1
|
Suloh H, Ojha SK, Kartawy M, Hamoudi W, Tripathi MK, Bazbaz W, Schottlender N, Ashery U, Khaliulin I, Amal H. Shared early molecular mechanisms revealed in P301S and 5xFAD Alzheimer's disease mouse models. Transl Psychiatry 2025; 15:97. [PMID: 40140365 PMCID: PMC11947184 DOI: 10.1038/s41398-025-03321-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 02/21/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder characterized by early molecular events that influence disease progression. Still, the molecular mechanisms caused by different mutations of AD are not understood. We have performed a multidisciplinary study to investigate and compare the early stages of the pathology in two transgenic AD mouse models: P301S and 5xFAD. Using SNOTRAP-based mass spectrometry, we assessed changes in S-nitrosylation, a nitric oxide-mediated post-translational modification, of proteins in both models during their juvenile age. The increased levels of 3-nitrotyrosine confirmed nitrosative stress in the mutant mice. Systems biology analysis revealed shared processes between the models, particularly in the γ-aminobutyric acid (GABA)ergic and glutamatergic neurotransmission processes. In the P301S model, we identified 273 S-nitrosylated (SNOed) proteins in the cortex, with 244 proteins uniquely SNOed in the diseased mice. In the 5xFAD model, 309 SNOed proteins were identified. We have found altered proteins expression of different glutamate/GABA-related markers in the cortex and hippocampus of both AD mouse models. Additionally, the phosphorylation levels of the mTOR signaling components revealed hyperactivation of this pathway in P301S mice. Conversely, 5xFAD mice showed no significant changes in mTOR signaling except for elevated phosphorylation of the ribosomal protein S6 in the cortex. Our findings revealed key molecular mechanisms in the two AD mouse models during their early stages. These mechanisms could serve as potential biomarkers and therapeutic targets for early-stage AD.
Collapse
Affiliation(s)
- Huda Suloh
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Shashank Kumar Ojha
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maryam Kartawy
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Manish Kumar Tripathi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wisam Bazbaz
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nofar Schottlender
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Uri Ashery
- School of Neurobiology, Biochemistry and Biophysics, Life Sciences Faculty, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Rosamund Stone Zander Translational Neuroscience Center, Boston Children's Hospital, Boston, Massachusetts; Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Tapken I, Schweitzer T, Paganin M, Schüning T, Detering NT, Sharma G, Niesert M, Saffari A, Kuhn D, Glynn A, Cieri F, Santonicola P, Cannet C, Gerstner F, Faller KME, Huang YT, Kothary R, Gillingwater TH, Di Schiavi E, Simon CM, Hensel N, Ziegler A, Viero G, Pich A, Claus P. The systemic complexity of a monogenic disease: the molecular network of spinal muscular atrophy. Brain 2025; 148:580-596. [PMID: 39183150 DOI: 10.1093/brain/awae272] [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: 03/07/2024] [Revised: 06/20/2024] [Accepted: 07/19/2024] [Indexed: 08/27/2024] Open
Abstract
Monogenic diseases are well-suited paradigms for the causal analysis of disease-driving molecular patterns. Spinal muscular atrophy (SMA) is one such monogenic model, caused by mutation or deletion of the survival of motor neuron 1 (SMN1) gene. Although several functions of the SMN protein have been studied, single functions and pathways alone do not allow the identification of crucial disease-driving molecules. Here, we analysed the systemic characteristics of SMA, using proteomics, phosphoproteomics, translatomics and interactomics, from two mouse models with different disease severities and genetics. This systems approach revealed subnetworks and proteins characterizing commonalities and differences of both models. To link the identified molecular networks with the disease-causing SMN protein, we combined SMN-interactome data with both proteomes, creating a comprehensive representation of SMA. By this approach, disease hubs and bottlenecks between SMN and downstream pathways could be identified. Linking a disease-causing molecule with widespread molecular dysregulations via multiomics is a concept for analyses of monogenic diseases.
Collapse
Affiliation(s)
- Ines Tapken
- SMATHERIA gGmbH-Non-Profit Biomedical Research Institute, Hannover 30625, Germany
- Center for Systems Neuroscience (ZSN), Hannover 30559, Germany
- Research Core Unit Proteomics, Hannover Medical School (MHH), Hannover 30625, Germany
| | - Theresa Schweitzer
- Research Core Unit Proteomics, Hannover Medical School (MHH), Hannover 30625, Germany
- Institute of Toxicology, Hannover Medical School (MHH), Hannover 30625, Germany
| | | | - Tobias Schüning
- SMATHERIA gGmbH-Non-Profit Biomedical Research Institute, Hannover 30625, Germany
| | - Nora T Detering
- SMATHERIA gGmbH-Non-Profit Biomedical Research Institute, Hannover 30625, Germany
- Center for Systems Neuroscience (ZSN), Hannover 30559, Germany
- Research Core Unit Proteomics, Hannover Medical School (MHH), Hannover 30625, Germany
| | - Gaurav Sharma
- CNR Unit, Institute of Biophysics, Trento 38123, Italy
| | - Moritz Niesert
- Department of Pediatrics I, Center for Pediatrics and Adolescent Medicine, Heidelberg University, Heidelberg 69120, Germany
| | - Afshin Saffari
- Department of Pediatrics I, Center for Pediatrics and Adolescent Medicine, Heidelberg University, Heidelberg 69120, Germany
| | - Daniela Kuhn
- SMATHERIA gGmbH-Non-Profit Biomedical Research Institute, Hannover 30625, Germany
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, Hannover 30625, Germany
| | - Amy Glynn
- SMATHERIA gGmbH-Non-Profit Biomedical Research Institute, Hannover 30625, Germany
| | - Federica Cieri
- CNR, Institute of Biosciences and Bioresources (IBBR), Naples 80131, Italy
- Department of Biology, University of Naples Federico II, Naples 80131, Italy
| | - Pamela Santonicola
- CNR, Institute of Biosciences and Bioresources (IBBR), Naples 80131, Italy
| | | | - Florian Gerstner
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig 04103, Germany
| | - Kiterie M E Faller
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Yu-Ting Huang
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Rashmi Kothary
- Faculty of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, K1H 8L6, Canada
| | - Thomas H Gillingwater
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Elia Di Schiavi
- CNR, Institute of Biosciences and Bioresources (IBBR), Naples 80131, Italy
| | - Christian M Simon
- Carl-Ludwig-Institute for Physiology, Leipzig University, Leipzig 04103, Germany
| | - Niko Hensel
- Department of Anatomy and Cell Biology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle (Saale) 06108, Germany
| | - Andreas Ziegler
- Department of Pediatrics I, Center for Pediatrics and Adolescent Medicine, Heidelberg University, Heidelberg 69120, Germany
| | | | - Andreas Pich
- Research Core Unit Proteomics, Hannover Medical School (MHH), Hannover 30625, Germany
- Institute of Toxicology, Hannover Medical School (MHH), Hannover 30625, Germany
| | - Peter Claus
- SMATHERIA gGmbH-Non-Profit Biomedical Research Institute, Hannover 30625, Germany
- Center for Systems Neuroscience (ZSN), Hannover 30559, Germany
- Institute of Functional and Applied Anatomy, Hannover Medical School, Hannover 30625, Germany
| |
Collapse
|
3
|
Penatzer J, Steele L, Breuer J, Fabia R, Hall M, Thakkar RK. FAS(APO), DAMP, and AKT Phosphoproteins Expression Predict the Development of Nosocomial Infection After Pediatric Burn Injury. J Burn Care Res 2024; 45:1607-1616. [PMID: 38863248 PMCID: PMC11565198 DOI: 10.1093/jbcr/irae111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Indexed: 06/13/2024]
Abstract
Pediatric burn injuries are a leading cause of morbidity with infections being the most common acute complication. Thermal injuries elicit a heightened cytokine response while suppressing immune function; however, the mechanisms leading to this dysfunction are still unknown. Our aim was to identify extracellular proteins and circulating phosphoprotein expression in the plasma after burn injury to predict the development of nosocomial infection (NI). Plasma was collected within 72 hours after injury from 64 pediatric burn subjects; of these, 18 went on to develop an NI. Extracellular damage-associated molecular proteins, FAS(APO), and protein kinase b (AKT) signaling phosphoproteins were analyzed. Subjects who went on to develop an NI had elevated high-mobility group box 1, heat shock protein 90 (HSP90), and FAS expression than those who did not develop an NI after injury (NoNI). Concurrently, phosphorylated (p-)AKT and mammalian target of rapamycin (p-mTOR) were elevated in those subjects who went on to develop an NI. Quadratic discriminant analysis revealed distinct differential profiles between NI and NoNI burn subjects using HSP90, FAS, and p-mTOR. The area under the receiver-operator characteristic curves displayed significant ability to distinguish between these 2 burn subject cohorts. These findings provide insight into predicting the signaling proteins involved in the development of NI in pediatric burn patients. Further, these proteins show promise as a diagnostic tool for pediatric burn patients at risk of developing infection while additional investigation may lead to potential therapeutics to prevent NI.
Collapse
Affiliation(s)
- Julia Penatzer
- Center for Clinical and Translation Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Lisa Steele
- Division of Critical Care Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Julie Breuer
- Division of Critical Care Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Renata Fabia
- Department of Pediatric Surgery, Burn Center, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Mark Hall
- Center for Clinical and Translation Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Division of Critical Care Medicine, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Rajan K Thakkar
- Center for Clinical and Translation Research, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatric Surgery, Burn Center, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| |
Collapse
|
4
|
Cascardo F, Vivanco M, Perrone MC, Werbach A, Enrico D, Mando P, Amat M, Martínez-Vazquez P, Burruchaga J, Mac Donnell M, Lanari C, Zwenger A, Waisberg F, Novaro V. Higher risk of recurrence in early-stage breast cancer patients with increased levels of ribosomal protein S6. Sci Rep 2024; 14:25136. [PMID: 39448637 PMCID: PMC11502685 DOI: 10.1038/s41598-024-75154-1] [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: 06/19/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024] Open
Abstract
PI3K/AKT/mTOR pathway is implicated in breast cancer progression and recurrence. The identification of PIK3CA and AKT1 mutations and loss of PTEN serve as selection criterion for targeted therapies involving selective inhibitors. However, they do not consistently align with pathway activation, and high-cost determinations limit their routine application. PI3K-downstream epigenetic regulatory mechanisms broaden the alterations that amplify pathway activity and, consequently, sensitivity to selective inhibitors. In this retrospective observational study, conducted within a cohort of early-stage breast cancer patients, we determined phosphorylated ribosomal protein S6 (pS6) at Ser240/244 by immunohistochemistry as an indicator of PI3K pathway activation. Log-Rank test and Cox proportional hazards regression were used to analyze the clinical relevance of pS6, alone and together with clinicopathological variables, regarding recurrence-free survival. ROC curves and the area under the curves were used to evaluate the calibration and discrimination properties of uni- and multivariate models. Our results show that a high percentage of pS6 positive tumor cells was associated with an unfavorable prognosis in a cohort of 129 hormone receptor positive/HER2 negative breast cancer patients (Hazard Ratio = 5.92; Log-Rank p = 9.5e-08; median follow-up = 53 months). When assessed in combination with lymph node status, the predictive capacity was higher compared to both univariate models individually. In conclusion, pS6 could represent a novel independent marker for predicting recurrence risk in luminal breast cancer.
Collapse
Affiliation(s)
- Florencia Cascardo
- Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Micaela Vivanco
- Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - María Cecilia Perrone
- Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Andrea Werbach
- Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Diego Enrico
- Instituto Alexander Fleming (IAF), Buenos Aires, Argentina
| | - Pablo Mando
- Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Buenos Aires, Argentina
| | - Mora Amat
- Instituto Alexander Fleming (IAF), Buenos Aires, Argentina
| | | | - Javier Burruchaga
- Hospital de Agudos "Magdalena V. de Martínez", General Pacheco, Buenos Aires, Argentina
| | - María Mac Donnell
- Hospital Provincial de Neuquén "Dr. Castro Rendón", Neuquén, Argentina
| | - Claudia Lanari
- Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Ariel Zwenger
- Hospital Provincial de Neuquén "Dr. Castro Rendón", Neuquén, Argentina
- Grupo Oncológico Cooperativo del Sur (GOCS), Neuquén, Argentina
| | | | - Virginia Novaro
- Instituto de Biología y Medicina Experimental (IBYME) - Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
5
|
Mei Y, Wang NN. New insights into the regulation of ethylene biosynthesis during leaf senescence in Arabidopsis. THE NEW PHYTOLOGIST 2024; 244:5-6. [PMID: 38840567 DOI: 10.1111/nph.19890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
This article is a Commentary on Zhu et al. (2024), 244: 116–130.
Collapse
Affiliation(s)
- Yuanyuan Mei
- Tianjin Key Laboratory of Protein Sciences, Department of Plant Biology and Ecology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ning Ning Wang
- Tianjin Key Laboratory of Protein Sciences, Department of Plant Biology and Ecology, College of Life Sciences, Nankai University, Tianjin, 300071, China
| |
Collapse
|
6
|
Thomas ACQ, Stead CA, Burniston JG, Phillips SM. Exercise-specific adaptations in human skeletal muscle: Molecular mechanisms of making muscles fit and mighty. Free Radic Biol Med 2024; 223:341-356. [PMID: 39147070 DOI: 10.1016/j.freeradbiomed.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
The mechanisms leading to a predominantly hypertrophied phenotype versus a predominantly oxidative phenotype, the hallmarks of resistance training (RT) or aerobic training (AT), respectively, are being unraveled. In humans, exposure of naïve persons to either AT or RT results in their skeletal muscle exhibiting generic 'exercise stress-related' signaling, transcription, and translation responses. However, with increasing engagement in AT or RT, the responses become refined, and the phenotype typically associated with each form of exercise emerges. Here, we review some of the mechanisms underpinning the adaptations of how muscles become, through AT, 'fit' and RT, 'mighty.' Much of our understanding of molecular exercise physiology has arisen from targeted analysis of post-translational modifications and measures of protein synthesis. Phosphorylation of specific residue sites has been a dominant focus, with canonical signaling pathways (AMPK and mTOR) studied extensively in the context of AT and RT, respectively. These alone, along with protein synthesis, have only begun to elucidate key differences in AT and RT signaling. Still, key yet uncharacterized differences exist in signaling and regulation of protein synthesis that drive unique adaptation to AT and RT. Omic studies are required to better understand the divergent relationship between exercise and phenotypic outcomes of training.
