1
|
Sharma S, Satheesan A, Majumdar A, Mukherjee S, Basu A. PARP-16 regulates the PERK and IRE-1α Mediated Unfolded Protein Response in Japanese Encephalitis Virus-Infected Neural Stem/Progenitor Cells. Mol Neurobiol 2025; 62:8084-8096. [PMID: 39979689 DOI: 10.1007/s12035-025-04748-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 02/06/2025] [Indexed: 02/22/2025]
Abstract
The viral infection and subsequent accumulation of viral proteins in the infected cells leads to endoplasmic reticulum (ER) stress. Japanese encephalitis virus (JEV) infection in the Central Nervous System (CNS) has been shown to induce unfolded protein response (UPR). The ER stress is resolved by the UPR which comprises certain signals that are transduced from the ER either to both the cytoplasm or nucleus, resulting in the adaptation for survival or may even lead to apoptosis. Here, we demonstrate that Poly ADP-ribose polymerase-16 (PARP-16) expression is regulating the ER stress response following JEV infection of Neural Stem/Progenitor cells (NSPCs) in the BALB/c mouse model. Activation of the key sensors of UPR, namely, protein kinase R (PKR)-like ER kinase (PERK) and Inositol-requiring enzyme-1α (IRE-1α) by PARP-16 upon JEV infection, led to the activation of their downstream signalling cascade. The siRNA-mediated in vitro downregulation of PARP-16 in NSPCs alleviated the overall UPR, as the abundance of UPR markers and their downstream modulators of signalling cascade was found to be downregulated. These results highlight an important role of PARP-16 during JEV infection of NSPCs.
Collapse
Affiliation(s)
- Shivangi Sharma
- National Brain Research Centre, Manesar, Haryana, 122052, India
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, R3E0J9, Canada
| | | | - Atreye Majumdar
- National Brain Research Centre, Manesar, Haryana, 122052, India
| | - Sriparna Mukherjee
- National Brain Research Centre, Manesar, Haryana, 122052, India
- Department of Pharmacology and Physiology, Pavilion Roger-Gaudry, Universite de Montréal, Montréal, Québec, Canada
| | - Anirban Basu
- National Brain Research Centre, Manesar, Haryana, 122052, India.
| |
Collapse
|
2
|
Xu H, Hutchinson T, Koganti S, Rousseau B, Xia D, McIntosh M, Bhaduri-McIntosh S. Leveraging the interconnected unfolded protein response and NLRP3 inflammasome pathways to reactivate Epstein-Barr virus in diffuse large B-cell lymphomas. NAR Cancer 2025; 7:zcaf017. [PMID: 40330549 PMCID: PMC12051106 DOI: 10.1093/narcan/zcaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/31/2025] [Accepted: 04/25/2025] [Indexed: 05/08/2025] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL), when associated with Epstein-Barr virus (EBV) in immunocompromised individuals such as AIDS patients, presents a significant treatment challenge. Lytic induction therapy, which reactivates latent EBV to directly kill tumor cells and sensitize them to nucleoside analogs that block viral replication and immune clearance, offers promise. However, little is known about EBV reactivation in DLBCL. Here, we examined four EBV-positive DLBCL cell lines and found variable, cell-line-specific responses to lytic stimuli, with most showing an abortive response-either before or after genome replication, without virus release. This is in contrast to commonly studied lymphoma cells in which EBV reactivation typically leads to a full lytic cycle. Mechanistically, we show that the unfolded protein response (UPR), via a splice variant of the transcription factor XBP1, upregulates TXNIP and NLRP3, activating the inflammasome and removing a barrier to transcription of the EBV latent-to-lytic switch gene BZLF1. Combining lytic induction with the nucleoside analog ganciclovir enhanced oncolytic cell death. This study identifies a pivotal link between two danger sensing pathways, the UPR and the inflammasome, in reactivating the virus resident in DLBCL and suggests that controlled lytic reactivation could provide a basis for EBV-targeted therapies to improve outcomes in this malignancy.
Collapse
MESH Headings
- Lymphoma, Large B-Cell, Diffuse/virology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Humans
- Unfolded Protein Response
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- Virus Activation/drug effects
- Herpesvirus 4, Human/physiology
- Inflammasomes/metabolism
- Epstein-Barr Virus Infections/virology
- Cell Line, Tumor
- X-Box Binding Protein 1/genetics
- X-Box Binding Protein 1/metabolism
- Carrier Proteins/metabolism
- Carrier Proteins/genetics
- Trans-Activators/genetics
- Trans-Activators/metabolism
- Virus Latency
- Signal Transduction
Collapse
Affiliation(s)
- Huanzhou Xu
- Division of Infectious Diseases, Department of Pediatrics, University of Florida, Gainesville, FL 32610, United States
| | - Tarun E Hutchinson
- Child Health Research Institute, Department of Pediatrics, University of Florida, Gainesville, FL 32610, United States
| | - Siva Koganti
- Division of Infectious Diseases, Department of Pediatrics, University of Florida, Gainesville, FL 32610, United States
| | - Beth A Rousseau
- Division of Infectious Diseases, Department of Pediatrics, University of Florida, Gainesville, FL 32610, United States
| | - Daniel Xia
- Child Health Research Institute, Department of Pediatrics, University of Florida, Gainesville, FL 32610, United States
| | - Michael T McIntosh
- Child Health Research Institute, Department of Pediatrics, University of Florida, Gainesville, FL 32610, United States
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, United States
| | - Sumita Bhaduri-McIntosh
- Division of Infectious Diseases, Department of Pediatrics, University of Florida, Gainesville, FL 32610, United States
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL 32610, United States
| |
Collapse
|
3
|
Zhang H, Jiang N, Xu M, Jing D, Dong T, Liu Q, Lv Q, Huo R, Chen P, Li L, Wang X. M2 macrophage derived exosomal miR-20a-5p ameliorates trophoblast pyroptosis and placental injuries in obstetric antiphospholipid syndrome via the TXNIP/NLRP3 axis. Life Sci 2025; 370:123561. [PMID: 40127859 DOI: 10.1016/j.lfs.2025.123561] [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: 12/07/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
AIM Obstetric antiphospholipid syndrome (OAPS) is a pregnancy-related complication characterized by trophoblast pyroptosis and placental injury induced by antiphospholipid antibodies (aPLs). M2-polarized macrophage-derived exosomes (M2-exos) exert anti-inflammatory, immunomodulatory, and growth-promoting effects in various autoimmune diseases and tumors. However, their role in OAPS is not yet clear. Therefore, in this study, we isolated M2-exos from M2 macrophages and investigated their effects on trophoblast proliferation, death, migration, invasion, and pyroptosis following stimulation using aPLs. MAIN METHODS First, we established an animal model of OAPS and thereafter treated the OAPS mice with exogenous M2-exos via injection through the tail vein. Then to clarify the roles of miR-20a-5p and thioredoxin-interacting protein (TXNIP) in OAPS, we performed gain- or loss-of-function assays, and used GraphPad Prism software to analyze the collected data with statistical significance set at P < 0.05. KEY FINDINGS MicroRNAs (miRNAs) sequencing revealed the enrichment of miR-20a-5p in M2-exos, and these M2-exos significantly alleviated aPLs-induced trophoblast dysfunction. Our results also indicated that M2-exos delivered miR-20a-5p to trophoblast cells directly targeted thioredoxin-interacting protein (TXNIP), and thus suppressed the TXNIP/NLRP3 pathway, reduced pyroptosis and inflammation in trophoblast cells, and improved placental function and fetal development. SIGNIFICANCE M2-exos improve pregnancy outcomes in OAPS via the miR-20a-5p/TXNIP/NLRP3 axis, and thus represent as a novel therapeutic approach for aPLs-induced OAPS.
Collapse
Affiliation(s)
- Hongyuan Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China
| | - Ning Jiang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Mingyang Xu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Die Jing
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Tingting Dong
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Qian Liu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Feixian County People's Hospital, Linyi 273400, Shandong, China
| | - Qingfeng Lv
- The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, Shandong, China
| | - Ruiheng Huo
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Pengzheng Chen
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China.
| | - Lei Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China.
| | - Xietong Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China.
| |
Collapse
|
4
|
Sun J, Lee K, Kutseikin S, Guerrero A, Rius B, Madhavan A, Buasakdi C, Cheong KN, Chatterjee P, Rosen DA, Yoon L, Ardejani MS, Mendoza A, Rosarda JD, Saez E, Kelly JW, Wiseman RL. Identification of a Selective Pharmacologic IRE1/XBP1s Activator with Enhanced Tissue Exposure. ACS Chem Biol 2025; 20:993-1003. [PMID: 40231944 DOI: 10.1021/acschembio.4c00867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Activation of the IRE1/XBP1s signaling arm of the unfolded protein response (UPR) has emerged as a promising strategy to mitigate etiologically diverse diseases. Despite this promise, few compounds are available to selectively activate IRE1/XBP1s signaling to probe the biologic and therapeutic implications of this pathway in human disease. Recently, we identified the compound IXA4 as a highly selective activator of protective IRE1/XBP1s signaling. While IXA4 has proven useful for increasing IRE1/XBP1s signaling in cultured cells and mouse liver, the utility of this compound is restricted by its limited activity in other tissues. To broaden our ability to pharmacologically interrogate the impact of IRE1/XBP1s signaling in vivo, we sought to identify IRE1/XBP1s activators with greater tissue activity than IXA4. We reanalyzed 'hits' from the high throughput screen used to identify IXA4, selecting compounds from structural classes not previously pursued. We then performed global RNAseq to confirm that these compounds showed transcriptome-wide selectivity for IRE1/XBP1s activation. Functional profiling revealed compound IXA62 as a selective IRE1/XBP1s activator that reduced Aβ secretion from CHO7PA2 cells and enhanced glucose-stimulated insulin secretion from rat insulinoma cells, mimicking the effects of IXA4 in these assays. IXA62 robustly and selectively activated IRE1/XBP1s signaling in the liver of mice dosed compound intraperitoneally or orally. In treated mice, IXA62 showed broader tissue activity, relative to IXA4, inducing expression of IRE1/XBP1s target genes in additional tissues such as kidney and lung. Collectively, our results designate IXA62 as a selective IRE1/XBP1s signaling activating compound with enhanced tissue activity, which increases our ability to pharmacologically probe the biologic significance and potential therapeutic utility of enhancing adaptive IRE1/XBP1s signaling in vivo.
Collapse
Affiliation(s)
- Jie Sun
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Kyunga Lee
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
| | - Sergei Kutseikin
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Adrian Guerrero
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
| | - Bibiana Rius
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Aparajita Madhavan
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Chavin Buasakdi
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Ka-Neng Cheong
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
- Department of Immunology and Microbial Science, Scripps Research, La Jolla, California 92037, United States
| | - Priyadarshini Chatterjee
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Dorian A Rosen
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Leonard Yoon
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
| | - Maziar S Ardejani
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
| | - Alejandra Mendoza
- Department of Immunology and Microbial Science, Scripps Research, La Jolla, California 92037, United States
| | - Jessica D Rosarda
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Enrique Saez
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Jeffery W Kelly
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
- The Skaggs Institute for Chemical Biology, Scripps Research, La Jolla, California 92037, United States
| | - R Luke Wiseman
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| |
Collapse
|
5
|
Mohammad-Sadeghipour M, Nematollahi MH, Sahebazzamani M, Ahmadinia H, Hajizadeh MR, Mahmoodi M, Sahebkar A. G protein-coupled estrogen receptor reduces the breast cancer cell survival by regulating the IRE1α/miR-17-5p/TXNIP pathway. J Steroid Biochem Mol Biol 2025; 252:106770. [PMID: 40328365 DOI: 10.1016/j.jsbmb.2025.106770] [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: 12/10/2024] [Revised: 05/01/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025]
Abstract
This study aimed to explore whether GPER can induce the UPR response in the SKBR3 cell line through ER and IREα activation, and to assess whether this response leads to cell survival or cell death. Additionally, the study sought to evaluate the impact of this response on cell behaviors such as apoptosis, migration, and drug resistance. To activate the UPR and induce ER stress, we treated the MCF10A cell line with 0.5 µg/ml TUN for 24 and 48 h. The expression levels of XBP-1 and C/EBP homology protein (CHOP) genes (ER stress markers) were measured using the qRT-PCR technique. The MCF10A + TUN cell line was used as a positive control. To determine the optimal doses of G1 and tamoxifen (TAM), we evaluated GPER expression using qRT-PCR analysis. Cells were then treated with various doses of G1 (1000 nM), G15 (1000 nM), and TAM (2000 nM), both individually and in combination (G1 + G15, TAM + G15, G1 + TAM), for 24 and 48 h. We measured the expression of GPER, IRE1α, MiR-17-5p, TXNIP, ABCB1, and ABCC1 genes. Apoptosis was assessed via flow cytometry, and cell migration was examined using the wound-healing assay. Our results demonstrated that GPER activation by G1 and TAM significantly increased IRE1α expression in SKBR3 cells. This activation, through its RIDD activity, cleaved miR-17-5p and initiated the UPR death response. The upregulation of the TXNIP gene expression enhanced apoptosis and chemotherapy sensitivity while decreasing cell migration. Interestingly, these effects were notably reversed by G15 treatment. In summary, the GPER/IRE1α/miR-17-5p/TXNIP axis plays a key role in the UPR pro-death response, promoting programmed cell death, reducing migration, and decreasing drug resistance in SKBR3 cells.
