1
|
Ciulla DA, Dranchak PK, Aitha M, van Neer RHP, Shah D, Tharakan R, Wilson KM, Wang Y, Braisted JC, Inglese J. A general assay platform to study protein pharmacology using ligand-dependent structural dynamics. Nat Commun 2025; 16:4342. [PMID: 40346061 PMCID: PMC12064818 DOI: 10.1038/s41467-025-59658-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
Drug design strategies represent a fundamental challenge in chemical biology that could benefit from the development of next-generation high-throughput assays. Here we demonstrate that structural dynamic changes induced by ligand binding can be transmitted to a sensor protein fused to a target protein terminus. Here, NanoLuc luciferase, used as the intact protein or its α-complementation peptide, was fused to seven proteins from distinct enzyme superfamilies resulting in sensitive ligand-dependent bioluminescent outputs. This finding allows a general non-competitive, function-independent, quantitative, isothermal gain-of-signal ligand binding readout. As applied to chemical library high throughput screening, we can observe multivariate pharmacologic outputs including cofactor-induced synergy in ligand binding, as well as an example of allosteric site binding. The structural dynamics response assay format described here can enable the investigation of proteins precluded from study due to cost-prohibitive, insensitive, or technically challenging assays, including from cell lysates containing endogenously expressed gene edited proteins.
Collapse
Affiliation(s)
- Daniel A Ciulla
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Patricia K Dranchak
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Mahesh Aitha
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Renier H P van Neer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Divia Shah
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Ravi Tharakan
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Kelli M Wilson
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Yuhong Wang
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - John C Braisted
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - James Inglese
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA.
- Metabolic Medicine Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
2
|
Irgit A, Kamıs R, Sever B, Tuyun AF, Otsuka M, Fujita M, Demirci H, Ciftci H. Structure and Dynamics of the ABL1 Tyrosine Kinase and Its Important Role in Chronic Myeloid Leukemia. Arch Pharm (Weinheim) 2025; 358:e70005. [PMID: 40346758 PMCID: PMC12064879 DOI: 10.1002/ardp.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/28/2025] [Accepted: 04/16/2025] [Indexed: 05/12/2025]
Abstract
Abelson (ABL1) tyrosine kinase is an essential component of non-receptor tyrosine kinases and is associated with numerous cellular processes, including differentiation and proliferation. The structural features of ABL1 include a distinct N-terminal cap region, a C-terminal tail, a bilobed kinase, SH2, and SH3 domains. These domains enable its engagement in several signaling cascades and dynamic control. The pathophysiology of chronic myeloid leukemia (CML) is mainly driven by the BCR-ABL1 oncoprotein, arising from dysregulation of ABL1 kinase, namely through its fusion to the breakpoint cluster region (BCR) gene. ABL1 is a crucial target in the treatment of CML as the BCR-ABL1 fusion causes uncontrolled cellular proliferation and resistance to apoptosis. Tyrosine kinase inhibitors (TKIs) targeting the ABL1 tyrosine kinase are playing a critical role in the treatment of CML through the inhibition of persistently activated signaling pathways mediated by the BCR-ABL1 fusion protein. The article examines the structural characteristics of ABL1, how they relate to CML, and the interactions between ABL1 and the current FDA-approved TKIs, emphasizing the kinase's critical function in carcinogenesis and its possible target status for tyrosine kinase inhibitors.
Collapse
MESH Headings
- Humans
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/chemistry
- Proto-Oncogene Proteins c-abl/chemistry
- Proto-Oncogene Proteins c-abl/metabolism
- Proto-Oncogene Proteins c-abl/antagonists & inhibitors
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/chemistry
- Animals
- Fusion Proteins, bcr-abl/metabolism
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Ayca Irgit
- Department of Molecular Biology and GeneticsKoc UniversityIstanbulTurkey
| | - Reyhan Kamıs
- Department of Molecular Biology and GeneticsKoc UniversityIstanbulTurkey
| | - Belgin Sever
- Department of Pharmaceutical Chemistry, Faculty of PharmacyAnadolu UniversityEskisehirTurkey
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Amaç Fatih Tuyun
- Department of Chemistry, Faculty of ScienceIstanbul University, FatihİstanbulTurkey
| | - Masami Otsuka
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
- Department of Drug DiscoveryScience Farm Ltd.KumamotoJapan
| | - Mikako Fujita
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Hasan Demirci
- Department of Molecular Biology and GeneticsKoc UniversityIstanbulTurkey
| | - Halilibrahim Ciftci
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life SciencesKumamoto UniversityKumamotoJapan
- Department of Drug DiscoveryScience Farm Ltd.KumamotoJapan
- Department of Molecular Biology and GeneticsBurdur Mehmet Akif Ersoy UniversityBurdurTurkey
- Department of Bioengineering SciencesIzmir Katip Celebi UniversityIzmirTurkey
| |
Collapse
|
3
|
Carreño-Flórez GP, Cuartas-López AM, Boudreau RL, Vicente-Manzanares M, Gallego-Gómez JC. Role of c-ABL in DENV-2 Infection and Actin Remodeling in Vero Cells. Int J Mol Sci 2025; 26:4206. [PMID: 40362443 PMCID: PMC12071696 DOI: 10.3390/ijms26094206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2025] [Revised: 04/19/2025] [Accepted: 04/26/2025] [Indexed: 05/15/2025] Open
Abstract
In this study, we address the role of c-ABL (cellular Abelson Tyr kinase) in the cytoskeletal rearrangements induced by DENV (Dengue virus) infection in mammalian cells. Using the specific inhibitor imatinib and targeted RNA interference, we show that c-ABL is necessary for viral entry and subsequent ENV (DENV envelope protein) accumulation in infected cells. In addition, c-ABL targeting attenuates F-actin reorganization induced by DENV infection. Together with the involvement of c-ABL in endothelial dysfunction induced by DENV and host secreted factors, our findings strongly suggest that c-ABL is a potential host-targeted antiviral that could control DENV infection and/or its evolution to more severe forms of the disease.
Collapse
Affiliation(s)
- Grace Paola Carreño-Flórez
- Translational Medicine Group—School of Medicine, Universidad de Antioquia, Medellín 050010, Colombia; (G.P.C.-F.); (A.M.C.-L.)
| | - Alexandra Milena Cuartas-López
- Translational Medicine Group—School of Medicine, Universidad de Antioquia, Medellín 050010, Colombia; (G.P.C.-F.); (A.M.C.-L.)
| | - Ryan L. Boudreau
- Department of Internal Medicine and Abboud Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Miguel Vicente-Manzanares
- Molecular Mechanisms Program, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-University of Salamanca, 37007 Salamanca, Spain
| | - Juan Carlos Gallego-Gómez
- Translational Medicine Group—School of Medicine, Universidad de Antioquia, Medellín 050010, Colombia; (G.P.C.-F.); (A.M.C.-L.)
| |
Collapse
|
4
|
Weeratunga P, Harman RM, Jager MC, Van de Walle GR. Footprint-free induced pluripotent stem cells can be successfully differentiated into mesenchymal stromal cells in the feline model. Stem Cell Res Ther 2025; 16:195. [PMID: 40254569 PMCID: PMC12010622 DOI: 10.1186/s13287-025-04325-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Induced pluripotent stem cells (iPSCs) can propagate indefinitely and give rise to every other cell type, rendering them invaluable for disease modelling, drug development research, and usage in regenerative medicine. While feline iPSCs have been described, there are currently no reports on generating genome integration (footprint)-free iPSCs from domestic cats. Therefore, the objective of this study was to generate feline iPSCs from fetal fibroblasts using non-integrative Sendai virus (SeV) vectors carrying human transcription factors. Moreover, these iPSCs were differentiated into mesenchymal stromal cells (MSCs), which can be used as an alternative to tissue-derived MSCs. METHODS Feline fetal fibroblasts were transduced with CytoTune-iPS 2.0 Sendai Reprogramming vectors at recommended multiplicity of infections (MOI) and cultured for about 6 days. At 7 days post transduction cells were dissociated, replated on inactivated feeder cells and maintained in iPSC medium for 28 days with daily medium change. Emerging iPSC colonies were mechanically passaged and transferred to fresh feeder cells and further passaged every 6-8 days. Four feline iPSC lines were generated, with two selected for further in-depth characterization. Feline iPSCs were then differentiated into MSCs using a serial plating strategy and an inhibitor of the transforming growth factor-β (TGF-β) type I receptor. RESULTS Feline iPSCs exhibited characteristic colony morphology, high nuclear-to-cytoplasmic ratio, positive alkaline phosphatase activity, and expressed feline OCT4, SOX2, and Nanog homeobox (NANOG) stem cell markers. Expression of SeV-derived transgenes decreased during passaging to be eventually lost from the host cells and feline iPSCs could be stably maintained for over 35 passages. Feline iPSCs differentiated into embryoid bodies in vitro and did not form fully differentiated teratomas; instead, they generated in vivo masses containing mesodermal tissue derivatives when injected into immunodeficient mice. Feline iPSC-derived MSCs were plastic adherent, displayed MSC-like morphology, expressed MSC-specific surface markers, and differentiated into cells from the mesodermal lineage in vitro. RNA deep sequencing identified 1,189 differentially expressed genes in feline iPSC-derived MSCs compared to feline iPSCs. CONCLUSION We demonstrated the generation of footprint-free iPSCs from domestic cats and their directed differentiation potential towards MSCs. These SeV-derived feline iPSCs and iPSC-derived MSCs will provide valuable models to study feline diseases and explore novel therapeutic strategies and can serve as translational models for human health, leading to increased knowledge on disease pathogenesis and improved therapeutic interventions.
Collapse
Affiliation(s)
- Prasanna Weeratunga
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14850, USA
| | - Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14850, USA
| | - Mason C Jager
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, 235 Hungerford Hill Road, Ithaca, NY, 14850, USA.
- Department of Veterinary Pathobiology, The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
5
|
Xiang L, Yang J, Rao J, Ma A, Liu C, Zhang Y, Huang A, Xie T, Xue H, Chen Z, Yuan J, Yan H. Integrating Machine Learning and Bulk and Single-Cell RNA Sequencing to Decipher Diverse Cell Death Patterns for Predicting the Prognosis of Neoadjuvant Chemotherapy in Breast Cancer. Int J Mol Sci 2025; 26:3682. [PMID: 40332226 PMCID: PMC12027272 DOI: 10.3390/ijms26083682] [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: 02/24/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Breast cancer (BRCA) continues to pose a serious risk to women's health worldwide. Neoadjuvant chemotherapy (NAC) is a critical treatment strategy. Nevertheless, the heterogeneity in treatment outcomes necessitates the identification of reliable biomarkers and prognostic models. Programmed cell death (PCD) pathways serve as a critical factor in tumor development and treatment response. However, the relationship between the diverse patterns of PCD and NAC in BRCA remains unclear. We integrated machine learning and multiple bioinformatics tools to explore the association between 19 PCD patterns and the prognosis of NAC within a cohort of 921 BRCA patients treated with NAC from seven multicenter cohorts. A prognostic risk model based on PCD-related genes (PRGs) was constructed and evaluated using a combination of 117 machine learning algorithms. Immune infiltration analysis, mutation analysis, pharmacological analysis, and single-cell RNA sequencing (scRNA-seq) were conducted to explore the genomic profile and clinical significance of these model genes in BRCA. Immunohistochemistry (IHC) was employed to validate the expression of select model genes (UGCG, BTG22, TNFRSF21, and MYB) in BRCA tissues. We constructed a PRGs prognostic risk model by using a signature comprising 20 PCD-related DEGs to forecast the clinical outcomes of NAC in BRCA patients. The prognostic model demonstrated excellent predictive accuracy, with a high concordance index (C-index) of 0.772, and was validated across multiple independent datasets. Our results demonstrated a strong association between the developed model and the survival prognosis, clinical pathological features, immune infiltration, tumor microenvironment (TME), gene mutations, and drug sensitivity of NAC for BRCA patients. Moreover, IHC studies further demonstrated that the expression of certain model genes in BRCA tissues was significantly associated with the efficacy of NAC and emerged as an autonomous predictor of outcomes influencing the outcome of patients. We are the first to integrate machine learning and bulk and scRNA-seq to decode various cell death mechanisms for the prognosis of NAC in BRCA. The developed unique prognostic model, based on PRGs, provides a novel and comprehensive strategy for predicting the NAC outcomes of BRCA patients. This model not only aids in understanding the mechanisms underlying NAC efficacy but also offers insights into personalized treatment strategies, potentially improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Honglin Yan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (L.X.); (J.Y.); (J.R.); (A.M.); (C.L.); (Y.Z.); (A.H.); (T.X.); (H.X.); (Z.C.); (J.Y.)
| |
Collapse
|
6
|
Jain AG, Cortes JE. Asciminib: the tyrosine kinase inhibitor with a unique mechanism of action. Expert Opin Pharmacother 2025; 26:677-684. [PMID: 40087828 DOI: 10.1080/14656566.2025.2480762] [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: 01/26/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/17/2025]
Abstract
INTRODUCTION Management of chronic phase chronic myeloid leukemia (CML-CP) was revolutionized with the development of tyrosine kinase inhibitors (TKIs). Imatinib (first generation), dasatinib, nilotinib and bosutinib (second generation), and ponatinib (third generation) are the five approved TKIs that inhibit BCR::ABL1 by binding to the ATP binding site of ABL1. About half of the resistance to TKIs develops through acquisition of mutations in the ATP binding site, including T315I. Hence, a novel TKI with a distinct mechanism of action that inhibits bcr-abl1 by specifically targeting the ABL1 myristoyl pocket (STAMP inhibitor) was developed. AREAS COVERED Asciminib was first approved for treatment of CML-CP in the third line setting or beyond and in patients with T315I mutation in October, 2021. More recently, in October, 2024, asciminib was approved for newly diagnosed CML-CP based on ASC4FIRST data showing MMR rate of 67.7% in the asciminib arm compared to 49% in the investigator choice TKI arm (p < 0.001) at 48 weeks. In this review we detail the mechanism of action, preclinical data, clinical data, safety, and tolerability of asciminib. EXPERT OPINION Due to its mechanism of action, asciminib has fewer off-target effects, resulting in an improved safety and tolerability profile.