Collapse
Affiliation(s)
- Aaron C Q Thomas
- Protein Metabolism Research Lab, Department of Kinesiology, McMaster University, Hamilton, ON, Canada; Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Connor A Stead
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Jatin G Burniston
- Research Institute for Sport & Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Stuart M Phillips
- Protein Metabolism Research Lab, Department of Kinesiology, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
7
|
Ramalho S, Dopler A, Faller W. Ribosome specialization in cancer: a spotlight on ribosomal proteins. NAR Cancer 2024; 6:zcae029. [PMID: 38989007 PMCID: PMC11231584 DOI: 10.1093/narcan/zcae029] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 06/11/2024] [Accepted: 06/19/2024] [Indexed: 07/12/2024] Open
Abstract
In the past few decades, our view of ribosomes has changed substantially. Rather than passive machines without significant variability, it is now acknowledged that they are heterogeneous, and have direct regulatory capacity. This 'ribosome heterogeneity' comes in many flavors, including in both the RNA and protein components of ribosomes, so there are many paths through which ribosome specialization could arise. It is easy to imagine that specialized ribosomes could have wide physiological roles, through the translation of specific mRNA populations, and there is now evidence for this in several contexts. Translation is highly dysregulated in cancer, needed to support oncogenic phenotypes and to overcome cellular stress. However, the role of ribosome specialization in this is not clear. In this review we focus on specialized ribosomes in cancer. Specifically, we assess the impact that post-translational modifications and differential ribosome incorporation of ribosomal proteins (RPs) have in this disease. We focus on studies that have shown a ribosome-mediated change in translation of specific mRNA populations, and hypothesize how such a process could be driving other phenotypes. We review the impact of RP-mediated heterogeneity in both intrinsic and extrinsic oncogenic processes, and consider how this knowledge could be leveraged to benefit patients.
Collapse
Affiliation(s)
- Sofia Ramalho
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Anna Dopler
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - William James Faller
- Division of Oncogenomics, The Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
8
|
Dheeraj A, Garcia Marques FJ, Tailor D, Bermudez A, Resendez A, Pandrala M, Grau B, Kumar P, Haley CB, Honkala A, Kujur P, Jeffrey SS, Pitteri S, Malhotra SV. Inhibition of protein translational machinery in triple-negative breast cancer as a promising therapeutic strategy. Cell Rep Med 2024; 5:101552. [PMID: 38729158 PMCID: PMC11148772 DOI: 10.1016/j.xcrm.2024.101552] [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: 08/18/2022] [Revised: 07/11/2023] [Accepted: 04/11/2024] [Indexed: 05/12/2024]
Abstract
Y-box binding protein-1 (YB-1) is a proto-oncogenic protein associated with protein translation regulation. It plays a crucial role in the development and progression of triple-negative breast cancer (TNBC). In this study, we describe a promising approach to inhibit YB-1 using SU056, a small-molecule inhibitor. SU056 physically interacts with YB-1 and reduces its expression, which helps to restrain the progression of TNBC. Proteome profiling analysis indicates that the inhibition of YB-1 by SU056 can alter the proteins that regulate protein translation, an essential process for cancer cell growth. Preclinical studies on human cells, mice, and patient-derived xenograft tumor models show the effectiveness of SU056. Moreover, toxicological studies have shown that SU056 treatment and dosing are well tolerated without any adverse effects. Overall, our study provides a strong foundation for the further development of SU056 as a potential treatment option for patients with TNBC by targeting YB-1.
Collapse
Affiliation(s)
- Arpit Dheeraj
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Fernando Jose Garcia Marques
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Dhanir Tailor
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Abel Bermudez
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Angel Resendez
- Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Mallesh Pandrala
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Benedikt Grau
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Praveen Kumar
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Carrsyn B Haley
- Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Alexander Honkala
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Praveen Kujur
- Department of Surgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Stefanie S Jeffrey
- Department of Surgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sharon Pitteri
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sanjay V Malhotra
- Department of Cell, Developmental and Cancer Biology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Center for Experimental Therapeutics, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA; Department of Radiation Oncology, Stanford University School of Medicine, Palo Alto, CA, USA.
| |
Collapse
|
9
|
Lin D, Dong X, Xiao X, Xiang Z, Lei X, Wang J. Proteomic and phosphoproteomic analysis of responses to enterovirus A71 infection reveals novel targets for antiviral and viral replication. Antiviral Res 2023; 220:105761. [PMID: 37992763 DOI: 10.1016/j.antiviral.2023.105761] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/06/2023] [Accepted: 11/17/2023] [Indexed: 11/24/2023]
Abstract
Hand, foot, and mouth disease (HFMD) is a common infectious disease in infants and children, especially those under five years of age. EV-A71 is a common pathogen that causes HFMD and the primary pathogen leading to severe or fatal HFMD, which is characterized by neurological complications. However, the underlying mechanisms of EV-A71 pathogenesis remain largely unknown. In this report, we used proteomic and phosphorylated proteomic methods to characterize the proteome and phosphoproteome profiles of EV-A71-infected human neuroblastoma SK-N-SH cells. More than 7744 host proteins and 10069 phosphorylation modification sites were successfully quantified. Among them, 974 proteins and 3648 phosphorylation modification sites were regulated significantly during EV-A71 infection. KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analysis revealed that EV-A71 altered cell biological processes, including protein synthesis, RNA splicing and metabolism in SK-N-SH cells. Notably, based on the prediction of upregulated kinases during EV-A71 infection, we identified specific kinase inhibitors approved by the FDA, with ceralasertib, bosutinib, flavin mononucleotide, minocycline, pimasertib and acetylcysteine inhibiting EV-A71 infection. Finally, EV-A71 proteins were found to be phosphorylated during infection, with one site (S184 on 3D polymerase) observed to be crucial for viral replication because a S184A mutation knocked out viral replication. The results improve our understanding of the host response to EV-A71 infection of neuroblastoma cells and provide potential targets for developing anti-EV-A71 strategies.
Collapse
Affiliation(s)
- Dandan Lin
- NHC Key Laboratory of System Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China
| | - Xiaojing Dong
- NHC Key Laboratory of System Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China
| | - Xia Xiao
- NHC Key Laboratory of System Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China
| | - Zichun Xiang
- NHC Key Laboratory of System Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China; State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China
| | - Xiaobo Lei
- NHC Key Laboratory of System Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China; State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, PR China.
| | - Jianwei Wang
- NHC Key Laboratory of System Biology of Pathogens and Christophe Mérieux Laboratory, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, PR China.
| |
Collapse
|
10
|
Lyons LC, Vanrobaeys Y, Abel T. Sleep and memory: The impact of sleep deprivation on transcription, translational control, and protein synthesis in the brain. J Neurochem 2023; 166:24-46. [PMID: 36802068 PMCID: PMC10919414 DOI: 10.1111/jnc.15787] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/31/2023] [Accepted: 02/07/2023] [Indexed: 02/20/2023]
Abstract
In countries around the world, sleep deprivation represents a widespread problem affecting school-age children, teenagers, and adults. Acute sleep deprivation and more chronic sleep restriction adversely affect individual health, impairing memory and cognitive performance as well as increasing the risk and progression of numerous diseases. In mammals, the hippocampus and hippocampus-dependent memory are vulnerable to the effects of acute sleep deprivation. Sleep deprivation induces changes in molecular signaling, gene expression and may cause changes in dendritic structure in neurons. Genome wide studies have shown that acute sleep deprivation alters gene transcription, although the pool of genes affected varies between brain regions. More recently, advances in research have drawn attention to differences in gene regulation between the level of the transcriptome compared with the pool of mRNA associated with ribosomes for protein translation following sleep deprivation. Thus, in addition to transcriptional changes, sleep deprivation also affects downstream processes to alter protein translation. In this review, we focus on the multiple levels through which acute sleep deprivation impacts gene regulation, highlighting potential post-transcriptional and translational processes that may be affected by sleep deprivation. Understanding the multiple levels of gene regulation impacted by sleep deprivation is essential for future development of therapeutics that may mitigate the effects of sleep loss.
Collapse
Affiliation(s)
- Lisa C Lyons
- Program in Neuroscience, Department of Biological Science, Florida State University, Tallahassee, Florida, USA
| | - Yann Vanrobaeys
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Iowa City, Iowa, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| | - Ted Abel
- Department of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
- Iowa Neuroscience Institute, Iowa City, Iowa, USA
| |
Collapse
|
11
|
Rao TJR, Mao G, Cuffari BJ, Billack B. Dysregulation of the mTOR pathway by mechlorethamine. Toxicology 2023; 486:153434. [PMID: 36708981 PMCID: PMC10266297 DOI: 10.1016/j.tox.2023.153434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
Mechlorethamine (HN2) is a derivative of the chemical warfare agent sulfur mustard (SM) and cutaneous exposure to HN2 is associated with dermal-epidermal junction (DEJ) disruption (vesication). The primary purpose of the present study was to investigate the effect of HN2 on the mammalian target of rapamycin (mTOR) signaling pathway using an in vivo mouse ear vesicant model (MEVM). To this end, the ears of male C57BL/ 6 J mice were exposed to a single topical dose of HN2 (100 mM) or vehicle control (DMSO). Mice were then euthanized 30 min, 1 h or 24 h following exposure. Mouse ear skin exposed to HN2 and biopsied 24 h thereafter exhibited increased tissue expression of Raptor, an important member of the mTORC1 complex, relative to vehicle treated samples. HN2 reduced the downstream effectors phospho S6 (Ser 240/244) ribosomal protein and phospho 4E-BP1 (Thr 37/46) of the mTOR pathway in the epidermis at 30 min, 1 h and 24 h following HN2 exposure but not in the dermis. These results support the hypothesis that HN2-mediated cutaneous toxicity involves dysregulation of the mTOR signaling pathway in the epidermis.
Collapse
Affiliation(s)
| | - Ganming Mao
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, NY, USA
| | - Benedette J Cuffari
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, NY, USA
| | - Blase Billack
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Jamaica, NY, USA.
| |
Collapse
|
12
|
Olagoke OC, Segatto ALA, Afolabi BA, Ardisson-Araujo D, Aschner M, Rocha JBT. RPS6 transcriptional modulation in neural tissues of Nauphoeta cinerea during streptozotocin-associated sugar metabolism impairment. Comp Biochem Physiol B Biochem Mol Biol 2023; 263:110785. [PMID: 35931314 DOI: 10.1016/j.cbpb.2022.110785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/29/2022] [Accepted: 07/31/2022] [Indexed: 11/19/2022]
Abstract
The use of insects to model molecular events that characterize degenerative conditions was originally met with scepticism. However, the discovery of insect insulin-like peptides in the 1970's and the demonstration of evolutionary conservation of insulin-related signalling from insects to mammals have highlighted the importance and reduced cost of insect models in biomedical research. Here, we expand on our earlier described modelling of streptozotocin-induced brain glucose metabolic disruption in Nauphoeta cinerea, using RNA-sequencing analysis to study the transcriptional and genetic signatures of degeneration and stress signalling when glucose levels are elevated in the brain of the lobster cockroach. Nymphs were randomly divided into three groups: Control (0.8% NaCl), and two single streptozotocin injection doses (74 nmol and 740 nmol). The transcriptional analyses featured a dysregulation of 226 genes at high dose STZ treatment and 278 genes at the low dose. Our mRNA-sequencing data showed that ribosomal protein genes were the most upregulated genes at both 74 and 740 nmol STZ treatment. We therefore used RT-qPCR and relative transcriptional methods to validate our proposed mechanism of brain glucose toxicity-induced degeneration in Nauphoeta cinerea, which involved the upregulation of ribosomal proteins and rpS6 regulators (mTORC1, protein kinases, casein kinase 1 and Death-associated protein kinase), the upregulation of MAPK cascades (RAS, ERK, P38 and JNK), alongside the downregulation of the PI3K/AKT cascade. Taken together, this study highlights the remarkable opportunity for Nauphoeta cinerea use as an experimental organism in hyperglycaemia, degeneration, and stress signalling.
Collapse
Affiliation(s)
- Olawande C Olagoke
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences (CCNE), Federal University of Santa Maria, 97105-900 Santa Maria, RS, Brazil; Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Ana L A Segatto
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences (CCNE), Federal University of Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | | | - Daniel Ardisson-Araujo
- Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, DF 70910-900, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - João B T Rocha
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences (CCNE), Federal University of Santa Maria, 97105-900 Santa Maria, RS, Brazil
| |
Collapse
|
13
|
de Vasconcelos DAA, Nachbar RT, Pinheiro CH, do Amaral CL, Crisma AR, Vitzel KF, Abreu P, Alonso-Vale MI, Lopes AB, Bento-Santos A, Falcão-Tebas F, de Santana DF, do Nascimento E, Curi R, Pithon-Curi TC, Hirabara SM, Leandro CG. Maternal low-protein diet reduces skeletal muscle protein synthesis and mass via Akt-mTOR pathway in adult rats. Front Nutr 2022; 9:947458. [PMID: 36110404 PMCID: PMC9468266 DOI: 10.3389/fnut.2022.947458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022] Open
Abstract
Several studies have demonstrated that a maternal low-protein diet induces long-term metabolic disorders, but the involved mechanisms are unclear. This study investigated the molecular effects of a low-protein diet during pregnancy and lactation on glucose and protein metabolism in soleus muscle isolated from adult male rats. Female rats were fed either a normal protein diet or low-protein diet during gestation and lactation. After weaning, all pups were fed a normal protein diet until the 210th day postpartum. In the 7th month of life, mass, contractile function, protein and glucose metabolism, and the Akt-mTOR pathway were measured in the soleus muscles of male pups. Dry weight and contractile function of soleus muscle in the low-protein diet group rats were found to be lower compared to the control group. Lipid synthesis was evaluated by measuring palmitate incorporation in white adipose tissue. Palmitate incorporation was higher in the white adipose tissue of the low-protein diet group. When incubated soleus muscles were stimulated with insulin, protein synthesis, total amino acid incorporation and free amino acid content, glucose incorporation and uptake, and glycogen synthesis were found to be reduced in low-protein diet group rats. Fasting glycemia was higher in the low-protein diet group. These metabolic changes were associated with a decrease in Akt and GSK-3β signaling responses to insulin and a reduction in RPS6 in the absence of the hormone. There was also notably lower expression of Akt in the isolated soleus muscle of low-protein diet group rats. This study is the first to demonstrate how maternal diet restriction can reduce skeletal muscle protein and mass by downregulating the Akt-mTOR pathway in adulthood.