Collapse
Affiliation(s)
- Maryam Mohammad-Sadeghipour
- Molecular Medicine Research Center, Inistitute of Basic Medical Sciences Research, Rafsanjan University of Medical Sciences, Rafsanjan City, Rafsanjan Province, Iran; Department of Clinical Biochemistry, Afzalipoor Faculty of Medicine, Kerman University of Medical Sciences, Kerman City, Kerman Province, Iran
| | - Mohammad Hadi Nematollahi
- Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman City, Kerman Province, Iran
| | - Maryam Sahebazzamani
- Department of Clinical Biochemistry, Afzalipoor Faculty of Medicine, Kerman University of Medical Sciences, Kerman City, Kerman Province, Iran
| | - Hassan Ahmadinia
- Department of Biostatistics, Rafsanjan University of Medical Sciences, Rafsanjan City, Rafsanjan Province, Iran
| | - Mohammad Reza Hajizadeh
- Molecular Medicine Research Center, Inistitute of Basic Medical Sciences Research, Rafsanjan University of Medical Sciences, Rafsanjan City, Rafsanjan Province, Iran; Department of Clinical Biochemistry, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan City, Rafsanjan Province, Iran
| | - Mehdi Mahmoodi
- Molecular Medicine Research Center, Inistitute of Basic Medical Sciences Research, Rafsanjan University of Medical Sciences, Rafsanjan City, Rafsanjan Province, Iran; Department of Clinical Biochemistry, Afzalipoor Faculty of Medicine, Kerman University of Medical Sciences, Kerman City, Kerman Province, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
He F, Zheng Y, Elsabagh M, Fan K, Zha X, Zhang B, Wang M, Zhang H. Gut microbiota modulate intestinal inflammation by endoplasmic reticulum stress-autophagy-cell death signaling axis. J Anim Sci Biotechnol 2025; 16:63. [PMID: 40312439 PMCID: PMC12046778 DOI: 10.1186/s40104-025-01196-8] [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: 11/20/2024] [Accepted: 03/17/2025] [Indexed: 05/03/2025] Open
Abstract
The intestinal tract, a complex organ responsible for nutrient absorption and digestion, relies heavily on a balanced gut microbiome to maintain its integrity. Disruptions to this delicate microbial ecosystem can lead to intestinal inflammation, a hallmark of inflammatory bowel disease (IBD). While the role of the gut microbiome in IBD is increasingly recognized, the underlying mechanisms, particularly those involving endoplasmic reticulum (ER) stress, autophagy, and cell death, remain incompletely understood. ER stress, a cellular response to various stressors, can trigger inflammation and cell death. Autophagy, a cellular degradation process, can either alleviate or exacerbate ER stress-induced inflammation, depending on the specific context. The gut microbiome can influence both ER stress and autophagy pathways, further complicating the interplay between these processes. This review delves into the intricate relationship between ER stress, autophagy, and the gut microbiome in the context of intestinal inflammation. By exploring the molecular mechanisms underlying these interactions, we aim to provide a comprehensive theoretical framework for developing novel therapeutic strategies for IBD. A deeper understanding of the ER stress-autophagy axis, the gut microbial-ER stress axis, and the gut microbial-autophagy axis may pave the way for targeted interventions to restore intestinal health and mitigate the impact of IBD.
Collapse
Affiliation(s)
- Feiyang He
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
- Key Laboratory of Fujian Universities Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, 364012, P. R. China
| | - Yi Zheng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Mabrouk Elsabagh
- Department of Animal Production and Technology, Faculty of Agricultural Sciences and Technologies, Niğde Ömermer Halisdemir University, Nigde, 51240, Turkey
| | - Kewei Fan
- Key Laboratory of Fujian Universities Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, 364012, P. R. China
| | - Xia Zha
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Bei Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Science, Shihezi, 832000, P. R. China
| | - Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China.
| |
Collapse
|
7
|
Wang C, Ji X, Wang X, Song Y, Pan C, Qian M, Jin Y. The endoplasmic reticulum-mitochondrial crosstalk involved in nanoplastics and di(2-ethylhexyl) phthalate co-exposure induced the damage to mouse mammary epithelial cells. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 372:126014. [PMID: 40057162 DOI: 10.1016/j.envpol.2025.126014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
Abstract
With the extensive use of plastic products, significant amounts of microplastics, nanoplastic particles (NPs), and plasticizers such as Di(2-ethylhexyl) phthalate (DEHP) are continuously released into the environment. However, the toxic effects of NPs alone or in combination with DEHP on mammary glands remain unreported. This study investigates the impacts of NPs and DEHP on the structure and function of mouse mammary epithelial cells and elucidates the underlying molecular mechanisms. We found that co-exposure to NPs and DEHP induced severe pyroptosis, inflammation and oxidative stress in HC11 cells. Co-exposure also caused mitochondrial damage, as evidenced by changes in mitochondrial membrane potential, increase in mitochondrial ROS and inhibition of ATP production. Moreover, NPs and DEHP co-exposure increased the transcriptional levels of endoplasmic reticulum (ER) stress-related genes, activated the inflammation-related NLRP3 signaling pathway, and damaged the cell membrane integrity. Notably, Co-exposure enhanced the ER-mitochondria crosstalk in HC11 cells, as evidenced by the upregulated transcriptional levels of ER Ca2+ channel proteins (Ip3r1, Grp75 and Vdac1), increased mitochondrial Ca2+ levels, and expanded mitochondrial-ER contact areas. In summary, this study revealed that NPs and DEHP co-exposure had the potential to induce pyroptosis and inflammation by enhancing the ER-mitochondria crosstalk, ultimately resulting in injury to mammary glands. These findings would provide some new insights into the molecular mechanisms underlying the toxic effects of NPs and DEHP to mammary glands.
Collapse
Affiliation(s)
- Caihong Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Xiang Ji
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Xiaoya Wang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Yunmeng Song
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Chunqiang Pan
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| | - Mingrong Qian
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Interdisciplinary Research Academy, Zhejiang Shuren University, Hangzhou, 310015, China.
| | - Yuanxiang Jin
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310032, China
| |
Collapse
|
8
|
Sharma A, Heffernan LM, Hoang K, Jeyaseelan S, Beavers WN, Abuaita BH. Activation of the endoplasmic reticulum stress regulator IRE1α compromises pulmonary host defenses. Cell Rep 2025; 44:115632. [PMID: 40315054 DOI: 10.1016/j.celrep.2025.115632] [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: 10/07/2024] [Revised: 01/29/2025] [Accepted: 04/09/2025] [Indexed: 05/04/2025] Open
Abstract
The endoplasmic reticulum (ER) stress sensor inositol-requiring enzyme 1α (IRE1α) is associated with lung infections where innate immune cells are drivers for progression and resolution ammatory cytokinesflammation. Yet, the role of IRE1α in pulmonary innate immune host defense during acute respiratory infection remains unexplored. Here, we found that activation of IRE1α in infected lungs compromises immunity against methicillin-resistant Staphylococcus aureus (MRSA)-induced primary and secondary pneumonia. Moreover, activation of IRE1α in MRSA-infected lungs and alveolar macrophages (AMs) leads to exacerbated production of inflammatory mediators followed by cell death. Ablation of myeloid IRE1α or global IRE1α inhibition confers protection against MRSA-induced pneumonia with improved survival, bacterial clearance, cytokine reduction, and lung injury. In addition, loss of myeloid IRE1α protects mice against MRSA-induced secondary to influenza pneumonia by promoting AM survival. Thus, activation of IRE1α is detrimental to pneumonia, and therefore, it shows potential as a target to control excessive unresolved lung inflammation.
Collapse
Affiliation(s)
- Amit Sharma
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Linda M Heffernan
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Ky Hoang
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Samithamby Jeyaseelan
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - William N Beavers
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA; Mass Spectrometry Resource Center, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA
| | - Basel H Abuaita
- Department of Pathobiological Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA 70803, USA.
| |
Collapse
|
9
|
Moreews M, Karlsson MCI. Endoplasmic reticulum stress: A key player in immune cell regulation and autoimmune disorders. Semin Immunol 2025; 78:101954. [PMID: 40267701 DOI: 10.1016/j.smim.2025.101954] [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: 10/14/2024] [Revised: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 04/25/2025]
Abstract
The endoplasmic reticulum (ER) is a large organelle, found in all eukaryotes, that is essential for normal cellular function. This function encompasses protein folding and quality control, post-translational modifications, lipid regulation, and the storage of intracellular calcium, among others. These diverse processes are essential for maintaining proteome stability. Therefore, a robust surveillance system is established under stress to ensure cell homeostasis. Sources of stress can originate from the cellular environment, including nutrient deprivation, hypoxia, and low pH, as well as from endogenous signals within the cell, such as metabolic challenges and increased demands for protein production. When cellular homeostasis is altered by one of these triggers, ER primary functions are altered which leads to the accumulation of misfolded proteins. These impaired proteins trigger the activation of the Unfolded Protein Response (UPR) pathway. This response aims at reducing ER stress by implementing the induction of complex programs to restore cell homeostasis. However, extended ER stress can modify the UPR response, shifting its signals from promoting survival to triggering pathways that reprogram or eliminate affected cells.
Collapse
Affiliation(s)
- Marion Moreews
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm SE-171 77, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm SE-171 77, Sweden.
| |
Collapse
|
10
|
Wu D, Eeda V, Maria Z, Rawal K, Wang A, Herlea-Pana O, Babu Undi R, Lim HY, Wang W. Targeting IRE1α improves insulin sensitivity and thermogenesis and suppresses metabolically active adipose tissue macrophages in male obese mice. eLife 2025; 13:RP100581. [PMID: 40244655 PMCID: PMC12005715 DOI: 10.7554/elife.100581] [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] [Indexed: 04/18/2025] Open
Abstract
Overnutrition engenders the expansion of adipose tissue and the accumulation of immune cells, in particular, macrophages, in the adipose tissue, leading to chronic low-grade inflammation and insulin resistance. In obesity, several proinflammatory subpopulations of adipose tissue macrophages (ATMs) identified hitherto include the conventional 'M1-like' CD11C-expressing ATM and the newly discovered metabolically activated CD9-expressing ATM; however, the relationship among ATM subpopulations is unclear. The ER stress sensor inositol-requiring enzyme 1α (IRE1α) is activated in the adipocytes and immune cells under obesity. It is unknown whether targeting IRE1α is capable of reversing insulin resistance and obesity and modulating the metabolically activated ATMs. We report that pharmacological inhibition of IRE1α RNase significantly ameliorates insulin resistance and glucose intolerance in male mice with diet-induced obesity. IRE1α inhibition also increases thermogenesis and energy expenditure, and hence protects against high fat diet-induced obesity. Our study shows that the 'M1-like' CD11c+ ATMs are largely overlapping with but yet non-identical to CD9+ ATMs in obese white adipose tissue. Notably, IRE1α inhibition diminishes the accumulation of obesity-induced metabolically activated ATMs and 'M1-like' ATMs, resulting in the curtailment of adipose inflammation and ensuing reactivation of thermogenesis, without augmentation of the alternatively activated M2 macrophage population. Our findings suggest the potential of targeting IRE1α for the therapeutic treatment of insulin resistance and obesity.
Collapse
Affiliation(s)
- Dan Wu
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at BirminghamBirminghamUnited States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Venkateswararao Eeda
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Zahra Maria
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Komal Rawal
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | | | - Oana Herlea-Pana
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Ram Babu Undi
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Hui-Ying Lim
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at BirminghamBirminghamUnited States
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| | - Weidong Wang
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at BirminghamBirminghamUnited States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Sciences CenterOklahoma CityUnited States
| |
Collapse
|
11
|
Soczewski E, Schafir A, Castagnola L, Materazzi L, Fernández L, Marcial A, Presa J, Saravia F, Grasso E, Vota D, Pérez Leirós C, Ramhorst R, Gori S. Exacerbated endoplasmic reticulum stress transmitted by endometrial stromal cells alters the conditioning of tolerogenic dendritic cells affecting trophoblast migration. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:795-807. [PMID: 40112190 DOI: 10.1093/jimmun/vkae065] [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: 07/19/2024] [Accepted: 12/17/2024] [Indexed: 03/22/2025]
Abstract
Endometrial stromal cells acquire a secretory profile associated with endoplasmic reticulum stress (ERS) and unfolded protein response (UPR) related to the onset of a sterile inflammatory response essential for sustaining embryo implantation. However, exacerbated stromal ERS/UPR is associated with reproductive complications. Given the ability of dendritic cells (DCs) to sense stress signals and be conditioned by stromal cells, here we investigated the transmission of ERS (TERS) from stromal cells to monocytes and its impact on tolerogenic DCs conditioning. Blood monocytes were differentiated into DCs (rhGM-CSF+rhIL-4, 5 d) in the presence or absence of conditioned media derived from either thapsigargin-treated (stressed) or nonstressed human endometrial stromal cell line. Soluble factors released by stressed stromal cells impaired CD1a+CD14- DC differentiation and induced a proinflammatory profile, increasing the CD86high cell population, COX-2 expression, and tumor necrosis factor (TNF)-α, interleukin (IL)-8 and IL-1β secretion. Additionally, TERS was observed in these cultures, with increased expression of IRE1α, PERK, and ATF4. Even the splicing of the adaptive UPR marker XBP1 was increased though at low levels, its nuclear translocation was unchanged. These effects on spliced XBP1, coupled with a decreased GRP78/BiP and heightened CHOP expression, suggest the triggering of terminal UPR over adaptive UPR, confirmed by the induction of lytic cell death in stressed cultures. Finally, exacerbated TERS negatively impacted trophoblast migration in a blastocyst-like spheroid in vitro model. These findings suggest that exacerbated stromal ERS can be transmitted to monocytes, altering their differentiation, immune profile, and viability, which could ultimately impair trophoblast migration.
