The UPR aims to restore ER homeostasis by involving key regulators inositol-requiring enzyme-1(IRE1), PKR-like ER kinase (PERK), and activating transcription factor 6 (ATF6). Additionally, ER-phagy, a selective form of autophagy, eliminates ER components under stress conditions. Understanding the interplay between hypoxia, ER stress, UPR, and autophagy in the tumor microenvironment is crucial for developing effective cancer therapies to overcome drug resistance. Targeting the components of the UPR and modulating ER-phagy could potentially improve the efficacy of existing cancer therapies. Future research should define the conditions under which ER stress responses and ER-phagy act as pro-survival versus pro-death mechanisms and develop precise methods to quantify ER-phagic flux in tumor cells.
Collapse
Affiliation(s)
- Sameer Kumar Panda
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, 80138, Italy
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
| | | | - Ayesha Rehman
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, 80138, Italy
| | - Vitale Del Vecchio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, 80138, Italy
- Department of Life Sciences, Health, and Health Professions, Link Campus University, Via del Casale Di San Pio V 4, Rome, 00165, Italia
| | - Luigi Mele
- University of Basilicata, Via Dell'Ateneo Lucano 10, Potenza, 85100, Italy
| | - Sandhya Chipurupalli
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
- Department of Pediatrics & Biochemistry and Molecular Biology, Dalhousie University, Halifax, NS, Canada
| | - Nirmal Robinson
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Vincenzo Desiderio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, 80138, Italy.
| |
Collapse
|
14
|
Bin H, Wen W. Metformin attenuates endoplasmic reticulum stress in diabetic kidney disease: mechanistic insights and future perspectives. Int Urol Nephrol 2025:10.1007/s11255-025-04562-7. [PMID: 40343634 DOI: 10.1007/s11255-025-04562-7] [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: 02/07/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
Diabetic kidney disease (DKD) is a common microvascular complication of diabetes that can lead to end-stage renal failure. Emerging evidence suggests that endoplasmic reticulum (ER) stress plays a crucial role in the pathogenesis of DKD by affecting various renal parenchymal cells, including endothelial cells, podocytes, and mesangial cells. This review comprehensively examines the relationship between ER stress and DKD, focusing on how metformin, a first-line antidiabetic medication, ameliorates ER stress-induced kidney injury. Multiple factors, including reactive oxygen species (ROS), proteinuria, and advanced glycation end products (AGEs), contribute to ER stress in DKD. Metformin's renoprotective effects are primarily mediated through activation of the AMPK signaling pathway, which modulates ER stress response, reduction of oxidative stress and its impact on ER function, and improvement of mitochondrial function. These mechanisms collectively lead to decreased proteinuria, reduced cell apoptosis, and attenuated epithelial-mesenchymal transition in diabetic kidneys. Understanding these molecular mechanisms provides new insights into the therapeutic potential of metformin in DKD treatment. However, further research is needed to elucidate the precise molecular pathways through which metformin regulates ER stress in different renal cell types under diabetic conditions.
Collapse
Affiliation(s)
- Huang Bin
- Department of Endocrinology, Division of Life Science and Medicine, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, 230000, China
| | - Wenjie Wen
- Anhui Province Engineering Research Center for Dental Materials and Application, School of Stomatology, Wannan Medical College, Wuhu, 241002, China.
| |
Collapse
|
15
|
Chen YF, Teng YC, Yang JH, Kao CH, Tsai TF. Cisd1 synergizes with Cisd2 to modulate protein processing by maintaining mitochondrial and ER homeostasis. Aging (Albany NY) 2025; 17:206249. [PMID: 40349253 DOI: 10.18632/aging.206249] [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: 05/22/2024] [Accepted: 03/24/2025] [Indexed: 05/14/2025]
Abstract
Connection and crosstalk among the organelles critically contribute to cellular functions. Destruction of any kind of organelle is likely to induce a series of intracellular disorders and finally lead to cell death. Because of its subcellular locations, CDGSH iron-sulfur domain-containing protein 1 (Cisd1) and Cisd2 have functions that are related to maintaining mitochondria and ER homeostasis. As previous reports have shown, Cisd2 knockout mice have a decreased body weight and poor survival rate, and the primary defects were conducted in skeletal muscle. Our previous findings indicated that Cisd1 deletion causes a range of skeletal muscle defects in mice with Cisd2 deficiency, including mitochondrial degeneration, endoplasmic reticulum (ER) stress, and alteration of protein process, as well as programmed cell death. In Cisd1 and Cisd2 deficient condition, the whole of the protein biosynthesis was damaged, including translation, modification, transport, and degradation. Changes in the immune response, redox regulation, and metabolism were also present in Cisd1 and Cisd2 double knockout mice. Overall, we have demonstrated that Cisd1 and Cisd2 knockout have a synergistic effect on skeletal muscles, and that Cisd2 plays a more critical role than Cisd1. These synergistic effects impact signaling regulation and interrupt the crosstalk and homeostasis of organelles. This creates severe disorders in various tissues and organs.
Collapse
Affiliation(s)
- Yi-Fan Chen
- International Master Program for Translation Science, College of Medical Science and Technology, Taipei Medical University, New Taipei City 23564, Taiwan
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11529, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
- Master Program in Clinical Genomics and Proteomics, School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Yuan-Chi Teng
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112, Taiwan
| | - Jian-Hsin Yang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Cheng-Heng Kao
- Center of General Education, Chang Gung University, Taoyuan 333, Taiwan
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Center for Healthy Longevity and Aging Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan 350, Taiwan
| |
Collapse
|
16
|
Li X, Hu F, Lu T, Wu S, Ma G, Lin Y, Zhang H. Endoplasmic reticulum stress in non-small cell lung cancer. Am J Cancer Res 2025; 15:1829-1851. [PMID: 40371139 PMCID: PMC12070083 DOI: 10.62347/rgrq7608] [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: 01/21/2025] [Accepted: 04/16/2025] [Indexed: 05/16/2025] Open
Abstract
The Endoplasmic reticulum (ER), an organelle present in various eukaryotic cells, is responsible for protein synthesis, modification, folding, and transport, as well as for the regulation of lipid metabolism and Ca2+ homeostasis. ER stress plays a pivotal role in the pathogenesis and therapeutic response of non-small cell lung cancer (NSCLC), significantly influencing cellular fate decisions through its unique sensing and regulatory mechanisms. This review aims to elucidate the key role of ER stress sensors and to explore how they mediate cell autophagy, apoptosis, and non-apoptotic modes of cell death in the context of drug-treated NSCLC. This investigation lays a solid foundation for optimizing future treatment strategies for NSCLC.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| | - Fangning Hu
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| | - Tong Lu
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| | - Shuo Wu
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| | - Guanqiang Ma
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| | - Yani Lin
- Shandong Provincial Hospital Affiliated to Shandong First Medical University and Shandong Academy of Medical Sciences, School of Laboratory Animal and Shandong Laboratory Animal Center, Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical SciencesJinan, Shandong, China
| | - Hua Zhang
- Department of Thoracic Surgery, Shandong Provincial Public Health Clinical CenterJinan, Shandong, China
| |
Collapse
|
17
|
Jiang KC, Zhu YH, Jiang ZL, Liu Y, Hussain W, Luo HY, Sun WH, Ji XY, Li DX. Regulation of PEST-containing nuclear proteins in cancer cells: implications for cancer biology and therapy. Front Oncol 2025; 15:1548886. [PMID: 40330830 PMCID: PMC12052563 DOI: 10.3389/fonc.2025.1548886] [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: 12/20/2024] [Accepted: 04/01/2025] [Indexed: 05/08/2025] Open
Abstract
The PEST-containing nuclear protein (PCNP) is a nuclear protein involved in the regulation of cell cycle progression, protein degradation, and tumorigenesis. PCNP contains a PEST sequence, a polypeptide structural motif rich in proline (P), glutamic acid (E), serine (S), and threonine (T), which serves as a proteolytic recognition signal. The degradation of specific proteins via the PEST sequence plays a crucial role in modulating signaling pathways that control cell growth, differentiation, apoptosis, and stress responses. PCNP is primarily degraded through the ubiquitin-proteasome system (UPS) and the calpain pathway, with phosphorylation of threonine and serine residues further accelerating its degradation. The ubiquitination of PCNP by the ring finger protein NIRF in an E3 ligase-dependent manner is well documented, along with its involvement in the MAPK and PI3K/AKT/mTOR signaling pathways. Additionally, PCNP is implicated in p53-mediated cell cycle arrest and apoptosis, which are essential for inhibiting tumor growth. To explore the role of PCNP in cancer, this review examines its effects on cell growth, differentiation, proliferation, and apoptosis in lung adenocarcinoma, thyroid cancer, ovarian cancer, and other malignancies derived from glandular epithelial cells. By focusing on PCNP and its regulatory mechanisms, this study provides a scientific basis for further research on the biological functions of the PEST sequence in tumor development and cancer progression.
Collapse
Affiliation(s)
- Kai-Chun Jiang
- Department of Traditional Chinese Medicine, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China
| | - Yong-Hao Zhu
- School of Stomatology, Henan University, Kaifeng, Henan, China
| | - Zhi-Liang Jiang
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Yi Liu
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Wahab Hussain
- School of Stomatology, Henan University, Kaifeng, Henan, China
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
| | - Huang-Yin Luo
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Wei-Hang Sun
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Urology, Institute of Urology, Sichuan University, Chengdu, China
| | - Xin-Ying Ji
- Kaifeng Municipal Key Laboratory for Infection and Biosafety, Henan International Joint Laboratory of Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University College of Medicine, Kaifeng, Henan, China
- Department of Oncology, Huaxian County Hospital, Anyang, Henan, China
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, China
| | - Ding-Xi Li
- The Affiliated Cancer Hospital, Zhengzhou University & Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
18
|
Zhu L, Yang Y, Tan J, Lin Y, Qing J, Li X, Zeng L. Effect of 2,5-hexanedione on rat ovarian granulosa cell apoptosis involves endoplasmic reticulum stress-dependent m-TOR signaling pathway. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:319-328. [PMID: 39668517 DOI: 10.1080/15287394.2024.2438832] [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: 12/14/2024]
Abstract
Occupational exposure to N-hexane/2,5-hexanedione (2,5-HD) was found to adversely affect reproductive functions in females. However, there are few studies regarding the mechanisms underlying reproductive system damage initiated by 2,5-HD. Several studies demonstrated that 2,5-HD exerts hormonal dysfunctions in females by promoting apoptosis using rat ovarian granulosa cells (GCs) as a model. The endoplasmic reticulum (ER) plays a key role in cellular processes such as protein folding and modification, Ca2+ storage, and lipid synthesis, which are known to involve the activation of stress (ERS)-dependent m-TOR signaling pathway. Thus, the aim of this study was to examine the effects of 2,5-HD on ER and the associated activation of stress (ERS)-dependent m-TOR signaling pathway resulting in consequent apoptosis of ovarian GCs. Data demonstrated that after intraperitoneal treatment with 100, 200, or 400 mg/kg 2,5-HD for 6 consecutive weeks, 5 times per week, a decrease in body weight, ovarian weight, and relative ovary weight was found. Terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay showed that 2,5-HD promoted apoptosis of ovarian GCs, which involved enhanced relative protein expression levels of m-TOR/p-mTOR. Our findings demonstrated that 2,5-HD (1) elevated expression levels of pro-apoptosis-related genes Bax and Caspase 3, (2) decreased expression levels of the anti-apoptosis gene Bcl-2, and (3) activated the protein expression of glucose-regulatory protein 78 (GRP78), inositol-requiring enzyme-1 (IRE1), and c-Jun terminal kinase (JNK) associated with increased apoptosis. Evidence indicates that chronic exposure to 2,5-HD induced apoptosis of ovarian GCs, and the possible mechanism underlying this effect involves the ERS-dependent m-TOR signaling pathway.
