1
|
Buchholz HE, Martin SA, Dorweiler JE, Radtke CM, Knier AS, Beans NB, Manogaran AL. Hsp70 chaperones, Ssa1 and Ssa2, limit poly(A) binding protein aggregation. Mol Biol Cell 2025; 36:ar66. [PMID: 40202836 DOI: 10.1091/mbc.e25-01-0027] [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: 04/11/2025] Open
Abstract
Molecular chaperones play a central role in maintaining protein homeostasis. The highly conserved Hsp70 family of chaperones have major functions in folding of nascent peptides, protein refolding, and protein aggregate disassembly. In yeast, loss of two Hsp70 proteins, Ssa1 and Ssa2, is associated with decreased cellular growth and shortened lifespan. While heterologous or mutant temperature-sensitive proteins form anomalous large cytoplasmic inclusions in ssa1Δssa2Δ strains, it is unclear how endogenous wild-type proteins behave and are regulated in the presence of limiting Hsp70s. Using the wild-type yeast Poly A binding protein (Pab1), which is involved in mRNA binding and forms stress granules (SGs) upon heat shock, Pab1 forms large inclusions in approximately half of ssa1Δssa2Δ cells in the absence of stress. Overexpression of Ssa1, Hsp104, and Sis1 almost completely limits the formation of these large inclusions in ssa1Δssa2Δ, suggesting that excess Ssa1, Hsp104, and Sis1 can each compensate for the lower levels of Ssa proteins. Upon heat shock, SGs also form in cells whether large Pab1 inclusions are present or not. Surprisingly, cells containing only SGs disassemble faster than wild type, whereas cells with both large inclusions disassemble slower albeit completely. We suspect that disassembly of these large inclusions is linked to the elevated heat shock response and elevated Hsp104 and Sis1 levels in ssa1Δssa2Δ strains. We also observed that wild-type cultures grown to saturation also form large Pab1-GFP inclusions. These inclusions can be partially rescued by overexpression of Ssa1. Taken together, our data suggest that Hsp70 not only plays a role in limiting unwanted protein aggregation in normal cells, but as cells age, the depletion of active Hsp70 possibly underlies the age-related aggregation of endogenous proteins.
Collapse
Affiliation(s)
- Hannah E Buchholz
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53201-1881
| | - Sean A Martin
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53201-1881
| | - Jane E Dorweiler
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53201-1881
| | - Claire M Radtke
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53201-1881
| | - Adam S Knier
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53201-1881
| | - Natalia B Beans
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53201-1881
| | - Anita L Manogaran
- Department of Biological Sciences, Marquette University, Milwaukee, WI 53201-1881
| |
Collapse
|
2
|
Nabariya DK, Knüpfer LM, Hartwich P, Killian MS, Centler F, Krauß S. Transcriptomic analysis of intracellular RNA granules and small extracellular vesicles: Unmasking their overlap in a cell model of Huntington's disease. Mol Cell Probes 2025; 81:102026. [PMID: 40090627 DOI: 10.1016/j.mcp.2025.102026] [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: 07/05/2024] [Revised: 03/13/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Huntington's disease (HD) arises from the abnormal expansion of a CAG repeat in the HTT gene. The mutant CAG repeat triggers aberrant RNA-protein interactions and translates into toxic aggregate-prone polyglutamine protein. These aberrant RNA-protein ineractions also seed the formation of cytoplasmic liquid-like granules, such as stress granules. Emerging evidence demonstrates that granules formed via liquid-liquid phase separation can mature into gel-like inclusions that persist within the cell and may act as precursor to aggregates that occur in patients' tissue. Thus, deregulation of RNA granules is an important component of neurodegeneration. Interestingly, both the formation of intracellular membrane-less organelles like stress granules and the secretion of small extracellular vesicles (sEVs) increase upon stress and under disease conditions. sEVs are lipid membrane-bound particles that are secreted from all cell types and may participate in the spreading of misfolded proteins and aberrant RNA-protein complexes across the central nervous system in neurodegenerative diseases like HD. In this study, we performed a comparative transcriptomic analysis of sEVs and RNA granules in an HD model. RNA granules and sEVs were isolated from an inducible HD cell model. Both sEVs and RNA granules were isolated from induced (HD) and non-induced (control) cells and analyzed by RNA sequencing. Our comparative analysis between the transcriptomics data of HD RNA granules and sEVs showed that: (I) intracellular RNA granules and extracellular RNA vesicles share content, (II) several non-coding RNAs translocate to RNA granules, and (III) the composition of RNA granules and sEVs is affected in HD cells. Our data showing common transcripts in intracellular RNA granules and extracellular sEVs suggest that formation of RNA granules and sEV loading may be related. Moreover, we found a high abundance of lncRNAs in both control and HD samples, with several transcripts under REST regulation, highlighting their potential role in HD pathogenesis and selective incorporation into sEVs. The transcriptome cargo of RNA granules or sEVs may serve as a source for diagnostic strategies. For example, disease-specific RNA-signatures of sEVs can serve as biomarker of central nervous system diseases. Therefore, we compared our dataset to transcriptomic data from HD patient sEVs in blood. However, our data suggest that the cell-type specific signature of sEV-secreted RNAs as well as their high variability may make it difficult to detect these biomarkers in blood.
Collapse
Affiliation(s)
- Deepti Kailash Nabariya
- Human and Neurobiology, Department of Chemistry and Biology, University of Siegen, Siegen, Germany
| | - Lisa Maria Knüpfer
- Human and Neurobiology, Department of Chemistry and Biology, University of Siegen, Siegen, Germany
| | - Patrick Hartwich
- Chemistry and Structure of Novel Materials, Department of Chemistry and Biology, University of Siegen, Siegen, Germany
| | - Manuela S Killian
- Chemistry and Structure of Novel Materials, Department of Chemistry and Biology, University of Siegen, Siegen, Germany
| | - Florian Centler
- Bioinformatics, School of Life Sciences, University of Siegen, Siegen, Germany
| | - Sybille Krauß
- Human and Neurobiology, Department of Chemistry and Biology, University of Siegen, Siegen, Germany.
| |
Collapse
|
3
|
Ma G, Ruan X, Yang B, Li N, Su D, Sun S, Chen S, Xu K, Ying Z, Wang H. Abnormal regulation of membrane-less organelles contributes to profilin1-associated ALS. J Biol Chem 2025:110259. [PMID: 40409555 DOI: 10.1016/j.jbc.2025.110259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/25/2025] Open
Abstract
Profilin 1 (PFN1) is a key cytoskeletal protein that regulates actin dynamics by incorporating monomeric actin into linear filaments. PFN1 deletion or mutations have been linked to numerous neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS). However, the contribution of PFN1 to neurodegenerative pathologies is poorly understood. Recent studies have implicated the role of aberrant cellular membrane-less organelles (MLOs) in neurodegenerative pathogenesis. Here, we demonstrate that PFN1 is involved in the assembly of MLOs, including Cajal bodies and Stress granules. Specifically, depletion of PFN1 leads to abnormal Cajal body accumulation and accelerated maturation into a gel-like state, consequently dysregulating snRNP biogenesis and impairing pre-mRNA splicing efficiency in both neuronal and non-neuronal cells. Similarly, we show that PFN1 knockdown accelerates the assembly of Stress granules in stressed cells. Furthermore, we demonstrate that the ALS-linked PFN1-C71G mutant exhibits a loss of function in the context of MLO biogenesis. We further reveal that the PFN1 deficiency-induced Cajal body dysregulation, but not Stress granule assembly, is caused by cellular actin filament depolymerization. Importantly, the actin filament agonist CN04 rescues Cajal body properties in PFN1-depleted cells. Taken together, our findings shed light on the role of PFN1 in MLO biogenesis and suggest its involvement in neurodegenerative pathogenesis.
Collapse
Affiliation(s)
- Guoqiang Ma
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xiye Ruan
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Bojun Yang
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ningning Li
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Dan Su
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Shan Sun
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Siqian Chen
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Kangjia Xu
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zheng Ying
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; MOE Key Laboratory of Geriatric Diseases and Immunology, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou, Jiangsu 215123, China.
| | - Hongfeng Wang
- Jiangsu Key Laboratory of Drug Discovery and Translational Research for Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China; MOE Key Laboratory of Geriatric Diseases and Immunology, College of Pharmaceutical Sciences, Suzhou Medical College of Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
4
|
Biancon G, Busarello E, Cheng M, Halene S, Tebaldi T. Dissecting the stress granule RNA world: dynamics, strategies, and data. RNA (NEW YORK, N.Y.) 2025; 31:743-755. [PMID: 40086831 DOI: 10.1261/rna.080409.125] [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: 02/01/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Stress granules (SGs) are cytoplasmic ribonucleoprotein granules that commonly nucleate from the interaction of translationally stalled mRNAs and RNA-binding proteins. SGs are involved in the cellular adaptation to stress conditions participating in the regulation of gene expression and cell signaling. While dysregulation of SG dynamics has been increasingly implicated in human disease, a comprehensive understanding of SG composition, particularly of the RNA component, across various conditions remains elusive. Here, we review the physiological and pathological aspects of SGs, discuss current and future experimental strategies to identify SG components, and provide insights into the SG RNA world through the meta-analysis of 26 human SG transcriptome data sets.
Collapse
Affiliation(s)
- Giulia Biancon
- Section of Hematology, Department of Internal Medicine, Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06511, USA
- Hematology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Emma Busarello
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, 38123, Italy
| | - Matthew Cheng
- Section of Hematology, Department of Internal Medicine, Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06511, USA
| | - Toma Tebaldi
- Section of Hematology, Department of Internal Medicine, Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, Connecticut 06511, USA
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, 38123, Italy
| |
Collapse
|
5
|
Stickelman Z, Sarfraz N, Rice MK, Lambeck BJ, Milkovich S, Braselmann E. Expanding the Riboglow-FLIM Toolbox with Different Fluorescence Lifetime-Producing RNA Tags. Biochemistry 2025. [PMID: 40372768 DOI: 10.1021/acs.biochem.4c00567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
RNAs are essential elements of biology with subcellular localizations critical for function. Genetically tagging fluorescent RNA reporters to an RNA of interest allows for investigating RNA spatiotemporal dynamics. We previously developed a fluorescence lifetime imaging microscopy (FLIM)-based RNA-tagging platform, Riboglow-FLIM. Here, a genetically encoded RNA tag binds a fluorescent probe, causing an increase in both fluorescence intensity and fluorescence lifetime. Importantly, the Riboglow platform is derived from a bacterial riboswitch RNA family and different riboswitch sequences from nature may build the basis for multiplexing capabilities. We previously observed fluorescence lifetime differences for two RNA tags in vitro and in live mammalian cells as a proof-of-concept demonstration. As an in-depth expansion for multiplexing capabilities, here we evaluate the performance of different RNA sequences in vitro for systematically expanding the RNA tag sequence space of Riboglow-FLIM. We use two methods of varying the genetic tag to evaluate multiplexing capabilities, a literature-guided and a rational design approach. The literature-guided approach includes riboswitch sequences with both indirect and direct evidence of probe binding. For this, a phylogenetic tree of riboswitch-derived tags from indirect binding results was constructed, and RNA members from different branches were characterized. We also designed RNA mutations rationally based on insights from established Riboglow RNA tags. Together, nine different RNA tags yielded a wide range of fluorescence lifetimes for the Riboglow-FLIM platform, building the foundation to tag and track several different RNAs simultaneously. These findings will serve as the basis for achieving multiplexed RNA imaging in live cells using a fluorescence lifetime sensor.
Collapse
Affiliation(s)
- Zachary Stickelman
- Department of Chemistry, Georgetown University, Washington, District of Columbia 20057, United States
| | - Nadia Sarfraz
- Department of Chemistry, Georgetown University, Washington, District of Columbia 20057, United States
| | - Morgan K Rice
- Department of Chemistry, Georgetown University, Washington, District of Columbia 20057, United States
| | - Ben J Lambeck
- Department of Chemistry, Georgetown University, Washington, District of Columbia 20057, United States
| | - Sonja Milkovich
- Department of Chemistry, Georgetown University, Washington, District of Columbia 20057, United States
| | - Esther Braselmann
- Department of Chemistry, Georgetown University, Washington, District of Columbia 20057, United States
| |
Collapse
|
6
|
Rajachandran S, Xu Q, Cao Q, Zhang X, Chen F, Mangiameli SM, Chen H. Subcellular level spatial transcriptomics with PHOTON. Nat Commun 2025; 16:4457. [PMID: 40368943 PMCID: PMC12078482 DOI: 10.1038/s41467-025-59801-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
The subcellular localization of RNA is closely linked to its function. Many RNA species are partitioned into organelles and other subcellular compartments for storage, processing, translation, or degradation. Thus, capturing the subcellular spatial distribution of RNA would directly contribute to the understanding of RNA functions and regulation. Here, we present PHOTON, a method which combines high resolution imaging with high throughput sequencing to achieve spatial transcriptome profiling at subcellular resolution. We demonstrate PHOTON as a versatile tool to accurately capture the transcriptome of target cell types in situ at the tissue level such as granulosa cells in the ovary, as well as RNA content within subcellular compartments such as the nucleoli, the mitochondria, and the stress granules. Using PHOTON, we also reveal the functional role of m6A modifications on mRNA partitioning into stress granules. These results collectively demonstrate that PHOTON is a flexible and generalizable platform for understanding subcellular molecular dynamics through the transcriptomic lens.