Collapse
MESH Headings
- Humans
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/administration & dosage
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Fusion Proteins, bcr-abl/antagonists & inhibitors
- Fusion Proteins, bcr-abl/genetics
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/adverse effects
- Animals
- Pyrazoles/pharmacology
- Pyrazoles/adverse effects
- Drug Resistance, Neoplasm
- Mutation
- Tyrosine Kinase Inhibitors
- Niacinamide/analogs & derivatives
Collapse
Affiliation(s)
- Akriti G Jain
- Leukemia and Myeloid Disorders, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
- Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Jorge E Cortes
- Georgia Cancer Center, Cecil F. Whitaker Jr. GRA Eminent Scholar Chair in Cancer, Augusta, GA, USA
| |
Collapse
|
7
|
Imani S, Aminnezhad S, Alikarami M, Abedi Z, Mosleh IS, Maghsoudloo M, Taheri Z. Exploration of drug repurposing for Mpox outbreaks targeting gene signatures and host-pathogen interactions. Sci Rep 2024; 14:29436. [PMID: 39604570 PMCID: PMC11603026 DOI: 10.1038/s41598-024-79897-9] [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: 09/05/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024] Open
Abstract
Monkeypox (Mpox) is a growing public health concern, with complex interactions within host systems contributing to its impact. This study employs multi-omics approaches to uncover therapeutic targets and potential drug repurposing opportunities to better understand Mpox's molecular pathogenesis. We developed an in silico host-pathogen interaction (HPI) network and applied weighted gene co-expression network analysis (WGCNA) to explore interactions between Mpox and host proteins. Subtype-specific host-pathogen protein-protein interaction networks were constructed, and key modules from the HPI and WGCNA were integrated to identify significant host proteins. To predict upstream signaling pathways and transcription factors, we used eXpression2Kinases and ChIP-X Enrichment Analysis. The multi-Steiner trees method was applied to compare our findings with those from FDA-approved antiviral drugs. Analysis of 55 differentially expressed genes in Mpox infection revealed 11 kinases and 15 transcription factors as key regulators. We identified 16 potential drug targets, categorized into 8 proviral genes (ESR2, ERK1, ERK2, P38, JNK1, CDK4, GSK3B, STAT3) designated for inhibition, and 8 antiviral genes (IKKA, HDAC1, HIPK2, TF65, CSK21, HIPK2, ESR2, GSK3B) designated for activation. Proviral genes are involved in the AKT, Wnt, and STAT3 pathways, while antiviral genes impact the AP-1, NF-κB, apoptosis, and IFN pathways. Promising FDA-approved candidates were identified, including kinase inhibitors, steroid hormone receptor agonists, STAT3 inhibitors, and notably Niclosamide. This study enhances our understanding of Mpox by identifying key therapeutic targets and potential repurposable drugs, providing a valuable framework for developing new treatments.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China.
| | - Sargol Aminnezhad
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Moslem Alikarami
- Research and Development Center, Dina Pharmed Exir Salamat Co, Tehran, Iran
| | - Zahra Abedi
- School of Biotechnology College of Science, University of Tehran, Tehran, Iran
| | - Iman Samei Mosleh
- Plant Functional Genomics Lab, Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Vienna, Austria
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, the Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zahra Taheri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
8
|
Ban F, Zhou L, Yang Z, Liu Y, Zhang Y. Aspergillusidone G Potentiates the Anti-Inflammatory Effects of Polaprezinc in LPS-Induced BV2 Microglia: A Bioinformatics and Experimental Study. Mar Drugs 2024; 22:324. [PMID: 39057433 PMCID: PMC11278036 DOI: 10.3390/md22070324] [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: 06/27/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Neuroinflammation is one of the main mechanisms involved in the progression of neurodegenerative diseases (NDs), and microglial activation is the main feature of neuroinflammation. Polaprezinc (Pol), a chelator of L-carnosine and zinc, is widely used as a clinical drug for gastric ulcers. However, its potential effects on NDs remain unexplored. In LPS-induced BV-2 microglia, we found that Pol reduced the generation of NO and ROS and revealed inhibited expression of iNOS, COX-2, and inflammatory factors such as IL-6, TNF-α, and 1L-1β by Pol using qRT-PCR and Western blotting. These effects were found to be associated with the suppression of the NF-κB signaling pathway. Moreover, we evaluated the potential synergistic effects of aspergillusidone G (Asp G) when combined with Pol. Remarkably, co-treatment with low doses of Asp G enhanced the NO inhibition by Pol from approximately 30% to 80% in LPS-induced BV2 microglia, indicating a synergistic anti-inflammatory effect. A bioinformatics analysis suggested that the synergistic mechanism of Asp G and Pol might be attributed to several targets, including NFκB1, NRF2, ABL1, TLR4, and PPARα. These findings highlight the anti-neuroinflammatory properties of Pol and its enhanced efficacy when combined with Asp G, proposing a novel therapeutic strategy for managing neuroinflammation in NDs.
Collapse
Affiliation(s)
- Fangfang Ban
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (F.B.); (Z.Y.); (Y.L.)
| | - Longjian Zhou
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (F.B.); (Z.Y.); (Y.L.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Zhiyou Yang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (F.B.); (Z.Y.); (Y.L.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Yayue Liu
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (F.B.); (Z.Y.); (Y.L.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| | - Yi Zhang
- Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Laboratory for Marine Biological Products, Guangdong Provincial Center for Modern Agricultural Scientific Innovation, Shenzhen Institute of Guangdong Ocean University, Zhanjiang Municipal Key Laboratory of Marine Drugs and Nutrition for Brain Health, Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China; (F.B.); (Z.Y.); (Y.L.)
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524088, China
- Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
9
|
Yao X, Chen R, Chen H, Koleske A, Xiao X. Impact of Abl2/Arg deficiency on anxiety and depressive behaviors in mice. Behav Brain Res 2024; 468:115022. [PMID: 38697301 DOI: 10.1016/j.bbr.2024.115022] [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: 02/11/2024] [Revised: 04/19/2024] [Accepted: 04/26/2024] [Indexed: 05/04/2024]
Abstract
Abl2/Arg (ABL-related gene) is a member of the Abelson family of nonreceptor tyrosine kinases, known for its role in tumor progression, metastasis, tissue injury responses, inflammation, neural degeneration, and other diseases. In this study, we developed Abl2/Arg knockout (abl2-/-) mice to explore its impact on sensory/motor functions and emotion-related behaviors. Our findings show that abl2-/- mice exhibit normal growth and phenotypic characteristics, closely resembling their wild-type (WT) counterparts. Behavioral tests, including the elevated plus maze, marble-burying behavior test, and open field test, indicated pronounced anxiety-like behaviors in abl2-/- mice compared to WT mice. Furthermore, in the tail suspension test, abl2-/- mice showed a significant decrease in mobility time, suggesting depressive-like behavior. Conversely, in the Y-maze and cliff avoidance reaction tests, no notable differences were observed between abl2-/- and WT mice, suggesting the absence of working memory deficits and impulsivity in abl2-/- mice. Proteomic analysis of the hippocampus in abl2-/- mice highlighted significant alterations in proteins related to anxiety and depression, especially those associated with the GABAergic synapse in inhibitory neurotransmission. The expression of Gabbr2 was significantly reduced in the hippocampus of abl2-/- compared to WT mice, and intraperitoneal treatment of GABA receptor agonist Gaboxadol normalized anxiety/depression-related behaviors of abl2-/- mice. These findings underscore the potential role of Abl2/Arg in influencing anxiety and depressive-like behaviors, thereby contributing valuable insights into its broader physiological and pathological functions.
Collapse
Affiliation(s)
- Xiaojuan Yao
- Institute of Science and Technology for Brain-Inspired Intelligence, Behavioral and Cognitive Neuroscience Center, Fudan University, Shanghai 200433, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Behavioral and Cognitive Neuroscience Center, Fudan University, Shanghai 200433, China
| | - Ruiying Chen
- Institute of Science and Technology for Brain-Inspired Intelligence, Behavioral and Cognitive Neuroscience Center, Fudan University, Shanghai 200433, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Behavioral and Cognitive Neuroscience Center, Fudan University, Shanghai 200433, China
| | - Hongting Chen
- Institute of Science and Technology for Brain-Inspired Intelligence, Behavioral and Cognitive Neuroscience Center, Fudan University, Shanghai 200433, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Behavioral and Cognitive Neuroscience Center, Fudan University, Shanghai 200433, China
| | - Anthony Koleske
- Departments of Molecular Biophysics and Biochemistry and Neuroscience, Yale University, New Haven, CT 06520, USA
| | - Xiao Xiao
- Institute of Science and Technology for Brain-Inspired Intelligence, Behavioral and Cognitive Neuroscience Center, Fudan University, Shanghai 200433, China; Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Ministry of Education, Behavioral and Cognitive Neuroscience Center, Fudan University, Shanghai 200433, China.
| |
Collapse
|
10
|
Ayub A, Hasan MK, Mahmud Z, Hossain MS, Kabir Y. Dissecting the multifaceted roles of autophagy in cancer initiation, growth, and metastasis: from molecular mechanisms to therapeutic applications. Med Oncol 2024; 41:183. [PMID: 38902544 DOI: 10.1007/s12032-024-02417-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
Autophagy is a cytoplasmic defense mechanism that cells use to break and reprocess their intracellular components. This utilization of autophagy is regarded as a savior in nutrient-deficient and other stressful conditions. Hence, autophagy keeps contact with and responds to miscellaneous cellular tensions and diverse pathways of signal transductions, such as growth signaling and cellular death. Importantly, autophagy is regarded as an effective tumor suppressor because regular autophagic breakdown is essential for cellular maintenance and minimizing cellular damage. However, paradoxically, autophagy has also been observed to promote the events of malignancies. This review discussed the dual role of autophagy in cancer, emphasizing its influence on tumor survival and progression. Possessing such a dual contribution to the malignant establishment, the prevention of autophagy can potentially advocate for the advancement of malignant transformation. In contrast, for the context of the instituted tumor, the agents of preventing autophagy potently inhibit the advancement of the tumor. Key regulators, including calpain 1, mTORC1, and AMPK, modulate autophagy in response to nutritional conditions and stress. Oncogenic mutations like RAS and B-RAF underscore autophagy's pivotal role in cancer development. The review also delves into autophagy's context-dependent roles in tumorigenesis, metastasis, and the tumor microenvironment (TME). It also discusses the therapeutic effectiveness of autophagy for several cancers. The recent implication of autophagy in the control of both innate and antibody-mediated immune systems made it a center of attention to evaluating its role concerning tumor antigens and treatments of cancer.
Collapse
Affiliation(s)
- Afia Ayub
- Department of Biochemistry and Molecular Biology, Tejgaon College, National University, Gazipur, 1704, Bangladesh
| | - Md Kamrul Hasan
- Department of Biochemistry and Molecular Biology, Tejgaon College, National University, Gazipur, 1704, Bangladesh.
- Department of Health Research Methods, Evidence, and Impact, McMaster University, 1280 Main St. W., Hamilton, L8S 4K1, Canada.
- Department of Public Health, North South University, Dhaka, Bangladesh.
| | - Zimam Mahmud
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| | - Md Sabbir Hossain
- Department of Biochemistry and Molecular Biology, Tejgaon College, National University, Gazipur, 1704, Bangladesh
| | - Yearul Kabir
- Department of Biochemistry and Molecular Biology, University of Dhaka, Dhaka, 1000, Bangladesh.
| |
Collapse
|
11
|
Schumacher JA, Wright ZA, Rufin Florat D, Anand SK, Dasyani M, Batta SPR, Laverde V, Ferrari K, Klimkaite L, Bredemeier NO, Gurung S, Koller GM, Aguera KN, Chadwick GP, Johnson RD, Davis GE, Sumanas S. SH2 domain protein E and ABL signaling regulate blood vessel size. PLoS Genet 2024; 20:e1010851. [PMID: 38190417 PMCID: PMC10798624 DOI: 10.1371/journal.pgen.1010851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/19/2024] [Accepted: 12/20/2023] [Indexed: 01/10/2024] Open
Abstract
Blood vessels in different vascular beds vary in size, which is essential for their function and fluid flow along the vascular network. Molecular mechanisms involved in the formation of a vascular lumen of appropriate size, or tubulogenesis, are still only partially understood. Src homology 2 domain containing E (She) protein was previously identified in a screen for proteins that interact with Abelson (Abl)-kinase. However, its biological role has remained unknown. Here we demonstrate that She and Abl signaling regulate vessel size in zebrafish embryos and human endothelial cell culture. Zebrafish she mutants displayed increased endothelial cell number and enlarged lumen size of the dorsal aorta (DA) and defects in blood flow, eventually leading to the DA collapse. Vascular endothelial specific overexpression of she resulted in a reduced diameter of the DA, which correlated with the reduced arterial cell number and lower endothelial cell proliferation. Chemical inhibition of Abl signaling in zebrafish embryos caused a similar reduction in the DA diameter and alleviated the she mutant phenotype, suggesting that She acts as a negative regulator of Abl signaling. Enlargement of the DA size in she mutants correlated with an increased endothelial expression of claudin 5a (cldn5a), which encodes a protein enriched in tight junctions. Inhibition of cldn5a expression partially rescued the enlarged DA in she mutants, suggesting that She regulates DA size, in part, by promoting cldn5a expression. SHE knockdown in human endothelial umbilical vein cells resulted in a similar increase in the diameter of vascular tubes, and also increased phosphorylation of a known ABL downstream effector CRKL. These results argue that SHE functions as an evolutionarily conserved inhibitor of ABL signaling and regulates vessel and lumen size during vascular tubulogenesis.
