1
|
Bianco E, Bonassera M, Uliana F, Tilma J, Winkler M, Zencir S, Gossert A, Oborská-Oplová M, Dechant R, Hugener J, Panse VG, Pilhofer M, Albert B, Kimmig P, Peter M. Stm1 regulates Ifh1 activity revealing crosstalk between ribosome biogenesis and ribosome dormancy. Mol Cell 2025; 85:1806-1823.e17. [PMID: 40315826 DOI: 10.1016/j.molcel.2025.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 01/26/2025] [Accepted: 04/04/2025] [Indexed: 05/04/2025]
Abstract
Nutrient abundance boosts ribosome biogenesis, whereas ribosome dormancy factors limit ribosome degradation upon starvation. The equilibrium between the two pathways governs cell growth. In this study, we identified suppressor of Tom1 (Stm1) as a molecular link between ribosome protection and biogenesis in Saccharomyces cerevisiae. While Stm1 was previously described as a dormancy factor, we show that it activates Ifh1, a transcriptional activator of ribosomal protein genes. Stm1 transiently localizes to the nucleolus, where it interacts with pre-ribosomes and directly binds RNA and Ifh1 through its C-terminal intrinsically disordered region (IDR). Although the IDR is dispensable for ribosome protection, its loss compromises cell growth. The IDR is phosphorylated upon nutrient starvation, which disrupts its interaction with Ifh1. Our findings reveal a molecular pathway sensing and adjusting ribosome abundance in response to nutrient availability, reinforcing the relevance of regulated ribosome homeostasis in physiology and disease.
Collapse
Affiliation(s)
- Eliana Bianco
- Department of Biology, Institute of Biochemistry, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland.
| | - Martina Bonassera
- Department of Biology, Institute of Biochemistry, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland; Department of Biology, Institute of Molecular Biology and Biophysics, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland
| | - Federico Uliana
- Department of Biology, Institute of Biochemistry, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland
| | - Janny Tilma
- Department of Biology, Institute of Biochemistry, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland
| | - Martin Winkler
- Department of Biology, Institute of Biochemistry, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland
| | - Sevil Zencir
- Department of Cell Biology Sciences III, Université de Genève, 1211 Geneva, Switzerland
| | - Alvar Gossert
- Department of Biology, Institute of Biochemistry, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland; Biomolecular NMR Spectroscopy Platform (BNSP), Department of Biology, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland
| | | | - Reinhard Dechant
- Department of Biology, Institute of Biochemistry, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland
| | - Jannik Hugener
- Department of Biology, Institute of Molecular Biology and Biophysics, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland
| | - Vikram Govind Panse
- Institute of Medical Microbiology, University of Zürich, 8006 Zürich, Switzerland; Faculty of Science, University of Zürich, Zürich, Switzerland
| | - Martin Pilhofer
- Department of Biology, Institute of Molecular Biology and Biophysics, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland
| | - Benjamin Albert
- Molecular, Cellular and Developmental Biology Unit (MCD), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31062 Toulouse, France
| | - Philipp Kimmig
- Department of Biology, Institute of Biochemistry, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland
| | - Matthias Peter
- Department of Biology, Institute of Biochemistry, Eidgenössische Technische Hochschule (ETH), 8093 Zürich, Switzerland.
