1
|
Dong Y, Liu G, Situ X, Xia L, Zhang T, Zhu X, Jin H, Liu Y, Shou S. Non-Canonical STING-PERK Pathway Modulation of Cellular Senescence and Therapeutic Response in Sepsis-Associated Acute Kidney Injury. Inflammation 2025; 48:696-712. [PMID: 38913144 DOI: 10.1007/s10753-024-02081-8] [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: 05/07/2024] [Revised: 06/05/2024] [Accepted: 06/11/2024] [Indexed: 06/25/2024]
Abstract
Abstract-This study explored the role of the non-canonical STING-PERK signaling pathway in sepsis-associated acute kidney injury (SA-AKI). Gene expression data from the GEO database and serum STING protein levels in patients with SA-AKI were analyzed. An LPS-induced mouse model and an in vitro model using HK-2 cells were used to investigate the role of STING in SA-AKI. STING expression was suppressed using shRNA silencing technology and the STING inhibitor C176. Kidney function, inflammatory markers, apoptosis, and senescence were measured. The role of the STING-PERK pathway was investigated by silencing PERK in HK-2 cells and administering the PERK inhibitor GSK2606414. STING mRNA expression and serum STING protein levels were significantly higher in patients with SA-AKI. Suppressing STING expression improved kidney function, reduced inflammation, and inhibited apoptosis and senescence. Silencing PERK or administering GSK2606414 suppressed the inflammatory response, cell apoptosis, and senescence, suggesting that PERK is a downstream effector in the STING signaling pathway. The STING-PERK signaling pathway exacerbates cell senescence and apoptosis in SA-AKI. Inhibiting this pathway could provide potential therapeutic targets for SA-AKI treatment.
Collapse
Affiliation(s)
- Yuxin Dong
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Guanghe Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xiaonan Situ
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Lei Xia
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Tianyi Zhang
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xiangxi Zhu
- Zunyi Medical University, No. 368 Jinwan Road, Jinhaian Community, Sanzao Town, Jinwan District, Zhuhai, 519041, Guangdong, China
| | - Heng Jin
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Yancun Liu
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Songtao Shou
- Department of Emergency Medicine, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
2
|
De Robertis M, Bozic T, Santek I, Marzano F, Markelc B, Silvestris DA, Tullo A, Pesole G, Cemazar M, Signori E. Transcriptomic analysis of the immune response to in vivo gene electrotransfer in colorectal cancer. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102448. [PMID: 39967849 PMCID: PMC11834060 DOI: 10.1016/j.omtn.2025.102448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 01/10/2025] [Indexed: 02/20/2025]
Abstract
Gene electrotransfer (GET) has recently emerged as an effective nonviral approach for plasmid DNA (pDNA) delivery in gene therapy for several pathologies, including cancer. Multiple mechanisms have been identified that influence cell biology after GET, as electroporation significantly increases pDNA uptake and immunogenicity, which may directly influence target cell death. However, the molecular effects of in vivo electroporation-mediated DNA delivery have yet to be fully elucidated. In this study, we evaluated the transcriptomes of murine colorectal tumors treated with two protocols, short- and high-voltage (SHV) electric pulses or an adapted high-voltage-low-voltage (HV-LV) pulse protocol, both of which are used for reversible electroporation. Although no significant differences in clinical outcomes were observed, variations in intratumoral macrophage infiltration were reported between the two treatment methods. Transcriptomic analysis revealed that apoptosis is a predominant mode of cell death after GET by SHV pulses, whereas GET by HV-LV pulses is associated with immunogenic necrotic pathways as well as the activation of both the innate and adaptive immune response. We demonstrated that specific pulse parameters can induce distinct immunomodulatory profiles in the tumor microenvironment; therefore, these aspects should be considered carefully when selecting the most suitable GET-based approach for antitumor immunization.
Collapse
Affiliation(s)
- Mariangela De Robertis
- Department of Biosciences, Biotechnology, and Environment, University of Bari “Aldo Moro”, 70126 Bari, Italy
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Tim Bozic
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
| | - Iva Santek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| | - Flaviana Marzano
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Bostjan Markelc
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva ulica 101, 1000 Ljubljana, Slovenia
| | | | - Apollonia Tullo
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Graziano Pesole
- Department of Biosciences, Biotechnology, and Environment, University of Bari “Aldo Moro”, 70126 Bari, Italy
- Institute of Biomembranes, Bioenergetics, and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, 70126 Bari, Italy
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Zaloska cesta 2, 1000 Ljubljana, Slovenia
- Faculty of Health Sciences, University of Primorska, Polje 42, 6310 Izola, Slovenia
| | - Emanuela Signori
- Laboratory of Molecular Pathology and Experimental Oncology, Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche, 0133 Rome, Italy
| |
Collapse
|
3
|
Zdradzinski MD, Vaughn LS, Matoo S, Trumbull K, Loomis A, Thames E, Lee SJ, Perrone-Bizzozero N, Lu Q, Larsen JM, Twiss JL. KHSRP-mediated Decay of Axonally Localized Prenyl-Cdc42 mRNA Slows Nerve Regeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636857. [PMID: 39975228 PMCID: PMC11839134 DOI: 10.1101/2025.02.06.636857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The small GTPase CDC42 promotes axon growth through actin filament polymerization and this growth is driven by axonal localization of the mRNA encoding the prenylated CDC42 isoform (Prenyl-Cdc42). Here, we show that axonal Prenyl-Cdc42 mRNA transport and translation are decreased by growth-inhibiting stimulation and increased by growth-promoting stimulation. In contrast, axonal RhoA mRNA transport and translation are increased by growth inhibition but unaffected by growth promotion. Localized increase in KHSRP in response to growth inhibitory stimulation, through elevation of intracellular Ca2+, promotes decay of axonal Prenyl-Cdc42 mRNA. Distinct 3'UTR motifs regulate transport and stability of axonal Prenyl-Cdc42 mRNA. KHSRP protein binds to a Prenyl-Cdc42 mRNA motif within nt 801-875 and the mRNA is remarkably increased in axons of Khsrp -/- mice. Selective depletion of Prenyl-Cdc42 mRNA from axons reverses the accelerated axon regeneration seen in Khsrp -/- mice.
Collapse
Affiliation(s)
- M D Zdradzinski
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208 USA
| | - Lauren S Vaughn
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208 USA
| | - Samaneh Matoo
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208 USA
| | - Kayleigh Trumbull
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC USA 29634
| | - Ashley Loomis
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208 USA
| | - Elizabeth Thames
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208 USA
| | - Seung Joon Lee
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208 USA
- Genomic Medicine, Biogen, Cambridge, MA, 02142 USA
| | | | - Qun Lu
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208 USA
- South Carolina SmartState Centers for Neurotherapeutics, University of South Carolina, Columbia, SC 29208 USA
| | - Jessica M Larsen
- Department of Chemical and Biomolecular Engineering, Clemson University, Clemson, SC USA 29634
| | - J L Twiss
- Department of Biological Sciences, University of South Carolina, Columbia, SC 20208 USA
- South Carolina SmartState Center for Childhood Neurotherapeutics, University of South Carolina, Columbia, SC 29208 USA
- Carolina Autism and Neurodevelopment Center, University of South Carolina, Columbia, SC 29208 USA
| |
Collapse
|
4
|
Wang X, Dai C, Cheng W, Wang J, Cui X, Pan G, Chen Y, Han Y, Guo X, Jiang F. Repressing cytokine storm-like response in macrophages by targeting the eIF2α-integrated stress response pathway. Int Immunopharmacol 2025; 147:113965. [PMID: 39752757 DOI: 10.1016/j.intimp.2024.113965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/21/2024] [Accepted: 12/26/2024] [Indexed: 01/29/2025]
Abstract
Cytokine storm is a life-threatening systemic hyper-inflammatory state caused by different etiologies, in which the bulk production of pro-inflammatory cytokines from activated macrophages has a central role. Integrated stress response (ISR) comprises several protective signaling pathways, leading to phosphorylation of eukaryotic initiation factor 2α (eIF2α) and repression of protein translation. Emerging evidence suggests that ISR induction may elicit anti-inflammatory effects. Currently, however, it is unclear whether targeting eIF2α phosphorylation is sufficient to inhibit the cytokine storm-like response in macrophages. Here we carried out a proof-of-concept study, employing two approaches: (1) ectopic expression of the eIF2α-S51D mutant (mimicking the phosphorylated eIF2α); (2) treatment with salubrinal, a small molecule inhibitor of eIF2α dephosphorylation. Experiments were performed in lipopolysaccharides (LPS)-stimulated macrophages and in murine models with LPS-induced acute endotoxemia. We demonstrated that in macrophages, ectopic expression of eIF2α-S51D, treatment with salubrinal, and gene silencing of PP1/GADD34 (the phosphatase holoenzyme mediating eIF2α dephosphorylation) significantly inhibited LPS-induced cytokine productions without changing their mRNA levels. Polysome PCR and puromycin incorporation assays confirmed that salubrinal suppressed de novo protein translation of the cytokines. In vivo, salubrinal pre-treatment mitigated LPS-induced acute lung injury and significantly reduced the concentration of circulating TNF-α. Salubrinal did not exhibit any effects on the Toll-like receptor 4-mediated signaling or the activation of mammalian target of rapamycin (mTOR). Our data suggest that direct manipulation of eIF2α phosphorylation, thereby bypassing all associated upstream signaling events, may suppress the cytokine storm-like response in activated macrophages, likely by decoupling the gene transcription and protein translation. Inhibiting eIF2α dephosphorylation with small molecule inhibitors may be a viable therapeutic strategy to treat disorders involving cytokine storm-like responses.
Collapse
Affiliation(s)
- Xiaoyun Wang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China; Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Chaochao Dai
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China
| | - Wen Cheng
- Department of Traditional Chinese Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Jianli Wang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Xiaopei Cui
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China; Jinan Clinical Research Center for Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Guopin Pan
- College of Pharmacy, Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, Xinxiang Key Laboratory of Vascular Remodeling Intervention and Molecular Targeted Therapy Drug Development, Xinxiang Medical University, Xinxiang, Henan Province, China
| | - Ye Chen
- Laboratory Medicine Center, The Second Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yu Han
- Department of Pathology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong Province, China
| | - Xiaosun Guo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Fan Jiang
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
5
|
Gao H, Gu T, Gao X, Song Z, Liu J, Song Y, Zhang G, Sun Y. African swine fever virus enhances viral replication by increasing intracellular reduced glutathione levels, which suppresses stress granule formation. Vet Res 2024; 55:172. [PMID: 39707514 DOI: 10.1186/s13567-024-01433-4] [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: 06/15/2024] [Accepted: 09/27/2024] [Indexed: 12/23/2024] Open
Abstract
African swine fever virus (ASFV) is a DNA virus that has significantly impacted the global swine industry. Currently, there are no effective therapies or vaccines against ASFV. Stress granules (SGs), known for their antiviral properties, are not induced during ASFV infection, even though reactive oxygen species (ROS) are generated. The mechanism by which ASFV regulates SGs formation remains unclear. This study demonstrates that ASFV antagonises SGs formation and increases intracellular levels of reduced glutathione (GSH) levels. The use of the GSH inhibitor BSO and the activator NAC confirmed that the ASFV-induced increase in GSH helps to suppress SGs formation and influences viral replication. Additionally, this study revealed that ASFV enhances GSH by upregulating the antioxidant transcription factor NRF2, as well as factors involved in GSH synthesis and regeneration, such as GCLC, and those related to the ferroptosis pathway, such as SLC7A11. Furthermore, the study uncovered that ASFV manipulates intracellular GSH levels by activating the mitochondrial protein AIFM1. This regulatory mechanism helps the virus inhibit the formation of intracellular SGs, thereby creating an optimal environment for viral replication. These findings provide new insights into the molecular strategies employed by ASFV.
