1
|
He F, Zheng Y, Elsabagh M, Fan K, Zha X, Zhang B, Wang M, Zhang H. Gut microbiota modulate intestinal inflammation by endoplasmic reticulum stress-autophagy-cell death signaling axis. J Anim Sci Biotechnol 2025; 16:63. [PMID: 40312439 PMCID: PMC12046778 DOI: 10.1186/s40104-025-01196-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/17/2025] [Indexed: 05/03/2025] Open
Abstract
The intestinal tract, a complex organ responsible for nutrient absorption and digestion, relies heavily on a balanced gut microbiome to maintain its integrity. Disruptions to this delicate microbial ecosystem can lead to intestinal inflammation, a hallmark of inflammatory bowel disease (IBD). While the role of the gut microbiome in IBD is increasingly recognized, the underlying mechanisms, particularly those involving endoplasmic reticulum (ER) stress, autophagy, and cell death, remain incompletely understood. ER stress, a cellular response to various stressors, can trigger inflammation and cell death. Autophagy, a cellular degradation process, can either alleviate or exacerbate ER stress-induced inflammation, depending on the specific context. The gut microbiome can influence both ER stress and autophagy pathways, further complicating the interplay between these processes. This review delves into the intricate relationship between ER stress, autophagy, and the gut microbiome in the context of intestinal inflammation. By exploring the molecular mechanisms underlying these interactions, we aim to provide a comprehensive theoretical framework for developing novel therapeutic strategies for IBD. A deeper understanding of the ER stress-autophagy axis, the gut microbial-ER stress axis, and the gut microbial-autophagy axis may pave the way for targeted interventions to restore intestinal health and mitigate the impact of IBD.
Collapse
Affiliation(s)
- Feiyang He
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
- Key Laboratory of Fujian Universities Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, 364012, P. R. China
| | - Yi Zheng
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Mabrouk Elsabagh
- Department of Animal Production and Technology, Faculty of Agricultural Sciences and Technologies, Niğde Ömermer Halisdemir University, Nigde, 51240, Turkey
| | - Kewei Fan
- Key Laboratory of Fujian Universities Preventive Veterinary Medicine and Biotechnology, Longyan University, Longyan, 364012, P. R. China
| | - Xia Zha
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Bei Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China
- State Key Laboratory of Sheep Genetic Improvement and Healthy Production, Xinjiang Academy of Agricultural Reclamation Science, Shihezi, 832000, P. R. China
| | - Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, P. R. China.
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, 225009, P. R. China.
| |
Collapse
|
2
|
Renner DM, Parenti NA, Bracci N, Weiss SR. Betacoronaviruses Differentially Activate the Integrated Stress Response to Optimize Viral Replication in Lung-Derived Cell Lines. Viruses 2025; 17:120. [PMID: 39861909 PMCID: PMC11769277 DOI: 10.3390/v17010120] [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: 12/23/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The betacoronavirus genus contains five of the seven human coronaviruses, making it a particularly critical area of research to prepare for future viral emergence. We utilized three human betacoronaviruses, one from each subgenus-HCoV-OC43 (embecovirus), SARS-CoV-2 (sarbecovirus), and MERS-CoV (merbecovirus)-, to study betacoronavirus interactions with the PKR-like ER kinase (PERK) pathway of the integrated stress response (ISR)/unfolded protein response (UPR). The PERK pathway becomes activated by an abundance of unfolded proteins within the endoplasmic reticulum (ER), leading to phosphorylation of eIF2α and translational attenuation. We demonstrate that MERS-CoV, HCoV-OC43, and SARS-CoV-2 all activate PERK and induce responses downstream of p-eIF2α, while only SARS-CoV-2 induces detectable p-eIF2α during infection. Using a small molecule inhibitor of eIF2α dephosphorylation, we provide evidence that MERS-CoV and HCoV-OC43 maximize viral replication through p-eIF2α dephosphorylation. Interestingly, genetic ablation of growth arrest and DNA damage-inducible protein (GADD34) expression, an inducible protein phosphatase 1 (PP1)-interacting partner targeting eIF2α for dephosphorylation, did not significantly alter HCoV-OC43 or SARS-CoV-2 replication, while siRNA knockdown of the constitutive PP1 partner, constitutive repressor of eIF2α phosphorylation (CReP), dramatically reduced HCoV-OC43 replication. Combining GADD34 knockout with CReP knockdown had the maximum impact on HCoV-OC43 replication, while SARS-CoV-2 replication was unaffected. Overall, we conclude that eIF2α dephosphorylation is critical for efficient protein production and replication during MERS-CoV and HCoV-OC43 infection. SARS-CoV-2, however, appears to be insensitive to p-eIF2α and, during infection, may even downregulate dephosphorylation to limit host translation.
Collapse
Affiliation(s)
- David M. Renner
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (D.M.R.); (N.A.P.); (N.B.)
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicholas A. Parenti
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (D.M.R.); (N.A.P.); (N.B.)
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nicole Bracci
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (D.M.R.); (N.A.P.); (N.B.)
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susan R. Weiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (D.M.R.); (N.A.P.); (N.B.)
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
3
|
Renner DM, Parenti NA, Weiss SR. BETACORONAVIRUSES DIFFERENTIALLY ACTIVATE THE INTEGRATED STRESS RESPONSE TO OPTIMIZE VIRAL REPLICATION IN LUNG DERIVED CELL LINES. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614975. [PMID: 39386680 PMCID: PMC11463420 DOI: 10.1101/2024.09.25.614975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The betacoronavirus genus contains five of the seven human viruses, making it a particularly critical area of research to prepare for future viral emergence. We utilized three human betacoronaviruses, one from each subgenus- HCoV-OC43 (embecovirus), SARS-CoV-2 (sarbecovirus) and MERS-CoV (merbecovirus)- to study betacoronavirus interaction with the PKR-like ER kinase (PERK) pathway of the integrated stress response (ISR)/unfolded protein response (UPR). The PERK pathway becomes activated by an abundance of unfolded proteins within the endoplasmic reticulum (ER), leading to phosphorylation of eIF2α and translational attenuation in lung derived cell lines. We demonstrate that MERS-CoV, HCoV-OC43, and SARS-CoV-2 all activate PERK and induce responses downstream of p-eIF2α, while only SARS-CoV-2 induces detectable p-eIF2α during infection. Using a small molecule inhibitor of eIF2α dephosphorylation, we provide evidence that MERS-CoV and HCoV-OC43 maximize replication through p-eIF2α dephosphorylation. Interestingly, genetic ablation of GADD34 expression, an inducible phosphatase 1 (PP1)-interacting partner targeting eIF2α for dephosphorylation, did not significantly alter HCoV-OC43 or SARS-CoV-2 replication, while siRNA knockdown of the constitutive PP1 partner, CReP, dramatically reduced HCoV-OC43 replication. Combining growth arrest and DNA damage-inducible protein (GADD34) knockout with peripheral ER membrane-targeted protein (CReP) knockdown had the maximum impact on HCoV-OC43 replication, while SARS-CoV-2 replication was unaffected. Overall, we conclude that eIF2α dephosphorylation is critical for efficient protein production and replication during MERS-CoV and HCoV-OC43 infection. SARS-CoV-2, however, appears to be insensitive to p-eIF2α and, during infection, may even downregulate dephosphorylation to limit host translation. IMPORTANCE Lethal human betacoronaviruses have emerged three times in the last two decades, causing two epidemics and a pandemic. Here, we demonstrate differences in how these viruses interact with cellular translational control mechanisms. Utilizing inhibitory compounds and genetic ablation, we demonstrate that MERS-CoV and HCoV-OC43 benefit from keeping p-eIF2α levels low to maintain high rates of virus translation while SARS-CoV-2 tolerates high levels of p-eIF2α. We utilized a PP1:GADD34/CReP inhibitor, GADD34 KO cells, and CReP-targeting siRNA to investigate the therapeutic potential of these pathways. While ineffective for SARS-CoV-2, we found that HCoV-OC43 seems to primarily utilize CReP to limit p-eIF2a accumulation. This work highlights the need to consider differences amongst these viruses, which may inform the development of host-directed pan-coronavirus therapeutics.