| |
Collapse
|
2
|
Salmazo MIDBF, Alonso JCC, de Arruda Camargo GC, de Oliveira G, da Silva Santos A, Ávila M, Roberto IM, de Freitas LLL, Bottene MC, Lestingi JFP, Caria PHF, Durán N, Kobarg J, Fávaro WJ. Clinical and immunohistochemical effects of OncoTherad (MRB-CFI-1) nanoimmunotherapy on SERBP1, HABP4, CD44 and Ki-67 in BCG-unresponsive non-muscle invasive bladder cancer. Tissue Cell 2025; 93:102783. [PMID: 39938427 DOI: 10.1016/j.tice.2025.102783] [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: 10/18/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/14/2025]
Abstract
Non-muscle-invasive bladder cancer (NMIBC) is a malignancy with a high recurrence and progression rate, particularly in patients who fail to respond to standard Bacillus Calmette-Guérin (BCG) therapy. OncoTherad (MRB-CFI-1) nanoimmunotherapy has emerged as a promising therapeutic option, with potential to modulate immune responses and inhibit tumor progression. This study evaluated the clinical efficacy of OncoTherad (MRB-CFI-1) nanoimmunotherapy in patients with BCG-unresponsive NMIBC and investigated correlations between therapeutic outcomes and histopathological and molecular findings. In this retrospective cross-sectional study, 20 patients with BCG-unresponsive NMIBC were treated with OncoTherad (MRB-CFI-1) across two clinical centers. Bladder tissue samples were collected before and after treatment, and immunohistochemical analyses were performed to assess the expression of SERBP1, HABP4, CD44, and Ki-67. Primary endpoints included pathological complete response (pCR), recurrence-free survival (RFS), and duration of response (DoR), which were analyzed in relation to immunohistochemical biomarker findings. Our results demonstrated that high Ki-67 proliferative index and elevated immunoreactivity for CD44 and SERBP1 were associated with shorter RFS. Treatment with OncoTherad (MRB-CFI-1) significantly reduced (p < 0.05) the immunoreactivity of SERBP1 and CD44, which was accompanied by a marked decrease in Ki-67 proliferative index, indicating effective suppression of tumor activity. Conversely, a significant increase (p < 0.05) in HABP4 immunoreactivity was observed, suggesting a protective role against NMIBC recurrence and progression. A pCR was achieved in 65 % of patients, with a median RFS of 21.1 months and a median DoR of 15.7 months, underscoring the clinical efficacy of OncoTherad (MRB-CFI-1). These findings suggest that OncoTherad (MRB-CFI-1) nanoimmunotherapy offers a novel and effective treatment strategy for patients with BCG-unresponsive NMIBC, providing a promising alternative to radical cystectomy and significantly improving patient outcomes.
Collapse
Affiliation(s)
- Maria Izabel de Barros Frazão Salmazo
- Laboratory of Urogenital Carcinogenesis and Immunotherapy (LCURGIN), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil
| | - João Carlos Cardoso Alonso
- Laboratory of Urogenital Carcinogenesis and Immunotherapy (LCURGIN), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil; Paulínia Municipal Hospital, Paulínia City, São Paulo State, Brazil
| | - Gabriela Cardoso de Arruda Camargo
- Laboratory of Urogenital Carcinogenesis and Immunotherapy (LCURGIN), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil
| | - Gabriela de Oliveira
- Laboratory of Urogenital Carcinogenesis and Immunotherapy (LCURGIN), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil
| | - André da Silva Santos
- Laboratory of Urogenital Carcinogenesis and Immunotherapy (LCURGIN), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil
| | - Monaliza Ávila
- Laboratory of Urogenital Carcinogenesis and Immunotherapy (LCURGIN), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil
| | - Isadora Manzato Roberto
- Laboratory of Urogenital Carcinogenesis and Immunotherapy (LCURGIN), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil
| | - Leandro Luiz Lopes de Freitas
- Pathology Department, Medical School, Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil
| | | | - Jean Felipe Prodocimo Lestingi
- São Vicente de Paulo Charity Hospital, Jundiaí City, São Paulo State, Brazil; Division of Urology, University of Sao Paulo Medical School, São Paulo City, São Paulo State, Brazil
| | - Paulo Henrique Ferreira Caria
- Laboratory of Urogenital Carcinogenesis and Immunotherapy (LCURGIN), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil
| | - Nelson Durán
- Laboratory of Urogenital Carcinogenesis and Immunotherapy (LCURGIN), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil
| | - Jörg Kobarg
- Laboratory of Signal Mechanisms, School of Pharmaceutical Sciences (FCF), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil
| | - Wagner José Fávaro
- Laboratory of Urogenital Carcinogenesis and Immunotherapy (LCURGIN), Universidade Estadual de Campinas (UNICAMP), Campinas City, São Paulo State, Brazil.