Collapse
Affiliation(s)
- Lemei Zhu
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
| | - Yue Yang
- Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Basic Medical Sciences, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Jingsi Tan
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
| | - Yibo Lin
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
| | - Jiaqi Qing
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
| | - Xin Li
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
| | - Lingfeng Zeng
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations & School of Public Health, Changsha Medical University, Changsha, China
- Department of Pharmacology and Toxicology, Hunan Key Laboratory of Pharmacodynamics and Safety Evaluation of New Drugs & Hunan Provincial Research Center for Safety Evaluation of Drugs, Changsha, China
| |
Collapse
|
19
|
Albini A, Di Paola L, Mei G, Baci D, Fusco N, Corso G, Noonan D. Inflammation and cancer cell survival: TRAF2 as a key player. Cell Death Dis 2025; 16:292. [PMID: 40229245 PMCID: PMC11997178 DOI: 10.1038/s41419-025-07609-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 03/25/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025]
Abstract
TNF receptor-associated factor 2 (TRAF2) plays a crucial role in both physiological and pathological processes. It takes part in the regulation of cell survival and death, tissue regeneration, development, endoplasmic reticulum stress response, autophagy, homeostasis of the epithelial barrier and regulation of adaptive and innate immunity. Initially identified for its interaction with TNF receptor 2 (TNFR2), TRAF2 contains a TRAF domain that enables homo- and hetero-oligomerization, allowing it to interact with multiple receptors and signaling molecules. While best known for mediating TNFR1 and TNFR2 signaling, TRAF2 also modulates other receptor pathways, including MAPK, NF-κB, and Wnt/β-catenin cascades. By regulating NF-κB-inducing kinase (NIK), TRAF2 is a key activator of the alternative NF-κB pathway, linking it to inflammatory diseases, immune dysfunction, and tumorigenesis. In the innate immune system, TRAF2 influences macrophage differentiation, activation, and survival and stimulates natural killer cell cytotoxicity. In the adaptive immune system, it represses effector B- and T-cell activity while sustaining regulatory T-cell function, thus promoting immune suppression. The lack of fine-tuning of TRAF2 activity leads to excessive NF-kB activation, driving chronic inflammation and autoimmunity. Although TRAF2 can act as a tumor suppressor, it is predominantly described as a tumor promoter, as its expression has been correlated with increased metastatic potential and poorer prognosis in several types of cancer. Targeting TRAF2 or TRAF2-dependent signaling pathways might represent a promising anti-cancer therapeutic strategy.
Collapse
Grants
- The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
- PRIN 2022, grant 2022PJKF88 The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
- PRIN 2022 The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
- "Umberto Veronesi" Foundation project: "Massive CDH1 genetic screening in the so-called hereditary breast-gastric cancer syndrome". The work was also supported by the Italian Ministry of Health Ricerca Corrente to IRCCS IEO, European Institute of Oncology, and IRCCS MultiMedica, Italy.
Collapse
Affiliation(s)
- Adriana Albini
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.
| | - Luisa Di Paola
- Unit of Chemical-Physics Fundamentals in Chemical Engineering, Faculty Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico, Rome, Italy
| | - Giampiero Mei
- Department of Experimental Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Denisa Baci
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- Molecular Cardiology Laboratory, IRCCS-Policlinico San Donato, Milan, Italy
| | - Nicola Fusco
- European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy
| | - Giovanni Corso
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
- Division of Breast Surgery, European Institute of Oncology (IEO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Milan, Italy.
| | - Douglas Noonan
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
- IRCCS MultiMedica, Milan, Italy
| |
Collapse
|
20
|
Lu M, Ren Y, Feng S, Wang S, Xia W, Gu B, Shen Y, Yue A, Li N, Zhang Y, Zhong J. MDM2 inhibitor induces apoptosis in colon cancer cells through activation of the CHOP-DR5 pathway, independent of p53 phenotype. Front Pharmacol 2025; 16:1508421. [PMID: 40264676 PMCID: PMC12011796 DOI: 10.3389/fphar.2025.1508421] [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: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Murine double minute 2 (MDM2), a key negative regulator of p53, forms a feedback loop with p53 to drive tumor progression, including colorectal cancer. Nutlin-3a, an MDM2 inhibitor, induces apoptosis in wild-type p53 tumors, but its effects on p53-mutated cancers and potential p53-independent apoptotic mechanisms remain unclear. Methods We investigated Nutlin-3a's effects on colon cancer cells with varying p53 phenotypes. Endoplasmic reticulum (ER) stress-associated CHOP was detected and knocked down to explore mechanisms. In vitro and in vivo experiments assessed Nutlin-3a's synergy with 5-fluorouracil and TRAIL. Results Nutlin-3a activated caspase-8-dependent extrinsic apoptosis in colon cancer cells via DR5 upregulation, independent of p53 status. ER stress and CHOP activation mediated DR5 induction, driven by calcium release. Combined Nutlin-3a treatment enhanced sensitivity to 5-fluorouracil and TRAIL in vitro and in vivo through caspase-8 pathway activation. Discussion These findings reveal a novel p53-independent apoptotic mechanism of Nutlin-3a involving ER stress and death receptor signaling. This pathway highlights Nutlin-3a's potential as an adjuvant therapy for colon cancer, even in p53-mutated tumors, by enhancing chemotherapeutic efficacy through extrinsic apoptosis.
Collapse
Affiliation(s)
- Manman Lu
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yingli Ren
- College of Medicine, Henan Polytechnic University, Jiaozuo, China
| | - Sijia Feng
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Shenggen Wang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Weiyue Xia
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Baoru Gu
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yuhou Shen
- Department of Abdominal Surgical Oncology Ward 2, Xinxiang Central Hospital, Xinxiang, China
- Department of Abdominal Surgical Oncology Ward 2, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Aimin Yue
- Department of Abdominal Surgical Oncology Ward 2, Xinxiang Central Hospital, Xinxiang, China
- Department of Abdominal Surgical Oncology Ward 2, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, China
| | - Na Li
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yongxi Zhang
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jiateng Zhong
- Department of Oncology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Henan Province Engineering Technology Research Center of Tumor Diagnostic Biomarkers and RNA Interference Drugs, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
21
|
Kropp EM, Matono S, Wang OY, Robida AM, Kandarpa M, Grant JL, Oleson BJ, Alt A, Talpaz M, Pianko M, Li Q. Identification of Omaveloxolone as An Endoplasmic Reticulum Associated Degradation Inhibitor That Induces Early Apoptotic Signaling in Multiple Myeloma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646787. [PMID: 40236186 PMCID: PMC11996520 DOI: 10.1101/2025.04.02.646787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Endoplasmic reticulum-associated degradation (ERAD) is essential for maintaining protein homeostasis, yet its regulatory mechanisms remain poorly understood. A major challenge in studying ERAD is the lack of specific inhibitors targeting the ERAD complex. To address this, we conducted a cell-based high-throughput screen using the FDA-repurposing library and identified omaveloxolone (RTA408) as a potent ERAD inhibitor that selectively impairs the degradation of ER luminal and membrane substrates. Beyond its utility in identifying ERAD substrates, RTA408 exhibits strong cytotoxic effects in multiple myeloma (MM), an incurable plasma cell malignancy. RTA408 inhibits ERAD activity and rapidly induces apoptotic signaling via caspase 8 and the death-inducing signaling complex (DISC). Notably, RTA408 is cytotoxic to malignant plasma cells, including those resistant to proteasome inhibitors, and demonstrates in vivo anti-myeloma activity. Our findings establish ERAD inhibitors as valuable tools for dissecting ERAD regulation while also highlighting their potential as therapeutic agents for MM. Highlights We developed a cell-based screening approach to identify novel modulators of endoplasmic reticulum associated degradation (ERAD), with implications in studying ERAD biology and targeting plasma cell neoplasms.Screening of the FDA-repurposing library identified Omaveloxolone (RTA408) as an inhibitor of luminal and membrane ERAD substrate degradation, which can be leveraged to identify ERAD substrates.maveloxolone treatment rapidly induces the unfolded protein response and apoptosis that is dependent on caspase 8 and death-inducing signaling complex (DISC) in multiple myeloma cells. maveloxolone exhibits cytotoxic effects against multiple myeloma cells in vitro and in vivo and induces apoptosis in primary plasma cells from patients with relapsed/refractory myeloma.
Collapse
|
22
|
Zhang Y, Liu H, Liu D, Zhang H, Ma Y, Li N, Zhang C, Xue M, Wang F, Jia X, Zhang H, Tang K, Xu X, Wang S, Wei Y, Yang X, Zuo J, Chen L, Jin B, Zhang Y. Hantaan virus infection induces human mucosal-associated invariant T cell pyroptosis through IRE1α pathway. Commun Biol 2025; 8:538. [PMID: 40169922 PMCID: PMC11961572 DOI: 10.1038/s42003-025-07979-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
Hantaan virus (HTNV) triggers an epidemic of hemorrhagic fever with renal syndrome (HFRS), which is predominantly prevalent in Asia. Mucosal-associated invariant T (MAIT) cells, categorized as innate-like T lymphocytes, perform crucial functions in the innate host defense mechanism during virus infection. We previously showed that MAIT cells played antiviral roles in vitro. But marked reduction of MAIT cells was present in the peripheral blood of HFRS patients. Till now, the role of MAIT cells in vivo and the mechanisms of HTNV-induced the MAIT cell deficiency have not yet been fully explored. In this study, by combining the clinical samples, MAIT deficiency mice and in vitro infected MAIT cell models, we find that pyroptosis was the main reason of MAIT cell loss in the peripheral blood of HFRS patients. The molecular mechanisms are related to the overload of calcium in the endoplasmic reticulum (ER) of MAIT cells, which subsequently induces inosital-requiring enzyme-1α (IRE1α)-mediated ER-stress and following pyroptosis. ER-stress inhibitor can reverse the pyroptosis of MAIT cells during HTNV infection. In conclusion, this study firstly reveals the underlying molecular mechanisms for the deficiency of MAIT cells during HTNV infection, and suggests a potential way to stabilize the MAIT cells population in HFRS.