Collapse
Affiliation(s)
- Han Gao
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
- School of Animal Science and Technology, Foshan University, Foshan, 528225, China
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Animal Science and Technology, Foshan University, Foshan, 528225, China
| | - Taoming Gu
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Xiaopeng Gao
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Zebu Song
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Jing Liu
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Yi Song
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China
| | - Guihong Zhang
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China.
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China.
| | - Yankuo Sun
- African Swine Fever Regional Laboratory of China (Guangzhou), South China Agricultural University, Guangzhou, 510642, China.
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
- Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming, 525000, China.
| |
Collapse
|
6
|
Bhatter N, Ivanov P. A general framework to analyze potential roles of tDRs in mammalian protein synthesis. Methods Enzymol 2024; 711:29-46. [PMID: 39952711 DOI: 10.1016/bs.mie.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2025]
Abstract
tRNA-derived RNAs (tDRs) are a heterogeneous class of small non-coding RNAs that have been implicated in numerous biological processes including the regulation of mRNA translation. A subclass of tDRs called tRNA-derived stress-induced RNAs (tiRNAs) have been shown to participate in translational control under stress where specific tiRNAs repress protein synthesis. Here, we use a prototypical tiRNA (5'-tiRNAAla) that inhibits mRNA translation in vitro and in cells as a model to study potential roles of tDRs in translational control. Specifically, we propose to use commercially available and custom-made in vitro translation systems together with sensitive luciferase-based mRNA reporters as well as transfection studies to determine potential effects of a given tDR on various aspects of protein synthesis. We overview methods to probe the capacity of specific tDRs to target specific steps of mRNA translation initiation, the most regulated step in translational control. Using 5'-tiRNAAla as an example, we analyze its effects on the integrity of the m7GTP (cap)-bound eIF4F complex and phosphorylation of eIF2α, the key regulatory molecule of the Integrated Stress Response. Using transfection studies, we also monitor whether tDRs can promote formation of stress granules (SGs), RNA granules are often formed in response to global translation repression in live cells. This simple workflow offers fast, scalable, and reliable analyses of a potential involvement of specific tDRs in the modulation of protein synthesis and provides initial hints on molecular mechanisms that underline such mRNA translation regulation.
Collapse
Affiliation(s)
- Nupur Bhatter
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's, Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Pavel Ivanov
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's, Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
7
|
Sugahara S, Unuma K, Wen S, Funakoshi T, Aki T, Uemura K. Dissociation of mitochondrial and ribosomal biogenesis during thallium administration in rat kidney. PLoS One 2024; 19:e0311884. [PMID: 39630634 PMCID: PMC11616847 DOI: 10.1371/journal.pone.0311884] [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: 04/30/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024] Open
Abstract
Thallium (Tl) is a heavy metal with toxicity comparative to other heavy metals such as As, Cd, and Hg. Nevertheless, fewer studies have been reported concerning the molecular mechanism of Tl toxicity as compared to other heavy metals. To obtain insight into Tl toxicity in the kidney, rats were intraperitoneally administered Tl2SO4 (30 mg/kg), and the kidneys were removed 2 or 5 days later to examine the effects of Tl. Transcriptome analysis using DNA microarray of the rat kidney 2 and 5 days after Tl administration showed that cytoplasmic ribosomal proteins are the most upregulated category; many of the genes involved in ribosome biosynthesis were upregulated by Tl administration. This upregulation was associated with the activation of eukaryotic transcription initiation factor 2α (eIF2α), implying that increased ribosome biogenesis was linked to the subsequent activation of protein translation. In contrast, decreased mitochondrial biogenesis was revealed via proteomic analysis. Although we found an increase in Myc, a positive regulator of both ribosomal and mitochondrial biogenesis, decreased levels of NRF1 and TFAM, positive regulators of mitochondrial biogenesis whose gene expression is directory activated by Myc, were paradoxically observed. Taken together, differing responses of ribosomes and mitochondria to Tl toxicity were observed. Failure of transmission of the Myc signal to NRF1/TFAM might be involved in the observed disruption of coordinated responses in mitochondria and ribosomes during Tl administration in rat kidney.
Collapse
Affiliation(s)
- Sho Sugahara
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kana Unuma
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Shuheng Wen
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Takeshi Funakoshi
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Toshihiko Aki
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Koichi Uemura
- Department of Forensic Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
8
|
Splichal RC, Chen K, Walton SP, Chan C. The Role of Endoplasmic Reticulum Stress on Reducing Recombinant Protein Production in Mammalian Cells. Biochem Eng J 2024; 210:109434. [PMID: 39220803 PMCID: PMC11360842 DOI: 10.1016/j.bej.2024.109434] [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] [Indexed: 09/04/2024]
Abstract
Therapeutic recombinant protein production relies on industrial scale culture of mammalian cells to produce active proteins in quantities sufficient for clinical use. The combination of stresses from industrial cell culture environment and recombinant protein production can overwhelm the protein synthesis machinery in the endoplasmic reticulum (ER). This leads to a buildup of improperly folded proteins which induces ER stress. Cells respond to ER stress by activating the Unfolded Protein Response (UPR). To restore proteostasis, ER sensor proteins reduce global protein synthesis and increase chaperone protein synthesis, and if that is insufficient the proteins are degraded. If proteostasis is still not restored, apoptosis is initiated. Increasing evidence suggests crosstalk between ER proteostasis and DNA damage repair (DDR) pathways. External factors (e.g., metabolites) from the cellular environment as well as internal factors (e.g., transgene copy number) can impact genome stability. Failure to maintain genome integrity reduces cell viability and in turn protein production. This review focuses on the association between ER stress and processes that affect protein production and secretion. The processes mediated by ER stress, including inhibition of global protein translation, chaperone protein production, degradation of misfolded proteins, DNA repair, and protein secretion, impact recombinant protein production. Recombinant protein production can be reduced by ER stress through increased autophagy and protein degradation, reduced protein secretion, and reduced DDR response.
Collapse
Affiliation(s)
- R. Chauncey Splichal
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
| | - Kevin Chen
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
| | - S. Patrick Walton
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
| | - Christina Chan
- Department of Chemical Engineering and Materials Science, Michigan State University, MI, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, MI, USA
- Department of Computer Science and Engineering, Michigan State University, MI, USA
- Institute for Quantitative Health Science and Engineering, Division of Medical Devices, Michigan State University, MI, USA
| |
Collapse
|
9
|
Zhang C, Xu H, Tang Q, Duan Y, Xia H, Huang H, Ye D, Bi F. CaMKII suppresses proteotoxicity by phosphorylating BAG3 in response to proteasomal dysfunction. EMBO Rep 2024; 25:4488-4514. [PMID: 39261742 PMCID: PMC11466968 DOI: 10.1038/s44319-024-00248-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 09/13/2024] Open
Abstract
Protein quality control serves as the primary defense mechanism for cells against proteotoxicity induced by proteasome dysfunction. While cells can limit the build-up of ubiquitinated misfolded proteins during proteasome inhibition, the precise mechanism is unclear. Here, we find that protein kinase Ca2+/Calmodulin (CaM)-dependent protein kinase II (CaMKII) maintains proteostasis during proteasome inhibition. We show that proteasome inhibition activates CaMKII, which phosphorylates B-cell lymphoma 2 (Bcl-2)-associated athanogene 3 (BAG3) at residues S173, S377, and S386. Phosphorylated BAG3 activates the heme-regulated inhibitor (HRI)- eukaryotic initiation factor-2α (eIF2α) signaling pathway, suppressing protein synthesis and the production of aggregated ubiquitinated misfolded proteins, ultimately mitigating the proteotoxic crisis. Inhibition of CaMKII exacerbates the accumulation of aggregated misfolded proteins and paraptosis induced by proteasome inhibitors. Based on these findings, we validate that combined targeting of proteasome and CaMKII accelerates tumor cell death and enhances the efficacy of proteasome inhibitors in tumor treatment. Our data unveil a new proteasomal inhibition-induced misfolded protein quality control mechanism and propose a novel therapeutic intervention for proteasome inhibitor-mediated tumor treatment.
Collapse
Affiliation(s)
- Chenliang Zhang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Huanji Xu
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiulin Tang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yichun Duan
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hongwei Xia
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Huixi Huang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Di Ye
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Feng Bi
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
10
|
Naseem S, Sun L, Qiu J. Stress granules in atherosclerosis: Insights and therapeutic opportunities. Curr Probl Cardiol 2024; 49:102760. [PMID: 39059785 DOI: 10.1016/j.cpcardiol.2024.102760] [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: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Atherosclerosis, a complex inflammatory and metabolic disorder, is the underlying cause of several life-threatening cardiovascular diseases. Stress granules (SG) are biomolecular condensates composed of proteins and mRNA that form in response to stress. Recent studies suggest a potential link between SG and atherosclerosis development. However, there remain gaps in understanding SG role in atherosclerosis development. Here we provide a thorough analysis of the role of SG in atherosclerosis, covering cellular stresses stimulation, core components, and regulatory genes in SG formation. Furthermore, we explore atherosclerosis induced factors such as inflammation, low or oscillatory shear stress (OSS), and oxidative stress (OS) may impact SG formation and then the development of atherosclerotic lesions. We have assessed how changes in SG dynamics impact pro-atherogenic processes like endothelial dysfunction, lipid metabolism, and immune cell recruitment in atherosclerosis. In summary, this review emphasizes the complex interplay between SG and atherosclerosis that could open innovative directions for targeted therapeutic strategies in preventing or treating atherosclerotic cardiovascular diseases.
Collapse
Affiliation(s)
- Sahar Naseem
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Lijuan Sun
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400030, China.
| |
Collapse
|
11
|
Martinez JC, Seluanov A, Gorbunova V. LINE1 and its host: one cell's junk is another cell's treasure. EMBO J 2024; 43:3547-3549. [PMID: 39075273 PMCID: PMC11377821 DOI: 10.1038/s44318-024-00175-5] [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: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/31/2024] Open
Abstract
New findings show that expression of the LINE1 retrotransposon RNA stimulates bone repair in mice and humans.
Collapse
Affiliation(s)
- John C Martinez
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Andrei Seluanov
- Department of Biology, University of Rochester, Rochester, NY, USA.
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| | - Vera Gorbunova
- Department of Biology, University of Rochester, Rochester, NY, USA.
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
12
|
Schloßhauer JL, Tholen L, Körner A, Kubick S, Chatzopoulou S, Hönow A, Zemella A. Promoting the production of challenging proteins via induced expression in CHO cells and modified cell-free lysates harboring T7 RNA polymerase and mutant eIF2α. Synth Syst Biotechnol 2024; 9:416-424. [PMID: 38601208 PMCID: PMC11004649 DOI: 10.1016/j.synbio.2024.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/12/2024] [Accepted: 03/16/2024] [Indexed: 04/12/2024] Open
Abstract
Chinese hamster ovary (CHO) cells are crucial in biopharmaceutical production due to their scalability and capacity for human-like post-translational modifications. However, toxic proteins and membrane proteins are often difficult-to-express in living cells. Alternatively, cell-free protein synthesis can be employed. This study explores innovative strategies for enhancing the production of challenging proteins through the modification of CHO cells by investigating both, cell-based and cell-free approaches. A major result in our study involves the integration of a mutant eIF2 translation initiation factor and T7 RNA polymerase into CHO cell lysates for cell-free protein synthesis. This resulted in elevated yields, while eliminating the necessity for exogenous additions during cell-free production, thereby substantially enhancing efficiency. Additionally, we explore the potential of the Rosa26 genomic site for the integration of T7 RNA polymerase and cell-based tetracycline-controlled protein expression. These findings provide promising advancements in bioproduction technologies, offering flexibility to switch between cell-free and cell-based protein production as needed.