Collapse
Affiliation(s)
- Diogo Antonio Alves de Vasconcelos
- Department of Nutrition, Center of Health Sciences, Federal University of Pernambuco, Recife, Brazil
- Post-graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Vitória de Santo Antão, Vitória de Santo Antão, Brazil
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Renato Tadeu Nachbar
- Quebec Heart and Lung Institute Research Center, Laval University, Quebec City, QC, Canada
| | - Carlos Hermano Pinheiro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Cátia Lira do Amaral
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Amanda Rabello Crisma
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Kaio Fernando Vitzel
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- School of Health Sciences, College of Health, Massey University, Auckland, New Zealand
| | - Phablo Abreu
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Isabel Alonso-Vale
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Andressa Bolsoni Lopes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Adriano Bento-Santos
- Post-graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Vitória de Santo Antão, Vitória de Santo Antão, Brazil
| | - Filippe Falcão-Tebas
- The Ritchie Centre, Hudson Institute of Medical Research, Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - David Filipe de Santana
- Post-graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Vitória de Santo Antão, Vitória de Santo Antão, Brazil
| | - Elizabeth do Nascimento
- Department of Nutrition, Center of Health Sciences, Federal University of Pernambuco, Recife, Brazil
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Tania Cristina Pithon-Curi
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Sandro Massao Hirabara
- Interdisciplinary Post-graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, Brazil
| | - Carol Góis Leandro
- Post-graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Vitória de Santo Antão, Vitória de Santo Antão, Brazil
| |
Collapse
|
14
|
Beyond controlling cell size: functional analyses of S6K in tumorigenesis. Cell Death Dis 2022; 13:646. [PMID: 35879299 PMCID: PMC9314331 DOI: 10.1038/s41419-022-05081-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 01/21/2023]
Abstract
As a substrate and major effector of the mammalian target of rapamycin complex 1 (mTORC1), the biological functions of ribosomal protein S6 kinase (S6K) have been canonically assigned for cell size control by facilitating mRNA transcription, splicing, and protein synthesis. However, accumulating evidence implies that diverse stimuli and upstream regulators modulate S6K kinase activity, leading to the activation of a plethora of downstream substrates for distinct pathobiological functions. Beyond controlling cell size, S6K simultaneously plays crucial roles in directing cell apoptosis, metabolism, and feedback regulation of its upstream signals. Thus, we comprehensively summarize the emerging upstream regulators, downstream substrates, mouse models, clinical relevance, and candidate inhibitors for S6K and shed light on S6K as a potential therapeutic target for cancers.
Collapse
|
15
|
Li F, Fang Y, Zhuang Q, Cheng M, Moronge D, Jue H, Meyuhas O, Ding X, Zhang Z, Chen JK, Wu H. Blocking ribosomal protein S6 phosphorylation inhibits podocyte hypertrophy and focal segmental glomerulosclerosis. Kidney Int 2022; 102:121-135. [PMID: 35483522 PMCID: PMC10711420 DOI: 10.1016/j.kint.2022.02.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 02/01/2022] [Accepted: 02/17/2022] [Indexed: 10/18/2022]
Abstract
Ribosomal protein S6 (rpS6) phosphorylation mediates the hypertrophic growth of kidney proximal tubule cells. However, the role of rpS6 phosphorylation in podocyte hypertrophy and podocyte loss during the pathogenesis of focal segmental glomerulosclerosis (FSGS) remains undefined. Here, we examined rpS6 phosphorylation levels in kidney biopsy specimens from patients with FSGS and in podocytes from mouse kidneys with Adriamycin-induced FSGS. Using genetic and pharmacologic approaches in the mouse model of FSGS, we investigated the role of rpS6 phosphorylation in podocyte hypertrophy and loss during development and progression of FSGS. Phosphorylated rpS6 was found to be markedly increased in the podocytes of patients with FSGS and Adriamycin-induced FSGS mice. Genetic deletion of the Tuberous sclerosis 1 gene in kidney glomerular podocytes activated mammalian target of rapamycin complex 1 signaling to rpS6 phosphorylation, resulting in podocyte hypertrophy and pathologic features similar to those of patients with FSGS including podocyte loss, leading to segmental glomerulosclerosis. Since protein phosphatase 1 is known to negatively regulate rpS6 phosphorylation, treatment with an inhibitor increased phospho-rpS6 levels, promoted podocyte hypertrophy and exacerbated formation of FSGS lesions. Importantly, blocking rpS6 phosphorylation (either by generating congenic rpS6 knock-in mice expressing non-phosphorylatable rpS6 or by inhibiting ribosomal protein S6 kinase 1-mediated rpS6 phosphorylation with an inhibitor) significantly blunted podocyte hypertrophy, inhibited podocyte loss, and attenuated formation of FSGS lesions. Thus, our study provides genetic and pharmacologic evidence indicating that specifically targeting rpS6 phosphorylation can attenuate the development of FSGS lesions by inhibiting podocyte hypertrophy and associated podocyte depletion.
Collapse
Affiliation(s)
- Fang Li
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Cellular Biology and Anatomy Medical College of Georgia, Augusta University, Augusta, Georgia, USA; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yili Fang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qiyuan Zhuang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meichu Cheng
- Department of Cellular Biology and Anatomy Medical College of Georgia, Augusta University, Augusta, Georgia, USA; Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Desmond Moronge
- Department of Cellular Biology and Anatomy Medical College of Georgia, Augusta University, Augusta, Georgia, USA; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Hao Jue
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Oded Meyuhas
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Xiaoqiang Ding
- Department of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhigang Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Jian-Kang Chen
- Department of Cellular Biology and Anatomy Medical College of Georgia, Augusta University, Augusta, Georgia, USA; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Huijuan Wu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
16
|
Mapping PP1c and Its Inhibitor 2 Interactomes Reveals Conserved and Specific Networks in Asexual and Sexual Stages of Plasmodium. Int J Mol Sci 2022; 23:ijms23031069. [PMID: 35162991 PMCID: PMC8835298 DOI: 10.3390/ijms23031069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/10/2022] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
Malaria parasites require multiple phosphorylation and dephosphorylation steps to drive signaling pathways for proper differentiation and transformation. Several protein phosphatases, including protein phosphatase 1 (PP1), one of the main dephosphorylation enzymes, have been shown to be indispensable for the Plasmodium life cycle. The catalytic subunit of PP1 (PP1c) participates in cellular processes via dynamic interactions with a vast number of binding partners that contribute to its diversity of action. In this study, we used Plasmodium berghei transgenic parasite strains stably expressing PP1c or its inhibitor 2 (I2) tagged with mCherry, combined with the mCherry affinity pulldown of proteins from asexual and sexual stages, followed by mass spectrometry analyses. Mapped proteins were used to identify interactomes and to cluster functionally related proteins. Our findings confirm previously known physical interactions of PP1c and reveal enrichment of common biological processes linked to cellular component assembly in both schizonts and gametocytes to biosynthetic processes/translation in schizonts and to protein transport exclusively in gametocytes. Further, our analysis of PP1c and I2 interactomes revealed that nuclear export mediator factor and peptidyl-prolyl cis-trans isomerase, suggested to be essential in P. falciparum, could be potential targets of the complex PP1c/I2 in both asexual and sexual stages. Our study emphasizes the adaptability of Plasmodium PP1 and provides a fundamental study of the protein interaction landscapes involved in a myriad of events in Plasmodium, suggesting why it is crucial to the parasite and a source for alternative therapeutic strategies.
Collapse
|
17
|
Yi YW, You KS, Park JS, Lee SG, Seong YS. Ribosomal Protein S6: A Potential Therapeutic Target against Cancer? Int J Mol Sci 2021; 23:ijms23010048. [PMID: 35008473 PMCID: PMC8744729 DOI: 10.3390/ijms23010048] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 12/19/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
Ribosomal protein S6 (RPS6) is a component of the 40S small ribosomal subunit and participates in the control of mRNA translation. Additionally, phospho (p)-RPS6 has been recognized as a surrogate marker for the activated PI3K/AKT/mTORC1 pathway, which occurs in many cancer types. However, downstream mechanisms regulated by RPS6 or p-RPS remains elusive, and the therapeutic implication of RPS6 is underappreciated despite an approximately half a century history of research on this protein. In addition, substantial evidence from RPS6 knockdown experiments suggests the potential role of RPS6 in maintaining cancer cell proliferation. This motivates us to investigate the current knowledge of RPS6 functions in cancer. In this review article, we reviewed the current information about the transcriptional regulation, upstream regulators, and extra-ribosomal roles of RPS6, with a focus on its involvement in cancer. We also discussed the therapeutic potential of RPS6 in cancer.
Collapse
Affiliation(s)
- Yong Weon Yi
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (K.S.Y.); (J.-S.P.)
- Department of Nanobiomedical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
| | - Kyu Sic You
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (K.S.Y.); (J.-S.P.)
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
| | - Jeong-Soo Park
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (K.S.Y.); (J.-S.P.)
| | - Seok-Geun Lee
- Graduate School, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (S.-G.L.); (Y.-S.S.); Tel.: +82-2-961-2355 (S.-G.L.); +82-41-550-3875 (Y.-S.S.); Fax: +82-2-961-9623 (S.-G.L.)
| | - Yeon-Sun Seong
- Department of Biochemistry, College of Medicine, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea; (Y.W.Y.); (K.S.Y.); (J.-S.P.)
- Graduate School of Convergence Medical Science, Dankook University, Cheonan 31116, Chungcheongnam-do, Korea
- Correspondence: (S.-G.L.); (Y.-S.S.); Tel.: +82-2-961-2355 (S.-G.L.); +82-41-550-3875 (Y.-S.S.); Fax: +82-2-961-9623 (S.-G.L.)
| |
Collapse
|
18
|
Hippocampal neurons' cytosolic and membrane-bound ribosomal transcript profiles are differentially regulated by learning and subsequent sleep. Proc Natl Acad Sci U S A 2021; 118:2108534118. [PMID: 34819370 PMCID: PMC8640746 DOI: 10.1073/pnas.2108534118] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 12/25/2022] Open
Abstract
Sleep loss disrupts consolidation of hippocampus-dependent memory. To understand the cellular basis for this effect, we quantified RNAs associated with translating ribosomes in cytosol and on cellular membranes of different hippocampal neuron populations. Our analysis suggests that while sleep loss (but not learning) alters numerous ribosomal transcripts in cytosol, learning has dramatic effects on transcript profiles for less–well-characterized membrane-bound ribosomes. We demonstrate that postlearning sleep deprivation occludes already minimal learning-driven changes on cytosolic ribosomes. It simultaneously alters transcripts associated with metabolic and biosynthetic processes in membrane-bound ribosomes in excitatory hippocampal neurons and highly active, putative “engram” neurons, respectively. Together, these findings provide insights into the cellular mechanisms altered by learning and their disruption by subsequent sleep loss. The hippocampus is essential for consolidating transient experiences into long-lasting memories. Memory consolidation is facilitated by postlearning sleep, although the underlying cellular mechanisms are largely unknown. We took an unbiased approach to this question by using a mouse model of hippocampally mediated, sleep-dependent memory consolidation (contextual fear memory). Because synaptic plasticity is associated with changes to both neuronal cell membranes (e.g., receptors) and cytosol (e.g., cytoskeletal elements), we characterized how these cell compartments are affected by learning and subsequent sleep or sleep deprivation (SD). Translating ribosome affinity purification was used to profile ribosome-associated RNAs in different subcellular compartments (cytosol and membrane) and in different cell populations (whole hippocampus, Camk2a+ neurons, or highly active neurons with phosphorylated ribosomal subunit S6 [pS6+]). We examined how transcript profiles change as a function of sleep versus SD and prior learning (contextual fear conditioning; CFC). While sleep loss altered many cytosolic ribosomal transcripts, CFC altered almost none, and CFC-driven changes were occluded by subsequent SD. In striking contrast, SD altered few transcripts on membrane-bound (MB) ribosomes, while learning altered many more (including long non-coding RNAs [lncRNAs]). The cellular pathways most affected by CFC were involved in structural remodeling. Comparisons of post-CFC MB transcript profiles between sleeping and SD mice implicated changes in cellular metabolism in Camk2a+ neurons and protein synthesis in highly active pS6+ (putative “engram”) neurons as biological processes disrupted by SD. These findings provide insights into how learning affects hippocampal neurons and suggest that the effects of SD on memory consolidation are cell type and subcellular compartment specific.
Collapse
|
19
|
Zhu FY, Wang LL, Meng TG, Wang RL, Yang ZX, Cao Y, Zhu GY, Jin Z, Gao LL, Zeng WT, Wang ZB, Sun QY, Zhang D. Inhibiting bridge integrator 2 phosphorylation leads to improved oocyte quality, ovarian health and fertility in aging and after chemotherapy in mice. NATURE AGING 2021; 1:1010-1023. [PMID: 37118338 DOI: 10.1038/s43587-021-00133-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 10/04/2021] [Indexed: 04/30/2023]
Abstract
Female ovaries degenerate about 20 years earlier than testes leading to reduced primordial follicle reserve and a reduction in oocyte quality. Here we found that bridge integrator 2 (BIN2) is enriched in mouse ovaries and oocytes and that global knockout of this protein improves both female fertility and oocyte quality. Quantitative ovarian proteomics and phosphoproteomics showed that Bin2 knockout led to a decrease in phosphorylated ribosomal protein S6 (p-RPS6), a component of the mammalian target of rapamycin pathway and greatly increased nicotinamide nucleotide transhydrogenase (NNT), the free-radical detoxifier. Mechanistically, we find that phosphorylation of BIN2 at Thr423 and Ser424 leads to its translocation from the membrane to the cytoplasm, subsequent phosphorylation of RPS6 and inhibition of Nnt translation. We synthesized a BIN2-penetrating peptide (BPP) designed to inhibit BIN2 phosphorylation and found that a 3-week BPP treatment improved primordial follicle reserve and oocyte quality in aging and after chemotherapy-induced premature ovarian failure without discernible side effects.
Collapse
Affiliation(s)
- Feng-Yu Zhu
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Li-Li Wang
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Tie-Gang Meng
- Fertility Preservation Lab and Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China
- State Key Lab of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ruo-Lei Wang
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Zhi-Xia Yang
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Ying Cao
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Gang-Yi Zhu
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Zhen Jin
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Lei-Lei Gao
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China
| | - Wen-Tao Zeng
- Animal Core Facility, Nanjing Medical University, Nanjing, China
| | - Zhen-Bo Wang
- State Key Lab of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Qing-Yuan Sun
- Fertility Preservation Lab and Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, China.
| | - Dong Zhang
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China.