Collapse
Affiliation(s)
- Yusi Zhang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - He Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Dalu Liu
- Department of Radiation Medicine and Protection, Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, School of Public Health, Fourth Military Medical University, Xi'an, 710032, China
| | - Huiyuan Zhang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Ying Ma
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Na Li
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Chunmei Zhang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Manling Xue
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | | | | | - Hui Zhang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Kang Tang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaoyue Xu
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Department of Immunology, School of Basic Medical Sciences, Yan'an university, Yan'an, 716000, China
| | - Shijia Wang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Department of Immunology, School of Basic Medical Sciences, Yan'an university, Yan'an, 716000, China
| | - Yiwen Wei
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Yan'an university, Yan'an, 716000, China
| | - Xiaojing Yang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- School of Life Sciences, Yan'an university, Yan'an, 716000, China
| | - Jiajia Zuo
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
- Department of Immunology, School of Basic Medical Sciences, Yan'an university, Yan'an, 716000, China
| | - Lihua Chen
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Boquan Jin
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Yun Zhang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
23
|
Mao W, Zhang Z. The Hsa_circ_0105558/miR-182-5p/ATF6 Cascade Affects H 2O 2-Triggered Oxidative Damage and Apoptosis of Human Lens Epithelial Cells. Biochem Genet 2025; 63:1241-1257. [PMID: 38530576 DOI: 10.1007/s10528-024-10753-1] [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: 07/26/2023] [Accepted: 02/20/2024] [Indexed: 03/28/2024]
Abstract
Age-related cataract (ARC) is the prevalent cause of useful vision loss. Circular RNAs are related to ARC pathogenesis partly through their competing endogenous RNA (ceRNA) activity. Herein, we defined the action of hsa_circ_0105558 in hydrogen peroxide (H2O2)-driven apoptosis and oxidative damage in human lens epithelial SRA01/04 cells. Hsa_circ_0105558, microRNA (miR)-182-5p and activating transcription factor 6 (ATF6) were evaluated by a qRT-PCR or immunoblotting method. The hsa_circ_0105558/miR-182-5p and miR-182-5p/ATF6 relationships were predicted by bioinformatics analysis and confirmed by dual-luciferase reporter assay. Reactive oxygen species level, glutathione peroxidase level, superoxide dismutase activity, and malondialdehyde level were measured using the matched assay kits. Hsa_circ_0105558 was upregulated in human ARC lens and H2O2-exposed SRA01/04 cells. Suppression of hsa_circ_0105558 attenuated H2O2-driven SRA01/04 cell apoptosis and oxidative damage. Hsa_circ_0105558 targeted miR-182-5p, and reduced miR-182-5p expression reversed the influence of hsa_circ_0105558 depletion on H2O2-driven oxidative damage and apoptosis. ATF6 was a target of miR-182-5p, and miR-182-5p-driven downregulation of ATF6 regulated cell oxidative damage and apoptosis under H2O2 insult. Moreover, hsa_circ_0105558 functioned as a ceRNA to post-transcriptionally control ATF6 expression through miR-182-5p competition. Our study demonstrates that hsa_circ_0105558 modulates SRA01/04 cell oxidative damage and apoptosis under H2O2 insult through the miR-182-5p/ATF6 cascade.
Collapse
Affiliation(s)
- Wei Mao
- Refractive Surgery Department, Ningbo Eye Hospital, Ningbo, 315010, Zhejiang, China.
| | - Zhe Zhang
- Refractive Surgery Department, Ningbo Eye Hospital, Ningbo, 315010, Zhejiang, China
| |
Collapse
|
24
|
Saulle I, Vitalyos AV, D’Agate D, Clerici M, Biasin M. Unveiling the impact of ERAP1 and ERAP2 on migration, angiogenesis and ER stress response. Front Cell Dev Biol 2025; 13:1564649. [PMID: 40226591 PMCID: PMC11985534 DOI: 10.3389/fcell.2025.1564649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/12/2025] [Indexed: 04/15/2025] Open
Abstract
Recent studies have investigated the key roles exerted by ERAP1 and ERAP2 in maintaining cellular homeostasis, emphasizing their functions beyond traditional antigen processing and presentation. In particular, genetic variants of these IFNγ-inducible aminopeptidases significantly impact critical cellular pathways, including migration, angiogenesis, and autophagy, which are essential in immune responses and disease processes. ERAP1's influence on endothelial cell migration and VEGF-driven angiogenesis, along with ERAP2's role in managing stress-induced autophagy via the UPR, highlights their importance in cellular adaptation to stress and disease outcomes, including autoimmune diseases, cancer progression, and infections. By presenting recent insights into ERAP1 and ERAP2 functions, this review underscores their potential as therapeutic targets in immune regulation and cellular stress-response pathways.
Collapse
Affiliation(s)
- Irma Saulle
- Università degli Studi di Milano, Dipartimento di Scienze Biomediche e Cliniche, Milano, Italy
- Università degli Studi di Milano, Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Milano, Italy
| | | | - Daniel D’Agate
- Università degli Studi di Milano, Dipartimento di Scienze Biomediche e Cliniche, Milano, Italy
| | - Mario Clerici
- Università degli Studi di Milano, Dipartimento di Fisiopatologia Medico-Chirurgica e dei Trapianti, Milano, Italy
- IRCCS, Fondazione Don Carlo Gnocchi, Milano, Italy
| | - Mara Biasin
- Università degli Studi di Milano, Dipartimento di Scienze Biomediche e Cliniche, Milano, Italy
| |
Collapse
|
25
|
He Z, Liu Q, Wang Y, Zhao B, Zhang L, Yang X, Wang Z. The role of endoplasmic reticulum stress in type 2 diabetes mellitus mechanisms and impact on islet function. PeerJ 2025; 13:e19192. [PMID: 40166045 PMCID: PMC11956770 DOI: 10.7717/peerj.19192] [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: 12/18/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a globally prevalent metabolic disorder characterized by insulin resistance and dysfunction of islet cells. Endoplasmic reticulum (ER) stress plays a crucial role in the pathogenesis and progression of T2DM, especially in the function and survival of β-cells. β-cells are particularly sensitive to ER stress because they require substantial insulin synthesis and secretion energy. In the early stages of T2DM, the increased demand for insulin exacerbates β-cell ER stress. Although the unfolded protein response (UPR) can temporarily alleviate this stress, prolonged or excessive stress leads to pancreatic cell dysfunction and apoptosis, resulting in insufficient insulin secretion. This review explores the mechanisms of ER stress in T2DM, particularly its impact on islet cells. We discuss how ER stress activates UPR signaling pathways to regulate protein folding and degradation, but when stress becomes excessive, these pathways may contribute to β-cell death. A deeper understanding of how ER stress impacts islet cells could lead to the development of novel T2DM treatment strategies aimed at improving islet function and slowing disease progression.
Collapse
Affiliation(s)
- Zhaxicao He
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Qian Liu
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Yan Wang
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Bing Zhao
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Lumei Zhang
- Gansu University of Chinese Medicine, Lanzhou, China
| | - Xia Yang
- Tianshui Hospital of Traditional Chinese Medicine, Tianshui, China
| | - Zhigang Wang
- Gansu University of Chinese Medicine, Lanzhou, China
- Tianshui Hospital of Traditional Chinese Medicine, Tianshui, China
| |
Collapse
|
26
|
Zarrella S, Miranda MR, Covelli V, Restivo I, Novi S, Pepe G, Tesoriere L, Rodriquez M, Bertamino A, Campiglia P, Tecce MF, Vestuto V. Endoplasmic Reticulum Stress and Its Role in Metabolic Reprogramming of Cancer. Metabolites 2025; 15:221. [PMID: 40278350 PMCID: PMC12029571 DOI: 10.3390/metabo15040221] [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: 02/17/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Endoplasmic reticulum (ER) stress occurs when ER homeostasis is disrupted, leading to the accumulation of misfolded or unfolded proteins. This condition activates the unfolded protein response (UPR), which aims to restore balance or trigger cell death if homeostasis cannot be achieved. In cancer, ER stress plays a key role due to the heightened metabolic demands of tumor cells. This review explores how metabolomics can provide insights into ER stress-related metabolic alterations and their implications for cancer therapy. Methods: A comprehensive literature review was conducted to analyze recent findings on ER stress, metabolomics, and cancer metabolism. Studies examining metabolic profiling of cancer cells under ER stress conditions were selected, with a focus on identifying potential biomarkers and therapeutic targets. Results: Metabolomic studies highlight significant shifts in lipid metabolism, protein synthesis, and oxidative stress management in response to ER stress. These metabolic alterations are crucial for tumor adaptation and survival. Additionally, targeting ER stress-related metabolic pathways has shown potential in preclinical models, suggesting new therapeutic strategies. Conclusions: Understanding the metabolic impact of ER stress in cancer provides valuable opportunities for drug development. Metabolomics-based approaches may help identify novel biomarkers and therapeutic targets, enhancing the effectiveness of antitumor therapies.
Collapse
Affiliation(s)
- Salvatore Zarrella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Verdiana Covelli
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (V.C.); (M.R.)
| | - Ignazio Restivo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (L.T.)
| | - Sara Novi
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (L.T.)
| | - Manuela Rodriquez
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (V.C.); (M.R.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Mario Felice Tecce
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| |
Collapse
|
27
|
Kumari B. Cellular Stress Responses and Associated Diseases: A Focus on Heat Shock Proteins. Cell Biochem Biophys 2025:10.1007/s12013-025-01724-3. [PMID: 40126823 DOI: 10.1007/s12013-025-01724-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/08/2025] [Indexed: 03/26/2025]
Abstract
Cellular stress response is the response of the cell at molecular level in order to combat various environmental stressors / viral infections. These stressors can be either intra or extracellular. In the beginning of the insult cell tries to recoup from these adverse events by various mechanism like heat shock protein response, unfolded protein response, mitochondrial stress signaling, DNA damage response etc. However, if these stressors exceed the cellular capacity to coup with it, it leads to programmed cell death and senescence. Also, chronic stress and cortisol released in response to cellular stress decreases telomerase activity which is needed to replenish telomeres which are protective casing at the end of a strand of DNA. Too low telomeres lead to cell death or cell become pro-inflammatory leading to aging process and other health associated risks like cardiovascular diseases neurodegenerative diseases, autoimmune diseases, cancers etc.
Collapse
Affiliation(s)
- Bandana Kumari
- Associate Professor, Department of Biochemistry, All India Institute of Medical Sciences, Patna, Bihar, India.
| |
Collapse
|
28
|
Sato S, Ogawa Y, Shimizu E, Asai K, Negishi K, Tsubota K, Hirayama M. Endoplasmic reticulum stress contributes to the development of ocular graft-vs-host disease in the eyelids and the ocular surface. Ocul Surf 2025; 37:115-131. [PMID: 40127761 DOI: 10.1016/j.jtos.2025.03.003] [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: 10/24/2024] [Revised: 02/27/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND While endoplasmic reticulum (ER) stress has been implicated in various aspects of graft-versus-host disease (GVHD), its effects on the eyelids and ocular surface in patients with chronic GVHD (cGVHD) remains poorly understood. We aimed to investigate the relationship between ER stress and ocular GVHD using the ER stress suppressor, 4-phenylbutyric acid (PBA). METHODS The study used allogeneic bone marrow transplantation (BMT) and syngeneic BMT to establish a cGVHD mouse model. cGVHD mice were treated with either intraperitoneal administration of PBA or 2 % PBA eye drops following BMT. RESULTS The Intraperitoneal PBA-treated (PBAip) group retained a larger meibomian gland (MG) area and corneal epithelial damage and inflammatory and fibrotic cell infiltration in the ocular surface was attenuated compared to vehicle-treated cGVHD mice. The expression of unfolded protein response markers was significantly elevated in the vehicle group compared to the syngeneic control and the PBAip group. Electron microscopy and immunohistochemistry revealed that fibroblasts and macrophages infiltrated the eyelids and ocular surface of cGVHD mice under ER stress. The corneal fluorescein staining score was significantly lower in the PBA eye drop-treated group than in the vehicle-treated group. The numbers of leukocyte marker CD45-, T cell marker CD4-, and macrophage marker F4/80-positive cells were significantly reduced in the PBA eye drop-treated group compared to the vehicle group. CONCLUSIONS The study suggests that the ER stress response, which is triggered by cGVHD in ocular surface tissues, can be suppressed by PBA, an ER stress suppressor, potentially offering therapeutic benefits in ocular GVHD.