Collapse
Affiliation(s)
- Jeffrey L. Schloßhauer
- Fraunhofer Project Group PZ-Syn of the Fraunhofer Institute for Cell Therapy and Immunology-IZI, Branch Bioanalytics and Bioprocesses-IZI-BB, Am Mühlenberg, Potsdam, Germany
- Fraunhofer Institute for Cell Therapy and Immunology-IZI, Branch Bioanalytics and Bioprocesses-IZI-BB, Am Mühlenberg, Potsdam, Germany
- Institute for Chemistry and Biochemistry, Laboratory of Protein Biochemistry, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Lena Tholen
- Fraunhofer Institute for Cell Therapy and Immunology-IZI, Branch Bioanalytics and Bioprocesses-IZI-BB, Am Mühlenberg, Potsdam, Germany
| | - Alexander Körner
- Fraunhofer Institute for Cell Therapy and Immunology-IZI, Branch Bioanalytics and Bioprocesses-IZI-BB, Am Mühlenberg, Potsdam, Germany
- Institute of Biotechnology, Technische Universität Berlin, Straße des 17. Juni 135, 10623, Berlin, Germany
| | - Stefan Kubick
- Fraunhofer Institute for Cell Therapy and Immunology-IZI, Branch Bioanalytics and Bioprocesses-IZI-BB, Am Mühlenberg, Potsdam, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus –Senftenberg, The Brandenburg Medical School Theodor Fontane, University of Potsdam, Potsdam, Germany
- Institute for Chemistry and Biochemistry, Laboratory of Protein Biochemistry, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Sofia Chatzopoulou
- Fraunhofer Institute for Cell Therapy and Immunology-IZI, Branch Bioanalytics and Bioprocesses-IZI-BB, Am Mühlenberg, Potsdam, Germany
| | - Anja Hönow
- New/era/mabs GmbH, August-Bebel-Str. 89, 14482, Potsdam, Germany
| | - Anne Zemella
- Fraunhofer Institute for Cell Therapy and Immunology-IZI, Branch Bioanalytics and Bioprocesses-IZI-BB, Am Mühlenberg, Potsdam, Germany
| |
Collapse
|
13
|
Zhu B, Liu S, David NL, Dion W, Doshi NK, Siegel LB, Amorim T, Andrews RE, Kumar GVN, Li H, Irfan S, Pesaresi T, Sharma AX, Sun M, Fazeli PK, Steinhauser ML. Evidence for ~12-h ultradian gene programs in humans. NPJ BIOLOGICAL TIMING AND SLEEP 2024; 1:4. [PMID: 39148626 PMCID: PMC11325440 DOI: 10.1038/s44323-024-00005-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/11/2024] [Indexed: 08/17/2024]
Abstract
Mice and many marine organisms exhibit ~12-h ultradian rhythms, however, direct evidence of ~12-h ultradian rhythms in humans is lacking. Here, we performed prospective, temporal transcriptome profiling of peripheral white blood cells from three healthy humans. All three participants independently exhibited robust ~12-h transcriptional rhythms in molecular programs involved in RNA and protein metabolism, with strong homology to circatidal gene programs previously identified in Cnidarian marine species.
Collapse
Affiliation(s)
- Bokai Zhu
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA USA
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Silvia Liu
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Natalie L. David
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - William Dion
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Nandini K. Doshi
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Lauren B. Siegel
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Tânia Amorim
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Rosemary E. Andrews
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - G. V. Naveen Kumar
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Hanwen Li
- Department of Statistics, Kenneth P. Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA USA
| | - Saad Irfan
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Tristan Pesaresi
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Ankit X. Sharma
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Michelle Sun
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Pouneh K. Fazeli
- Neuroendocrinology Unit, Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| | - Matthew L. Steinhauser
- Aging Institute of UPMC, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Center for Human Integrative Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA USA
| |
Collapse
|
14
|
Kadonosawa Y, Yokoyama M, Tatara Y, Fujita T, Yokoyama Y. Overexpression of carbonyl reductase 1 in ovarian cancer cells suppresses proliferation and activates the eIF2 signaling pathway. Oncol Lett 2024; 28:359. [PMID: 38881711 PMCID: PMC11177172 DOI: 10.3892/ol.2024.14492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 04/16/2024] [Indexed: 06/18/2024] Open
Abstract
High expression of carbonyl reductase 1 (CBR1) protein in ovarian cancer cells inhibits tumor growth and metastasis. However, the underlying mechanism is unknown. To investigate the mechanism by which CBR1 suppresses tumor growth, the present study generated ovarian cancer cells that constitutively overexpress human CBR1 (hCBR1) protein. Ovarian cancer cell lines (OVCAR-3 and SK-OV-3) were transfected with a plasmid encoding hCBR1, followed by selection with G418 to isolate hCBR1-overexpressing lines. The proliferation rates of hCBR1-overexpressing cells were then compared with those of negative control and wild-type cells. Overexpression of hCBR1 led to significant inhibition of proliferation (P<0.05). Subsequently, to investigate changes in intracellular signaling pathways, cellular proteins were extracted and subjected to proteome analysis using liquid chromatography followed by mass spectrometry. There was an inverse correlation between CBR1 protein expression and cell proliferation. In addition, Ingenuity Pathway Analysis of hCBR1-overexpressing cell lines was performed, which revealed changes in the expression of proteins involved in signaling pathways related to growth regulation. Of these, the eukaryotic translation initiation factor 2 (eIF2) signaling pathway was upregulated most prominently. Thus, alterations in multiple tumor-related signaling pathways, including eIF2 signaling, may lead to growth suppression. Taken together, the present data may lead to the development of new drugs that target CBR1 and related signaling pathways, thereby improving outcomes for patients with ovarian cancer.
Collapse
Affiliation(s)
- Yuka Kadonosawa
- Department of Obstetrics and Gynecology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Minako Yokoyama
- Department of Obstetrics and Gynecology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Yota Tatara
- Department of Stress Response Science, Center for Advanced Medical Research, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Toshitsugu Fujita
- Department of Biochemistry and Genome Biology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| | - Yoshihito Yokoyama
- Department of Obstetrics and Gynecology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori 036-8562, Japan
| |
Collapse
|
15
|
Rocha AL, Pai V, Perkins G, Chang T, Ma J, De Souza EV, Chu Q, Vaughan JM, Diedrich JK, Ellisman MH, Saghatelian A. An Inner Mitochondrial Membrane Microprotein from the SLC35A4 Upstream ORF Regulates Cellular Metabolism. J Mol Biol 2024; 436:168559. [PMID: 38580077 PMCID: PMC11292582 DOI: 10.1016/j.jmb.2024.168559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 04/07/2024]
Abstract
Upstream open reading frames (uORFs) are cis-acting elements that can dynamically regulate the translation of downstream ORFs by suppressing downstream translation under basal conditions and, in some cases, increasing downstream translation under stress conditions. Computational and empirical methods have identified uORFs in the 5'-UTRs of approximately half of all mouse and human transcripts, making uORFs one of the largest regulatory elements known. Because the prevailing dogma was that eukaryotic mRNAs produce a single functional protein, the peptides and small proteins, or microproteins, encoded by uORFs were rarely studied. We hypothesized that a uORF in the SLC35A4 mRNA is producing a functional microprotein (SLC35A4-MP) because of its conserved amino acid sequence. Through a series of biochemical and cellular experiments, we find that the 103-amino acid SLC35A4-MP is a single-pass transmembrane inner mitochondrial membrane (IMM) microprotein. The IMM contains the protein machinery crucial for cellular respiration and ATP generation, and loss of function studies with SLC35A4-MP significantly diminish maximal cellular respiration, indicating a vital role for this microprotein in cellular metabolism. The findings add SLC35A4-MP to the growing list of functional microproteins and, more generally, indicate that uORFs that encode conserved microproteins are an untapped reservoir of functional microproteins.
Collapse
Affiliation(s)
- Andréa L Rocha
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Victor Pai
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Guy Perkins
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tina Chang
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jiao Ma
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Eduardo V De Souza
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Qian Chu
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Joan M Vaughan
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jolene K Diedrich
- Mass Spectrometry Core for Proteomics and Metabolomics, The Salk Institute for Biological Studies, 10010 North Torrey Pines Road, La Jolla, CA, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Alan Saghatelian
- Clayton Foundation Laboratories for Peptide Biology, Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
16
|
Qu S, Yang C, Sun X, Huang H, Li J, Zhu Y, Zhang Y, Li L, Liang H, Zen K. Blockade of pan-viral propagation by inhibition of host cell PNPT1. Int J Antimicrob Agents 2024; 63:107124. [PMID: 38412930 DOI: 10.1016/j.ijantimicag.2024.107124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 12/29/2023] [Accepted: 02/21/2024] [Indexed: 02/29/2024]
Abstract
For successful viral propagation within infected cells, the virus needs to overcome the cellular integrated stress response (ISR), triggered during viral infection, which, in turn, inhibits general protein translation. This paper reports a tactic employed by viruses to suppress the ISR by upregulating host cell polyribonucleotide nucleotidyltransferase 1 (PNPT1). The propagation of adenovirus, murine cytomegalovirus and hepatovirus within their respective host cells induces PNPT1 expression. Notably, when PNPT1 is knocked down, the propagation of all three viruses is prevented. Mechanistically, the inhibition of PNPT1 facilitates the relocation of mitochondrial double-stranded RNAs (mt-dsRNAs) to the cytoplasm, where they activate RNA-activated protein kinase (PKR). This activation leads to eukaryotic initiation factor 2α (eIF2α) phosphorylation, resulting in the suppression of translation. Furthermore, by scrutinizing the PNPT1 recognition element and screening 17,728 drugs and bioactive compounds approved by the US Food and Drug Administration, lanatoside C was identified as a potent PNPT1 inhibitor. This compound impedes the propagation of adenovirus, murine cytomegalovirus and hepatovirus, and suppresses production of the severe acute respiratory syndrome coronavirus-2 spike protein. These discoveries shed light on a novel strategy to impede pan-viral propagation by activating the host cell mt-dsRNA-PKR-eIF2α signalling axis.