Collapse
Affiliation(s)
- David M. Renner
- Departments of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA 19104-6076
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA 19104-6076
| | - Nicholas A. Parenti
- Departments of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA 19104-6076
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA 19104-6076
| | - Susan R. Weiss
- Departments of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA 19104-6076
- Penn Center for Research on Coronaviruses and Other Emerging Pathogens, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA 19104-6076
| |
Collapse
|
4
|
Hicks D, Giresh K, Wrischnik LA, Weiser DC. The PPP1R15 Family of eIF2-alpha Phosphatase Targeting Subunits (GADD34 and CReP). Int J Mol Sci 2023; 24:17321. [PMID: 38139150 PMCID: PMC10743859 DOI: 10.3390/ijms242417321] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The vertebrate PPP1R15 family consists of the proteins GADD34 (growth arrest and DNA damage-inducible protein 34, the product of the PPP1R15A gene) and CReP (constitutive repressor of eIF2α phosphorylation, the product of the PPP1R15B gene), both of which function as targeting/regulatory subunits for protein phosphatase 1 (PP1) by regulating subcellular localization, modulating substrate specificity and assembling complexes with target proteins. The primary cellular function of these proteins is to facilitate the dephosphorylation of eukaryotic initiation factor 2-alpha (eIF2α) by PP1 during cell stress. In this review, we will provide a comprehensive overview of the cellular function, biochemistry and pharmacology of GADD34 and CReP, starting with a brief introduction of eIF2α phosphorylation via the integrated protein response (ISR). We discuss the roles GADD34 and CReP play as feedback inhibitors of the unfolded protein response (UPR) and highlight the critical function they serve as inhibitors of the PERK-dependent branch, which is particularly important since it can mediate cell survival or cell death, depending on how long the stressful stimuli lasts, and GADD34 and CReP play key roles in fine-tuning this cellular decision. We briefly discuss the roles of GADD34 and CReP homologs in model systems and then focus on what we have learned about their function from knockout mice and human patients, followed by a brief review of several diseases in which GADD34 and CReP have been implicated, including cancer, diabetes and especially neurodegenerative disease. Because of the potential importance of GADD34 and CReP in aspects of human health and disease, we will discuss several pharmacological inhibitors of GADD34 and/or CReP that show promise as treatments and the controversies as to their mechanism of action. This review will finish with a discussion of the biochemical properties of GADD34 and CReP, their regulation and the additional interacting partners that may provide insight into the roles these proteins may play in other cellular pathways. We will conclude with a brief outline of critical areas for future study.
Collapse
Affiliation(s)
- Danielle Hicks
- Department of Science, Mathematics and Engineering, Modesto Junior College, Modesto, CA 95350, USA
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Krithika Giresh
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Lisa A. Wrischnik
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Douglas C. Weiser
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| |
Collapse
|
5
|
Wan L, Chen Z, Yang J, Wu G, Xu Y, Cui J, Zhao X. Identification of endoplasmic reticulum stress-related signature characterizes the tumor microenvironment and predicts prognosis in lung adenocarcinoma. Sci Rep 2023; 13:19462. [PMID: 37945620 PMCID: PMC10636162 DOI: 10.1038/s41598-023-45690-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Lung adenocarcinoma (LUAD) remains one of the most lethal malignancies worldwide, with a high mortality rate and unfavorable prognosis. Endoplasmic reticulum (ER) stress is a key regulator of tumour growth, metastasis, and the response to chemotherapy, targeted therapies and immune response. It acts via responding to misfolded proteins and triggering abnormal activation of ER stress sensors and downstream signalling pathways. Notably, the expression patterns of ER-stress-related-genes (ERSRGs) are indicative of survival outcomes, especially in the context of immune infiltration. Through consensus clustering of prognosis-associated ERSRGs, we delineated two distinct LUAD subtypes: Cluster 1 and Cluster 2. Comprehensive analyses revealed significant disparities between these subtypes in terms of prognosis, immune cell infiltration, and tumor progression. Leveraging the robustness of LASSO regression and Multivariate stepwise regression, we constructed and validated an ER Stress-associated risk signature for LUAD. This signature underwent assessments for its prognostic value, correlation with clinical attributes, and interaction within the tumour immune microenvironment. By integrating this signature with multivariate cox analysis of distinct pathological stages, we devised an enhanced nomogram, validated through various statistical metrics, with an area under the curve for overall survival at 1, 3, and 5 years post-diagnosis being 0.79, 0.80, and 0.81, respectively. In conclusion, our findings introduce a composite signature of 11 pivotal ERSRGs, holding promise as a potent prognostic tool for LUAD, and offering insights for immunotherapeutic and targeted intervention strategies.
Collapse
Affiliation(s)
- Li Wan
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, China
| | - Zhike Chen
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Gaotian Wu
- Soochow University Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, Suzhou, China
| | - Yao Xu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian Cui
- Department of Thoracic Surgery, Wuzhong District People's Hospital, Suzhou, China.
| | - Xueping Zhao
- School of Nursing, Medical College of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
6
|
Kumar A, Karuppagounder SS, Chen Y, Corona C, Kawaguchi R, Cheng Y, Balkaya M, Sagdullaev BT, Wen Z, Stuart C, Cho S, Ming GL, Tuvikene J, Timmusk T, Geschwind DH, Ratan RR. 2-Deoxyglucose drives plasticity via an adaptive ER stress-ATF4 pathway and elicits stroke recovery and Alzheimer's resilience. Neuron 2023; 111:2831-2846.e10. [PMID: 37453419 PMCID: PMC10528360 DOI: 10.1016/j.neuron.2023.06.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 05/10/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023]
Abstract
Intermittent fasting (IF) is a diet with salutary effects on cognitive aging, Alzheimer's disease (AD), and stroke. IF restricts a number of nutrient components, including glucose. 2-deoxyglucose (2-DG), a glucose analog, can be used to mimic glucose restriction. 2-DG induced transcription of the pro-plasticity factor, Bdnf, in the brain without ketosis. Accordingly, 2-DG enhanced memory in an AD model (5xFAD) and functional recovery in an ischemic stroke model. 2-DG increased Bdnf transcription via reduced N-linked glycosylation, consequent ER stress, and activity of ATF4 at an enhancer of the Bdnf gene, as well as other regulatory regions of plasticity/regeneration (e.g., Creb5, Cdc42bpa, Ppp3cc, and Atf3) genes. These findings demonstrate an unrecognized role for N-linked glycosylation as an adaptive sensor to reduced glucose availability. They further demonstrate that ER stress induced by 2-DG can, in the absence of ketosis, lead to the transcription of genes involved in plasticity and cognitive resilience as well as proteostasis.