Collapse
Affiliation(s)
- Elizabeth Soczewski
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Jena, Germany
| | - Ana Schafir
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Lara Castagnola
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Lourdes Materazzi
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Laura Fernández
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Agustina Marcial
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Jessica Presa
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Flavia Saravia
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
- Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Esteban Grasso
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Daiana Vota
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Rosanna Ramhorst
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| | - Soledad Gori
- Universidad de Buenos Aires (UBA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN-CONICET), Buenos Aires, Argentina
| |
Collapse
|
12
|
Jo S, Jing G, Chen J, Xu G, Shalev A. Oral TIX100 protects against obesity-associated glucose intolerance and diet-induced adiposity. Diabetes Obes Metab 2025; 27:2223-2231. [PMID: 39895486 PMCID: PMC11885086 DOI: 10.1111/dom.16223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 01/11/2025] [Accepted: 01/21/2025] [Indexed: 02/04/2025]
Abstract
AIMS Glucagon-like peptide 1 receptor agonists and dual agonists have changed the treatment landscape of obesity and type 2 diabetes (T2D), but significant limitations have emerged due to their gastrointestinal side effects, loss of lean mass, and necessity for ongoing subcutaneous injections. Our objective was, therefore, to test a novel small molecule as a different and potentially better tolerated oral medications to improve obesity-associated impairment in glucose homeostasis. MATERIALS AND METHODS High-fat diet (HFD)-fed mice or severely obese, leptin-deficient ob/ob mice were randomly assigned to serve as controls or receive oral TIX100, a novel thioredoxin-interacting protein (TXNIP) inhibitor just approved by the FDA as an investigational new drug for type 1 diabetes (T1D). The TIX100 effects on glucose intolerance and weight control were then assessed. RESULTS TIX100 protected against HFD-induced glucose intolerance, hyperinsulinemia, and hyperglucagonemia. TIX100 also reduced diet-induced adiposity resulting in 15% lower weight in treated mice as compared with controls on HFD (p <0.05), while preserving lean mass. Even though the TIX100 weight effects were lost in ob/ob mice, TIX100 improved glucose control leading to a dramatic 2.3% reduction in HbA1C (p <0.05), independent of any weight loss. This is consistent with the beneficial effects of TIX100 in non-obese diabetes models and its protection against elevated TXNIP and islet cell stress common to all diabetes types. CONCLUSIONS Thus, TIX100 may provide a novel, oral therapy for T2D that targets underlying disease pathology including islet cell dysfunction and hyperglucagonemia and promotes metabolic health and weight control without aggressive weight loss.
Collapse
Affiliation(s)
- SeongHo Jo
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Gu Jing
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Junqin Chen
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Guanlan Xu
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Anath Shalev
- Comprehensive Diabetes Center, Department of Medicine, Division of Endocrinology, Diabetes, and MetabolismUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
13
|
Li C, Fang Y, Chen YM. Beyond Redox Regulation: Novel Roles of TXNIP in the Pathogenesis and Therapeutic Targeting of Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:615-625. [PMID: 39814099 PMCID: PMC11959421 DOI: 10.1016/j.ajpath.2024.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
Cellular stress, such as oxidative and endoplasmic reticulum (ER) stresses, contributes to the development of various kidney diseases. Oxidative stress is prompted by reactive oxygen species accumulation and delicately mitigated by glutathione and thioredoxin (Trx) antioxidant systems. Initially identified as a Trx-binding partner, Trx-interacting protein (TXNIP) is significantly up-regulated and activated by oxidative and ER stresses. The function of TXNIP is closely linked to its subcellular localizations. Under normal physiological conditions, TXNIP primarily localizes to the nucleus. When exposed to reactive oxygen species or ER stress, TXNIP relocates to mitochondria and binds to mitochondrial Trx2, which releases Trx-tethered apoptosis signal-regulating kinase 1 and activates apoptosis signal-regulating kinase 1-mediated apoptosis. Oxidative and ER stresses are also closely associated with autophagy. TXNIP can promote or inhibit autophagy depending on context. Although recent studies have highlighted the indispensable role of TXNIP in the etiology and progression of kidney disease, TXNIP-targeted therapy is still missing. This review focuses on the following: i) oxidative and ER stresses; ii) regulation and function of TXNIP during cellular stress; iii) TXNIP in stress-regulated autophagy; iv) TXNIP in kidney diseases (nephrotic syndrome, diabetic nephropathy and chronic kidney disease, acute kidney injury, and kidney aging); and v) novel treatment agents targeting TXNIP in kidney disease. Current advances in chemical compounds and RNA-based therapy suppressing TXNIP are also reviewed.
Collapse
Affiliation(s)
- Chuang Li
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Yili Fang
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Ying Maggie Chen
- Division of Nephrology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri; Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, Missouri.
| |
Collapse
|
14
|
Abdel Mageed SS, Elimam H, Elesawy AE, Abulsoud AI, Raouf AA, Tabaa MME, Mohammed OA, Zaki MB, Abd-Elmawla MA, El-Dakroury WA, Mangoura SA, Elrebehy MA, Elballal MS, Mohamed AA, Ashraf A, Abdel-Reheim MA, Eleragi AMS, Abdellatif H, Doghish AS. Unraveling the impact of miRNAs on gouty arthritis: diagnostic significance and therapeutic opportunities. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3433-3450. [PMID: 39560752 PMCID: PMC11978694 DOI: 10.1007/s00210-024-03603-9] [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: 06/18/2024] [Accepted: 11/03/2024] [Indexed: 11/20/2024]
Abstract
Gouty arthritis is a prevalent inflammatory illness. Gout attacks begin when there is an imbalance in the body's uric acid metabolism, which leads to urate buildup and the development of the ailment. A family of conserved, short non-coding RNAs known as microRNAs (miRNAs) can regulate post-transcriptional protein synthesis by attaching to the 3' untranslated region (UTR) of messenger RNA (mRNA). An increasing amount of research is pointing to miRNAs as potential players in several inflammatory diseases, including gouty arthritis. miRNAs may influence the progression of the disease by regulating immune function and inflammatory responses. This review mainly focused on miRNAs and how they contribute to gouty arthritis. It also looked at how miRNAs could be used as diagnostic, prognostic, and potential therapeutic targets.
Collapse
Affiliation(s)
- Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Hanan Elimam
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| | - Ahmed Amr Raouf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Manar Mohammed El Tabaa
- Pharmacology & Environmental Toxicology, Environmental Studies & Research Institute (ESRI), University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Mohamed Bakr Zaki
- Biochemistry, Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Sadat City, 32897, Menoufia, Egypt
| | - Mai A Abd-Elmawla
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Safwat Abdelhady Mangoura
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Galala University, New Galala City, 43713, Suez,, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang, Republic of Korea
| | - Aya A Mohamed
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Alaa Ashraf
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | | | - Ali M S Eleragi
- Department of Microorganisms and Clinical Parasitology, College of Medicine, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Hussein Abdellatif
- Department of Human and Clinical Anatomy, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
- Department of Anatomy and Embryology, Faculty of Medicine, University of Mansoura, Mansoura, 35516, Egypt
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt.
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt.
| |
Collapse
|
15
|
Hansen MS, Pokharel P, Piganelli J, Sussel L. The Chicken or the Egg Dilemma: Understanding the Interplay between the Immune System and the β Cell in Type 1 Diabetes. Cold Spring Harb Perspect Med 2025; 15:a041591. [PMID: 38951031 PMCID: PMC11960692 DOI: 10.1101/cshperspect.a041591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
In this review, we explore the complex interplay between the immune system and pancreatic β cells in the context of type 1 diabetes (T1D). While T1D is predominantly considered a T-cell-mediated autoimmune disease, the inability of human leukocyte antigen (HLA)-risk alleles alone to explain disease development suggests a role for β cells in initiating and/or propagating disease. This review delves into the vulnerability of β cells, emphasizing their susceptibility to endoplasmic reticulum (ER) stress and protein modifications, which may give rise to neoantigens. Additionally, we discuss the role of viral infections as contributors to T1D onset, and of genetic factors with dual impacts on the immune system and β cells. A greater understanding of the interplay between environmental triggers, autoimmunity, and the β cell will not only lead to insight as to why the islet β cells are specifically targeted by the immune system in T1D but may also reveal potential novel therapeutic interventions.
Collapse
Affiliation(s)
- Maria Skjøtt Hansen
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| | - Pravil Pokharel
- Division of Endocrinology Diabetes and Metabolism, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Jon Piganelli
- Division of Endocrinology Diabetes and Metabolism, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | - Lori Sussel
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| |
Collapse
|
16
|
Liu J, Chen X, Liu J, Peng C, Wang F, Huang X, Li S, Liu Y, Shou W, Cao D, Li X. Prenatal Inflammatory Exposure Predisposes Offspring to Chronic Kidney Diseases Via the Activation of the eIF2α-ATF4 Pathway. Inflammation 2025; 48:747-759. [PMID: 38913145 DOI: 10.1007/s10753-024-02084-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2024] [Indexed: 06/25/2024]
Abstract
It has recently become more recognized that renal diseases in adults can originate from adverse intrauterine (maternal) environmental exposures. Previously, we found that prenatal lipopolysaccharide (LPS) exposure can result in chronic renal inflammation, which leads to renal damage in older offspring rats. To test whether prenatal inflammatory exposure predisposes offspring to renal damage, a mouse model of oral adenine consumption-induced chronic kidney disease (CKD) was applied to offspring from prenatal LPS-treated mothers (offspring-pLPS) and age-matched control offspring of prenatal saline-treated mothers (offspring-pSaline). We found that offspring-pLPS mice presented with more severe renal collagen deposition and renal dysfunction after 4 weeks of adenine consumption than sex- and treatment-matched offspring-pSaline controls. To illustrate the underlying molecular mechanism, we subjected offspring-pLPS and offspring-pSaline kidneys to genome-wide transcriptomic analysis. Bioinformatic analysis of the sequencing data, together with further experimental confirmation, revealed a strong activation of the PERK-eIF2α-ATF4-mediated unfolded protein response (UPR) in offspring-pLPS kidneys, which likely contributed to the CKD predisposition seen in offspring-pLPS mice. More importantly, the specific eIF2α-ATF4 signaling inhibitor ISIRB was able to prevent adenine-induced CKD in the offspring-pLPS mice. Our findings suggest that the eIF2α-ATF4-mediated UPR, but not PERK, is likely the major disease-causing pathway in prenatal inflammatory exposure-induced CKD predisposition. Our study also suggests that targeting this signaling pathway is a potentially promising approach for CKD treatment.
Collapse
Affiliation(s)
- Jie Liu
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - Xin Chen
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - Jie Liu
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - Cuiping Peng
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - Fangjie Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Xiaoyong Huang
- Institute of Immunology, PLA, Army Medical University, Chongqing, China
| | - Shuhui Li
- Department of Clinical Biochemistry, College of Pharmacy, Army Medical University, Chongqing, China
| | - Ying Liu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Weinian Shou
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dayan Cao
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China.
| | - Xiaohui Li
- Institute of Materia Medica and Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China.
| |
Collapse
|
17
|
Shalev A. Target Discovery to Diabetes Therapy-TXNIP From Bench to Bedside With NIDDK. Endocrinology 2025; 166:bqaf055. [PMID: 40105688 PMCID: PMC11949688 DOI: 10.1210/endocr/bqaf055] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 03/10/2025] [Accepted: 03/15/2025] [Indexed: 03/20/2025]
Abstract
Diabetes is the most expensive chronic disease in the United States, with more than $400 billion in annual costs, and it affects more than 38 million Americans. While major advances in drug treatment have been made for type 2 diabetes (T2D) and the often-associated obesity, there are still no approved and effective medications targeting β-cell loss or islet dysfunction, which is one of the major underlying causes of both type 1 diabetes (T1D) and T2D. In addition, there are no oral medications for T1D approved in the United States more than 100 years after the discovery of insulin, and attractive therapeutic targets are only starting to emerge. As we celebrate the 75th anniversary of the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), progress is finally being made in this area with NIDDK support. This mini-review follows the discovery of thioredoxin-interacting protein inhibitors as an example of a methodical approach to identify and develop an oral β-cell treatment for T1D. It further discusses how the initial molecular findings were translated into novel clinical treatment approaches that promote the patient's own islet health and β-cell function using drug repurposing as well as new drug discovery.
Collapse
Affiliation(s)
- Anath Shalev
- Department of Medicine, Comprehensive Diabetes Center and Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL 35294-2182, USA
| |
Collapse
|
18
|
Zaher A, Stephens SB. Breaking the Feedback Loop of β-Cell Failure: Insight into the Pancreatic β-Cell's ER-Mitochondria Redox Balance. Cells 2025; 14:399. [PMID: 40136648 PMCID: PMC11941261 DOI: 10.3390/cells14060399] [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: 02/10/2025] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Pancreatic β-cells rely on a delicate balance between the endoplasmic reticulum (ER) and mitochondria to maintain sufficient insulin stores for the regulation of whole animal glucose homeostasis. The ER supports proinsulin maturation through oxidative protein folding, while mitochondria supply the energy and redox buffering that maintain ER proteostasis. In the development of Type 2 diabetes (T2D), the progressive decline of β-cell function is closely linked to disruptions in ER-mitochondrial communication. Mitochondrial dysfunction is a well-established driver of β-cell failure, whereas the downstream consequences for ER redox homeostasis have only recently emerged. This interdependence of ER-mitochondrial functions suggests that an imbalance is both a cause and consequence of metabolic dysfunction. In this review, we discuss the regulatory mechanisms of ER redox control and requirements for mitochondrial function. In addition, we describe how ER redox imbalances may trigger mitochondrial dysfunction in a vicious feed forward cycle that accelerates β-cell dysfunction and T2D onset.
Collapse
Affiliation(s)
- Amira Zaher
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52246, USA;
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52246, USA
| | - Samuel B. Stephens
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52246, USA;
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52246, USA
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52246, USA
| |
Collapse
|
19
|
Dabsan S, Twito G, Biadsy S, Igbaria A. Less is better: various means to reduce protein load in the endoplasmic reticulum. FEBS J 2025; 292:976-989. [PMID: 38865586 PMCID: PMC11880973 DOI: 10.1111/febs.17201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/08/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
The endoplasmic reticulum (ER) is an important organelle that controls the intracellular and extracellular environments. The ER is responsible for folding almost one-third of the total protein population in the eukaryotic cell. Disruption of ER-protein folding is associated with numerous human diseases, including metabolic disorders, neurodegenerative diseases, and cancer. During ER perturbations, the cells deploy various mechanisms to increase the ER-folding capacity and reduce ER-protein load by minimizing the number of substrates entering the ER to regain homeostasis. These mechanisms include signaling pathways, degradation mechanisms, and other processes that mediate the reflux of ER content to the cytosol. In this review, we will discuss the recent discoveries of five different ER quality control mechanisms, including the unfolded protein response (UPR), ER-associated-degradation (ERAD), pre-emptive quality control, ER-phagy and ER to cytosol signaling (ERCYS). We will discuss the roles of these processes in decreasing ER-protein load and inter-mechanism crosstalk.