Collapse
Affiliation(s)
- Shinri Sato
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Yoko Ogawa
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Eisuke Shimizu
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuki Asai
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuno Negishi
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazuo Tsubota
- Tsubota Laboratory, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Masatoshi Hirayama
- Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| |
Collapse
|
29
|
Hou L, Yang X, Liu C, Yu J, Wu Z, Wang Y, Zeng P, Guo J, Shi Y, Zhou J, Liu J. Seneca Valley virus induces mitochondrial apoptosis by activating ER stress or the PERK pathway based on Ca 2+ transfer from ER to mitochondria. J Virol 2025; 99:e0217724. [PMID: 39912666 PMCID: PMC11915807 DOI: 10.1128/jvi.02177-24] [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: 12/11/2024] [Accepted: 01/18/2025] [Indexed: 02/07/2025] Open
Abstract
Seneca Valley virus (SVV), also known as Senecavirus A, a porcine pathogen that causes vesicular diseases, is prevalent in pig herds worldwide. SVV infection induces endoplasmic reticulum (ER) stress in PK-15 and BHK-21 cells, accompanied by activation of the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) and activating transcription factor 6 (ATF6) pathways, which in turn facilitates SVV replication. ER stress is associated with the regulation of Ca2+ homeostasis and mitochondrial apoptosis. However, the precise role of Ca2+ in SVV-induced apoptosis remains unclear. In this study, western blotting, flow cytometry, and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling (TUNEL) detection revealed that either ER stress or the PERK pathway is involved in the apoptosis of SVV-infected cells treated with specific inhibitors. Furthermore, SVV-mediated ER stress markedly contributed to the transfer of Ca2+ from the ER to mitochondria. The subsequent increase in mitochondrial Ca2+ content was accompanied by an increased number of ER membranes near the mitochondria. Finally, the inhibition of mitochondrial Ca2+ overload, ER stress, and the PERK pathway substantially attenuated SVV-mediated mitochondrial dysfunction, as evidenced by analyzing mitochondrial membrane potential (MMP), mitochondrial permeability transition poremPTP, reactive oxygen speciesROS, and adenosine 5'-triphosphate ATP, and the levels of mitochondrial apoptosis. These findings demonstrate that SVV induces mitochondrial apoptosis, which is dependent on ER stress-mediated transmission of Ca2+ from the ER to the mitochondria. IMPORTANCE Viruses have developed multiple mechanisms to facilitate their proliferation or persistence through manipulating various organelles in cells. Seneca Valley virus (SVV), as a novel emerging pathogen associated with vesicular disease, is clinically and economically important infections that affect farm animals. Previously, we had confirmed that SVV-induced endoplasmic reticulum (ER) stress benefited for viral replication. Ca2+, as an intracellular signaling messenger mainly stored in the ER, is regulated by ER stress and then involved in apoptosis. However, the precise mechanism that Ca2+ transfer induced by SVV infection triggered apoptosis remained unclear. Here, we found that SVV infection triggered the Ca2+ transform from ER to mitochondria, resulting in mitochondrial dysfunction, and finally induced mitochondrial apoptosis. Our study shed light on a novel mechanism revealing how ER stress manipulates Ca2+ homeostasis to induce mitochondrial apoptosis and regulate viral proliferation.
Collapse
Affiliation(s)
- Lei Hou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Xiaoyu Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Changzhe Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Ju Yu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zhi Wu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yong Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Penghui Zeng
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jinshuo Guo
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yongyan Shi
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jianwei Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Jue Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| |
Collapse
|
30
|
Stilkerich A, Schicht G, Seidemann L, Hänsel R, Friebel A, Hoehme S, Seehofer D, Damm G. Cell Homeostasis or Cell Death-The Balancing Act Between Autophagy and Apoptosis Caused by Steatosis-Induced Endoplasmic Reticulum (ER) Stress. Cells 2025; 14:449. [PMID: 40136698 PMCID: PMC11941029 DOI: 10.3390/cells14060449] [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: 12/20/2024] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic-dysfunction-associated steatotic liver disease (MASLD) is a prevalent liver condition with potential progression to cirrhosis and impaired regeneration post-resection. A key mechanism underlying lipotoxicity is endoplasmic reticulum (ER) stress, particularly the activation of the unfolded protein response (UPR). This study investigates the interplay between lipid accumulation, endoplasmic reticulum (ER) stress, and cellular outcomes, focusing on the balance between autophagy and apoptosis. We cultured primary human hepatocytes (PHH) in a free fatty acid (FFA)-enriched medium for 120 h, assessing lipid accumulation, metabolism, and the expression of selected UPR markers. Additionally, we investigated the effects of lipid load on cell activity and growth in proliferating HepG2 cells. We observed that FFA uptake consistently induced ER stress, shifting cellular responses toward apoptosis under high lipid loads. Donor-specific differences were evident, particularly in lipid storage, excretion, and sensitivity to lipotoxicity. Some donors exhibited limited triglyceride (TAG) storage and excretion, leading to an excess of FFA whose metabolic fate remains unclear. Proliferation was more sensitive to lipid accumulation than overall cell activity, with even low FFA concentrations impairing growth, highlighting the vulnerability of regenerative processes to steatosis. The study elucidates how ER stress pathways, such as PERK-CHOP and IRE1α-JNK, are differentially activated in response to lipid overload, tipping the balance toward apoptosis in severe cases. The limited activation of repair mechanisms, such as autophagy, further emphasizes the critical role of ER stress in determining hepatocyte fate. The donor-dependent variability highlights the need for personalized strategies to mitigate lipotoxic effects and enhance liver regeneration in steatosis-related conditions.
Collapse
Affiliation(s)
- Anna Stilkerich
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.S.); (G.S.); (L.S.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany; (R.H.); (A.F.); (S.H.)
| | - Gerda Schicht
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.S.); (G.S.); (L.S.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany; (R.H.); (A.F.); (S.H.)
| | - Lena Seidemann
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.S.); (G.S.); (L.S.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany; (R.H.); (A.F.); (S.H.)
| | - René Hänsel
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany; (R.H.); (A.F.); (S.H.)
| | - Adrian Friebel
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany; (R.H.); (A.F.); (S.H.)
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, Haertelstraße 16-18, 04107 Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), 04105 Leipzig, Germany
| | - Stefan Hoehme
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany; (R.H.); (A.F.); (S.H.)
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, Haertelstraße 16-18, 04107 Leipzig, Germany
- Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), 04105 Leipzig, Germany
| | - Daniel Seehofer
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.S.); (G.S.); (L.S.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany; (R.H.); (A.F.); (S.H.)
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, 04103 Leipzig, Germany; (A.S.); (G.S.); (L.S.); (D.S.)
- Saxonian Incubator for Clinical Translation (SIKT), Leipzig University, 04103 Leipzig, Germany; (R.H.); (A.F.); (S.H.)
| |
Collapse
|
31
|
Tsai CY, Lee CY, Chen JH, Chiang CK. Chronic Antibody-Mediated Rejection and Plasma Cell ER Stress: Opportunities and Challenges with Calcineurin Inhibitors. Int J Mol Sci 2025; 26:2711. [PMID: 40141353 PMCID: PMC11943340 DOI: 10.3390/ijms26062711] [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: 12/03/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Chronic alloantibody-mediated rejection (cAMR) remains a major challenge in transplant immunology, with no FDA-approved targeted therapies currently available. Despite advancements in cellular immunosuppression, effective strategies to mitigate alloantibody-mediated rejection are still lacking. This review provides a comprehensive overview of transplant rejection with a particular focus on the pathophysiology and therapeutic landscape of cAMR. We highlight the role of plasma cell-driven alloantibody production and its susceptibility to endoplasmic reticulum (ER) stress, a pathway with potential for therapeutic intervention. Special attention is given to calcineurin inhibitors (CNIs), which, beyond their well-established T-cell inhibitory effects, exhibit differential impacts on ER stress and plasma cell viability. By delineating the mechanistic differences between cyclosporine and tacrolimus in regulating ER stress responses, we propose potential therapeutic implications for optimizing cAMR management. This review underscores the need for innovative strategies targeting plasma cell biology to improve long-term transplant outcomes.
Collapse
Affiliation(s)
- Ching-Yi Tsai
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100233, Taiwan; (C.-Y.T.); (J.-H.C.)
- Department of Medical Research, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Chih-Yuan Lee
- Department of Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan;
- Organ Transplant Center, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Jia-Huang Chen
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100233, Taiwan; (C.-Y.T.); (J.-H.C.)
| | - Chih-Kang Chiang
- Graduate Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100233, Taiwan; (C.-Y.T.); (J.-H.C.)
- Department of Integrated Diagnostics & Therapeutics, National Taiwan University Hospital, Taipei 100225, Taiwan
| |
Collapse
|
32
|
Ventura ALM, Silva TM, França GR. Cannabinoids Activate Endoplasmic Reticulum Stress Response and Promote the Death of Avian Retinal Müller Cells in Culture. Brain Sci 2025; 15:291. [PMID: 40149812 PMCID: PMC11940308 DOI: 10.3390/brainsci15030291] [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: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Activation of cannabinoid CB1 or CB2 receptors induces the death of glial progenitors from the chick retina in culture. Here, by using an enriched retinal glial cell culture, we characterized some mechanisms underlying glial death promoted by cannabinoids. METHODS AND RESULTS Retinal cultures obtained from 8-day-old (E8) chick embryos and maintained for 12-15 days (C12-15) were used. MTT assays revealed that the CB1/CB2 agonist WIN 55,212-2 (WIN) decreased cell viability in the cultures in a time-dependent manner, with a concomitant increase in extracellular LDH activity, suggesting membrane integrity loss. Cell death was also dose-dependently induced by cannabidiol (CBD), Δ9-tetrahydrocannabinol (THC), and CP55940, another CB1/CB2 agonist. In contrast to WIN-induced cell death that was not blocked by either antagonist, the deleterious effect of CBD was blocked by the CB2 receptor antagonist SR144528, but not by PF514273, a CB1 receptor antagonist. WIN-treated cultures showed glial cells with large vacuoles in cytoplasm that were absent in cultures incubated with WIN plus 4-phenyl-butyrate (PBA), a chemical chaperone. Since cannabinoids induced the phosphorylation of eukaryotic initiation factor 2-alfa (eIF2α), these results suggest a process of endoplasmic reticulum (ER) swelling and stress. Incubation of the cultures with WIN for 4 h induced a ~five-fold increase in the number of cells labeled with the ROS indicator CM-H2DCFDA. WIN induced the phosphorylation of JNK but not of p38 in the cultures, and also induced an increase in the number of glial cells expressing cleaved-caspase 3 (c-CASP3). The decrease in cell viability and the expression of c-CASP3 was blocked by salubrinal, an inhibitor of eIF2α dephosphorylation. CONCLUSIONS These data suggest that cannabinoids induce the apoptosis of glial cells in culture by promoting ROS production, ER stress, JNK phosphorylation, and caspase-3 processing. The graphical abstract was created at Biorender.com.