| |
Collapse
|
3
|
Hao T, Li Y, Ren Q, Zeng Y, Gao L, Zhu W, Liang J, Lin Y, Hu J, Yan G, Sun S, Cai J. circ-1584 selectively promotes the antitumor activity of the oncolytic virus M1 on pancreatic cancer. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200919. [PMID: 39866243 PMCID: PMC11760297 DOI: 10.1016/j.omton.2024.200919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/25/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025]
Abstract
Pancreatic cancer is among the most challenging tumors to treat, and due to its immune tolerance characteristics, existing immunotherapy methods are not effective in alleviating the disease. Oncolytic virus therapy, a potential new strategy for treating pancreatic cancer, also faces the limitation of being ineffective when used alone. Elucidating the key host endogenous circular RNAs (circRNAs) involved in M1 virus-mediated killing of pancreatic ductal adenocarcinoma (PDAC) cells may help overcome this limitation. Here, we report that the oncolytic virus M1, a nonpathogenic alphavirus, exhibits different cell viability-inhibitory effects on different pancreatic cancer cells in the clinical stage. Through high-throughput circRNA sequencing, we found that circRNA expression varies among these cells. Further gain-of-function and loss-of-function experiments have shown that circ-1584 can selectively enhance the anti-pancreatic cancer effects of the M1 virus in vitro and in vivo. Additionally, circ-1584 may negatively regulate miR-578 to modulate the anti-pancreatic cancer effects of the M1 virus. Our findings lay the foundation for using circRNA as an adjuvant to enhance the M1 virus efficacy against pancreatic cancer.
Collapse
Affiliation(s)
- Taofang Hao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuanyuan Li
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Qianyao Ren
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ying Zeng
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Leyi Gao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wenbo Zhu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jiankai Liang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yuan Lin
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, The First Affiliated Hospital-Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Human Microbiome and Elderly Chronic Diseases, Ministry of Education, Beijing, China
| | - Jun Hu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shuxin Sun
- Pancreatic Center, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Jing Cai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Molecular Biology and Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
4
|
Bhandare P, Narain A, Hofstetter J, Rummel T, Wenzel J, Schülein-Völk C, Lamer S, Eilers U, Schlosser A, Eilers M, Erhard F, Wolf E. Phenotypic screens identify SCAF1 as critical activator of RNAPII elongation and global transcription. Nucleic Acids Res 2025; 53:gkae1219. [PMID: 39698826 PMCID: PMC11879057 DOI: 10.1093/nar/gkae1219] [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: 08/01/2024] [Revised: 10/30/2024] [Accepted: 12/03/2024] [Indexed: 12/20/2024] Open
Abstract
Transcripts produced by RNA polymerase II (RNAPII) are fundamental for cellular responses to environmental changes. It is therefore no surprise that there exist multiple avenues for the regulation of this process. To explore the regulation mediated by RNAPII-interacting proteins, we used a small interfering RNA (siRNA)-based screen to systematically evaluate their influence on RNA synthesis. We identified several proteins that strongly affected RNAPII activity. We evaluated one of the top hits, SCAF1 (SR-related C-terminal domain-associated factor 1), using an auxin-inducible degradation system and sequencing approaches. In agreement with our screen results, acute depletion of SCAF1 decreased RNA synthesis, and showed an increase of Serine-2 phosphorylated-RNAPII (pS2-RNAPII). We found that the accumulation of pS2-RNAPII within the gene body occurred at GC-rich regions and was indicative of stalled RNAPII complexes. The accumulation of stalled RNAPII complexes was accompanied by reduced recruitment of initiating RNAPII, explaining the observed global decrease in transcriptional output. Furthermore, upon SCAF1 depletion, RNAPII complexes showed increased association with components of the proteasomal-degradation machinery. We concluded that in cells lacking SCAF1, RNAPII undergoes a rather interrupted passage, resulting in intervention by the proteasomal-degradation machinery to clear stalled RNAPII. While cells survive the compromised transcription caused by absence of SCAF1, further inhibition of proteasomal-degradation machinery is synthetically lethal.