Collapse
Affiliation(s)
- Salam Dabsan
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Gal Twito
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Suma Biadsy
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael
| | - Aeid Igbaria
- Department of Life SciencesBen‐Gurion University of the NegevBeer ShevaIsrael
| |
Collapse
|
20
|
Acosta-Alvear D, Harnoss JM, Walter P, Ashkenazi A. Homeostasis control in health and disease by the unfolded protein response. Nat Rev Mol Cell Biol 2025; 26:193-212. [PMID: 39501044 DOI: 10.1038/s41580-024-00794-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 02/27/2025]
Abstract
Cells rely on the endoplasmic reticulum (ER) to fold and assemble newly synthesized transmembrane and secretory proteins - essential for cellular structure-function and for both intracellular and intercellular communication. To ensure the operative fidelity of the ER, eukaryotic cells leverage the unfolded protein response (UPR) - a stress-sensing and signalling network that maintains homeostasis by rebalancing the biosynthetic capacity of the ER according to need. The metazoan UPR can also redirect signalling from cytoprotective adaptation to programmed cell death if homeostasis restoration fails. As such, the UPR benefits multicellular organisms by preserving optimally functioning cells while removing damaged ones. Nevertheless, dysregulation of the UPR can be harmful. In this Review, we discuss the UPR and its regulatory processes as a paradigm in health and disease. We highlight important recent advances in molecular and mechanistic understanding of the UPR that enable greater precision in designing and developing innovative strategies to harness its potential for therapeutic gain. We underscore the rheostatic character of the UPR, its contextual nature and critical open questions for its further elucidation.
Collapse
Affiliation(s)
| | - Jonathan M Harnoss
- Department of General, Visceral, Thoracic and Transplant Surgery, University Hospital Giessen, Giessen, Germany
| | - Peter Walter
- Altos Labs, Inc., Bay Area Institute of Science, Redwood City, CA, USA.
| | - Avi Ashkenazi
- Research Oncology, Genentech, Inc., South San Francisco, CA, USA.
| |
Collapse
|
21
|
Liu X, Zhou D, Su Y, Liu H, Su Q, Shen T, Zhang M, Mi X, Zhang Y, Yue S, Zhang Z, Wang D, Tan X. PDIA4 targets IRE1α/sXBP1 to alleviate NLRP3 inflammasome activation and renal tubular injury in diabetic kidney disease. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167645. [PMID: 39743023 DOI: 10.1016/j.bbadis.2024.167645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
The role of ER stress in the pathogenesis of diabetic kidney diseases (DKD) remains unclear. We employed bioinformatics to identify the UPR pathway activation, inflammation, and programmed cell death patterns in diabetic tubules. Levels of IRE1α/sXBP1 signaling, NLRP3 inflammasome activity and pyroptosis in tubular cells under high glucose conditions were measured. IRE1α knockdown was used to determine its role in glucose-triggered activation of the NLRP3 inflammasome and pyroptosis. PDIA4 overexpression and silencing were used to assess its impact on the IRE1α/sXBP1 pathway. The dynamic interaction among PDIA4, GRP78, and IRE1α under high glucose were analyzed using immunoprecipitation and crosslinking assays. In STZ-induced and db/db mouse models of DKD, the regulatory role of PDIA4 on IRE1α/sXBP1 signaling and diabetic tubular inflammation and injury were evaluated. Our study showed that IRE1α/sXBP1, NLRP3 inflammasome, and pyroptosis are activated in the renal tubules of DKD patients. Induction of IRE1α pathway mediated the glucose-triggered activation of the NLRP3 inflammasome and pyroptosis. Moreover, overexpression of PDIA4 decreased the activation of IRE1α/sXBP1 under high glucose conditions. High glucose leads to the release of GRP78 from IRE1α and an increased interaction between IRE1α and PDIA4. In mouse models of DKD, overexpressing PDIA4 mitigated diabetic tubular injury and inflammation, marked by decreased IRE1α/sXBP1 and NLRP3 inflammasome. In conclusion, our findings demonstrate that high glucose triggers NLRP3 inflammasome and pyroptosis via the IRE1α/sXBP1 pathway in renal tubular cells. Overexpression of PDIA4 suppresses IRE1α signaling by binding to its oligomeric form, implying a promising therapeutic intervention for DKD.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Donghui Zhou
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Yu Su
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Hongling Liu
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Qiuyue Su
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Tianyu Shen
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Mianzhi Zhang
- Dongfang Hospital of Beijing University of Chinese medicine, Beijing, China
| | - Xue Mi
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Yuying Zhang
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Shijing Yue
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Zhujun Zhang
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China
| | - Dekun Wang
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China.
| | - Xiaoyue Tan
- Department of Pathology, School of Medicine, Nankai University, Tianjin, China.
| |
Collapse
|
22
|
Liu Z, Liu Q, Zeng A, Song L. Regulatory function of endoplasmic reticulum stress in colorectal cancer: Mechanism, facts, and perspectives. Int Immunopharmacol 2025; 147:114024. [PMID: 39764998 DOI: 10.1016/j.intimp.2025.114024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/29/2025]
Abstract
Colorectal cancer (CRC) is an exceedingly common and profoundly impactful malignancy of the digestive system, posing a grave threat to human health. Endoplasmic reticulum stress (ERS) is an intracellular biological reaction that mobilizes the unfolded protein response (UPR) to tackling dysregulation in protein homeostasis. This process subtly modulates the cell to either restore normal cellular function or steer it towards apoptosis. The high metabolic demands of CRC cells sculpt a rigorous tumor microenvironment (TME), compelling CRC cells to experience ERS. Adaptive responses induced by mild ERS furnish the necessary conditions for the survival of CRC cells, whereas the cell death mechanisms triggered by sustained ERS could be considered a prospective strategy for cancer therapy. Considering the complex regulation of ERS in cancer development, this article offers a comprehensive review of the molecular mechanisms through which ERS influences CRC fate. It provides crucial insights for exploring the role of ERS in the occurrence and progression of CRC, laying a new theoretical foundation for devising precise therapeutic strategies targeting ERS. Furthermore, by synthesizing extensive clinical and preclinical studies, we delve into therapeutic strategies targeting ERS, including the potential of targeting ERS in immunotherapy, the utilization of native compounds, advancements in proteasome inhibitors, and the potential synergies of these strategies with traditional chemotherapy agents and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Zihan Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anqi Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Academy of Chinese Medicine Sciences, Sichuan Institute for Translational Chinese Medicine, Chengdu, Sichuan 610041, China.
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
23
|
Shen C, Zhang Q. Forsythiaside A alleviates myocardial injury in streptozotocin-induced diabetes via endoplasmic reticulum stress-NLRP3 inflammasome pathway. Int Immunopharmacol 2025; 147:113956. [PMID: 39742724 DOI: 10.1016/j.intimp.2024.113956] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/16/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025]
Abstract
The aim of this study was to evaluate for the effects of forsythiaside A (FA) on myocardial injury in streptozotocin (STZ)-induced diabetes mice. Blood glucose (BG), serum triglycerides (TG), lactate dehydrogenase (LDH), creatine kinase isoenzyme (CK-MB), cardiac troponin (cTnI), malondialdehyde (MDA), superoxide dismutase (SOD) levels were detected in STZ mice. The structure and function of heart was observed via cardiac ultrasound. Cytokine levels in mouse serum and heart were detected using enzyme-linked immunosorbent assay (ELISA) as well as TG, LDH, CK-MB, cTnI, MDA and SOD in high glucose (Glu) induced H9c2 cells. Western blot detection of the expression of endoplasmic reticulum stress-related TXNIP/NLRP3 inflammasome pathways (GRP78, PERK, P-PERK, EIF-α, P-EIF-α, XBP1, ATF6, TXNIP and NLRP3) in SCD mice and LCG induced H9c2 cells. Endoplasmic reticulum stress activator tunicamycin (TM) was used to validate the above pathway for FA. It was also found that FA had protective effects on myocardial injury in STZ mice via restored heart function, improved cardiac pathological changes and suppressed inflammatory response as well as in Glu induced H9c2 cells. In conclusion, FA alleviated myocardial injury in diabetes via endoplasmic reticulum stress-NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Chuanpu Shen
- Anhui Provincial laboratory of inflammatory and immunity disease, Anhui Institute of Innovative Drugs, School of pharmacy, Anhui Medical University, Hefei 230032, PR China
| | - Qing Zhang
- Department of Cardiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, PR China.
| |
Collapse
|
24
|
Wu D, Eeda V, Maria Z, Rawal K, Wang A, Herlea-Pana O, Undi RB, Lim HY, Wang W. Targeting IRE1α improves insulin sensitivity and thermogenesis and suppresses metabolically active adipose tissue macrophages in male obese mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.07.17.603931. [PMID: 39071288 PMCID: PMC11275733 DOI: 10.1101/2024.07.17.603931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Overnutrition engenders the expansion of adipose tissue and the accumulation of immune cells, in particular, macrophages, in the adipose tissue, leading to chronic low-grade inflammation and insulin resistance. In obesity, several proinflammatory subpopulations of adipose tissue macrophages (ATMs) identified hitherto include the conventional "M1-like" CD11C-expressing ATM and the newly discovered metabolically activated CD9-expressing ATM; however, the relationship among ATM subpopulations is unclear. The ER stress sensor inositol-requiring enzyme 1α (IRE1α) is activated in the adipocytes and immune cells under obesity. It is unknown whether targeting IRE1α is capable of reversing insulin resistance and obesity and modulating the metabolically activated ATMs. We report that pharmacological inhibition of IRE1α RNase significantly ameliorates insulin resistance and glucose intolerance in male mice with diet-induced obesity. IRE1α inhibition also increases thermogenesis and energy expenditure, and hence protects against high fat diet-induced obesity. Our study shows that the "M1-like" CD11c+ ATMs are largely overlapping with but yet non-identical to CD9+ ATMs in obese white adipose tissue. Notably, IRE1α inhibition diminishes the accumulation of obesity-induced metabolically activated ATMs and "M1-like" ATMs, resulting in the curtailment of adipose inflammation and ensuing reactivation of thermogenesis, without augmentation of the alternatively activated M2 macrophage population. Our findings suggest the potential of targeting IRE1α for the therapeutic treatment of insulin resistance and obesity.
Collapse
Affiliation(s)
- Dan Wu
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, Alabama, 35233, United States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Venkateswararao Eeda
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Zahra Maria
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Komal Rawal
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Audrey Wang
- Indian Springs School, 190 Woodward Dr, Pelham, Alabama 35124
| | - Oana Herlea-Pana
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Ram Babu Undi
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Hui-Ying Lim
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, Alabama, 35233, United States
- Department of Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| | - Weidong Wang
- Department of Genetics, Heersink School of Medicine, UAB Comprehensive Diabetes Center, University of Alabama at Birmingham, 1918 University Blvd, Birmingham, Alabama, 35233, United States
- Department of Medicine, Division of Endocrinology, The University of Oklahoma Health Science Center, 941 Stanton L. Young Boulevard, Oklahoma City, Oklahoma 73104, United States
| |
Collapse
|
25
|
Spector L, Subramanian N. Revealing the dance of NLRP3: spatiotemporal patterns in inflammasome activation. IMMUNOMETABOLISM (COBHAM, SURREY) 2025; 7:e00053. [PMID: 39816134 PMCID: PMC11731036 DOI: 10.1097/in9.0000000000000053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/01/2024] [Indexed: 01/18/2025]
Abstract
The nucleotide-binding domain, leucine-rich repeat, and pyrin domain containing-protein 3 (NLRP3) inflammasome is a multiprotein complex that plays a critical role in the innate immune response to both infections and sterile stressors. Dysregulated NLRP3 activation has been implicated in a variety of autoimmune and inflammatory diseases, including cryopyrin-associated periodic fever syndromes, diabetes, atherosclerosis, Alzheimer's disease, inflammatory bowel disease, and cancer. Consequently, fine-tuning NLRP3 activity holds significant therapeutic potential. Studies have implicated several organelles, including mitochondria, lysosomes, the endoplasmic reticulum (ER), the Golgi apparatus, endosomes, and the centrosome, in NLRP3 localization and inflammasome assembly. However, reports of conflict and many factors regulating interactions between NLRP3 and subcellular organelles remain unknown. This review synthesizes the current understanding of NLRP3 spatiotemporal dynamics, focusing on recent literature that elucidates the roles of subcellular localization and organelle stress in NLRP3 signaling and its crosstalk with other innate immune pathways converging at these organelles.