Collapse
Affiliation(s)
- Amit Kumar
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Saravanan S Karuppagounder
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Yingxin Chen
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Carlo Corona
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Riki Kawaguchi
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yuyan Cheng
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mustafa Balkaya
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Botir T Sagdullaev
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA; Regeneron Pharmaceuticals, Tarrytown, New York, NY, USA
| | - Zhexing Wen
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Charles Stuart
- East Tennessee State University Quillen College of Medicine, Johnson City, TN, USA
| | - Sunghee Cho
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA
| | - Guo-Li Ming
- Department of Neuroscience, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jürgen Tuvikene
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Tõnis Timmusk
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Tallinn, Estonia
| | - Daniel H Geschwind
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Rajiv R Ratan
- Burke Neurological Institute and Brain and Mind Research Institute, Weill Cornell Medicine, 785 Mamaroneck Ave, White Plains, NY, USA.
| |
Collapse
|
7
|
Mingjie Y, Yair A, Tali G. The RIDD activity of C. elegans IRE1 modifies neuroendocrine signaling in anticipation of environment stress to ensure survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.10.552841. [PMID: 37609168 PMCID: PMC10441387 DOI: 10.1101/2023.08.10.552841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Xbp1 splicing and regulated IRE1-dependent RNA decay (RIDD) are two RNase activities of the ER stress sensor IRE1. While Xbp1 splicing has important roles in stress responses and animal physiology, the physiological role(s) of RIDD remain enigmatic. Genetic evidence in C. elegans connects XBP1-independent IRE1 activity to organismal stress adaptation, but whether this is via RIDD, and what are the targets is yet unknown. We show that cytosolic kinase/RNase domain of C. elegans IRE1 is indeed capable of RIDD in human cells, and that sensory neurons use RIDD to signal environmental stress, by degrading mRNA of TGFβ-like growth factor DAF-7. daf-7 was degraded in human cells by both human and worm IRE1 RNAse activity with same efficiency and specificity as Blos1, confirming daf-7 as RIDD substrate. Surprisingly, daf-7 degradation in vivo was triggered by concentrations of ER stressor tunicamycin too low for xbp-1 splicing. Decrease in DAF-7 normally signals food limitation and harsh environment, triggering adaptive changes to promote population survival. Because C. elegans is a bacteriovore, and tunicamycin, like other common ER stressors, is an antibiotic secreted by Streptomyces spp., we asked whether daf-7 degradation by RIDD could signal pending food deprivation. Indeed, pre-emptive tunicamycin exposure increased survival of C. elegans populations under food limiting/high temperature stress, and this protection was abrogated by overexpression of DAF-7. Thus, C. elegans uses stress-inducing metabolites in its environment as danger signals, and employs IRE1's RIDD activity to modulate the neuroendocrine signaling for survival of upcoming environmental challenge.
Collapse
Affiliation(s)
- Ying Mingjie
- Department of Biology, Drexel University, Philadelphia, PA
- Department of Pathology and Lab Medicine, The Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | - Argon Yair
- Department of Pathology and Lab Medicine, The Children's Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
8
|
Liu S, Gao Q, Li Y, Lun J, Yu M, Zhang H, Fang J. XBP1s acts as a transcription factor of IRE1α and promotes proliferation of colon cancer cells. Arch Biochem Biophys 2023; 737:109552. [PMID: 36828260 DOI: 10.1016/j.abb.2023.109552] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/14/2023] [Accepted: 02/21/2023] [Indexed: 02/24/2023]
Abstract
Upon ER stress, IRE1α is activated to splice XBP1 mRNA to generate XBP1s, a transcription factor that induces the expression of genes to cope with the stress. Expression of IRE1α is elevated in cancers and the IRE1α-XBP1s axis plays an important role in proliferation of cancer cells. However, the underlying mechanism is not well known. We found that ER stressors induced the expression of IRE1α, which was inhibited by depletion of XBP1s. XBP1s bound IRE1α promoter and initiated the transcription of IRE1α. These data indicate that XBP1s acts as a transcription factor of IRE1α. Overexpression of XBP1s increased the phosphorylation of JNK, a substrate of IRE1α kinase, which was inhibited by IRE1α kinase inhibitor Kira8. Overexpression of XBP1s also activated the regulated IRE1-dependent decay of mRNAs, which was suppressed by IRE1α RNase inhibitor STF083010. Moreover, we found that expression of XBP1s promoted proliferation of colon cancer cells, which was abrogated by Kira8 and STF083010. The results suggest that XBP1s functions to induce IRE1α expression and promote cancer cell proliferation. Our findings reveal a previously unknown mechanism of IRE1α expression by XBP1s and highlight the role of this regulation in proliferation of colon cancer cells, suggesting that IRE1α-targeting is a potential therapeutic strategy for colon cancer.
Collapse
Affiliation(s)
- Shuting Liu
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao 266061, China
| | - Qiang Gao
- Shanghai Institute of Nutrition and Health, CAS, Shanghai, 200031, China
| | - Yuyao Li
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao 266061, China
| | - Jie Lun
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao 266061, China
| | - Mengchao Yu
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao, 266071, China
| | - Hongwei Zhang
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250014, China.
| | - Jing Fang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, School of Basic Medicine, Qingdao University, Qingdao 266061, China.
| |
Collapse
|
9
|
Liu X, Taylor SA, Gromer KD, Zhang D, Hubchak SC, LeCuyer BE, Iwawaki T, Shi Z, Rockey DC, Green RM. Mechanisms of liver injury in high fat sugar diet fed mice that lack hepatocyte X-box binding protein 1. PLoS One 2022; 17:e0261789. [PMID: 35030194 PMCID: PMC8759640 DOI: 10.1371/journal.pone.0261789] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 12/09/2021] [Indexed: 11/26/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most common causes of liver diseases in the United States and can progress to cirrhosis, end-stage liver disease and need for liver transplantation. There are limited therapies for NAFLD, in part, due to incomplete understanding of the disease pathogenesis, which involves different cell populations in the liver. Endoplasmic reticulum stress and its adaptative unfolded protein response (UPR) signaling pathway have been implicated in the progression from simple hepatic steatosis to nonalcoholic steatohepatitis (NASH). We have previously shown that mice lacking the UPR protein X-box binding protein 1 (XBP1) in the liver demonstrated enhanced liver injury and fibrosis in a high fat sugar (HFS) dietary model of NAFLD. In this study, to better understand the role of liver XBP1 in the pathobiology of NAFLD, we fed hepatocyte XBP1 deficient mice a HFS diet or chow and investigated UPR and other cell signaling pathways in hepatocytes, hepatic stellate cells and immune cells. We demonstrate that loss of XBP1 in hepatocytes increased inflammatory pathway expression and altered expression of the UPR signaling in hepatocytes and was associated with enhanced hepatic stellate cell activation after HFS feeding. We believe that a better understanding of liver cell-specific signaling in the pathogenesis of NASH may allow us to identify new therapeutic targets.