Collapse
Affiliation(s)
- Jennifer A. Schumacher
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, Ohio, United States of America
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - Zoë A. Wright
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Diandra Rufin Florat
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Surendra K. Anand
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Manish Dasyani
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Surya Prakash Rao Batta
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Valentina Laverde
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Kaitlin Ferrari
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Laurita Klimkaite
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Nina O. Bredemeier
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
| | - Suman Gurung
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| | - Gretchen M. Koller
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Kalia N. Aguera
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Griffin P. Chadwick
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - Riley D. Johnson
- Department of Biological Sciences, Miami University, Hamilton, Ohio, United States of America
| | - George E. Davis
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, Florida, United States of America
| | - Saulius Sumanas
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, Ohio, United States of America
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, Ohio, United States of America
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, Florida, United States of America
| |
Collapse
|
12
|
Niu ZX, Wang YT, Sun JF, Nie P, Herdewijn P. Recent advance of clinically approved small-molecule drugs for the treatment of myeloid leukemia. Eur J Med Chem 2023; 261:115827. [PMID: 37757658 DOI: 10.1016/j.ejmech.2023.115827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023]
Abstract
Myeloid leukemia denotes a hematologic malignancy characterized by aberrant proliferation and impaired differentiation of blood progenitor cells within the bone marrow. Despite the availability of several treatment options, the clinical outlook for individuals afflicted with myeloid leukemia continues to be unfavorable, making it a challenging disease to manage. Over the past, substantial endeavors have been dedicated to the identification of novel targets and the advancement of enhanced therapeutic modalities to ameliorate the management of this disease, resulting in the discovery of many clinically approved small-molecule drugs for myeloid leukemia, including histone deacetylase inhibitors, hypomethylating agents, and tyrosine kinase inhibitors. This comprehensive review succinctly presents an up-to-date assessment of the application and synthetic routes of clinically sanctioned small-molecule drugs employed in the treatment of myeloid leukemia. Additionally, it provides a concise exploration of the pertinent challenges and prospects encompassing drug resistance and toxicity. Overall, this review effectively underscores the considerable promise exhibited by clinically endorsed small-molecule drugs in the therapeutic realm of myeloid leukemia, while concurrently shedding light on the prospective avenues that may shape the future landscape of drug development within this domain.
Collapse
Affiliation(s)
- Zhen-Xi Niu
- Department of Pharmacy, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Ya-Tao Wang
- First People's Hospital of Shangqiu, Henan Province, Shangqiu, 476100, China; Department of Orthopedics, China-Japan Union Hospital, Jilin University, Changchun, 130033, China.
| | - Jin-Feng Sun
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, College of Pharmacy, Yanji, Jilin, 133002, China.
| | - Peng Nie
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| | - Piet Herdewijn
- Rega Institute for Medical Research, Medicinal Chemistry, KU Leuven, Herestraat 49-Box 1041, 3000, Leuven, Belgium.
| |
Collapse
|
13
|
Yeh TJ, Wang HC, Cho SF, Wu CC, Hsieh TY, Huang CT, Wang MH, Chuang TM, Gau YC, Du JS, Liu YC, Hsiao HH, Pan MR, Chen LT, Moi SH. The Prognosis Performance of a Neutrophil- and Lymphocyte-Associated Gene Mutation Score in a Head and Neck Cancer Cohort. Biomedicines 2023; 11:3113. [PMID: 38137334 PMCID: PMC10741104 DOI: 10.3390/biomedicines11123113] [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: 05/08/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
The treatment of head and neck squamous cell carcinomas (HNSCCs) is multimodal, and chemoradiotherapy (CRT) is a critical component. However, the availability of predictive or prognostic markers in patients with HNSCC is limited. Inflammation is a well-documented factor in cancer, and several parameters have been studied, with the neutrophil-to-lymphocyte ratio (NLR) being the most promising. The NLR is the most extensively researched clinical biomarker in various solid tumors, including HNSCC. In our study, we collected clinical and next-generation sequencing (NGS) data with targeted sequencing information from 107 patients with HNSCC who underwent CRT. The difference in the NLR between the good response group and the poor response group was significant, with more patients having a high NLR in the poor response group. We also examined the genetic alterations linked to the NLR and found a total of 41 associated genes across eight common pathways searched from the KEGG database. The overall mutation rate was low, and there was no significant mutation difference between the low- and high-NLR groups. Using a multivariate binomial generalized linear model, we identified three candidate genes (MAP2K2, MAP2K4, and ABL1) that showed significant results and were used to create a gene mutation score (GMS). Using the NLR-GMS category, we noticed that the high-NLR-GMS group had significantly shorter relapse-free survival compared to the intermediate- or low-NLR-GMS groups.
Collapse
Affiliation(s)
- Tsung-Jang Yeh
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Hui-Ching Wang
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Shih-Feng Cho
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chun-Chieh Wu
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Tzu-Yu Hsieh
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
| | - Chien-Tzu Huang
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Min-Hong Wang
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Tzer-Ming Chuang
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
| | - Yuh-Ching Gau
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Jeng-Shiun Du
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Yi-Chang Liu
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hui-Hua Hsiao
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (T.-J.Y.); (H.-C.W.); (S.-F.C.); (T.-Y.H.); (C.-T.H.); (M.-H.W.); (T.-M.C.); (Y.-C.G.); (J.-S.D.); (Y.-C.L.); (H.-H.H.)
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Mei-Ren Pan
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Li-Tzong Chen
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan
| | - Sin-Hua Moi
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Research Center for Precision Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
14
|
Martinez A, Lamaizon CM, Valls C, Llambi F, Leal N, Fitzgerald P, Guy C, Kamiński MM, Inestrosa NC, van Zundert B, Cancino GI, Dulcey AE, Zanlungo S, Marugan JJ, Hetz C, Green DR, Alvarez AR. c-Abl Phosphorylates MFN2 to Regulate Mitochondrial Morphology in Cells under Endoplasmic Reticulum and Oxidative Stress, Impacting Cell Survival and Neurodegeneration. Antioxidants (Basel) 2023; 12:2007. [PMID: 38001860 PMCID: PMC10669615 DOI: 10.3390/antiox12112007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/17/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
The endoplasmic reticulum is a subcellular organelle key in the control of synthesis, folding, and sorting of proteins. Under endoplasmic reticulum stress, an adaptative unfolded protein response is activated; however, if this activation is prolonged, cells can undergo cell death, in part due to oxidative stress and mitochondrial fragmentation. Here, we report that endoplasmic reticulum stress activates c-Abl tyrosine kinase, inducing its translocation to mitochondria. We found that endoplasmic reticulum stress-activated c-Abl interacts with and phosphorylates the mitochondrial fusion protein MFN2, resulting in mitochondrial fragmentation and apoptosis. Moreover, the pharmacological or genetic inhibition of c-Abl prevents MFN2 phosphorylation, mitochondrial fragmentation, and apoptosis in cells under endoplasmic reticulum stress. Finally, in the amyotrophic lateral sclerosis mouse model, where endoplasmic reticulum and oxidative stress has been linked to neuronal cell death, we demonstrated that the administration of c-Abl inhibitor neurotinib delays the onset of symptoms. Our results uncovered a function of c-Abl in the crosstalk between endoplasmic reticulum stress and mitochondrial dynamics via MFN2 phosphorylation.
Collapse
Affiliation(s)
- Alexis Martinez
- Cell Signaling Laboratory, Department of Cell and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Basal Center for Aging and Regeneration, Pontificia Universidad Católica de Chile (CARE UC), Santiago 8331150, Chile
| | - Cristian M. Lamaizon
- Cell Signaling Laboratory, Department of Cell and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Millennium Institute on Immunology and Immunotherapy, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Cristian Valls
- Cell Signaling Laboratory, Department of Cell and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Fabien Llambi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Nancy Leal
- Cell Signaling Laboratory, Department of Cell and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Patrick Fitzgerald
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Cliff Guy
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Marcin M. Kamiński
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Nibaldo C. Inestrosa
- Basal Center for Aging and Regeneration, Pontificia Universidad Católica de Chile (CARE UC), Santiago 8331150, Chile
- Center of Excellence in Biomedicine of Magallanes (CEBIMA), University of Magallanes, Punta Arenas 6210427, Chile
| | - Brigitte van Zundert
- Basal Center for Aging and Regeneration, Pontificia Universidad Católica de Chile (CARE UC), Santiago 8331150, Chile
- Institute of Biomedical Sciences, Faculty of Medicine & Faculty of Life Sciences, Universidad Andres Bello, Santiago 8370146, Chile
- Department of Neurology, University of Massachusetts Chan Medical School (UMMS), Worcester, MA 01655, USA
| | - Gonzalo I. Cancino
- Laboratory of Neurobiology, Department of Cell and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Andrés E. Dulcey
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Silvana Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O’Higgins 340, Santiago 8331150, Chile
| | - Juan J. Marugan
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago 8330015, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago 8380453, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago 8330015, Chile
- The Buck Institute for Research in Aging, Novato, CA 94945, USA
| | - Douglas R. Green
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Alejandra R. Alvarez
- Cell Signaling Laboratory, Department of Cell and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Basal Center for Aging and Regeneration, Pontificia Universidad Católica de Chile (CARE UC), Santiago 8331150, Chile
- Millennium Institute on Immunology and Immunotherapy, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| |
Collapse
|
15
|
Duan D, Lyu W, Chai P, Ma S, Wu K, Wu C, Xiong Y, Sestan N, Zhang K, Koleske AJ. Abl2 repairs microtubules and phase separates with tubulin to promote microtubule nucleation. Curr Biol 2023; 33:4582-4598.e10. [PMID: 37858340 PMCID: PMC10877310 DOI: 10.1016/j.cub.2023.09.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 07/07/2023] [Accepted: 09/06/2023] [Indexed: 10/21/2023]
Abstract
Abl family kinases are evolutionarily conserved regulators of cell migration and morphogenesis. Genetic experiments in Drosophila suggest that Abl family kinases interact functionally with microtubules to regulate axon guidance and neuronal morphogenesis. Vertebrate Abl2 binds to microtubules and promotes their plus-end elongation, both in vitro and in cells, but the molecular mechanisms by which Abl2 regulates microtubule (MT) dynamics are unclear. We report here that Abl2 regulates MT assembly via condensation and direct interactions with both the MT lattice and tubulin dimers. We find that Abl2 promotes MT nucleation, which is further facilitated by the ability of the Abl2 C-terminal half to undergo liquid-liquid phase separation (LLPS) and form co-condensates with tubulin. Abl2 binds to regions adjacent to MT damage, facilitates MT repair via fresh tubulin recruitment, and increases MT rescue frequency and lifetime. Cryo-EM analyses strongly support a model in which Abl2 engages tubulin C-terminal tails along an extended MT lattice conformation at damage sites to facilitate repair via fresh tubulin recruitment. Abl2Δ688-790, which closely mimics a naturally occurring splice isoform, retains binding to the MT lattice but does not bind tubulin, promote MT nucleation, or increase rescue frequency. In COS-7 cells, MT reassembly after nocodazole treatment is greatly slowed in Abl2 knockout COS-7 cells compared with wild-type cells, and these defects are rescued by re-expression of Abl2, but not Abl2Δ688-790. We propose that Abl2 locally concentrates tubulin to promote MT nucleation and recruits it to defects in the MT lattice to enable repair and rescue.
Collapse
Affiliation(s)
- Daisy Duan
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Wanqing Lyu
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Pengxin Chai
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Shaojie Ma
- Department of Neuroscience, Yale University, New Haven, CT 06510, USA
| | - Kuanlin Wu
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Chunxiang Wu
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Yong Xiong
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Nenad Sestan
- Department of Neuroscience, Yale University, New Haven, CT 06510, USA; Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT 06510, USA; Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06510, USA; Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale School of Medicine, New Haven, CT 06510, USA; Yale Child Study Center, Yale School of Medicine, New Haven, CT 06510, USA
| | - Kai Zhang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA; Department of Neuroscience, Yale University, New Haven, CT 06510, USA.
| |
Collapse
|
16
|
de Buhr S, Gräter F. Myristoyl's dual role in allosterically regulating and localizing Abl kinase. eLife 2023; 12:e85216. [PMID: 37843155 PMCID: PMC10619977 DOI: 10.7554/elife.85216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 10/15/2023] [Indexed: 10/17/2023] Open
Abstract
c-Abl kinase, a key signaling hub in many biological processes ranging from cell development to proliferation, is tightly regulated by two inhibitory Src homology domains. An N-terminal myristoyl modification can bind to a hydrophobic pocket in the kinase C-lobe, which stabilizes the autoinhibitory assembly. Activation is triggered by myristoyl release. We used molecular dynamics simulations to show how both myristoyl and the Src homology domains are required to impose the full inhibitory effect on the kinase domain and reveal the allosteric transmission pathway at residue-level resolution. Importantly, we find myristoyl insertion into a membrane to thermodynamically compete with binding to c-Abl. Myristoyl thus not only localizes the protein to the cellular membrane, but membrane attachment at the same time enhances activation of c-Abl by stabilizing its preactivated state. Our data put forward a model in which lipidation tightly couples kinase localization and regulation, a scheme that currently appears to be unique for this non-receptor tyrosine kinase.