Collapse
Affiliation(s)
- Lauren Spector
- Institute for Systems Biology, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Naeha Subramanian
- Institute for Systems Biology, Seattle, WA, USA
- Department of Immunology, University of Washington, Seattle, WA, USA
| |
Collapse
|
26
|
Khan MS, Tak J, Kim YS, Lee SG, Lee EB, Kim SG. Chronic hyperglycemia induces hepatocyte pyroptosis via Gα 12/Gα 13-associated endoplasmic reticulum stress: Effect of pharmacological intervention. Life Sci 2025; 360:123180. [PMID: 39561875 DOI: 10.1016/j.lfs.2024.123180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/17/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024]
Abstract
AIMS Hyperglycemia induces pathophysiological changes. Endoplasmic reticulum (ER) stress with Gα12 overexpression may promote hepatocyte death. This study investigated whether sustained hyperglycemia triggers ER stress-associated pyroptosis and fibrosis in the liver alongside an overexpression of Gα12, and examined the potential link with VEGF-A levels. MAIN METHODS Mice were subjected to a high-fat diet (60 kcal% fat) with streptozotocin (50 mg/kg body weight, three consecutive times, between 12-13th weeks). AZ2 (a functional Gα12 inhibitor) was treated at 10 mg/kg body weight (5 times/week, 3 weeks). Immunoblotting and immunohistochemistry analyses were performed. KEY FINDINGS Hepatic Gα12/Gα13 were overexpressed in the diabetic mice. The following proteins downstream from the Gα12 axis were upregulated: PGC1α, PPARα, and SIRT1. Sustained hyperglycemia promoted ER stress marker levels. Histopathological and biochemical assays showed large-sized lipid droplet accumulation, hepatocyte degeneration, and damage as blood transaminase activities increased. Moreover, the diabetic condition increased IL-1β, caspase-1, and NLRP3 levels, which were supportive of pyroptosis. Consistently, the intensities of Masson's trichrome, collagen-1A1, α-SMA, vimentin, and fibronectin all increased. VEGF-A and VEGFR2 levels also increased in the liver and/or sera. The levels of hepatic pigment epithelial-derived factor (PEDF), a physiological antagonist of VEGF-A, decreased with its reciprocal increase in serum. These events were reversed by AZ2 treatment, supporting the role of Gα12 in hyperglycemic stress in the liver. SIGNIFICANCE Chronic hyperglycemia causes hepatic pyroptosis and fibrosis related to ER stress with Gα12/Gα13 and VEGF overexpression, which may be overcome by AZ2 treatments.
Collapse
Affiliation(s)
- Muhammad Sohaib Khan
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Jihoon Tak
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do 10326, Republic of Korea
| | - Yun Seok Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sang Gil Lee
- Center of Research and Development, A Pharma Inc, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Eun Byul Lee
- Center of Research and Development, A Pharma Inc, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang-si, Gyeonggi-do 10326, Republic of Korea.
| |
Collapse
|
27
|
Liu M, Yuan M, Ma Y, Wang J, Cheng X, Shi Y, Shang J, He M, Bai L, Du L, Tang H. Wild-Type and rtA181T/sW172* Mutant Strains of Hepatitis B Virus Drive Hepatocarcinogenesis via Distinct GRP78-Mediated ER Stress Pathways. J Med Virol 2025; 97:e70151. [PMID: 39749680 DOI: 10.1002/jmv.70151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/14/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025]
Abstract
Glucose-regulated protein 78 kDa (GRP78), a key marker of endoplasmic reticulum stress (ERS), is upregulated in hepatocellular carcinoma (HCC) tissues, but its role in hepatitis B virus (HBV)-induced tumorigenesis remains unclear. This study aimed to investigate the contribution of GRP78 to HBV-associated tumor development and explore the ERS pathways involved. The results showed that increased GRP78 expression in patients with HBV-related HCC was associated with a poor prognosis within the first 2 years following diagnosis. Furthermore, using wild-type HBV strain and the oncogenic HBV rtA181T/sW172* mutant, this study demonstrated that the HBV-induced GRP78 expression correlated with elevated reactive oxygen species (ROS) levels. Moreover, GRP78 expression enhanced hepatocyte proliferation and resistance to apoptosis. In wild-type HBV-infected hepatocytes, GRP78 suppressed apoptosis by inhibiting the PERK/p38 pathway. In contrast, the HBV rtA181T/sW172* mutation led to increased GRP78 expression and inhibition of cell apoptosis through activation of the IRE-1α/XBP1/BCL-2 pathway. In conclusion, GRP78 plays a pivotal role in HBV-induced hepatocarcinogenesis by modulating distinct ERS pathways. Targeting these pathways may aid in the therapeutic management of HBV-associated hepatocarcinogenesis.
Collapse
Affiliation(s)
- Miao Liu
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Man Yuan
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yuanji Ma
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Jiayi Wang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Xing Cheng
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Ying Shi
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jin Shang
- Liver Transplantation Center and HBP Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Min He
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lang Bai
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Lingyao Du
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
28
|
Tak J, Kim YS, Kim SG. Roles of X-box binding protein 1 in liver pathogenesis. Clin Mol Hepatol 2025; 31:1-31. [PMID: 39355873 PMCID: PMC11791611 DOI: 10.3350/cmh.2024.0441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/06/2024] [Accepted: 09/27/2024] [Indexed: 10/03/2024] Open
Abstract
The prevalence of drug-induced liver injury (DILI) and viral liver infections presents significant challenges in modern healthcare and contributes to considerable morbidity and mortality worldwide. Concurrently, metabolic dysfunctionassociated steatotic liver disease (MASLD) has emerged as a major public health concern, reflecting the increasing rates of obesity and leading to more severe complications such as fibrosis and hepatocellular carcinoma. X-box binding protein 1 (XBP1) is a distinct transcription factor with a basic-region leucine zipper structure, whose activity is regulated by alternative splicing in response to disruptions in endoplasmic reticulum (ER) homeostasis and the unfolded protein response (UPR) activation. XBP1 interacts with a key signaling component of the highly conserved UPR and is critical in determining cell fate when responding to ER stress in liver diseases. This review aims to elucidate the emerging roles and molecular mechanisms of XBP1 in liver pathogenesis, focusing on its involvement in DILI, viral liver infections, MASLD, fibrosis/cirrhosis, and liver cancer. Understanding the multifaceted functions of XBP1 in these liver diseases offers insights into potential therapeutic strategies to restore ER homeostasis and mitigate liver damage.
Collapse
Affiliation(s)
- Jihoon Tak
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Korea
| | - Yun Seok Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine, Seoul, Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Korea
| | - Sang Geon Kim
- College of Pharmacy and Integrated Research Institute for Drug Development, Dongguk University-Seoul, Goyang, Korea
| |
Collapse
|
29
|
Nong WJ, Tong XY, Ouyang JM. Comparison of Endoplasmic Reticulum Stress and Pyroptosis Induced by Pathogenic Calcium Oxalate Monohydrate and Physiologic Calcium Oxalate Dihydrate Crystals in HK-2 Cells: Insights into Kidney Stone Formation. Cells 2024; 13:2070. [PMID: 39768161 PMCID: PMC11674083 DOI: 10.3390/cells13242070] [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: 11/23/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Endoplasmic reticulum stress (ERS) can activate pyroptosis through CHOP and TXNIP; however, the correlation between this process and the formation of kidney stones has not been reported. The purpose is to investigate the effects of calcium oxalate monohydrate (COM) and calcium oxalate dihydrate (COD) on ERS and pyroptosis in HK-2 cells and to explore the formation mechanism of calcium oxalate stones. HK-2 cells were injured by 3 μm COM and COD. COM and COD significantly upregulated the expression levels of GRP78, CHOP, TXNIP, and pyroptosis-related proteins (NLRP3, caspase-1, GSDMD-N, and IL-1β). Fluorescence colocalization revealed that COM induced pyroptosis by inducing the interaction between TXNIP and NLRP3. Both COM and COD crystals can induce ERS and pyroptosis in HK-2 cells. COM induces the interaction with NLRP3 by the upregulation of CHOP and TXNIP and then promotes pyroptosis, while COD only promotes pyroptosis by the upregulation of CHOP. The cytotoxicity and the ability of COM to promote crystal adhesion and aggregation are higher than COD, suggesting that COM is more dangerous for calcium oxalate kidney stone formation.
Collapse
Affiliation(s)
| | | | - Jian-Ming Ouyang
- Institute of Biomineralization and Lithiasis Research, College of Chemistry and Materials Science, Jinan University, Guangzhou 510632, China
| |
Collapse
|
30
|
Han N, Chang XY, Yuan ZL, Wang YZ. Expression and correlation analysis of silent information regulator 1 (SIRT1), sterol regulatory element-binding protein-1 (SREBP1), and pyroptosis factor in gestational diabetes mellitus. J Matern Fetal Neonatal Med 2024; 37:2311809. [PMID: 38326276 DOI: 10.1080/14767058.2024.2311809] [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: 09/20/2023] [Accepted: 01/24/2024] [Indexed: 02/09/2024]
Abstract
BACKGROUND AND AIM Globally, the prevalence of gestational diabetes mellitus (GDM) is rising each year, yet its pathophysiology is still unclear. To shed new light on the pathogenesis of gestational diabetes mellitus and perhaps uncover new therapeutic targets, this study looked at the expression levels and correlations of SIRT1, SREBP1, and pyroptosis factors like NLRP3, Caspase-1, IL-1, and IL-18 in patients with GDM. METHODS This study involved a comparative analysis between two groups. The GDM group consisted of 50 GDM patients and the control group included 50 pregnant women with normal pregnancies. Detailed case data were collected for all participants. We utilized real-time quantitative PCR and Western Blot techniques to assess the expression levels of SIRT1 and SREBP1 in placental tissues from both groups. Additionally, we employed an enzyme-linked immunosorbent assay to measure the serum levels of SIRT1, SREBP1, and pyroptosis factors, namely NLRP3, Caspase-1, IL-1β, and IL-18, in the patients of both groups. Subsequently, we analyzed the correlations between these factors and clinical. RESULTS The results showed that there were significantly lower expression levels of SIRT1 in both GDM group placental tissue and serum compared to the control group (p < 0.01). In contrast, the expression of SREBP1 was significantly higher in the GDM group than in the control group (p < 0.05). Additionally, the serum levels of NLRP3, Caspase-1, IL-1β, and IL-18 were significantly elevated in the GDM group compared to the control group (p < 0.01). The expression of SIRT1 exhibited negative correlations with the expression of FPG, OGTT-1h, FINS, HOMA-IR, SREBP1, IL-1β, and IL-18. However, there was no significant correlation between SIRT1 expression and OGTT-2h, NLRP3, or Caspase-1. On the other hand, the expression of SREBP1 was positively correlated with the expression of IL-1β, Caspase-1, and IL-18, but has no apparent correlation with NLRP3. CONCLUSIONS Low SIRT1 levels and high SREBP1 levels in placental tissue and serum, coupled with elevated levels of pyroptosis factors NLRP3, Caspase-1, IL-1β, and IL-18 in serum, may be linked to the development of gestational diabetes mellitus. Furthermore, these three factors appear to correlate with each other in the pathogenesis of GDM, offering potential directions for future research and therapeutic strategies.
Collapse
Affiliation(s)
- Ning Han
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xin-Yuan Chang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Zi-Li Yuan
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yi-Zhan Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
31
|
Payea MJ, Dar SA, Anerillas C, Martindale JL, Belair C, Munk R, Malla S, Fan J, Piao Y, Yang X, Rehman A, Banskota N, Abdelmohsen K, Gorospe M, Maragkakis M. Senescence suppresses the integrated stress response and activates a stress-remodeled secretory phenotype. Mol Cell 2024; 84:4454-4469.e7. [PMID: 39481386 PMCID: PMC11585442 DOI: 10.1016/j.molcel.2024.10.003] [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: 11/20/2023] [Revised: 07/30/2024] [Accepted: 10/01/2024] [Indexed: 11/02/2024]
Abstract
Senescence is a state of indefinite cell-cycle arrest associated with aging, cancer, and age-related diseases. Here, we find that translational deregulation, together with a corresponding maladaptive integrated stress response (ISR), is a hallmark of senescence that desensitizes senescent cells to stress. We present evidence that senescent cells maintain high levels of eIF2α phosphorylation, typical of ISR activation, but translationally repress production of the stress response activating transcription factor 4 (ATF4) by ineffective bypass of the inhibitory upstream open reading frames (uORFs). Surprisingly, ATF4 translation remains inhibited even after acute proteotoxic and amino acid starvation stressors, resulting in a highly diminished stress response. We also find that stress augments the senescence-associated secretory phenotype with sustained remodeling of inflammatory factors expression that is suppressed by non-uORF carrying ATF4 mRNA expression. Our results thus show that senescent cells possess a unique response to stress, which entails an increase in their inflammatory profile.
Collapse
Affiliation(s)
- Matthew J Payea
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA.
| | - Showkat A Dar
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Carlos Anerillas
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jennifer L Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Cedric Belair
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Sulochan Malla
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Jinshui Fan
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Yulan Piao
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Xiaoling Yang
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Abid Rehman
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Nirad Banskota
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Manolis Maragkakis
- Laboratory of Genetics and Genomics, National Institute on Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
32
|
Soh LJ, Lee SY, Roebuck MM, Wong PF. Unravelling the interplay between ER stress, UPR and the cGAS-STING pathway: Implications for osteoarthritis pathogenesis and treatment strategy. Life Sci 2024; 357:123112. [PMID: 39378929 DOI: 10.1016/j.lfs.2024.123112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/20/2024] [Accepted: 10/03/2024] [Indexed: 10/10/2024]
Abstract
Osteoarthritis (OA) is a debilitating chronic degenerative disease affecting the whole joint organ leading to pain and disability. Cellular stress and injuries trigger inflammation and the onset of pathophysiological changes ensue after irreparable damage and inability to resolve inflammation, impeding the completion of the healing process. Extracellular matrix (ECM) degradation leads to dysregulated joint tissue metabolism. The reparative effort induces the proliferation of hypertrophic chondrocytes and matrix protein synthesis. Aberrant protein synthesis leads to endoplasmic reticulum (ER) stress and chondrocyte apoptosis with consequent cartilage matrix loss. These events in a vicious cycle perpetuate inflammation, hindering the restoration of normal tissue homeostasis. Recent evidence suggests that inflammatory responses and chondrocyte apoptosis could be caused by the activation of the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) signalling axis in response to DNA damage. It has been reported that there is a crosstalk between ER stress and cGAS-STING signalling in cellular senescence and other diseases. Based on recent evidence, this review discusses the role of ER stress, Unfolded Protein Response (UPR) and cGAS-STING pathway in mediating inflammatory responses in OA.