Collapse
Affiliation(s)
- Ana Lúcia Marques Ventura
- Neuroscience Program, Department of Neurobiology, Federal Fluminense University, Niterói CEP 24210-201, RJ, Brazil;
| | - Thayane Martins Silva
- Neuroscience Program, Department of Neurobiology, Federal Fluminense University, Niterói CEP 24210-201, RJ, Brazil;
| | - Guilherme Rapozeiro França
- Department of Physiological Sciences, Federal University of the State of Rio de Janeiro, Rio de Janeiro CEP 20211-040, RJ, Brazil;
| |
Collapse
|
33
|
Li Y, Li X, Wu Q, Puig M, Moulin F, Choudhuri S, Gingrich J, Guo L, Chen S. 7-Hydroxycannabidiol and 7-carboxycannabidiol induced cytotoxicity via apoptosis and endoplasmic reticulum stress in human hepatic cells. Arch Toxicol 2025:10.1007/s00204-025-04001-7. [PMID: 40029368 DOI: 10.1007/s00204-025-04001-7] [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: 01/17/2025] [Accepted: 02/19/2025] [Indexed: 03/05/2025]
Abstract
Cannabidiol (CBD), a major component of extract from the plant Cannabis sativa L., has demonstrated efficacy in treating childhood-onset epilepsy; however, animal studies and clinical trials have reported elevated liver enzymes after CBD use, suggesting potential liver toxicity. In a previous study, we demonstrated that CBD caused cytotoxicity with apoptosis and endoplasmic reticulum (ER) stress in human hepatic cells. In the present study, we investigated the toxicity profile of the two main metabolites of CBD, 7-hydroxy-CBD and 7-carboxy-CBD, in primary human hepatocytes and HepG2 cells. Our findings indicated that both metabolites induced cellular damage similar to the parent drug in these cells. 7-Hydroxy-CBD and 7-carboxy-CBD also caused cell cycle disturbances, apoptosis, and ER stress in HepG2 cells. Additionally, we explored the role of cytochrome P450 (CYP) in the metabolism of 7-hydroxy-CBD and 7-carboxy-CBD using HepG2 cell lines expressing 14 individual CYPs. We determined that 7-hydroxy-CBD is metabolized by CYP2D6, and CYP2D6-mediated metabolism attenuated the cytotoxicity, apoptosis, and ER stress induced by 7-hydroxy-CBD. The CYPs did not metabolize 7-carboxy-CBD. In summary, our findings highlight the mechanisms underlying cytotoxicity induced by 7-hydroxy-CBD and 7-carboxy-CBD in hepatic cells.
Collapse
Affiliation(s)
- Yuxi Li
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Xilin Li
- Division of Genetic and Molecular Toxicology, NCTR, U.S. FDA, Jefferson, AR, 72079, USA
| | - Qiangen Wu
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Montserrat Puig
- Division of Biotechnology Review and Research III, Office of Biotechnology Products, Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, 20993, USA
| | - Frederic Moulin
- Division of Hepatology and Nutrition, Office of New Drugs, Center for Drug Evaluation and Research, U.S. FDA, Silver Spring, MD, 20993, USA
| | - Supratim Choudhuri
- Division of Food Ingredients, Office of Pre-Market and Additive Safety, Office of Food Chemical Safety, Dietary Supplements, and Innovation, Human Foods Program, U.S. FDA, College Park, MD, 20740, USA
| | - Jeremy Gingrich
- Division of Food Ingredients, Office of Pre-Market and Additive Safety, Office of Food Chemical Safety, Dietary Supplements, and Innovation, Human Foods Program, U.S. FDA, College Park, MD, 20740, USA
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Si Chen
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), 3900 NCTR Road, Jefferson, AR, 72079, USA.
| |
Collapse
|
34
|
Nghiem THT, Nguyen KA, Kusuma F, Park S, Park J, Joe Y, Han J, Chung HT. The PERK-eIF2α-ATF4 Axis Is Involved in Mediating ER-Stress-Induced Ferroptosis via DDIT4-mTORC1 Inhibition and Acetaminophen-Induced Hepatotoxicity. Antioxidants (Basel) 2025; 14:307. [PMID: 40227255 PMCID: PMC11939615 DOI: 10.3390/antiox14030307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 04/15/2025] Open
Abstract
Ferroptosis, a regulated form of cell death characterized by lipid peroxidation and iron accumulation, is increasingly recognized for its role in disease pathogenesis. The unfolded protein response (UPR) has been implicated in both endoplasmic reticulum (ER) stress and ferroptosis-mediated cell fate decisions; yet, the specific mechanism remains poorly understood. In this study, we demonstrated that ER stress induced by tunicamycin and ferroptosis triggered by erastin both activate the UPR, leading to the induction of ferroptotic cell death. This cell death was mitigated by the application of chemical chaperones and a ferroptosis inhibitor. Among the three arms of the UPR, the PERK-eIF2α-ATF4 signaling axis was identified as a crucial mediator in this process. Mechanistically, the ATF4-driven induction of DDIT4 plays a pivotal role, facilitating ferroptosis via the inhibition of the mTORC1 pathway. Furthermore, acetaminophen (APAP)-induced hepatotoxicity was investigated as a model of eIF2α-ATF4-mediated ferroptosis. Our findings reveal that the inhibition of eIF2α-ATF4 or ferroptosis protects against APAP-induced liver damage, underscoring the therapeutic potential of targeting these pathways. Overall, this study not only clarifies the intricate role of the PERK-eIF2α-ATF4 axis in ER-stress-and erastin-induced ferroptosis but also extends these findings to a clinically relevant model, providing a foundation for potential therapeutic interventions in conditions characterized by dysregulated ferroptosis and ER stress.
Collapse
Affiliation(s)
- Thu-Hang Thi Nghiem
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea;
| | - Kim Anh Nguyen
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea; (K.A.N.); (F.K.); (S.P.)
| | - Fedho Kusuma
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea; (K.A.N.); (F.K.); (S.P.)
| | - Soyoung Park
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea; (K.A.N.); (F.K.); (S.P.)
| | - Jeongmin Park
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea; (J.P.); (Y.J.)
| | - Yeonsoo Joe
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea; (J.P.); (Y.J.)
| | - Jaeseok Han
- Department of Integrated Biomedical Science, Soonchunhyang University, Cheonan 31151, Republic of Korea; (K.A.N.); (F.K.); (S.P.)
- Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Republic of Korea
| | - Hun Taeg Chung
- College of Korean Medicine, Daegu Haany University, Gyeongsan 38610, Republic of Korea; (J.P.); (Y.J.)
| |
Collapse
|
35
|
Roy PK, Deepak K, Das CK, Das A, Biswas A, Jena BC, Mandal M. PSMC2 promotes resistance against temozolomide in glioblastoma via suppressing JNK-mediated autophagic cell death. Biochem Pharmacol 2025; 233:116755. [PMID: 39824465 DOI: 10.1016/j.bcp.2025.116755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Temozolomide is universally used to treat glioblastoma due to its unique ability to cross the blood-brain barrier and inhibit tumor growth through DNA alkylation. However, over time, the inevitable emergence of resistance to temozolomide impedes successful treatment of this cancer. As a result, there is an urgent need to identify new therapeutic targets to improve treatment outcomes for this malignancy. In this work, acquired temozolomide-resistant glioblastoma cell lines LN18 (LN18-TR) and T98G (T98G-TR) exhibited stronger aggressiveness and lower endoplasmic reticulum (ER) stress than their parental cells.. Besides, temozolomide resistance was associated with elevated proteasome activity that suppressed ER stress, which was restored upon inhibition of the proteasome with MG132. Specifically, our study revealed that the 19S proteasomal regulatory subunit PSMC2, which was overexpressed in adapted temozolomide-resistant glioblastoma cells, reduced pro-death autophagy and decreased temozolomide sensitivity in parental cells when overexpressed. While autophagy increased in parental cells following temozolomide treatment, it was not elevated in temozolomide-resistant glioblastoma cells. Genetic suppression of PSMC2 triggered the JNK signalling pathway causing phosphorylation of BCL2, allowing Beclin1 to be released from the BCL2-Beclin1 complex. This boosted autophagosome nucleation, increased pro-death autophagy, and restored apoptosis in temozolomide-resistant glioblastoma cells. Finally, targeting PSMC2 provided a unique method for interrupting autophagy-mediated ER stress maintenance and temozolomide resistance in glioblastoma.
Collapse
Affiliation(s)
- Pritam Kumar Roy
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - K Deepak
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Chandan Kanta Das
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, 421 Curie Boulevard, BRBII/III, Philadelphia, PA, 19104, USA
| | - Abhijit Das
- School of Bioscience, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Angana Biswas
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Bikash Chandra Jena
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47907, USA
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur, India.
| |
Collapse
|
36
|
Sugiura S, Yoshida H, Sugiura H, Uehara M, Sugiura Y, Maruo Y, Hayashi Y, Yamamoto T, Kato T, Fujimoto N, Udagawa J. Increased intracellular stress responses and decreased KLF2 in adult patients with atopic dermatitis. Cell Stress Chaperones 2025; 30:84-99. [PMID: 39938773 PMCID: PMC11891603 DOI: 10.1016/j.cstres.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 01/23/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025] Open
Abstract
Atopic dermatitis (AD) is prone to exacerbations in response to various triggering factors and flare-ups after remission. We searched for molecules associated with relapse/exacerbation of AD among molecules with altered gene expression in the skin of patients with AD. Microarray analyses were performed on lesional and nonlesional skin of adolescent or adult patients with recalcitrant AD and healthy controls. Five chaperones involved in intracellular stress responses, namely heat shock protein family A (Hsp70) member 9 (HSPA9), heat shock protein 90 beta family member 1 (HSP90B1), calnexin (CANX), malectin (MLEC; endoplasmic reticulum-associated degradation), and heat shock protein family D (Hsp60) member 1 (HSPD1), were consistently upregulated in involved and uninvolved skin of patients with AD. Damage-associated molecular patterns were upregulated in involved skin. KLF transcription factor 2 (KLF2) was decreased in involved skin and exhibited a decreasing trend in uninvolved skin of patients with AD. CD4(+)/CD8(+) double-positive cells (1.4% of T cells) were detected in lesions with declined KLF2 levels. WNT inhibitory factor 1 (WIF1) was downregulated in involved skin. Prolactin-induced protein was upregulated in only uninvolved skin of patients with AD. We found increased intracellular stress responses and decreased expression of KLF2 in the skin of patients with AD. Multifactorial genetic diseases, such as asthma, inflammatory bowel disease, type 2 diabetes, and rheumatoid arthritis, are associated with intracellular stress. Intracellular abnormalities may also be responsible for AD. Further research on AD may incorporate enhanced intracellular stress response and the decreased expression of KLF2 into the mechanism underlying AD.
Collapse
Affiliation(s)
- Shuji Sugiura
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan; Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan.
| | - Hiderou Yoshida
- Department of Molecular Biochemistry, Graduate School of Life Science, University of Hyogo, Ako, Japan
| | - Hisashi Sugiura
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan; Department of Dermatology, Sugiura Dermatology Clinic, Kusatsu, Japan
| | - Masami Uehara
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Yasuo Sugiura
- International Health Care Center, National Center for Global Health and Medicine, Tokyo, Japan; Department of Pediatrics, Navitas Clinic, Tokyo, Japan
| | - Yoshihiro Maruo
- Department of Pediatrics, Shiga University of Medical Science, Otsu, Japan
| | - Yuji Hayashi
- Hospital Division of Diagnostic Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Takefumi Yamamoto
- Central Research Laboratory, Shiga University of Medical Science, Otsu, Japan
| | - Takeshi Kato
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Noriki Fujimoto
- Department of Dermatology, Shiga University of Medical Science, Otsu, Japan
| | - Jun Udagawa
- Division of Anatomy and Cell Biology, Department of Anatomy, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
37
|
Xu X, Ge C, Wang S, Gao L, Wang C, Dai F, Wang Y, Xie S. Polyamine-modified naphthalimide derivative 9C inhibits colorectal cancer through ROS-mediated ER stress, migration and invasion. Toxicol In Vitro 2025; 103:105974. [PMID: 39586364 DOI: 10.1016/j.tiv.2024.105974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
Mounting evidence over the past decades has demonstrated the therapeutic potential of targeting endoplasmic reticulum (ER) stress signaling in cancer. Naphthalimdes exert their anti-cancer activities in a variety of ways. However, the effects of naphthalimides on ER stress are rarely reported. In this study, based on RNA-sequencing analysis, we observed that 9C, a naphthalimide derivative, could trigger ER stress to activate death receptor signaling and autophagy. Pretreatment of ER stress inhibitor, such as salubrinal, and autophagy inhibitor, such as 3-methyladenine (3-MA), partially reversed 9C-induced inhibition of cell growth. Furthermore, our results unveiled a reactive oxygen species (ROS)-dependent inhibitory effect of 9C. In addition, 9C inhibited colorectal cancer (CRC) cells migration and invasion. Removal of ROS using N-acetyl-L-cysteine (NAC) attenuated the expression of ATF4, CHOP, death receptors, E-cadherin, and the apoptosis and autophagy related proteins. Taken together, our results suggested that ROS-mediated ER stress, migration, and invasion is responsible for the therapeutic potential of naphthalimides including 9C in CRC.