Collapse
Affiliation(s)
- Svenja de Buhr
- Heidelberg Institute for Theoretical Studies (HITS), Heidelberg UniversityHeidelbergGermany
| | - Frauke Gräter
- Heidelberg Institute for Theoretical Studies (HITS), Heidelberg UniversityHeidelbergGermany
- Institute for Scientific Computing (IWR), Heidelberg UniversityHeidelbergGermany
| |
Collapse
|
17
|
Marchal MA, Moose DL, Varzavand A, Jordan NE, Taylor D, Tanas MR, Brown JA, Henry MD, Stipp CS. Abl kinases can function as suppressors of tumor progression and metastasis. Front Oncol 2023; 13:1241056. [PMID: 37746268 PMCID: PMC10514900 DOI: 10.3389/fonc.2023.1241056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Abl family kinases function as proto-oncogenes in various leukemias, and pro-tumor functions have been discovered for Abl kinases in many solid tumors as well. However, a growing body of evidence indicates that Abl kinases can function to suppress tumor cell proliferation and motility and tumor growth in vivo in some settings. Methods To investigate the role of Abl kinases in tumor progression, we used RNAi to generate Abl-deficient cells in a model of androgen receptor-indifferent, metastatic prostate cancer. The effect of Abl kinase depletion on tumor progression and metastasis was studied in an in vivo orthotopic model, and tumor cell motility, 3D growth, and signaling was studied in vitro. Results Reduced Abl family kinase expression resulted in a highly aggressive, metastatic phenotype in vivo that was associated with AKT pathway activation, increased growth on 3D collagen matrix, and enhanced cell motility in vitro. Inhibiting AKT pathway signaling abolished the increased 3D growth of Abl-deficient cells, while treatment with the Abl kinase inhibitor, imatinib, promoted 3D growth of multiple additional tumor cell types. Moreover, Abl kinase inhibition also promoted soft-agar colony formation by pre-malignant fibroblasts. Conclusions Collectively, our data reveal that Abl family kinases can function to suppress malignant cell phenotypes in vitro, and tumor progression and metastasis in vivo.
Collapse
Affiliation(s)
- Melissa A Marchal
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, United States
| | - Devon L Moose
- Department of Molecular Physiology & Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Afshin Varzavand
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, United States
| | - Nicole E Jordan
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, United States
| | - Destiney Taylor
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, United States
| | - Munir R Tanas
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - James A Brown
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Urology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Michael D Henry
- Department of Molecular Physiology & Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Christopher S Stipp
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
18
|
Sporbeck K, Haas ML, Pastor-Maldonado CJ, Schüssele DS, Hunter C, Takacs Z, Diogo de Oliveira AL, Franz-Wachtel M, Charsou C, Pfisterer SG, Gubas A, Haller PK, Knorr RL, Kaulich M, Macek B, Eskelinen EL, Simonsen A, Proikas-Cezanne T. The ABL-MYC axis controls WIPI1-enhanced autophagy in lifespan extension. Commun Biol 2023; 6:872. [PMID: 37620393 PMCID: PMC10449903 DOI: 10.1038/s42003-023-05236-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 08/10/2023] [Indexed: 08/26/2023] Open
Abstract
Human WIPI β-propellers function as PI3P effectors in autophagy, with WIPI4 and WIPI3 being able to link autophagy control by AMPK and TORC1 to the formation of autophagosomes. WIPI1, instead, assists WIPI2 in efficiently recruiting the ATG16L1 complex at the nascent autophagosome, which in turn promotes lipidation of LC3/GABARAP and autophagosome maturation. However, the specific role of WIPI1 and its regulation are unknown. Here, we discovered the ABL-ERK-MYC signalling axis controlling WIPI1. As a result of this signalling, MYC binds to the WIPI1 promoter and represses WIPI1 gene expression. When ABL-ERK-MYC signalling is counteracted, increased WIPI1 gene expression enhances the formation of autophagic membranes capable of migrating through tunnelling nanotubes to neighbouring cells with low autophagic activity. ABL-regulated WIPI1 function is relevant to lifespan control, as ABL deficiency in C. elegans increased gene expression of the WIPI1 orthologue ATG-18 and prolonged lifespan in a manner dependent on ATG-18. We propose that WIPI1 acts as an enhancer of autophagy that is physiologically relevant for regulating the level of autophagic activity over the lifespan.
Collapse
Affiliation(s)
- Katharina Sporbeck
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Maximilian L Haas
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Carmen J Pastor-Maldonado
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - David S Schüssele
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Catherine Hunter
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Zsuzsanna Takacs
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Institute of Molecular Biotechnology, A-1030, Vienna, Austria
| | - Ana L Diogo de Oliveira
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Mirita Franz-Wachtel
- Proteome Center Tübingen, Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Chara Charsou
- Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway
| | - Simon G Pfisterer
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Department of Anatomy, Faculty of Medicine, University of Helsinki, FI-00290, Helsinki, Finland
| | - Andrea Gubas
- Institute of Biochemistry II, Frankfurt Cancer Institute, Goethe University Medical School, D-60590, Frankfurt, Germany
| | - Patricia K Haller
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Roland L Knorr
- Humboldt University of Berlin, Institute of Biology, D-10115, Berlin, Germany
- Graduate School and Faculty of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
- International Research Frontiers Initiative, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, 226-8503, Japan
| | - Manuel Kaulich
- Institute of Biochemistry II, Frankfurt Cancer Institute, Goethe University Medical School, D-60590, Frankfurt, Germany
| | - Boris Macek
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Proteome Center Tübingen, Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Eeva-Liisa Eskelinen
- Department of Biosciences, University of Helsinki, Fl-00790, Helsinki, Finland
- Institute of Biomedicine, University of Turku, FI-20520, Turku, Finland
| | - Anne Simonsen
- Institute of Basic Medical Sciences, University of Oslo, 0372, Oslo, Norway
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway
| | - Tassula Proikas-Cezanne
- Interfaculty Institute of Cell Biology, Eberhard Karls University Tübingen, D-72076, Tübingen, Germany.
- International Max Planck Research School 'From Molecules to Organisms', Max Planck Institute for Biology and Eberhard Karls University Tübingen, D-72076, Tübingen, Germany.
| |
Collapse
|
19
|
Schumacher JA, Wright ZA, Florat DR, Anand SK, Dasyani M, Klimkaite L, Bredemeier NO, Gurung S, Koller GM, Aguera KN, Chadwick GP, Johnson RD, Davis GE, Sumanas S. SH2 domain protein E (SHE) and ABL signaling regulate blood vessel size. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.03.547455. [PMID: 37461480 PMCID: PMC10349984 DOI: 10.1101/2023.07.03.547455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Blood vessels in different vascular beds vary in lumen diameter, which is essential for their function and fluid flow along the vascular network. Molecular mechanisms involved in the formation of a vascular lumen of appropriate size, or tubulogenesis, are still only partially understood. Src homology 2 domain containing E (She) protein was previously identified in a screen for proteins that interact with Abelson (Abl)-kinase. However, its biological role has remained unknown. Here we demonstrate that She and Abl signaling regulate vascular lumen size in zebrafish embryos and human endothelial cell culture. Zebrafish she mutants displayed increased endothelial cell number and enlarged lumen size of the dorsal aorta (DA) and defects in blood flow. Vascular endothelial specific overexpression of she resulted in a reduced diameter of the DA lumen, which correlated with the reduced arterial cell number and lower endothelial cell proliferation. Chemical inhibition of Abl signaling in zebrafish embryos caused a similar reduction in the DA diameter and alleviated the she mutant phenotype, suggesting that She acts as a negative regulator of Abl signaling. Enlargement of the DA lumen in she mutants correlated with an increased endothelial expression of claudin 5a and 5b (cldn5a / cldn5b), which encode proteins enriched in tight junctions. Inhibition of cldn5a expression partially rescued the enlarged DA in she mutants, suggesting that She regulates DA lumen size, in part, by promoting cldn5a expression. SHE knockdown in human endothelial umbilical vein cells resulted in a similar increase in the diameter of vascular tubes, and also increased phosphorylation of a known ABL downstream effector CRKL. These results argue that SHE functions as an evolutionarily conserved inhibitor of ABL signaling and regulates lumen size during vascular tubulogenesis.
Collapse
Affiliation(s)
- Jennifer A. Schumacher
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, OH 45267, USA
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - Zoë A. Wright
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Diandra Rufin Florat
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Surendra K. Anand
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Manish Dasyani
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Laurita Klimkaite
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Nina O. Bredemeier
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
| | - Suman Gurung
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| | - Gretchen M. Koller
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Kalia N. Aguera
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Griffin P. Chadwick
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - Riley D. Johnson
- Department of Biological Sciences, Miami University, Hamilton, OH 45011, USA
| | - George E. Davis
- University of South Florida, Department of Molecular Pharmacology and Physiology, Tampa, FL 33612, USA
| | - Saulius Sumanas
- Cincinnati Children’s Hospital Medical Center, Division of Developmental Biology, Cincinnati, OH 45229, USA
- University of Cincinnati College of Medicine, Department of Pediatrics, Cincinnati, OH 45267, USA
- University of South Florida, Department of Pathology and Cell Biology, USF Health Heart Institute, Tampa, FL 33602, USA
| |
Collapse
|
20
|
Cunningham A, Brown M, Dresselhuis J, Robinson N, Hervie K, Cox ME, Mills J. Combination Effects of Integrin-linked Kinase and Abelson Kinase Inhibition on Aberrant Mitosis and Cell Death in Glioblastoma Cells. BIOLOGY 2023; 12:906. [PMID: 37508338 PMCID: PMC10376030 DOI: 10.3390/biology12070906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/21/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023]
Abstract
In cancer cells, inhibition of integrin-linked kinase (ILK) increases centrosome declustering causing mitotic arrest and cell death. Yet, not all cancer cells are susceptible to anti-ILK treatment alone. We investigate a combination drug strategy targeting ILK and another oncogenic kinase, Abelson kinase (ABL). Drug-concentration viability assays (i.e., MTT assays) indicate that ILK and ABL inhibitors in combination decreased the viability of glioblastoma cells over the ILK drug QLT-0267 alone. Combination strategies also increased aberrant mitoses and cell death over QLT-0267 alone. This was evident from an increase in mitotic arrest, apoptosis and a sub-G1 peak following FAC analysis. In vitro, ILK and ABL localized to the centrosome and the putative ILK kinase domain was important for this localization. Increased levels of cytosolic ABL are associated with its transformative abilities. ILK inhibitor effects on survival correlated with its ability to decrease cytosolic ABL levels and inhibit ABL's localization to mitotic centrosomes in glioblastoma cells. ILK inhibitor effects on ABL's centrosomal localization were reversed by the proteasomal inhibitor MG132 (a drug that inhibits ABL degradation). These results indicate that ILK regulates ABL at mitotic centrosomes and that combination treatments targeting ILK and ABL are more effective then QLT-0267 alone at decreasing the survival of dividing glioblastoma cells.
Collapse
Affiliation(s)
- Abigail Cunningham
- Department of Biology, Trinity Western University, Langley, BC V2Y 1Y1, Canada
| | - Maddisen Brown
- Department of Biology, Trinity Western University, Langley, BC V2Y 1Y1, Canada
| | | | - Nicole Robinson
- Vancouver Prostate Center and Vancouver Coastal Health Research Institute, Vancouver, BC V6T 1Z3, Canada
| | - Keni Hervie
- Department of Biology, Trinity Western University, Langley, BC V2Y 1Y1, Canada
| | - Michael E Cox
- Vancouver Prostate Center and Vancouver Coastal Health Research Institute, Vancouver, BC V6T 1Z3, Canada
| | - Julia Mills
- Department of Biology, Trinity Western University, Langley, BC V2Y 1Y1, Canada
| |
Collapse
|
21
|
León R, Gutiérrez DA, Pinto C, Morales C, de la Fuente C, Riquelme C, Cortés BI, González-Martin A, Chamorro D, Espinosa N, Fuentealba P, Cancino GI, Zanlungo S, Dulcey AE, Marugan JJ, Álvarez Rojas A. c-Abl tyrosine kinase down-regulation as target for memory improvement in Alzheimer's disease. Front Aging Neurosci 2023; 15:1180987. [PMID: 37358955 PMCID: PMC10289333 DOI: 10.3389/fnagi.2023.1180987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/12/2023] [Indexed: 06/28/2023] Open
Abstract
Background Growing evidence suggests that the non-receptor tyrosine kinase, c-Abl, plays a significant role in the pathogenesis of Alzheimer's disease (AD). Here, we analyzed the effect of c-Abl on the cognitive performance decline of APPSwe/PSEN1ΔE9 (APP/PS1) mouse model for AD. Methods We used the conditional genetic ablation of c-Abl in the brain (c-Abl-KO) and pharmacological treatment with neurotinib, a novel allosteric c-Abl inhibitor with high brain penetrance, imbued in rodent's chow. Results We found that APP/PS1/c-Abl-KO mice and APP/PS1 neurotinib-fed mice had improved performance in hippocampus-dependent tasks. In the object location and Barnes-maze tests, they recognized the displaced object and learned the location of the escape hole faster than APP/PS1 mice. Also, APP/PS1 neurotinib-fed mice required fewer trials to reach the learning criterion in the memory flexibility test. Accordingly, c-Abl absence and inhibition caused fewer amyloid plaques, reduced astrogliosis, and preserved neurons in the hippocampus. Discussion Our results further validate c-Abl as a target for AD, and the neurotinib, a novel c-Abl inhibitor, as a suitable preclinical candidate for AD therapies.