- Animal Core Facility, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
20
|
Pfeiffer S, Tomašcová A, Mamrak U, Haunsberger SJ, Connolly NMC, Resler A, Düssmann H, Weisová P, Jirström E, D'Orsi B, Chen G, Cremona M, Hennessy BT, Plesnila N, Prehn JHM. AMPK-regulated miRNA-210-3p is activated during ischaemic neuronal injury and modulates PI3K-p70S6K signalling. J Neurochem 2021; 159:710-728. [PMID: 33694332 DOI: 10.1111/jnc.15347] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/12/2021] [Accepted: 03/05/2021] [Indexed: 12/14/2022]
Abstract
Progressive neuronal injury following ischaemic stroke is associated with glutamate-induced depolarization, energetic stress and activation of AMP-activated protein kinase (AMPK). We here identify a molecular signature associated with neuronal AMPK activation, as a critical regulator of cellular response to energetic stress following ischaemia. We report a robust induction of microRNA miR-210-3p both in vitro in primary cortical neurons in response to acute AMPK activation and following ischaemic stroke in vivo. Bioinformatics and reverse phase protein array analysis of neuronal protein expression changes in vivo following administration of a miR-210-3p mimic revealed altered expression of phosphatase and tensin homolog (PTEN), 3-phosphoinositide-dependent protein kinase 1 (PDK1), ribosomal protein S6 kinase (p70S6K) and ribosomal protein S6 (RPS6) signalling in response to increasing miR-210-3p. In vivo, we observed a corresponding reduction in p70S6K activity following ischaemic stroke. Utilizing models of glutamate receptor over-activation in primary neurons, we demonstrated that induction of miR-210-3p was accompanied by sustained suppression of p70S6K activity and that this effect was reversed by miR-210-3p inhibition. Collectively, these results provide new molecular insight into the regulation of cell signalling during ischaemic injury, and suggest a novel mechanism whereby AMPK regulates miR-210-3p to control p70S6K activity in ischaemic stroke and excitotoxic injury.
Collapse
Affiliation(s)
- Shona Pfeiffer
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Anna Tomašcová
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Biomedical Centre Martin, Comenius University in Bratislava, Bratislava, Slovakia
| | - Uta Mamrak
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
| | - Stefan J Haunsberger
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Niamh M C Connolly
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alexa Resler
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Heiko Düssmann
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Petronela Weisová
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Elisabeth Jirström
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- FutureNeuro SFI Research Center, Royal College of Surgeons Ireland, Dublin, Ireland
| | - Beatrice D'Orsi
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Institute of Neuroscience, Italian National Research Council (CNR), Pisa, Italy
| | - Gang Chen
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mattia Cremona
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Dept of Molecular Medicine (Medical Oncology group), Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Bryan T Hennessy
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- Dept of Molecular Medicine (Medical Oncology group), Royal College of Surgeons in Ireland, Dublin, Ireland
- Department of Medical Oncology, Beaumont Hospital, Dublin, Ireland
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (ISD), Munich, Germany
- Munich Cluster of Systems Neurology (Synergy), Munich, Germany
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
- FutureNeuro SFI Research Center, Royal College of Surgeons Ireland, Dublin, Ireland
| |
Collapse
|
21
|
Sun L, Morikawa K, Sogo Y, Sugiura Y. MHY1485 enhances X-irradiation-induced apoptosis and senescence in tumor cells. JOURNAL OF RADIATION RESEARCH 2021; 62:782-792. [PMID: 34265852 PMCID: PMC8438247 DOI: 10.1093/jrr/rrab057] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 05/27/2021] [Indexed: 06/13/2023]
Abstract
The mammalian target of rapamycin (mTOR) is a sensor of nutrient status and plays an important role in cell growth and metabolism. Although inhibition of mTOR signaling promotes tumor cell death and several mTOR inhibitors have been used clinically, recent reports have shown that co-treatment with MHY1485, an mTOR activator, enhances the anti-cancer effects of anti-PD-1 antibody and 5-fluorouracil. However, it remains unclear whether MHY1485 treatment alters the effects of radiation on tumor cells. In this study, the radiosensitizing effects of MHY1485 were investigated using murine CT26 and LLC cell lines. We examined mTOR signaling, tumor cell growth, colony formation, apoptosis, senescence, oxidative stress, p21 accumulation and endoplasmic reticulum (ER) stress levels in cells treated with MHY1485 and radiation, either alone or together. We found that MHY1485 treatment inhibited growth and colony formation in both cell lines under irradiation and no-irradiation conditions, results that were not fully consistent with MHY1485's known role in activating mTOR signaling. Furthermore, we found that combined treatment with MHY1485 and radiation significantly increased apoptosis and senescence in tumor cells in association with oxidative stress, ER stress and p21 stabilization, compared to radiation treatment alone. Our results suggested that MHY1485 enhances the radiosensitivity of tumor cells by a mechanism that may differ from MHY1485's role in mTOR activation.
Collapse
Affiliation(s)
- Lue Sun
- Corresponding author. Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan. Tel: +81-29-849-1564; Fax: +81-29-861-6149; E-mail:
| | - Kumi Morikawa
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Yu Sogo
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Central 6, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Yuki Sugiura
- Health and Medical Research Institute, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), 2217-14, Hayashi-cho, Takamatsu, Kagawa 761-0895, Japan
| |
Collapse
|
22
|
Mancera-Martínez E, Dong Y, Makarian J, Srour O, Thiébeauld O, Jamsheer M, Chicher J, Hammann P, Schepetilnikov M, Ryabova LA. Phosphorylation of a reinitiation supporting protein, RISP, determines its function in translation reinitiation. Nucleic Acids Res 2021; 49:6908-6924. [PMID: 34133725 PMCID: PMC8266674 DOI: 10.1093/nar/gkab501] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 05/14/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
Reinitiation supporting protein, RISP, interacts with 60S (60S ribosomal subunit) and eIF3 (eukaryotic initiation factor 3) in plants. TOR (target-of-rapamycin) mediates RISP phosphorylation at residue Ser267, favoring its binding to eL24 (60S ribosomal protein L24). In a viral context, RISP, when phosphorylated, binds the CaMV transactivator/ viroplasmin, TAV, to assist in an exceptional mechanism of reinitiation after long ORF translation. Moreover, we show here that RISP interacts with eIF2 via eIF2β and TOR downstream target 40S ribosomal protein eS6. A RISP phosphorylation knockout, RISP-S267A, binds preferentially eIF2β, and both form a ternary complex with eIF3a in vitro. Accordingly, transient overexpression in plant protoplasts of RISP-S267A, but not a RISP phosphorylation mimic, RISP-S267D, favors translation initiation. In contrast, RISP-S267D preferentially binds eS6, and, when bound to the C-terminus of eS6, can capture 60S in a highly specific manner in vitro, suggesting that it mediates 60S loading during reinitiation. Indeed, eS6-deficient plants are highly resistant to CaMV due to their reduced reinitiation capacity. Strikingly, an eS6 phosphomimic, when stably expressed in eS6-deficient plants, can fully restore the reinitiation deficiency of these plants in cellular and viral contexts. These results suggest that RISP function in translation (re)initiation is regulated by phosphorylation at Ser267.
Collapse
Affiliation(s)
- Eder Mancera-Martínez
- Institut de biologie de moléculaire des plantes UPR2357 du CNRS, Université de Strasbourg, Strasbourg, France
| | - Yihan Dong
- Institut de biologie de moléculaire des plantes UPR2357 du CNRS, Université de Strasbourg, Strasbourg, France
| | - Joelle Makarian
- Institut de biologie de moléculaire des plantes UPR2357 du CNRS, Université de Strasbourg, Strasbourg, France
| | - Ola Srour
- Institut de biologie de moléculaire des plantes UPR2357 du CNRS, Université de Strasbourg, Strasbourg, France
| | - Odon Thiébeauld
- Institut de biologie de moléculaire des plantes UPR2357 du CNRS, Université de Strasbourg, Strasbourg, France
| | - Muhammed Jamsheer
- Institut de biologie de moléculaire des plantes UPR2357 du CNRS, Université de Strasbourg, Strasbourg, France
| | - Johana Chicher
- Plateforme protéomique Strasbourg Esplanade FRC1589 du CNRS, Université de Strasbourg, Strasbourg, France
| | - Philippe Hammann
- Plateforme protéomique Strasbourg Esplanade FRC1589 du CNRS, Université de Strasbourg, Strasbourg, France
| | - Mikhail Schepetilnikov
- Institut de biologie de moléculaire des plantes UPR2357 du CNRS, Université de Strasbourg, Strasbourg, France
| | - Lyubov A Ryabova
- Institut de biologie de moléculaire des plantes UPR2357 du CNRS, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
23
|
Li Y, Zhang J, Sun H, Chen Y, Li W, Yu X, Zhao X, Zhang L, Yang J, Xin W, Jiang Y, Wang G, Shi W, Zhu D. lnc-Rps4l-encoded peptide RPS4XL regulates RPS6 phosphorylation and inhibits the proliferation of PASMCs caused by hypoxia. Mol Ther 2021; 29:1411-1424. [PMID: 33429084 PMCID: PMC8058491 DOI: 10.1016/j.ymthe.2021.01.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/29/2020] [Accepted: 01/02/2021] [Indexed: 02/06/2023] Open
Abstract
Pulmonary artery smooth muscle cells (PASMCs) proliferation caused by hypoxia is an important pathological process of pulmonary hypertension (PH). Prevention of PASMCs proliferation can effectively reduce PH mortality. Long non-coding RNAs (lncRNAs) are involved in the proliferation process. Recent evidence has demonstrated that functional peptides encoded by lncRNAs play important roles in cell pathophysiological process. Our previous study has demonstrated that lnc-Rps4l with high coding ability mediates the PASMCs proliferation under hypoxic conditions. We hypothesize in this study that a lnc-Rps4l-encoded peptide is involved in hypoxic-induced PASMCs proliferation. The presence of peptide 40S ribosomal protein S4 X isoform-like (RPS4XL) encoded by lnc-Rps4l in PASMCs under hypoxic conditions was confirmed by bioinformatics, immunofluorescence, and immunohistochemistry. Inhibition of proliferation by the peptide RPS4XL was demonstrated in hypoxic PASMCs by MTT, bromodeoxyuridine (BrdU) incorporation, and immunofluorescence assays. By using the bioinformatics, coimmunoprecipitation (coIP), and mass spectrometry, RPS6 was identified to interact with RPS4XL. Furthermore, lnc-Rps4l-encoded peptide RPS4XL inhibited the RPS6 process via binding to RPS6 and inhibiting RPS6 phosphorylation at p-RPS6 (Ser240+Ser244) phosphorylation site. These results systematically elucidate the role and regulatory network of Rps4l-encoded peptide RPS4XL in PASMCs proliferation. These discoveries provide potential targets for early diagnosis and a leading compound for treatment of hypoxic PH.
Collapse
Affiliation(s)
- Yiying Li
- Biopharmaceutical Key Laboratory of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province 150081, P.R. China
| | - Junting Zhang
- Biopharmaceutical Key Laboratory of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province 150081, P.R. China
| | - Hanliang Sun
- Biopharmaceutical Key Laboratory of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province 150081, P.R. China
| | - Yujie Chen
- Biopharmaceutical Key Laboratory of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province 150081, P.R. China
| | - Wendi Li
- College of Pharmacy, Harbin University of Commerce, Harbin, Heilongjiang Province 150081, P.R. China
| | - Xiufeng Yu
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Xijuan Zhao
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Lixin Zhang
- Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China
| | - Jianfeng Yang
- College of Pharmacy, Harbin Medical University, Daqing 163319, P.R. China
| | - Wei Xin
- Biopharmaceutical Key Laboratory of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province 150081, P.R. China
| | - Yuan Jiang
- Biopharmaceutical Key Laboratory of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province 150081, P.R. China
| | - Guilin Wang
- College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, Heilongjiang Province 163319, P.R. China
| | - Wenbin Shi
- College of Medical Laboratory Science and Technology, Harbin Medical University, Daqing, Heilongjiang Province 163319, P.R. China
| | - Daling Zhu
- Biopharmaceutical Key Laboratory of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin, Heilongjiang Province 150081, P.R. China; Central Laboratory of Harbin Medical University (Daqing), Daqing 163319, P.R. China.
| |
Collapse
|
24
|
Abstract
We set out to identify novel neuronal populations controlling feeding behaviors. Therefore, we focused on identifying novel neuronal populations modulating food intake and body weight. In this article, we report that DMHPpp1r17 neurons are activated by increased food intake and that activating them results in decreased food intake and body weight, while inhibiting them leads to increased body weight and food intake. These data suggest that DMHPpp1r17 neurons restrict binges of eating. In addition to its basic science importance, these findings could have therapeutic applications, as they suggest that pharmacologic activation of PPP1R17 neurons could potentially reduce weight in settings of obesity and binge-like eating. Leptin-deficient ob/ob mice eat voraciously, and their food intake is markedly reduced by leptin treatment. In order to identify potentially novel sites of leptin action, we used PhosphoTRAP to molecularly profile leptin-responsive neurons in the hypothalamus and brainstem. In addition to identifying several known leptin responsive populations, we found that neurons in the dorsomedial hypothalamus (DMH) of ob/ob mice expressing protein phosphatase 1 regulatory subunit 17 (PPP1R17) constitutively express cFos and that this is suppressed by leptin treatment. Because ob mice are hyperphagic, we hypothesized that activating PPP1R17 neurons would increase food intake. However, chemogenetic activation of PPP1R17 neurons decreased food intake and body weight of ob/ob mice while inhibition of PPP1R17 neurons increased them. Similarly, in a scheduled feeding protocol that elicits increased consumption, mice also ate more when PPP1R17 neurons were inhibited and ate less when they were activated. Finally, we found that pair-feeding of ob mice reduced cFos expression to a similar extent as leptin and that reducing the amount of food available during scheduled feeding in DMHPpp1r17 neurons also decreased cFos in DMHPpp1r17 neurons. Finally, these neurons do not express the leptin receptor, suggesting that the effect of leptin on these neurons is indirect and secondary to reduced food intake. In aggregate, these results show that PPP1R17 neurons in the DMH are activated by increased food intake and in turn restrict intake to limit overconsumption, suggesting that they function to constrain binges of eating.