Collapse
Affiliation(s)
- Xiaojuan Xu
- School of Pharmacy, Henan University, Kaifeng 475004, Henan, China
| | - Chaochao Ge
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, Henan, China; School of Pharmacy, Heze University, Heze 274015, Shandong, China
| | - Senzhen Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, Henan, China; School of Life Sciences, Henan University, Kaifeng 475004, Henan, China
| | - Lei Gao
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, Henan, China
| | - Chaojie Wang
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, Henan, China
| | - Fujun Dai
- Henan Key Laboratory of Natural Medicine Innovation and Transformation, Henan University, Kaifeng 475004, Henan, China.
| | - Yuxia Wang
- College of Chemistry and Chemical Engineering, Henan University, Kaifeng 475004, Henan, China.
| | - Songqiang Xie
- School of Pharmacy, Henan University, Kaifeng 475004, Henan, China.
| |
Collapse
|
38
|
Yin M, Zheng X, Shi L. Targeting p38 MAPK: A potential bridge between ER stress and age-related bone loss. Cell Signal 2025; 127:111549. [PMID: 39638139 DOI: 10.1016/j.cellsig.2024.111549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/21/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
The endoplasmic reticulum (ER) is crucial in the development of numerous age-related bone disorders. Notably, ER stress can precipitate bone loss by orchestrating inflammatory responses, apoptosis, and autophagy through the activation of the p38 MAPK pathway. Age-related bone loss diseases pose a significant burden on society and healthcare as the global population ages. This review provides a comprehensive analysis of recent research advancements, delving into the critical role of ER stress-activated p38 MAPK in inflammation, apoptosis, and autophagy, as well as its impact on bone formation and bone resorption. This review elucidates the molecular mechanisms underlying the involvement of ER stress-activated p38 MAPK in osteoporosis, rheumatoid arthritis, periodontitis, and osteoarthritis and discusses the therapeutic potential of targeting p38 MAPK. Furthermore, this review provides a scientific foundation for new therapeutic strategies by highlighting prospective research directions.
Collapse
Affiliation(s)
- Meng Yin
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China; Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xin Zheng
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Shi
- Neck-Shoulder and Lumbocrural Pain Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
39
|
Jeong S, Doo M, Sung K, Kim YJ, Lee JH, Ha JH. Aruncus Dioicus Var. Kamtschaticus Extract Prevents Ocular Endoplasmic Reticulum Stress, Inflammation, and Oxidative Stress In Vitro. J Med Food 2025; 28:281-293. [PMID: 39973273 DOI: 10.1089/jmf.2024.k.0240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025] Open
Abstract
The aim of this study was to determine the anti-inflammatory and anti-endoplasmic reticulum (ER) stress effects of Aruncus dioicus var. kamtschaticus (ADK) extract on ARPE-19 cells. Pretreatment with ADK effectively mitigated thapsigargin (Tg)-induced increases in vascular endothelial growth factor protein secretion and intracellular calcium levels. Furthermore, pretreatment with ADK suppressed ocular ER stress-related protein expression in a dose-dependent manner, inhibited the loss of tight junctions, and suppressed interleukin-6 gene expression. Moreover, ADK pretreatment significantly prevented lipopolysaccharide-inducible proinflammatory cytokine gene expression at the transcription level and the phosphorylation of proteins involved in the mitogen-activated protein kinase-nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) axis at the posttranslational level. Additionally, ADK extract enhanced antioxidant activity, as evidenced by increased heme oxygenase-1 protein expression and increased 2,2-diphenyl-1-picrylhydrazyl radical scavenging and ferric-reducing antioxidant power. In conclusion, ADK extract effectively protected ARPE-19 cells from ocular ER stress, inflammation, and oxidative stress, demonstrating its potential as a nutraceutical intervention for ocular diseases.
Collapse
Affiliation(s)
- Sunyoung Jeong
- Bioanalytical and Pharmacokinetic Research Group, Korea Institute of Toxicology, Daejeon, Korea
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, Korea
| | - Miae Doo
- Department of Food and Nutrition, Kunsan National University, Gunsan, Korea
| | - Kihun Sung
- Department of Food Science and Nutrition, Dankook University, Cheonan, Korea
| | - Young Jun Kim
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| | - Jong-Hwa Lee
- Bioanalytical and Pharmacokinetic Research Group, Korea Institute of Toxicology, Daejeon, Korea
- Department of Human and Environmental Toxicology, University of Science and Technology, Daejeon, Korea
| | - Jung-Heun Ha
- Department of Food Science and Nutrition, Dankook University, Cheonan, Korea
- Research Center for Industrialization of Natural Neutralization, Dankook University, Yongin, Korea
| |
Collapse
|
40
|
Huang C, Qu QR, Hoque MT, Bendayan R. Dolutegravir induces endoplasmic reticulum stress at the blood-brain barrier. FASEB J 2025; 39:e70377. [PMID: 39985305 PMCID: PMC11846018 DOI: 10.1096/fj.202402677rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 01/17/2025] [Accepted: 01/27/2025] [Indexed: 02/24/2025]
Abstract
Dolutegravir (DTG)-based antiretroviral therapy is the contemporary first-line therapy to treat HIV infection. Despite its efficacy, mounting evidence has suggested a higher risk of neuropsychiatric adverse effect (NPAE) associated with DTG use, with a limited understanding of the underlying mechanisms. Our laboratory has previously reported a toxic effect of DTG but not bictegravir (BTG) in disrupting the blood-brain barrier (BBB) integrity. The current study aimed to investigate the underlying mechanism of DTG toxicity. Primary cultures of mouse brain microvascular endothelial cells were treated with DTG and BTG at therapeutically relevant concentrations. RNA sequencing, qPCR, western blot analysis, and cell stress assays (Ca2+ flux, H2DCFDA, TMRE, MTT) were applied to assess the results. The gene ontology (GO) analysis revealed an enriched transcriptome signature of endoplasmic reticulum (ER) stress following DTG treatment. We demonstrated that therapeutic concentrations of DTG but not BTG activated the ER stress sensor proteins (PERK, IRE1, p-IRE1) and downstream ER stress markers (eIF2α, p-eIF2α, Hspa5, Atf4, Ddit3, Ppp1r15a, Xbp1, spliced-Xbp1). In addition, DTG treatment resulted in a transient Ca2+ flux, an aberrant mitochondrial membrane potential, and a significant increase in reactive oxygen species in treated cells. Furthermore, we found that prior treatment with ER sensor or ER stress inhibitors significantly mitigated the DTG-induced downregulation of tight junction proteins (Zo-1, Ocln, Cldn5) and elevation of pro-inflammatory cytokines and chemokines (Il6, Il23a, Il12b, Cxcl1, Cxcl2). The current study provides valuable insights into DTG-mediated cellular toxicity mechanisms, which may serve as a potential explanation for DTG-associated NPAEs in the clinic.
Collapse
Affiliation(s)
- Chang Huang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of PharmacyUniversity of TorontoTorontoOntarioCanada
| | - Qing Rui Qu
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of PharmacyUniversity of TorontoTorontoOntarioCanada
| | - Md. Tozammel Hoque
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of PharmacyUniversity of TorontoTorontoOntarioCanada
| | - Reina Bendayan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of PharmacyUniversity of TorontoTorontoOntarioCanada
| |
Collapse
|
41
|
Wu JY, Han B, Yang T, Zheng L, Guo YX, Li JY, Guo XY, Yin HH, Xie RJ. CHOP aggravates hepatocyte apoptosis upon endoplasmic reticulum stress by downregulating autophagy. Cell Stress Chaperones 2025; 30:109-118. [PMID: 40023477 PMCID: PMC11968278 DOI: 10.1016/j.cstres.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/24/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
Endoplasmic reticulum (ER) stress-induced apoptosis plays a crucial role in various liver diseases. Hepatocytes respond to ER stress by activating the unfolded protein response and autophagy, which is essential for maintaining ER homeostasis. However, failure to restore ER balance via autophagy contributes to apoptosis. In this study, we aimed to explore the role of C/EBP homologous protein (CHOP) in regulating ER stress-induced apoptosis in rat hepatocytes. We found that CHOP downregulates autophagy, aggravating apoptosis. Our results revealed that inhibition of CHOP expression enhanced autophagy and reduced DTT-induced apoptosis in BRL-3A cells, whereas CHOP overexpression worsened apoptosis. Chromatin immunoprecipitation assays revealed that CHOP negatively regulates autophagy-related genes, such as ATG12, ATG5, and LC3. These findings suggest that CHOP modulation plays a crucial role in ER stress-induced hepatocyte apoptosis by regulating autophagy.
Collapse
Affiliation(s)
- Jia-Yu Wu
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Bing Han
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Ting Yang
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Lu Zheng
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Yi-Xin Guo
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Jia-Yao Li
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Xiao-Yu Guo
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Huan-Huan Yin
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Ru-Jia Xie
- Department of Pathophysiology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou Province, China.
| |
Collapse
|
42
|
Rodrigues BL, Pascoal LB, Genaro LM, Warrak LSCA, Rodrigues BAG, Coope A, Camargo MG, Oliveira PDSP, Ayrizono MDLS, Velloso LA, Leal RF. In Vitro Inhibition of Endoplasmic Reticulum Stress: A Promising Therapeutic Strategy for Patients with Crohn's Disease. Cells 2025; 14:270. [PMID: 39996742 PMCID: PMC11853800 DOI: 10.3390/cells14040270] [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: 12/18/2024] [Revised: 01/04/2025] [Accepted: 01/07/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Crohn's disease (CD) is an inflammatory bowel disease marked by an abnormal immune response and excessive pro-inflammatory cytokines, leading to impaired protein processing and endoplasmic reticulum (ER) stress. This stress, caused by the accumulation of misfolded proteins, triggers the unfolded protein response (UPR) through IRE1/Xbp-1, PERK/eIF2α, and ATF6 pathways, which are linked to intestinal inflammation. This study aimed to investigate ER stress in CD patients' intestinal mucosa and evaluate phenylbutyrate (PBA) as an ER stress inhibitor. METHODS Colon biopsies from CD patients and controls were cultured under five conditions, including 4-PBA treatments. Real-time PCR, cytokine level, and immunohistochemistry were performed. RESULTS Immunohistochemistry revealed that ER stress was activated in CD patients' intestinal epithelial cells and lamina propria cells. PERK/eIF2α, but not IRE1/Xbp-1 or ATF6, was upregulated in CD patients compared to controls. UPR-related genes (STC2, CALR, HSPA5, HSP90B1) were also elevated in CD patients. PBA treatment significantly reduced ER stress and UPR markers while decreasing apoptotic markers like DDIT3. Pro-inflammatory cytokines, such as IL-1β, IL-6, IL-17, TNF- α, and sCD40L, were significantly reduced after PBA treatment. CONCLUSION ER stress and UPR pathways are activated in CD colonic mucosa, and PBA reduces these markers, suggesting potential therapeutic benefits for CD-related inflammation.