Collapse
Affiliation(s)
- Rilda León
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela A. Gutiérrez
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudio Pinto
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian Morales
- Laboratory for Brain-Machine Interfaces and Neuromodulation, Facultad de Ingeniería, Instituto de Ingeniería Biológica y Médica, Pontificia Universidad Católica de Chile, Santiago, Chile
- Laboratory of Neural Circuits, Department of Psychiatry, Neuroscience Interdisciplinary Centre, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina de la Fuente
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristóbal Riquelme
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bastián I. Cortés
- Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Adrián González-Martin
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - David Chamorro
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nelson Espinosa
- Laboratory of Neural Circuits, Department of Psychiatry, Neuroscience Interdisciplinary Centre, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Fuentealba
- Laboratory of Neural Circuits, Department of Psychiatry, Neuroscience Interdisciplinary Centre, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gonzalo I. Cancino
- Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Silvana Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Andrés E. Dulcey
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, United States
| | - Juan J. Marugan
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, United States
| | - Alejandra Álvarez Rojas
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Biological Sciences Faculty, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
22
|
Fan S, Shen Y, Li S, Xiang X, Li N, Li Y, Xu J, Cui M, Han X, Xia J, Huang Y. The S2 Subunit of Infectious Bronchitis Virus Affects Abl2-Mediated Syncytium Formation. Viruses 2023; 15:1246. [PMID: 37376546 DOI: 10.3390/v15061246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The S2 subunit serves a crucial role in infectious bronchitis virus (IBV) infection, particularly in facilitating membrane fusion. Using reverse genetic techniques, mutant strains of the S2 locus exhibited substantially different syncytium-forming abilities in chick embryonic kidney cells. To determine the precise formation mechanism of syncytium, we demonstrated the co-ordinated role of Abl2 and its mediated cytoskeletal regulatory pathway within the S2 subunit. Using a combination of fluorescence quantification, RNA silencing, and protein profiling techniques, the functional role of S2 subunits in IBV-infected cells was exhaustively determined. Our findings imply that Abl2 is not the primary cytoskeletal regulator, the viral S2 component is involved in indirect regulation, and the three different viral strains activate various cytoskeletal regulatory pathways through Abl2. CRK, CRKL, ABI1, NCKAP1, and ENAH also play a role in cytoskeleton regulation. Our research provides a point of reference for the development of an intracellular regulatory network for the S2 subunit and a foundation for the rational design of antiviral drug targets against Abl2.
Collapse
Affiliation(s)
- Shunyi Fan
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Yuxi Shen
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Shuyun Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Xuelian Xiang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Nianling Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Yongxin Li
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Jing Xu
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Min Cui
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Xinfeng Han
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Jing Xia
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| | - Yong Huang
- College of Veterinary Medicine, Sichuan Agricultural University, Huimin Road, Wenjiang, Chengdu 611130, China
| |
Collapse
|
23
|
Asano Y, Matsumoto Y, Wada J, Rottapel R. E3-ubiquitin ligases and recent progress in osteoimmunology. Front Immunol 2023; 14:1120710. [PMID: 36911671 PMCID: PMC9996189 DOI: 10.3389/fimmu.2023.1120710] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Ubiquitin-mediated proteasomal degradation is a post-transcriptional protein modification that is comprised of various components including the 76-amino acid protein ubiquitin (Ub), Ub-activating enzyme (E1), Ub-conjugating enzyme (E2), ubiquitin ligase (E3), deubiquitinating enzyme (DUB) and proteasome. We and others have recently provided genetic evidence showing that E3-ubiquitin ligases are associated with bone metabolism, the immune system and inflammation through ubiquitylation and subsequent degradation of their substrates. Dysregulation of the E3-ubiquitin ligase RNF146-mediated degradation of the adaptor protein 3BP2 (SH3 domain-binding protein 2) causes cherubism, an autosomal dominant disorder associated with severe inflammatory craniofacial dysmorphia syndrome in children. In this review, on the basis of our discoveries in cherubism, we summarize new insights into the roles of E3-ubiquitin ligases in the development of human disorders caused by an abnormal osteoimmune system by highlighting recent genetic evidence obtained in both human and animal model studies.
Collapse
Affiliation(s)
- Yosuke Asano
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yoshinori Matsumoto
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Robert Rottapel
- Princess Margaret Cancer Center, University Health Network, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Division of Rheumatology, St. Michael’s Hospital, Toronto, ON, Canada
| |
Collapse
|
24
|
Kumar D, Kumar H, Kumar V, Deep A, Sharma A, Marwaha MG, Marwaha RK. Mechanism-based approaches of 1,3,4 thiadiazole scaffolds as potent enzyme inhibitors for cytotoxicity and antiviral activity. MEDICINE IN DRUG DISCOVERY 2023. [DOI: 10.1016/j.medidd.2022.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
25
|
Nauth T, Bazgir F, Voß H, Brandenstein LI, Mosaddeghzadeh N, Rickassel V, Deden S, Gorzelanny C, Schlüter H, Ahmadian MR, Rosenberger G. Cutaneous manifestations in Costello syndrome: HRAS p.Gly12Ser affects RIN1-mediated integrin trafficking in immortalized epidermal keratinocytes. Hum Mol Genet 2023; 32:304-318. [PMID: 35981076 DOI: 10.1093/hmg/ddac188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 07/15/2022] [Accepted: 08/07/2022] [Indexed: 01/18/2023] Open
Abstract
Heterozygous germline missense variants in the HRAS gene underlie Costello syndrome (CS). The molecular basis for cutaneous manifestations in CS is largely unknown. We used an immortalized human cell line, HaCaT keratinocytes, stably expressing wild-type or CS-associated (p.Gly12Ser) HRAS and defined RIN1 as quantitatively most prominent, high-affinity effector of active HRAS in these cells. As an exchange factor for RAB5 GTPases, RIN1 is involved in endosomal sorting of cell-adhesion integrins. RIN1-dependent RAB5A activation was strongly increased by HRASGly12Ser, and HRAS-RIN1-ABL1/2 signaling was induced in HRASWT- and HRASGly12Ser-expressing cells. Along with that, HRASGly12Ser expression decreased total integrin levels and enriched β1 integrin in RAB5- and EEA1-positive early endosomes. The intracellular level of active β1 integrin was increased in HRASGly12Ser HaCaT keratinocytes due to impaired recycling, whereas RIN1 disruption raised β1 integrin cell surface distribution. HRASGly12Ser induced co-localization of β1 integrin with SNX17 and RAB7 in early/sorting and late endosomes, respectively. Thus, by retaining β1 integrin in intracellular endosomal compartments, HRAS-RIN1 signaling affects the subcellular availability of β1 integrin. This may interfere with integrin-dependent processes as we detected for HRASGly12Ser cells spreading on fibronectin. We conclude that dysregulation of receptor trafficking and integrin-dependent processes such as cell adhesion are relevant in the pathobiology of CS.
Collapse
Affiliation(s)
- Theresa Nauth
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Farhad Bazgir
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Hannah Voß
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Laura I Brandenstein
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Niloufar Mosaddeghzadeh
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Verena Rickassel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sophia Deden
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Christian Gorzelanny
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, Section Mass Spectrometry and Proteomics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Mohammad R Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - Georg Rosenberger
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| |
Collapse
|
26
|
Jones JK, Zhang H, Lyne AM, Cavalli FMG, Hassen WE, Stevenson K, Kornahrens R, Yang Y, Li S, Dell S, Reitman ZJ, Herndon JE, Hoj J, Pendergast AM, Thompson EM. ABL1 and ABL2 promote medulloblastoma leptomeningeal dissemination. Neurooncol Adv 2023; 5:vdad095. [PMID: 37781087 PMCID: PMC10540884 DOI: 10.1093/noajnl/vdad095] [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] [Indexed: 10/03/2023] Open
Abstract
Background Medulloblastoma is the most common malignant pediatric brain tumor, and leptomeningeal dissemination (LMD) of medulloblastoma both portends a poorer prognosis at diagnosis and is incurable at recurrence. The biological mechanisms underlying LMD are unclear. The Abelson (ABL) tyrosine kinase family members, ABL1 and ABL2, have been implicated in cancer cell migration, invasion, adhesion, metastasis, and chemotherapy resistance, and are upstream mediators of the oncogene c-MYC in fibroblasts and lung cancer cells. However, their role in medulloblastoma has not yet been explored. The purpose of this work was to elucidate the role of ABL1/2 in medulloblastoma LMD. Methods ABL1 and ABL2 mRNA expression of patient specimens was analyzed. shRNA knockdowns of ABL1/2 and pharmacologic inhibition of ABL1/2 were used for in vitro and in vivo analyses of medulloblastoma LMD. RNA sequencing of ABL1/2 genetic knockdown versus scrambled control medulloblastoma was completed. Results ABL1/2 mRNA is highly expressed in human medulloblastoma and pharmacologic inhibition of ABL kinases resulted in cytotoxicity. Knockdown of ABL1/2 resulted in decreased adhesion of medulloblastoma cells to the extracellular matrix protein, vitronectin (P = .0013), and significantly decreased tumor burden in a mouse model of medulloblastoma LMD with improved overall survival (P = .0044). Furthermore, both pharmacologic inhibition of ABL1/2 and ABL1/2 knockdown resulted in decreased expression of c-MYC, identifying a putative signaling pathway, and genes/pathways related to oncogenesis and neurodevelopment were differentially expressed between ABL1/2 knockdown and control medulloblastoma cells. Conclusions ABL1 and ABL2 have potential roles in medulloblastoma LMD upstream of c-MYC expression.
Collapse
Affiliation(s)
- Jill K Jones
- Harvard/MIT MD-PhD Program, Boston, MA, USA
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Hengshan Zhang
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Anne-Marie Lyne
- Institut Curie, PSL Research University, Paris, France
- Inserm, U900, Paris, France
- MINES ParisTech, CBI – Centre for Computational Biology, PL Research University, Paris, France
| | - Florence M G Cavalli
- Institut Curie, PSL Research University, Paris, France
- Inserm, U900, Paris, France
- MINES ParisTech, CBI – Centre for Computational Biology, PL Research University, Paris, France
| | - Wafa E Hassen
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Kevin Stevenson
- Duke University Molecular Physiology Institute, Durham, NC, USA
| | - Reb Kornahrens
- Department of Neurosurgery, Duke University, Durham, NC, USA
| | - Yuanfan Yang
- Department of Neurosurgery, Duke University, Durham, NC, USA
- Department of Neurosurgery, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sean Li
- Department of Neurosurgery, Duke University, Durham, NC, USA
- Case Western University School of Medicine, Cleveland, OH, USA
| | - Samuel Dell
- Department of Neurosurgery, Duke University, Durham, NC, USA
- Division of Hematologic Malignancies and Cellular Therapy, Duke Cancer Institute
| | - Zachary J Reitman
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - James E Herndon
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Jacob Hoj
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | | | - Eric M Thompson
- Department of Neurosurgery, Duke University, Durham, NC, USA
- Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC, USA
- Department of Neurosurgery, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
27
|
Wang X, Yuan Y, Liu Y, Zhang L. Arm race between Rift Valley fever virus and host. Front Immunol 2022; 13:1084230. [PMID: 36618346 PMCID: PMC9813963 DOI: 10.3389/fimmu.2022.1084230] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
Rift Valley fever (RVF) is a zoonotic disease caused by Rift Valley fever virus (RVFV), an emerging arbovirus within the Phenuiviridae family of Bunyavirales that has potential to cause severe diseases in both humans and livestock. It increases the incidence of abortion or foetal malformation in ruminants and leads to clinical manifestations like encephalitis or haemorrhagic fever in humans. Upon virus invasion, the innate immune system from the cell or the organism is activated to produce interferon (IFN) and prevent virus proliferation. Meanwhile, RVFV initiates countermeasures to limit antiviral responses at transcriptional and protein levels. RVFV nonstructural proteins (NSs) are the key virulent factors that not only perform immune evasion but also impact the cell replication cycle and has cytopathic effects. In this review, we summarize the innate immunity host cells employ depending on IFN signal transduction pathways, as well as the immune evasion mechanisms developed by RVFV primarily with the inhibitory activity of NSs protein. Clarifying the arms race between host innate immunity and RVFV immune evasion provides new avenues for drug target screening and offers possible solutions to current and future epidemics.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yupei Yuan
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yihan Liu
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Leiliang Zhang
- Department of Infectious Diseases, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
28
|
Allosteric Inhibition of c-Abl to Induce Unfolded Protein Response and Cell Death in Multiple Myeloma. Int J Mol Sci 2022; 23:ijms232416162. [PMID: 36555805 PMCID: PMC9786043 DOI: 10.3390/ijms232416162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/03/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Endoplasmic reticulum stress activates inositol-requiring enzyme 1α (IRE1α) and protein kinase, R-like endoplasmic reticulum kinase (PERK), the two principal regulators of the unfolded protein response (UPR). In multiple myeloma, adaptive IRE1α signaling is predominantly activated and regulates cell fate along with PERK. Recently, we demonstrated that GNF-2, an allosteric c-Abl inhibitor, rheostatically enhanced IRE1α activity and induced apoptosis through c-Abl conformational changes in pancreatic β cells. Herein, we analyzed whether the pharmacological modulation of c-Abl conformation resulted in anti-myeloma effects. First, we investigated the effects of GNF-2 on IRE1α activity and cell fate, followed by an investigation of the anti-myeloma effects of asciminib, a new allosteric c-Abl inhibitor. Finally, we performed RNA sequencing to characterize the signaling profiles of asciminib. We observed that both GNF-2 and asciminib decreased cell viability and induced XBP1 mRNA splicing in primary human myeloma cells and myeloma cell lines. RNA sequencing identified the induction of UPR- and apoptosis-related genes by asciminib. Asciminib re-localized c-Abl to the endoplasmic reticulum, and its combination with a specific IRE1α inhibitor, KIRA8, enhanced cell death with the reciprocal induction of CHOP mRNA expression. Together, the allosteric inhibition of c-Abl-activated UPR with anti-myeloma effects; this could be a novel therapeutic target for multiple myeloma.