Collapse
Affiliation(s)
- Shuang Qu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Chen Yang
- Department of Gastroenterology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xinlei Sun
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hai Huang
- Department of Gastroenterology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Jiacheng Li
- Department of Gastroenterology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yujie Zhu
- Department of Gastroenterology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yaliang Zhang
- Department of Gastroenterology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Limin Li
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Hongwei Liang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China.
| | - Ke Zen
- Department of Gastroenterology, Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University Medical School, Nanjing, Jiangsu, China.
| |
Collapse
|
17
|
Chen Y, McDonald JA. Collective cell migration relies on PPP1R15-mediated regulation of the endoplasmic reticulum stress response. Curr Biol 2024; 34:1390-1402.e4. [PMID: 38428416 PMCID: PMC11003853 DOI: 10.1016/j.cub.2024.02.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 12/19/2023] [Accepted: 02/07/2024] [Indexed: 03/03/2024]
Abstract
Collective cell migration is integral to many developmental and disease processes. Previously, we discovered that protein phosphatase 1 (Pp1) promotes border cell collective migration in the Drosophila ovary. We now report that the Pp1 phosphatase regulatory subunit dPPP1R15 is a critical regulator of border cell migration. dPPP1R15 is an ortholog of mammalian PPP1R15 proteins that attenuate the endoplasmic reticulum (ER) stress response. We show that, in collectively migrating border cells, dPPP1R15 phosphatase restrains an active physiological protein kinase R-like ER kinase- (PERK)-eIF2α-activating transcription factor 4 (ATF4) stress pathway. RNAi knockdown of dPPP1R15 blocks border cell delamination from the epithelium and subsequent migration, increases eIF2α phosphorylation, reduces translation, and drives expression of the stress response transcription factor ATF4. We observe similar defects upon overexpression of ATF4 or the eIF2α kinase PERK. Furthermore, we show that normal border cells express markers of the PERK-dependent ER stress response and require PERK and ATF4 for efficient migration. In many other cell types, unresolved ER stress induces initiation of apoptosis. In contrast, border cells with chronic RNAi knockdown of dPPP1R15 survive. Together, our results demonstrate that the PERK-eIF2α-ATF4 pathway, regulated by dPPP1R15 activity, counteracts the physiological ER stress that occurs during collective border cell migration. We propose that in vivo collective cell migration is intrinsically "stressful," requiring tight homeostatic control of the ER stress response for collective cell cohesion, dynamics, and movement.
Collapse
Affiliation(s)
- Yujun Chen
- Division of Biology, Kansas State University, 1717 Claflin Road, Manhattan, KS 66506, USA
| | - Jocelyn A McDonald
- Division of Biology, Kansas State University, 1717 Claflin Road, Manhattan, KS 66506, USA.
| |
Collapse
|
18
|
Gao H, Gao X, Liu X, Luo Y, Zhong J, Liu J, Yan L, Wang H, Gong L, Zhang G, Zheng Z, Sun Y. African swine fever virus maintains de novo global cellular protein synthesis and inhibits stress granules formation via dephosphorylating eIF2α. Vet Microbiol 2024; 290:109988. [PMID: 38244395 DOI: 10.1016/j.vetmic.2024.109988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 01/22/2024]
Abstract
African swine fever virus (ASFV) has caused enormous economic losses since its first reported detection, and there is still no effective vaccines or drug treatment. During infection, viruses may employ various strategies, such as regulating the host endoplasmic reticulum stress/unfolded protein response or the formation of stress granules (SGs), to form an optimal environment for virus replication. However, how ASFV infection regulates host endoplasmic reticulum stress, eIF2α-regulated protein synthesis, and the formation of SGs remains unclear. Here, we evaluated the activation of ER stress and its three downstream axes during ASFV infection and identified a powerful dephosphorylation of eIF2α by ASFV ex vivo. This strong dephosphorylation property could maintain the efficiency of eIF2α-mediated de novo global protein synthesis, thus ensuring efficient viral protein synthesis at early stage. In addition, the powerful dephosphorylation of eIF2α by ASFV upon infection could also inhibit the formation of SGs induced by sodium arsenite. In addition, a specific eIF2α dephosphorylation inhibitor, salubrinal, could partially counteract ASFV-mediated eIF2α dephosphorylation and inhibit viral replication. Our results provide new insights into the areas of ASFV`s escape from host immunity and hijacking of the host protein translation system.
Collapse
Affiliation(s)
- Han Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510000, China
| | - Xiaopeng Gao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China
| | - Xing Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510000, China
| | - Yizhuo Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510000, China
| | - Jianhao Zhong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510000, China
| | - Jing Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510000, China
| | - Luling Yan
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510000, China
| | - Heng Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510000, China
| | - Lang Gong
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China
| | - Guihong Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510000, China
| | - Zezhong Zheng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510000, China.
| | - Yankuo Sun
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; African Swine Fever Regional Laboratory of China (Guangzhou), Guangzhou 510642, China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, Guangzhou 510000, China.
| |
Collapse
|
19
|
Paul PK, Umarvaish S, Bajaj S, S. RF, Mohan H, Annaert W, Chaudhary V. Maintenance of proteostasis by Drosophila Rer1 is essential for competitive cell survival and Myc-driven overgrowth. PLoS Genet 2024; 20:e1011171. [PMID: 38408084 PMCID: PMC10919865 DOI: 10.1371/journal.pgen.1011171] [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: 05/17/2023] [Revised: 03/07/2024] [Accepted: 02/05/2024] [Indexed: 02/28/2024] Open
Abstract
Defects in protein homeostasis can induce proteotoxic stress, affecting cellular fitness and, consequently, overall tissue health. In various growing tissues, cell competition based mechanisms facilitate detection and elimination of these compromised, often referred to as 'loser', cells by the healthier neighbors. The precise connection between proteotoxic stress and competitive cell survival remains largely elusive. Here, we reveal the function of an endoplasmic reticulum (ER) and Golgi localized protein Rer1 in the regulation of protein homeostasis in the developing Drosophila wing epithelium. Our results show that loss of Rer1 leads to proteotoxic stress and PERK-mediated phosphorylation of eukaryotic initiation factor 2α. Clonal analysis showed that rer1 mutant cells are identified as losers and eliminated through cell competition. Interestingly, we find that Rer1 levels are upregulated upon Myc-overexpression that causes overgrowth, albeit under high proteotoxic stress. Our results suggest that increased levels of Rer1 provide cytoprotection to Myc-overexpressing cells by alleviating the proteotoxic stress and thereby supporting Myc-driven overgrowth. In summary, these observations demonstrate that Rer1 acts as a novel regulator of proteostasis in Drosophila and reveal its role in competitive cell survival.
Collapse
Affiliation(s)
- Pranab Kumar Paul
- Cell and developmental signaling laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh, India
| | - Shruti Umarvaish
- Cell and developmental signaling laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh, India
| | - Shivani Bajaj
- Cell and developmental signaling laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh, India
| | - Rishana Farin S.
- Cell and developmental signaling laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh, India
| | - Hrudya Mohan
- Cell and developmental signaling laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh, India
| | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, KU Leuven, Leuven, Belgium, and Department of Neurosciences, KU Leuven, Gasthuisberg, Leuven, Belgium
| | - Varun Chaudhary
- Cell and developmental signaling laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research Bhopal, Bhopal, Madhya Pradesh, India
| |
Collapse
|
20
|
Lyu J, Zhuang Y, Lin Y. Circadian regulation of translation. RNA Biol 2024; 21:14-24. [PMID: 39324589 PMCID: PMC11441039 DOI: 10.1080/15476286.2024.2408524] [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] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/27/2024] Open
Abstract
Most, if not all organisms exhibit robust rhythmicity of their biological functions, allowing a perpetual adaptation to external clues within the daily 24 hours-cycle. Studies on circadian rhythm regulation primarily focused on transcriptional level, considering mRNA levels to represent the primary determinant of oscillations of intracellular protein levels. However, a plethora of emerging evidence suggests that post-transcriptional regulation, particularly rhythmic mRNA translation, is not solely reliant on the oscillation of transcription. Instead, the circadian regulation of mRNA translation plays a critical role as well. A comprehensive understanding of these mechanisms underlying rhythmic translation and its regulation should bridge the gap in rhythm regulation beyond RNA fluctuations in research, and greatly enhance our comprehension of rhythm generation and maintenance. In this review, we summarize the major mechanisms of circadian regulation of translation, including regulation of translation initiation, elongation, and the alteration in rhythmic translation to external stresses, such as endoplasmic reticulum (ER) stress and ageing. We also illuminate the complex interplay between phase separation and mRNA translation. Together, we have summarized various facets of mRNA translation in circadian regulation, to set on forthcoming studies into the intricate regulatory mechanisms underpinning circadian rhythms and their implications for associated disorders.
Collapse
Affiliation(s)
- Jiali Lyu
- State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Centre for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yanrong Zhuang
- State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Centre for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Yi Lin
- State Key Laboratory of Membrane Biology, IDG/McGovern Institute for Brain Research, Tsinghua-Peking Joint Centre for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
21
|
Ando R, Ishikawa Y, Kamada Y, Izawa S. Contribution of the yeast bi-chaperone system in the restoration of the RNA helicase Ded1 and translational activity under severe ethanol stress. J Biol Chem 2023; 299:105472. [PMID: 37979914 PMCID: PMC10746526 DOI: 10.1016/j.jbc.2023.105472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
Preexposure to mild stress often improves cellular tolerance to subsequent severe stress. Severe ethanol stress (10% v/v) causes persistent and pronounced translation repression in Saccharomyces cerevisiae. However, it remains unclear whether preexposure to mild stress can mitigate translation repression in yeast cells under severe ethanol stress. We found that the translational activity of yeast cells pretreated with 6% (v/v) ethanol was initially significantly repressed under subsequent 10% ethanol but was then gradually restored even under severe ethanol stress. We also found that 10% ethanol caused the aggregation of Ded1, which plays a key role in translation initiation as a DEAD-box RNA helicase. Pretreatment with 6% ethanol led to the gradual disaggregation of Ded1 under subsequent 10% ethanol treatment in wild-type cells but not in fes1Δhsp104Δ cells, which are deficient in Hsp104 with significantly reduced capacity for Hsp70. Hsp104 and Hsp70 are key components of the bi-chaperone system that play a role in yeast protein quality control. fes1Δhsp104Δ cells did not restore translational activity under 10% ethanol, even after pretreatment with 6% ethanol. These results indicate that the regeneration of Ded1 through the bi-chaperone system leads to the gradual restoration of translational activity under continuous severe stress. This study provides new insights into the acquired tolerance of yeast cells to severe ethanol stress and the resilience of their translational activity.
Collapse
Affiliation(s)
- Ryoko Ando
- Graduate School of Science and Technology, Kyoto Institute of Technology, Sakyo-ku, Kyoto, Japan
| | - Yu Ishikawa
- Graduate School of Science and Technology, Kyoto Institute of Technology, Sakyo-ku, Kyoto, Japan
| | | | - Shingo Izawa
- Graduate School of Science and Technology, Kyoto Institute of Technology, Sakyo-ku, Kyoto, Japan.
| |
Collapse
|
22
|
Zhang L, Zhi K, Su Y, Peng W, Meng X. Effect of eIF2α in Neuronal Injury Induced by High Glucose and the Protective Mechanism of Resveratrol. Mol Neurobiol 2023; 60:6043-6059. [PMID: 37410333 DOI: 10.1007/s12035-023-03457-x] [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: 05/09/2023] [Accepted: 06/21/2023] [Indexed: 07/07/2023]
Abstract
Diabetes mellitus (DM) is a type of metabolic disease characterized by chronic hyperglycemia, which can lead to different degrees of cognitive decline. Therefore, it is crucial to explore the molecular biological mechanisms of neuronal injury. In this study, we investigated the effect of high glucose on eIF2α expression and the mechanism of neuronal injury, and on this basis, the protective mechanism of resveratrol is explored. Treatment with 50 mM high glucose in cortical neurons increased the levels of eIF2α phosphorylation; the expressions of ATF4 and CHOP increased. ISRIB alleviated high glucose-induced neuronal injury by reducing eIF2α phosphorylation when neurons were pretreated with ISRIB before high glucose treatment. Compared with the high glucose-treated group, resveratrol pretreatment reduced eIF2α phosphorylation, the levels of its downstream molecules ATF4 and CHOP, and LDH release. Resveratrol reduced the level of cortical eIF2α phosphorylation and the expression of its downstream molecules in DM mice and improved the ability of spatial memory and learning in DM mice without affecting anxiety and motor performance. Meanwhile, resveratrol modulated the expression of Bcl-2 protein and also effectively decreased the DM-induced up-regulation of Bax, caspase-3, p53, p21, and p16. Taken together, these results suggested that high glucose caused neuronal injury through the eIF2α/ATF4/CHOP pathway which was inhibited by ISRIB and resveratrol. The present study indicates that eIF2α is the new target for the treatment of high glucose-induced neuronal injury, and resveratrol is a potential new medicine to treat diabetes encephalopathy.