Collapse
Affiliation(s)
- Bruno Lima Rodrigues
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Lívia Bitencourt Pascoal
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Lívia Moreira Genaro
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Leonardo Saint Clair Assad Warrak
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Beatriz Alves Guerra Rodrigues
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Andressa Coope
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Michel Gardere Camargo
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Priscilla de Sene Portel Oliveira
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Maria de Lourdes Setsuko Ayrizono
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| | - Lício Augusto Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-864, Brazil
| | - Raquel Franco Leal
- Inflammatory Bowel Disease Research Laboratory, Colorectal Surgery Unit, Gastrocenter, School of Medical Sciences, University of Campinas (Unicamp), São Paulo 13083-878, Brazil
| |
Collapse
|
43
|
Ammons DT, Harris RA, Chow L, Dow S. Characterization of canine tumor-infiltrating leukocyte transcriptomic signatures reveals conserved expression patterns with human osteosarcoma. Cancer Immunol Immunother 2025; 74:105. [PMID: 39932553 PMCID: PMC11813853 DOI: 10.1007/s00262-025-03950-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 01/20/2025] [Indexed: 02/14/2025]
Abstract
Immune cells play key roles in host responses to malignant tumors. The selective pressure that immune cells elicit on tumors promotes immune escape, while tumor-associated modulation of immune cells creates an environment favorable to tumor growth and progression. In this study we used publicly available single-cell RNA sequencing (scRNA-seq) data from the translationally relevant canine osteosarcoma (OS) model to compare tumor-infiltrating immune cells to circulating leukocytes. Through computational analysis we investigated the differences in cell type proportions and how the OS TME impacted infiltrating immune cell transcriptomic profiles relative to circulating leukocytes. Differential abundance analysis revealed increased proportions of follicular helper T cells, regulatory T cells, and mature regulatory dendritic cells (mregDCs) in the OS TME. Differential gene expression analysis identified exhaustion markers (LAG3, HAVCR2, PDCD1) to be upregulated in CD4 and CD8 T cells within the OS TME. Comparisons of B cell gene expression profiles revealed an enrichment of protein processing and endoplasmic reticulum pathways, suggesting infiltrating B cells were activated following tumor infiltration. Gene expression changes within myeloid cells identified increased expression of immune suppressive molecules (CD274, OSM, MSR1) in the OS TME, indicating the TME skews myeloid cells toward an immunosuppressive phenotype. Comparisons to human literature and analysis of human scRNA-seq data revealed conserved transcriptomic responses to tumor infiltration, while also identifying species differences. Overall, the analysis presented here provides new insights into how the OS TME impacts the transcriptional programs of major immune cell populations in dogs and acts as a resource for comparative immuno-oncology research.
Collapse
Affiliation(s)
- Dylan T Ammons
- Flint Animal Cancer Center, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
| | - R Adam Harris
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Lyndah Chow
- Flint Animal Cancer Center, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Steven Dow
- Flint Animal Cancer Center, Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
- Department of Microbiology, Immunology and Pathology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, 80523, USA.
| |
Collapse
|
44
|
Mazzolini L, Touriol C. PERK-Olating Through Cancer: A Brew of Cellular Decisions. Biomolecules 2025; 15:248. [PMID: 40001551 PMCID: PMC11852789 DOI: 10.3390/biom15020248] [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: 12/20/2024] [Revised: 01/24/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
The type I protein kinase PERK is an endoplasmic reticulum (ER) transmembrane protein that plays a multifaceted role in cancer development and progression, influencing tumor growth, metastasis, and cellular stress responses. The activation of PERK represents one of the three signaling pathways induced during the unfolded protein response (UPR), which is triggered, in particular, in tumor cells that constitutively experience various intracellular and extracellular stresses that impair protein folding within the ER. PERK activation can lead to both pro-survival and proapoptotic outcomes, depending on the cellular context and the extent of ER stress. It helps the reprogramming of the gene expression in cancer cells, thereby ensuring survival in the face of oncogenic stress, such as replicative stress and DNA damage, and also microenvironmental challenges, including hypoxia, angiogenesis, and metastasis. Consequently, PERK contributes to tumor initiation, transformation, adaptation to the microenvironment, and chemoresistance. However, sustained PERK activation in cells can also impair cell proliferation and promote apoptotic death by various interconnected processes, including mitochondrial dysfunction, translational inhibition, the accumulation of various cellular stresses, and the specific induction of multifunctional proapoptotic factors, such as CHOP. The dual role of PERK in promoting both tumor progression and suppression makes it a complex target for therapeutic interventions. A comprehensive understanding of the intricacies of PERK pathway activation and their impact is essential for the development of effective therapeutic strategies, particularly in diseases like cancer, where the ER stress response is deregulated in most, if not all, of the solid and liquid tumors. This article provides an overview of the knowledge acquired from the study of animal models of cancer and tumor cell lines cultured in vitro on PERK's intracellular functions and their impact on cancer cells and their microenvironment, thus highlighting potential new therapeutic avenues that could target this protein.
Collapse
|
45
|
Ahn CH, Myong JS, Ahmed KR, Rahman MA, Fahim MMH, Choi M, Rahman M, Choi J, Kim K, Moon S, Dalli M, Syahputra RA, Shin SW, Harrath AH, Park MN, Kim B, Yoo HS. A pharmacoinformatic approach for studying Atractylodes Lancea DC's anticancer potential and control ROS-mediated apoptosis against prostate cancer cells. Front Oncol 2025; 15:1471110. [PMID: 39980567 PMCID: PMC11841406 DOI: 10.3389/fonc.2025.1471110] [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: 07/26/2024] [Accepted: 01/07/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Prostate cancer (PCa) is a malignancy characterized by abnormal cell proliferation in the prostate gland, a critical component of the male reproductive system. Atractylodes lancea DC. (ALD), a medicinal herb commonly used in traditional Asian medicine, is highly regarded for its antioxidant, antidiabetic, and anticancer properties. Virtual docking stud-ies have identified Atractylenolide II and III as active components of ALD, demonstrating strong binding potential to inhibit androgen receptor (AR) activity, with docking scores of -8.9 and -9.3, respectively. These findings suggest that ALD may exert a synergistic effect comparable to or greater than that of enzalutamide (ENZ) in inhibiting AR. How-ever, its specific anticancer and anti-metastatic mechanisms in prostate cancer remain unclear. Methods The cytotoxic effects of ALD were evaluated on PC3 and DU145 prostate cancer cells, as well as on the normal prostate cell line BPH-1. Cell viability was assessed using the EZ-Cytotoxic kit, while colony formation assays and TUNEL staining were used to meas-ure proliferation and apoptosis, respectively. Apoptosis was further analyzed through an-nexin V-FITC/PI staining and quantified by flow cytometry (FACS). Western blotting was performed to elucidate the underlying molecular mechanisms. Additionally, mito-chondrial membrane potential (ΔΨm) and intracellular calcium levels were measured to evaluate mitochondrial function, while reactive oxygen species (ROS) generation was assessed with and without pretreatment with N-acetylcysteine (NAC) . Results ALD selectively reduced the viability of PC3 and DU145 prostate cancer cells while spar-ing BPH-1 normal prostate cells, demonstrating cancer-selective cytotoxicity. ALD dis-rupted mitochondrial function by reducing ΔΨm and increasing intracellular calcium lev-els. A concentration-dependent increase in ROS generation was observed in PC3 and DU145 cells, which was completely inhibited by NAC pretreatment, confirming a ROS-mediated mechanism. Colony formation assays revealed a significant reduction in prolif-eration, while TUNEL and annexin V-FITC/PI staining indicated enhanced apoptosis. Western blot analysis showed that ALD modulates critical survival pathways, leading to apoptotic cell death. Discussion These findings demonstrate that ALD exerts potent anticancer effects against metastatic prostate cancer cells through ROS-mediated apoptosis and mitochondrial dysfunction, while exhibiting minimal cytotoxicity toward normal prostate cells. The presence of ac-tive compounds such as Atractylenolide II and III suggests a synergistic interaction that enhances AR inhibition and promotes apoptosis. ALD's ability to engage multiple path-ways highlights its therapeutic potential as a selective and multifaceted treatment for ag-gressive prostate cancer.