Collapse
Affiliation(s)
- Pranjali Bhandare
- Institute of Biochemistry, University of Kiel, Rudolf-Höber-Straße 1, Kiel 24118, Germany
- Cancer Systems Biology Group, Theodor Boveri Institute, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Ashwin Narain
- Cancer Systems Biology Group, Theodor Boveri Institute, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Julia Hofstetter
- Cancer Systems Biology Group, Theodor Boveri Institute, University of Würzburg, Am Hubland, Würzburg 97074, Germany
- Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Teresa Rummel
- Faculty for Informatics and Data Science, University of Regensburg, Bajuwarenstraße 4, Regensburg 93040, Germany
| | - Julia Wenzel
- Institute of Biochemistry, University of Kiel, Rudolf-Höber-Straße 1, Kiel 24118, Germany
| | - Christina Schülein-Völk
- Core Unit High-Content Microscopy, Biocenter, Theodor Boveri Institute, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Stephanie Lamer
- Rudolf-Virchow-Zentrum - Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Straße 2, Würzburg 97080, Germany
| | - Ursula Eilers
- Core Unit High-Content Microscopy, Biocenter, Theodor Boveri Institute, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Andreas Schlosser
- Rudolf-Virchow-Zentrum - Center for Integrative and Translational Bioimaging, University of Würzburg, Josef-Schneider-Straße 2, Würzburg 97080, Germany
| | - Martin Eilers
- Chair of Biochemistry and Molecular Biology, Theodor Boveri Institute, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| | - Florian Erhard
- Faculty for Informatics and Data Science, University of Regensburg, Bajuwarenstraße 4, Regensburg 93040, Germany
| | - Elmar Wolf
- Institute of Biochemistry, University of Kiel, Rudolf-Höber-Straße 1, Kiel 24118, Germany
- Cancer Systems Biology Group, Theodor Boveri Institute, University of Würzburg, Am Hubland, Würzburg 97074, Germany
| |
Collapse
|
5
|
Cui N, Han X, Yang X, Zhao X, Huang Q, Xu C, Su S. Avian leukosis virus usurps the cellular SERBP1 protein to enhance its transcription and promote productive infections in avian cells. Poult Sci 2024; 103:103755. [PMID: 38663206 PMCID: PMC11068620 DOI: 10.1016/j.psj.2024.103755] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/02/2024] [Accepted: 04/06/2024] [Indexed: 05/07/2024] Open
Abstract
Avian leukosis virus subgroup K (ALV-K) is composed of newly emerging isolates, which cluster separately from the well-characterized subgroups A, B, C, D, E, and J in sequence analysis, and exhibits a specific host range and a unique pattern of superinfection interference. Avian leukosis virus subgroup K replicate more slowly in avian cells than other ALV strains, leading to escaped detection during ALV eradication, but the underlying mechanism are largely unknown. In our previous study, we have reported that JS11C1 and most of other suspected ALV-K strains possessed unique mutations in the U3 region. Here, we selected 5 mutations in some important transcriptional regulation elements to explore the possible factor contributing for the lower activity of LTR, including CA-TG mutation in the CAAT box, 21 nt deletion in the CAAT box, A-G and A-T mutations in the CArG boxes, 11 nt insertion in the PRE boxes, and C-T mutation in the TATA box. On the basis of infectious clone of JS11C1, we demonstrated that the 11 nt fragment in the PRE boxes was associated with the transcription activity of LTR, the enhancer ability of U3, and the replication capacity of the virus. Notably, we determined the differential U3-protein interaction profile of ALVs and found that the 11 nt fragment specifically binds to cellular SERPINE1 mRNA binding protein 1 (SERBP1) to increase the LTR activity and enhance virus replication. Collectively, these findings reveal that a 11 nt fragment in the U3 gene contributed to its binding ability to the cellular SERBP1 to enhance its transcription and the infectious virus productions in avian cells. This study highlighted the vital role of host factor in retrovirus replication and thus provides a new perspective to elucidate the interaction between retrovirus and its host and a molecular basis to develop efficient strategies against retroviruses.