Collapse
Affiliation(s)
- Xiaoying Liu
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Sarah A. Taylor
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
| | - Kyle D. Gromer
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, United States of America
| | - Danny Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Susan C. Hubchak
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Brian E. LeCuyer
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, Japan
| | - Zengdun Shi
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Don C. Rockey
- Digestive Disease Research Center, Medical University of South Carolina, Charleston, South Carolina, United States of America
| | - Richard M. Green
- Division of Gastroenterology and Hepatology, Department of Medicine, Northwestern University, Chicago, Illinois, United States of America
| |
Collapse
|
10
|
Ricci D, Gidalevitz T, Argon Y. The special unfolded protein response in plasma cells. Immunol Rev 2021; 303:35-51. [PMID: 34368957 DOI: 10.1111/imr.13012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 06/08/2021] [Indexed: 12/11/2022]
Abstract
The high rate of antibody production places considerable metabolic and folding stress on plasma cells (PC). Not surprisingly, they rely on the unfolded protein response (UPR), a universal signaling, and transcriptional network that monitors the health of the secretory pathway and mounts cellular responses to stress. Typically, the UPR utilizes three distinct stress sensors in the ER membrane, each regulating a subset of targets to re-establish homeostasis. PC use a specialized UPR scheme-they preemptively trigger the UPR via developmental signals and suppress two of the sensors, PERK and ATF6, relying on IRE1 alone. The specialized PC UPR program is tuned to the specific needs at every stage of development-from early biogenesis of secretory apparatus, to massive immunoglobulin expression later. Furthermore, the UPR in PC integrates with other pathways essential in a highly secretory cell-mTOR pathway that ensures efficient synthesis, autophagosomes that recycle components of the synthetic machinery, and apoptotic signaling that controls cell fate in the face of excessive folding stress. This specialized PC program is not shared with other secretory cells, for reasons yet to be defined. In this review, we give a perspective into how and why PC need such a unique UPR program.
Collapse
Affiliation(s)
- Daniela Ricci
- Department of Pathology and Lab Medicine, The Childrens' Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| | - Tali Gidalevitz
- Department of Biology, Drexel University, Philadelphia, PA, USA
| | - Yair Argon
- Department of Pathology and Lab Medicine, The Childrens' Hospital of Philadelphia and the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Park SM, Kang TI, So JS. Roles of XBP1s in Transcriptional Regulation of Target Genes. Biomedicines 2021; 9:biomedicines9070791. [PMID: 34356855 PMCID: PMC8301375 DOI: 10.3390/biomedicines9070791] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 12/17/2022] Open
Abstract
The spliced form of X-box binding protein 1 (XBP1s) is an active transcription factor that plays a vital role in the unfolded protein response (UPR). Under endoplasmic reticulum (ER) stress, unspliced Xbp1 mRNA is cleaved by the activated stress sensor IRE1α and converted to the mature form encoding spliced XBP1 (XBP1s). Translated XBP1s migrates to the nucleus and regulates the transcriptional programs of UPR target genes encoding ER molecular chaperones, folding enzymes, and ER-associated protein degradation (ERAD) components to decrease ER stress. Moreover, studies have shown that XBP1s regulates the transcription of diverse genes that are involved in lipid and glucose metabolism and immune responses. Therefore, XBP1s has been considered an important therapeutic target in studying various diseases, including cancer, diabetes, and autoimmune and inflammatory diseases. XBP1s is involved in several unique mechanisms to regulate the transcription of different target genes by interacting with other proteins to modulate their activity. Although recent studies discovered numerous target genes of XBP1s via genome-wide analyses, how XBP1s regulates their transcription remains unclear. This review discusses the roles of XBP1s in target genes transcriptional regulation. More in-depth knowledge of XBP1s target genes and transcriptional regulatory mechanisms in the future will help develop new therapeutic targets for each disease.
Collapse
|
12
|
Duwaerts CC, Siao K, Soon RK, Her C, Iwawaki T, Kohno K, Mattis AN, Maher JJ. Hepatocyte-specific deletion of XBP1 sensitizes mice to liver injury through hyperactivation of IRE1α. Cell Death Differ 2020; 28:1455-1465. [PMID: 33219328 DOI: 10.1038/s41418-020-00671-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 10/27/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
X-box binding protein-1 (XBP1) is a transcription factor that plays a central role in controlling cellular responses to endoplasmic reticulum (ER) stress. Under stress conditions, the transcriptionally active form of XBP1 is generated via splicing of Xbp1 mRNA by the ER-resident protein inositol-requiring enzyme-1 (IRE1α). Genetic deletion of XBP1 has multiple consequences: some resulting from the loss of the transcription factor per se, and others related to compensatory activation of IRE1α. The objective of the current study was to investigate the effects of XBP1 deletion in adult mouse liver and determine to what extent they are direct or indirect. XBP1 was deleted from hepatocytes in adult Xbp1fl/fl mice using AAV8-Transthyretin-Cre (Xbp1Δhep). Xbp1Δhep mice exhibited no liver disease at baseline, but developed acute biochemical and histologic liver injury in response to a dietary challenge with fructose for 4 weeks. Fructose-mediated liver injury in Xbp1Δhep mice coincided with heightened IRE1α activity, as demonstrated by Xbp1 mRNA splicing, JNK activation, and regulated IRE1α-dependent RNA decay (RIDD). Activation of eIF2α was also evident, with associated up-regulation of the pro-apoptotic molecules CHOP, BIM, and PUMA. To determine whether the adverse consequences of liver-specific XBP1 deletion were due to XBP1 loss or heightened IRE1α activity, we repeated a fructose challenge in mice with liver-specific deletion of both XBP1 and IRE1α (Xbp1Δhep;Ire1aΔhep). Xbp1Δhep;Ire1aΔhep mice were protected from fructose-mediated liver injury and failed to exhibit any of the signs of ER stress seen in mice lacking XBP1 alone. The protective effect of IRE1α deletion persisted even with long-term exposure to fructose. Xbp1Δhep mice developed liver fibrosis at 16 weeks, but Xbp1Δhep;Ire1aΔhep mice did not. Overall, the results indicate that the deleterious effects of hepatocyte-specific XBP1 deletion are due primarily to hyperactivation of IRE1α. They support further exploration of IRE1α as a contributor to acute and chronic liver diseases.