Collapse
Affiliation(s)
- Shreya Rajachandran
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qianlan Xu
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qiqi Cao
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xin Zhang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Fei Chen
- Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Sarah M Mangiameli
- Gene Regulation Observatory, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Haiqi Chen
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
7
|
Methawasin M, Zhang Y, Gregorich ZR, He Y, Liu C, Muldoon J, Hourani Z, Smith JE, Granzier H, Guo W. Reducing Granules Without Splicing Restoration Alleviates RBM20 Cardiomyopathy. Circ Res 2025; 136:1134-1146. [PMID: 40242865 PMCID: PMC12064375 DOI: 10.1161/circresaha.124.324781] [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: 04/19/2024] [Revised: 03/12/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND RBM20 (RNA binding motif protein 20) cardiomyopathy is a severe form of dilated cardiomyopathy (DCM). Genetic variants in the nuclear localization signal of Rbm20 hinder its nuclear import and promote cytoplasmic pathogenic RNP (ribonucleoprotein) granules. We aimed to investigate whether reducing RNP granules by inhibiting Rbm20 expression could alleviate the DCM phenotype in Rbm20 S639G (Rbm20S639G) knock-in mice. METHODS We downregulated Rbm20, utilizing antisense oligonucleotides (ASOs) that specifically inhibit Rbm20 expression. We administered Rbm20-ASOs in Rbm20S639G mice that carry a serine-to-glycine substitution in the nuclear localization signal of RBM20. The Rbm20-ASOs were administered subcutaneously at 25 mg/kg once a week for 8 weeks in both young (14-day-old) mice before the onset of DCM and adult (3-month-old) mice with established DCM phenotype. In vivo cardiac function was assessed by echocardiography. RNP granules were identified through fluorescent immunohistochemical staining, and the number and size of RNP granules were quantified using Cell Profiler software. Alternative splicing of RBM20 target genes was determined by reverse transcription polymerase chain reaction, and titin isoform expression was analyzed by gel electrophoresis. Cardiomyocyte Ca2+ release-reuptake kinetics and mouse electrocardiography were also studied. RESULTS The results revealed that reducing the level of Rbm20 expression through treatment with ASOs significantly decreased the cytoplasmic RNP granules within the Rbm20S639G cardiomyocytes. ASO treatment reduced the severity of DCM developed when treatment was initiated before the onset of the disease. Importantly, ASO treatment reversed cardiac dysfunction and remodeling when treatment was commenced in mice with established DCM as shown by a significant improvement in ejection fraction and a decrease in the severity of left ventricular chamber dilation. Treatment with ASOs also effectively mitigated left ventricular hypertrophic remodeling and improved ECG parameters observed as normalized P wave and QRS durations. These beneficial effects occur without the restoration of mis-splicing of RBM20 target genes, including the primary target gene Ttn, and other genes such as Camk2d, Ryr2, and Ank3. CONCLUSIONS The findings of this study demonstrated that RNP granules serve as a critical driver for RBM20 cardiomyopathy, and reduction of RNP granules through treatment with ASOs is a possible therapeutic option for RBM20 cardiomyopathy in patients carrying RBM20 genetic variants in the nuclear localization signal region.
Collapse
Affiliation(s)
- Mei Methawasin
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| | - Yanghai Zhang
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Zachery R. Gregorich
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Yaqin He
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Chunling Liu
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Julia Muldoon
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - Zaynab Hourani
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - John E. Smith
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - Henk Granzier
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ 85721, USA
| | - Wei Guo
- Department of Animal and Dairy Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA
- Cardiovascular Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| |
Collapse
|
8
|
Mohanan G, Roy R, Malka-Mahieu H, Lamba S, Fabbri L, Kalia S, Biswas A, Martineau S, M Labbé C, Vagner S, Rajyaguru PI. Genotoxic stress triggers Scd6-dependent regulation of translation to modulate the DNA damage response. EMBO Rep 2025:10.1038/s44319-025-00443-3. [PMID: 40275106 DOI: 10.1038/s44319-025-00443-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025] Open
Abstract
The role of mRNA translation and decay in the genotoxic stress response remains poorly explored. Here, we identify the role of yeast RGG motif-containing RNA binding protein Scd6 and its human ortholog LSM14A in genotoxic stress response. Scd6 localizes to cytoplasmic puncta upon cell treatment with various genotoxic agents. Scd6 genetically interacts with SRS2, a DNA helicase with an anti-recombination role in DNA damage repair under HU stress. Scd6 directly interacts with the SRS2 mRNA to repress its translation in cytoplasmic granules upon HU stress in an eIF4G1-independent manner. Scd6-SRS2 interaction is modulated by arginine methylation and the LSm-domain of Scd6, which acts as a cis-regulator of Scd6 arginine methylation. LSM14A regulates the translation of mRNAs encoding key NHEJ (Non-homologous end-joining) proteins such as RTEL1 (SRS2 functional homolog) and LIG4. NHEJ activity in yeast and mammalian cells is regulated by Scd6 and LSM14A, respectively. Overall, this report unveils the role of RNA binding proteins in regulating the translation of specific mRNAs coding for DNA damage response proteins upon genotoxic stress.
Collapse
Affiliation(s)
- Gayatri Mohanan
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Raju Roy
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
- University of Pennsylvania, Philadelphia, PA, USA
| | - Hélène Malka-Mahieu
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France
| | - Swati Lamba
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Lucilla Fabbri
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France
| | - Sidhant Kalia
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France
| | - Anusmita Biswas
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Sylvain Martineau
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France
| | - Céline M Labbé
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France
| | - Stéphan Vagner
- Institut Curie, PSL Research University, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France.
- Université Paris Sud, Université Paris-Saclay, CNRS UMR3348, INSERM U1278, F-91405, Orsay, France.
| | | |
Collapse
|
9
|
Jimenez Granda ER, Karoui H, Brilland X, Baret JC, Martin N. Light-Responsive Mononucleotide Coacervates. Chemistry 2025:e202501109. [PMID: 40244931 DOI: 10.1002/chem.202501109] [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: 04/07/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/19/2025]
Abstract
Liquid-liquid phase separation (LLPS) is central to the formation of biomolecular condensates in modern cells and is also explored as a mechanism for assembling protocells. Phase-separated droplets provide dynamic micro-environments that concentrate reactants, enhance reactions, and allow molecular exchange, essential for both cellular regulation, and adaptive compartmentalization. While modern cells achieve dynamic LLPS through enzymatic and metabolic pathways, protocells may be designed to harness external stimuli such as pH, temperature, redox potential, or light. However, existing studies of light-responsive coacervates rely on complex biomolecules, limiting their use as minimal platforms of energy-driven phase separation. Here, we develop a minimal system of light-responsive coacervates composed of low-molecular-weight species: mononucleotides and azobenzene-based amphiphiles with varying charge valency. These coacervates undergo reversible phase transitions through azobenzene photoisomerization, with their behavior governed by charge valency and nucleotide structure. We demonstrate that coacervates formed with high-valency azobenzenes remain stable under UV irradiation but exhibit significant property changes, while those with low-valency azobenzenes dissolve, enabling light-controlled nucleotide release. Additionally, we achieved hierarchical droplet organization and light-actuated biomolecular localization within multiphase systems. Overall, these findings establish a minimal platform for designing light-responsive synthetic protocells, providing insights into dynamic compartmentalization in de novo life-like systems.
Collapse
Affiliation(s)
- Edison Rafael Jimenez Granda
- Univ. Bordeaux, CNRS, Centre de Recherche Paul Pascal, UMR 5031, 115 avenue du Dr. Schweitzer, Pessac, 33600, France
| | - Hedi Karoui
- Univ. Bordeaux, CNRS, Centre de Recherche Paul Pascal, UMR 5031, 115 avenue du Dr. Schweitzer, Pessac, 33600, France
| | - Xavier Brilland
- Univ. Bordeaux, CNRS, Centre de Recherche Paul Pascal, UMR 5031, 115 avenue du Dr. Schweitzer, Pessac, 33600, France
| | - Jean-Christophe Baret
- Univ. Bordeaux, CNRS, Centre de Recherche Paul Pascal, UMR 5031, 115 avenue du Dr. Schweitzer, Pessac, 33600, France
- Institut Universitaire de France, Paris, 75005, France
| | - Nicolas Martin
- Univ. Bordeaux, CNRS, Centre de Recherche Paul Pascal, UMR 5031, 115 avenue du Dr. Schweitzer, Pessac, 33600, France
| |
Collapse
|
10
|
Chen R, Grill S, Lin B, Saiduddin M, Lehmann R. Origin and establishment of the germline in Drosophila melanogaster. Genetics 2025; 229:iyae217. [PMID: 40180587 PMCID: PMC12005264 DOI: 10.1093/genetics/iyae217] [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: 05/14/2024] [Accepted: 12/09/2024] [Indexed: 04/05/2025] Open
Abstract
The continuity of a species depends on germ cells. Germ cells are different from all the other cell types of the body (somatic cells) as they are solely destined to develop into gametes (sperm or egg) to create the next generation. In this review, we will touch on 4 areas of embryonic germ cell development in Drosophila melanogaster: the assembly and function of germplasm, which houses the determinants for germ cell specification and fate and the mitochondria of the next generation; the process of pole cell formation, which will give rise to primordial germ cells (PGCs); the specification of pole cells toward the PGC fate; and finally, the migration of PGCs to the somatic gonadal precursors, where they, together with somatic gonadal precursors, form the embryonic testis and ovary.
Collapse
Affiliation(s)
- Ruoyu Chen
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Vilcek Institute of Graduate Studies, Department of Cell Biology, NYU School of Medicine, New York University, New York, NY 10016, USA
| | - Sherilyn Grill
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Benjamin Lin
- Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Mariyah Saiduddin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Vilcek Institute of Graduate Studies, Department of Cell Biology, NYU School of Medicine, New York University, New York, NY 10016, USA
| | - Ruth Lehmann
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| |
Collapse
|
11
|
Yang G, Wang Y, Guo J, Rui T. Connecting the Dots: Stress Granule and Cardiovascular Diseases. J Cardiovasc Transl Res 2025:10.1007/s12265-025-10619-w. [PMID: 40229624 DOI: 10.1007/s12265-025-10619-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Stress granules (SGs) are membrane-less cytoplasmic assemblies composed of mRNAs and RNA-binding proteins (RBPs) that transiently form to cope with various cellular stressors by halting mRNA translation and, consequently, protein synthesis. SG formation plays a crucial role in regulating multiple cellular processes, including cellular senescence, inflammatory responses, and adaptation to oxidative stress under both physiological and pathological conditions. Dysregulation of SG assembly and disassembly has been implicated in the pathogenesis of various diseases, including cardiovascular diseases (CVDs), cancer, viral and bacterial infections, and degenerative diseases. In this review, we survey the key aspects of SGs biogenesis and biological functions, with a particular focus on their causal involvement in CVDs. Furthermore, we summarized several SG-modulating compounds and discussed the therapeutic potential of small molecules targeting SG-related diseases in clinical settings.
Collapse
Affiliation(s)
- Gaowei Yang
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu, 212002, People's Republic of China
| | - Yiming Wang
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu, 212002, People's Republic of China
| | - Junfang Guo
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu, 212002, People's Republic of China
| | - Tao Rui
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu, 212002, People's Republic of China.
| |
Collapse
|
12
|
Kim SW, Lee J, Jo KW, Jeong YH, Shin WS, Kim KT. RNF144A-VRK2-G3BP1 axis regulates stress granule assembly. Cell Death Discov 2025; 11:158. [PMID: 40204710 PMCID: PMC11982375 DOI: 10.1038/s41420-025-02460-6] [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: 10/21/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/11/2025] Open
Abstract
Under the cellular stress, stress granules (SGs) help survival and proliferation of the cell. Unfortunately, the same SGs help unwanted cancer cells under stressful environment, including anti-cancer chemotherapy treatment. While SGs elevate the cancer cell's resistance to chemotherapy, the mechanism behind the formation of SGs in cancer cell under chemotherapy treatment is still to be revealed. Here, we identified that the level of VRK2 and the phosphorylation of its novel substrate, G3BP1, are reduced when the cellular stress was increased by sodium arsenite (SA) or cisplatin treatment. We also demonstrated that the level of RNF144A is increased in response to the stress and further downregulates VRK2 through proteasomal degradation in various types of cancer cells. Furthermore, inhibition of SG formation by the overexpression of VRK2 sensitized the cells to the stress and chemotherapy. Together, our study establishes an RNF144A-VRK2-G3BP1 axis that regulates SG formation and suggest its potential usage in anti-cancer therapy.
Collapse
Affiliation(s)
- Sung Wook Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Jae Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Kyung Won Jo
- Hesed Bio Corporation, Pohang, Gyeongbuk, Republic of Korea
| | - Young-Hun Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Won Sik Shin
- Department of Life Sciences, Pohang University of Science and Technology, Pohang, Gyeongbuk, Republic of Korea
| | - Kyong-Tai Kim
- Generative Genomics Research Center, Global Green Research & Development Center, Handong Global University, Pohang, Gyeongbuk, Republic of Korea.
| |
Collapse
|
13
|
Xu S, Ouyang Y, Qin Y, Chen D, Duan Z, Song D, Harries D, Xia F, Willner I, Huang F. Spatiotemporal dynamic and catalytically mediated reconfiguration of compartmentalized cyanuric acid/polyadenine DNA microdroplet condensates. Nat Commun 2025; 16:3352. [PMID: 40204808 PMCID: PMC11982331 DOI: 10.1038/s41467-025-58650-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
Native cells possess membrane-bound subcompartments, organelles, such as mitochondria and lysosomes, that intercommunicate and regulate cellular functions. Extensive efforts are directed to develop synthetic cells, or protocells, that replicate these structures and functions. Among these approaches, phase-separated coacervate microdroplets composed of polymers, polysaccharides, proteins, or nucleic acids are gaining interest as cell-mimicking systems. Particularly, compartmentalization of the synthetic protocell assemblies and the integration of functional constituents in the containments allowing signaling, programmed transfer of chemical agents, and spatiotemporal controlled catalytic transformations across the protocell subdomains, are challenging goals in developing artificial cells. Here, we report the assembly of compartmentalized, phase-separated cyanuric acid/polyadenine coacervate microdroplets. Hierarchical, co-centric compartmentalization is achieved through the dynamic and competitive spatiotemporal occupation of pre-engineered barcode domains within the polyadenine microdroplet framework by invading DNA strands. By encoding structural and functional information within these DNA-invaded compartments, the light-triggered, switchable reconfiguration of compartments, switchable catalytic reconfiguration of containments, and reversible aggregation/deaggregation of the compartmentalized microdroplets are demonstrated.