Collapse
Affiliation(s)
- Ning Cui
- Shandong Key Laboratory of Animal Disease Control and Breeding; Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, Jinan, China
| | - Xiaoxia Han
- Shandong Key Laboratory of Animal Disease Control and Breeding; Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, Jinan, China; College of Life Sciences, Shandong Normal University, Jinan, China
| | - Xiao Yang
- Shandong Key Laboratory of Animal Disease Control and Breeding; Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, Jinan, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention; College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Xiaoran Zhao
- Shandong Key Laboratory of Animal Disease Control and Breeding; Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, Jinan, China; Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention; College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Qinghua Huang
- Shandong Key Laboratory of Animal Disease Control and Breeding; Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, Jinan, China
| | - Chuantian Xu
- Shandong Key Laboratory of Animal Disease Control and Breeding; Institute of Animal Science and Veterinary Medicine, Shandong Academy of Agricultural Sciences, Jinan, China; Key Laboratory of Livestock and Poultry Multi-omics of MARA, Jinan, China.
| | - Shuai Su
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention; College of Veterinary Medicine, Shandong Agricultural University, Tai'an, China.
| |
Collapse
|
6
|
Bleckmann A, Spitzlberger N, Denninger P, Ehrnsberger HF, Wang L, Bruckmann A, Reich S, Holzinger P, Medenbach J, Grasser KD, Dresselhaus T. Cytosolic RGG RNA-binding proteins are temperature sensitive flowering time regulators in Arabidopsis. Biol Chem 2023; 404:1069-1084. [PMID: 37674329 DOI: 10.1515/hsz-2023-0171] [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: 04/03/2023] [Accepted: 08/03/2023] [Indexed: 09/08/2023]
Abstract
mRNA translation is tightly regulated by various classes of RNA-binding proteins (RBPs) during development and in response to changing environmental conditions. In this study, we characterize the arginine-glycine-glycine (RGG) motif containing RBP family of Arabidopsis thaliana representing homologues of the multifunctional translation regulators and ribosomal preservation factors Stm1 from yeast (ScStm1) and human SERBP1 (HsSERBP1). The Arabidopsis genome encodes three RGG proteins named AtRGGA, AtRGGB and AtRGGC. While AtRGGA is ubiquitously expressed, AtRGGB and AtRGGC are enriched in dividing cells. All AtRGGs localize almost exclusively to the cytoplasm and bind with high affinity to ssRNA, while being capable to interact with most nucleic acids, except dsRNA. A protein-interactome study shows that AtRGGs interact with ribosomal proteins and proteins involved in RNA processing and transport. In contrast to ScStm1, AtRGGs are enriched in ribosome-free fractions in polysome profiles, suggesting additional plant-specific functions. Mutant studies show that AtRGG proteins differentially regulate flowering time, with a distinct and complex temperature dependency for each AtRGG protein. In conclusion, we suggest that AtRGGs function in fine-tuning translation efficiency to control flowering time and potentially other developmental processes in response to environmental changes.
Collapse
Affiliation(s)
- Andrea Bleckmann
- Cell Biology and Plant Biochemistry, University of Regensburg, D-93053 Regensburg, Germany
| | - Nicole Spitzlberger
- Cell Biology and Plant Biochemistry, University of Regensburg, D-93053 Regensburg, Germany
| | - Philipp Denninger
- Cell Biology and Plant Biochemistry, University of Regensburg, D-93053 Regensburg, Germany
| | - Hans F Ehrnsberger
- Cell Biology and Plant Biochemistry, University of Regensburg, D-93053 Regensburg, Germany
| | - Lele Wang
- Cell Biology and Plant Biochemistry, University of Regensburg, D-93053 Regensburg, Germany
| | - Astrid Bruckmann
- Biochemistry I, University of Regensburg, D-93053 Regensburg, Germany
| | - Stefan Reich
- Biochemistry I, University of Regensburg, D-93053 Regensburg, Germany
| | - Philipp Holzinger
- Cell Biology and Plant Biochemistry, University of Regensburg, D-93053 Regensburg, Germany
| | - Jan Medenbach
- Biochemistry I, University of Regensburg, D-93053 Regensburg, Germany
| | - Klaus D Grasser
- Cell Biology and Plant Biochemistry, University of Regensburg, D-93053 Regensburg, Germany
| | - Thomas Dresselhaus
