1
|
Desai M, Gulati K, Agrawal M, Ghumra S, Sahoo PK. Stress granules: Guardians of cellular health and triggers of disease. Neural Regen Res 2026; 21:588-597. [PMID: 39995077 DOI: 10.4103/nrr.nrr-d-24-01196] [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] [Accepted: 01/15/2025] [Indexed: 02/26/2025] Open
Abstract
Stress granules are membraneless organelles that serve as a protective cellular response to external stressors by sequestering non-translating messenger RNAs (mRNAs) and regulating protein synthesis. Stress granules formation mechanism is conserved across species, from yeast to mammals, and they play a critical role in minimizing cellular damage during stress. Composed of heterogeneous ribonucleoprotein complexes, stress granules are enriched not only in mRNAs but also in noncoding RNAs and various proteins, including translation initiation factors and RNA-binding proteins. Genetic mutations affecting stress granule assembly and disassembly can lead to abnormal stress granule accumulation, contributing to the progression of several diseases. Recent research indicates that stress granule dynamics are pivotal in determining their physiological and pathological functions, with acute stress granule formation offering protection and chronic stress granule accumulation being detrimental. This review focuses on the multifaceted roles of stress granules under diverse physiological conditions, such as regulation of mRNA transport, mRNA translation, apoptosis, germ cell development, phase separation processes that govern stress granule formation, and their emerging implications in pathophysiological scenarios, such as viral infections, cancer, neurodevelopmental disorders, neurodegeneration, and neuronal trauma.
Collapse
Affiliation(s)
- Meghal Desai
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| | - Keya Gulati
- College of Science and Liberal Arts, New Jersey Institute of Technology, Newark, NJ, USA
| | - Manasi Agrawal
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| | - Shruti Ghumra
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| | - Pabitra K Sahoo
- Department of Biological Sciences, Rutgers University - Newark, Newark, NJ, USA
| |
Collapse
|
2
|
Mohan HM, Fernandez MG, Huang C, Lin R, Ryou JH, Seyfried D, Grotewold N, Whiteley AM, Barmada SJ, Basrur V, Mosalaganti S, Paulson HL, Sharkey LM. Endogenous retrovirus-like proteins recruit UBQLN2 to stress granules and alter their functional properties. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.620053. [PMID: 39484508 PMCID: PMC11527177 DOI: 10.1101/2024.10.24.620053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
The human genome is replete with sequences derived from foreign elements including endogenous retrovirus-like proteins of unknown function. Here we show that UBQLN2, a ubiquitin-proteasome shuttle factor implicated in neurodegenerative diseases, is regulated by the linked actions of two retrovirus-like proteins, RTL8 and PEG10. RTL8 confers on UBQLN2 the ability to complex with and regulate PEG10. PEG10, a core component of stress granules, drives the recruitment of UBQLN2 to stress granules under various stress conditions, but can only do so when RTL8 is present. Changes in PEG10 levels further remodel the kinetics of stress granule disassembly and overall composition by incorporating select extracellular vesicle proteins. Within stress granules, PEG10 forms virus-like particles, underscoring the structural heterogeneity of this class of biomolecular condensates. Together, these results reveal an unexpected link between pathways of cellular proteostasis and endogenous retrovirus-like proteins.
Collapse
|
3
|
Rao X, Li Z, Zhang Q, Lai Y, Liu J, Li L, Cheng H, Shen W, Sun D. α-Hederin induces paraptosis by targeting GPCRs to activate Ca 2+/MAPK signaling pathway in colorectal cancer. Cancer Med 2024; 13:e7202. [PMID: 38659391 PMCID: PMC11043672 DOI: 10.1002/cam4.7202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/03/2024] [Accepted: 04/06/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Non-apoptotic cell death is presently emerging as a potential direction to overcome the apoptosis resistance of cancer cells. In the current study, a natural plant agent α-hederin (α-hed) induces caspase-independent paraptotic modes of cell death. PURPOSE The present study is aimed to investigate the role of α-hed induces paraptosis and the associated mechanism of it. METHODS The cell proliferation was detected by CCK-8. The cytoplasm organelles were observed under electron microscope. Calcium (Ca2+) level was detected by flow cytometry. Swiss Target Prediction tool analyzed the potential molecule targets of α-hed. Molecular docking methods were used to evaluate binding abilities of α-hed with targets. The expressions of genes and proteins were analyzed by RT-qPCR, western blotting, immunofluorescence, and immunohistochemistry. Xenograft models in nude mice were established to evaluate the anticancer effects in vivo. RESULTS α-hed exerted significant cytotoxicity against a panel of CRC cell lines by inhibiting proliferation. Besides, it induced cytoplasmic vacuolation in all CRC cells. Electron microscopy images showed the aberrant dilation of endoplasmic reticulum and mitochondria. Both mRNA and protein expressions of Alg-2 interacting proteinX (Alix), the marker of paraptosis, were inhibited by α-hed. Besides, both Swiss prediction and molecular docking showed that the structure of α-hed could tightly target to GPCRs. GPCRs were reported to activate the phospholipase C (PLC)-β3/ inositol 1,4,5-trisphosphate receptor (IP3R)/ Ca2+/ protein kinase C alpha (PKCα) pathway, and we then found all proteins and mRNA expressions of PLCβ3, IP3R, and PKCα were increased by α-hed. After blocking the GPCR signaling, α-hed could not elevate Ca2+ level and showed less CRC cell cytotoxicity. MAPK cascade is the symbol of paraptosis, and we then demonstrated that α-hed activated MAPK cascade by elevating Ca2+ flux. Since non-apoptotic cell death is presently emerging as a potential direction to overcome chemo-drug resistance, we then found α-hed also induced paraptosis in 5-fluorouracil-resistant (5-FU-R) CRC cells, and it reduced the growth of 5-FU-R CRC xenografts. CONCLUSIONS Collectively, our findings proved α-hed as a promising candidate for inducing non-apoptotic cell death, paraptosis. It may overcome the resistance of apoptotic-based chemo-resistance in CRC.