Collapse
Affiliation(s)
- Shijun Xu
- State Key Laboratory of Geomicrobiology and Environmental Changes, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China
| | - Yu Ouyang
- Institute of Chemistry, Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yunlong Qin
- Institute of Chemistry, Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Danlong Chen
- State Key Laboratory of Geomicrobiology and Environmental Changes, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China
| | - Zhijuan Duan
- State Key Laboratory of Geomicrobiology and Environmental Changes, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China
| | - Dongxing Song
- School of Mechanics and Safety Engineering, Zhengzhou University, Zhengzhou, Henan, China.
| | - Daniel Harries
- Institute of Chemistry, The Fritz Haber Research Center, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Fan Xia
- State Key Laboratory of Geomicrobiology and Environmental Changes, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China.
| | - Itamar Willner
- Institute of Chemistry, Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Fujian Huang
- State Key Laboratory of Geomicrobiology and Environmental Changes, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, China.
| |
Collapse
|
14
|
Tang L, Zhang W, Liao Y, Wang W, Deng X, Wang C, Shi W. Autophagy: a double-edged sword in ischemia-reperfusion injury. Cell Mol Biol Lett 2025; 30:42. [PMID: 40197222 PMCID: PMC11978130 DOI: 10.1186/s11658-025-00713-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Ischemia-reperfusion (I/R) injury describes the pathological process wherein tissue damage, initially caused by insufficient blood supply (ischemia), is exacerbated upon the restoration of blood flow (reperfusion). This phenomenon can lead to irreversible tissue damage and is commonly observed in contexts such as cardiac surgery and stroke, where blood supply is temporarily obstructed. During ischemic conditions, the anaerobic metabolism of tissues and organs results in compromised enzyme activity. Subsequent reperfusion exacerbates mitochondrial dysfunction, leading to increased oxidative stress and the accumulation of reactive oxygen species (ROS). This cascade ultimately triggers cell death through mechanisms such as autophagy and mitophagy. Autophagy constitutes a crucial catabolic mechanism within eukaryotic cells, facilitating the degradation and recycling of damaged, aged, or superfluous organelles and proteins via the lysosomal pathway. This process is essential for maintaining cellular homeostasis and adapting to diverse stress conditions. As a cellular self-degradation and clearance mechanism, autophagy exhibits a dualistic function: it can confer protection during the initial phases of cellular injury, yet potentially exacerbate damage in the later stages. This paper aims to elucidate the fundamental mechanisms of autophagy in I/R injury, highlighting its dual role in regulation and its effects on both organ-specific and systemic responses. By comprehending the dual mechanisms of autophagy and their implications for organ function, this study seeks to explore the potential for therapeutic interventions through the modulation of autophagy within clinical settings.
Collapse
Affiliation(s)
- Lingxuan Tang
- Basic Medical University, Naval Medical University, Shanghai, 200433, China
| | - Wangzheqi Zhang
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yan Liao
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Weijie Wang
- Basic Medical University, Naval Medical University, Shanghai, 200433, China
| | - Xiaoming Deng
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Changli Wang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
| | - Wenwen Shi
- School of Nursing, Navy Military Medical University, Shanghai, China.
| |
Collapse
|
15
|
Manjunath L, Santiago G, Ortega P, Sanchez A, Oh S, Garcia A, Li J, Duong D, Bournique E, Bouin A, Semler BL, Setiaputra D, Buisson R. Cooperative role of PACT and ADAR1 in preventing aberrant PKR activation by self-derived double-stranded RNA. Nat Commun 2025; 16:3246. [PMID: 40185749 PMCID: PMC11971382 DOI: 10.1038/s41467-025-58412-2] [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: 11/17/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Double-stranded RNAs (dsRNAs) produced during viral infections are recognized by the innate immune sensor protein kinase R (PKR), triggering a host translation shutoff that inhibits viral replication and propagation. Given the harmful effects of uncontrolled PKR activation, cells must tightly regulate PKR to ensure that its activation occurs only in response to viral infections, not endogenous dsRNAs. Here, we use CRISPR-Translate, a FACS-based genome-wide CRISPR-Cas9 knockout screening method that exploits translation levels as a readout and identifies PACT as a key inhibitor of PKR during viral infection. We find that PACT-deficient cells hyperactivate PKR in response to different RNA viruses, raising the question of why cells need to limit PKR activity. Our results demonstrate that PACT cooperates with ADAR1 to suppress PKR activation from self-dsRNAs in uninfected cells. The simultaneous deletion of PACT and ADAR1 results in synthetic lethality, which can be fully rescued in PKR-deficient cells. We propose that both PACT and ADAR1 act as essential barriers against PKR, creating a threshold of tolerable levels to endogenous dsRNA in cells without activating PKR-mediated translation shutdown and cell death.
Collapse
Affiliation(s)
- Lavanya Manjunath
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, USA
- Center for Virus Research, University of California Irvine, Irvine, California, USA
| | - Gisselle Santiago
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, USA
- Center for Virus Research, University of California Irvine, Irvine, California, USA
| | - Pedro Ortega
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, USA
- Center for Virus Research, University of California Irvine, Irvine, California, USA
| | - Ambrocio Sanchez
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, USA
- Center for Virus Research, University of California Irvine, Irvine, California, USA
| | - Sunwoo Oh
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, USA
- Center for Virus Research, University of California Irvine, Irvine, California, USA
| | - Alexander Garcia
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, USA
- Center for Virus Research, University of California Irvine, Irvine, California, USA
| | - Junyi Li
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, USA
- Center for Virus Research, University of California Irvine, Irvine, California, USA
| | - Dana Duong
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, USA
- Center for Virus Research, University of California Irvine, Irvine, California, USA
| | - Elodie Bournique
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, USA
- Center for Virus Research, University of California Irvine, Irvine, California, USA
| | - Alexis Bouin
- Center for Virus Research, University of California Irvine, Irvine, California, USA
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, California, USA
| | - Bert L Semler
- Center for Virus Research, University of California Irvine, Irvine, California, USA
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, California, USA
| | - Dheva Setiaputra
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Rémi Buisson
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, California, USA.
- Center for Virus Research, University of California Irvine, Irvine, California, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California Irvine, Irvine, California, USA.
| |
Collapse
|
16
|
Pan CR, Knutson SD, Huth SW, MacMillan DWC. µMap proximity labeling in living cells reveals stress granule disassembly mechanisms. Nat Chem Biol 2025; 21:490-500. [PMID: 39215100 PMCID: PMC11868469 DOI: 10.1038/s41589-024-01721-2] [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: 01/05/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024]
Abstract
Phase-separated condensates are membrane-less intracellular structures comprising dynamic protein interactions that organize essential biological processes. Understanding the composition and dynamics of these organelles advances our knowledge of cellular behaviors and disease pathologies related to granule dysregulation. In this study, we apply microenvironment mapping with a HaloTag-based platform (HaloMap) to characterize intracellular stress granule dynamics in living cells. After validating the robustness and sensitivity of this approach, we then profile the stress granule proteome throughout the formation and disassembly and under pharmacological perturbation. These experiments reveal several ubiquitin-related modulators, including the HECT (homologous to E6AP C terminus) E3 ligases ITCH and NEDD4L, as well as the ubiquitin receptor toll-interacting protein TOLLIP, as key mediators of granule disassembly. In addition, we identify an autophagy-related pathway that promotes granule clearance. Collectively, this work establishes a general photoproximity labeling approach for unraveling intracellular protein interactomes and uncovers previously unexplored regulatory mechanisms of stress granule dynamics.
Collapse
Affiliation(s)
- Chenmengxiao Roderick Pan
- Merck Center for Catalysis at Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Steve D Knutson
- Merck Center for Catalysis at Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Sean W Huth
- Merck Center for Catalysis at Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - David W C MacMillan
- Merck Center for Catalysis at Princeton University, Princeton, NJ, USA.
- Department of Chemistry, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
17
|
Mejías D, Seballos V, Lafon-Hughes L. Liquid‒liquid phase separation and poly(ADP‒ribosyl)ation in the context of ultraviolet radiation-induced stress in mammalian cells. Biophys Rev 2025; 17:499-510. [PMID: 40376396 PMCID: PMC12075067 DOI: 10.1007/s12551-025-01294-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 02/21/2025] [Indexed: 05/18/2025] Open
Abstract
Poly(ADP‒ribose) polymerases (PARPs) consume NAD+ to synthesize poly(ADP‒ribose) (PAR) primarily via post-translational modification. PAR is degraded mainly by poly (ADP-ribose) glycohydrolase (PARG). PAR can be linear or branched and can have up to 200 monomers. With two phosphates per monomer, PAR is highly negatively charged. PAR can be recognized by specific protein domains and has been described as a "glue" or scaffold for the assembly of multiprotein complexes. PAR is involved in several diverse cellular structures and functions, including DNA replication, transcription, DNA repair, chromatin structure and imprinting regulation, mitotic spindle assembly, cell‒cell junctions, cytoplasmic granule formation, biomineralization and the formation of pathological aggregates. Here, we review the effects of ultraviolet radiation (UVR) on mammalian cells, emphasizing the participation of PAR metabolism in the novel paradigm of liquid‒liquid phase separation (LLPS). Further studies demand interdisciplinary approaches, undoubtedly requiring contributions from biophysicists. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-025-01294-x.
Collapse
Affiliation(s)
- Daniela Mejías
- Biophysical Chemistry Group, Biological Sciences Department, Centro Universitario Litoral Norte, Salto, Universidad de La República (UdelaR), Rivera1350, ZIP 50.000, Montevideo, Uruguay
| | - Valentina Seballos
- Biophysical Chemistry Group, Biological Sciences Department, Centro Universitario Litoral Norte, Salto, Universidad de La República (UdelaR), Rivera1350, ZIP 50.000, Montevideo, Uruguay
| | - Laura Lafon-Hughes
- Biophysical Chemistry Group, Biological Sciences Department, Centro Universitario Litoral Norte, Salto, Universidad de La República (UdelaR), Rivera1350, ZIP 50.000, Montevideo, Uruguay
| |
Collapse
|
18
|
Ricolo D, Casanova J, Giannios P. Drosophila and human Headcase define a new family of ribonucleotide granule proteins required for stress response. SCIENCE ADVANCES 2025; 11:eads2086. [PMID: 40153502 PMCID: PMC11952099 DOI: 10.1126/sciadv.ads2086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 02/25/2025] [Indexed: 03/30/2025]
Abstract
Cells have means to adapt to environmental stresses such as temperature fluctuations, toxins, or nutrient availability. Stress responses, being dynamic, extend beyond transcriptional control and encompass post-transcriptional mechanisms allowing for rapid changes in protein synthesis. Previous research has established headcase as a fundamental gene for stress responses and survival of the Drosophila adult progenitor cells (APCs). However, the molecular role of Headcase has remained elusive. Here, we identify Headcase as a component of ribonucleoprotein (RNP) granules. We also show that, Headcase is required for proper RNP granule formation and remodeling upon stress and is crucial for translation control. Likewise, the human Headcase homolog (HECA) is identified as a component of RNP granules and has similar roles in translational regulation and stress protection. Thus, Headcase proteins define a new family contributing to specific roles among the RNP heterogeneous network.
Collapse
Affiliation(s)
- Delia Ricolo
- Institut de Biologia Molecular de Barcelona (CSIC), Baldiri Reixac 4, 08028 Barcelona, Catalonia, Spain
- Institut de Recerca Biomèdica de Barcelona, (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
| | - Jordi Casanova
- Institut de Biologia Molecular de Barcelona (CSIC), Baldiri Reixac 4, 08028 Barcelona, Catalonia, Spain
- Institut de Recerca Biomèdica de Barcelona, (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
| | - Panagiotis Giannios
- Institut de Biologia Molecular de Barcelona (CSIC), Baldiri Reixac 4, 08028 Barcelona, Catalonia, Spain
- Institut de Recerca Biomèdica de Barcelona, (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 10, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
19
|
Feng J, Osmekhina E, Timonen JVI, Linder MB. Effects of Sup35 overexpression on the formation, morphology, and physiological functions of intracellular Sup35 assemblies. Appl Environ Microbiol 2025; 91:e0170324. [PMID: 39912644 PMCID: PMC11921396 DOI: 10.1128/aem.01703-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/16/2025] [Indexed: 02/07/2025] Open
Abstract
The yeast prion protein Sup35 is aggregation-prone at high concentrations. De novo Sup35 prion formation occurs at a significantly increased rate after transient overexpression of Sup35 in the presence of another prion, [PIN+], but it is still a rare event. Recent studies uncovered an additional and seemingly more prevalent role of Sup35: at its physiological level, it undergoes phase separation to form reversible condensates in response to transient stress. Stress-induced reversible Sup35 condensation in the [psi-] strain enhances cellular fitness after stress ceases, whereas irreversible Sup35 aggregates in the [PSI+] strain do not confer this advantage. However, how Sup35 overexpression, which could potentially lead to irreversible aggregation, affects its condensation under stress conditions remains unclear. In this study, we used a combinatorial method to examine how different levels of Sup35 overproduction and cellular conditions affect the nature, formation, and physical properties of Sup35 assemblies in yeast cells, as well as their impacts on cellular growth. We observed notable morphological distinctions between irreversible Sup35 aggregates and reversible Sup35 condensates, possibly indicating different formation mechanisms. In addition, Sup35 aggregation caused by a very high overexpression level can strongly inhibit cell growth, diminish the formation of stress-induced condensates when Sup35 is completely aggregated, and impair cellular recovery from stress. Together, this study advances our fundamental understanding of the physical properties and formation mechanism of different Sup35 assemblies and their impacts on cellular growth. We conclude that in vivo studies are sensitive to overexpression and can lead to assembly routes that strongly affect functions. IMPORTANCE The role of condensates in living cells is often studied by overexpression. For understanding their physiological role, this can be problematic. Overexpression can shift cellular functions, thereby changing the system under study, and overexpression can also affect the phase behavior of condensates by shifting the position of the system in the underlying phase diagram. Our detailed study of overexpression of Sup35 in S. cerevisiae shows the interplay between these factors and highlights basic features of intracellular condensation such as the balance between condensation and aggregation as well as how cellular localization and responsiveness depend on protein levels. We also apply super-resolution microscopy to highlight details within the cells.