Collapse
|
25
|
Alonso-Barroso E, Pérez B, Desviat LR, Richard E. Cardiomyocytes Derived from Induced Pluripotent Stem Cells as a Disease Model for Propionic Acidemia. Int J Mol Sci 2021; 22:ijms22031161. [PMID: 33503868 PMCID: PMC7865492 DOI: 10.3390/ijms22031161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
Propionic acidemia (PA), one of the most frequent life-threatening organic acidemias, is caused by mutations in either the PCCA or PCCB genes encoding both subunits of the mitochondrial propionyl-CoA carboxylase (PCC) enzyme. Cardiac alterations (hypertrophy, dilated cardiomyopathy, long QT) are one of the major causes of mortality in patients surviving the neonatal period. To overcome limitations of current cellular models of PA, we generated induced pluripotent stem cells (iPSCs) from a PA patient with defects in the PCCA gene, and successfully differentiated them into cardiomyocytes. PCCA iPSC-derived cardiomyocytes exhibited reduced oxygen consumption, an accumulation of residual bodies and lipid droplets, and increased ribosomal biogenesis. Furthermore, we found increased protein levels of HERP, GRP78, GRP75, SIG-1R and MFN2, suggesting endoplasmic reticulum stress and calcium perturbations in these cells. We also analyzed a series of heart-enriched miRNAs previously found deregulated in the heart tissue of a PA murine model and confirmed their altered expression. Our novel results show that PA iPSC-cardiomyocytes represent a promising model for investigating the pathological mechanisms underlying PA cardiomyopathies, also serving as an ex vivo platform for therapeutic evaluation.
Collapse
Affiliation(s)
- Esmeralda Alonso-Barroso
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (E.A.-B.); (B.P.); (L.R.D.)
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, 28029 Madrid, Spain
| | - Belén Pérez
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (E.A.-B.); (B.P.); (L.R.D.)
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, 28029 Madrid, Spain
| | - Lourdes Ruiz Desviat
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (E.A.-B.); (B.P.); (L.R.D.)
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, 28029 Madrid, Spain
| | - Eva Richard
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, Universidad Autónoma de Madrid, 28049 Madrid, Spain; (E.A.-B.); (B.P.); (L.R.D.)
- Centro de Diagnóstico de Enfermedades Moleculares (CEDEM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, 28029 Madrid, Spain
- Instituto de Investigación Sanitaria Hospital La Paz (IdiPaz), ISCIII, 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
26
|
Green SR, Al-Attar R, McKechnie AE, Naidoo S, Storey KB. Role of Akt signaling pathway regulation in the speckled mousebird (Colius striatus) during torpor displays tissue specific responses. Cell Signal 2020; 75:109763. [PMID: 32871209 DOI: 10.1016/j.cellsig.2020.109763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/06/2020] [Accepted: 08/25/2020] [Indexed: 10/23/2022]
Abstract
Pronounced heterothermic responses are relatively rare among birds. Along with taxa such as hummingbirds and caprimulgids, the order Coliiformes (mousebirds) is known to possess the physiological capacity for torpor. During torpor, body temperature is greatly reduced and a bird becomes unresponsive to external stimuli until ambient temperatures return to more favorable conditions. Under such conditions, these birds are forced to rely only on their internal fuel storage for energy and show great reduction in metabolic rates by decreasing energy-expensive processes. This study investigated the role of the key insulin-Akt signaling kinase pathway involved in regulating energy metabolism and protein translation in the liver, kidney, heart, skeletal muscle, and brain of the speckled mousebird (Colius striatus). The degree of phosphorylation of well-conserved target residues with important regulatory function was examined in both the euthermic control and torpid birds. The results demonstrated marked differences in responses between the tissues with decreases in RPS6 S235/236 phosphorylation in the kidney (0.52 fold of euthermic) and muscle (0.29 fold of euthermic) as well as decreases in GS3K3β S9 in muscle (0.60 fold of euthermic) and GSK3α S21 (0.71 fold of euthermic) phosphorylation in kidney during torpor, suggesting a downregulation of this pathway. Interestingly, the liver demonstrated an increase in RPS6 S235/236 (2.89 fold increase) and P70S6K T412 (1.44 fold increase) phosphorylation in the torpor group suggesting that protein translation is maintained in this tissue. This study demonstrates that avian torpor is a complex phenomenon and alterations in this signaling pathway follow a tissue specific pattern.
Collapse
Affiliation(s)
- Stuart R Green
- Institute of Biochemistry & Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa K1S 5B6, Ontario, Canada
| | - Rasha Al-Attar
- Institute of Biochemistry & Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa K1S 5B6, Ontario, Canada
| | - Andrew E McKechnie
- South African Research Chair in Conservation Physiology, National Zoological Garden, South African National Biodiversity Institute, Pretoria, South Africa; DST-NRF Centre of Excellence, FitzPatrick Institute, Department of Zoology and Entomology, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Samantha Naidoo
- South African Research Chair in Conservation Physiology, National Zoological Garden, South African National Biodiversity Institute, Pretoria, South Africa; DST-NRF Centre of Excellence, FitzPatrick Institute, Department of Zoology and Entomology, University of Pretoria, Private Bag X20, Hatfield 0028, South Africa
| | - Kenneth B Storey
- Institute of Biochemistry & Department of Biology, Carleton University, 1125 Colonel By Drive, Ottawa K1S 5B6, Ontario, Canada.
| |
Collapse
|
27
|
Klofas LK, Short BP, Snow JP, Sinnaeve J, Rushing GV, Westlake G, Weinstein W, Ihrie RA, Ess KC, Carson RP. DEPDC5 haploinsufficiency drives increased mTORC1 signaling and abnormal morphology in human iPSC-derived cortical neurons. Neurobiol Dis 2020; 143:104975. [PMID: 32574724 PMCID: PMC7462127 DOI: 10.1016/j.nbd.2020.104975] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 05/21/2020] [Accepted: 06/13/2020] [Indexed: 01/21/2023] Open
Abstract
Mutations in the DEPDC5 gene can cause epilepsy, including forms with and without brain malformations. The goal of this study was to investigate the contribution of DEPDC5 gene dosage to the underlying neuropathology of DEPDC5-related epilepsies. We generated induced pluripotent stem cells (iPSCs) from epilepsy patients harboring heterozygous loss of function mutations in DEPDC5. Patient iPSCs displayed increases in both phosphorylation of ribosomal protein S6 and proliferation rate, consistent with elevated mTORC1 activation. In line with these findings, we observed increased soma size in patient iPSC-derived cortical neurons that was rescued with rapamycin treatment. These data indicate that human cells heterozygous for DEPDC5 loss-of-function mutations are haploinsufficient for control of mTORC1 signaling. Our findings suggest that human pathology differs from mouse models of DEPDC5-related epilepsies, which do not show consistent phenotypic differences in heterozygous neurons, and support the need for human-based models to affirm and augment the findings from animal models of DEPDC5-related epilepsy.
Collapse
Affiliation(s)
- Lindsay K Klofas
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA
| | - Brittany P Short
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John P Snow
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Justine Sinnaeve
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Grant Westlake
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Will Weinstein
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca A Ihrie
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kevin C Ess
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Robert P Carson
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN, USA; Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
28
|
Costa RO, Martins H, Martins LF, Cwetsch AW, Mele M, Pedro JR, Tomé D, Jeon NL, Cancedda L, Jaffrey SR, Almeida RD. Synaptogenesis Stimulates a Proteasome-Mediated Ribosome Reduction in Axons. Cell Rep 2020; 28:864-876.e6. [PMID: 31340150 PMCID: PMC6686882 DOI: 10.1016/j.celrep.2019.06.080] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 12/21/2018] [Accepted: 06/21/2019] [Indexed: 11/19/2022] Open
Abstract
Ribosomes and a subset of cellular mRNAs are trafficked into axons of developing neurons. The axonal localization of translational machinery allows new proteins to be rapidly and locally synthesized during axonal growth and pathfinding. However, in mature neurons, axonal ribosomes are significantly reduced or even absent. The mechanism that elicits this removal is currently unknown. Here, we demonstrate that synapse formation is the trigger for ribosome reduction in mature axons. In vivo analysis shows that axonal ribosome levels decrease in rat brain at a developmental stage coincident with synapse formation. Next, we observe in vitro that different synaptogenic inducers trigger an overall decrease of ribosomal proteins and rRNA in the axons of spinal motor neurons. We further observe that this process is dependent on the ubiquitin-proteasome system but not on autophagy. Together, these data identify synaptogenesis as the long missing biological trigger that leads to ribosome disappearance during axonal maturation. The mechanism behind the striking loss of ribosomes from axons during neuronal maturation is unknown. Using in vivo and in vitro models, including neuron-muscle co-cultures and combining biochemistry and imaging techniques, Costa et al. demonstrate that synapse formation triggers ribosome reduction by a mechanism involving the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Rui O Costa
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - Helena Martins
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Luís F Martins
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal; PhD Programme in Experimental Biology and Biomedicine (PDBEB), Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Andrzej W Cwetsch
- NBT - Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy
| | - Miranda Mele
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Joana R Pedro
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
| | - Diogo Tomé
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; iBiMED - Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Noo Li Jeon
- Institute of Advanced Machinery and Design, Seoul National University, Seoul, Republic of Korea; Department of Mechanical and Aerospace Engineering, Seoul National University, Seoul, Republic of Korea
| | - Laura Cancedda
- NBT - Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Genoa, Italy; Dulbecco Telethon Institute, Roma, Italy
| | - Samie R Jaffrey
- Department of Pharmacology, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Ramiro D Almeida
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal; Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal; iBiMED - Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
29
|
LIN28B/ let-7 control the ability of neonatal murine auditory supporting cells to generate hair cells through mTOR signaling. Proc Natl Acad Sci U S A 2020; 117:22225-22236. [PMID: 32826333 DOI: 10.1073/pnas.2000417117] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mechano-sensory hair cells within the inner ear cochlea are essential for the detection of sound. In mammals, cochlear hair cells are only produced during development and their loss, due to disease or trauma, is a leading cause of deafness. In the immature cochlea, prior to the onset of hearing, hair cell loss stimulates neighboring supporting cells to act as hair cell progenitors and produce new hair cells. However, for reasons unknown, such regenerative capacity (plasticity) is lost once supporting cells undergo maturation. Here, we demonstrate that the RNA binding protein LIN28B plays an important role in the production of hair cells by supporting cells and provide evidence that the developmental drop in supporting cell plasticity in the mammalian cochlea is, at least in part, a product of declining LIN28B-mammalian target of rapamycin (mTOR) activity. Employing murine cochlear organoid and explant cultures to model mitotic and nonmitotic mechanisms of hair cell generation, we show that loss of LIN28B function, due to its conditional deletion, or due to overexpression of the antagonistic miRNA let-7g, suppressed Akt-mTOR complex 1 (mTORC1) activity and renders young, immature supporting cells incapable of generating hair cells. Conversely, we found that LIN28B overexpression increased Akt-mTORC1 activity and allowed supporting cells that were undergoing maturation to de-differentiate into progenitor-like cells and to produce hair cells via mitotic and nonmitotic mechanisms. Finally, using the mTORC1 inhibitor rapamycin, we demonstrate that LIN28B promotes supporting cell plasticity in an mTORC1-dependent manner.
Collapse
|
30
|
Miao ZF, Lewis MA, Cho CJ, Adkins-Threats M, Park D, Brown JW, Sun JX, Burclaff JR, Kennedy S, Lu J, Mahar M, Vietor I, Huber LA, Davidson NO, Cavalli V, Rubin DC, Wang ZN, Mills JC. A Dedicated Evolutionarily Conserved Molecular Network Licenses Differentiated Cells to Return to the Cell Cycle. Dev Cell 2020; 55:178-194.e7. [PMID: 32768422 DOI: 10.1016/j.devcel.2020.07.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/04/2020] [Accepted: 07/11/2020] [Indexed: 02/06/2023]
Abstract
Differentiated cells can re-enter the cell cycle to repair tissue damage via a series of discrete morphological and molecular stages coordinated by the cellular energetics regulator mTORC1. We previously proposed the term "paligenosis" to describe this conserved cellular regeneration program. Here, we detail a molecular network regulating mTORC1 during paligenosis in both mouse pancreatic acinar and gastric chief cells. DDIT4 initially suppresses mTORC1 to induce autodegradation of differentiated cell components and damaged organelles. Later in paligenosis, IFRD1 suppresses p53 accumulation. Ifrd1-/- cells do not complete paligenosis because persistent p53 prevents mTORC1 reactivation and cell proliferation. Ddit4-/- cells never suppress mTORC1 and bypass the IFRD1 checkpoint on proliferation. Previous reports and our current data implicate DDIT4/IFRD1 in governing paligenosis in multiple organs and species. Thus, we propose that an evolutionarily conserved, dedicated molecular network has evolved to allow differentiated cells to re-enter the cell cycle (i.e., undergo paligenosis) after tissue injury. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Zhi-Feng Miao
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, First Hospital of China Medical University, Shenyang 110001, China
| | - Mark A Lewis
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Charles J Cho
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mahliyah Adkins-Threats
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dongkook Park
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey W Brown
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jing-Xu Sun
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, First Hospital of China Medical University, Shenyang 110001, China
| | - Joseph R Burclaff
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan Kennedy
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jianyun Lu
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marcus Mahar
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Ilja Vietor
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Lukas A Huber
- Division of Cell Biology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Nicholas O Davidson
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Valeria Cavalli
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Deborah C Rubin
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhen-Ning Wang
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, First Hospital of China Medical University, Shenyang 110001, China.
| | - Jason C Mills
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
31
|
Dommann N, Sánchez-Taltavull D, Eggs L, Birrer F, Brodie T, Salm L, Baier FA, Medová M, Humbert M, Tschan MP, Beldi G, Candinas D, Stroka D. The LIM Protein Ajuba Augments Tumor Metastasis in Colon Cancer. Cancers (Basel) 2020; 12:cancers12071913. [PMID: 32679899 PMCID: PMC7409172 DOI: 10.3390/cancers12071913] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer, along with its high potential for recurrence and metastasis, is a major health burden. Uncovering proteins and pathways required for tumor cell growth is necessary for the development of novel targeted therapies. Ajuba is a member of the LIM domain family of proteins whose expression is positively associated with numerous cancers. Our data shows that Ajuba is highly expressed in human colon cancer tissue and cell lines. Publicly available data from The Cancer Genome Atlas shows a negative correlation between survival and Ajuba expression in patients with colon cancer. To investigate its function, we transduced SW480 human colon cancer cells, with lentiviral constructs to knockdown or overexpress Ajuba protein. The transcriptome of the modified cell lines was analyzed by RNA sequencing. Among the pathways enriched in the differentially expressed genes, were cell proliferation, migration and differentiation. We confirmed our sequencing data with biological assays; cells depleted of Ajuba were less proliferative, more sensitive to irradiation, migrated less and were less efficient in colony formation. In addition, loss of Ajuba expression decreased the tumor burden in a murine model of colorectal metastasis to the liver. Taken together, our data supports that Ajuba promotes colon cancer growth, migration and metastasis and therefore is a potential candidate for targeted therapy.