Collapse
Affiliation(s)
- Li-Jen Soh
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Siam-Yee Lee
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Margaret M Roebuck
- Department of Musculoskeletal & Ageing Science, Institute of Life Course & Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK; Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool L3 9TA, UK
| | - Pooi-Fong Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia.
| |
Collapse
|
33
|
Chen X, Yang Y, Zhou Z, Yu H, Zhang S, Huang S, Wei Z, Ren K, Jin Y. Unraveling the complex interplay between Mitochondria-Associated Membranes (MAMs) and cardiovascular Inflammation: Molecular mechanisms and therapeutic implications. Int Immunopharmacol 2024; 141:112930. [PMID: 39146786 DOI: 10.1016/j.intimp.2024.112930] [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: 07/04/2024] [Revised: 07/26/2024] [Accepted: 08/10/2024] [Indexed: 08/17/2024]
Abstract
Cardiovascular diseases (CVDs) represent a significant public health concern because of their associations with inflammation, oxidative stress, and abnormal remodeling of the heart and blood vessels. In this review, we discuss the intricate interplay between mitochondria-associated membranes (MAMs) and cardiovascular inflammation, highlighting their role in key cellular processes such as calcium homeostasis, lipid metabolism, oxidative stress management, and ERS. We explored how these functions impact the pathogenesis and progression of various CVDs, including myocardial ischemia-reperfusion injury, atherosclerosis, diabetic cardiomyopathy, cardiovascular aging, heart failure, and pulmonary hypertension. Additionally, we examined current therapeutic strategies targeting MAM-related pathways and proteins, emphasizing the potential of MAMs as therapeutic targets. Our review aims to provide new insights into the mechanisms of cardiovascular inflammation and propose novel therapeutic approaches to improve cardiovascular health outcomes.
Collapse
Affiliation(s)
- Xing Chen
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Yang Yang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Zheng Zhou
- Department of Geriatric Endocrinology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Haihan Yu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Shuwei Zhang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Siyuan Huang
- Department of Translational Medicine Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Ziqing Wei
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China.
| |
Collapse
|
34
|
Shang B, Dong Y, Feng B, Zhao J, Wang Z, Crans DC, Yang X. Combination therapy enhances efficacy and overcomes toxicity of metal-based anti-diabetic agent. Br J Pharmacol 2024; 181:4214-4228. [PMID: 38965763 DOI: 10.1111/bph.16485] [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: 11/12/2023] [Revised: 05/09/2024] [Accepted: 05/22/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND AND PURPOSE Metal-based therapeutic agents are limited by the required concentration of metal-based agents. Hereby, we determined if combination with 17β-oestradiol (E2) could reduce such levels and the therapy still be effective in type 2 diabetes mellitus (T2DM). EXPERIMENTAL APPROACH The metal-based agent (vanadyl acetylacetonate [VAC])- 17β-oestradiol (E2) combination is administered using the membrane-permeable graphene quantum dots (GQD), the vehicle, to form the active GQD-E2-VAC complexes, which was characterized by fluorescence spectra, infrared spectra and X-ray photoelectron spectroscopy. In db/db type 2 diabetic mice, the anti-diabetic effects of GQD-E2-VAC complexes were evaluated using blood glucose levels, oral glucose tolerance test (OGTT), serum insulin levels, homeostasis model assessment (homeostasis model assessment of insulin resistance [HOMA-IR] and homeostasis model assessment of β-cell function [HOMA-β]), histochemical assays and western blot. KEY RESULTS In diabetic mice, GQD-E2-VAC complex had comprehensive anti-diabetic effects, including control of hyperglycaemia, improved insulin sensitivity, correction of hyperinsulinaemia and prevention of β-cell loss. Co-regulation of thioredoxin interacting protein (TXNIP) activation by the combination of metal complex and 17β-oestradiol contributed to the enhanced anti-diabetic effects. Furthermore, a potent mitochondrial protective antioxidant, coniferaldehyde, significantly potentiates the protective effects of GQD-E2-VAC complexes. CONCLUSION AND IMPLICATIONS A metal complex-E2 combinatorial approach achieved simultaneously the protection of β cells and insulin enhancement at an unprecedented low dose, similar to the daily intake of dietary metals in vitamin supplements. This study demonstrates the positive effects of combination and multi-modal therapies towards type 2 diabetes treatment.
Collapse
Affiliation(s)
- Bing Shang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Yaqiong Dong
- Institute of Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Bo Feng
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Jingyan Zhao
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Zhi Wang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
| | - Debbie C Crans
- Department of Chemistry and Cell and Molecular Biology Program, College of Natural Science, Colorado State University, Fort Collins, Colorado, USA
| | - Xiaoda Yang
- State Key Laboratory of Natural and Biomimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China
- SATCM Key Laboratory of Compound Drug Detoxification, Peking University Health Science Center, Beijing, China
| |
Collapse
|
35
|
Noh SG, Kim HW, Kim S, Chung KW, Jung YS, Yoon JH, Yu BP, Lee J, Chung HY. Senoinflammation as the underlying mechanism of aging and its modulation by calorie restriction. Ageing Res Rev 2024; 101:102503. [PMID: 39284417 DOI: 10.1016/j.arr.2024.102503] [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: 07/04/2024] [Revised: 08/25/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024]
Abstract
Senoinflammation is characterized by an unresolved low-grade inflammatory process that affects multiple organs and systemic functions. This review begins with a brief overview of the fundamental concepts and frameworks of senoinflammation. It is widely involved in the aging of various organs and ultimately leads to progressive systemic degeneration. Senoinflammation underlying age-related inflammation, is causally related to metabolic dysregulation and the formation of senescence-associated secretory phenotype (SASP) during aging and age-related diseases. This review discusses the biochemical evidence and molecular biology data supporting the concept of senoinflammation and its regulatory processes, highlighting the anti-aging and anti-inflammatory effects of calorie restriction (CR). Experimental data from CR studies demonstrated effective suppression of various pro-inflammatory cytokines and chemokines, lipid accumulation, and SASP during aging. In conclusion, senoinflammation represents the basic mechanism that creates a microenvironment conducive to aging and age-related diseases. Furthermore, it serves as a potential therapeutic target for mitigating aging and age-related diseases.
Collapse
Affiliation(s)
- Sang Gyun Noh
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Hyun Woo Kim
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Seungwoo Kim
- Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Ki Wung Chung
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Young-Suk Jung
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Jeong-Hyun Yoon
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Byung Pal Yu
- Department of Physiology, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jaewon Lee
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea.
| | - Hae Young Chung
- Research Institute for Drug Development, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea; Department of Pharmacy, College of Pharmacy, Pusan National University, 2 Busandaehak-ro 63beon-gil, Geumjeong-gu, Busan 46241, Republic of Korea.
| |
Collapse
|
36
|
Fernández L, Kong CS, Alkhoury M, Tryfonos M, Brighton PJ, Rawlings TM, Muter J, Gori MS, Leirós CP, Lucas ES, Brosens JJ, Ramhorst R. The endoplasmic reticulum protein HSPA5/BiP is essential for decidual transformation of human endometrial stromal cells. Sci Rep 2024; 14:25992. [PMID: 39472623 PMCID: PMC11522507 DOI: 10.1038/s41598-024-76241-z] [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: 06/11/2024] [Accepted: 10/11/2024] [Indexed: 11/02/2024] Open
Abstract
Decidualization denotes the process of inflammatory reprogramming of endometrial stromal cells (EnSC) into specialized decidual cells (DC). During this process, EnSC are subjected to endoplasmic reticulum (ER) stress as well as acute cellular senescence. Both processes contribute to the proinflammatory mid-luteal implantation window and their dysregulation has been implicated in reproductive failure. Here, we evaluated the link between ER stress, decidual differentiation and senescence. In-silico analysis identified HSPA5 gene, codifying the ER chaperone BiP, as a potentially critical regulator of cell fate divergence of decidualizing EnSC into anti-inflammatory DC and pro-inflammatory senescent decidual cells (snDC). Knockdown of HSPA5 in primary EnSC resulted both in decreased expression of DC marker genes and attenuated induction of senescence associated β-galactosidase activity, a marker of snDC. Stalling of the decidual reaction upon HSPA5 knockdown was apparent at 8 days of differentiation and was preceded by the upregulation of ER stress associated proteins IRE1α and PERK. Further, HSPA5 knockdown impaired colony-forming unit activity of primary EnSC, indicative of loss of cellular plasticity. Together, our results point to a key role for HSPA5/BiP in decidual transformation of EnSCs and highlight the importance of constraining ER stress levels during this process.
Collapse
Affiliation(s)
- Laura Fernández
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales IQUIBICEN, Buenos Aires, Argentina
- Warwick Medical School, Division of Biomedical Sciences, University of Warwick, Coventry, UK
| | - Chow-Seng Kong
- Warwick Medical School, Division of Biomedical Sciences, University of Warwick, Coventry, UK
| | - Majd Alkhoury
- Tommy's National Centre for Miscarriage Research, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
| | - Maria Tryfonos
- Warwick Medical School, Division of Biomedical Sciences, University of Warwick, Coventry, UK
| | - Paul J Brighton
- Warwick Medical School, Division of Biomedical Sciences, University of Warwick, Coventry, UK
| | - Thomas M Rawlings
- Warwick Medical School, Division of Biomedical Sciences, University of Warwick, Coventry, UK
| | - Joanne Muter
- Warwick Medical School, Division of Biomedical Sciences, University of Warwick, Coventry, UK
| | - Maria Soledad Gori
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales IQUIBICEN, Buenos Aires, Argentina
| | - Claudia Pérez Leirós
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales IQUIBICEN, Buenos Aires, Argentina
| | - Emma S Lucas
- Warwick Medical School, Division of Biomedical Sciences, University of Warwick, Coventry, UK
- Faculty of Health, University of Sheffield, Sheffield, UK
| | - Jan J Brosens
- Warwick Medical School, Division of Biomedical Sciences, University of Warwick, Coventry, UK
- Tommy's National Centre for Miscarriage Research, University Hospitals Coventry and Warwickshire NHS Trust, Coventry, CV2 2DX, UK
| | - Rosanna Ramhorst
- CONICET, Universidad de Buenos Aires, Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales IQUIBICEN, Buenos Aires, Argentina.
- School of Sciences, University of Buenos Aires, IQUIBICEN-CONICET, Int. Guiraldes 2160, Ciudad Universitaria, Pabellón 2 Piso 4, C1428EHA, Buenos Aires, Argentina.
| |
Collapse
|
37
|
Capolupo I, Miranda MR, Musella S, Di Sarno V, Manfra M, Ostacolo C, Bertamino A, Campiglia P, Ciaglia T. Exploring Endocannabinoid System: Unveiling New Roles in Modulating ER Stress. Antioxidants (Basel) 2024; 13:1284. [PMID: 39594426 PMCID: PMC11591047 DOI: 10.3390/antiox13111284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/28/2024] Open
Abstract
The endoplasmic reticulum (ER) is the organelle mainly involved in maintaining cellular homeostasis and driving correct protein folding. ER-dependent defects or dysfunctions are associated with the genesis/progression of several pathological conditions, including cancer, inflammation, and neurodegenerative disorders, that are directly or indirectly correlated to a wide set of events collectively named under the term "ER stress". Despite the recent increase in interest concerning ER activity, further research studies are needed to highlight all the mechanisms responsible for ER failure. In this field, recent discoveries paved the way for the comprehension of the strong interaction between ER stress development and the endocannabinoid system. The activity of the endocannabinoid system is mediated by the activation of cannabinoid receptors (CB), G protein-coupled receptors that induce a decrease in cAMP levels, with downstream anti-inflammatory effects. CB activation drives, in most cases, the recovery of ER homeostasis through the regulation of ER stress hallmarks PERK, ATF6, and IRE1. In this review, we focus on the CB role in modulating ER stress, with particular attention to the cellular processes leading to UPR activation and oxidative stress response extinguishment, and to the mechanisms underlying natural cannabinoids' modulation of this complex cellular machine.
Collapse
Affiliation(s)
- Ilaria Capolupo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, Fisciano, 84084 Salerno, Italy; (I.C.); (M.R.M.); (S.M.); (V.D.S.); (C.O.); (A.B.); (P.C.)
- PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, 84084 Salerno, Italy
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, Fisciano, 84084 Salerno, Italy; (I.C.); (M.R.M.); (S.M.); (V.D.S.); (C.O.); (A.B.); (P.C.)
- PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, 84084 Salerno, Italy
- NBFC—National Biodiversity Future Center, 90133 Palermo, Italy
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, Fisciano, 84084 Salerno, Italy; (I.C.); (M.R.M.); (S.M.); (V.D.S.); (C.O.); (A.B.); (P.C.)
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II, Fisciano, 84084 Salerno, Italy; (I.C.); (M.R.M.); (S.M.); (V.D.S.); (C.O.); (A.B.); (P.C.)
| | - Michele Manfra
- Department of Health Science, University of Basilicata, Viale dell’Ateneo Lucano 10, 85100 Potenza, Italy;
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, Fisciano, 84084 Salerno, Italy; (I.C.); (M.R.M.); (S.M.); (V.D.S.); (C.O.); (A.B.); (P.C.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, Fisciano, 84084 Salerno, Italy; (I.C.); (M.R.M.); (S.M.); (V.D.S.); (C.O.); (A.B.); (P.C.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, Fisciano, 84084 Salerno, Italy; (I.C.); (M.R.M.); (S.M.); (V.D.S.); (C.O.); (A.B.); (P.C.)
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, Fisciano, 84084 Salerno, Italy; (I.C.); (M.R.M.); (S.M.); (V.D.S.); (C.O.); (A.B.); (P.C.)
| |
Collapse
|
38
|
Jing G, Jo S, Shalev A. A novel class of oral, non-immunosuppressive, beta cell-targeting, TXNIP-inhibiting T1D drugs is emerging. Front Endocrinol (Lausanne) 2024; 15:1476444. [PMID: 39429740 PMCID: PMC11486709 DOI: 10.3389/fendo.2024.1476444] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 09/19/2024] [Indexed: 10/22/2024] Open
Abstract
Diabetes treatment options have improved dramatically over the last 100 years, however, close to 2 million individuals in the U.S. alone live with type 1 diabetes (T1D) and are still dependent on multiple daily insulin injections and/or continuous insulin infusion with a pump to stay alive and no oral medications are available. After decades of focusing on immunosuppressive/immunomodulatory approaches for T1D, it has now become apparent that at least after disease onset, this by itself may not be sufficient, and in order to be effective, therapies need to also address beta cell health. This Perspective article discusses the emergence of such a beta cell-targeting, novel class of oral T1D drugs targeting thioredoxin-interacting protein (TXNIP) and some very recent advances in this field that start to address this unmet medical need. It thereby focuses on repurposing of the antihypertensive drug, verapamil found to non-specifically inhibit TXNIP and on TIX100, a new chemical entity specifically developed as an oral anti-diabetic drug to inhibit TXNIP. Both have shown striking anti-diabetic effects in preclinical studies. Verapamil has also proven to be beneficial in adults and children with recent onset T1D, while TIX100 has just been cleared by the U.S. Food and Drug Administration (FDA) to proceed to clinical trials. Taken together, we propose that such non-immunosuppressive, adjunctive therapies to insulin, alone or in combination with immune modulatory approaches, are critical in order to achieve effective and durable disease-modifying treatments for T1D.