Collapse
Affiliation(s)
- Jianhui Feng
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo, Finland
- The Center of Excellence in Life-Inspired Hybrid Materials (LIBER), Aalto University, Espoo, Finland
| | - Ekaterina Osmekhina
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo, Finland
- The Center of Excellence in Life-Inspired Hybrid Materials (LIBER), Aalto University, Espoo, Finland
| | - Jaakko V. I. Timonen
- The Center of Excellence in Life-Inspired Hybrid Materials (LIBER), Aalto University, Espoo, Finland
- Department of Applied Physics, School of Science, Aalto University, Espoo, Finland
| | - Markus B. Linder
- Department of Bioproducts and Biosystems, School of Chemical Engineering, Aalto University, Espoo, Finland
- The Center of Excellence in Life-Inspired Hybrid Materials (LIBER), Aalto University, Espoo, Finland
| |
Collapse
|
20
|
Biayna J, Dumbović G. Decoding subcellular RNA localization one molecule at a time. Genome Biol 2025; 26:45. [PMID: 40033325 PMCID: PMC11874642 DOI: 10.1186/s13059-025-03507-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 02/13/2025] [Indexed: 03/05/2025] Open
Abstract
Eukaryotic cells are highly structured and composed of multiple membrane-bound and membraneless organelles. Subcellular RNA localization is a critical regulator of RNA function, influencing various biological processes. At any given moment, RNAs must accurately navigate the three-dimensional subcellular environment to ensure proper localization and function, governed by numerous factors, including splicing, RNA stability, modifications, and localizing sequences. Aberrant RNA localization can contribute to the development of numerous diseases. Here, we explore diverse RNA localization mechanisms and summarize advancements in methods for determining subcellular RNA localization, highlighting imaging techniques transforming our ability to study RNA dynamics at the single-molecule level.
Collapse
Affiliation(s)
- Josep Biayna
- Goethe University Frankfurt, Center for Molecular Medicine, Institute for Cardiovascular Regeneration, Frankfurt, Germany
| | - Gabrijela Dumbović
- Goethe University Frankfurt, Center for Molecular Medicine, Institute for Cardiovascular Regeneration, Frankfurt, Germany.
- Cardio-Pulmonary Institute (CPI), Goethe University, Frankfurt, Frankfurt, Germany.
- German Center of Cardiovascular Research (DZHK), Partner Site Rhein/Main, Frankfurt, Germany.
| |
Collapse
|
21
|
Gong X, Feng S, Wang J, Gao B, Xue W, Chu H, Fang S, Yuan Y, Cheng Y, Liao M, Sun Y, Tan L, Song C, Qiu X, Ding C, Tijhaar E, Forlenza M, Liao Y. Coronavirus endoribonuclease nsp15 suppresses host protein synthesis and evades PKR-eIF2α-mediated translation shutoff to ensure viral protein synthesis. PLoS Pathog 2025; 21:e1012987. [PMID: 40096172 PMCID: PMC11975131 DOI: 10.1371/journal.ppat.1012987] [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: 08/21/2024] [Revised: 04/07/2025] [Accepted: 02/16/2025] [Indexed: 03/19/2025] Open
Abstract
The endoribonuclease (EndoU) nsp15 of coronaviruses plays a crucial role in evading host innate immune responses by reducing the abundance of viral double-stranded RNA (dsRNA). However, our understanding of its interactions with host cellular targets remains limited. In this study, we demonstrate that overexpression of nsp15 from four coronavirus genera inhibits cellular protein synthesis and causes nuclear retention of PABPC1. Mutation analysis confirms the essential role of EndoU activity in these processes. Fluorescence in situ hybridization (FISH) analysis shows that cellular mRNA co-localizes with nsp15 in certain cells. Real time RT-PCR indicates that the mRNA levels of several antiviral genes decrease in cells expressing nsp15, and this reduction depends on the EndoU activity of nsp15. Using infectious bronchitis virus (IBV) as a model, we investigate the inhibitory effect of nsp15 on protein translation during infection. We find that infection with IBV with functional nsp15 suppresses protein synthesis in a PKR-eIF2α independent manner, with PABPC1 mainly located in the cytoplasm. However, infection with EndoU activity-deficiency mutant virus rIBV-nsp15-H238A results in the accumulation of viral dsRNA, triggering a PKR-eIF2α-dependent shutdown of protein synthesis and leading to the nuclear relocation of PABPC1. In the absence of the PKR-eIF2α pathway, IBV is still able to suppress host protein synthesis, while the inhibitory effect of rIBV-nsp15-H238A on protein synthesis was significantly reduced. Although nsp15 locates to replication-transcription complex (RTC) during infection, RNA immunoprecipitation (RIP)-Seq analysis confirms that IBV nsp15 binds to six viral RNAs and 237 cellular RNAs. The proteins encoded by the nsp15-associated cellular RNAs predominantly involved in translation. Additionally, proteomic analysis of the nsp15 interactome identifies 809 cellular proteins, which are significantly enriched in pathways related to ribosome biogenesis, RNA processing, and translation. Therefore, nsp15 helps virus circumvent the detrimental PKR-eIF2α pathway by reducing viral dsRNA accumulation and suppresses host protein synthesis by targeting host RNAs and proteins. This study reveals unique yet conserved mechanisms of protein synthesis shutdown by catalytically active nsp15 EndoU, shedding light on how coronaviruses regulate host protein expression.
Collapse
Affiliation(s)
- Xiaoqian Gong
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
- Cell Biology and Immunology Group, Wageningen University and Research, Department of Animal Sciences, Wageningen, the Netherlands,
| | - Shanhuan Feng
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| | - Jiehuang Wang
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| | - Bo Gao
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| | - Wenxiang Xue
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| | - Hongyan Chu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| | - Shouguo Fang
- College of Agriculture, College of Animal Sciences, Yangtze University, Jingzhou, China,
| | - Yanmei Yuan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| | - Yuqiang Cheng
- Shanghai Key Laboratory of Veterinary Biotechnology, Key Laboratory of Urban Agriculture (South), School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China,
| | - Min Liao
- Key Laboratory of Animal Virology of Ministry of Agriculture, Zhejiang University, Hangzhou, China,
| | - Yingjie Sun
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| | - Lei Tan
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| | - Cuiping Song
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| | - Xusheng Qiu
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| | - Chan Ding
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China,
| | - Edwin Tijhaar
- Cell Biology and Immunology Group, Wageningen University and Research, Department of Animal Sciences, Wageningen, the Netherlands,
| | - Maria Forlenza
- Cell Biology and Immunology Group, Wageningen University and Research, Department of Animal Sciences, Wageningen, the Netherlands,
- Host-Microbe Interactomics Group, Wageningen University and Research, Department of Animal Sciences, Wageningen, the Netherlands
| | - Ying Liao
- Department of Avian Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China,
| |
Collapse
|
22
|
Firdaus MER, Dukhno E, Kapoor R, Gerlach P. Two Birds With One Stone: RNA Virus Strategies to Manipulate G3BP1 and Other Stress Granule Components. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70005. [PMID: 40170442 PMCID: PMC11962251 DOI: 10.1002/wrna.70005] [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] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 04/03/2025]
Abstract
Stress granules (SGs) are membrane-less organelles forming in the cytoplasm in response to various types of stress, including viral infection. SGs and SG-associated proteins can play either a proviral role, by facilitating viral replication, or an antiviral role, by limiting the translation capacity, sequestering viral RNA, or contributing to the innate immune response of the cell. Consequently, viruses frequently target stress granules while counteracting cellular translation shut-off and the antiviral response. One strategy is to sequester SG components, not only to impair their assembly but also to repurpose and incorporate them into viral replication sites. G3BP1 is a key SG protein, driving its nucleation through protein-protein and protein-RNA interactions. Many cellular proteins, including other SG components, interact with G3BP1 via their ΦxFG motifs. Notably, SARS-CoV N proteins and alphaviral nsP3 proteins contain similar motifs, allowing them to compete for G3BP1. Several SG proteins have been shown to interact with the flaviviral capsid protein, which is primarily responsible for anchoring the viral genome inside the virion. There are also numerous examples of structured elements within coronaviral and flaviviral RNAs recruiting or sponging SG proteins. Despite these insights, the structural and biochemical details of SG-virus interactions remain largely unexplored and are known only for a handful of cases. Exploring their molecular relevance for infection and discovering new examples of direct SG-virus contacts is highly important, as advances in this area will open new possibilities for the design of targeted therapies and potentially broad-spectrum antivirals.
Collapse
Affiliation(s)
- Moh Egy Rahman Firdaus
- IMol Polish Academy of SciencesWarsawPoland
- ReMedy International Research Agenda UnitIMol Polish Academy of SciencesWarsawPoland
| | - Eliana Dukhno
- IMol Polish Academy of SciencesWarsawPoland
- ReMedy International Research Agenda UnitIMol Polish Academy of SciencesWarsawPoland
| | | | - Piotr Gerlach
- IMol Polish Academy of SciencesWarsawPoland
- ReMedy International Research Agenda UnitIMol Polish Academy of SciencesWarsawPoland
| |
Collapse
|
23
|
Boraas LC, Hu M, Martino P, Thornton L, Vejnar CE, Zhen G, Zeng L, Parker DM, Cox AL, Giraldez AJ, Su X, Mayr C, Wang S, Nicoli S. G3BP1 ribonucleoprotein complexes regulate focal adhesion protein mobility and cell migration. Cell Rep 2025; 44:115237. [PMID: 39883578 PMCID: PMC11923778 DOI: 10.1016/j.celrep.2025.115237] [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: 04/17/2024] [Revised: 11/05/2024] [Accepted: 01/06/2025] [Indexed: 02/01/2025] Open
Abstract
The subcellular localization of mRNAs plays a pivotal role in biological processes, including cell migration. For instance, β-actin mRNA and its associated RNA-binding protein (RBP), ZBP1/IGF2BP1, are recruited to focal adhesions (FAs) to support localized β-actin synthesis, crucial for cell migration. However, whether other mRNAs and RBPs also localize at FAs remains unclear. Here, we identify hundreds of mRNAs that are enriched at FAs (FA-mRNAs). FA-mRNAs share characteristics with stress granule (SG) mRNAs and are found in ribonucleoprotein (RNP) complexes with the SG RBP. Mechanistically, G3BP1 binds to FA proteins in an RNA-dependent manner, and its RNA-binding and dimerization domains, essential for G3BP1 to form RNPs in SG, are required for FA localization and cell migration. We find that G3BP1 RNPs promote cell speed by enhancing FA protein mobility and FA size. These findings suggest a previously unappreciated role for G3BP1 RNPs in regulating FA function under non-stress conditions.
Collapse
Affiliation(s)
- Liana C Boraas
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Mengwei Hu
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Pieter Martino
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Lauren Thornton
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Charles E Vejnar
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Gang Zhen
- Laboratory of Genome Integrity, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Longhui Zeng
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Dylan M Parker
- Department of Biochemistry and Howard Hughes Medical Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Andy L Cox
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Antonio J Giraldez
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Xiaolei Su
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA; Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Christine Mayr
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Siyuan Wang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA.
| | - Stefania Nicoli
- Yale Cardiovascular Research Center, Department of Internal Medicine, Section of Cardiology, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
24
|
Lin RJ, Lin LH, Chen ZP, Liu BC, Ko PC, Liao CL. The zinc finger protein ZFP36L2 inhibits flavivirus infection via the 5'-3' XRN1-mediated RNA decay pathway in the replication complexes. J Biomed Sci 2025; 32:27. [PMID: 39972499 PMCID: PMC11841009 DOI: 10.1186/s12929-025-01122-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 01/24/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND The zinc finger protein 36-like (ZFP36L) family is a CCCH-type group consisting of RNA-binding proteins, i.e., ZFP36L1 and ZFP36L2, which regulate cellular mRNA through the RNA decay pathway. ZFP36L1 combats flavivirus infections through the 5'-3' XRN1 and 3'-5' RNA exosome decay pathways. The present study clarified the role of human ZFP36L2 in the defense response of the host against flavivirus infection. METHODS Cell lines with overexpression or knockdown of ZFP36L2 were established using lentiviral vectors carrying genes for overexpression and short-hairpin RNA targeting specific genes, respectively. A plaque assay was employed to determine the viral titer. Immunofluorescence and real-time quantitative polymerase chain reaction were used to measure the viral RNA levels. The in vitro-transcribed RNA transcript derived from a replication-dead Japanese encephalitis virus (JEV) replicon containing the renilla luciferase reporter gene (J-R2A-NS5mt) was used to assess the stability of the flavivirus RNA. An RNA immunoprecipitation assay was used to detect the protein-RNA binding ability. Confocal microscopic images were captured to analyze protein colocalization. RESULTS ZFP36L2 served as an innate host defender against JEV and dengue virus. ZFP36L2 inhibited flavivirus infection solely through the 5'-3' XRN1 RNA decay pathway, whereas ZFP36L1 inhibited JEV infection via the 5'-3' XRN1 and 3'-5' RNA exosome RNA decay pathways. The direct binding between viral RNA and ZFP36L2 via its CCCH-type zinc finger motifs facilitated the degradation of flavivirus RNA mediated by 5'-3' XRN1. Furthermore, ZFP36L2 was localized in processing bodies (PBs), which participate in the 5'-3' XRN1-mediated RNA decay pathway. Nonetheless, the disruption of PBs did not affect the antiviral activity of ZFP36L2, suggesting that its localization is not essential for the function of the protein. Interestingly, the colocalization of ZFP36L2 and XRN1 with viral RNA and NS3 revealed that the antiviral activity of ZFP36L2 occurred within the replication complexes (RCs). CONCLUSIONS In summary, ZFP36L2 bound to and degraded viral RNA through the XRN1-mediated RNA decay pathway in the RCs, thereby inhibiting flavivirus replication. These findings provide valuable insights into the diverse antiviral mechanisms of the ZFP36-like family of proteins in the innate immune response against flavivirus infection.