Collapse
Affiliation(s)
- Caroline C Duwaerts
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.,The Liver Center, University of California San Francisco, San Francisco, CA, USA
| | - Kevin Siao
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.,The Liver Center, University of California San Francisco, San Francisco, CA, USA
| | - Russell K Soon
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.,The Liver Center, University of California San Francisco, San Francisco, CA, USA.,BioMarin Pharmaceutical Inc. 105 Digital Drive, Novato, CA, 94949, USA
| | - Chris Her
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA.,The Liver Center, University of California San Francisco, San Francisco, CA, USA
| | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa, 920-0293, Japan
| | - Kenji Kohno
- Institute for Research Initiatives, Nara Institute of Science and Technology, Ikoma, Nara, 630-0192, Japan
| | - Aras N Mattis
- The Liver Center, University of California San Francisco, San Francisco, CA, USA.,Department of Pathology, University of California San Francisco, San Francisco, CA, USA
| | - Jacquelyn J Maher
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA. .,The Liver Center, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
13
|
Riaz TA, Junjappa RP, Handigund M, Ferdous J, Kim HR, Chae HJ. Role of Endoplasmic Reticulum Stress Sensor IRE1α in Cellular Physiology, Calcium, ROS Signaling, and Metaflammation. Cells 2020; 9:E1160. [PMID: 32397116 PMCID: PMC7290600 DOI: 10.3390/cells9051160] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/27/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Inositol-requiring transmembrane kinase endoribonuclease-1α (IRE1α) is the most prominent and evolutionarily conserved unfolded protein response (UPR) signal transducer during endoplasmic reticulum functional upset (ER stress). A IRE1α signal pathway arbitrates yin and yang of cellular fate in objectionable conditions. It plays several roles in fundamental cellular physiology as well as in several pathological conditions such as diabetes, obesity, inflammation, cancer, neurodegeneration, and in many other diseases. Thus, further understanding of its molecular structure and mechanism of action during different cell insults helps in designing and developing better therapeutic strategies for the above-mentioned chronic diseases. In this review, recent insights into structure and mechanism of activation of IRE1α along with its complex regulating network were discussed in relation to their basic cellular physiological function. Addressing different binding partners that can modulate IRE1α function, UPRosome triggers different downstream pathways depending on the cellular backdrop. Furthermore, IRE1α are in normal cell activities outside the dominion of ER stress and activities under the weather of inflammation, diabetes, and obesity-related metaflammation. Thus, IRE1 as an ER stress sensor needs to be understood from a wider perspective for comprehensive functional meaning, which facilitates us with assembling future needs and therapeutic benefits.
Collapse
Affiliation(s)
- Thoufiqul Alam Riaz
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Jeonbuk National University, Jeonju 54907, Korea; (T.A.R.); (R.P.J.)
| | - Raghu Patil Junjappa
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Jeonbuk National University, Jeonju 54907, Korea; (T.A.R.); (R.P.J.)
| | - Mallikarjun Handigund
- Department of Laboratory Medicine, Jeonbuk National University, Medical School, Jeonju 54907, Korea;
| | - Jannatul Ferdous
- Department of Radiology and Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Korea;
| | - Hyung-Ryong Kim
- College of Dentistry, Dankook University, Cheonan 31116, Korea
| | - Han-Jung Chae
- Department of Pharmacology, School of Medicine, Institute of New Drug Development, Jeonbuk National University, Jeonju 54907, Korea; (T.A.R.); (R.P.J.)
| |
Collapse
|
14
|
Sprooten J, Garg AD. Type I interferons and endoplasmic reticulum stress in health and disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 350:63-118. [PMID: 32138904 PMCID: PMC7104985 DOI: 10.1016/bs.ircmb.2019.10.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Type I interferons (IFNs) comprise of pro-inflammatory cytokines created, as well as sensed, by all nucleated cells with the main objective of blocking pathogens-driven infections. Owing to this broad range of influence, type I IFNs also exhibit critical functions in many sterile inflammatory diseases and immunopathologies, especially those associated with endoplasmic reticulum (ER) stress-driven signaling pathways. Indeed, over the years accumulating evidence has indicated that the presence of ER stress can influence the production, or sensing of, type I IFNs induced by perturbations like pattern recognition receptor (PRR) agonists, infections (bacterial, viral or parasitic) or autoimmunity. In this article we discuss the link between type I IFNs and ER stress in various diseased contexts. We describe how ER stress regulates type I IFNs production or sensing, or how type I IFNs may induce ER stress, in various circumstances like microbial infections, autoimmunity, diabetes, cancer and other ER stress-related contexts.
Collapse
Affiliation(s)
- Jenny Sprooten
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Department for Cellular and Molecular Medicine, Cell Death Research & Therapy (CDRT) Unit, KU Leuven, Leuven, Belgium.
| |
Collapse
|
15
|
Carew NT, Nelson AM, Liang Z, Smith SM, Milcarek C. Linking Endoplasmic Reticular Stress and Alternative Splicing. Int J Mol Sci 2018; 19:ijms19123919. [PMID: 30544499 PMCID: PMC6321306 DOI: 10.3390/ijms19123919] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/04/2018] [Accepted: 12/05/2018] [Indexed: 12/16/2022] Open
Abstract
RNA splicing patterns in antibody-secreting cells are shaped by endoplasmic reticulum stress, ELL2 (eleven-nineteen lysine-rich leukemia gene 2) induction, and changes in the levels of snRNAs. Endoplasmic reticulum stress induces the unfolded protein response comprising a highly conserved set of genes crucial for cell survival; among these is Ire1, whose auto-phosphorylation drives it to acquire a regulated mRNA decay activity. The mRNA-modifying function of phosphorylated Ire1 non-canonically splices Xbp1 mRNA and yet degrades other cellular mRNAs with related motifs. Naïve splenic B cells will activate Ire1 phosphorylation early on after lipopolysaccharide (LPS) stimulation, within 18 h; large-scale changes in mRNA content and splicing patterns result. Inhibition of the mRNA-degradation function of Ire1 is correlated with further differences in the splicing patterns and a reduction in the mRNA factors for snRNA transcription. Some of the >4000 splicing changes seen at 18 h after LPS stimulation persist into the late stages of antibody secretion, up to 72 h. Meanwhile some early splicing changes are supplanted by new splicing changes introduced by the up-regulation of ELL2, a transcription elongation factor. ELL2 is necessary for immunoglobulin secretion and does this by changing mRNA processing patterns of immunoglobulin heavy chain and >5000 other genes.
Collapse
Affiliation(s)
- Nolan T Carew
- School of Medicine, Department of Immunology, University of Pittsburgh, E1059 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| | - Ashley M Nelson
- School of Medicine, Department of Immunology, University of Pittsburgh, E1059 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| | - Zhitao Liang
- School of Medicine, Department of Immunology, University of Pittsburgh, E1059 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| | - Sage M Smith
- School of Medicine, Department of Immunology, University of Pittsburgh, E1059 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| | - Christine Milcarek
- School of Medicine, Department of Immunology, University of Pittsburgh, E1059 Biomedical Science Tower, Pittsburgh, PA 15261, USA.
| |
Collapse
|
16
|
Xu X, Krumm C, So JS, Bare CJ, Holman C, Gromada J, Cohen DE, Lee AH. Preemptive Activation of the Integrated Stress Response Protects Mice From Diet-Induced Obesity and Insulin Resistance by Fibroblast Growth Factor 21 Induction. Hepatology 2018; 68:2167-2181. [PMID: 29698569 PMCID: PMC6203669 DOI: 10.1002/hep.30060] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 04/23/2018] [Indexed: 12/11/2022]
Abstract
Integrated stress response (ISR) is a signaling system in which phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) by stress-specific kinases and subsequent activation of activation transcription factor (ATF) 4 help restore cellular homeostasis following exposure to environmental stresses. ISR activation has been observed in metabolic diseases, including hepatic steatosis (HS), steatohepatitis (SH), and insulin resistance (IR), but it remains unclear whether ISR contributes to disease pathogenesis or represents an innate defense mechanism against metabolic stresses. Constitutive repressor of eIF2α phosphorylation (CReP) is a critical regulatory subunit of the eIF2α phosphatase complex. Here, we show that CReP ablation causes constitutive eIF2α phosphorylation in the liver, which leads to activation of the ATF4 transcriptional program including increased fibroblast growth factor 21 (FGF21) production. Liver-specific CReP knockout (CRePLKO ) mice exhibited marked browning of white adipose tissue (WAT) and increased energy expenditure and insulin sensitivity in an FGF21-dependent manner. Furthermore, CRePLKO mice were protected from high-fat diet (HFD)-induced obesity, HS, and IR. Acute CReP ablation in liver of HFD-induced obese mice also reduced adiposity and improved glucose homeostasis. Conclusion: These data suggest that CReP abundance is a critical determinant for eIF2α phosphorylation and ensuing ISR activation in the liver. Constitutive ISR activation in the liver induces FGF21 and confers protection from HFD-induced adiposity, IR, and HS in mice. Augmenting hepatic ISR may represent a therapeutic approach to treat metabolic disorders.