- Cell Biology and Plant Biochemistry, University of Regensburg, D-93053 Regensburg, Germany
| |
Collapse
|
7
|
Wang F, Wang L, Gan S, Feng S, Ouyang S, Wang X, Yuan S. SERBP1 Promotes Stress Granule Clearance by Regulating 26S Proteasome Activity and G3BP1 Ubiquitination and Protects Male Germ Cells from Thermostimuli Damage. RESEARCH (WASHINGTON, D.C.) 2023; 6:0091. [PMID: 37223481 PMCID: PMC10202183 DOI: 10.34133/research.0091] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/15/2023] [Indexed: 04/18/2024]
Abstract
Stress granules (SGs) are membraneless cytoplasmic condensates that dynamically assemble in response to various stressors and reversibly disassemble after stimulus removal; however, the mechanisms underlying SG dynamics and their physiological roles in germ cell development are elusive. Here, we show that SERBP1 (SERPINE1 mRNA binding protein 1) is a universal SG component and conserved regulator of SG clearance in somatic and male germ cells. SERBP1 interacts with the SG core component G3BP1 and 26S proteasome proteins PSMD10 and PSMA3 and recruits them to SGs. In the absence of SERBP1, reduced 20S proteasome activity, mislocalized valosin containing protein (VCP) and Fas associated factor family member 2 (FAF2), and diminished K63-linked polyubiquitination of G3BP1 during the SG recovery period were observed. Interestingly, the depletion of SERBP1 in testicular cells in vivo causes increased germ cell apoptosis upon scrotal heat stress. Accordingly, we propose that a SERBP1-mediated mechanism regulates 26S proteasome activity and G3BP1 ubiquitination to facilitate SG clearance in both somatic and germ cell lines.
Collapse
Affiliation(s)
- Fengli Wang
- Institute of Reproductive Health, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lingjuan Wang
- Institute of Reproductive Health, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shiming Gan
- Institute of Reproductive Health, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shenglei Feng
- Institute of Reproductive Health, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430030, China
| | - Sijin Ouyang
- Institute of Reproductive Health, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoli Wang
- Institute of Reproductive Health, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430030, China
- Laboratory of Animal Center,
Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
8
|
Barbato C, Frisone P, Braccini L, D’Aguanno S, Pieroni L, Ciotti MT, Catalanotto C, Cogoni C, Ruberti F. Silencing of Ago-2 Interacting Protein SERBP1 Relieves KCC2 Repression by miR-92 in Neurons. Cells 2022; 11:1052. [PMID: 35326503 PMCID: PMC8947033 DOI: 10.3390/cells11061052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/11/2022] [Accepted: 03/18/2022] [Indexed: 12/13/2022] Open
Abstract
RNA-binding proteins (RBPs) play important roles in modulating miRNA-mediated mRNA target repression. Argonaute2 (Ago2) is an essential component of the RNA-induced silencing complex (RISC) that plays a central role in silencing mechanisms via small non-coding RNA molecules known as siRNAs and miRNAs. Small RNAs loaded into Argonaute proteins catalyze endoribonucleolytic cleavage of target RNAs or recruit factors responsible for translational silencing and mRNA target destabilization. In previous studies we have shown that KCC2, a neuronal Cl (-) extruding K (+) Cl (-) co-transporter 2, is regulated by miR-92 in neuronal cells. Searching for Ago2 partners by immunoprecipitation and LC-MS/MS analysis, we isolated among other proteins the Serpine mRNA binding protein 1 (SERBP1) from SH-SY5Y neuroblastoma cells. Exploring the role of SERBP1 in miRNA-mediated gene silencing in SH-SY5Y cells and primary hippocampal neurons, we demonstrated that SERBP1 silencing regulates KCC2 expression through the 3' untranslated region (UTR). In addition, we found that SERBP1 as well as Ago2/miR-92 complex bind to KCC2 3'UTR. Finally, we demonstrated the attenuation of miR-92-mediated repression of KCC2 3'UTR by SERBP1 silencing. These findings advance our knowledge regarding the miR-92-mediated modulation of KCC2 translation in neuronal cells and highlight SERBP1 as a key component of this gene regulation.
Collapse
Affiliation(s)
- Christian Barbato
- Institute of Biochemistry and Cell Biology, National Research Council CNR, Department of Sense Organs, University of Rome Sapienza, 00161 Roma, Italy
| | - Paola Frisone
- Institute of Biochemistry and Cell Biology CNR, Campus A. Buzzati-Traverso, 00015 Monterotondo (RM), Italy; (P.F.); (M.T.C.)