Collapse
Affiliation(s)
| | | | - Anath Shalev
- Comprehensive Diabetes Center and Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
39
|
İpek E, Ahsan U, Özsoy B, Ekren Aşıcı GS, Tatar M, Özpilavcı BN, Epikmen ET, Özsoy ŞY, Khamseh EK, Petracci M. Endoplasmic reticulum stress and associated apoptosis are linked with the pathogenesis of white striping in broiler breast muscles. Poult Sci 2024; 103:104103. [PMID: 39094495 PMCID: PMC11345576 DOI: 10.1016/j.psj.2024.104103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/05/2024] [Accepted: 07/11/2024] [Indexed: 08/04/2024] Open
Abstract
White striping (WS) that appears as white stripes parallel to the muscle fibrils is an emerging growth-related abnormality of broiler breast meat. The pathomechanism of this defect has not been fully understood despite intensive studies over the past decade. In the present study, endoplasmic reticulum (ER) stress and its associated apoptotic pathways were investigated to elucidate the potential role of these pathways in the development of WS. To this end, a total of 60 Pectoralis major (Pm) muscle samples were collected from 55-d-old Ross 308 male broiler chickens according to the severity of gross WS lesions (normal, mild, and severe). Histopathological and molecular analyses were conducted to evaluate the lesions and genes involved in the ER stress and related apoptosis. All the Pm samples, both with and without macroscopic WS lesions, showed varying degrees of myodegenerative lesions. Molecular analysis revealed that the transcript abundances of many components related to protein kinase R-like ER kinase (PERK) and inositol-requiring enzyme type 1 (IRE-1) signals of the ER stress response were significantly greater in severely WS-affected breast tissues compared to their mildly affected and normal counterparts. Similarly, the transcript abundances of apoptotic markers related to both signaling pathways were significantly greater in severe WS lesions than those of mildly affected and normal Pm tissues. Besides these, a significant increase in caspase-3 transcript abundance was seen in severe WS lesions in comparison with mild WS and normal breast muscles. Findings of this study suggest that ER stress response and its related apoptotic pathways are possibly activated in the breast muscle of broiler chickens with severe WS lesions. Based on these findings, it is speculated that ER stress-mediated apoptosis occupies a central role in the progression of WS in broiler chickens.
Collapse
Affiliation(s)
- Emrah İpek
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın 09016, Türkiye
| | - Umair Ahsan
- Department of Plant and Animal Production, Burdur Vocational School of Food, Agriculture and Livestock, Burdur Mehmet Akif Ersoy University, Burdur 15030, Türkiye; Center for Agriculture, Livestock and Food Research Burdur Mehmet Akif Ersoy University, Burdur 15030, Türkiye
| | - Bülent Özsoy
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın 09016, Türkiye
| | - Gamze Sevri Ekren Aşıcı
- Department of Biochemistry, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın 09016, Türkiye
| | - Musa Tatar
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Kastamonu University, Kastamonu, Türkiye
| | - Beyza Nur Özpilavcı
- Department of Biochemistry, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın 09016, Türkiye
| | - Erkmen Tuğrul Epikmen
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın 09016, Türkiye
| | - Şule Yurdagül Özsoy
- Department of Pathology, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın 09016, Türkiye
| | - Ehsan Karimiyan Khamseh
- Department of Animal Nutrition and Nutritional Diseases, Faculty of Veterinary Medicine, Aydın Adnan Menderes University, Aydın 09016, Türkiye
| | - Massimiliano Petracci
- Department of Agricultural and Food Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy.
| |
Collapse
|
40
|
Berkel C. Inducers and Inhibitors of Pyroptotic Death of Granulosa Cells in Models of Premature Ovarian Insufficiency and Polycystic Ovary Syndrome. Reprod Sci 2024; 31:2972-2992. [PMID: 39026050 PMCID: PMC11438836 DOI: 10.1007/s43032-024-01643-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024]
Abstract
Granulosa cells (GCs), the largest cell population and primary source of steroid hormones in the ovary, are the important somatic ovarian components. They have critical roles in folliculogenesis by supporting oocyte, facilitating its growth, and providing a microenvironment suitable for follicular development and oocyte maturation, thus having essential functions in maintaining female fertility and in reproductive health in general. Pyroptotic death of GCs and associated inflammation have been implicated in the pathogenesis of several reproductive disorders in females including Premature Ovarian Insufficiency (POI) and Polycystic Ovary Syndrome (PCOS). Here, I reviewed factors, either intrinsic or extrinsic, that induce or inhibit pyroptosis in GCs in various models of these disorders, both in vitro and in vivo, and also covered associated molecular mechanisms. Most of these studied factors influence NLRP3 inflammasome- and GSDMD (Gasdermin D)-mediated pyroptosis in GCs, compared to other inflammasomes and gasdermins (GSDMs). I conclude that a more complete mechanistic understanding of these factors in terms of GC pyroptosis is required to be able to develop novel strategies targeting inflammatory cell death in the ovary.
Collapse
Affiliation(s)
- Caglar Berkel
- Department of Molecular Biology and Genetics, Tokat Gaziosmanpasa University, Tokat, Türkiye.
| |
Collapse
|
41
|
Yu C, Zhang Z, Xiao L, Ai M, Qing Y, Zhang Z, Xu L, Yu OY, Cao Y, Liu Y, Song K. IRE1α pathway: A potential bone metabolism mediator. Cell Prolif 2024; 57:e13654. [PMID: 38736291 PMCID: PMC11471397 DOI: 10.1111/cpr.13654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 04/07/2024] [Accepted: 05/01/2024] [Indexed: 05/14/2024] Open
Abstract
Osteoblasts and osteoclasts collaborate in bone metabolism, facilitating bone development, maintaining normal bone density and strength, and aiding in the repair of pathological damage. Endoplasmic reticulum stress (ERS) can disrupt the intracellular equilibrium between osteoclast and osteoblast, resulting in dysfunctional bone metabolism. The inositol-requiring enzyme-1α (IRE1α) pathway-the most conservative unfolded protein response pathway activated by ERS-is crucial in regulating cell metabolism. This involvement encompasses functions such as inflammation, autophagy, and apoptosis. Many studies have highlighted the potential roles of the IRE1α pathway in osteoblasts, chondrocytes, and osteoclasts and its implication in certain bone-related diseases. These findings suggest that it may serve as a mediator for bone metabolism. However, relevant reviews on the role of the IRE1α pathway in bone metabolism remain unavailable. Therefore, this review aims to explore recent research that elucidated the intricate roles of the IRE1α pathway in bone metabolism, specifically in osteogenesis, chondrogenesis, osteoclastogenesis, and osteo-immunology. The findings may provide novel insights into regulating bone metabolism and treating bone-related diseases.
Collapse
Affiliation(s)
- Chengbo Yu
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Zhixiang Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Li Xiao
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Mi Ai
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Ying Qing
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Zhixing Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Lianyi Xu
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Ollie Yiru Yu
- Faculty of DentistryThe University of Hong KongHong Kong SARChina
| | - Yingguang Cao
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| | - Yong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Frontier Science Center for Immunology and Metabolism, and the Institute for Advanced StudiesWuhan UniversityWuhanHubeiChina
| | - Ke Song
- Department of Stomatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Prosthodontics and Implantology, School of Stomatology, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and RegenerationWuhanChina
| |
Collapse
|
42
|
Liu LJ, O'Donoghue AJ, Caffrey CR. The proteasome as a drug target for treatment of parasitic diseases. ADVANCES IN PARASITOLOGY 2024; 126:53-96. [PMID: 39448194 DOI: 10.1016/bs.apar.2024.04.001] [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: 10/26/2024]
Abstract
The proteasome is a proteolytically active molecular machine comprising many different protein subunits. It is essential for growth and survival in eukaryotic cells and has long been considered a drug target. Here, we summarize the biology of the proteasome, the early research relating to the development of specific proteasome inhibitors (PIs) for treatment of various cancers, and their translation and eventual evolution as exciting therapies for parasitic diseases. We also highlight the development and adaptation of technologies that have allowed for a deep understanding of the idiosyncrasies of individual parasite proteasomes, as well as the preclinical and clinical advancement of PIs with remarkable therapeutic indices.
Collapse
Affiliation(s)
- Lawrence J Liu
- Center for Discovery and Innovation in Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, United States; Department of Chemistry and Biochemistry, University of California, San Diego, CA, United States.
| | - Anthony J O'Donoghue
- Center for Discovery and Innovation in Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, United States
| | - Conor R Caffrey
- Center for Discovery and Innovation in Diseases (CDIPD), Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, CA, United States
| |
Collapse
|
43
|
Holst-Hansen T, Nielsen PY, Jensen MH, Mandrup-Poulsen T, Trusina A. Tipping-point transition from transient to persistent inflammation in pancreatic islets. NPJ Syst Biol Appl 2024; 10:102. [PMID: 39266581 PMCID: PMC11393080 DOI: 10.1038/s41540-024-00427-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/19/2024] [Indexed: 09/14/2024] Open
Abstract
Type 2 diabetes (T2D) is associated with a systemic increase in the pro-inflammatory cytokine IL-1β. While transient exposure to low IL-1β concentrations improves insulin secretion and β-cell proliferation in pancreatic islets, prolonged exposure leads to impaired insulin secretion and collective β-cell death. IL-1 is secreted locally by islet-resident macrophages and β-cells; however, it is unknown if and how the two opposing modes may emerge at single islet level. We investigated the duality of IL-1β with a quantitative in silico model of the IL-1 regulatory network in pancreatic islets. We find that the network can produce either transient or persistent IL-1 responses when induced by pro-inflammatory and metabolic cues. This suggests that the duality of IL-1 may be regulated at the single islet level. We use two core feedbacks in the IL-1 regulation to explain both modes: First, a fast positive feedback in which IL-1 induces its own production through the IL-1R/IKK/NF-κB pathway. Second, a slow negative feedback where NF-κB upregulates inhibitors acting at different levels along the IL-1R/IKK/NF-κB pathway-IL-1 receptor antagonist and A20, among others. A transient response ensues when the two feedbacks are balanced. When the positive feedback dominates over the negative, islets transit into the persistent inflammation mode. Consistent with several observations, where the size of islets was implicated in its inflammatory state, we find that large islets and islets with high density of IL-1β amplifying cells are more prone to transit into persistent IL-1β mode. Our results are likely not limited to IL-1β but are general for the combined effect of multiple pro-inflammatory cytokines and chemokines. Generalizing complex regulations in terms of two feedback mechanisms of opposing nature and acting on different time scales provides a number of testable predictions. Taking islet architecture and cellular heterogeneity into consideration, further dynamic monitoring and experimental validation in actual islet samples will be crucial to verify the model predictions and enhance its utility in clinical applications.
Collapse
Affiliation(s)
| | - Pernille Yde Nielsen
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Lyngby, Denmark
| | - Mogens H Jensen
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark.
| | - Thomas Mandrup-Poulsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Ala Trusina
- Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
44
|
Umashankar B, Eliasson L, Ooi CY, Kim KW, Shaw JAM, Waters SA. Beyond insulin: Unraveling the complex interplay of ER stress, oxidative damage, and CFTR modulation in CFRD. J Cyst Fibros 2024; 23:842-852. [PMID: 38897882 DOI: 10.1016/j.jcf.2024.06.004] [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: 03/04/2024] [Revised: 05/10/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
CF-related diabetes (CFRD) is a prevalent comorbidity in people with Cystic Fibrosis (CF), significantly impacting morbidity and mortality rates. This review article critically evaluates the current understanding of CFRD molecular mechanisms, including the role of CFTR protein, oxidative stress, unfolded protein response (UPR) and intracellular communication. CFRD manifests from a complex interplay between exocrine pancreatic damage and intrinsic endocrine dysfunction, further complicated by the deleterious effects of misfolded CFTR protein on insulin secretion and action. Studies indicate that ER stress and subsequent UPR activation play critical roles in both exocrine and endocrine pancreatic cell dysfunction, contributing to β-cell loss and insulin insufficiency. Additionally, oxidative stress and altered calcium flux, exacerbated by CFTR dysfunction, impair β-cell survival and function, highlighting the significance of antioxidant pathways in CFRD pathogenesis. Emerging evidence underscores the importance of exosomal microRNAs (miRNAs) in mediating inflammatory and stress responses, offering novel insights into CFRD's molecular landscape. Despite insulin therapy remaining the cornerstone of CFRD management, the variability in response to CFTR modulators underscores the need for personalized treatment approaches. The review advocates for further research into non-CFTR therapeutic targets, emphasizing the need to address the multifaceted pathophysiology of CFRD. Understanding the intricate mechanisms underlying CFRD will pave the way for innovative treatments, moving beyond insulin therapy to target the disease's root causes and improve the quality of life for individuals with CF.