Collapse
Affiliation(s)
- Noëlle Dommann
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Daniel Sánchez-Taltavull
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Linda Eggs
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Fabienne Birrer
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Tess Brodie
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Lilian Salm
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Felix Alexander Baier
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Michaela Medová
- Department of Biomedical Research, Radiation Oncology, University of Bern, Bern University Hospital, 3008 Bern, Switzerland;
| | - Magali Humbert
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (M.H.); (M.P.T.)
| | - Mario P. Tschan
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; (M.H.); (M.P.T.)
| | - Guido Beldi
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Daniel Candinas
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
| | - Deborah Stroka
- Department of Biomedical Research, Visceral and Transplantation Surgery, University of Bern, Clinic of Visceral Surgery and Medicine, Bern University Hospital, 3008 Bern, Switzerland; (N.D.); (D.S.-T.); (L.E.); (F.B.); (T.B.); (L.S.); (F.A.B.); (G.B.); (D.C.)
- Correspondence: ; Tel.: +41-31-632-27-48
| |
Collapse
|
32
|
Song J, Song M. Identification of hydatidosis-related modules and key regulatory genes. PeerJ 2020; 8:e9280. [PMID: 32596042 PMCID: PMC7306218 DOI: 10.7717/peerj.9280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 05/12/2020] [Indexed: 12/15/2022] Open
Abstract
Background Echinococcosis caused by larval of Echinococcus is prevalent all over the world. Although clinical experience showed that the presence of tapeworms could not be found in liver lesions, the repeated infection and aggravation of lesions still occur in the host. Here, this study constructed a multifactor-driven disease-related dysfunction network to explore the potential molecular pathogenesis mechanism in different hosts after E.multilocularis infection. Method First, iTRAQ sequencing was performed on human liver infected with E.multilocularis. Second, obtained microRNAs(miRNAs) expression profiles of humans and canine infected with Echinococcus from the GEO database. In addition, we also performed differential expression analysis, protein interaction network analysis, enrichment analysis, and crosstalk analysis to obtain genes and modules related to E.multilocularis infection. Pivot analysis is used to calculate the potential regulatory effects of multiple factors on the module and identify related non-coding RNAs(ncRNAs) and transcription factors(TFs). Finally, we screened the target genes of miRNAs of Echinococcus to further explore its infection mechanism. Results A total of 267 differentially expressed proteins from humans and 3,635 differentially expressed genes from canine were obtained. They participated in 16 human-related dysfunction modules and five canine-related dysfunction modules, respectively. Both human and canine dysfunction modules are significantly involved in BMP signaling pathway and TGF-beta signaling pathway. In addition, pivot analysis found that 1,129 ncRNAs and 110 TFs significantly regulated human dysfunction modules, 158 ncRNAs and nine TFs significantly regulated canine dysfunction modules. Surprisingly, the Echinococcus miR-184 plays a role in the pathogenicity regulation by targeting nine TFs and one ncRNA in humans. Similarly, miR-184 can also cause physiological dysfunction by regulating two transcription factors in canine. Conclusion The results show that the miRNA-184 of Echinococcus can regulate the pathogenic process through various biological functions and pathways. The results laid a solid theoretical foundation for biologists to further explore the pathogenic mechanism of Echinococcosis.
Collapse
Affiliation(s)
- Jijun Song
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China.,Harbin Weike Biotechnology Co., Ltd., Harbin Veterinary Research Institute, CAAS, Harbin, Heilongjiang, China
| | - Mingxin Song
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China.,Heilongjiang Key Laboratory for Zoonosis, Harbin, China
| |
Collapse
|
33
|
Ma J, Li M, Chai J, Wang K, Li P, Liu Y, Zhao D, Xu J, Yu K, Yan Q, Guo S, Wang Z, Fan L. Expression of RSK4, CD44 and MMP-9 is upregulated and positively correlated in metastatic ccRCC. Diagn Pathol 2020; 15:28. [PMID: 32209138 PMCID: PMC7093975 DOI: 10.1186/s13000-020-00948-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 03/19/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND To investigate the expression and function of RSK4, MMP-9 and CD44 in primary clear cell renal cell carcinoma (primary ccRCC) and metastatic clear cell renal cell carcinoma (metastatic ccRCC), as well as the correlation with clinicopathological features of patients. METHOD The expression levels of RSK4, CD44 and MMP-9 in 52 primary ccRCC samples and 48 metastatic ccRCC samples were detected by immunohistochemistry, and the relationship between RSK4, CD44 and MMP-9 expression and clinicopathological features as well as prognosis of metastatic ccRCC patients was statistically analysed. Ectopic RSK4 expression in ccRCC cell lines was performed to determine its effect on cell cycle regulation, tumour invasiveness, and metastatic capability. RESULTS The positive rates of RSK4, MMP-9 and CD44 expression in metastatic ccRCC tissues were 75, 68.75 and 91.7%, respectively, while the rates in primary ccRCC tissues were 44.2, 34.6 and 69.2%, respectively. Thus, the positive rates in metastatic ccRCC were higher than those in primary ccRCC (PRSK4 = 0. 002; PMMP-9 = 0. 002; PCD44 = 0. 001). However, the expression of RSK4, CD44 and MMP-9 was unrelated to age, gender, or metastatic sites (P > 0.05) but was related to WHO/ISUP nucleolar grade (PRSK4 = 0.019; PCD44 = 0.026; PMMP-9 = 0.049). In metastatic ccRCC, expression among the three proteins showed a positive correlation (P = 0.008). Moreover, expression between RSK4 and CD44 (P = 0.019) and MMP-9 and CD44 (P = 0.05) also showed positive correlations, whereas RSK4 and MMP-9 showed no significant correlation (P = 1.00). Molecular studies showed that overexpression of RSK4 could enhance the invasive and migratory abilities of ccRCC cell lines through the regulation of CD44 and MMP-9 expression and vice versa. CONCLUSIONS The overexpression of RSK4, MMP-9 and CD44 is associated with the invasion and metastasis of ccRCC, indicating that they could be potential prognostic factors and serve as new potential therapeutic targets for ccRCC.
Collapse
Affiliation(s)
- Jing Ma
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Jia Chai
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Kaijing Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Peifeng Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
- Department of Pathology, The 960th Hospital of PLA, Jinan, Shandong China
| | - Yixiong Liu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Danhui Zhao
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Junpeng Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Kangjie Yu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Qingguo Yan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Shuangping Guo
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Zhe Wang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| | - Linni Fan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Changle West Road #169, Xi’an, 710032 Shaan Xi Province China
| |
Collapse
|
34
|
Al-Attar R, Childers CL, Nguyen VC, Pamenter ME, Storey KB. Differential protein phosphorylation is responsible for hypoxia-induced regulation of the Akt/mTOR pathway in naked mole rats. Comp Biochem Physiol A Mol Integr Physiol 2020; 242:110653. [PMID: 31926299 DOI: 10.1016/j.cbpa.2020.110653] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/25/2019] [Accepted: 01/06/2020] [Indexed: 12/21/2022]
Abstract
Naked mole rats (NMRs, Heterocephalus glaber) are among the most hypoxia-tolerant mammals known. They can reduce their metabolic rate (>85%) under severe hypoxia, remain moderately active and recover with no obvious signs of damage. Hence, NMRs are an excellent model for studying mammalian hypoxia tolerance. The current study characterized the involvement of posttranslational modifications in regulating the Akt/mTOR pathway that regulates protein synthesis, and the responses of key ribosomal proteins in order to assess tissue-specific responses to 4 h exposure to 7% O2 (compared to controls at 21% O2). Results showed a tissue-specific regulation of the Akt/mTOR pathway via differential phosphorylation. Relative amounts of p-TSC(S939) in brain and of p-TSC(S939), p-Akt(473) and p-PTEN(S380) in liver increased under hypoxia, whereas levels of IGF1R(Y1135/1136) in liver decreased. In skeletal muscle, levels of p-Akt(S473) and p-PTEN(S380) decreased during hypoxia, whereas lungs showed an increase in p-mTOR(S2884) content but a decrease in p-RPS6(S235-236) under the same conditions. Analysis of the phosphorylation states of ribosomal proteins revealed increases in p-4E-BP1(T37/46) content in brain and lungs under hypoxia, as well as a rise in total 4E-BP1 protein level in liver. Phosphorylated eIF-4B(S422) content also increased in liver while levels of p-eIF-2α(S51), and eIF-4E(S209) decreased during hypoxia in liver. Overall, hypoxia altered the Akt/mTOR pathway, which correlated with a general decrease in activity of the ribosomal protein biosynthesis machinery in muscle, lung, and brain of NMRs. However, the increase in eIF-4B in liver suggests the potential promotion of cap-independent mRNA translation mechanism operating under hypoxic stress.
Collapse
Affiliation(s)
- Rasha Al-Attar
- Department of Biology, Carleton University, Ottawa, ON, Canada
| | | | - Vu C Nguyen
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Matthew E Pamenter
- Department of Biology, University of Ottawa, Ottawa, ON, Canada; uOttawa Brain and Mind Research Institute, Canada
| | | |
Collapse
|
35
|
Yoshikawa M, Morifuji T, Matsumoto T, Maeshige N, Tanaka M, Fujino H. Effects of combined treatment with blood flow restriction and low-current electrical stimulation on muscle hypertrophy in rats. J Appl Physiol (1985) 2019; 127:1288-1296. [PMID: 31556832 DOI: 10.1152/japplphysiol.00070.2019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
This study aimed to clarify the effects of a combined treatment comprising blood flow restriction and low-current electrical stimulation on skeletal muscle hypertrophy in rats. Male Wistar rats were divided into control (Cont), blood flow restriction (Bfr), electrical stimulation (Es), or Bfr with Es (Bfr + Es) groups. Pressure cuffs (80 mmHg) were placed around the thighs of Bfr and Bfr + Es rats. Low-current Es was applied to calf muscles in the Es and Bfr + Es rats. In experiment 1, a 1-day treatment regimen (5-min stimulation, followed by 5-min rest) was delivered four times to study the acute effects. In experiment 2, the same treatment regimen was delivered three times/wk for 8 wk. Body weight, muscle mass, changes in maximal isometric contraction, fiber cross-sectional area of the soleus muscle, expression of phosphorylated and total-ERK1/2, phosphorylated-rpS6 Ser235/236, phosphorylated and total Akt, and phosphorylated-rpS6 Ser240/244 were measured. Bfr and Es treatment alone failed to induce muscle hypertrophy and increase the expression of phosphorylated rpS6 Ser240/244. Combined Bfr + Es upregulated muscle mass, increased the fiber cross-sectional area, and increased phosphorylated rpS6 Ser240/244 expression and phosphorylated rpS6 Ser235/236 expression compared with controls. Combined treatment with Bfr and low-current Es can induce muscle hypertrophy via activation of two protein synthesis signaling pathways. This treatment should be introduced for older patients with sarcopenia and others with muscle weakness.NEW & NOTEWORTHY We investigated the acute and chronic effect of low-current electrical stimulation with blood flow restriction on skeletal muscle hypertrophy and the mechanisms controlling the hypertrophic response. Low-current electrical stimulation could not induce skeletal muscle hypertrophy, but a combination treatment did. Blood lactate and growth hormone levels were increased in the early response. Moreover, activation of ERK1/2 and mTOR pathways were observed in both the acute and chronic response, which contribute to muscle hypertrophy.
Collapse
Affiliation(s)
- Madoka Yoshikawa
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Takeshi Morifuji
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan.,Department of Rehabilitation Science, Osaka Kawasaki Rehabilitation University, Kaizuka, Japan
| | - Tomohiro Matsumoto
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Noriaki Maeshige
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan
| | - Minoru Tanaka
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan.,Department of Rehabilitation Science, Osaka Health Science University, Osaka, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Science, Kobe, Japan
| |
Collapse
|
36
|
Berndsen K, Lis P, Yeshaw WM, Wawro PS, Nirujogi RS, Wightman M, Macartney T, Dorward M, Knebel A, Tonelli F, Pfeffer SR, Alessi DR. PPM1H phosphatase counteracts LRRK2 signaling by selectively dephosphorylating Rab proteins. eLife 2019; 8:e50416. [PMID: 31663853 PMCID: PMC6850886 DOI: 10.7554/elife.50416] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/30/2019] [Indexed: 12/19/2022] Open
Abstract
Mutations that activate LRRK2 protein kinase cause Parkinson's disease. LRRK2 phosphorylates a subset of Rab GTPases within their Switch-II motif controlling interaction with effectors. An siRNA screen of all human protein phosphatases revealed that a poorly studied protein phosphatase, PPM1H, counteracts LRRK2 signaling by specifically dephosphorylating Rab proteins. PPM1H knockout increased endogenous Rab phosphorylation and inhibited Rab dephosphorylation in human A549 cells. Overexpression of PPM1H suppressed LRRK2-mediated Rab phosphorylation. PPM1H also efficiently and directly dephosphorylated Rab8A in biochemical studies. A "substrate-trapping" PPM1H mutant (Asp288Ala) binds with high affinity to endogenous, LRRK2-phosphorylated Rab proteins, thereby blocking dephosphorylation seen upon addition of LRRK2 inhibitors. PPM1H is localized to the Golgi and its knockdown suppresses primary cilia formation, similar to pathogenic LRRK2. Thus, PPM1H acts as a key modulator of LRRK2 signaling by controlling dephosphorylation of Rab proteins. PPM1H activity enhancers could offer a new therapeutic approach to prevent or treat Parkinson's disease.