- Department of Molecular Medicine, University of Rome Sapienza, 00161 Roma, Italy; (L.B.); (C.C.)
| | - Laura Braccini
- Department of Molecular Medicine, University of Rome Sapienza, 00161 Roma, Italy; (L.B.); (C.C.)
| | - Simona D’Aguanno
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Luisa Pieroni
- Department of Experimental Neuroscience, Proteomics and Metabolomics Unit, IRCCS-Fondazione Santa Lucia, 00143 Rome, Italy;
| | - Maria Teresa Ciotti
- Institute of Biochemistry and Cell Biology CNR, Campus A. Buzzati-Traverso, 00015 Monterotondo (RM), Italy; (P.F.); (M.T.C.)
| | - Caterina Catalanotto
- Department of Molecular Medicine, University of Rome Sapienza, 00161 Roma, Italy; (L.B.); (C.C.)
| | - Carlo Cogoni
- Department of Molecular Medicine, University of Rome Sapienza, 00161 Roma, Italy; (L.B.); (C.C.)
| | - Francesca Ruberti
- Institute of Biochemistry and Cell Biology CNR, Campus A. Buzzati-Traverso, 00015 Monterotondo (RM), Italy; (P.F.); (M.T.C.)
| |
Collapse
|
9
|
Baudin A, Xu X, Libich DS. The 1H, 15N and 13C resonance assignments of the C-terminal domain of Serpine mRNA binding protein 1 (SERBP1). BIOMOLECULAR NMR ASSIGNMENTS 2021; 15:461-466. [PMID: 34436734 PMCID: PMC9990118 DOI: 10.1007/s12104-021-10046-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/19/2021] [Indexed: 06/13/2023]
Abstract
SERBP1 is a multifunctional mRNA-binding protein that has been shown to play a regulatory role in a number of biological processes such as thrombosis, DNA damage repair, and the cellular response to nutrient deprivation. Additionally, SERBP1 is upregulated in glioblastoma, leukemia as well as liver, prostrate and ovarian cancers where it has been implicated in metastatic disease and poor patient outcomes. SERBP1 binds target mRNA, stabilizing and regulating the post-translational expression of the transcript. Since SERBP1 lacks canonical RNA-binding motifs such as RRM domains or zinc fingers, its target recognition and binding mechanisms are not well understood. Recent reports suggest that it is capable of recognizing both RNA sequence motifs and structured domains. Here we report the production and purification of the intrinsically disordered C-terminal domain of SERBP1, the assignment of the 1H, 13C, 15N backbone resonances of the protein by solution-state NMR, and secondary structure predictions. We show that the protein is not entirely disordered and identify an α-helix that was stable under the experimental conditions. This work is the first step toward understanding the structural basis underpinning the molecular mechanisms of SERBP1 functions, particularly interactions with mRNA targets.
Collapse
Affiliation(s)
- Antoine Baudin
- Department of Biochemistry and Structural Biology, and Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, 8403 Floyd Curl Dr., San Antonio, TX, 78229, USA
| | - Xiaoping Xu
- Department of Biochemistry and Structural Biology, and Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, 8403 Floyd Curl Dr., San Antonio, TX, 78229, USA
| | - David S Libich
- Department of Biochemistry and Structural Biology, and Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, 8403 Floyd Curl Dr., San Antonio, TX, 78229, USA.