Collapse
Affiliation(s)
- Chi-Hoon Ahn
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ji Soo Myong
- East West Cancer Center, Seoul Korean Medicine Hospital, Daejeon University, Seoul, Republic of Korea
| | - Kazi Rejvee Ahmed
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Md Ataur Rahman
- Department of Oncology, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI, United States
| | | | - Min Choi
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Muntajin Rahman
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jinwon Choi
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Kiryang Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seungjoon Moon
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Chansol Hospital of Korean Medicine, Incheon, Republic of Korea
| | - Mohammed Dalli
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, University Mohammed the First, Oujda, Morocco
- Higher Institute of Nursing Professions and Health Techniques, Oujda, Morocco
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Sumatera Utara, Indonesia
| | - Sang-Won Shin
- Department of Humanities & Social Medicine, School of Korean Medicine, Pusan National University, Yangsan-si, Gyeongsangnam-do, Republic of Korea
| | - Abdel Halim Harrath
- College of Science, Department of Zoology, King Saud University, Riyadh, Saudi Arabia
| | - Moon Nyeo Park
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hwa-Seung Yoo
- East West Cancer Center, Seoul Korean Medicine Hospital, Daejeon University, Seoul, Republic of Korea
| |
Collapse
|
46
|
Lee EJ, Kim K, Diaz-Aguilar MS, Min H, Chavez E, Steinbergs KJ, Safarta LA, Zhang G, Ryan AF, Lin JH. Mutations in unfolded protein response regulator ATF6 cause hearing and vision loss syndrome. J Clin Invest 2025; 135:e175562. [PMID: 39570676 PMCID: PMC11785932 DOI: 10.1172/jci175562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/19/2024] [Indexed: 02/04/2025] Open
Abstract
Activating transcription factor 6 (ATF6) is a key regulator of the unfolded protein response (UPR) and is important for ER function and protein homeostasis in metazoan cells. Patients carrying loss-of-function ATF6 disease alleles develop the cone dysfunction disorder achromatopsia. The effect of loss of ATF6 function on other cell types, organs, and diseases in people remains unclear. Here, we report that progressive sensorineural hearing loss was a notable complaint in some patients carrying ATF6 disease alleles and that Atf6-/- mice also showed progressive auditory deficits affecting both sexes. In mice with hearing deficits, we found disorganized stereocilia on hair cells and focal loss of outer hair cells. Transcriptomics analysis of Atf6-/- cochleae revealed a marked induction of the UPR, especially through the protein kinase RNA-like endoplasmic reticulum kinase (PERK) arm. These findings identify ATF6 as an essential regulator of cochlear health and function. Furthermore, they support the idea that ATF6 inactivation in people causes progressive sensorineural hearing loss as part of a blindness-deafness genetic syndrome targeting hair cells and cone photoreceptors. Last, our genetic findings indicate that ER stress is an important pathomechanism underlying cochlear damage and hearing loss, with clinical implications for patient lifestyle modifications that minimize environmental and physiological sources of ER stress to the ear.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Departments of Pathology and
- Ophthalmology, Stanford University School of Medicine, Stanford, California, USA
- VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Kyle Kim
- Departments of Pathology and
- VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Monica Sophia Diaz-Aguilar
- Departments of Pathology and
- Ophthalmology, Stanford University School of Medicine, Stanford, California, USA
- VA Palo Alto Healthcare System, Palo Alto, California, USA
- Rush University Medical College, Chicago, Illinois, USA
| | - Hyejung Min
- Departments of Pathology and
- Ophthalmology, Stanford University School of Medicine, Stanford, California, USA
- VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Eduardo Chavez
- Departments of Otolaryngology and Neuroscience, UCSD and Veterans Administration Medical Center, La Jolla, California, USA
| | - Korina J. Steinbergs
- Departments of Pathology and
- Ophthalmology, Stanford University School of Medicine, Stanford, California, USA
| | - Lance A. Safarta
- Departments of Pathology and
- Ophthalmology, Stanford University School of Medicine, Stanford, California, USA
| | - Guirong Zhang
- Departments of Pathology and
- VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Allen F. Ryan
- Departments of Otolaryngology and Neuroscience, UCSD and Veterans Administration Medical Center, La Jolla, California, USA
| | - Jonathan H. Lin
- Departments of Pathology and
- Ophthalmology, Stanford University School of Medicine, Stanford, California, USA
- VA Palo Alto Healthcare System, Palo Alto, California, USA
| |
Collapse
|
47
|
İlhan İ, Asci H, Ozmen O, Buyukbayram Hİ, Arlıoglu M, Kurtbolat O. The renoprotective effects of cannabidiol on lipopolysaccharide-induced systemic inflammation model of rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1841-1851. [PMID: 39180672 DOI: 10.1007/s00210-024-03391-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024]
Abstract
Sepsis-induced renal damage poses a significant threat, necessitating effective therapeutic strategies. Cannabidiol (CBD) has beneficial effects on tissues and their functions by exhibiting antioxidant and anti-inflammatory effects. This study investigates the potential protective effects of CBD in mitigating lipopolysaccharide (LPS)-induced renal injury in Wistar Albino rats. Thirty-two Wistar Albino rats were categorized into control, LPS (5 mg/kg i.p.), LPS + CBD, and CBD (5 mg/kg i.p.) groups. After the experiment, samples were collected for biochemical, genetic, histopathological, and immunohistochemical analyses. Oxidative stress markers as total oxidant status (TOS) and total antioxidant status (TAS), oxidative stress index (OSI), superoxide dismutase (SOD), glutathione peroxidase (GPx), malondialdehyde (MDA), immune staining as tumor necrosis factor alpha (TNF-α), interleukin-10 (IL-10), caspase-3, gene expressions as nuclear factor erythroid 2-related factor 2 (NRF2), C/EBP homologous protein (CHOP), caspase-9, glucose-regulating protein 78 (GRP78), B-cell leukemia/lymphoma 2 (Bcl2), and tissue histology have been examined. The LPS-exposed group exhibited significant renal abnormalities, mitigated by CBD intervention in the LPS + CBD group. CBD reduced immunoexpression scores for TNF-α, caspase-3, and IL-10. Biochemically, CBD induced a positive shift in the oxidative balance, increasing TAS, SOD, and GPx, while decreasing TOS, OSI, and MDA levels. Genetic analyses highlighted CBD's regulatory impact on NRF2, CHOP, caspase-9, GRP78, and Bcl2, providing molecular insights into its protective role against LPS-induced renal damage. This study underscores CBD as a promising protective agent against sepsis-induced renal damage. Our findings could provide valuable insights into potential therapeutic avenues for addressing renal complications in sepsis.
Collapse
Affiliation(s)
- İlter İlhan
- Department of Biochemistry, Faculty of Medicine, Suleyman Demirel University, Isparta, 32200, Turkey.
| | - Halil Asci
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Ozlem Ozmen
- Department of Pathology, Faculty of Veterinary, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Halil İbrahim Buyukbayram
- Department of Biochemistry, Faculty of Medicine, Suleyman Demirel University, Isparta, 32200, Turkey
| | - Melih Arlıoglu
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Okan Kurtbolat
- Department of Pharmacology, Institute of Medicine, Suleyman Demirel University, Isparta, Turkey
| |
Collapse
|
48
|
Yeganeh Markid T, Pourahmadiyan A, Hamzeh S, Sharifi-Bonab M, Asadi MR, Jalaiei A, Rezazadeh M, Ghafouri-Fard S. A special focus on polyadenylation and alternative polyadenylation in neurodegenerative diseases: A systematic review. J Neurochem 2025; 169:e16255. [PMID: 39556113 DOI: 10.1111/jnc.16255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/14/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024]
Abstract
Neurodegenerative diseases (NDDs) are one of the prevailing conditions characterized by progressive neuronal loss. Polyadenylation (PA) and alternative polyadenylation (APA) are the two main post-transcriptional events that regulate neuronal gene expression and protein production. This systematic review analyzed the available literature on the role of PA and APA in NDDs, with an emphasis on their contributions to disease development. A comprehensive literature search was performed using the PubMed, Scopus, Cochrane, Google Scholar, Embase, Web of Science, and ProQuest databases. The search strategy was developed based on the framework introduced by Arksey and O'Malley and supplemented by the inclusion and exclusion criteria. The study selection was performed by two independent reviewers. Extraction and data organization were performed in accordance with the predefined variables. Subsequently, quantitative and qualitative analyses were performed. Forty-seven studies were included, related to a variety of NDDs, namely Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. Disease induction was performed using different models, including human tissues, animal models, and cultured cells. Most investigations were related to PA, although some were related to APA or both. Amyloid precursor protein (APP), Tau, SNCA, and STMN2 were the major genes identified; most of the altered PA patterns were related to mRNA stability and translation efficiency. This review particularly underscores the key roles of PA and APA in the pathogenesis of NDDs through their mechanisms that contribute to gene expression dysregulation, protein aggregation, and neuronal dysfunction. Insights into these mechanisms may lead to new therapeutic strategies focused on the modulation of PA and APA activities. Further research is required to investigate the translational potential of targeting these pathways for NDD treatment.
Collapse
Affiliation(s)
- Tarlan Yeganeh Markid
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azam Pourahmadiyan
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Soroosh Hamzeh
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mirmohsen Sharifi-Bonab
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohamad Reza Asadi
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Jalaiei
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Rezazadeh
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
García-Juárez M, García-Rodríguez A, Cruz-Carrillo G, Flores-Maldonado O, Becerril-Garcia M, Garza-Ocañas L, Torre-Villalvazo I, Camacho-Morales A. Intermittent Fasting Improves Social Interaction and Decreases Inflammatory Markers in Cortex and Hippocampus. Mol Neurobiol 2025; 62:1511-1535. [PMID: 39002056 DOI: 10.1007/s12035-024-04340-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 06/28/2024] [Indexed: 07/15/2024]
Abstract
Autism spectrum disorder (ASD) is a psychiatric condition characterized by reduced social interaction, anxiety, and stereotypic behaviors related to neuroinflammation and microglia activation. We demonstrated that maternal exposure to Western diet (cafeteria diet or CAF) induced microglia activation, systemic proinflammatory profile, and ASD-like behavior in the offspring. Here, we aimed to identify the effect of alternate day fasting (ADF) as a non-pharmacologic strategy to modulate neuroinflammation and ASD-like behavior in the offspring prenatally exposed to CAF diet. We found that ADF increased plasma beta-hydroxybutyrate (BHB) levels in the offspring exposed to control and CAF diets but not in the cortex (Cx) and hippocampus (Hpp). We observed that ADF increased the CD45 + cells in Cx of both groups; In control individuals, ADF promoted accumulation of CD206 + microglia cells in choroid plexus (CP) and increased in CD45 + macrophages cells and lymphocytes in the Cx. Gestational exposure to CAF diet promoted defective sociability in the offspring; ADF improved social interaction and increased microglia CD206 + in the Hpp and microglia complexity in the dentate gyrus. Additionally, ADF led to attenuation of the ER stress markers (Bip/ATF6/p-JNK) in the Cx and Hpp. Finally, biological modeling showed that fasting promotes higher microglia complexity in Cx, which is related to improvement in social interaction, whereas in dentate gyrus sociability is correlated with less microglia complexity. These data suggest a contribution of intermittent fasting as a physiological stimulus capable of modulating microglia phenotype and complexity in the brain, and social interaction in male mice.
Collapse
Affiliation(s)
- Martín García-Juárez
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico
| | - Adamary García-Rodríguez
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico
| | - Gabriela Cruz-Carrillo
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico
| | - Orlando Flores-Maldonado
- Facultad de Medicina, Departamento de Microbiología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Miguel Becerril-Garcia
- Facultad de Medicina, Departamento de Microbiología, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Lourdes Garza-Ocañas
- Department of Pharmacology and Toxicology, College of Medicine, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, México
| | - Ivan Torre-Villalvazo
- Departamento de Fisiología de La Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (INCMNSZ), 14080, Mexico City, Mexico
| | - Alberto Camacho-Morales
- Facultad de Medicina, Departamento de Bioquímica, Universidad Autónoma de Nuevo León, Madero y Dr. Aguirre Pequeño. Col. Mitras Centro, C.P. 64460, Monterrey, Nuevo León, Mexico.
- Centro de Investigación y Desarrollo en Ciencias de La Salud, Universidad Autónoma de Nuevo León, Unidad de Neurometabolismo, Monterrey, Nuevo León, Mexico.
- College of Medicine, Universidad Autónoma de Nuevo Leon, San Nicolás de los Garza, NL, Mexico.
| |
Collapse
|
50
|
Shin GS, Park Y, Kim JY, Kim CH, An MJ, Lee HM, Jo AR, Kim J, Hwangbo Y, Kim JW. Propylparaben-induced endoplasmic reticulum stress triggers G2/M phase cell cycle arrest and initiates caspase-3-dependent apoptosis in human lung cells. Genes Genomics 2025; 47:223-233. [PMID: 39699851 DOI: 10.1007/s13258-024-01605-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/01/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Propylparaben (PrP) is commonly used as an antimicrobial agent in food, cosmetics, and pharmaceuticals. While recent studies have shown that PrP exposure can cause various disruptions in cellular physiology, the precise mechanisms behind these effects remain unclear. OBJECTIVE In this study, we sought to examine the cytotoxic effects of PrP exposure on human lung cells in a dose- and time-dependent manner. We utilized flow cytometry to analyze the expression of proteins associated with the cell cycle and apoptosis at the single-cell level. RESULTS Our results showed that PrP treatment leads to a significant upregulation of genes related to ER stress. The activation of ER stress results in a decrease in cyclin B1 levels, which subsequently causes cell cycle arrest at the G2/M phase. After 48 h of PrP exposure, the unfolded protein response (UPR) triggers an apoptotic signaling pathway, increasing the number of cells undergoing caspase-3-mediated apoptosis. Together, these physiological changes lead to a reduction in cell viability in the presence of PrP. CONCLUSION These findings suggest that PrP exerts harmful effects on human lung cells by activating ER stress, which can lead to apoptosis and cell cycle arrest.
Collapse
Affiliation(s)
- Geun-Seup Shin
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yuna Park
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ji-Young Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Chul-Hong Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Mi-Jin An
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hyun-Min Lee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Ah-Ra Jo
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jinho Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Yujeong Hwangbo
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jung-Woong Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|