Collapse
Affiliation(s)
- Lijing Zhang
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Kaining Zhi
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanfang Su
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenpeng Peng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xianfang Meng
- Department of Neurobiology, Institute of Brain Research, School of Basic Medical Sciences, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
23
|
Zhao N, Ho JSY, Meng F, Zheng S, Kurland AP, Tian L, Rea-Moreno M, Song X, Seo JS, Kaniskan HÜ, Te Velthuis AJW, Tortorella D, Chen YW, Johnson JR, Jin J, Marazzi I. Generation of host-directed and virus-specific antivirals using targeted protein degradation promoted by small molecules and viral RNA mimics. Cell Host Microbe 2023; 31:1154-1169.e10. [PMID: 37339625 PMCID: PMC10528416 DOI: 10.1016/j.chom.2023.05.030] [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: 08/01/2022] [Revised: 04/24/2023] [Accepted: 05/30/2023] [Indexed: 06/22/2023]
Abstract
Targeted protein degradation (TPD), as exemplified by proteolysis-targeting chimera (PROTAC), is an emerging drug discovery platform. PROTAC molecules, which typically contain a target protein ligand linked to an E3 ligase ligand, recruit a target protein to the E3 ligase to induce its ubiquitination and degradation. Here, we applied PROTAC approaches to develop broad-spectrum antivirals targeting key host factors for many viruses and virus-specific antivirals targeting unique viral proteins. For host-directed antivirals, we identified a small-molecule degrader, FM-74-103, that elicits selective degradation of human GSPT1, a translation termination factor. FM-74-103-mediated GSPT1 degradation inhibits both RNA and DNA viruses. Among virus-specific antivirals, we developed viral RNA oligonucleotide-based bifunctional molecules (Destroyers). As a proof of principle, RNA mimics of viral promoter sequences were used as heterobifunctional molecules to recruit and target influenza viral polymerase for degradation. This work highlights the broad utility of TPD to rationally design and develop next-generation antivirals.
Collapse
Affiliation(s)
- Nan Zhao
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Sook Yuin Ho
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Fanye Meng
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Simin Zheng
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrew P Kurland
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lu Tian
- Department of Otolaryngology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Martha Rea-Moreno
- Department of Otolaryngology, Master of Science in Biomedical Science Program, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xiangyang Song
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ji-Seon Seo
- Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - H Ümit Kaniskan
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Aartjan J W Te Velthuis
- Lewis Thomas Laboratory, Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Domenico Tortorella
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ya-Wen Chen
- Department of Otolaryngology, Department of Cell, Developmental and Regenerative Biology, Black Family Stem Cell Institute, Institute for Airway Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jeffrey R Johnson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Ivan Marazzi
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
24
|
Schmal C, Maier B, Ashwal-Fluss R, Bartok O, Finger AM, Bange T, Koutsouli S, Robles MS, Kadener S, Herzel H, Kramer A. Alternative polyadenylation factor CPSF6 regulates temperature compensation of the mammalian circadian clock. PLoS Biol 2023; 21:e3002164. [PMID: 37379316 DOI: 10.1371/journal.pbio.3002164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 05/15/2023] [Indexed: 06/30/2023] Open
Abstract
A defining property of circadian clocks is temperature compensation, characterized by the resilience of their near 24-hour free-running periods against changes in environmental temperature within the physiological range. While temperature compensation is evolutionary conserved across different taxa of life and has been studied within many model organisms, its molecular underpinnings remain elusive. Posttranscriptional regulations such as temperature-sensitive alternative splicing or phosphorylation have been described as underlying reactions. Here, we show that knockdown of cleavage and polyadenylation specificity factor subunit 6 (CPSF6), a key regulator of 3'-end cleavage and polyadenylation, significantly alters circadian temperature compensation in human U-2 OS cells. We apply a combination of 3'-end-RNA-seq and mass spectrometry-based proteomics to globally quantify changes in 3' UTR length as well as gene and protein expression between wild-type and CPSF6 knockdown cells and their dependency on temperature. Since changes in temperature compensation behavior should be reflected in alterations of temperature responses within one or all of the 3 regulatory layers, we statistically assess differential responses upon changes in ambient temperature between wild-type and CPSF6 knockdown cells. By this means, we reveal candidate genes underlying circadian temperature compensation, including eukaryotic translation initiation factor 2 subunit 1 (EIF2S1).
Collapse
Affiliation(s)
- Christoph Schmal
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Bert Maier
- Laboratory of Chronobiology, Institute for Medical immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Reut Ashwal-Fluss
- Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Osnat Bartok
- Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anna-Marie Finger
- Laboratory of Chronobiology, Institute for Medical immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Tanja Bange
- Institute of Medical Psychology, Faculty of Medicine, Ludwig-Maximilians-Universität München, München, Germany
| | - Stella Koutsouli
- Institute of Medical Psychology, Faculty of Medicine, Ludwig-Maximilians-Universität München, München, Germany
| | - Maria S Robles
- Institute of Medical Psychology, Faculty of Medicine, Ludwig-Maximilians-Universität München, München, Germany
| | - Sebastian Kadener
- Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Hanspeter Herzel
- Institute for Theoretical Biology, Humboldt-Universität zu Berlin and Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Achim Kramer
- Laboratory of Chronobiology, Institute for Medical immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
25
|
Yang H, Tan M, Gao Z, Wang S, Lyu L, Ding H. Role of Hydrogen Sulfide and Hypoxia in Hepatic Angiogenesis of Portal Hypertension. J Clin Transl Hepatol 2023; 11:675-681. [PMID: 36969894 PMCID: PMC10037502 DOI: 10.14218/jcth.2022.00217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/23/2022] [Accepted: 09/21/2022] [Indexed: 01/05/2023] Open
Abstract
The pathogenesis of portal hypertension remains unclear, and is believed to involve dysfunction of liver sinusoidal endothelial cells (LSEC), activation of hepatic stellate cells (HSC), dysregulation of endogenous hydrogen sulfide (H2S) synthesis, and hypoxia-induced angiogenic responses. H2S, a novel gas transmitter, plays an important role in various pathophysiological processes, especially in hepatic angiogenesis. Inhibition of endogenous H2S synthase by pharmaceutical agents or gene silencing may enhance the angiogenic response of endothelial cells. Hypoxia-inducible factor-1 (HIF-1) is the main transcription factor of hypoxia, which induces hepatic angiogenesis through up-regulation of vascular endothelial growth factor (VEGF) in HSC and LSEC. H2S has also been shown to be involved in the regulation of VEGF-mediated angiogenesis. Therefore, H2S and HIF-1 may be potential therapeutic targets for portal hypertension. The effects of H2S donors or prodrugs on the hemodynamics of portal hypertension and the mechanism of H2S-induced angiogenesis are promising areas for future research.
Collapse
Affiliation(s)
- Huaxiang Yang
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| | - Mingjie Tan
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| | - Zhuqing Gao
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| | - Shanshan Wang
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
- Cell Biology Laboratory, Beijing Institute of Hepatology, Beijing, China
| | - Lingna Lyu
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| | - Huiguo Ding
- Department of Gastroenterology and Hepatology, Beijing You’an Hospital Affiliated to Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Lin DW, Hsu YC, Chang CC, Hsieh CC, Lin CL. Insights into the Molecular Mechanisms of NRF2 in Kidney Injury and Diseases. Int J Mol Sci 2023; 24:6053. [PMID: 37047024 PMCID: PMC10094034 DOI: 10.3390/ijms24076053] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Redox is a constant phenomenon in organisms. From the signaling pathway transduction to the oxidative stress during the inflammation and disease process, all are related to reduction-oxidation (redox). Nuclear factor erythroid 2-related factor 2 (NRF2) is a transcription factor targeting many antioxidant genes. In non-stressed conditions, NRF2 maintains the hemostasis of redox with housekeeping work. It expresses constitutively with basal activity, maintained by Kelch-like-ECH-associated protein 1 (KEAP1)-associated ubiquitination and degradation. When encountering stress, it can be up-regulated by several mechanisms to exert its anti-oxidative ability in diseases or inflammatory processes to protect tissues and organs from further damage. From acute kidney injury to chronic kidney diseases, such as diabetic nephropathy or glomerular disease, many results of studies have suggested that, as a master of regulating redox, NRF2 is a therapeutic option. It was not until the early termination of the clinical phase 3 trial of diabetic nephropathy due to heart failure as an unexpected side effect that we renewed our understanding of NRF2. NRF2 is not just a simple antioxidant capacity but has pleiotropic activities, harmful or helpful, depending on the conditions and backgrounds.
Collapse
Affiliation(s)
- Da-Wei Lin
- Department of Internal Medicine, St. Martin de Porres Hospital, Chiayi 600, Taiwan;
| | - Yung-Chien Hsu
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan
| | - Cheng-Chih Chang
- Department of Surgery, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (C.-C.C.); (C.-C.H.)
| | - Ching-Chuan Hsieh
- Department of Surgery, Chang Gung Memorial Hospital, Chiayi 613, Taiwan; (C.-C.C.); (C.-C.H.)
| | - Chun-Liang Lin
- Department of Nephrology, Chang Gung Memorial Hospital, Chiayi 613, Taiwan
- Kidney and Diabetic Complications Research Team (KDCRT), Chang Gung Memorial Hospital, Chiayi 613, Taiwan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
- Kidney Research Center, Chang Gung Memorial Hospital, Taipei 105, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| |
Collapse
|
27
|
Vijayalalitha R, Archita T, Juanitaa GR, Jayasuriya R, Amin KN, Ramkumar KM. Role of Long Non-Coding RNA in Regulating ER Stress Response to the Progression of Diabetic Complications. Curr Gene Ther 2023; 23:96-110. [PMID: 35927920 DOI: 10.2174/1566523222666220801141450] [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: 01/06/2022] [Revised: 04/08/2022] [Accepted: 04/22/2022] [Indexed: 11/22/2022]
Abstract
Chronic hyperglycemia damages the nerves and blood vessels, culminating in other vascular complications. Such complications enhance cytokine, oxidative and endoplasmic reticulum (ER) stress. ER is the primary organelle where proteins are synthesised and attains confirmatory changes before its site of destination. Perturbation of ER homeostasis activates signaling sensors within its lumen, the unfolded protein response (UPR) that orchestrates ER stress and is extensively studied. Increased ER stress markers are reported in diabetic complications in addition to lncRNA that acts as an upstream marker inducing ER stress response. This review focuses on the mechanisms of lncRNA that regulate ER stress markers, especially during the progression of diabetic complications. Through this systemic review, we showcase the dysfunctional lncRNAs that act as a leading cause of ER stress response to the progression of diabetic complications.