Collapse
Affiliation(s)
- Xu Xu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Christopher Krumm
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jae-Seon So
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065
- Department of Medical Biotechnology, Dongguk University Gyeongju, Gyeongju-si, Korea
| | - Curtis J. Bare
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Corey Holman
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Jesper Gromada
- Regeneron Pharmaceuticals, Tarrytown, New York 10591, USA
| | - David E. Cohen
- Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Ann-Hwee Lee
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065
- Regeneron Pharmaceuticals, Tarrytown, New York 10591, USA
| |
Collapse
|
17
|
So JS. Roles of Endoplasmic Reticulum Stress in Immune Responses. Mol Cells 2018; 41:705-716. [PMID: 30078231 PMCID: PMC6125421 DOI: 10.14348/molcells.2018.0241] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/03/2018] [Accepted: 07/19/2018] [Indexed: 02/06/2023] Open
Abstract
The endoplasmic reticulum (ER) is a critical organelle for protein synthesis, folding and modification, and lipid synthesis and calcium storage. Dysregulation of ER functions leads to the accumulation of misfolded- or unfolded-protein in the ER lumen, and this triggers the unfolded protein response (UPR), which restores ER homeostasis. The UPR is characterized by three distinct downstream signaling pathways that promote cell survival or apoptosis depending on the stressor, the intensity and duration of ER stress, and the cell type. Mammalian cells express the UPR transducers IRE1, PERK, and ATF6, which control transcriptional and translational responses to ER stress. Direct links between ER stress and immune responses are also evident, but the mechanisms by which UPR signaling cascades are coordinated with immunity remain unclear. This review discusses recent investigations of the roles of ER stress in immune responses that lead to differentiation, maturation, and cytokine expression in immune cells. Further understanding of how ER stress contributes to the pathogenesis of immune disorders will facilitate the development of novel therapies that target UPR pathways.
Collapse
Affiliation(s)
- Jae-Seon So
- Department of Medical Biotechnology, Dongguk University Gyeongju, Gyeongju 38066,
Korea
| |
Collapse
|
18
|
Abdullah A, Ravanan P. The unknown face of IRE1α - Beyond ER stress. Eur J Cell Biol 2018; 97:359-368. [PMID: 29747876 DOI: 10.1016/j.ejcb.2018.05.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/03/2018] [Accepted: 05/03/2018] [Indexed: 12/16/2022] Open
Abstract
IRE1α (Inositol Requiring kinase Enzyme 1 alpha), a transmembrane protein localized to the endoplasmic reticulum (ER) is a master regulator of the unfolded protein response (UPR) pathway. The fate determining steps during ER stress-induced apoptosis are greatly attributed to IRE1α's endoribonuclease and kinase activities. Apart from its role as a chief executioner in ER stress, recent studies have shown that upon activation in the presence or absence of ER stress, IRE1α executes multiple cellular processes such as differentiation, immune response, progression and repression of the cell cycle. Besides its crucial role in protein misfolding, the versatile contributions of IRE1α in other cellular functions are greatly unknown. In this review, we have discussed the structural conservation of IRE1 among eukaryotes, the mechanisms underlying its activation and the recent understandings of the non-apoptotic functions of IRE1 other than ER stress-induced cell death.
Collapse
Affiliation(s)
- Ahmad Abdullah
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India
| | - Palaniyandi Ravanan
- Apoptosis and Cell Survival Research Lab, Department of Biosciences, School of Biosciences and Technology, VIT University, Vellore, Tamil Nadu, India.
| |
Collapse
|
19
|
Wu L, Liu H, Li L, Xu D, Gao Y, Guan Y, Chen Q. 5,7,3',4'-Tetramethoxyflavone protects chondrocytes from ER stress-induced apoptosis through regulation of the IRE1α pathway. Connect Tissue Res 2018; 59:157-166. [PMID: 28436754 PMCID: PMC6104397 DOI: 10.1080/03008207.2017.1321639] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
AIM OF THE STUDY To investigate the roles of endoplasmic reticulum (ER) transmembrane sensor inositol-requiring enzyme-1 (IRE1)α signaling in ER stress-induced chondrocyte apoptosis, and to determine the molecular mechanisms underlying chondroprotective activity of 5,7,3',4'-tetramethoxyflavone (TMF) from Murraya exotica. MATERIALS AND METHODS IRE1α was knocked down by siRNA transfection in chondrocytes, which were harvested from rats' knee cartilages. Chondrocytes with IRE1α deficiency were administrated with tunicamycin (TM) and TMF. Chondrocyte apoptosis was quantified by flow cytometry and DAPI/TUNEL staining. Expression of mRNA and proteins was quantified by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western-blot, respectively. RESULTS IRE1α deficiency significantly increased the rate of TM-induced chondrocyte apoptosis, down-regulated the expression of pro-survival factors XBP1S and Bcl-2, and up-regulated pro-apoptotic factors CHOP, p-JNK, and caspase-3. TMF suppressed TM-induced chondrocyte apoptosis by activating the expression of IRE1α, which reversed the expression patterns of downstream pro-survival and pro-apoptotic factors due to IRE1α deficiency. CONCLUSION The mechanism of TMF in protecting chondrocytes against ER stress-induced apoptosis might be associated with regulating the activity of ER sensor IRE1α and its downstream pathway.
Collapse
Affiliation(s)
- Longhuo Wu
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA;,College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Haiqing Liu
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA;,College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Linfu Li
- College of Pharmacy, Gannan Medical University, Ganzhou, China
| | - Daohua Xu
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA;,Department of Pharmacology, Guangdong Medical University, Dongguan, China
| | - Yun Gao
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA
| | - Yingjie Guan
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA
| | - Qian Chen
- Department of Orthopaedics, Warren Alpert Medical School of Brown University/Rhode Island Hospital, Providence, RI, USA
| |
Collapse
|
20
|
Regulated IRE1-dependent mRNA decay sets the threshold for dendritic cell survival. Nat Cell Biol 2017; 19:698-710. [PMID: 28459443 DOI: 10.1038/ncb3518] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 03/21/2017] [Indexed: 12/23/2022]
Abstract
The IRE1-XBP1 signalling pathway is part of a cellular programme that protects against endoplasmic reticulum (ER) stress, but also controls development and survival of immune cells. Loss of XBP1 in splenic type 1 conventional dendritic cells (cDC1s) results in functional alterations without affecting cell survival. However, in mucosal cDC1s, loss of XBP1 impaired survival in a tissue-specific manner-while lung cDC1s die, intestinal cDC1s survive. This was not caused by differential activation of ER stress cell-death regulators CHOP or JNK. Rather, survival of intestinal cDC1s was associated with their ability to shut down protein synthesis through a protective integrated stress response and their marked increase in regulated IRE1-dependent messenger RNA decay. Furthermore, loss of IRE1 endonuclease on top of XBP1 led to cDC1 loss in the intestine. Thus, mucosal DCs differentially mount ATF4- and IRE1-dependent adaptive mechanisms to survive in the face of ER stress.