Collapse
Affiliation(s)
- Ren-Jye Lin
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institute, Taipei, Taiwan.
| | - Li-Hsiung Lin
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institute, Taipei, Taiwan
| | - Zih-Ping Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Bing-Cheng Liu
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei, Taiwan
| | - Pin-Chen Ko
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Ching-Len Liao
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
25
|
Bujisic B, Lee HG, Xu L, Weissbein U, Rivera C, Topisirovic I, Lee JT. 7SL RNA and signal recognition particle orchestrate a global cellular response to acute thermal stress. Nat Commun 2025; 16:1630. [PMID: 39952919 PMCID: PMC11828898 DOI: 10.1038/s41467-025-56351-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/16/2025] [Indexed: 02/17/2025] Open
Abstract
Non-coding 7SL RNA is an ancestor to mammalian Alu and B1 SINE RNAs and is thought to function exclusively within the Signal Recognition Particle (SRP), aiding in the translocation of secretory proteins into the endoplasmic reticulum for export. Here, we discover a function of 7SL/SRP unrelated to protein secretion. Under acute heat shock, 7SL and SRP together selectively arrest cellular transcription and translation machineries during early response to stress. Under thermal stress, 7SL is upregulated, accumulates in the nucleus, and binds to target genes repressed by heat shock. Concurrently, in the cytosol, SRP binds to ribosomes and inhibits new protein synthesis. Translational suppression occurs independently of the signal peptide and is abrogated by depleting SRP. Translation inhibition extends to the mitochondria, as nuclear-encoded genes with mitochondrial functions are enriched among SRP targets. Thus, apart from its role in protein export, 7SL/SRP orchestrates a global response to acute stress that encompasses the nucleus, cytosol, and mitochondria across transcription and translation.
Collapse
Affiliation(s)
- Bojan Bujisic
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Hun-Goo Lee
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Lilei Xu
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Uri Weissbein
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Carlos Rivera
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114, USA
- Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA, 02114, USA
| | - Ivan Topisirovic
- Lady Davis Institute, Gerald Bronfman Department of Oncology and Departments of Biochemistry and Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Jeannie T Lee
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, 02114, USA.
- Department of Genetics, The Blavatnik Institute, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
26
|
Sulaj E, Sandell FL, Schwaigerlehner L, Marzban G, Dohm JC, Kunert R. Systems Biology of Recombinant 2G12 and 353/11 mAb Production in CHO-K1 Cell Lines at Phosphoproteome Level. Proteomes 2025; 13:9. [PMID: 39982319 PMCID: PMC11843875 DOI: 10.3390/proteomes13010009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/22/2025] Open
Abstract
Background: Chinese hamster ovary (CHO) cells are extensively used in the pharmaceutical industry for producing complex proteins, primarily because of their ability to perform human-like post-translational modifications. However, the efficiency of high-quality protein production can vary significantly for monoclonal antibody-producing cell lines, within the CHO host cell lines or by extrinsic factors. Methods: To investigate the complex cellular mechanisms underlying this variability, a phosphoproteomics analysis was performed using label-free quantitative liquid chromatography after a phosphopeptide enrichment of recombinant CHO cells producing two different antibodies and a tunicamycin treatment experiment. Using MaxQuant and Perseus for data analysis, we identified 2109 proteins and quantified 4059 phosphosites. Results: Significant phosphorylation dynamics were observed in nuclear proteins of cells producing the difficult-to-produce 2G12 mAb. It suggests that the expression of 2G12 regulates nuclear pathways based on increases and decreases in phosphorylation abundance. Furthermore, a substantial number of changes in the phosphorylation pattern related to tunicamycin treatment have been detected. TM treatment affects, among other phosphoproteins, the eukaryotic elongation factor 2 kinase (Eef2k). Conclusions: The alterations in the phosphorylation landscape of key proteins involved in cellular processes highlight the mechanisms behind stress-induced cellular responses.
Collapse
Affiliation(s)
- Eldi Sulaj
- Department of Biotechnology and Food Science, Institute of Animal Cell Technology and Systems Biology (IACTSB), BOKU University, Muthgasse 18, 1190 Vienna, Austria; (E.S.); (L.S.); (R.K.)
| | - Felix L. Sandell
- Department of Biotechnology and Food Science, Institute of Computational Biology (ICB), BOKU University, Muthgasse 18, 1190 Vienna, Austria; (F.L.S.)
| | - Linda Schwaigerlehner
- Department of Biotechnology and Food Science, Institute of Animal Cell Technology and Systems Biology (IACTSB), BOKU University, Muthgasse 18, 1190 Vienna, Austria; (E.S.); (L.S.); (R.K.)
| | - Gorji Marzban
- Department of Biotechnology and Food Science, Institute of Bioprocess Science and Engineering (IBSE), BOKU University, Muthgasse 18, 1190 Vienna, Austria
| | - Juliane C. Dohm
- Department of Biotechnology and Food Science, Institute of Computational Biology (ICB), BOKU University, Muthgasse 18, 1190 Vienna, Austria; (F.L.S.)
| | - Renate Kunert
- Department of Biotechnology and Food Science, Institute of Animal Cell Technology and Systems Biology (IACTSB), BOKU University, Muthgasse 18, 1190 Vienna, Austria; (E.S.); (L.S.); (R.K.)
| |
Collapse
|
27
|
Challa S, Nandu T, Kim HB, Gong X, Renshaw CW, Li WC, Tan X, Aljardali MW, Camacho CV, Chen J, Kraus WL. RACK1 MARylation regulates translation and stress granules in ovarian cancer cells. J Cell Biol 2025; 224:e202401101. [PMID: 39760726 DOI: 10.1083/jcb.202401101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 09/09/2024] [Accepted: 11/11/2024] [Indexed: 01/07/2025] Open
Abstract
Mono(ADP-ribosyl)ation (MARylation) is emerging as a critical regulator of ribosome function and translation. Herein, we demonstrate that RACK1, an integral component of the ribosome, is MARylated by the mono(ADP-ribosyl) transferase (MART) PARP14 in ovarian cancer cells. MARylation of RACK1 is required for stress granule formation and promotes the colocalization of RACK1 in stress granules with G3BP1, eIF3η, and 40S ribosomal proteins. In parallel, we observed reduced translation of a subset of mRNAs, including those encoding key cancer regulators (e.g., AKT). Treatment with a PARP14 inhibitor or mutation of the sites of MARylation on RACK1 blocks these outcomes, as well as the growth of ovarian cancer cells in culture and in vivo. To reset the system after prolonged stress and recovery, the ADP-ribosyl hydrolase TARG1 deMARylates RACK1, leading to the dissociation of the stress granules and the restoration of translation. Collectively, our results demonstrate a therapeutically targetable pathway that controls polysome assembly, translation, and stress granule dynamics in ovarian cancer cells.
Collapse
Affiliation(s)
- Sridevi Challa
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tulip Nandu
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hyung Bum Kim
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Graduate Program in Genetics, Development, and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xuan Gong
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Charles W Renshaw
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wan-Chen Li
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xinrui Tan
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Section of Laboratory Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marwa W Aljardali
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cristel V Camacho
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Section of Laboratory Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jin Chen
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - W Lee Kraus
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Graduate Program in Genetics, Development, and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Section of Laboratory Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
28
|
Rajendran A, Castañeda CA. Protein quality control machinery: regulators of condensate architecture and functionality. Trends Biochem Sci 2025; 50:106-120. [PMID: 39755440 PMCID: PMC11805624 DOI: 10.1016/j.tibs.2024.12.003] [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: 07/10/2024] [Revised: 11/23/2024] [Accepted: 12/02/2024] [Indexed: 01/06/2025]
Abstract
Protein quality control (PQC) mechanisms including the ubiquitin (Ub)-proteasome system (UPS), autophagy, and chaperone-mediated refolding are essential to maintain protein homeostasis in cells. Recent studies show that these PQC mechanisms are further modulated by biomolecular condensates that sequester PQC components and compartmentalize reactions. Accumulating evidence points towards the PQC machinery playing a pivotal role in regulating the assembly, disassembly, and viscoelastic properties of several condensates. Here, we discuss how the PQC machinery can form their own condensates and also be recruited to known condensates under physiological or stress-induced conditions. We present molecular insights into how the multivalent architecture of polyUb chains, Ub-binding adaptor proteins, and other PQC machinery contribute to condensate assembly, leading to the regulation of downstream PQC outcomes and therapeutic potential.
Collapse
Affiliation(s)
- Anitha Rajendran
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA
| | - Carlos A Castañeda
- Department of Chemistry, Syracuse University, Syracuse, NY 13244, USA; Department of Biology, Syracuse University, Syracuse, NY 13244, USA; Bioinspired Institute, Syracuse University, Syracuse, NY 13244, USA; Interdisciplinary Neuroscience Program, Syracuse University, Syracuse, NY 13244, USA.
| |
Collapse
|
29
|
Liao Y, Fan C, Zheng J, Liu C, Zhu W, Xu Y, Qian X, Yang Y. Enhanced liquid-liquid phase separation of stress granules in a reconstructed model and their cytoplasmic targeting using a DNA nanodevice. J Mater Chem B 2025; 13:1744-1752. [PMID: 39704478 DOI: 10.1039/d4tb02161d] [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: 12/21/2024]
Abstract
Biomolecular condensates (BCs) are crucial membraneless organelles formed through the process of liquid-liquid phase separation (LLPS) involving proteins and nucleic acids. These LLPS processes are tightly linked with essential cellular activities. Stress granules (SGs), functioning as cytoplasmic BCs, play indispensable roles in maintaining cellular homeostasis and are implicated in diseases like cancers and neurodegenerative disorders. However, devices that can regulate SG LLPS are lacking. Herein, a triangular prism-shaped DNA nanostructure containing polythymidine (ΔDNA(polyT)) is presented as a nanodevice to investigate the LLPS process of in vitro reconstructed SGs (rSGs), a mixture of marker protein G3BP1 and total RNAs. Our observations reveal that the concentration threshold required for rSG LLPS decreases upon addition of ΔDNA(polyT), suggesting an enhancement in SG LLPS efficiency. It is speculated that ΔDNA(polyT) can concentrate mRNAs onto its surface via polyT hybridization with poly-adenosine sequences (polyA) in mRNAs. This alteration in the spatial distribution of mRNAs subsequently affects the multivalency interactions between G3BP1 and mRNAs. Furthermore, ΔDNA(polyT) exhibits excellent colocalization with cytoplasmic SGs under stressed conditions. This DNA-based nanodevice presents a new artificial approach for the targeted regulation of BC LLPS and holds promise for future studies focusing on BCs.
Collapse
Affiliation(s)
- Yue Liao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Chunyu Fan
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Jiaxin Zheng
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Caixia Liu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Weiping Zhu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
- Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yufang Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| | - Xuhong Qian
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yangyang Yang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
30
|
Tahmasebinia F, Tang Y, Tang R, Zhang Y, Bonderer W, de Oliveira M, Laboret B, Chen S, Jian R, Jiang L, Snyder M, Chen CH, Shen Y, Liu Q, Liu B, Wu Z. The 40S ribosomal subunit recycling complex modulates mitochondrial dynamics and endoplasmic reticulum - mitochondria tethering at mitochondrial fission/fusion hotspots. Nat Commun 2025; 16:1021. [PMID: 39863576 PMCID: PMC11762756 DOI: 10.1038/s41467-025-56346-3] [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: 04/22/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
The 40S ribosomal subunit recycling pathway is an integral link in the cellular quality control network, occurring after translational errors have been corrected by the ribosome-associated quality control (RQC) machinery. Despite our understanding of its role, the impact of translation quality control on cellular metabolism remains poorly understood. Here, we reveal a conserved role of the 40S ribosomal subunit recycling (USP10-G3BP1) complex in regulating mitochondrial dynamics and function. The complex binds to fission-fusion proteins located at mitochondrial hotspots, regulating the functional assembly of endoplasmic reticulum-mitochondria contact sites (ERMCSs). Furthermore, it alters the activity of mTORC1/2 pathways, suggesting a link between quality control and energy fluctuations. Effective communication is essential for resolving proteostasis-related stresses. Our study illustrates that the USP10-G3BP1 complex acts as a hub that interacts with various pathways to adapt to environmental stimuli promptly. It advances our molecular understanding of RQC regulation and helps explain the pathogenesis of human proteostasis and mitochondrial dysfunction diseases.
Collapse
Affiliation(s)
- Foozhan Tahmasebinia
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, 75275, USA
| | - Yinglu Tang
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, 75275, USA
| | - Rushi Tang
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore
| | - Yi Zhang
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, 75275, USA
| | - Will Bonderer
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, 75275, USA
| | - Maisa de Oliveira
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, 75275, USA
| | - Bretton Laboret
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, 75275, USA
| | - Songjie Chen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ruiqi Jian
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Lihua Jiang
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, NHRI, Miaoli, 350401, Taiwan
| | - Yawei Shen
- Department of Biological Sciences, Clemson University, Clemson, SC, 29634, USA
- Center for Human Genetics, Clemson University, Greenwood, SC, 29646, USA
| | - Qing Liu
- Department of Biological Sciences, Clemson University, Clemson, SC, 29634, USA
- Center for Human Genetics, Clemson University, Greenwood, SC, 29646, USA
| | - Boxiang Liu
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, 117543, Singapore.
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117543, Singapore.
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Cardiovascular-Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117543, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117543, Singapore.