Collapse
Affiliation(s)
- Bala Umashankar
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Lena Eliasson
- Department of Clinical Sciences, Unit of Islet Cell Exocytosis, Lund University Diabetes Centre, Scania University Hospital, Malmö, Scania, Sweden
| | - Chee Y Ooi
- Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Department of Gastroenterology, Sydney Children's Hospital Randwick, NSW, Australia
| | - Ki Wook Kim
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Virology and Serology Division (SaViD), New South Wales Health Pathology, Prince of Wales Hospital, Randwick, NSW, Australia
| | - James A M Shaw
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Shafagh A Waters
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia; Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
45
|
Sak F, Sengul F, Vatansev H. The Role of Endoplasmic Reticulum Stress in Metabolic Diseases. Metab Syndr Relat Disord 2024; 22:487-493. [PMID: 38666441 DOI: 10.1089/met.2024.0013] [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: 09/13/2024] Open
Abstract
The endoplasmic reticulum (ER), the center of protein folding, also controls the cell's life-and-death signaling mechanisms. ER stress caused by unfolded or misfolded proteins leads to the activation of the unfolded protein response (UPR) in the cell. The UPR utilizes three main signaling pathways to restore disrupted ER homeostasis. These signaling pathways are protein kinase R-like endoplasmic reticulum kinase, inositol-requiring enzyme 1, and activating transcription factor 6. Studies have reported that ER stress (ERS) plays a role in the pathogenesis of metabolic disorders such as diabetes, obesity, atherosclerosis, and nonalcoholic liver disease. This review will briefly discuss the ERS response in these metabolic diseases.
Collapse
Affiliation(s)
- Firdevs Sak
- Faculty of Medicine, Department of Medical Biochemistry, University of Selçuk, Konya, Turkey
| | - Fatma Sengul
- Faculty of Pharmacy, Department of Biochemistry, University of Adiyaman, Adiyaman, Turkey
| | - Husamettin Vatansev
- Faculty of Medicine, Department of Medical Biochemistry, University of Selçuk, Konya, Turkey
| |
Collapse
|
46
|
Galli F, Bartolini D, Ronco C. Oxidative stress, defective proteostasis and immunometabolic complications in critically ill patients. Eur J Clin Invest 2024; 54:e14229. [PMID: 38676423 DOI: 10.1111/eci.14229] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/31/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024]
Abstract
Oxidative stress (OS) develops in critically ill patients as a metabolic consequence of the immunoinflammatory and degenerative processes of the tissues. These induce increased and/or dysregulated fluxes of reactive species enhancing their pro-oxidant activity and toxicity. At the same time, OS sustains its own inflammatory and immunometabolic pathogenesis, leading to a pervasive and vitious cycle of events that contribute to defective immunity, organ dysfunction and poor prognosis. Protein damage is a key player of these OS effects; it generates increased levels of protein oxidation products and misfolded proteins in both the cellular and extracellular environment, and contributes to forms DAMPs and other proteinaceous material to be removed by endocytosis and proteostasis processes of different cell types, as endothelial cells, tissue resident monocytes-macrophages and peripheral immune cells. An excess of OS and protein damage in critical illness can overwhelm such cellular processes ultimately interfering with systemic proteostasis, and consequently with innate immunity and cell death pathways of the tissues thus sustaining organ dysfunction mechanisms. Extracorporeal therapies based on biocompatible/bioactive membranes and new adsorption techniques may hold some potential in reducing the impact of OS on the defective proteostasis of patients with critical illness. These can help neutralizing reactive and toxic species, also removing solutes in a wide spectrum of molecular weights thus improving proteostasis and its immunometabolic corelates. Pharmacological therapy is also moving steps forward which could help to enhance the efficacy of extracorporeal treatments. This narrative review article explores the aspects behind the origin and pathogenic role of OS in intensive care and critically ill patients, with a focus on protein damage as a cause of impaired systemic proteostasis and immune dysfunction in critical illness.
Collapse
Affiliation(s)
- Francesco Galli
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Desirée Bartolini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Claudio Ronco
- Department of Medicine, International Renal Research Institute of Vicenza, University of Padova, San Bortolo Hospital Vicenza, Vicenza, Italy
| |
Collapse
|
47
|
Kim JH, Yu H, Kang JH, Hong EH, Kang MH, Seong M, Cho H, Shin YU. MicroRNA Regulation for Inflammasomes in High Glucose-Treated ARPE-19 Cells. J Ophthalmol 2024; 2024:3654690. [PMID: 39220230 PMCID: PMC11366061 DOI: 10.1155/2024/3654690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/22/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Purpose This study aimed to evaluate the expression of microRNAs (miRNAs) and inflammasomes in diabetes-induced retinal cells and to determine their role in the pathogenesis of diabetic retinopathy (DR). Methods To establish diabetes-induced cell models, ARPE-19 cells were treated with high glucose. The expression levels of five miRNAs (miR-185, miR-17, miR-20a, miR-15a, and miR-15b) were measured in high glucose-treated ARPE-19 cells using real-time quantitative polymerase chain reaction. Western blotting was performed to measure inflammasome expression in cellular models. miR-17 was selected as the target miRNA, and inflammasome expression was measured following the transfection of an miR-17 mimic into high glucose-treated ARPE-19 cells. Results In high glucose-treated ARPE-19 cells, miRNA expression was substantially downregulated, whereas that of inflammasome components was significantly increased. Following the transfection of the miR-17 mimic into high glucose-treated ARPE-19 cells, the levels of inflammasome components were significantly decreased. Conclusions This study investigated the relationship between miRNAs and inflammasomes in diabetes-induced cells using high glucose-treated ARPE-19 cells. These findings suggested that miR-17 suppresses inflammasomes, thereby reducing the subsequent inflammatory response and indicating that miRNAs and inflammasomes could serve as new therapeutic targets for DR.
Collapse
Affiliation(s)
- Ji Hong Kim
- Department of OphthalmologyHanyang University College of Medicine, Seoul, Republic of Korea
- Department of OphthalmologyHanyang University Seoul Hospital, Seoul, Republic of Korea
| | - Hyoseon Yu
- Department of OphthalmologyHanyang University College of Medicine, Seoul, Republic of Korea
| | - Ji Hye Kang
- Department of OphthalmologyHanyang University College of Medicine, Seoul, Republic of Korea
| | - Eun Hee Hong
- Department of OphthalmologyHanyang University College of Medicine, Seoul, Republic of Korea
- Department of OphthalmologyHanyang University Guri Hospital, Guri, Gyeonggi-do, Republic of Korea
- Hanyang Institute of Bioscience and BiotechnologyHanyang University, Seoul, Republic of Korea
| | - Min Ho Kang
- Department of OphthalmologyHanyang University College of Medicine, Seoul, Republic of Korea
- Department of OphthalmologyHanyang University Guri Hospital, Guri, Gyeonggi-do, Republic of Korea
| | - Mincheol Seong
- Department of OphthalmologyHanyang University College of Medicine, Seoul, Republic of Korea
- Department of OphthalmologyHanyang University Guri Hospital, Guri, Gyeonggi-do, Republic of Korea
- NOON Eye Clinic, Guri, Gyeonggi-do, Republic of Korea
| | - Heeyoon Cho
- Department of OphthalmologyHanyang University College of Medicine, Seoul, Republic of Korea
- Department of OphthalmologyHanyang University Guri Hospital, Guri, Gyeonggi-do, Republic of Korea
- NOON Eye Clinic, Guri, Gyeonggi-do, Republic of Korea
| | - Yong Un Shin
- Department of OphthalmologyHanyang University College of Medicine, Seoul, Republic of Korea
- Department of OphthalmologyHanyang University Guri Hospital, Guri, Gyeonggi-do, Republic of Korea
- Hanyang Institute of Bioscience and BiotechnologyHanyang University, Seoul, Republic of Korea
| |
Collapse
|
48
|
Hu Q, Zhang L, Tao Y, Xie S, Wang A, Luo C, Yang R, Shen Z, He B, Fang Y, Chen P. Semaglutide Ameliorates Hepatocyte Steatosis in a Cell Co-Culture System by Downregulating the IRE1α-XBP1-C/EBPα Signaling Pathway in Macrophages. Pharmacology 2024; 110:26-35. [PMID: 39089233 DOI: 10.1159/000540654] [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: 11/06/2023] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
INTRODUCTION Non-alcoholic fatty liver disease (NAFLD) is currently the most common type of chronic liver disease. Semaglutide is a glucose-lowering drug administered for the treatment of type 2 diabetes mellitus (T2DM) and is clinically effective in the treatment of NAFLD. X-box binding protein 1 (XBP1) is related to the pathogenesis of both NAFLD and T2DM. The aim of the present study was to demonstrate whether the underlying mechanism of semaglutide treatment for NAFLD is via downregulation of the inositol-requiring transmembrane kinase/endonuclease-1α (IRE1α)-XBP1-CCAAT/enhancer binding protein α (C/EBPα) signaling pathway in macrophages. METHODS In the present study, NAFLD cell modeling was induced by oleic acid (0.4 mm) and palmitic acid (0.2 mm). Hepatocytes (AML12) and macrophages (RAW264.7) were co-cultured in 6-well Transwell plates. Semaglutide (60 or 140 nm) was administrated for 24 h, while pioglitazone (2 μm) and toyocamycin (200 nm) were used as a positive control drug and a XBP1 inhibitor, respectively. Autophagy and apoptosis of AML12 cells were detected by transmission electron microscopy and Western blotting (WB). Hepatocyte steatosis was evaluated by adopting total intracellular triglyceride determination, analysis of the relative expression of proteins and genes associated with lipid metabolism and hepatocyte Oil red O staining. Detection of inflammation factors was conducted by ELISA and WB. To explore the underlying mechanism of NAFLD treatment with semaglutide, the relative expression of related proteins and genes were tested. RESULTS Our study demonstrated that semaglutide treatment improved autophagy and inhibited apoptosis of hepatocytes, while notably ameliorating steatosis of hepatocytes. In addition, inflammation was attenuated in the NAFLD cell co-culture model after semaglutide administration. Semaglutide also significantly reduced the protein and gene expression levels of the IRE1α-XBP1-C/EBPα signaling pathway in macrophages. CONCLUSION Semaglutide partially ameliorated NAFLD by downregulating the IRE1α-XBP1-C/EBPα signaling pathway in macrophages. These findings may provide a potential theoretical basis for semaglutide therapy for NAFLD.
Collapse
Affiliation(s)
- Qin Hu
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
- Department of Endocrinology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Li Zhang
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - YiTing Tao
- Department of Nephrology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - ShuangLin Xie
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - AiYun Wang
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - Caiying Luo
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - RenHua Yang
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - Zhiqiang Shen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - Bo He
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
| | - Yu Fang
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Peng Chen
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming, China
- College of Modern Biomedical Industry, Kunming Medical University, Kunming, China
| |
Collapse
|
49
|
Lo CW, Lii CK, Lin KS, Li CC, Liu KL, Yang YC, Chen HW. Luteolin, apigenin, and chrysin inhibit lipotoxicity-induced NLRP3 inflammasome activation and autophagy damage in macrophages by suppressing endoplasmic reticulum stress. ENVIRONMENTAL TOXICOLOGY 2024; 39:4120-4133. [PMID: 38654489 DOI: 10.1002/tox.24289] [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: 01/25/2024] [Revised: 03/14/2024] [Accepted: 03/31/2024] [Indexed: 04/26/2024]
Abstract
Lipotoxicity leads to numerous metabolic disorders such as nonalcoholic steatohepatitis. Luteolin, apigenin, and chrysin are three flavones with known antioxidant and anti-inflammatory properties, but whether they inhibit lipotoxicity-mediated NLRP3 inflammasome activation was unclear. To address this question, we used J774A.1 macrophages and Kupffer cells stimulated with 100 μM palmitate (PA) in the presence or absence of 20 μM of each flavone. PA increased p-PERK, p-IRE1α, p-JNK1/2, CHOP, and TXNIP as well as p62 and LC3-II expression and induced autophagic flux damage. Caspase-1 activation and IL-1β release were also noted after 24 h of exposure to PA. In the presence of the PERK inhibitor GSK2656157, PA-induced CHOP and TXNIP expression and caspase-1 activation were mitigated. Compared with PA treatment alone, Bcl-2 coupled to beclin-1 was elevated and autophagy was reversed by the JNK inhibitor SP600125. With luteolin, apigenin, and chrysin treatment, PA-induced ROS production, ER stress, TXNIP expression, autophagic flux damage, and apoptosis were ameliorated. Moreover, TXNIP binding to NLRP3 and IL-1β release in response to LPS/PA challenge were reduced. These results suggest that luteolin, apigenin, and chrysin protect hepatic macrophages against PA-induced NLRP3 inflammasome activation and autophagy damage by attenuating endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Chia-Wen Lo
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Chong-Kuei Lii
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Kuan-Shuan Lin
- Department of Nutrition, China Medical University, Taichung, Taiwan
| | - Chien-Chun Li
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Kai-Li Liu
- Department of Nutrition, Chung Shan Medical University, Taichung, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ya-Chen Yang
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung, Taiwan
| | - Haw-Wen Chen
- Department of Nutrition, China Medical University, Taichung, Taiwan
| |
Collapse
|
50
|
Ni L, Yang L, Lin Y. Recent progress of endoplasmic reticulum stress in the mechanism of atherosclerosis. Front Cardiovasc Med 2024; 11:1413441. [PMID: 39070554 PMCID: PMC11282489 DOI: 10.3389/fcvm.2024.1413441] [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: 05/01/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
The research progress of endoplasmic reticulum (ER) stress in atherosclerosis (AS) is of great concern. The ER, a critical cellular organelle, plays a role in important biological processes including protein synthesis, folding, and modification. Various pathological factors may cause ER stress, and sustained or excessive ER stress triggers the unfolded protein response, ultimately resulting in apoptosis and disease. Recently, researchers have discovered the importance of ER stress in the onset and advancement of AS. ER stress contributes to the occurrence of AS through different pathways such as apoptosis, inflammatory response, oxidative stress, and autophagy. Therefore, this review focuses on the mechanisms of ER stress in the development of AS and related therapeutic targets, which will contribute to a deeper understanding of the disease's pathogenesis and provide novel strategies for preventing and treating AS.
Collapse
Affiliation(s)
| | | | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|