Collapse
|
29
|
Gros K, Matkovič U, Parato G, Miš K, Luin E, Bernareggi A, Sciancalepore M, Marš T, Lorenzon P, Pirkmajer S. Neuronal Agrin Promotes Proliferation of Primary Human Myoblasts in an Age-Dependent Manner. Int J Mol Sci 2022; 23:ijms231911784. [PMID: 36233091 PMCID: PMC9570459 DOI: 10.3390/ijms231911784] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/06/2022] [Accepted: 09/22/2022] [Indexed: 12/02/2022] Open
Abstract
Neuronal agrin, a heparan sulphate proteoglycan secreted by the α-motor neurons, promotes the formation and maintenance of the neuromuscular junction by binding to Lrp4 and activating muscle-specific kinase (MuSK). Neuronal agrin also promotes myogenesis by enhancing differentiation and maturation of myotubes, but its effect on proliferating human myoblasts, which are often considered to be unresponsive to agrin, remains unclear. Using primary human myoblasts, we determined that neuronal agrin induced transient dephosphorylation of ERK1/2, while c-Abl, STAT3, and focal adhesion kinase were unresponsive. Gene silencing of Lrp4 and MuSK markedly reduced the BrdU incorporation, suggesting the functional importance of the Lrp4/MuSK complex for myoblast proliferation. Acute and chronic treatments with neuronal agrin increased the proliferation of human myoblasts in old donors, but they did not affect the proliferation of myoblasts in young donors. The C-terminal fragment of agrin which lacks the Lrp4-binding site and cannot activate MuSK had a similar age-dependent effect, indicating that the age-dependent signalling pathways activated by neuronal agrin involve the Lrp4/MuSK receptor complex as well as an Lrp4/MuSK-independent pathway which remained unknown. Collectively, our results highlight an age-dependent role for neuronal agrin in promoting the proliferation of human myoblasts.
Collapse
Affiliation(s)
- Katarina Gros
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Urška Matkovič
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Giulia Parato
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- The B.R.A.I.N. Centre for Neuroscience, University of Trieste, 34127 Trieste, Italy
| | - Katarina Miš
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Elisa Luin
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- The B.R.A.I.N. Centre for Neuroscience, University of Trieste, 34127 Trieste, Italy
| | - Annalisa Bernareggi
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- The B.R.A.I.N. Centre for Neuroscience, University of Trieste, 34127 Trieste, Italy
| | - Marina Sciancalepore
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- The B.R.A.I.N. Centre for Neuroscience, University of Trieste, 34127 Trieste, Italy
| | - Tomaž Marš
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Paola Lorenzon
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
- The B.R.A.I.N. Centre for Neuroscience, University of Trieste, 34127 Trieste, Italy
- Correspondence: (P.L.); (S.P.)
| | - Sergej Pirkmajer
- Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Correspondence: (P.L.); (S.P.)
| |
Collapse
|
30
|
Erol A. Genotoxicity-Stimulated and CYLD-Driven Malignant Transformation. Cancer Manag Res 2022; 14:2339-2356. [PMID: 35958947 PMCID: PMC9362849 DOI: 10.2147/cmar.s373557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/28/2022] [Indexed: 11/23/2022] Open
Abstract
Oxidative stress, which can cause DNA damage, can both activate TNF-R1 directly in the absence of TNF stimulation and phosphorylate c-Abl, thus promoting its cytoplasmic translocation. Persistent cytoplasmic localization of c-Abl has been associated with cellular transformation. c-Abl phosphorylates OTULIN at tyrosine 56, thereby disrupting its relationship with LUBAC. OTULIN-released LUBAC interacts with SPATA2 and is recruited to the TNF-R1sc, facilitating SPATA2-CYLD interaction. All these interactions are required for the activation of IKKβ to stimulate NF-κB transcriptional activity following genotoxic stress. IKKβ also induces the critical phosphorylation of CYLD at serine 568 to increase its deubiquitinating (DUB) activity required for the termination of signaling cascades. Contrary to the widespread belief that CYLD is an absolute tumor suppressor, CYLD initiates and terminates NF-κB activity by alternately using its oncoprotein and tumor suppressor activities, respectively. If IKKβ fails to achieve the DUB activity-inducing phosphorylation at serine 568, CYLD would operate in a sustained mode of oncogenic activity. The resulting dysregulated NF-κB activation and other accompanying pathologies will disrupt cellular homeostasis in favor of transformation.
Collapse
Affiliation(s)
- Adnan Erol
- Independent Researcher, Istanbul, Turkey
| |
Collapse
|
31
|
Jiang P, Tang S, Hudgins H, Smalligan T, Zhou X, Kamat A, Dharmarpandi J, Naguib T, Liu X, Dai Z. The Abl/Abi signaling links WAVE regulatory complex to Cbl E3 ubiquitin ligase and is essential for breast cancer cell metastasis. Neoplasia 2022; 32:100819. [PMID: 35839699 PMCID: PMC9287790 DOI: 10.1016/j.neo.2022.100819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/29/2022] [Indexed: 12/04/2022]
Abstract
A Cbl-TKB binding motif regulates the stability of Abi and WAVE regulatory complex. Abl kinases serve as a switch to activate Cbl-mediated Abi/WRC degradation. Depletion of Abi1 impairs EGFR and Src family kinases signaling. Abi1 is essential for breast cancer cell invasion and lung metastasis.
The family of Abelson interactor (Abi) proteins is a component of WAVE regulatory complex (WRC) and a downstream target of Abelson (Abl) tyrosine kinase. The fact that Abi proteins also interact with diverse membrane proteins and intracellular signaling molecules places these proteins at a central position in the network that controls cytoskeletal functions and cancer cell metastasis. Here, we identified a motif in Abi proteins that conforms to consensus sequences found in a cohort of receptor and non-receptor tyrosine kinases that bind to Cbl-tyrosine kinase binding domain. The phosphorylation of tyrosine 213 in this motif is essential for Abi degradation. Double knockout of c-Cbl and Cbl B in Bcr-Abl-transformed leukemic cells abolishes Abi1, Abi2, and WAVE2 degradation. Moreover, knockout of Abi1 reduces Src family kinase Lyn activation in Bcr-Abl-positive leukemic cells and promotes EGF-induced EGF receptor downregulation in breast cancer cells. Importantly, Abi1 depletion impeded breast cancer cell invasion in vitro and metastasis in mouse xenografts. Together, these studies uncover a novel mechanism by which the WRC and receptor/non-receptor tyrosine kinases are regulated and identify Abi1 as a potential therapeutic target for metastatic breast cancer.
Collapse
Affiliation(s)
- Peixin Jiang
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, Amarillo, TX 79106, USA
| | - Suni Tang
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, USA
| | - Hogan Hudgins
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, Amarillo, TX 79106, USA
| | - Tate Smalligan
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, Amarillo, TX 79106, USA
| | - Xue Zhou
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, USA
| | - Anuja Kamat
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, Amarillo, TX 79106, USA
| | - Janaki Dharmarpandi
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, Amarillo, TX 79106, USA
| | - Tarek Naguib
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, Amarillo, TX 79106, USA
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, TX, 77204, USA.
| | - Zonghan Dai
- Department of Internal Medicine, Texas Tech University Health Sciences Center School of Medicine, Amarillo, TX 79106, USA.
| |
Collapse
|
32
|
Luo L, Jiang P, Chen Q, Chang J, Jing Y, Luo X, Gu H, Huang Y, Chen R, Liu J, Kang D, Liu Q, Wang Y, Fang G, Zhu Y, Guan F, Lei J, Yang L, Liu C, Dai X. c-Abl controls BCR signaling and B cell differentiation by promoting B cell metabolism. Immunology 2022; 167:181-196. [PMID: 35753034 DOI: 10.1111/imm.13525] [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: 10/10/2021] [Accepted: 03/15/2022] [Indexed: 11/30/2022] Open
Abstract
As a non-receptor tyrosine kinase, c-Abl was first studied in chronic myelogenous leukemia, and its role in lymphocytes has been well characterized. c-Abl is involved in B cell development and CD19 associated B cell antigen receptor (BCR) signaling. Although c-Abl regulates different metabolic pathways, the role of c-Abl is still unknown in B cell metabolism. In this study, B cell specific c-Abl knockout (KO) mice (Mb1Cre+/- c-Ablfl/fl ) were used to investigate how c-Abl regulates B cell metabolism and BCR signaling. We found that the levels of activation positive BCR signaling proximal molecules, phosphorylated spleen tyrosine kinase (pSYK) and phosphorylated Bruton tyrosine kinase (pBTK), were decreased, while the level of key negative regulator, phosphorylated SH2-containing inositol phosphatase (pSHIP1), was increased in Mb1Cre+/- c-Ablfl/fl mice. Furthermore, we found c-Abl deficiency weakened the B cell spreading, formation of BCR signalosomes, and the polymerization of actin during BCR activation, and also impaired the differentiation of germinal center (GC) B cells both in quiescent condition and after immunization. Moreover, B cell mitochondrial respiration and the expression of B cell metabolism regulating molecules were downregulated in c-Abl deficiency mice. Overall, c-Abl, which involved in actin remodeling and B cell metabolism, positively regulates BCR signaling and promotes GC differentiation. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Li Luo
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Panpan Jiang
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianglin Chen
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Jiang Chang
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yukai Jing
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Luo
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heng Gu
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanmei Huang
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ran Chen
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ju Liu
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danqing Kang
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Liu
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
| | - Yi Wang
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guofeng Fang
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingzi Zhu
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Guan
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Lei
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Yang
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaohong Liu
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Dai
- Department of Pathogen Biology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Oncogenic Signalling of PEAK2 Pseudokinase in Colon Cancer. Cancers (Basel) 2022; 14:cancers14122981. [PMID: 35740644 PMCID: PMC9221080 DOI: 10.3390/cancers14122981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 06/05/2022] [Accepted: 06/13/2022] [Indexed: 11/22/2022] Open
Abstract
Simple Summary Catalytically inactive kinases, also named pseudokinases, play important roles in the regulation of cell growth and adhesion. While frequently deregulated in human cancer, their role in tumour development is partially elucidated. Here, we report an important tumour function for the pseudokinase PEAK2 in colorectal cancer (CRC) and propose that PEAK2 upregulation can affect cancer cell adhesive properties through an ABL-dependent mechanism to enable cancer progression. Therefore, targeting PEAK2 oncogenic activity with small tyrosine kinases (TK) inhibitors may be of therapeutic interest in colorectal cancer (CRC). Abstract The PEAK family pseudokinases are essential components of tyrosine kinase (TK) pathways that regulate cell growth and adhesion; however, their role in human cancer remains unclear. Here, we report an oncogenic activity of the pseudokinase PEAK2 in colorectal cancer (CRC). Notably, high PRAG1 expression, which encodes PEAK2, was associated with a bad prognosis in CRC patients. Functionally, PEAK2 depletion reduced CRC cell growth and invasion in vitro, while its overexpression increased these transforming effects. PEAK2 depletion also reduced CRC development in nude mice. Mechanistically, PEAK2 expression induced cellular protein tyrosine phosphorylation, despite its catalytic inactivity. Phosphoproteomic analysis identified regulators of cell adhesion and F-actin dynamics as PEAK2 targets. Additionally, PEAK2 was identified as a novel ABL TK activator. In line with this, PEAK2 expression localized at focal adhesions of CRC cells and induced ABL-dependent formation of actin-rich plasma membrane protrusions filopodia that function to drive cell invasion. Interestingly, all these PEAK2 transforming activities were regulated by its main phosphorylation site, Tyr413, which implicates the SRC oncogene. Thus, our results uncover a protumoural function of PEAK2 in CRC and suggest that its deregulation affects adhesive properties of CRC cells to enable cancer progression.