Collapse
Affiliation(s)
- Xiwu Rao
- The First Clinical Medical College of Nanjing University of Chinese MedicineCollaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese MedicineNanjingChina
- Department of OncologyThe First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhouChina
- Guangzhou University of Chinese MedicineGuangzhouChina
- Postdoctoral Research Station of Guangzhou University of Chinese MedicineGuangzhouChina
| | - Ziwen Li
- The First Clinical Medical College of Nanjing University of Chinese MedicineCollaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese MedicineNanjingChina
| | - Qinchang Zhang
- The First Clinical Medical College of Nanjing University of Chinese MedicineCollaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese MedicineNanjingChina
| | - Yueyang Lai
- The First Clinical Medical College of Nanjing University of Chinese MedicineCollaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese MedicineNanjingChina
| | - Jianrong Liu
- Department of Infectious DiseaseNanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese MedicineNanjingChina
| | - Liu Li
- The First Clinical Medical College of Nanjing University of Chinese MedicineCollaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese MedicineNanjingChina
| | - Haibo Cheng
- The First Clinical Medical College of Nanjing University of Chinese MedicineCollaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese MedicineNanjingChina
| | - Weixing Shen
- The First Clinical Medical College of Nanjing University of Chinese MedicineCollaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese MedicineNanjingChina
| | - Dongdong Sun
- The First Clinical Medical College of Nanjing University of Chinese MedicineCollaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese MedicineNanjingChina
| |
Collapse
|
4
|
Duncan C, Mata J. Translation-complex profiling of fission yeast cells reveals dynamic rearrangements of scanning ribosomal subunits upon nutritional stress. Nucleic Acids Res 2022; 50:13011-13025. [PMID: 36478272 PMCID: PMC9825154 DOI: 10.1093/nar/gkac1140] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 12/13/2022] Open
Abstract
Control of mRNA translation is key for stress responses. Translation initiation is usually rate-limiting and, in eukaryotes, involves mRNA scanning by the small ribosomal subunit. Despite its importance, many aspects of translation in vivo have not been explored fully, especially at the transcriptome-wide level. A recent method termed translation-complex profiling (TCP-seq) allows transcriptome-wide views of scanning ribosomal subunits. We applied TCP-seq to nutritional stress in the fission yeast Schizosaccharomyces pombe. At initiation sites, we observed multiple complexes resembling those of mammals, and consistent with queuing of scanning subunits. In 5' UTRs, small subunit accumulations were common and may reflect impediments to scanning. A key mediator of stress responses in S. pombe is the Fil1 transcription factor, which is regulated translationally by a poorly-understood mechanism involving upstream Open Reading Frames (uORFs). TCP-seq data of fil1 shows that stress allows scanning subunits to by-pass specific uORFs and reach the fil1 coding sequence. The integration of these observations with reporter assays revealed that fil1 translational control is mediated by a combination of scanning reinitiation-repressive and permissive uORFs, and establishes fil1 as a model for uORF-mediated translational control. Altogether, our transcriptome-wide study reveals general and gene-specific features of translation in a model eukaryote.
Collapse
Affiliation(s)
| | - Juan Mata
- To whom correspondence should be addressed. Tel: +44 01223360467;
| |
Collapse
|
5
|
Hu HY, Liu YJ. Sequestration of cellular native factors by biomolecular assemblies: Physiological or pathological? BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119360. [PMID: 36087810 DOI: 10.1016/j.bbamcr.2022.119360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 09/01/2022] [Accepted: 09/02/2022] [Indexed: 06/15/2023]
Abstract
In addition to native-state structures, biomolecules often form condensed supramolecular assemblies or cellular membraneless organelles that are critical for cell life. These biomolecular assemblies, generally including liquid-like droplets (condensates) and amyloid-like aggregates, can sequester or recruit their interacting partners, so as to either modulate various cellular behaviors or even cause disorders. This review article summarizes recent advances in the sequestration of native factors by biomolecular assemblies and discusses their potential consequences on cellular function, homeostasis, and disease pathology.