Collapse
|
21
|
Abstract
Hepatic steatosis, the first step in the progression of nonalcoholic fatty liver disease, is characterized by triglyceride accumulation in hepatocytes and is highly prevalent in people with obesity. Although initially asymptomatic, hepatic steatosis is an important risk factor for the development of hepatic insulin resistance and type 2 diabetes mellitus and can also progress to more severe pathologies such as nonalcoholic steatohepatitis, liver fibrosis and cirrhosis; hepatic steatosis has, therefore, received considerable research interest in the past 20 years. The lipid accumulation that defines hepatic steatosis disturbs the function of the endoplasmic reticulum (ER) in hepatocytes, thereby generating chronic ER stress that interferes with normal cellular function. Although ubiquitous stress response mechanisms (namely, ER-associated degradation, unfolded protein response and autophagy) are the main processes for restoring cellular proteostasis, these mechanisms are unable to alleviate ER stress in the context of the fatty liver. Furthermore, ER stress and ER stress responses can promote lipid accumulation in hepatocytes in a counter-productive manner and could, therefore, be the origin of a vicious pathological cycle.
Collapse
Affiliation(s)
- Andrei Baiceanu
- Institut National de la Santé et de la Recherche Médicale, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Centre de Recherche des Cordeliers, 15 rue de l'école de médecine, F-75006, Paris, France
- University of Medicine and Pharmacy Iuliu Hat¸ieganu, Faculty of Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania
| | - Pierre Mesdom
- Institut National de la Santé et de la Recherche Médicale, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Centre de Recherche des Cordeliers, 15 rue de l'école de médecine, F-75006, Paris, France
| | - Marie Lagouge
- Institut National de la Santé et de la Recherche Médicale, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Centre de Recherche des Cordeliers, 15 rue de l'école de médecine, F-75006, Paris, France
| | - Fabienne Foufelle
- Institut National de la Santé et de la Recherche Médicale, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMRS 1138, Centre de Recherche des Cordeliers, F-75006, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, UMRS 1138, Centre de Recherche des Cordeliers, 15 rue de l'école de médecine, F-75006, Paris, France
| |
Collapse
|
22
|
Nissinen TA, Degerman J, Räsänen M, Poikonen AR, Koskinen S, Mervaala E, Pasternack A, Ritvos O, Kivelä R, Hulmi JJ. Systemic blockade of ACVR2B ligands prevents chemotherapy-induced muscle wasting by restoring muscle protein synthesis without affecting oxidative capacity or atrogenes. Sci Rep 2016; 6:32695. [PMID: 27666826 PMCID: PMC5036092 DOI: 10.1038/srep32695] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/12/2016] [Indexed: 12/26/2022] Open
Abstract
Doxorubicin is a widely used and effective chemotherapy drug. However, cardiac and skeletal muscle toxicity of doxorubicin limits its use. Inhibiting myostatin/activin signalling can prevent muscle atrophy, but its effects in chemotherapy-induced muscle wasting are unknown. In the present study we investigated the effects of doxorubicin administration alone or combined with activin receptor ligand pathway blockade by soluble activin receptor IIB (sACVR2B-Fc). Doxorubicin administration decreased body mass, muscle size and bone mineral density/content in mice. However, these effects were prevented by sACVR2B-Fc administration. Unlike in many other wasting situations, doxorubicin induced muscle atrophy without markedly increasing typical atrogenes or protein degradation pathways. Instead, doxorubicin decreased muscle protein synthesis which was completely restored by sACVR2B-Fc. Doxorubicin administration also resulted in impaired running performance without effects on skeletal muscle mitochondrial capacity/function or capillary density. Running performance and mitochondrial function were unaltered by sACVR2B-Fc administration. Tumour experiment using Lewis lung carcinoma cells demonstrated that sACVR2B-Fc decreased the cachectic effects of chemotherapy without affecting tumour growth. These results demonstrate that blocking ACVR2B signalling may be a promising strategy to counteract chemotherapy-induced muscle wasting without damage to skeletal muscle oxidative capacity or cancer treatment.
Collapse
Affiliation(s)
- T A Nissinen
- Department of Biology of Physical Activity, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - J Degerman
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - M Räsänen
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - A R Poikonen
- Department of Biology of Physical Activity, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - S Koskinen
- LIKES Research Center for Sport and Health Sciences, Jyväskylä, Finland
| | - E Mervaala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - A Pasternack
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - O Ritvos
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - R Kivelä
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - J J Hulmi
- Department of Biology of Physical Activity, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
23
|
Complementary Roles of GADD34- and CReP-Containing Eukaryotic Initiation Factor 2α Phosphatases during the Unfolded Protein Response. Mol Cell Biol 2016; 36:1868-80. [PMID: 27161320 DOI: 10.1128/mcb.00190-16] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 04/27/2016] [Indexed: 02/02/2023] Open
Abstract
Phosphorylation of eukaryotic initiation factor 2α (eIF2α) controls transcriptome-wide changes in mRNA translation in stressed cells. While phosphorylated eIF2α (P-eIF2α) attenuates global protein synthesis, mRNAs encoding stress proteins are more efficiently translated. Two eIF2α phosphatases, containing GADD34 and CReP, catalyze P-eIF2α dephosphorylation. The current view of GADD34, whose transcription is stress induced, is that it functions in a feedback loop to resolve cell stress. In contrast, CReP, which is constitutively expressed, controls basal P-eIF2α levels in unstressed cells. Our studies show that GADD34 drives substantial changes in mRNA translation in unstressed cells, particularly targeting the secretome. Following activation of the unfolded protein response (UPR), rapid translation of GADD34 mRNA occurs and GADD34 is essential for UPR progression. In the absence of GADD34, eIF2α phosphorylation is persistently enhanced and the UPR translational program is significantly attenuated. This "stalled" UPR is relieved by the subsequent activation of compensatory mechanisms that include AKT-mediated suppression of PKR-like kinase (PERK) and increased expression of CReP mRNA, partially restoring protein synthesis. Our studies highlight the coordinate regulation of UPR by the GADD34- and CReP-containing eIF2α phosphatases to control cell viability.