Collapse
Affiliation(s)
- Ramanarayanan Vijayalalitha
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Tca Archita
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - George Raj Juanitaa
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Ravichandran Jayasuriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Karan Naresh Amin
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| |
Collapse
|
28
|
Datta C, Truesdell SS, Wu KQ, Bukhari SIA, Ngue H, Buchanan B, Le Tonqueze O, Lee S, Kollu S, Granovetter MA, Boukhali M, Kreuzer J, Batool MS, Balaj L, Haas W, Vasudevan S. Ribosome changes reprogram translation for chemosurvival in G0 leukemic cells. SCIENCE ADVANCES 2022; 8:eabo1304. [PMID: 36306353 PMCID: PMC9616492 DOI: 10.1126/sciadv.abo1304] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Quiescent leukemic cells survive chemotherapy, with translation changes. Our data reveal that FXR1, a protein amplified in several aggressive cancers, is elevated in quiescent and chemo-treated leukemic cells and promotes chemosurvival. This suggests undiscovered roles for this RNA- and ribosome-associated protein in chemosurvival. We find that FXR1 depletion reduces translation, with altered rRNAs, snoRNAs, and ribosomal proteins (RPs). FXR1 regulates factors that promote transcription and processing of ribosomal genes and snoRNAs. Ribosome changes in FXR1-overexpressing cells, including RPLP0/uL10 levels, activate eIF2α kinases. Accordingly, phospho-eIF2α increases, enabling selective translation of survival and immune regulators in FXR1-overexpressing cells. Overriding these genes or phospho-eIF2α with inhibitors reduces chemosurvival. Thus, elevated FXR1 in quiescent or chemo-treated leukemic cells alters ribosomes that trigger stress signals to redirect translation for chemosurvival.
Collapse
Affiliation(s)
- Chandreyee Datta
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Samuel S. Truesdell
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Keith Q. Wu
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Syed I. A. Bukhari
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Harrison Ngue
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Brienna Buchanan
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Olivier Le Tonqueze
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Sooncheol Lee
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Swapna Kollu
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Madeleine A. Granovetter
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Myriam Boukhali
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Johannes Kreuzer
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Maheen S. Batool
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Leonora Balaj
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Wilhelm Haas
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Shobha Vasudevan
- Massachusetts General Hospital Cancer Center, Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
- Corresponding author.
| |
Collapse
|
29
|
Wang T, Chen P, Weir S, Baltezor M, Schoenen FJ, Chen Q. Novel compound C150 inhibits pancreatic cancer through induction of ER stress and proteosome assembly. Front Oncol 2022; 12:870473. [PMID: 36276125 PMCID: PMC9579335 DOI: 10.3389/fonc.2022.870473] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022] Open
Abstract
Pancreatic cancer is a devastating disease with a dismal prognosis and poor treatment outcomes. Searching for new agents for pancreatic cancer treatment is of great significance. We previously identified a novel activity of compound C150 to inhibit pancreatic cancer epithelial-to-mesenchymal transition (EMT). Here, we further revealed its mechanism of action. C150 induced ER stress in pancreatic cancer cells and subsequently increased proteasome activity by enhancing proteasome assembly, which subsequently enhanced the degradation of critical EMT transcription factors (EMT-TFs). In addition, as cellular responses to ER stress, autophagy was elevated, and general protein synthesis was inhibited in pancreatic cancer cells. Besides EMT inhibition, the C150-induced ER stress resulted in G2/M cell cycle arrest, which halted cell proliferation and led to cellular senescence. In an orthotopic syngeneic mouse model, an oral dose of C150 at 150 mg/kg 3× weekly significantly increased survival of mice bearing pancreatic tumors, and reduced tumor growth and ascites occurrence. These results suggested that compound C150 holds promises in comprehensively inhibiting pancreatic cancer progression.
Collapse
Affiliation(s)
- Tao Wang
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas, KS, United States
| | - Ping Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas, KS, United States
| | - Scott Weir
- Department of Cancer Biology, University of Kansas Medical Center, Kansas, KS, United States
| | - Michael Baltezor
- Biotechnology Innovation and Optimization Center, University of Kansas, Lawrence, KS, United States
| | - Frank J. Schoenen
- Higuchi Biosciences Center, University of Kansas, Lawrence, KS, United States
- Medicinal Chemistry Core Laboratory, Lead Development and Optimization Shared Resource, University of Kansas Cancer Center, Lawrence, KS, United States
| | - Qi Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas, KS, United States
- *Correspondence: Qi Chen,
| |
Collapse
|
30
|
Zhang C, Yang B, Biazik JM, Webster RF, Xie W, Tang J, Allioux FM, Abbasi R, Mousavi M, Goldys EM, Kilian KA, Chandrawati R, Esrafilzadeh D, Kalantar-Zadeh K. Gallium Nanodroplets are Anti-Inflammatory without Interfering with Iron Homeostasis. ACS NANO 2022; 16:8891-8903. [PMID: 35613428 DOI: 10.1021/acsnano.1c10981] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Gallium (Ga) compounds, as the source of Ga ions (Ga3+), have been historically used as anti-inflammatories. Currently, the widely accepted mechanisms of the anti-inflammatory effects for Ga3+ are rationalized on the basis of their similarities to ferric ions (Fe3+), which permits Ga3+ to bind with Fe-binding proteins and subsequently disturbs the Fe homeostasis in the immune cells. Here in contrast to the classic views, our study presents the mechanisms of Ga as anti-inflammatory by delivering Ga nanodroplets (GNDs) into lipopolysaccharide-induced macrophages and exploring the processes. The GNDs show a selective inhibition of nitric oxide (NO) production without affecting the accumulation of pro-inflammatory mediators. This is explained by GNDs disrupting the synthesis of inducible NO synthase in the activated macrophages by upregulating the levels of eIF2α phosphorylation, without interfering with the Fe homeostasis. The Fe3+ transferrin receptor-independent endocytosis of GNDs by the cells prompts a fundamentally different mechanism as anti-inflammatories in comparison to that imparted by Ga3+. This study reveals the fundamental molecular basis of GND-macrophage interactions, which may provide additional avenues for the use of Ga for anti-inflammatory and future biomedical and pharmaceutical applications.
Collapse
Affiliation(s)
- Chengchen Zhang
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Biyao Yang
- ARC Centre of Excellence for Nanoscale BioPhotonics, Graduate School of Biomedical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Joanna M Biazik
- Electron Microscope Unit, Mark Wainwright Analytical Centre, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Richard F Webster
- Electron Microscope Unit, Mark Wainwright Analytical Centre, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Wanjie Xie
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Jianbo Tang
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Francois-Marie Allioux
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Roozbeh Abbasi
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Maedehsadat Mousavi
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Ewa M Goldys
- ARC Centre of Excellence for Nanoscale BioPhotonics, Graduate School of Biomedical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Kristopher A Kilian
- School of Chemistry, School of Materials Science and Engineering, Australian Centre for NanoMedicine, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Rona Chandrawati
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Dorna Esrafilzadeh
- Graduate School of Biomedical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Kourosh Kalantar-Zadeh
- School of Chemical Engineering, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| |
Collapse
|
31
|
Tibolone Pre-Treatment Ameliorates the Dysregulation of Protein Translation and Transport Generated by Palmitic Acid-Induced Lipotoxicity in Human Astrocytes: A Label-Free MS-Based Proteomics and Network Analysis. Int J Mol Sci 2022; 23:ijms23126454. [PMID: 35742897 PMCID: PMC9223656 DOI: 10.3390/ijms23126454] [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: 04/29/2022] [Revised: 05/27/2022] [Accepted: 05/30/2022] [Indexed: 01/27/2023] Open
Abstract
Excessive accumulation and release of fatty acids (FAs) in adipose and non-adipose tissue are characteristic of obesity and are associated with the leading causes of death worldwide. Chronic exposure to high concentrations of FAs such as palmitic acid (pal) is a risk factor for developing different neurodegenerative diseases (NDs) through several mechanisms. In the brain, astrocytic dysregulation plays an essential role in detrimental processes like metabolic inflammatory state, oxidative stress, endoplasmic reticulum stress, and autophagy impairment. Evidence shows that tibolone, a synthetic steroid, induces neuroprotective effects, but its molecular mechanisms upon exposure to pal remain largely unknown. Due to the capacity of identifying changes in the whole data-set of proteins and their interaction allowing a deeper understanding, we used a proteomic approach on normal human astrocytes under supraphysiological levels of pal as a model to induce cytotoxicity, finding changes of expression in proteins related to translation, transport, autophagy, and apoptosis. Additionally, tibolone pre-treatment showed protective effects by restoring those same pal-altered processes and increasing the expression of proteins from cell survival processes. Interestingly, ARF3 and IPO7 were identified as relevant proteins, presenting a high weight in the protein-protein interaction network and significant differences in expression levels. These proteins are related to transport and translation processes, and their expression was restored by tibolone. This work suggests that the damage caused by pal in astrocytes simultaneously involves different mechanisms that the tibolone can partially revert, making tibolone interesting for further research to understand how to modulate these damages.
Collapse
|
32
|
Kathjoo MB, Srivastava M. A link between DNA double‐strand breaks and regulation of global translation. FEBS J 2022; 289:3093-3096. [DOI: 10.1111/febs.16398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 12/01/2022]
|
33
|
Hernández-Elvira M, Sunnerhagen P. Post-transcriptional regulation during stress. FEMS Yeast Res 2022; 22:6585650. [PMID: 35561747 PMCID: PMC9246287 DOI: 10.1093/femsyr/foac025] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 11/12/2022] Open
Abstract
To remain competitive, cells exposed to stress of varying duration, rapidity of onset, and intensity, have to balance their expenditure on growth and proliferation versus stress protection. To a large degree dependent on the time scale of stress exposure, the different levels of gene expression control: transcriptional, post-transcriptional and post-translational, will be engaged in stress responses. The post-transcriptional level is appropriate for minute-scale responses to transient stress, and for recovery upon return to normal conditions. The turnover rate, translational activity, covalent modifications, and subcellular localisation of RNA species are regulated under stress by multiple cellular pathways. The interplay between these pathways is required to achieve the appropriate signalling intensity and prevent undue triggering of stress-activated pathways at low stress levels, avoid overshoot, and down-regulate the response in a timely fashion. As much of our understanding of post-transcriptional regulation has been gained in yeast, this review is written with a yeast bias, but attempts to generalise to other eukaryotes. It summarises aspects of how post-transcriptional events in eukaryotes mitigate short-term environmental stresses, and how different pathways interact to optimise the stress response under shifting external conditions.
Collapse
Affiliation(s)
- Mariana Hernández-Elvira
- Department of Chemistry and Molecular Biology, Lundberg Laboratory, University of Gothenburg, P.O. Box 462, S-405 30 Göteborg, Sweden
| | - Per Sunnerhagen
- Department of Chemistry and Molecular Biology, Lundberg Laboratory, University of Gothenburg, P.O. Box 462, S-405 30 Göteborg, Sweden
| |
Collapse
|
34
|
Zhang D, Zhu L, Wang Y, Li P, Gao Y. Translational Control of COVID-19 and Its Therapeutic Implication. Front Immunol 2022; 13:857490. [PMID: 35422818 PMCID: PMC9002053 DOI: 10.3389/fimmu.2022.857490] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/07/2022] [Indexed: 12/19/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of COVID-19, which has broken out worldwide for more than two years. However, due to limited treatment, new cases of infection are still rising. Therefore, there is an urgent need to understand the basic molecular biology of SARS-CoV-2 to control this virus. SARS-CoV-2 replication and spread depend on the recruitment of host ribosomes to translate viral messenger RNA (mRNA). To ensure the translation of their own mRNAs, the SARS-CoV-2 has developed multiple strategies to globally inhibit the translation of host mRNAs and block the cellular innate immune response. This review provides a comprehensive picture of recent advancements in our understanding of the molecular basis and complexity of SARS-CoV-2 protein translation. Specifically, we summarize how this viral infection inhibits host mRNA translation to better utilize translation elements for translation of its own mRNA. Finally, we discuss the potential of translational components as targets for therapeutic interventions.