Collapse
|
34
|
Zhang J, Sun JG, Xing X, Wu R, Zhou L, Zhang Y, Yuan F, Wang S, Yuan Z. c-Abl-induced Olig2 phosphorylation regulates the proliferation of oligodendrocyte precursor cells. Glia 2022; 70:1084-1099. [PMID: 35156232 DOI: 10.1002/glia.24157] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 11/12/2022]
Abstract
Oligodendrocytes (OLs), the myelinating cells in the central nervous system (CNS), are differentiated from OL progenitor cells (OPCs). The proliferation of existing OPCs is indispensable for myelination during CNS development and remyelination in response to demyelination stimulation. The transcription factor Olig2 is required for the specification of OLs and is expressed in the OL lineage. However, the post-translational modification of Olig2 in the proliferation of OPCs is poorly understood. Herein, we identified that c-Abl directly phosphorylates Olig2 mainly at the Tyr137 site, and that Olig2 phosphorylation is essential for OPC proliferation. The expression levels of c-Abl gradually decreased with brain development; moreover, c-Abl was highly expressed in OPCs. OL-specific c-Abl knockout at the developmental stage led to an insufficient proliferation of OPCs, a decreased expression of myelin-related genes, and myelination retardation. Accordingly, a c-Abl-specific kinase inhibitor suppressed OPC proliferation in vitro. Furthermore, we observed that OL-specific c-Abl knockout reduced OPC proliferation and remyelination in a cuprizone model of demyelination. In addition, we found that nilotinib, a clinically used c-Abl inhibitor, decreased the expression of myelin basic protein (Mbp) and motor coordination in mice, indicating a neurological side effect of a long-term administration of the c-Abl inhibitor. Thus, we identified the important role of c-Abl in OLs during developmental myelination and remyelination in a disease model.
Collapse
Affiliation(s)
- Jun Zhang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
- School of Basic Medicine, Qingdao University, Qingdao, China
| | - Jian-Guang Sun
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Xiaowen Xing
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Rong Wu
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lujun Zhou
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Ying Zhang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Fang Yuan
- Department of Oncology, The General Hospital of Chinese People's Liberation Army No.5 Medical Science Center, Beijing, China
| | - Shukun Wang
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Zengqiang Yuan
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing, China
- Center of Alzheimer's Disease, Beijing Institute of Brain Disorders, Beijing, China
| |
Collapse
|
35
|
Bogaard HJ, Aman J. Tyrosine Kinases and Endothelial Homeostasis in Pulmonary Arterial Hypertension (PAH): Too Hot to Handle? Am J Respir Cell Mol Biol 2022; 67:147-149. [PMID: 35580152 PMCID: PMC9348559 DOI: 10.1165/rcmb.2022-0122ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Harm Jan Bogaard
- VU University Medical Center, Pulmonary Medicine, Amsterdam, Netherlands;
| | - Jurjan Aman
- Amsterdam UMC - Locatie VUMC, 1209, Pulmonary Diseases, Amsterdam, Netherlands
| |
Collapse
|
36
|
Le Vely B, Phan C, Berrebeh N, Thuillet R, Ottaviani M, Chelgham MK, Chaumais MC, Amazit L, Humbert M, Huertas A, Guignabert C, Tu L. Loss of cAbl Tyrosine Kinase in Pulmonary Arterial Hypertension Causes Dysfunction of Vascular Endothelial Cells. Am J Respir Cell Mol Biol 2022; 67:215-226. [PMID: 35550008 DOI: 10.1165/rcmb.2021-0332oc] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a progressive and fatal disease characterized by the dysfunction of pulmonary endothelial cells (ECs) and obstructive vascular remodeling. The non-receptor tyrosine kinase c-Abelson (cAbl) plays central roles in regulating cell-cycle arrest, apoptosis, and senescence after cellular stress. We hypothesized that cAbl is down-activated in experimental and human PAH, thus leading to reduced DNA integrity and angiogenic capacity of pulmonary ECs from PAH patients (PAH-ECs). We found cAbl and phosphorylated cAbl levels to be lower in the endothelium of remodeled pulmonary vessels in the lungs of PAH patients than controls. Similar observations were obtained for the lungs of sugen+hypoxia (SuHx) and monocrotaline (MCT) rats with established pulmonary hypertension. These in situ abnormalities were also replicated in vitro, with cultured PAH-ECs displaying lower cAbl expression and activity and an altered DNA damage response and capacity of tube formation. Downregulation of cAbl by RNA-interference in Control-ECs or its inhibition with dasatinib resulted in genomic instability and the failure to form tubes, whereas upregulation of cAbl with DPH reduced DNA damage and apoptosis in PAH-ECs. Finally, we establish the existence of crosstalk between cAbl and bone morphogenetic protein receptor type II (BMPRII). This work identifies the loss of cAbl signaling as a novel contributor to pulmonary EC dysfunction associated with PAH.
Collapse
Affiliation(s)
- Benjamin Le Vely
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Carole Phan
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Nihel Berrebeh
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Raphaël Thuillet
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Mina Ottaviani
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Mustapha Kamel Chelgham
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Marie-Camille Chaumais
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France.,Université Paris-Saclay Faculté de Pharmacie, 70620, Chatenay-Malabry, France
| | - Larbi Amazit
- Institut Biomédical de Bicêtre, 46657, UMS_44, Villejuif, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Marc Humbert
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France.,Assistance Publique - Hopitaux de Paris, 26930, Service de Pneumologie et Soins Intensifs Respiratoires, Le Kremlin-Bicêtre, France
| | - Alice Huertas
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France.,Assistance Publique - Hopitaux de Paris, 26930, Service de Pneumologie et Soins Intensifs Respiratoires, Le Kremlin-Bicêtre, France
| | - Christophe Guignabert
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France
| | - Ly Tu
- INSERM, 27102, UMR_S 999, Le Plessis-Robinson, France.,Université Paris-Saclay Faculté de Médecine, 89691, UMR_S 999, Le Kremlin-Bicetre, France;
| |
Collapse
|
37
|
Strobelt R, Adler J, Paran N, Yahalom-Ronen Y, Melamed S, Politi B, Shulman Z, Schmiedel D, Shaul Y. Imatinib inhibits SARS-CoV-2 infection by an off-target-mechanism. Sci Rep 2022; 12:5758. [PMID: 35388061 PMCID: PMC8984672 DOI: 10.1038/s41598-022-09664-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 03/21/2022] [Indexed: 12/15/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causal agent of the COVID-19 pandemic. More than 274 million individuals have suffered from COVID-19 and over five million people have died from this disease so far. Therefore, there is an urgent need for therapeutic drugs. Repurposing FDA approved drugs should be favored since evaluation of safety and efficacy of de-novo drug design are both costly and time consuming. We report that imatinib, an Abl tyrosine kinase inhibitor, robustly decreases SARS-CoV-2 infection and uncover a mechanism of action. We show that imatinib inhibits the infection of SARS-CoV-2 and its surrogate lentivector pseudotype. In latter, imatinib inhibited both routes of viral entry, endocytosis and membrane-fusion. We utilized a system to quantify in real-time cell-cell membrane fusion mediated by the SARS-CoV-2 surface protein, Spike, and its receptor, hACE2, to demonstrate that imatinib inhibits this process in an Abl1 and Abl2 independent manner. Furthermore, cellular thermal shift assay revealed a direct imatinib-Spike interaction that affects Spike susceptibility to trypsin digest. Collectively, our data suggest that imatinib inhibits Spike mediated viral entry by an off-target mechanism. These findings mark imatinib as a promising therapeutic drug in inhibiting the early steps of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Romano Strobelt
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Julia Adler
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Nir Paran
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Yfat Yahalom-Ronen
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Sharon Melamed
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Boaz Politi
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness Ziona, Israel
| | - Ziv Shulman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Dominik Schmiedel
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Yosef Shaul
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
38
|
Anti-apoptotic HAX-1 suppresses cell apoptosis by promoting c-Abl kinase-involved ROS clearance. Cell Death Dis 2022; 13:298. [PMID: 35379774 PMCID: PMC8979985 DOI: 10.1038/s41419-022-04748-2] [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: 07/07/2021] [Revised: 03/05/2022] [Accepted: 03/18/2022] [Indexed: 11/08/2022]
Abstract
The anti-apoptotic protein HAX-1 has been proposed to modulate mitochondrial membrane potential, calcium signaling and actin remodeling. HAX-1 mutation or deficiency results in severe congenital neutropenia (SCN), loss of lymphocytes and neurological impairments by largely unknown mechanisms. Here, we demonstrate that the activation of c-Abl kinase in response to oxidative or genotoxic stress is dependent on HAX-1 association. Cellular reactive oxygen species (ROS) accumulation is inhibited by HAX-1-dependent c-Abl activation, which greatly contributes to the antiapoptotic role of HAX-1 in stress. HAX-1 (Q190X), a loss-of-function mutant responsible for SCN, fails to bind with and activate c-Abl, leading to dysregulated cellular ROS levels, damaged mitochondrial membrane potential and eventually apoptosis. The extensive apoptosis of lymphocytes and neurons in Hax-1-deficient mice could also be remarkably suppressed by c-Abl activation. These findings underline the important roles of ROS clearance in HAX-1-mediated anti-apoptosis by c-Abl kinase activation, providing new insight into the pathology and treatment of HAX-1-related hereditary disease or tumorigenesis.
Collapse
|
39
|
Miyamoto D, Takeuchi K, Chihara K, Fujieda S, Sada K. Protein tyrosine kinase Abl promotes hepatitis C virus particle assembly via interaction with viral substrate activator NS5A. J Biol Chem 2022; 298:101804. [PMID: 35257746 PMCID: PMC8980994 DOI: 10.1016/j.jbc.2022.101804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 02/26/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022] Open
Abstract
Previously, we reported that knockdown of Abl protein tyrosine kinase by shRNA or pharmacological inhibition suppresses particle assembly of J6/JFH1 strain–derived hepatitis C virus (HCV) in Huh-7.5 cells. However, the detailed mechanism by which Abl regulates HCV replication remained unclear. In this study, we established Abl-deficient (Abl−) cells through genome editing and compared HCV production between Abl− cells expressing WT or kinase-dead Abl and parental Huh-7.5 cells. Our findings revealed that Abl expression was not required from the stages of virus attachment and entry to viral gene expression; however, the kinase activity of Abl was necessary for the assembly of HCV particles. Reconstitution experiments using human embryonic kidney 293T cells revealed that phosphorylation of Tyr412 in the activation loop of Abl was enhanced by coexpression with the viral nonstructural protein 5A (NS5A) and was abrogated by the substitution of NS5A Tyr330 with Phe (Y330F), suggesting that NS5A functions as a substrate activator of Abl. Abl–NS5A association was also attenuated by the Y330F mutation of NS5A or the kinase-dead Abl, and Abl Tyr412 phosphorylation was not enhanced by NS5A bearing a mutation disabling homodimerization, although the association of Abl with NS5A was still observed. Taken together, these results demonstrate that Abl forms a phosphorylation-dependent complex with dimeric NS5A necessary for viral particle assembly, but that Abl is capable of complex formation with monomeric NS5A regardless of tyrosine phosphorylation. Our findings provide the foundation of a molecular basis for a new hepatitis C treatment strategy using Abl inhibitors.
Collapse
Affiliation(s)
- Daisuke Miyamoto
- Department of Otorhinolaryngology Head & Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Department of Genome Science and Microbiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
| | - Kenji Takeuchi
- Department of Genome Science and Microbiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Organization for Life Science Advancement Programs, University of Fukui, Fukui, Japan
| | - Kazuyasu Chihara
- Department of Genome Science and Microbiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Organization for Life Science Advancement Programs, University of Fukui, Fukui, Japan
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology Head & Neck Surgery, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Organization for Life Science Advancement Programs, University of Fukui, Fukui, Japan
| | - Kiyonao Sada
- Department of Genome Science and Microbiology, Faculty of Medical Sciences, University of Fukui, Fukui, Japan; Organization for Life Science Advancement Programs, University of Fukui, Fukui, Japan.
| |
Collapse
|
40
|
Population-based Targeted RNA Sequencing Reveals Novel Disease-related Gene Fusions in pediatric and adult T-ALL. Leuk Res 2022; 116:106825. [DOI: 10.1016/j.leukres.2022.106825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 03/04/2022] [Accepted: 03/10/2022] [Indexed: 11/21/2022]
|
41
|
Manley PW, Huth F, Moussaoui S, Schoepfer J. A kinase inhibitor which specifically targets the ABL myristate pocket (STAMP), but unlike asciminib crosses the blood–brain barrier. Bioorg Med Chem Lett 2022; 59:128577. [DOI: 10.1016/j.bmcl.2022.128577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/03/2022] [Accepted: 01/15/2022] [Indexed: 11/25/2022]
|
42
|
Alizadehmohajer N, Behmardi A, Najafgholian S, Moradi S, Mohammadi F, Nedaeinia R, Haghjooy Javanmard S, Sohrabi E, Salehi R, Ferns GA, Emami Nejad A, Manian M. Screening of potential inhibitors of COVID-19 with repurposing approach via molecular docking. NETWORK MODELING AND ANALYSIS IN HEALTH INFORMATICS AND BIOINFORMATICS 2022; 11:11. [PMID: 35136710 PMCID: PMC8814570 DOI: 10.1007/s13721-021-00341-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 07/14/2021] [Accepted: 10/01/2021] [Indexed: 01/20/2023]
Abstract
SARS-CoV-2 (COVID-19) is the causative organism for a pandemic disease with a high rate of infectivity and mortality. In this study, we aimed to assess the affinity between several available small molecule and proteins, including Abl kinase inhibitors, Janus kinase inhibitor, dipeptidyl peptidase 4 inhibitors, RNA-dependent RNA polymerase inhibitors, and Papain-like protease inhibitors, using binding simulation, to test whether they may be effective in inhibiting COVID-19 infection through several mechanisms. The efficiency of inhibitors was evaluated based on docking scores using AutoDock Vina software. Strong ligand-protein interactions were predicted among some of these drugs, that included: Imatinib, Remdesivir, and Telaprevir, and this may render these compounds promising candidates. Some candidate drugs might be efficient in disease control as potential inhibitors or lead compounds against the SARS-CoV-2. It is also worth highlighting the powerful immunomodulatory role of other drugs, such as Abivertinib that inhibits pro-inflammatory cytokine production associated with cytokine release syndrome (CRS) and the progression of COVID-19 infection. The potential role of other Abl kinase inhibitors, including Imatinib in reducing SARS-CoV and MERS-CoV viral titers, immune regulatory function and the development of acute respiratory distress syndrome (ARDS), indicate that this drug may be useful for COVID-19, as the SARS-CoV-2 genome is similar to SARS-CoV.