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Singapore, 138672, Singapore.
| | - Zhihao Wu
- Department of Biological Sciences, Dedman College of Humanities and Sciences, Southern Methodist University, Dallas, TX, 75275, USA.
| |
Collapse
|
31
|
Buchholz HE, Martin SA, Dorweiler JE, Radtke CM, Knier AS, Beans NB, Manogaran AL. Hsp70 chaperones, Ssa1 and Ssa2, limit poly(A) binding protein aggregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.17.633617. [PMID: 39896508 PMCID: PMC11785122 DOI: 10.1101/2025.01.17.633617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Molecular chaperones play a central role in maintaining protein homeostasis. The highly conserved Hsp70 family of chaperones have major functions in folding of nascent peptides, protein refolding, and protein aggregate disassembly. In yeast, loss of two Hsp70 proteins, Ssa1 and Ssa2, is associated with decreased cellular growth and shortened lifespan. While heterologous or mutant temperature sensitive proteins form anomalous large cytoplasmic inclusions in ssa1Δssa2Δ strains, it is unclear how endogenous wildtype proteins behave and are regulated in the presence of limiting Hsp70s. Using the wildtype yeast Poly A binding protein (Pab1), which is involved in mRNA binding and forms stress granules (SGs) upon heat shock, Pab1 forms large inclusions in approximately half of ssa1Δssa2Δ cells in the absence of stress. Overexpression of Ssa1, Hsp104, and Sis1 almost completely limits the formation of these large inclusions in ssa1Δssa2Δ, suggesting that excess Ssa1, Hsp104 and Sis1 can each compensate for the lower levels of Ssa proteins. Upon heat shock, SGs also form in cells whether large Pab1 inclusions are present or not. Surprisingly, cells containing only SGs disassemble faster than wildtype, whereas cells with both large inclusions disassemble slower albeit completely. We suspect that disassembly of these large inclusions is linked to the elevated heat shock response and elevated Hsp104 and Sis1 levels in ssa1Δssa2Δ strains. We also observed that wildtype cultures grown to saturation also form large Pab1-GFP inclusions. These inclusions can be partially rescued by overexpression of Ssa1. Taken together, our data suggests that Hsp70 not only plays a role in limiting unwanted protein aggregation in normal cells, but as cells age, the depletion of active Hsp70 possibly underlies the age-related aggregation of endogenous proteins.
Collapse
Affiliation(s)
- Hannah E Buchholz
- Department of Biological Sciences, Marquette University, Milwaukee, WI, 53201-1881 USA
| | - Sean A Martin
- Department of Biological Sciences, Marquette University, Milwaukee, WI, 53201-1881 USA
| | - Jane E Dorweiler
- Department of Biological Sciences, Marquette University, Milwaukee, WI, 53201-1881 USA
| | - Claire M Radtke
- Department of Biological Sciences, Marquette University, Milwaukee, WI, 53201-1881 USA
| | - Adam S Knier
- Department of Biological Sciences, Marquette University, Milwaukee, WI, 53201-1881 USA
| | - Natalia B Beans
- Department of Biological Sciences, Marquette University, Milwaukee, WI, 53201-1881 USA
| | - Anita L Manogaran
- Department of Biological Sciences, Marquette University, Milwaukee, WI, 53201-1881 USA
| |
Collapse
|
32
|
Cheng SJ, Gafaar T, Kuttiyatveetil JRA, Sverzhinsky A, Chen C, Xu M, Lilley A, Pascal JM, Leung AKL. Regulation of stress granule maturation and dynamics by poly(ADP-ribose) interaction with PARP13. Nat Commun 2025; 16:621. [PMID: 39805863 PMCID: PMC11731017 DOI: 10.1038/s41467-024-55666-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
Non-covalent interactions of poly(ADP-ribose) (PAR) facilitate condensate formation, yet the impact of these interactions on condensate properties remains unclear. Here, we demonstrate that PAR-mediated interactions through PARP13, specifically the PARP13.2 isoform, are essential for modulating the dynamics of stress granules-a class of cytoplasmic condensates that form upon stress, including types frequently observed in cancers. Single amino acid mutations in PARP13, which reduce its PAR-binding activity, lead to the formation of smaller yet more numerous stress granules than observed in the wild-type. This fragmented stress granule phenotype is also apparent in PARP13 variants with cancer-associated single-nucleotide polymorphisms (SNPs) that disrupt PAR binding. Notably, this fragmented phenotype is conserved across a variety of stresses that trigger stress granule formation via diverse pathways. Furthermore, this PAR-binding mutant diminishes condensate dynamics and impedes fusion. Overall, our study uncovers the important role of PAR-protein interactions in stress granule dynamics and maturation, mediated through PARP13.
Collapse
Affiliation(s)
- Shang-Jung Cheng
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Temitope Gafaar
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | | | - Aleksandr Sverzhinsky
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Carla Chen
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Minghui Xu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Allison Lilley
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - John M Pascal
- Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC, Canada
| | - Anthony K L Leung
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD, USA.
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University, Baltimore, MD, USA.
- Department of Oncology, School of Medicine, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
33
|
Li C, Liu Z, Fu C, Li H, He T, Wu G, Sheng Y, Shen M, Liu H. Hypoxia-induced degradation of FTO promotes apoptosis by unmasking RACK1-mediated activation of MTK1-JNK1/2 pathway. J Adv Res 2025:S2090-1232(25)00038-4. [PMID: 39805423 DOI: 10.1016/j.jare.2025.01.019] [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: 09/19/2024] [Revised: 01/09/2025] [Accepted: 01/11/2025] [Indexed: 01/16/2025] Open
Abstract
INTRODUCTION Hypoxia, a condition characterized by inadequate oxygen supply to tissues, triggers various cellular responses, including apoptosis. The RNA demethylase FTO has been shown to exert anti-apoptotic effects, but its functions independent of RNA demethylase-particularly those involving protein-protein interactions-during hypoxia remain unclear. OBJECTIVES This study aimed to elucidate the cytoprotective mechanism of FTO in preventing apoptosis under hypoxic stress. METHODS NIH/3T3 cells, MEF cells, and mouse granulosa cells were cultured under hypoxia (1 % O2) and treated with inhibitors (chloroquine, MG132, cycloheximide) to identify FTO degradation pathways. RNA interference was used to knock down atg7, nedd4, and fto. Mass spectrometry identified FTO-associated proteins, and their interactions with FTO were analyzed with immunoprecipitation assays. FTO localization was examined through nuclear and cytoplasmic fractionation and fluorescence microscopy. Apoptosis was evaluated by flow cytometry (annexin V/PI). The role of FTO independent of its m6A demethylase activity was determined by inhibiting FTO function using FB23-2 or an H228A/D230A mutant lacking m6A demethylase activity. RESULTS Upon hypoxia exposure, FTO relocated from the nucleus to the cytoplasm and underwent degradation through a regulatory pathway in which the E1-like ubiquitin-activating enzyme ATG7 and the E3 ubiquitin ligase NEDD4 cooperatively activated both the ubiquitin-proteasome system (UPS) and the autophagic-lysosomal pathway (ALP) in NIH/3T3 cells, MEF cells, and mouse granulosa cells. Furthermore, knocking down atg7 resulted in FTO accumulation in the cytoplasm, where FTO exerted its protective effect by binding with RACK1, which impairs the interaction between RACK1 and MTK1, thereby blocking activation of JNK1/2 and subsequently preventing apoptosis in hypoxic cells. CONCLUSION This study reveals a novel function of cytoplasmic FTO in disrupting the RACK1-MTK1-JNK1/2-apoptosis cascade during hypoxia, positioning the functional context of FTO at the layer of protein-protein interactions, which extends its mechanistic role beyond RNA demethylation.
Collapse
Affiliation(s)
- Chengyu Li
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Zhaojun Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Chen Fu
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Hongmin Li
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Tong He
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Gang Wu
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Yanan Sheng
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China
| | - Ming Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China.
| | - Honglin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Weigang 1, Nanjing 210095, China.
| |
Collapse
|
34
|
Akaree N, Secco V, Levy-Adam F, Younis A, Carra S, Shalgi R. Regulation of physiological and pathological condensates by molecular chaperones. FEBS J 2025. [PMID: 39756021 DOI: 10.1111/febs.17390] [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: 05/30/2024] [Revised: 11/17/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
Biomolecular condensates are dynamic membraneless compartments that regulate a myriad of cellular functions. A particular type of physiological condensate called stress granules (SGs) has gained increasing interest due to its role in the cellular stress response and various diseases. SGs, composed of several hundred RNA-binding proteins, form transiently in response to stress to protect mRNAs from translation and disassemble when the stress subsides. Interestingly, SGs contain several aggregation-prone proteins, such as TDP-43, FUS, hnRNPA1, and others, which are typically found in pathological inclusions seen in autopsy tissues from amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) patients. Moreover, mutations in these genes lead to the familial form of ALS and FTD. This has led researchers to propose that pathological aggregation is seeded by aberrant SGs: SGs that fail to properly disassemble, lose their dynamic properties, and become pathological condensates which finally 'mature' into aggregates. Here, we discuss the evidence supporting this model for various ALS/FTD-associated proteins. We further continue to focus on molecular chaperone-mediated regulation of ALS/FTD-associated physiological condensates on one hand, and pathological condensates on the other. In addition to SGs, we review ALS/FTD-relevant nuclear condensates, namely paraspeckles, anisosomes, and nucleolar amyloid bodies, and discuss their emerging regulation by chaperones. As the majority of chaperoning mechanisms regulate physiological condensate disassembly, we highlight parallel themes of physiological and pathological condensation regulation across different chaperone families, underscoring the potential for early disease intervention.
Collapse
Affiliation(s)
- Nadeen Akaree
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Valentina Secco
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy
| | - Flonia Levy-Adam
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Amal Younis
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Serena Carra
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Italy
| | - Reut Shalgi
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
35
|
Clark M, Farinha A, Morrison A, Lisi G. Structural, biological, and biomedical implications of mRNA interactions with the master regulator HuR. NAR MOLECULAR MEDICINE 2025; 2:ugaf002. [PMID: 39980665 PMCID: PMC11838611 DOI: 10.1093/narmme/ugaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/27/2025] [Accepted: 01/30/2025] [Indexed: 02/22/2025]
Abstract
Human antigen R (HuR) is a ubiquitously expressed RNA-binding protein (RBP) that has been implicated in a vast range of biological processes including stress response, angiogenesis, cell proliferation, and differentiation. Dysregulation of HuR has been linked to a number of pathological disorders including vascular disease, inflammation, and cancers such as those of the breast and colon. Like many RBPs, HuR is composed of multiple RNA-recognition motif (RRM) domains; however, HuR and the three other members of the Hu family (HuB, HuC, and HuD) possess a unique structural composition with two RRMs separated from a third C-terminal RRM by a long, unstructured hinge region. While there has been extensive research on the role of HuR in cellular, molecular, and developmental biology, there are fewer structural and biochemical studies of HuR and many questions still remain about the molecular mechanisms of HuR. In this review, we endeavor to synthesize existing HuR research spanning the last three decades in order to define known mechanistic roles of each domain, highlight remaining uncertainties, and provide a backdrop for ongoing research into the chemistry and biology of HuR and similar multi-RRM containing proteins.
Collapse
Affiliation(s)
- Madeline E Clark
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02903, United States
| | - Andrew Farinha
- Departments of Research and Medicine, Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, United States
| | - Alan R Morrison
- Departments of Research and Medicine, Vascular Research Laboratory, Providence VA Medical Center, Providence, RI 02908, United States
- Ocean State Research Institute, Inc., Providence, RI 02908, United States
- Department of Medicine, Section of Cardiology, Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - George P Lisi
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI 02903, United States
- Brown University RNA Center, Providence, RI 02903, United States
| |
Collapse
|
36
|
Friedson B, Willis SD, Shcherbik N, Campbell AN, Cooper KF. The CDK8 kinase module: A novel player in the transcription of translation initiation and ribosomal genes. Mol Biol Cell 2025; 36:ar2. [PMID: 39565680 PMCID: PMC11742111 DOI: 10.1091/mbc.e24-04-0164] [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: 04/12/2024] [Revised: 11/05/2024] [Accepted: 11/12/2024] [Indexed: 11/22/2024] Open
Abstract
Survival following stress is dependent upon reprogramming transcription and translation. Communication between these programs following stress is critical for adaptation but is not clearly understood. The Cdk8 kinase module (CKM) of the Mediator complex modulates the transcriptional response to various stresses. Its involvement in regulating translational machinery has yet to be elucidated, highlighting an existing gap in knowledge. Here, we report that the CKM positively regulates a subset of ribosomal protein (RP) and translation initiation factor (TIF)-encoding genes under physiological conditions in Saccharomyces cerevisiae. In mouse embryonic fibroblasts and HCT116 cells, the CKM regulates unique sets of RP and TIF genes, demonstrating some conservation of function across species. In yeast, this is mediated by Cdk8 phosphorylation of one or more transcription factors which control RP and TIF expression. Conversely, the CKM is disassembled following nutrition stress, permitting repression of RP and TIF genes. The CKM also plays a transcriptional role important for promoting cell survival, particularly during translational machinery stress triggered by ribosome-targeting antibiotics. Furthermore, in mammalian cells, the activity of CDK8 and its paralogue, CDK19, promotes cell survival following ribosome inhibition. These results provide mechanistic insights into the CKM's role in regulating expression of a subset of genes associated with translation.
Collapse
Affiliation(s)
- Brittany Friedson
- Department of Molecular Biology, Virtual Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| | - Stephen D. Willis
- Department of Molecular Biology, Virtual Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| | - Natalia Shcherbik
- Department of Molecular Biology, Virtual Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| | - Alicia N. Campbell
- Department of Molecular Biology, Virtual Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| | - Katrina F. Cooper
- Department of Molecular Biology, Virtual Health College of Medicine and Life Sciences, School of Osteopathic Medicine, Rowan University, Stratford, NJ 08084
| |
Collapse
|
37
|
Tu W, Theisen RQ, Jin P, Chenoweth DM, Patel AJ, Good MC. Delivery of Peptide Coacervates to Form Stable Interaction Hubs in Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.26.625566. [PMID: 39651133 PMCID: PMC11623604 DOI: 10.1101/2024.11.26.625566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Cells contain membrane-bound and membraneless organelles that operate as spatially distinct biochemical niches. However, these subcellular reaction centers lose fidelity with aging and as a result of disease. A grand challenge for biomedicine is restoring or augmenting cellular functionalities. Although commonly tackled by gene replacement therapy, an excited new strategy is the delivery of protein-based materials that can directly interact with and alter biological networks inside a cell. In this study we sought to develop long-lasting materials capable of cellular uptake and incorporation, akin to an artificial organelle or intracellular interaction hub. Drawing inspiration from protein-based membranelles organelles, we developed a new delivery method to transplant micron size peptide-based compartments into living cells. We determined conditions to form large stable coacervates that are efficiently taken up by a variety of useful cell types and demonstrate their intracellular stability over time. We developed tools to enhance the extent and spatial organization of cargo loading into these coacervates, including co-assembly of nanobodies that selectively bind to targets of interest. Combining them together, we demonstrate successful targeting of GFP protein inside cells. These results represent an important first step toward the development of deliverable synthetic organelles that can be fabricated in vitro and taken up by cells for applications in cell engineering and regenerative medicine.