| |
Collapse
|
10
|
Liu Y, Hu X, Hu L, Xu C, Liang X. Let-7i-5p enhances cell proliferation, migration and invasion of ccRCC by targeting HABP4. BMC Urol 2021; 21:49. [PMID: 33775245 PMCID: PMC8005230 DOI: 10.1186/s12894-021-00820-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/19/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is one of the best-characterized and most pervasive renal cancers. The present study aimed to explore the effects and potential mechanisms of let-7i-5p in ccRCC cells. METHODS Using bioinformatics analyses, we investigated the expression of let-7i-5p in The Cancer Genome Atlas (TCGA) database and predicted biological functions and possible target genes of let-7i-5p in ccRCC cells. Cell proliferation assay, wound healing assay and transwell invasion assay were conducted to characterize the effects of let-7i-5p in ccRCC cells. To verify the interactions between let-7i-5p and HABP4, dual-luciferase reporter assay, quantitative real-time polymerase chain reaction, and western blotting were conducted. Rescue experiments were used to investigate the relationship between let-7i-5p and HABP4. RESULTS TCGA data analysis revealed that ccRCC tissues had significantly increased let-7i-5p expression, which was robustly associated with poor overall survival. Further verification showed that ccRCC cell proliferation, migration and invasion were inhibited by let-7i-5p inhibitor but enhanced by let-7i-5p mimics. Subsequently, HABP4 was predicted to be the target gene of let-7i-5p. TCGA data showed that ccRCC tissues had decreased expression of HABP4 and that HABP4 expression was negatively correlated with let-7i-5p. Further verification showed that downregulation of HABP4 expression promoted cell proliferation, migration and invasion. The dual-luciferase reporter gene assay suggested that the let-7i-5p/HABP4 axis was responsible for the development of ccRCC. CONCLUSION Our results provide evidence that let-7i-5p functions as a tumor promoter in ccRCC and facilitates cell proliferation, migration and invasion by targeting HABP4. These results clarify the pathogenesis of ccRCC and offer a potential target for its treatment.
Collapse
Affiliation(s)
- Yujie Liu
- Department of Geriatric Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xing Hu
- Department of General Practice, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Liang Hu
- Department of Cardiovascular Surgery, The General Hospital of Western Theater Command, Chengdu, 610083, China
| | - Changjing Xu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xuemei Liang
- Department of Geriatric Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
11
|
Melo-Hanchuk TD, Colleti C, Saito Â, Mendes MCS, Carvalheira JBC, Vassallo J, Kobarg J. Intracellular hyaluronic acid-binding protein 4 (HABP4): a candidate tumor suppressor in colorectal cancer. Oncotarget 2020; 11:4325-4337. [PMID: 33245729 PMCID: PMC7679031 DOI: 10.18632/oncotarget.27804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Hyaluronic Acid-binding protein 4 (HABP4) is a regulatory protein of 57 kDa that is functionally involved in transcription regulation and RNA metabolism and shows several characteristics common to oncoproteins or tumor suppressors, including altered expression in cancer tissues, nucleus/cytoplasm shuttling, intrinsic lack of protein structure, complex interactomes and post translational modifications. Its gene has been found in a region on chromosome 9q22.3-31, which contains SNP haplotypes occurring in individuals with a high risk for familial colon cancer. To test a possible role of HABP4 in tumorigenesis we generated knockout mice by the CRISPR/Cas9 method and treated the animals with azoxymethane (AOM)/dextran sodium sulfate (DSS) for induction of colon tumors. HABP4-/- mice, compared to wild type mice, had more and larger tumors, and expressed more of the proliferation marker proteins Cyclin-D1, CDK4 and PCNA. Furthermore, the cells of the bottom of the colon crypts in the HABP4-/- mice divided more rapidly. Next, we generated also HABP4-/- HCT 116 cells, in cell culture and found again an increased proliferation in clonogenic assays in comparison to wild-type cells. Our study of the protein expression levels of HABP4 in human colon cancer samples, through immunohistochemistry assays, showed, that 30% of the tumors analyzed had low expression of HABP4. Our data suggest that HABP4 is involved in proliferation regulation of colon cells in vitro and in vivo and that it is a promising new candidate for a tumor suppressor protein that can be explored both in the diagnosis and possibly therapy of colon cancer.
Collapse
Affiliation(s)
- Talita Diniz Melo-Hanchuk
- Department of Biochemistry and Tissue Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- These authors contributed equally to this work
| | - Carolina Colleti
- School of Pharmaceutical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- These authors contributed equally to this work
| | - Ângela Saito
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, SP, Brazil
- These authors contributed equally to this work
| | - Maria Carolina Santos Mendes
- Division of Oncology, Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - José Barreto Campello Carvalheira
- Division of Oncology, Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Jose Vassallo
- Laboratory of Investigative Pathology, CIPED, Faculty of Medical Sciences, State University of Campinas, Campinas, SP, Brazil
| | - Jörg Kobarg
- School of Pharmaceutical Sciences, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|