Collapse
Affiliation(s)
- Hong-Yu Hu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, PR China.
| | - Ya-Jun Liu
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai 200031, PR China; University of Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
6
|
Wang K, Wang Z, Wang Z, Xie X, Zang L, Wang L, Che F. Stellera chamaejasme L. extracts in the treatment of glioblastoma cell lines: Biological verification based on a network pharmacology approach. Front Oncol 2022; 12:962970. [PMID: 36059675 PMCID: PMC9428724 DOI: 10.3389/fonc.2022.962970] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/28/2022] [Indexed: 11/21/2022] Open
Abstract
Background Stellera chamaejasme L (RXLD) has been demonstrated with good clinical effects and medicinal value in the treatment of cancer in vivo and in vitro. Specifically, RXLD can eliminate aggregation accumulation, which is depicted as a vital characteristic feature of intracranial tumors. The potential pharmacological mechanisms of anti-glioblastoma (GBM) have not been adequately identified. Methods The 3D structures of the chemical ingredients in RXLD were imported into the PharmMapper database to construct the pharmacophore models. The gene targets of GBM were obtained from databases. The pharmacophore-targets network and the protein-protein interactions (PPI) were constructed using the String database and were visualized by using Cytoscape. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) enrichment analyses were conducted using Bioconductor software. Cytoscape visualized the relationship of pathways and candidate genes to screen for key target genes. Software packages PyMOL, AutoDock, and Vina acquired the molecular docking results. In vitro experiments were undertaken to characterize RXLD extracts’ effects on A172 cell line proliferation, viability, apoptosis, cell cycle, cell wound healing, cell migration, reactive oxygen species generation, and mitochondrial membrane potential. The expression of core genes in the related pathways was detected by Western blotting. Results We identified 216 potential targets associated with GBM. The core components in RXLD were neochamaejasmin A, wikstrol A, isochamaejasmin, chamaejasmine, and subtoxin A. The undertaken GO enrichment analysis revealed that oxidative stress, cell proliferation, cell cycle, cell invasion, and cell migration were involved in the biological processes. The KEGG enrichment analysis revealed that the crucial pathway was MAPK pathway, while HRAS, PRKCB, MAPK9, CCND1, and TP53 were distributed in core locations. A total of seven RXLD pharmacophores demonstrated strong spontaneous docking activities with MAPK9. In vitro assays indicated that RXLD can induce apoptosis, block the cell cycle in the G2/M and S phases, inhibit cell migration via the Wnt/β-catenin pathway, and inhibited p62/Nrf2 pathway. Conclusions We speculate that the RAS/MAPK pathway might be an upstream pathway through which the RXLD exerts its anti-GBM effects and might be able to regulate further the Wnt/β-catenin, the oxidative stress, and the ferroptosis pathways.
Collapse
Affiliation(s)
- Kaiyue Wang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Neurology, Linyi People’s Hospital, Shandong University, Linyi, China
| | - Zengyong Wang
- Central Laboratory, Linyi People’s Hospital, Shandong University, Linyi, China
- Key Laboratory of Neurophysiology, Linyi, China
- Key Laboratory of Tumor Biology, Linyi, China
| | - Zhiqiang Wang
- Central Laboratory, Linyi People’s Hospital, Shandong University, Linyi, China
- Key Laboratory of Neurophysiology, Linyi, China
- Key Laboratory of Tumor Biology, Linyi, China
| | - Xiaoli Xie
- Central Laboratory, Linyi People’s Hospital, Shandong University, Linyi, China
- Key Laboratory of Neurophysiology, Linyi, China
- Key Laboratory of Tumor Biology, Linyi, China
| | - Lanlan Zang
- Central Laboratory, Linyi People’s Hospital, Shandong University, Linyi, China
- Key Laboratory of Neurophysiology, Linyi, China
- Key Laboratory of Tumor Biology, Linyi, China
- Clinical Pharmacological Laboratory, Linyi People’s Hospital, Shandong University, Linyi, China
- *Correspondence: Lanlan Zang, ; Lijuan Wang, ; Fengyuan Che,
| | - Lijuan Wang
- Central Laboratory, Linyi People’s Hospital, Shandong University, Linyi, China
- Key Laboratory of Neurophysiology, Linyi, China
- Key Laboratory of Tumor Biology, Linyi, China
- Department of Hematology, Linyi People’s Hospital, Shandong University, Linyi, China
- *Correspondence: Lanlan Zang, ; Lijuan Wang, ; Fengyuan Che,
| | - Fengyuan Che
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Neurology, Linyi People’s Hospital, Shandong University, Linyi, China
- Central Laboratory, Linyi People’s Hospital, Shandong University, Linyi, China
- Key Laboratory of Neurophysiology, Linyi, China
- Key Laboratory of Tumor Biology, Linyi, China
- *Correspondence: Lanlan Zang, ; Lijuan Wang, ; Fengyuan Che,
| |
Collapse
|