Collapse
Affiliation(s)
- Dejiu Zhang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Lei Zhu
- College of Basic Medical, Qingdao Binhai University, Qingdao, China
| | - Yin Wang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Peifeng Li
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| | - Yanyan Gao
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
35
|
Carey JL, Guo L. Liquid-Liquid Phase Separation of TDP-43 and FUS in Physiology and Pathology of Neurodegenerative Diseases. Front Mol Biosci 2022; 9:826719. [PMID: 35187086 PMCID: PMC8847598 DOI: 10.3389/fmolb.2022.826719] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Liquid-liquid phase separation of RNA-binding proteins mediates the formation of numerous membraneless organelles with essential cellular function. However, aberrant phase transition of these proteins leads to the formation of insoluble protein aggregates, which are pathological hallmarks of neurodegenerative diseases including ALS and FTD. TDP-43 and FUS are two such RNA-binding proteins that mislocalize and aggregate in patients of ALS and FTD. They have similar domain structures that provide multivalent interactions driving their phase separation in vitro and in the cellular environment. In this article, we review the factors that mediate and regulate phase separation of TDP-43 and FUS. We also review evidences that connect the phase separation property of TDP-43 and FUS to their functional roles in cells. Aberrant phase transition of TDP-43 and FUS leads to protein aggregation and disrupts their regular cell function. Therefore, restoration of functional protein phase of TDP-43 and FUS could be beneficial for neuronal cells. We discuss possible mechanisms for TDP-43 and FUS aberrant phase transition and aggregation while reviewing the methods that are currently being explored as potential therapeutic strategies to mitigate aberrant phase transition and aggregation of TDP-43 and FUS.
Collapse
Affiliation(s)
| | - Lin Guo
- Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
36
|
Verbelen B, Girardi T, Sulima SO, Vereecke S, Verstraete P, Verbeeck J, Royaert J, Cinque S, Montanaro L, Penzo M, Imbrechts M, Geukens N, Geuens T, Dierckx K, Pepe D, Kampen K, De Keersmaecker K. Exploitation of the ribosomal protein L10 R98S mutation to enhance recombinant protein production in mammalian cells. Eng Life Sci 2022; 22:100-114. [PMID: 35140557 PMCID: PMC8811726 DOI: 10.1002/elsc.202100124] [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: 09/20/2021] [Revised: 12/03/2021] [Accepted: 12/24/2021] [Indexed: 11/17/2022] Open
Abstract
Mammalian cells are commonly used to produce recombinant protein therapeutics, but suffer from a high cost per mg of protein produced. There is therefore great interest in improving protein yields to reduce production cost. We present an entirely novel approach to reach this goal through direct engineering of the cellular translation machinery by introducing the R98S point mutation in the catalytically essential ribosomal protein L10 (RPL10-R98S). Our data support that RPL10-R98S enhances translation levels and fidelity and reduces proteasomal activity in lymphoid Ba/F3 and Jurkat cell models. In HEK293T cells cultured in chemically defined medium, knock-in of RPL10-R98S was associated with a 1.7- to 2.5-fold increased production of four transiently expressed recombinant proteins and 1.7-fold for one out of two stably expressed proteins. In CHO-S cells, eGFP reached a 2-fold increased expression under stable but not transient conditions, but there was no production benefit for monoclonal antibodies. The RPL10-R98S associated production gain thus depends on culture conditions, cell type, and the nature of the expressed protein. Our study demonstrates the potential for using a ribosomal protein mutation for pharmaceutical protein production gains, and further research on how various factors influence RPL10-R98S phenotypes can maximize its exploitability for the mammalian protein production industry.
Collapse
Affiliation(s)
- Benno Verbelen
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Tiziana Girardi
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
- Flamingo TherapeuticsLeuvenBelgium
| | - Sergey O. Sulima
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
- Institute of Biological and Medical ImagingHelmholtz Zentrum München (GmbH)NeuherbergOberschleißheimGermany
- Center for Translational Cancer ResearchTechnical University of MunichMünchenGermany
| | - Stijn Vereecke
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Paulien Verstraete
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Jelle Verbeeck
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Jonathan Royaert
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Sonia Cinque
- Laboratory for RNA Cancer BiologyDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Lorenzo Montanaro
- IRCCS Azienda Ospedaliero‐Universitaria di BolognaBolognaItaly
- Department of ExperimentalDiagnostic and Specialty Medicine and Center for Applied Biomedical Research (CRBA)Alma Mater Studiorum‐University of BolognaBolognaItaly
| | - Marianna Penzo
- Department of ExperimentalDiagnostic and Specialty Medicine and Center for Applied Biomedical Research (CRBA)Alma Mater Studiorum‐University of BolognaBolognaItaly
| | - Maya Imbrechts
- Laboratory for Therapeutic and Diagnostic AntibodiesDepartment of Pharmaceutical and Pharmacological SciencesKU LeuvenLeuvenBelgium
| | - Nick Geukens
- Laboratory for Therapeutic and Diagnostic AntibodiesDepartment of Pharmaceutical and Pharmacological SciencesKU LeuvenLeuvenBelgium
- PharmAbsKU LeuvenLeuvenBelgium
| | | | | | - Daniele Pepe
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Kim Kampen
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
- Department of Radiotherapy, Maastricht Radiation Oncology (MAASTRO)Maastricht UniversityMaastrichtNetherlands
| | - Kim De Keersmaecker
- Laboratory for Disease Mechanisms in CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| |
Collapse
|
37
|
Noyes NC, Phan A, Davis RL. Memory suppressor genes: Modulating acquisition, consolidation, and forgetting. Neuron 2021; 109:3211-3227. [PMID: 34450024 PMCID: PMC8542634 DOI: 10.1016/j.neuron.2021.08.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/15/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023]
Abstract
The brain has a remarkable but underappreciated capacity to limit memory formation and expression. The term "memory suppressor gene" was coined in 1998 as an attempt to explain emerging reports that some genes appeared to limit memory. At that time, only a handful of memory suppressor genes were known, and they were understood to work by limiting cAMP-dependent consolidation. In the intervening decades, almost 100 memory suppressor genes with diverse functions have been discovered that affect not only consolidation but also acquisition and forgetting. Here we highlight the surprising extent to which biological limits are placed on memory formation through reviewing the literature on memory suppressor genes. In this review, we present memory suppressors within the framework of their actions on different memory operations: acquisition, consolidation, and forgetting. This is followed by a discussion of the reasons why there may be a biological need to limit memory formation.
Collapse
Affiliation(s)
- Nathaniel C Noyes
- Department of Neuroscience, Scripps Research Institute Florida, Jupiter, FL 33458, USA
| | - Anna Phan
- Department of Biological Sciences, University of Alberta, 11355 Saskatchewan Drive, Edmonton, AB T6G 2E9, Canada
| | - Ronald L Davis
- Department of Neuroscience, Scripps Research Institute Florida, Jupiter, FL 33458, USA.
| |
Collapse
|
38
|
Jennings MJ, Hathazi D, Nguyen CDL, Munro B, Münchberg U, Ahrends R, Schenck A, Eidhof I, Freier E, Synofzik M, Horvath R, Roos A. Intracellular Lipid Accumulation and Mitochondrial Dysfunction Accompanies Endoplasmic Reticulum Stress Caused by Loss of the Co-chaperone DNAJC3. Front Cell Dev Biol 2021; 9:710247. [PMID: 34692675 PMCID: PMC8526738 DOI: 10.3389/fcell.2021.710247] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 09/02/2021] [Indexed: 12/25/2022] Open
Abstract
Recessive mutations in DNAJC3, an endoplasmic reticulum (ER)-resident BiP co-chaperone, have been identified in patients with multisystemic neurodegeneration and diabetes mellitus. To further unravel these pathomechanisms, we employed a non-biased proteomic approach and identified dysregulation of several key cellular pathways, suggesting a pathophysiological interplay of perturbed lipid metabolism, mitochondrial bioenergetics, ER-Golgi function, and amyloid-beta processing. Further functional investigations in fibroblasts of patients with DNAJC3 mutations detected cellular accumulation of lipids and an increased sensitivity to cholesterol stress, which led to activation of the unfolded protein response (UPR), alterations of the ER-Golgi machinery, and a defect of amyloid precursor protein. In line with the results of previous studies, we describe here alterations in mitochondrial morphology and function, as a major contributor to the DNAJC3 pathophysiology. Hence, we propose that the loss of DNAJC3 affects lipid/cholesterol homeostasis, leading to UPR activation, β-amyloid accumulation, and impairment of mitochondrial oxidative phosphorylation.
Collapse
Affiliation(s)
- Matthew J. Jennings
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Denisa Hathazi
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Chi D. L. Nguyen
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Benjamin Munro
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Ute Münchberg
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Robert Ahrends
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Annette Schenck
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ilse Eidhof
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical Center, Nijmegen, Netherlands
| | - Erik Freier
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
| | - Matthis Synofzik
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
- German Centre for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Rita Horvath
- Department of Clinical Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Andreas Roos
- Leibniz-Institut für Analytische Wissenschaften – ISAS – e.V., Dortmund, Germany
- Department of Pediatric Neurology, Developmental Neurology and Social Pediatrics, Children’s Hospital University of Essen, Essen, Germany
| |
Collapse
|
39
|
Lyu X, Yang Q, Zhao F, Liu Y. Codon usage and protein length-dependent feedback from translation elongation regulates translation initiation and elongation speed. Nucleic Acids Res 2021; 49:9404-9423. [PMID: 34417614 PMCID: PMC8450115 DOI: 10.1093/nar/gkab729] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/26/2021] [Accepted: 08/17/2021] [Indexed: 12/26/2022] Open
Abstract
Essential cellular functions require efficient production of many large proteins but synthesis of large proteins encounters many obstacles in cells. Translational control is mostly known to be regulated at the initiation step. Whether translation elongation process can feedback to regulate initiation efficiency is unclear. Codon usage bias, a universal feature of all genomes, plays an important role in determining gene expression levels. Here, we discovered that there is a conserved but codon usage-dependent genome-wide negative correlation between protein abundance and CDS length. The codon usage effects on protein expression and ribosome flux on mRNAs are influenced by CDS length; optimal codon usage preferentially promotes production of large proteins. Translation of mRNAs with long CDS and non-optimal codon usage preferentially induces phosphorylation of initiation factor eIF2α, which inhibits translation initiation efficiency. Deletion of the eIF2α kinase CPC-3 (GCN2 homolog) in Neurospora preferentially up-regulates large proteins encoded by non-optimal codons. Surprisingly, CPC-3 also inhibits translation elongation rate in a codon usage and CDS length-dependent manner, resulting in slow elongation rates for long CDS mRNAs. Together, these results revealed a codon usage and CDS length-dependent feedback mechanism from translation elongation to regulate both translation initiation and elongation kinetics.