Collapse
Affiliation(s)
- Wangjie Tu
- Bioengineering Graduate Group, University of Pennsylvania, PA 19104
| | - Rachel Q. Theisen
- Department of Cell and Developmental Biology, University of Pennsylvania, PA 19104
| | - Pengfei Jin
- Chemistry Graduate Group, University of Pennsylvania, PA 19104
| | - David M. Chenoweth
- Chemistry Graduate Group, University of Pennsylvania, PA 19104
- Department of Chemistry, University of Pennsylvania, PA 19104
| | - Amish J. Patel
- Chemical and Biomolecular Engineering Department, University of Pennsylvania, PA 19104
| | - Matthew C. Good
- Bioengineering Graduate Group, University of Pennsylvania, PA 19104
- Department of Cell and Developmental Biology, University of Pennsylvania, PA 19104
- Department of Bioengineering, University of Pennsylvania, PA 19104
| |
Collapse
|
38
|
Kim J, Song CH. Stress Granules in Infectious Disease: Cellular Principles and Dynamic Roles in Immunity and Organelles. Int J Mol Sci 2024; 25:12950. [PMID: 39684660 DOI: 10.3390/ijms252312950] [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: 11/19/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Stress granules (SGs) are membrane-less aggregates that form in response to various cellular stimuli through a process called liquid-liquid phase separation (LLPS). Stimuli such as heat shock, osmotic stress, oxidative stress, and infections can induce the formation of SGs, which play crucial roles in regulating gene expression to help cells adapt to stress conditions. Various mRNAs and proteins are aggregated into SGs, particularly those associated with the protein translation machinery, which are frequently found in SGs. When induced by infections, SGs modulate immune cell activity, supporting the cellular response against infection. The roles of SGs differ in viral versus microbial infections, and depending on the type of immune cell involved, SGs function differently in response to infection. In this review, we summarize our current understanding of the implication of SGs in immunity and cellular organelles in the context of infectious diseases. Importantly, we explore insights into the regulatory functions of SGs in the context of host cells under infection.
Collapse
Affiliation(s)
- Jaewhan Kim
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Chang-Hwa Song
- Department of Medical Science, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
- Department of Microbiology, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| |
Collapse
|
39
|
Raval K, Jamshidi N, Seyran B, Salwinski L, Pillai R, Yang L, Ma F, Pellegrini M, Shin J, Yang X, Tudzarova S. Dysfunctional β-cell longevity in diabetes relies on energy conservation and positive epistasis. Life Sci Alliance 2024; 7:e202402743. [PMID: 39313296 PMCID: PMC11420665 DOI: 10.26508/lsa.202402743] [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: 03/27/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024] Open
Abstract
Long-lived PFKFB3-expressing β-cells are dysfunctional partly because of prevailing glycolysis that compromises metabolic coupling of insulin secretion. Their accumulation in type 2 diabetes (T2D) appears to be related to the loss of apoptotic competency of cell fitness competition that maintains islet function by favoring constant selection of healthy "winner" cells. To investigate how PFKFB3 can disguise the competitive traits of dysfunctional "loser" β-cells, we analyzed the overlap between human β-cells with bona fide "loser signature" across diabetes pathologies using the HPAP scRNA-seq and spatial transcriptomics of PFKFB3-positive β-cells from nPOD T2D pancreata. The overlapping transcriptional profile of "loser" β-cells was represented by down-regulated ribosomal biosynthesis and genes encoding for mitochondrial respiration. PFKFB3-positive "loser" β-cells had the reduced expression of HLA class I and II genes. Gene-gene interaction analysis revealed that PFKFB3 rs1983890 can interact with the anti-apoptotic gene MAIP1 implicating positive epistasis as a mechanism for prolonged survival of "loser" β-cells in T2D. Inhibition of PFKFB3 resulted in the clearance of dysfunctional "loser" β-cells leading to restored glucose tolerance in the mouse model of T2D.
Collapse
Affiliation(s)
- Kavit Raval
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Neema Jamshidi
- Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Berfin Seyran
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Lukasz Salwinski
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Raju Pillai
- Department of Pathology, City-of-Hope, Duarte, CA, USA
| | - Lixin Yang
- Department of Pathology, City-of-Hope, Duarte, CA, USA
| | - Feiyang Ma
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Matteo Pellegrini
- Molecular Cell and Developmental Biology, College of Life Sciences, University of California Los Angeles, Los Angeles, CA, USA
| | - Juliana Shin
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Xia Yang
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Slavica Tudzarova
- Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
40
|
Li T, Zhen H, Wu W, Yang F, Cao Z. tsRNAs: A Prospective, Effective Therapeutic Intervention for Neurodegenerative Diseases. CNS Neurosci Ther 2024; 30:e70177. [PMID: 39690867 DOI: 10.1111/cns.70177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/11/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Neurological disorders known as neurodegenerative diseases (NDDs) result in the slow loss of neurons in the central nervous system (CNS) or peripheral nervous system (PNS), as well as the collapse of neural networks in terms of structure and function. NDDs are expected to surpass cancer as the second biggest cause of mortality by 2040, according to World Health Organization (WHO) estimations. Neurons cannot effectively regenerate themselves because they are terminally differentiated. Accordingly, it is challenging to find medications that could stop or slow neurodegeneration. MAIN BODY The tsRNAs are a type of small non-coding RNAs derived from mature tRNAs or tRNA precursors. tsRNAs control gene expression and have a role in many physiological and pathological processes, including neurological illnesses. Antisense oligonucleotides are effective therapeutic agents for neurological diseases, and they may be the treatment of choice for neurodegenerative diseases in the future. Here, we review the biogenesis of tsRNA, its physiological and pathological functions in the central nervous system and neurological disorders, and its prospective use as a nucleic acid medication to treat NDDs, providing theoretical support and guidance for further exploration of tsRNAs in therapeutic intervention. CONCLUSION tsRNAs are emerging as important regulatory molecules in neurodegenerative diseases. Understanding the functions of tsRNAs in neurodegenerative diseases may provide new insights into disease mechanisms and lead to the development of novel treatment strategies.
Collapse
Affiliation(s)
- Tianqi Li
- School of Life Science and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Hui Zhen
- School of Life Science and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Weiwei Wu
- School of Life Science and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Fengtang Yang
- School of Life Science and Medicine, Shandong University of Technology, Zibo, Shandong, China
| | - Zhonghong Cao
- School of Life Science and Medicine, Shandong University of Technology, Zibo, Shandong, China
| |
Collapse
|
41
|
Li Z, Luo L, Ju X, Huang S, Lei L, Yu Y, Liu J, Zhang P, Chi T, Ma P, Huang C, Huang X, Ding Q, Zhang Y. Viral N protein hijacks deaminase-containing RNA granules to enhance SARS-CoV-2 mutagenesis. EMBO J 2024; 43:6444-6468. [PMID: 39567830 PMCID: PMC11649915 DOI: 10.1038/s44318-024-00314-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 10/28/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
Host cell-encoded deaminases act as antiviral restriction factors to impair viral replication and production through introducing mutations in the viral genome. We sought to understand whether deaminases are involved in SARS-CoV-2 mutation and replication, and how the viral factors interact with deaminases to trigger these processes. Here, we show that APOBEC and ADAR deaminases act as the driving forces for SARS-CoV-2 mutagenesis, thereby blocking viral infection and production. Mechanistically, SARS-CoV-2 nucleocapsid (N) protein, which is responsible for packaging viral genomic RNA, interacts with host deaminases and co-localizes with them at stress granules to facilitate viral RNA mutagenesis. N proteins from several coronaviruses interact with host deaminases at RNA granules in a manner dependent on its F17 residue, suggesting a conserved role in modulation of viral mutagenesis in other coronaviruses. Furthermore, mutant N protein bearing a F17A substitution cannot localize to deaminase-containing RNA granules and leads to reduced mutagenesis of viral RNA, providing support for its function in enhancing deaminase-dependent viral RNA editing. Our study thus provides further insight into virus-host cell interactions mediating SARS-CoV-2 evolution.
Collapse
Affiliation(s)
- Zhean Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingling Luo
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- The Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Xiaohui Ju
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Shisheng Huang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Liqun Lei
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yanying Yu
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Jia Liu
- Shanghai Institute for Advanced Immunochemical Studies, ShanghaiTech University, Shanghai, China
| | - Pumin Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian Chi
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China
| | - Peixiang Ma
- Shanghai Key Laboratory of Orthopedic Implants, Department of Orthopedic Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, Guangdong, China
| | - Cheng Huang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Xingxu Huang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China.
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
| | - Qiang Ding
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China.
| | - Yu Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, China.
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.
| |
Collapse
|
42
|
Li F, Wang W, Cheng H, Li M. Genome-wide analysis reveals the contributors to fast molecular evolution of the Chinese hook snout carp ( Opsariichthys bidens). Comput Struct Biotechnol J 2024; 23:2465-2477. [PMID: 38882676 PMCID: PMC11179538 DOI: 10.1016/j.csbj.2024.05.048] [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: 03/07/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/18/2024] Open
Abstract
Variations in molecular evolutionary rate have been widely investigated among lineages and genes. However, it remains an open question whether fast rate of molecular evolution is driven by natural selection or random drift, and how the fast rate is linked to metabolic rate. Additionally, previous studies on fast molecular evolution have been largely restricted to concatenated matrix of genes or a few specifically selected genes, but less is known for individual genes at the genome-wide level. Here we addressed these questions using more than 5000 single-copy orthologous (SCO) genes through comparative genomic and phylogenetic analyses among fishes, with a special focus on a newly-sequenced clupeocephalan fish the Chinese hook snout carp Opsariichthys bidens. We showed O. bidens displays significantly higher mean substitution rate and more fast-evolving SCO genes (2172 genes) than most fishes studied here. The rapidly evolving genes are enriched in highly conserved and very basic functions such as translation and ribosome that are critical for biological fitness. We further revealed that ∼25 % of these fast-evolving genes exhibit a constant increase of substitution rate from the common ancestor down to the present, suggesting a neglected but important contribution from ancestral states. Model fitting showed that ∼85 % of fast-evolving genes exclusive to O. bidens and related species follow the adaptive evolutionary model rather than random-drift model, and 7.6 % of fast-evolving genes identified in O. bidens have experienced positive selection, both indicating the reflection of adaptive selection. Finally, metabolic rate was observed to be linked with substitution rate in a gene-specific manner. Overall, our findings reveal fast molecular evolution of SCO genes at genome-wide level in O. bidens, and uncover the evolutionary and ecological contributors to it.
Collapse
Affiliation(s)
- Fengbo Li
- Zhejiang Institute of Freshwater Fisheries, 999 Hangchangqiao South Road, Huzhou 313001, China
| | - Wei Wang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, 1 Beichen West Road, Beijing 100101, China
| | - Haihua Cheng
- Zhejiang Institute of Freshwater Fisheries, 999 Hangchangqiao South Road, Huzhou 313001, China
| | - Ming Li
- Jinhua Fisheries Technology Extension Center, 828 Shuanglong South Street, Jinhua 321013, China
| |
Collapse
|
43
|
Cheng SS, Mody AC, Woo CM. Opportunities for Therapeutic Modulation of O-GlcNAc. Chem Rev 2024; 124:12918-13019. [PMID: 39509538 DOI: 10.1021/acs.chemrev.4c00417] [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: 11/15/2024]
Abstract
O-Linked β-N-acetylglucosamine (O-GlcNAc) is an essential, dynamic monosaccharide post-translational modification (PTM) found on serine and threonine residues of thousands of nucleocytoplasmic proteins. The installation and removal of O-GlcNAc is controlled by a single pair of enzymes, O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA), respectively. Since its discovery four decades ago, O-GlcNAc has been found on diverse classes of proteins, playing important functional roles in many cellular processes. Dysregulation of O-GlcNAc homeostasis has been implicated in the pathogenesis of disease, including neurodegeneration, X-linked intellectual disability (XLID), cancer, diabetes, and immunological disorders. These foundational studies of O-GlcNAc in disease biology have motivated efforts to target O-GlcNAc therapeutically, with multiple clinical candidates under evaluation. In this review, we describe the characterization and biochemistry of OGT and OGA, cellular O-GlcNAc regulation, development of OGT and OGA inhibitors, O-GlcNAc in pathophysiology, clinical progress of O-GlcNAc modulators, and emerging opportunities for targeting O-GlcNAc. This comprehensive resource should motivate further study into O-GlcNAc function and inspire strategies for therapeutic modulation of O-GlcNAc.
Collapse
Affiliation(s)
- Steven S Cheng
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Alison C Mody
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
| | - Christina M Woo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts 02138, United States
- Affiliate member of the Broad Institute, Cambridge, Massachusetts 02142, United States
| |
Collapse
|
44
|
Manjunath L, Santiago G, Ortega P, Sanchez A, Oh S, Garcia A, Bournique E, Bouin A, Semler BL, Setiaputra D, Buisson R. Cooperative Role of PACT and ADAR1 in Preventing Aberrant PKR Activation by Self-Derived Double-Stranded RNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625712. [PMID: 39651230 PMCID: PMC11623655 DOI: 10.1101/2024.11.27.625712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Double-stranded RNAs (dsRNAs) produced during viral infections are recognized by the innate immune sensor protein kinase R (PKR), triggering a host translation shutoff that inhibits viral replication and propagation. Given the harmful effects of uncontrolled PKR activation, cells must tightly regulate PKR to ensure that its activation occurs only in response to viral infections, not endogenous dsRNAs. Here, we use CRISPR-Translate, a FACS-based genome-wide CRISPR-Cas9 knockout screening method that exploits translation levels as a readout and identifies PACT as a key inhibitor of PKR during viral infection. We find that cells deficient for PACT hyperactivate PKR in response to several different RNA viruses, raising the question of why cells need to limit PKR activity. Our results demonstrate that PACT cooperates with ADAR1 to suppress PKR activation from self-dsRNAs in uninfected cells. The simultaneous deletion of PACT and ADAR1 results in synthetic lethality, which can be fully rescued in PKR-deficient cells. We propose that both PACT and ADAR1 act as essential barriers against PKR, creating a threshold of tolerable levels to endogenous dsRNA in cells without activating PKR-mediated translation shutdown and cell death.