Collapse
Affiliation(s)
- Kerryn Berndsen
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Pawel Lis
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Wondwossen M Yeshaw
- Department of BiochemistryStanford University School of MedicineStanfordUnited States
| | - Paulina S Wawro
- Department of BiochemistryStanford University School of MedicineStanfordUnited States
| | - Raja S Nirujogi
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Melanie Wightman
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Thomas Macartney
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Mark Dorward
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Axel Knebel
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Francesca Tonelli
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| | - Suzanne R Pfeffer
- Department of BiochemistryStanford University School of MedicineStanfordUnited States
| | - Dario R Alessi
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life SciencesUniversity of DundeeDundeeUnited Kingdom
| |
Collapse
|
37
|
Essential role of GEXP15, a specific Protein Phosphatase type 1 partner, in Plasmodium berghei in asexual erythrocytic proliferation and transmission. PLoS Pathog 2019; 15:e1007973. [PMID: 31348803 PMCID: PMC6685639 DOI: 10.1371/journal.ppat.1007973] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 08/07/2019] [Accepted: 07/10/2019] [Indexed: 12/21/2022] Open
Abstract
The essential and distinct functions of Protein Phosphatase type 1 (PP1) catalytic subunit in eukaryotes are exclusively achieved through its interaction with a myriad of regulatory partners. In this work, we report the molecular and functional characterization of Gametocyte EXported Protein 15 (GEXP15), a Plasmodium specific protein, as a regulator of PP1. In vitro interaction studies demonstrated that GEXP15 physically interacts with PP1 through the RVxF binding motif in P. berghei. Functional assays showed that GEXP15 was able to increase PP1 activity and the mutation of the RVxF motif completely abolished this regulation. Immunoprecipitation assays of tagged GEXP15 or PP1 in P. berghei followed by immunoblot or mass spectrometry analyses confirmed their interaction and showed that they are present both in schizont and gametocyte stages in shared protein complexes involved in the spliceosome and proteasome pathways and known to play essential role in parasite development. Phenotypic analysis of viable GEXP15 deficient P. berghei blood parasites showed that they were unable to develop lethal infection in BALB/c mice or to establish experimental cerebral malaria in C57BL/6 mice. Further, although deficient parasites produced gametocytes they did not produce any oocysts/sporozoites indicating a high fitness cost in the mosquito. Global proteomic and phosphoproteomic analyses of GEXP15 deficient schizonts revealed a profound defect with a significant decrease in the abundance and an impact on phosphorylation status of proteins involved in regulation of gene expression or invasion. Moreover, depletion of GEXP15 seemed to impact mainly the abundance of some specific proteins of female gametocytes. Our study provides the first insight into the contribution of a PP1 regulator to Plasmodium virulence and suggests that GEXP15 affects both the asexual and sexual life cycle. In the absence of an effective vaccine and the emerging resistance to artemisinin combination therapy, malaria is still a significant threat to human health. Increasing our understanding of the specific mechanisms of the biology of Plasmodium is essential to propose new strategies to control this infection. Here, we demonstrated that GEXP15, a specific protein in Plasmodium, was able to interact with the Protein Phosphatase 1 and regulate its activity. We showed that both proteins are implicated in common protein complexes involved in the mRNA splicing and proteasome pathways. We reported that the deletion of GEXP15 leads to a loss of parasite virulence during asexual stages and a total abolishment of the capacity of deficient parasites to develop in the mosquito. We also found that this deletion affects both protein phosphorylation status and significantly decreases the expression of essential proteins in schizont and gametocyte stages. This study characterizes for the first time a novel molecular pathway through the control of PP1 by an essential and specific Plasmodium regulator, which may contribute to the discovery of new therapeutic targets to control malaria.
Collapse
|
38
|
Romeo V, Gierke S, Edgar KA, Liu SD. Effects of PI3K Inhibition on Afucosylated Antibody-Driven FcγRIIIa Events and Phospho-S6 Activity in NK Cells. THE JOURNAL OF IMMUNOLOGY 2019; 203:137-147. [PMID: 31092639 DOI: 10.4049/jimmunol.1801418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 04/20/2019] [Indexed: 11/19/2022]
Abstract
PI3K is one of the most frequently mutated genes in cancers and has been the target of numerous anticancer therapies. With the additional development of therapeutics that mobilize the immune system, such as Abs with effector functions, bispecific Abs, and checkpoint inhibitors, many small molecule inhibitors that target PI3K are being combined with these immunomodulatory treatments. However, the PI3K pathway is also essential for lymphocyte function, and the presence of the PI3K inhibitor may render the immunomodulatory therapeutic ineffective in these combinatorial treatments. Therefore, therapeutics with enhanced activity, such as afucosylated Abs, which promote signaling and function, may be ideal in these types of treatments to offset the negative effect of PI3K inhibitors on immune cell function. Indeed, we show that afucosylated Abs can counterbalance these inhibitory effects on FcγRIIIa-driven signaling in human NK cells to produce signals similar to cells treated only with fucosylated Ab. Furthermore, NK cell activation, degranulation, chemokine/cytokine production, and Ab-dependent cellular cytotoxicity were similar between inhibitor-treated, afucosylated Ab-stimulated NK cells and cells activated only with its fucosylated counterpart. To our knowledge, these studies also identified a previously undefined role for phospho-S6 in human NK cells. In this study, a kinetic delay in PI3K-driven phosphorylation of S6 was observed to control transcription of the temporally regulated production of IFN-γ and TNF-α but not MIP-1α, MIP-1β, and RANTES. Together, these studies demonstrate the importance of the PI3K pathway for S6 phosphorylation in human NK cells and the need to combine PI3K inhibitors with therapeutic molecules that enhance immunomodulatory function for anticancer therapies.
Collapse
Affiliation(s)
- Valentina Romeo
- Department of Cancer Immunology, Genentech, Inc., South San Francisco, CA 94080
| | - Sarah Gierke
- Department of Pathology, Genentech, Inc., South San Francisco, CA 94080; and
| | - Kyle A Edgar
- Department of Translation Oncology, Genentech, Inc., South San Francisco, CA 94080
| | - Scot D Liu
- Department of Pathology, Genentech, Inc., South San Francisco, CA 94080; and
| |
Collapse
|
39
|
Paskaš S, Krajnović T, Basile MS, Dunđerović D, Cavalli E, Mangano K, Mammana S, Al-Abed Y, Nicoletti F, Mijatović S, Maksimović-Ivanić D. Senescence as a main mechanism of Ritonavir and Ritonavir-NO action against melanoma. Mol Carcinog 2019; 58:1362-1375. [PMID: 30997718 DOI: 10.1002/mc.23020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/26/2019] [Accepted: 04/01/2019] [Indexed: 12/19/2022]
Abstract
The main focus of this study is exploring the effect and mechanism of two HIV-protease inhibitors: Ritonavir and Ritonavir-nitric oxide (Ritonavir-NO) on in vitro growth of melanoma cell lines. NO modification significantly improved the antitumor potential of Ritonavir, as the IC50 values of Ritonavir-NO were approximately two times lower than IC50 values of the parental compound. Our results showed for the first time, that both compounds induced senescence in primary and metastatic melanoma cell lines. This transformation was manifested as a change in cell morphology, enlargement of nuclei, increased cellular granulation, upregulation of β-galactosidase activity, lipofuscin granules appearance, higher production of reactive oxygen species and persistent inhibition of proliferation. The expression of p53, as one of the key regulators of senescence, was upregulated after 48 hours of Ritonavir-NO treatment only in metastatic B16F10 cells, ranking it as a late-response event. The development of senescent phenotype was consistent with the alteration of the cytoskeleton-as we observed diminished expression of vinculin, α-actin, and β-tubulin. Permanent inhibition of S6 protein by Ritonavir-NO, but not Ritonavir, could be responsible for a stronger antiproliferative potential of the NO-modified compound. Taken together, induction of senescent phenotype may provide an excellent platform for developing therapeutic approaches based on selective killing of senescent cells.
Collapse
Affiliation(s)
- Svetlana Paskaš
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| | - Tamara Krajnović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| | - Maria S Basile
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia.,Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Duško Dunđerović
- Institute of Pathology, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Eugenio Cavalli
- Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Santa Mammana
- Department of Experimental Neurology, IRCCS Centro Neurolesi "Bonino-Pulejo", Messina, Italy
| | - Yousef Al-Abed
- Center for Molecular Innovation, The Feinstein Institute for Medical Research, Manhasset, New York
| | - Ferdinando Nicoletti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", Belgrade University, Belgrade, Serbia
| |
Collapse
|
40
|
Tanaka M, Morifuji T, Yoshikawa M, Nakanishi R, Fujino H. Effects of combined treatment with blood flow restriction and low-intensity electrical stimulation on diabetes mellitus-associated muscle atrophy in rats. J Diabetes 2019; 11:326-334. [PMID: 30225988 DOI: 10.1111/1753-0407.12857] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 08/12/2018] [Accepted: 09/10/2018] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Diabetes mellitus (DM) results in decreased muscle mass and harmful complications. Blood flow restriction (Bfr) and electrical stimulation (ES) increase muscle protein synthesis. We hypothesized that combined Bfr and low-intensity ES may be more effective in preventing diabetes-associated muscle atrophy by inhibiting the downregulation of protein synthesis-related pathways. In this study, the effects of combined Bfr and low-intensity ES on diabetes-associated muscle atrophy were investigated by evaluating advanced glycation end-products (AGEs) and receptor for AGEs (RAGE) in rats. METHODS Twenty-four Goto-Kakizaki (GK) rats were randomly divided into four treatment groups: sedentary DM, DM + Bfr (pressure cuffs placed around the thigh), DM + ES, and DM + Bfr + ES. Six Wistar rats were used as an age-matched control. Levels of AGEs and the expression of RAGE, extracellular signal-regulated kinase (ERK), and ribosomal protein S6 (rpS6) were determined in plantaris muscles. RESULTS Diabetes resulted in a loss of muscle mass and upregulation of AGEs and RAGE in the plantaris muscle compared with the control group. Treatment with Bfr and ES alone failed to attenuate diabetes-associated loss of muscle mass, and could not prevent the upregulation of AGEs. However, the combination of Bfr and ES prevented the diabetes-associated decrease in muscle mass and upregulation of AGEs. In addition, the combination treatment prevented diabetes-associated decreases in the expression of phosphorylated rpS6. CONCLUSIONS Combination treatment with Bfr and ES may prevent diabetes-associated muscle atrophy by upregulating inhibition of AGEs, which leads to the activation of protein synthesis.
Collapse
Affiliation(s)
- Minoru Tanaka
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
- Department of Rehabilitation Science, Osaka Health Science University, Osaka, Japan
| | - Takeshi Morifuji
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
- Department of Rehabilitation, Osaka Kawasaki Rehabilitation University, Kaizuka, Japan
| | - Madoka Yoshikawa
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Ryosuke Nakanishi
- Department of Rehabilitation, Kobe International University, Kobe, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
41
|
Lu T, Zhu Z, Wu J, She H, Han R, Xu H, Qin ZH. DRAM1 regulates autophagy and cell proliferation via inhibition of the phosphoinositide 3-kinase-Akt-mTOR-ribosomal protein S6 pathway. Cell Commun Signal 2019; 17:28. [PMID: 30902093 PMCID: PMC6431019 DOI: 10.1186/s12964-019-0341-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 03/14/2019] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Macroautophagy (hereafter autophagy) is a tightly regulated process that delivers cellular components to lysosomes for degradation. Damage-regulated autophagy modulator 1 (DRAM1) induces autophagy and is necessary for p53-mediated apoptosis. However, the signalling pathways regulated by DRAM1 are not fully understood. METHODS HEK293T cells were transfected with FLAG-DRAM1 plasmid. Autophagic proteins (LC3 and p62), phosphorylated p53 and the phosphorylated proteins of the class I PI3K-Akt-mTOR-ribosomal protein S6 (rpS6) signalling pathway were detected with Western blot analysis. Cellular distribution of DRAM1 was determined with immunostaining. DRAM1 was knocked down in HEK293T cells using siRNA oligos which is confirmed by quantitative RT-PCR. Cells were serum starved for 18 h after overexpression or knockdown of DRAM1 to decrease the rpS6 activity to the basal level, and then the cells were stimulated with insulin growth factor, epidermal growth factor or serum. rpS6 phosphorylation and rpS6 were detected with Western blotting. Similarly, after overexpression or knockdown of DRAM1, phosphorylation of IGF-1Rβ and IGF-1R were examined with Western blotting. Cell viability was determined with CCK-8 assay and colony formation assay. Finally, human cancer cells Hela, SW480, and HCT116 were transfected with the FLAG-DRAM1 plasmid and phosphorylated rpS6 and rpS6 were detected with Western blot analysis. RESULTS DRAM1 induced autophagy and inhibited rpS6 phosphorylation in an mTORC1-dependent manner in HEK293T cells. DRAM1 didn't affect the phosphorylated and total levels of p53. Furthermore, DRAM1 inhibited the activation of the PI3K-Akt pathway stimulated with growth factors or serum. DRAM1 was localized at the plasma membrane and regulate the phosphorylation of IGF-1 receptor. DRAM1 decreased cell viability and colony numbers upon serum starvation. Additionally, DRAM1 inhibited rpS6 phosphorylation in several human cancer cells. CONCLUSIONS Here we provided evidence that DRAM1 inhibited rpS6 phosphorylation in multiple cell types. DRAM1 inhibited the phosphorylation of Akt and the activation of Akt-rpS6 pathway stimulated with growth factors and serum. Furthermore, DRAM1 regulated the activation of IGF-1 receptor. Thus, our results identify that the class I PI3K-Akt-rpS6 pathway is regulated by DRAM1 and may provide new insight into the potential role of DRAM1 in human cancers.
Collapse
Affiliation(s)
- Ting Lu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China
| | - Zhou Zhu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China
| | - Junchao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China
| | - Hua She
- Department of Pharmacology and Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rong Han
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China
| | - Haidong Xu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China.
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China.
| |
Collapse
|
42
|
TOR and RPS6 transmit light signals to enhance protein translation in deetiolating Arabidopsis seedlings. Proc Natl Acad Sci U S A 2018; 115:12823-12828. [PMID: 30482859 PMCID: PMC6294885 DOI: 10.1073/pnas.1809526115] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Light enhances the translation efficiency of thousands of mRNAs during photomorphogenic development in Arabidopsis, but the underlying molecular mechanism remains elusive. Here we show that light activates the auxin-target of rapamycin (TOR)-ribosome protein S6 (RPS6) pathway to enhance translation in deetiolating Arabidopsis. We discovered that CONSTITUTIVE PHOTOMORPHOGENIC 1 (COP1) represses TOR activity in dark-grown seedlings. The perception of far-red and blue light by photoreceptors inactivates COP1, which leads to the derepression of the auxin-TOR-RPS6 pathway and enhanced de novo protein synthesis. Our study revealed a light-triggered signaling pathway for translational regulation. This sophisticated regulation also functions to ensure that young seedlings have strict skotomorphogenic development in the dark and a timely switch to photomorphogenic development. Deetiolation is an essential developmental process transforming young plant seedlings into the vegetative phase with photosynthetic activities. Light signals initiate this important developmental process by triggering massive reprogramming of the transcriptome and translatome. Compared with the wealth of knowledge of transcriptional regulation, the molecular mechanism underlying this light-triggered translational enhancement remains unclear. Here we show that light-enhanced translation is orchestrated by a light perception and signaling pathway composed of photoreceptors, CONSTITUTIVE PHOTOMORPHOGENESIS 1 (COP1), the phytohormone auxin, target of rapamycin (TOR), and ribosomal protein S6 (RPS6). In deetiolating Arabidopsis seedlings, photoreceptors, including phytochrome A and cryptochromes, perceive far-red and blue light to inactivate the negative regulator COP1, which leads to activation of the auxin pathway for TOR-dependent phosphorylation of RPS6. Arabidopsis mutants defective in TOR, RPS6A, or RPS6B exhibited delayed cotyledon opening, a characteristic of the deetiolating process to ensure timely vegetative development of a young seedling. This study provides a mechanistic view of light-triggered translational enhancement in deetiolating Arabidopsis.