Collapse
|
24
|
An inhibitor of HIV-1 protease modulates constitutive eIF2α dephosphorylation to trigger a specific integrated stress response. Proc Natl Acad Sci U S A 2015; 113:E117-26. [PMID: 26715744 DOI: 10.1073/pnas.1514076113] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Inhibitors of the HIV aspartyl protease [HIV protease inhibitors (HIV-PIs)] are the cornerstone of treatment for HIV. Beyond their well-defined antiretroviral activity, these drugs have additional effects that modulate cell viability and homeostasis. However, little is known about the virus-independent pathways engaged by these molecules. Here we show that the HIV-PI Nelfinavir decreases translation rates and promotes a transcriptional program characteristic of the integrated stress response (ISR). Mice treated with Nelfinavir display hallmarks of this stress response in the liver, including α subunit of translation initiation factor 2 (eIF2α) phosphorylation, activating transcription factor-4 (ATF4) induction, and increased expression of known downstream targets. Mechanistically, Nelfinavir-mediated ISR bypassed direct activation of the eIF2α stress kinases and instead relied on the inhibition of the constitutive eIF2α dephosphorylation and down-regulation of the phophatase cofactor CReP (Constitutive Repressor of eIF2α Phosphorylation; also known as PPP1R15B). These findings demonstrate that the modulation of eIF2α-specific phosphatase cofactor activity can be a rheostat of cellular homeostasis that initiates a functional ISR and suggest that the HIV-PIs could be repositioned as therapeutics in human diseases to modulate translation rates and stress responses.
Collapse
|
25
|
Vergara M, Berrios J, Martínez I, Díaz-Barrera A, Acevedo C, Reyes JG, Gonzalez R, Altamirano C. Endoplasmic Reticulum-Associated rht-PA Processing in CHO Cells: Influence of Mild Hypothermia and Specific Growth Rates in Batch and Chemostat Cultures. PLoS One 2015; 10:e0144224. [PMID: 26659083 PMCID: PMC4676689 DOI: 10.1371/journal.pone.0144224] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 11/16/2015] [Indexed: 12/11/2022] Open
Abstract
Background Chinese hamster ovary (CHO) cells are the main host for producing recombinant proteins with human therapeutic applications mainly because of their capability to perform proper folding and glycosylation processes. In addition, mild hypothermia is one of the main strategies for maximising the productivity of these systems. However, little information is available on the effect of culture temperature on the folding and degradation processes of recombinant proteins that takes place in the endoplasmic reticulum. Methods In order to evaluate the effect of the mild hypothermia on processing/endoplasmatic reticulum-associated degradation (ERAD) processes, batch cultures of CHO cells producing recombinant human tissue plasminogen activator (rht-PA) were carried out at two temperatures (37°C and 33°C) and treated with specific inhibitors of glycosylation and ERAD I (Ubiquitin/Proteasome system) or ERAD II (Autophagosoma/Lisosomal system) pathways. The effect of mild hypothermia was analysed separately from its indirect effect on specific cell growth rate. To do this, chemostat cultures were carried out at the same incubation conditions as the batch cultures, controlling cell growth at high (0.017 h-1) and low (0.012 h-1) dilution rates. For a better understanding of the investigated phenomenon, cell behaviour was also analysed using principal component analysis (PCA). Results and Conclusion Results suggest that rht-PA is susceptible to degradation by both ERAD pathways studied, revealing that processing and/or ERAD processes are sensitive to temperature cultivation in batch culture. Moreover, by isolating the effect of culture temperature from the effect of cell growth rate verifyed by using chemostat cultures, we have found that processing and/or ERAD processes are more sensitive to reduction in specific growth rate than low temperature, and that temperature reduction may have a positive effect on protein processing. Interestingly, PCA indicated that the integrated performance displayed by CHO cells is modulated predominantly by specific growth rate, indicating that the culture temperature has a lower weighted effect within the range of conditions evaluated in this work.
Collapse
Affiliation(s)
- Mauricio Vergara
- Institute of Chemistry, Pontificia Universidad Católica de Valparaíso, Av. Universidad 330, Curauma, Chile
- School of Biochemical Engineering, Pontificia Universidad Católica de Valparaíso, Av. Brasil 2085, Valparaíso, 4059, Chile
| | - Julio Berrios
- School of Biochemical Engineering, Pontificia Universidad Católica de Valparaíso, Av. Brasil 2085, Valparaíso, 4059, Chile
| | - Irene Martínez
- School of Biochemical Engineering, Pontificia Universidad Católica de Valparaíso, Av. Brasil 2085, Valparaíso, 4059, Chile
| | - Alvaro Díaz-Barrera
- School of Biochemical Engineering, Pontificia Universidad Católica de Valparaíso, Av. Brasil 2085, Valparaíso, 4059, Chile
| | - Cristian Acevedo
- Biotechnology Center “Dr. Daniel Alkalay Lowitt”, Universidad Técnica Federico Santa María, Av. España 1680, Valparaíso, Chile
| | - Juan G. Reyes
- Institute of Chemistry, Pontificia Universidad Católica de Valparaíso, Av. Universidad 330, Curauma, Chile
| | - Ramon Gonzalez
- Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas, United States of America
- Department of Bioengineering, Rice University, Houston, Texas, United States of America
| | - Claudia Altamirano
- School of Biochemical Engineering, Pontificia Universidad Católica de Valparaíso, Av. Brasil 2085, Valparaíso, 4059, Chile
- CREAS CONICYT Regional GORE Valparaíso R0GI1004. Av. Universidad, Curauma, Chile
- * E-mail:
| |
Collapse
|
26
|
Friend K, Brooks HA, Propson NE, Thomson JA, Kimble J. Embryonic Stem Cell Growth Factors Regulate eIF2α Phosphorylation. PLoS One 2015; 10:e0139076. [PMID: 26406898 PMCID: PMC4583406 DOI: 10.1371/journal.pone.0139076] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 09/09/2015] [Indexed: 12/18/2022] Open
Abstract
Growth factors and transcription factors are well known to regulate pluripotent stem cells, but less is known about translational control in stem cells. Here, we use embryonic stem cells (ESCs) to investigate a connection between ESC growth factors and eIF2α-mediated translational control (eIF2α phosphorylation promotes protein expression from mRNAs with upstream open-reading frames, or uORFs). We find abundant phosphorylated P-eIF2α (P-eIF2α) in both pluripotent mouse and human ESCs, but little P-eIF2α in ESCs triggered to differentiate. We show that the growth factors LIF (leukemia inhibitory factor) and BMP4 (bone morphogenic protein 4) both maintain P-eIF2α in mESCs, but use distinct mechanisms: LIF inhibits an eIF2α phosphatase whereas BMP4 activates an eIF2α kinase. The mRNAs encoding the pluripotency factors Nanog and c-Myc possess uORFs while Oct4 mRNA does not. We find that salubrinal, a chemical that increases eIF2α phosphorylation, promotes Nanog and c-Myc expression, but not Oct4 expression. These experiments connect ESC growth factors to eIF2α phosphorylation and suggest a chemical substitute for LIF to enhance Nanog and c-Myc expression.
Collapse
Affiliation(s)
- Kyle Friend
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
- Department of Chemistry and Biochemistry, Washington and Lee University, Lexington, Virginia, 24450, United States of America
- * E-mail: (KF); (JK)
| | - Hunter A. Brooks
- Department of Chemistry and Biochemistry, Washington and Lee University, Lexington, Virginia, 24450, United States of America
| | - Nicholas E. Propson
- The Morgridge Institute for Research, 309 North Orchard Street, Madison, Wisconsin, 53715, United States of America
| | - James A. Thomson
- The Morgridge Institute for Research, 309 North Orchard Street, Madison, Wisconsin, 53715, United States of America
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, 53706, United States of America
| | - Judith Kimble
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
- Howard Hughes Medical Institute, University of Wisconsin-Madison, Madison, Wisconsin, 53706, United States of America
- * E-mail: (KF); (JK)
| |
Collapse
|