Collapse
|
45
|
Patni D, Patil AD, Kirmire MS, Jha A, Jha SK. DNA-Mediated Formation of Phase-Separated Coacervates of the Nucleic Acid-Binding Domain of TAR DNA-Binding Protein (TDP-43) Prevents Its Amyloid-Like Misfolding. ACS Chem Neurosci 2024; 15:4105-4122. [PMID: 39471356 DOI: 10.1021/acschemneuro.4c00117] [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: 11/01/2024] Open
Abstract
Sequestration of protein molecules and nucleic acids to stress granules is one of the most promising strategies that cells employ to protect themselves from stress. In vitro, studies suggest that the nucleic acid-binding domain of TDP-43 (TDP-43tRRM) undergoes amyloid-like aggregation to β-sheet-rich structures in low pH stress. In contrast, we observed that the TDP-43tRRM undergoes complex coacervation in the presence of ssDNA to a dense and light phase, preventing its amyloid-like aggregation. The soluble light phase consists of monomeric native-like TDP-43tRRM. The microscopic data suggest that the dense phase consists of spherical coacervates with limited internal dynamics. We performed multiparametric analysis by employing various biophysical techniques and found that complex coacervation depends on the concentration and ratio of the participating biomolecules and is driven by multivalent interactions. The modulation of these forces due to environmental conditions or disease mutations regulates the extent of coacervation, and the weakening of interactions between TDP-43tRRM and ssDNA leads to amyloid-like aggregation of TDP-43tRRM. Our results highlight a competition among the native state, amyloid-like aggregates, and complex coacervates tuned by various environmental factors. Together, our results illuminate an alternate function of TDP-43tRRM in response to pH stress in the presence of the ssDNA.
Collapse
Affiliation(s)
- Divya Patni
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anjali D Patil
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mona S Kirmire
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Anjali Jha
- MIT School of Bioengineering Sciences and Research, MIT-ADT University, Loni Kalbhor, Pune 412 201, India
| | - Santosh Kumar Jha
- Physical and Materials Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411008, Maharashtra, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
46
|
Amico T, Dada ST, Lazzari A, Brezinova M, Trovato A, Vendruscolo M, Fuxreiter M, Maritan A. A scale-invariant log-normal droplet size distribution below the critical concentration for protein phase separation. eLife 2024; 13:RP94214. [PMID: 39556435 PMCID: PMC11573347 DOI: 10.7554/elife.94214] [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: 11/19/2024] Open
Abstract
Many proteins have been recently shown to undergo a process of phase separation that leads to the formation of biomolecular condensates. Intriguingly, it has been observed that some of these proteins form dense droplets of sizeable dimensions already below the critical concentration, which is the concentration at which phase separation occurs. To understand this phenomenon, which is not readily compatible with classical nucleation theory, we investigated the properties of the droplet size distributions as a function of protein concentration. We found that these distributions can be described by a scale-invariant log-normal function with an average that increases progressively as the concentration approaches the critical concentration from below. The results of this scaling analysis suggest the existence of a universal behaviour independent of the sequences and structures of the proteins undergoing phase separation. While we refrain from proposing a theoretical model here, we suggest that any model of protein phase separation should predict the scaling exponents that we reported here from the fitting of experimental measurements of droplet size distributions. Furthermore, based on these observations, we show that it is possible to use the scale invariance to estimate the critical concentration for protein phase separation.
Collapse
Affiliation(s)
- Tommaso Amico
- Department of Physics and Astronomy, University of PadovaPadovaItaly
| | - Samuel Toluwanimi Dada
- Centre for Misfolding Diseases, Department of Chemistry, University of CambridgeCambridgeUnited Kingdom
| | - Andrea Lazzari
- Department of Physics and Astronomy, University of PadovaPadovaItaly
| | - Michaela Brezinova
- Centre for Misfolding Diseases, Department of Chemistry, University of CambridgeCambridgeUnited Kingdom
| | - Antonio Trovato
- Department of Physics and Astronomy, University of PadovaPadovaItaly
- National Institute for Nuclear Physics (INFN), Padova SectionPadovaItaly
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of CambridgeCambridgeUnited Kingdom
| | - Monika Fuxreiter
- Department of Physics and Astronomy, University of PadovaPadovaItaly
- Department of Biomedical Sciences, University of PadovaPadovaItaly
| | - Amos Maritan
- Department of Physics and Astronomy, University of PadovaPadovaItaly
- National Institute for Nuclear Physics (INFN), Padova SectionPadovaItaly
| |
Collapse
|
47
|
He SL, Wang X, Kim SI, Kong L, Liu A, Wang L, Wang Y, Shan L, He P, Jang JC. Modulation of stress granule dynamics by phosphorylation and ubiquitination in plants. iScience 2024; 27:111162. [PMID: 39569378 PMCID: PMC11576400 DOI: 10.1016/j.isci.2024.111162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/22/2024] Open
Abstract
The Arabidopsis tandem CCCH zinc finger 1 (TZF1) is an RNA-binding protein that plays a pivotal role in plant growth and stress response. In this report, we show that TZF1 contains two intrinsically disordered regions necessary for its localization to stress granules (SGs). TZF1 recruits mitogen-activated protein kinase (MAPK) signaling components and an E3 ubiquitin ligase KEEP-ON-GOING (KEG) to SGs. TZF1 is phosphorylated by MPKs and ubiquitinated by KEG. Using a high throughput Arabidopsis protoplasts transient expression system, mutant studies reveal that the phosphorylation of specific residues plays differential roles in enhancing or reducing TZF1 SG assembly and protein-protein interaction with mitogen-activated kinase kinase 5 in SGs. Ubiquitination appears to play a positive role in TZF1 SG assembly, because mutations cause a reduction of typical SGs, while enhancing the assembly of large SGs encompassing the nucleus. Together, our results demonstrate that plant SG assembly is distinctively regulated by phosphorylation and ubiquitination.
Collapse
Affiliation(s)
- Siou-Luan He
- Department of Horticulture and Crop Science, Center for Applied Plant Sciences, and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Xiling Wang
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing 10093, China
| | - Sung-Il Kim
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085, USA
| | - Liang Kong
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085, USA
| | - Ailing Liu
- Department of Horticulture and Crop Science, Center for Applied Plant Sciences, and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Lei Wang
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing 10093, China
| | - Ying Wang
- Plant Pathology Department and Plant Molecular Cellular Biology Program, University of Florida, Gainesville, FL 32611, USA
| | - Libo Shan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085, USA
| | - Ping He
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085, USA
| | - Jyan-Chyun Jang
- Department of Horticulture and Crop Science, Center for Applied Plant Sciences, and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
48
|
Liboy-Lugo JM, Espinoza CA, Sheu-Gruttadauria J, Park JE, Xu A, Jowhar Z, Gao AL, Carmona-Negrón JA, Wittmann T, Jura N, Floor SN. G3BP isoforms differentially affect stress granule assembly and gene expression during cellular stress. Mol Biol Cell 2024; 35:ar140. [PMID: 39356796 PMCID: PMC11617104 DOI: 10.1091/mbc.e24-02-0062] [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: 02/07/2024] [Revised: 09/16/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
Stress granules (SGs) are macromolecular assemblies that form under cellular stress. Formation of these membraneless organelles is driven by the condensation of RNA and RNA-binding proteins such as G3BPs. G3BPs form SGs following stress-induced translational arrest. Three G3BP paralogues (G3BP1, G3BP2A, and G3BP2B) have been identified in vertebrates. However, the contribution of different G3BP paralogues to SG formation and gene expression changes is incompletely understood. Here, we probed the functions of G3BPs by identifying important residues for SG assembly at their N-terminal domain such as V11. This conserved amino acid is required for formation of the G3BP-Caprin-1 complex, hence promoting SG assembly. Total RNA sequencing and ribosome profiling revealed that a G3BPV11A mutant leads to changes in mRNA levels and ribosome engagement during the integrated stress response (ISR). Moreover, we found that G3BP2B preferentially forms SGs and promotes changes in mRNA expression under endoplasmic reticulum (ER) stress. Furthermore, our work is a resource for researchers to study gene expression changes under cellular stress. Together, this work suggests that perturbing protein-protein interactions mediated by G3BPs affect SG assembly and gene expression during the ISR, and such functions are differentially regulated by G3BP paralogues under ER stress.
Collapse
Affiliation(s)
- José M. Liboy-Lugo
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94158
| | - Carla A. Espinoza
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94158
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| | - Jessica Sheu-Gruttadauria
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
| | - Jesslyn E. Park
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
| | - Albert Xu
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
| | - Ziad Jowhar
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
- Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143
| | - Angela L. Gao
- Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA 94158
| | - José A. Carmona-Negrón
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158
- Department of Chemistry, University of Puerto Rico, Mayagüez, PR 00680
| | - Torsten Wittmann
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
| | - Natalia Jura
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA 94158
| | - Stephen N. Floor
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
49
|
Qi X, Zhao R, Yao X, Liu Q, Liu P, Zhu Z, Tu C, Gong W, Li X. Getah virus Nsp3 binds G3BP to block formation of bona fide stress granules. Int J Biol Macromol 2024; 279:135274. [PMID: 39226976 DOI: 10.1016/j.ijbiomac.2024.135274] [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: 03/17/2024] [Revised: 08/16/2024] [Accepted: 08/31/2024] [Indexed: 09/05/2024]
Abstract
Stress granules (SGs) are cytoplasmic aggregates of proteins and mRNA that form in response to diverse environmental stressors, including viral infections. Several viruses possess the ability to block the formation of stress granules by targeting the SGs marker protein G3BP. However, the molecular functions and mechanisms underlying the regulation of SGs formation by Getah virus (GETV) remain unclear. In this study, we found that GETV infection triggered the formation of Nsp3-G3BP aggregates, which differed in composition from SGs. Further studies revealed that the presence of these aggregates was dependent on the activation of the PKR/eIF2α signaling pathway. Interestingly, we found that Nsp3 HVD domain blocked the formation of SGs by binding to G3BP NTF2 domain. Moreover, knockout of G3BP in NCI-H1299 cells had no effect on GETV replication, while overexpression of G3BP to form the genuine SGs significantly inhibited GETV replication. Overall, our study elucidates a novel role GETV Nsp3 to change the composition of SG as well as cellular stress response.
Collapse
Affiliation(s)
- Xiaoyi Qi
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Ruihan Zhao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiaohui Yao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Qinqiu Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Panrao Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhenbang Zhu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Changchun Tu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Wenjie Gong
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiangdong Li
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
50
|
Shibuya R, Kajimoto S, Yaginuma H, Ariyoshi T, Okada Y, Nakabayashi T. Nucleic Acid-Rich Stress Granules Are Not Merely Crowded Condensates: A Quantitative Raman Imaging Study. Anal Chem 2024; 96:17078-17085. [PMID: 39405087 PMCID: PMC11525929 DOI: 10.1021/acs.analchem.4c01096] [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: 02/28/2024] [Revised: 09/25/2024] [Accepted: 10/04/2024] [Indexed: 10/30/2024]
Abstract
Liquid droplets, formed by intracellular liquid-liquid phase separation (LLPS), are called membraneless organelles. They provide transient enzymatic reaction fields for maintaining cellular homeostasis, although they might transform into aggregates, leading to neurodegenerative diseases. To understand the nature of intracellular droplets, it is crucial to quantify the liquid droplets inside a living cell as well as to elucidate the underlying biological mechanism. In this study, we performed near-infrared fluorescence and Raman imaging to quantify chemical components inside stress granules (SGs) formed by LLPS in living cells. The Raman images reveal that the nucleic acid concentration inside the SGs was more than 20% higher than the surrounding cytoplasm, whereas the lipid concentration was lower. Quantitative Raman intensity analysis using a water Raman band as an internal standard enables in situ concentration determination of nucleic acids in the SGs and other organelles. The intensity of the biomolecular C-H bands relative to the water band indicates that the crowding environment inside the SGs depends on the stress type; under oxidative stress, the inside of the SGs was nearly identical to the outside, whereas it was sparser in hyperosmotic stressed cells, suggesting that the high concentrations of nucleic acids play a pivotal role in maintaining the environments inside the SGs. These results demonstrate that intracellular droplets are not always highly condensed.
Collapse
Affiliation(s)
- Ren Shibuya
- Graduate
School of Pharmaceutical Sciences, Tohoku
University, Aoba-ku, Sendai 980-8578, Japan
| | - Shinji Kajimoto
- Graduate
School of Pharmaceutical Sciences, Tohoku
University, Aoba-ku, Sendai 980-8578, Japan
- JST
PRESTO, Kawaguchi, Saitama 332-0012, Japan
| | - Hideyuki Yaginuma
- Department
of Cell Biology and Physics, Universal Biology Institute and International
Research Center for Neurointelligence, The
University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory
for Cell Polarity Regulation, RIKEN Center
for Biosystems Dynamics Research, Suita, Osaka 565-0874, Japan
| | - Tetsuro Ariyoshi
- Department
of Cell Biology and Physics, Universal Biology Institute and International
Research Center for Neurointelligence, The
University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory
for Cell Polarity Regulation, RIKEN Center
for Biosystems Dynamics Research, Suita, Osaka 565-0874, Japan
| | - Yasushi Okada
- Department
of Cell Biology and Physics, Universal Biology Institute and International
Research Center for Neurointelligence, The
University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
- Laboratory
for Cell Polarity Regulation, RIKEN Center
for Biosystems Dynamics Research, Suita, Osaka 565-0874, Japan
| | - Takakazu Nakabayashi
- Graduate
School of Pharmaceutical Sciences, Tohoku
University, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|