Collapse
Affiliation(s)
- Xueliang Lyu
- Department of Physiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA.,State Key Laboratory of Agricultural Microbiology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Qian Yang
- Department of Physiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Fangzhou Zhao
- Department of Physiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| | - Yi Liu
- Department of Physiology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX 75390, USA
| |
Collapse
|
40
|
The Type and Source of Reactive Oxygen Species Influences the Outcome of Oxidative Stress in Cultured Cells. Cells 2021; 10:cells10051075. [PMID: 33946545 PMCID: PMC8147180 DOI: 10.3390/cells10051075] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 01/21/2023] Open
Abstract
Oxidative stress can be modeled using various different experimental approaches, such as exposing the cells or organisms to oxidative chemicals. However, the actual effects of these chemicals, outside of the immediate measured effect, have attracted relatively little attention. We show here that three commonly used oxidants, menadione, potassium bromate, and hydrogen peroxide, while known to function differently, also elicit different types of responses in HEK293T cells. Menadione and bromate exposure mainly trigger an integrated stress response, whereas hydrogen peroxide affects cellular processes more diversely. Interestingly, acute oxidative stress does not universally cause notable induction of DNA repair or antioxidant defense mechanisms. We also provide evidence that cells with previous experience of oxidative stress show adaptive changes in their responses when the stress is renewed. Our results urge caution when comparing studies where different sources of oxidative stress have been used or when generalizing the findings of these studies to other oxidant types or tissues.
Collapse
|
41
|
ATP-Independent Initiation during Cap-Independent Translation of m 6A-Modified mRNA. Int J Mol Sci 2021; 22:ijms22073662. [PMID: 33915887 PMCID: PMC8036370 DOI: 10.3390/ijms22073662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 11/16/2022] Open
Abstract
The methylation of adenosine in the N6 position (m6A) is a widely used modification of eukaryotic mRNAs. Its importance for the regulation of mRNA translation was put forward recently, essentially due to the ability of methylated mRNA to be translated in conditions of inhibited cap-dependent translation initiation, e.g., under stress. However, the peculiarities of translation initiation on m6A-modified mRNAs are not fully known. In this study, we used toeprinting and translation in a cell-free system to confirm that m6A-modified mRNAs can be translated in conditions of suppressed cap-dependent translation. We show for the first time that m6A-modified mRNAs display not only decreased elongation, but also a lower efficiency of translation initiation. Additionally, we report relative resistance of m6A-mRNA translation initiation in the absence of ATP and inhibited eIF4A activity. Our novel findings indicate that the scanning of m6A-modified leader sequences is performed by a noncanonical mechanism.
Collapse
|
42
|
Chen L, Bai J, Peng D, Gao Y, Cai X, Zhang J, Tang S, Niu L, Sun Y, Lou F, Zhou H, Yin Q, Wang Z, Sun L, Du X, Xu Z, Wang H, Li Q, Wang H. SZB120 Exhibits Immunomodulatory Effects by Targeting eIF2α to Suppress Th17 Cell Differentiation. THE JOURNAL OF IMMUNOLOGY 2021; 206:953-962. [PMID: 33483349 DOI: 10.4049/jimmunol.2000036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 12/11/2020] [Indexed: 11/19/2022]
Abstract
IL-17-secreting Th17 cells play an important role in the pathogenesis of various inflammatory and autoimmune diseases. IL-17-targeted biologics and small molecules are becoming promising treatments for these diseases. In this study, we report that SZB120, a derivative of the natural compound 3-acetyl-β-boswellic acid, inhibits murine Th17 cell differentiation by interacting with the α-subunit of eukaryotic initiation factor 2 (eIF2α). We showed that SZB120 directly interacts with eIF2α and contributes to serine 51 phosphorylation of eIF2α. The suppressive effect of SZB120 on Th17 cell differentiation was reversed by GSK2606414, an inhibitor of eIF2α phosphokinase. Phosphorylation of eIF2α induced by SZB120 decreased the protein expression of IκBζ, which is important for Th17 cell differentiation. Notably, interaction with eIF2α by SZB120 also impaired glucose uptake and glycolysis in T cells. In vivo, SZB120 treatment of C57BL/6 mice significantly attenuated IL-17/Th17-mediated autoimmune disease. Our study indicates that SZB120 is a promising drug candidate for IL-17/Th17-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Linjiao Chen
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Bai
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Danhong Peng
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.,Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuanyuan Gao
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaojie Cai
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junxun Zhang
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sibei Tang
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Liman Niu
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yang Sun
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Institute of Translational Medicine, Shanghai Institute of Immunology Center for Microbiota and Immune Related Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Fangzhou Lou
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hong Zhou
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Institute of Translational Medicine, Shanghai Institute of Immunology Center for Microbiota and Immune Related Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Qianqian Yin
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhikai Wang
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Libo Sun
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xuemei Du
- State Key Laboratory of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Pharmaceutical Co., Ltd., Nanjing 210042, China; and
| | - Zhenyao Xu
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.,Institute of Translational Medicine, Shanghai Institute of Immunology Center for Microbiota and Immune Related Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hong Wang
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qun Li
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Honglin Wang
- Department of Immunology and Microbiology, Key Laboratory of Cell Differentiation and Apoptosis, Shanghai Institute of Immunology, Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; .,Institute of Translational Medicine, Shanghai Institute of Immunology Center for Microbiota and Immune Related Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
43
|
Rubio A, Ghosh S, Mülleder M, Ralser M, Mata J. Ribosome profiling reveals ribosome stalling on tryptophan codons and ribosome queuing upon oxidative stress in fission yeast. Nucleic Acids Res 2021; 49:383-399. [PMID: 33313903 PMCID: PMC7797079 DOI: 10.1093/nar/gkaa1180] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 11/13/2020] [Accepted: 11/19/2020] [Indexed: 12/11/2022] Open
Abstract
Translational control is essential in response to stress. We investigated the translational programmes launched by the fission yeast Schizosaccharomyces pombe upon five environmental stresses. We also explored the contribution of defence pathways to these programmes: The Integrated Stress Response (ISR), which regulates translation initiation, and the stress-response MAPK pathway. We performed ribosome profiling of cells subjected to each stress, in wild type cells and in cells with the defence pathways inactivated. The transcription factor Fil1, a functional homologue of the yeast Gcn4 and the mammalian Atf4 proteins, was translationally upregulated and required for the response to most stresses. Moreover, many mRNAs encoding proteins required for ribosome biogenesis were translationally downregulated. Thus, several stresses trigger a universal translational response, including reduced ribosome production and a Fil1-mediated transcriptional programme. Surprisingly, ribosomes stalled on tryptophan codons upon oxidative stress, likely due to a decrease in charged tRNA-Tryptophan. Stalling caused ribosome accumulation upstream of tryptophan codons (ribosome queuing/collisions), demonstrating that stalled ribosomes affect translation elongation by other ribosomes. Consistently, tryptophan codon stalling led to reduced translation elongation and contributed to the ISR-mediated inhibition of initiation. We show that different stresses elicit common and specific translational responses, revealing a novel role in Tryptophan-tRNA availability.
Collapse
Affiliation(s)
- Angela Rubio
- Department of Biochemistry, University of Cambridge, UK
| | - Sanjay Ghosh
- Department of Biochemistry, University of Cambridge, UK
| | - Michael Mülleder
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Markus Ralser
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK.,Department of Biochemistry, Charité University Medicine, Berlin, Germany
| | - Juan Mata
- Department of Biochemistry, University of Cambridge, UK
| |
Collapse
|
44
|
Abstract
RNA-binding proteins are important regulators of RNA metabolism and are of critical importance in all steps of the gene expression cascade. The role of aberrantly expressed RBPs in human disease is an exciting research field and the potential application of RBPs as a therapeutic target or a diagnostic marker represents a fast-growing area of research.Aberrant overexpression of the human RNA-binding protein La has been found in various cancer entities including lung, cervical, head and neck, and chronic myelogenous leukaemia. Cancer-associated La protein supports tumour-promoting processes such as proliferation, mobility, invasiveness and tumour growth. Moreover, the La protein maintains the survival of cancer cells by supporting an anti-apoptotic state that may cause resistance to chemotherapeutic therapy.The human La protein represents a multifunctional post-translationally modified RNA-binding protein with RNA chaperone activity that promotes processing of non-coding precursor RNAs but also stimulates the translation of selective messenger RNAs encoding tumour-promoting and anti-apoptotic factors. In our model, La facilitates the expression of those factors and helps cancer cells to cope with cellular stress. In contrast to oncogenes, able to initiate tumorigenesis, we postulate that the aberrantly elevated expression of the human La protein contributes to the non-oncogenic addiction of cancer cells. In this review, we summarize the current understanding about the implications of the RNA-binding protein La in cancer progression and therapeutic resistance. The concept of exploiting the RBP La as a cancer drug target will be discussed.
Collapse
Affiliation(s)
- Gunhild Sommer
- Department for Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Regensburg, Regensburg, Germany
| | - Tilman Heise
- Department for Pediatric Hematology, Oncology and Stem Cell Transplantation, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
45
|
Taylor J, Yeomans AM, Packham G. Targeted inhibition of mRNA translation initiation factors as a novel therapeutic strategy for mature B-cell neoplasms. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:3-25. [PMID: 32924027 PMCID: PMC7116065 DOI: 10.37349/etat.2020.00002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 01/31/2020] [Indexed: 12/17/2022] Open
Abstract
Cancer development is frequently associated with dysregulation of mRNA translation to enhance both increased global protein synthesis and translation of specific mRNAs encoding oncoproteins. Thus, targeted inhibition of mRNA translation is viewed as a promising new approach for cancer therapy. In this article we review current progress in investigating dysregulation of mRNA translation initiation in mature B-cell neoplasms, focusing on chronic lymphocytic leukemia, follicular lymphoma and diffuse large B-cell lymphoma. We discuss mechanisms and regulation of mRNA translation, potential pathways by which genetic alterations and the tumor microenvironment alters mRNA translation in malignant B cells, preclinical evaluation of drugs targeted against specific eukaryotic initiation factors and current progress towards clinical development. Overall, inhibition of mRNA translation initiation factors is an exciting and promising area for development of novel targeted anti-tumor drugs.
Collapse
Affiliation(s)
- Joe Taylor
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, United Kingdom
| | - Alison M Yeomans
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, United Kingdom
| | - Graham Packham
- Cancer Research UK Centre, Cancer Sciences, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, United Kingdom
| |
Collapse
|
46
|
Kim HJ. Cell Fate Control by Translation: mRNA Translation Initiation as a Therapeutic Target for Cancer Development and Stem Cell Fate Control. Biomolecules 2019; 9:biom9110665. [PMID: 31671902 PMCID: PMC6921038 DOI: 10.3390/biom9110665] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 12/11/2022] Open
Abstract
Translation of mRNA is an important process that controls cell behavior and gene regulation because proteins are the functional molecules that determine cell types and function. Cancer develops as a result of genetic mutations, which lead to the production of abnormal proteins and the dysregulation of translation, which in turn, leads to aberrant protein synthesis. In addition, the machinery that is involved in protein synthesis plays critical roles in stem cell fate determination. In the current review, recent advances in the understanding of translational control, especially translational initiation in cancer development and stem cell fate control, are described. Therapeutic targets of mRNA translation such as eIF4E, 4EBP, and eIF2, for cancer treatment or stem cell fate regulation are reviewed. Upstream signaling pathways that regulate and affect translation initiation were introduced. It is important to regulate the expression of protein for normal cell behavior and development. mRNA translation initiation is a key step to regulate protein synthesis, therefore, identifying and targeting molecules that are critical for protein synthesis is necessary and beneficial to develop cancer therapeutics and stem cells fate regulation.
Collapse
Affiliation(s)
- Hyun-Jung Kim
- Laboratory of Molecular Stem Cell Pharmacology, College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea.
| |
Collapse
|