Collapse
|
43
|
Cromm PM, Samarasinghe KTG, Hines J, Crews CM. Addressing Kinase-Independent Functions of Fak via PROTAC-Mediated Degradation. J Am Chem Soc 2018; 140:17019-17026. [DOI: 10.1021/jacs.8b08008] [Citation(s) in RCA: 213] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Philipp M. Cromm
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
| | - Kusal T. G. Samarasinghe
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
| | - John Hines
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
| | - Craig M. Crews
- Department of Molecular, Cellular & Developmental Biology, Yale University, New Haven, Connecticut 06511, United States
- Department of Chemistry, Yale University, New Haven, Connecticut 06511, United States
- Department of Pharmacology, Yale University, New Haven, Connecticut 06511, United States
| |
Collapse
|
44
|
Cuperjani F, Gashi L, Kurshumliu F, Dreshaj S, Selimi F. Relationship between Ribosomal Protein S6-pS240 Expression and other Prognostic Factors in Non-Special Type Invasive Breast Cancer. Breast Care (Basel) 2018; 14:171-175. [PMID: 31316316 DOI: 10.1159/000491427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Background The aim of this study was to investigate the immunohistochemical expression of ribosomal protein (RP) S6-pS240 in non-special type invasive breast cancer in relation to other prognostic markers and gain new insights to facilitate more individualized treatment. Methods The following clinical and histopathological parameters of 120 patients were determined: S6-pS240 expression, age, menopausal status, tumor size and grade, TNM stage, Nottingham Prognostic Index (NPI), lymph node stage, estrogen and progesterone receptor (ER/PR) expression, HER2/neu amplification, lymphovascular invasion, and proliferative index as measured by Ki-67. Treatment protocol and disease-free survival were evaluated accordingly. Results Significant positive correlations were seen between S6-pS240 expression and Ki-67 values (rho = 0.530, p < 0.001), and NPI (rho = 0.370, p < 0.001) and HER2/neu amplification (rho = 0.368, p < 0.001). A negative correlation was found between S6-pS240 and ER/PR expression (rho = 0.362, p < 0.001). Patients with negative RP S6-pS240 expression had significantly longer disease-free survival (log-rank test, p = 0.005). Conclusion Immunohistochemical analysis of RP S6-pS240 is a valuable additional prognostic marker in patients with invasive breast cancer. Routine use of S6-pS240 immunohistochemistry is recommended.
Collapse
Affiliation(s)
- Frederik Cuperjani
- Thoracic Surgery Clinic, University Clinical Center Kosovo, Pristina, Kosovo
| | - Lumturije Gashi
- Pathology Institute, University Clinical Center Kosovo, Pristina, Kosovo
| | - Fisnik Kurshumliu
- Thoracic Surgery Clinic, University Clinical Center Kosovo, Pristina, Kosovo
| | - Shemsedin Dreshaj
- Infectious Diseases Clinic, University Clinical Center Kosovo, Pristina, Kosovo
| | - Fitim Selimi
- Thoracic Surgery Clinic, University Clinical Center Kosovo, Pristina, Kosovo
| |
Collapse
|
45
|
Pinkham C, Ahmed A, Bracci N, Narayanan A, Kehn-Hall K. Host-based processes as therapeutic targets for Rift Valley fever virus. Antiviral Res 2018; 160:64-78. [PMID: 30316916 DOI: 10.1016/j.antiviral.2018.10.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/27/2018] [Accepted: 10/05/2018] [Indexed: 12/28/2022]
Abstract
Rift Valley fever virus (RVFV) is an enveloped, segmented, negative sense RNA virus that replicates within the host's cytoplasm. To facilitate its replication, RVFV must utilize host cell processes and as such, these processes may serve as potential therapeutic targets. This review summarizes key host cell processes impacted by RVFV infection. Specifically the influence of RVFV on host transcriptional regulation, post-transcriptional regulation, protein half-life and availability, host signal transduction, trafficking and secretory pathways, cytoskeletal modulation, and mitochondrial processes and oxidative stress are discussed. Therapeutics targeted towards host processes that are essential for RVFV to thrive as well as their efficacy and importance to viral pathogenesis are highlighted.
Collapse
Affiliation(s)
- Chelsea Pinkham
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Aslaa Ahmed
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Nicole Bracci
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Aarthi Narayanan
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA
| | - Kylene Kehn-Hall
- National Center for Biodefense and Infectious Diseases, School of Systems Biology, George Mason University, Manassas, VA, USA.
| |
Collapse
|
46
|
Abel AM, Tiwari AA, Gerbec ZJ, Siebert JR, Yang C, Schloemer NJ, Dixon KJ, Thakar MS, Malarkannan S. IQ Domain-Containing GTPase-Activating Protein 1 Regulates Cytoskeletal Reorganization and Facilitates NKG2D-Mediated Mechanistic Target of Rapamycin Complex 1 Activation and Cytokine Gene Translation in Natural Killer Cells. Front Immunol 2018; 9:1168. [PMID: 29892299 PMCID: PMC5985319 DOI: 10.3389/fimmu.2018.01168] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 05/09/2018] [Indexed: 12/25/2022] Open
Abstract
Natural killer (NK) cells are innate lymphocytes that play essential roles in mediating antitumor immunity. NK cells respond to various inflammatory stimuli including cytokines and stress-induced cellular ligands which activate germline-encoded activation receptors (NKRs), such as NKG2D. The signaling molecules activated downstream of NKRs are well defined; however, the mechanisms that regulate these pathways are not fully understood. IQ domain-containing GTPase-activating protein 1 (IQGAP1) is a ubiquitously expressed scaffold protein. It regulates diverse cellular signaling programs in various physiological contexts, including immune cell activation and function. Therefore, we sought to investigate the role of IQGAP1 in NK cells. Development and maturation of NK cells from mice lacking IQGAP1 (Iqgap1-/- ) were mostly intact; however, the absolute number of splenic NK cells was significantly reduced. Phenotypic and functional characterization revealed a significant reduction in the egression of NK cells from the bone marrow of Iqagp1-/- mice altering their peripheral homeostasis. Lack of IQGAP1 resulted in reduced NK cell motility and their ability to mediate antitumor immunity in vivo. Activation of Iqgap1-/- NK cells via NKRs, including NKG2D, resulted in significantly reduced levels of inflammatory cytokines compared with wild-type (WT). This reduction in Iqgap1-/- NK cells is neither due to an impaired membrane proximal signaling nor a defect in gene transcription. The levels of Ifng transcripts were comparable between WT and Iqgap1-/- , suggesting that IQGAP1-dependent regulation of cytokine production is regulated by a post-transcriptional mechanism. To this end, Iqgap1-/- NK cells failed to fully induce S6 phosphorylation and showed significantly reduced protein translation following NKG2D-mediated activation, revealing a previously undefined regulatory function of IQGAP1 via the mechanistic target of rapamycin complex 1. Together, these results implicate IQGAP1 as an essential scaffold for NK cell homeostasis and function and provide novel mechanistic insights to the post-transcriptional regulation of inflammatory cytokine production.
Collapse
Affiliation(s)
- Alex M Abel
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States.,Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, United States
| | - Aradhana A Tiwari
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, United States
| | - Zachary J Gerbec
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States.,Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, United States
| | - Jason R Siebert
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States.,Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, United States
| | - Chao Yang
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States.,Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, United States
| | - Nathan J Schloemer
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Kate J Dixon
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States.,Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, United States
| | - Monica S Thakar
- Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Subramaniam Malarkannan
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI, United States.,Laboratory of Molecular Immunology and Immunotherapy, Blood Research Institute, Milwaukee, WI, United States.,Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
47
|
Jiang X, Feng S, Chen Y, Feng Y, Deng H. Proteomic analysis of mTOR inhibition-mediated phosphorylation changes in ribosomal proteins and eukaryotic translation initiation factors. Protein Cell 2018; 7:533-7. [PMID: 27278278 PMCID: PMC4930769 DOI: 10.1007/s13238-016-0279-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Xu Jiang
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Shan Feng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuling Chen
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Yun Feng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Haiteng Deng
- MOE Key Laboratory of Bioinformatics, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
48
|
Levenga J, Wong H, Milstead RA, Keller BN, LaPlante LE, Hoeffer CA. AKT isoforms have distinct hippocampal expression and roles in synaptic plasticity. eLife 2017; 6:30640. [PMID: 29173281 PMCID: PMC5722612 DOI: 10.7554/elife.30640] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/21/2017] [Indexed: 12/20/2022] Open
Abstract
AKT is a kinase regulating numerous cellular processes in the brain, and mutations in AKT are known to affect brain function. AKT is indirectly implicated in synaptic plasticity, but its direct role has not been studied. Moreover, three highly related AKT isoforms are expressed in the brain, but their individual roles are poorly understood. We find in Mus musculus, each AKT isoform has a unique expression pattern in the hippocampus, with AKT1 and AKT3 primarily in neurons but displaying local differences, while AKT2 is in astrocytes. We also find isoform-specific roles for AKT in multiple paradigms of hippocampal synaptic plasticity in area CA1. AKT1, but not AKT2 or AKT3, is required for L-LTP through regulating activity-induced protein synthesis. Interestingly, AKT activity inhibits mGluR-LTD, with overlapping functions for AKT1 and AKT3. In summary, our studies identify distinct expression patterns and roles in synaptic plasticity for AKT isoforms in the hippocampus.
Collapse
Affiliation(s)
- Josien Levenga
- Institute for Behavioral Genetics, University of Colorado-Boulder, Boulder, United States.,Linda Crnic Institute, Aurora, United States
| | - Helen Wong
- Institute for Behavioral Genetics, University of Colorado-Boulder, Boulder, United States
| | - Ryan A Milstead
- Department of Integrative Physiology, University of Colorado-Boulder, Boulder, United States
| | - Bailey N Keller
- Institute for Behavioral Genetics, University of Colorado-Boulder, Boulder, United States
| | - Lauren E LaPlante
- Institute for Behavioral Genetics, University of Colorado-Boulder, Boulder, United States
| | - Charles A Hoeffer
- Institute for Behavioral Genetics, University of Colorado-Boulder, Boulder, United States.,Linda Crnic Institute, Aurora, United States.,Department of Integrative Physiology, University of Colorado-Boulder, Boulder, United States
| |
Collapse
|
49
|
A regulatable AAV vector mediating GDNF biological effects at clinically-approved sub-antimicrobial doxycycline doses. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 5:16027. [PMID: 27069954 PMCID: PMC4813607 DOI: 10.1038/mtm.2016.27] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/26/2016] [Accepted: 02/26/2016] [Indexed: 12/19/2022]
Abstract
Preclinical and clinical data stress the importance of pharmacologically-controlling glial cell line-derived neurotrophic factor (GDNF) intracerebral administration to treat PD. The main challenge is finding a combination of a genetic switch and a drug which, when administered at a clinically-approved dose, reaches the brain in sufficient amounts to induce a therapeutic effect. We describe a highly-sensitive doxycycline-inducible adeno-associated virus (AAV) vector. This vector allowed for the first time a longitudinal analysis of inducible transgene expression in the brain using bioluminescence imaging. To evaluate the dose range of GDNF biological activity, the inducible AAV vector (8.0 × 10(9) viral genomes) was injected in the rat striatum at four delivery sites and increasing doxycycline doses administered orally. ERK/Akt signaling activation as well as tyrosine hydroxylase downregulation, a consequence of long-term GDNF treatment, were induced at plasmatic doxycycline concentrations of 140 and 320 ng/ml respectively, which are known not to increase antibiotic-resistant microorganisms in patients. In these conditions, GDNF covered the majority of the striatum. No behavioral abnormalities or weight loss were observed. Motor asymmetry resulting from unilateral GDNF treatment only appeared with a 2.5-fold higher vector and a 13-fold higher inducer doses. Our data suggest that using the herein-described inducible AAV vector, biological effects of GDNF can be obtained in response to sub-antimicrobial doxycycline doses.
Collapse
|
50
|
Pallis M, Harvey T, Russell N. Phenotypically Dormant and Immature Leukaemia Cells Display Increased Ribosomal Protein S6 Phosphorylation. PLoS One 2016; 11:e0151480. [PMID: 26985829 PMCID: PMC4795744 DOI: 10.1371/journal.pone.0151480] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/29/2016] [Indexed: 11/23/2022] Open
Abstract
Mechanistic/mammalian target of rapamycin (mTOR) activity drives a number of key metabolic processes including growth and protein synthesis. Inhibition of the mTOR pathway promotes cellular dormancy. Since cells from patients with acute myeloid leukaemia (AML) can be phenotypically dormant (quiescent), we examined biomarkers of their mTOR pathway activity concurrently with Ki-67 and CD71 (indicators of cycling cells) by quantitative flow cytometry. Using antibodies to phosphorylated epitopes of mTOR (S2448) and its downstream targets ribosomal protein S6 (rpS6, S235/236) and 4E-BP1 (T36/45), we documented that these phosphorylations were negligible in lymphocytes, but evident in dormant as well as proliferating subsets of both mobilised normal stem cell harvest CD34+ cells and AML blasts. Although mTOR phosphorylation in AML blasts was lower than that of the normal CD34+ cells, p-4E-BP1 was 2.6-fold higher and p-rpS6 was 22-fold higher. Moreover, in contrast to 4E-BP1, rpS6 phosphorylation was higher in dormant than proliferating AML blasts, and was also higher in the immature CD34+CD38- blast subset. Data from the Cancer Genome Atlas show that rpS6 expression is associated with that of respiratory chain enzymes in AML. We conclude that phenotypic quiescence markers do not necessarily predict metabolic dormancy and that elevated rpS6 ser235/236 phosphorylation is characteristic of AML.
Collapse
Affiliation(s)
- Monica Pallis
- Clinical Haematology, Nottingham University Hospitals, Nottingham, United Kingdom
- * E-mail:
| | - Tamsin Harvey
- Department of Haematology, University of Nottingham, Nottingham, United Kingdom
| | - Nigel Russell
- Clinical Haematology, Nottingham University Hospitals, Nottingham, United Kingdom
- Department of Haematology, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|