Collapse
Affiliation(s)
- Negin Alizadehmohajer
- Department of Medical Biotechnology and Translational Medicine, University of Milan, 20133 Milano, Italy
| | - Abtin Behmardi
- Cellular and Molecular Research Center, Research Institute for Prevention of Non-communicable, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Simin Najafgholian
- Department of Emergency Medicine, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Shabnam Moradi
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Forogh Mohammadi
- Department of Veterinary, Agriculture Faculty, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Ehsan Sohrabi
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Salehi
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Gordon A. Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Falmer, Brighton, BN1 9PH Sussex UK
| | - Asieh Emami Nejad
- Department of Biology, Payame Noor University (PNU), P.O. Box 19395-3697, Tehran, Iran
| | - Mostafa Manian
- Isfahan Neurosciences Research Center, Alzahra Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
43
|
Feng L, Fu S, Yao Y, Li Y, Xu L, Zhao Y, Luo L. Roles for c-Abl in postoperative neurodegeneration. Int J Med Sci 2022; 19:1753-1761. [PMID: 36313229 PMCID: PMC9608039 DOI: 10.7150/ijms.73740] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/01/2022] [Indexed: 11/07/2022] Open
Abstract
The nonreceptor tyrosine kinase c-Abl is inactive under normal conditions. Upon activation, c-Abl regulates signaling pathways related to cytoskeletal reorganization. It plays a vital role in modulating cell protrusion, cell migration, morphogenesis, adhesion, endocytosis and phagocytosis. A large number of studies have also found that abnormally activated c-Abl plays an important role in a variety of pathologies, including various inflammatory diseases and neurodegenerative diseases. c-Abl also plays a crucial role in neurodevelopment and neurodegenerative diseases, mainly through mechanisms such as neuroinflammation, oxidative stress (OS), and Tau protein phosphorylation. Inhibiting expression or activity of this kinase has certain neuroprotective and anti-inflammatory effects and can also improve cognition and behavior. Blockers of this kinase may have good preventive and treatment effects on neurodegenerative diseases. Cognitive dysfunction after anesthesia is also closely related to the abovementioned mechanisms. We infer that alterations in the expression and activity of c-Abl may underlie postoperative cognitive dysfunction (POCD). This article summarizes the current understanding and research progress on the mechanisms by which c-Abl may be related to postoperative neurodegeneration.
Collapse
Affiliation(s)
- Long Feng
- Department of Anesthesiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, China
| | - Shihui Fu
- Department of Cardiology, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, China.,Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yao Yao
- Center for the Study of Aging and Human Development and Geriatrics Division, Medical School of Duke University, North Carolina, USA.,Center for Healthy Aging and Development Studies, National School of Development, Peking University, Beijing, China
| | - Yulong Li
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Longhe Xu
- Department of Anesthesiology, The Third Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yali Zhao
- Central Laboratory, Hainan Hospital of Chinese People's Liberation Army General Hospital, Sanya, China
| | - Leiming Luo
- Department of Geriatric Cardiology, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
44
|
Mugoni V, Ciani Y, Nardella C, Demichelis F. Circulating RNAs in prostate cancer patients. Cancer Lett 2022; 524:57-69. [PMID: 34656688 DOI: 10.1016/j.canlet.2021.10.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 12/12/2022]
Abstract
Growing bodies of evidence have demonstrated that the identification of prostate cancer (PCa) biomarkers in the patients' blood and urine may remarkably improve PCa diagnosis and progression monitoring. Among diverse cancer-derived circulating materials, extracellular RNA molecules (exRNAs) represent a compelling component to investigate cancer-related alterations. Once outside the intracellular environment, exRNAs circulate in biofluids either in association with protein complexes or encapsulated inside extracellular vesicles (EVs). Notably, EV-associated RNAs (EV-RNAs) were used for the development of several assays (such as the FDA-approved Progensa Prostate Cancer Antigen 3 (PCA3 test) aiming at improving early PCa detection. EV-RNAs encompass a mixture of species, including small non-coding RNAs (e.g. miRNA and circRNA), lncRNAs and mRNAs. Several methods have been proposed to isolate EVs and relevant RNAs, and to perform RNA-Seq studies to identify potential cancer biomarkers. However, EVs in the circulation of a cancer patient include a multitude of diverse populations that are released by both cancer and normal cells from different tissues, thereby leading to a heterogeneous EV-RNA-associated transcriptional signal. Decrypting the complexity of such a composite signal is nowadays the major challenge faced in the identification of specific tumor-associated RNAs. Multiple deconvolution algorithms have been proposed so far to infer the enrichment of cancer-specific signals from gene expression data. However, novel strategies for EVs sorting and sequencing of RNA associated to single EVs populations will remarkably facilitate the identification of cancer-related molecules. Altogether, the studies summarized here demonstrate the high potential of using EV-RNA biomarkers in PCa and highlight the urgent need of improving technologies and computational approaches to characterize specific EVs populations and their relevant RNA cargo.
Collapse
Affiliation(s)
- Vera Mugoni
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Yari Ciani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Caterina Nardella
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy.
| |
Collapse
|
45
|
FDA-Approved Drugs for Hematological Malignancies-The Last Decade Review. Cancers (Basel) 2021; 14:cancers14010087. [PMID: 35008250 PMCID: PMC8750348 DOI: 10.3390/cancers14010087] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Hematological malignancies are diseases involving the abnormal production of blood cells. The aim of the study is to collect comprehensive information on new drugs used in the treatment of blood cancers which have introduced into therapy in the last decade. The approved drugs were analyzed for their structures and their biological activity mechanisms. Abstract Hematological malignancies, also referred to as blood cancers, are a group of diseases involving abnormal cell growth and persisting in the blood, lymph nodes, or bone marrow. The development of new targeted therapies including small molecule inhibitors, monoclonal antibodies, bispecific T cell engagers, antibody-drug conjugates, recombinant immunotoxins, and, finally, Chimeric Antigen Receptor T (CAR-T) cells has improved the clinical outcomes for blood cancers. In this review, we summarized 52 drugs that were divided into small molecule and macromolecule agents, approved by the Food and Drug Administration (FDA) in the period between 2011 and 2021 for the treatment of hematological malignancies. Forty of them have also been approved by the European Medicines Agency (EMA). We analyzed the FDA-approved drugs by investigating both their structures and mechanisms of action. It should be emphasized that the number of targeted drugs was significantly higher (46 drugs) than chemotherapy agents (6 drugs). We highlight recent advances in the design of drugs that are used to treat hematological malignancies, which make them more effective and less toxic.
Collapse
|
46
|
Bayazeid O, Rahman T. Correlation Analysis of Target Selectivity and Side Effects of FDA‐Approved Kinase Inhibitors**. ChemistrySelect 2021. [DOI: 10.1002/slct.202101367] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Omer Bayazeid
- Department of Pharmacognosy Faculty of Pharmacy Hacettepe University, Sihhiye 06100 Ankara Turkey
| | - Taufiq Rahman
- Department of Pharmacology University of Cambridge Tennis Court Road Cambridge CB2 1PD UK
| |
Collapse
|
47
|
Klejbor I, Shimshek DR, Klimaszewska-Łata J, Velasco-Estevez M, Moryś J, Karaszewski B, Szutowicz A, Rutkowska A. EBI2 is expressed in glial cells in multiple sclerosis lesions, and its knock-out modulates remyelination in the cuprizone model. Eur J Neurosci 2021; 54:5173-5188. [PMID: 34145920 DOI: 10.1111/ejn.15359] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 06/12/2021] [Indexed: 11/25/2022]
Abstract
EBI2 receptor regulates the immune system, and in multiple, sclerosis is upregulated in the central nervous system infiltrating lymphocytes. In newborn EBI2-deficient mice, myelin development is delayed, and its persistent antagonism inhibits remyelination in chemically demyelinated organotypic cerebellar slices. We used the cuprizone model of multiple sclerosis to elucidate the role of central nervous system-expressed EBI2 in de- and remyelination. The wild-type and EBI2 knock-out mice were fed 0.2% cuprizone in chow for 5 weeks and allowed to recover on a normal diet for 2 weeks. The data showed less efficient recovery of myelin, attenuated oligodendrocyte loss, fewer astrocytes and increased total cholesterol levels in the EBI2 knock-out mice after recovery. Moreover, the wild-type mice upregulated EBI2 expression after recovery confirming the involvement of EBI2 signalling during recovery from demyelination in the cuprizone model. The pro-inflammatory cytokine levels were at comparable levels in the wild-type and EBI2 knock-out mice, with only minor differences in TNFα and IL1β levels either at peak or during recovery. The neuroinflammatory signalling molecules, Abl1 kinase and NFКB1 (p105/p50) subunit, were significantly downregulated in the EBI2 knock-out mice at peak of disease. Immunohistochemical investigations of EBI2 receptor distribution in the central nervous system (CNS) cells in multiple sclerosis (MS) brain revealed strong expression of EBI2 in astrocytes and microglia inside the plaques implicating glia-expressed EBI2 in multiple sclerosis pathophysiology. Taken together, these findings demonstrate the involvement of EBI2 signalling in the recovery from demyelination rather than in demyelination and as such warrant further research into the role of EBI2 in remyelination.
Collapse
Affiliation(s)
- Ilona Klejbor
- Department of Anatomy and Physiology, Pomeranian University in Słupsk, Słupsk, Poland
| | - Derya R Shimshek
- Neuroscience, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | | | | | - Janusz Moryś
- Department of Anatomy and Neurobiology, Medical University of Gdańsk, Gdańsk, Poland
| | - Bartosz Karaszewski
- Division of Neurology, Department of Adult Neurology, Medical University of Gdańsk, Gdańsk, Poland
| | - Andrzej Szutowicz
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Rutkowska
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland.,Department of Anatomy and Neurobiology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
48
|
Wu K, Zhai X, Huang S, Jiang L, Yu Z, Huang J. Protein Kinases: Potential Drug Targets Against Schistosoma japonicum. Front Cell Infect Microbiol 2021; 11:691757. [PMID: 34277472 PMCID: PMC8282181 DOI: 10.3389/fcimb.2021.691757] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 06/18/2021] [Indexed: 12/24/2022] Open
Abstract
Schistosoma japonicum (S. japonicum) infection can induce serious organ damage and cause schistosomiasis japonica which is mainly prevalent in Asia and currently one of the most seriously neglected tropical diseases. Treatment of schistosomiasis largely depends on the drug praziquantel (PZQ). However, PZQ exhibits low killing efficacy on juvenile worms and the potential emergence of its drug resistance is a continual concern. Protein kinases (PKs) are enzymes that catalyze the phosphorylation of proteins and can participate in many signaling pathways in vivo. Recent studies confirmed the essential roles of PKs in the growth and development of S. japonicum, as well as in schistosome-host interactions, and researches have screened drug targets about PKs from S. japonicum (SjPKs), which provide new opportunities of developing new treatments on schistosomiasis. The aim of this review is to present the current progress on SjPKs from classification, different functions and their potential to become drug targets compared with other schistosomes. The efficiency of related protein kinase inhibitors on schistosomes is highlighted. Finally, the current challenges and problems in the study of SjPKs are proposed, which can provide future guidance for developing anti-schistosomiasis drugs and vaccines.
Collapse
Affiliation(s)
- Kaijuan Wu
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Xingyu Zhai
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Shuaiqin Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
| | - Liping Jiang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Zheng Yu
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China.,Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Jing Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China.,China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| |
Collapse
|
49
|
Wu K, Wu H, Lyu W, Kim Y, Furdui CM, Anderson KS, Koleske AJ. Platelet-derived growth factor receptor beta activates Abl2 via direct binding and phosphorylation. J Biol Chem 2021; 297:100883. [PMID: 34144039 PMCID: PMC8259415 DOI: 10.1016/j.jbc.2021.100883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 11/27/2022] Open
Abstract
Abl family kinases are nonreceptor tyrosine kinases activated by diverse cellular stimuli that regulate cytoskeleton organization, morphogenesis, and adhesion. The catalytic activity of Abl family kinases is tightly regulated in cells by a complex set of intramolecular and intermolecular interactions and post-translational modifications. For example, the platelet-derived growth factor receptor beta (PDGFRβ), important for cell proliferation and chemotaxis, is a potent activator of Abl family kinases. However, the molecular mechanism by which PDGFRβ engages and activates Abl family kinases is not known. We show here that the Abl2 Src homology 2 domain directly binds to phosphotyrosine Y771 in the PDGFRβ cytoplasmic domain. PDGFRβ directly phosphorylates multiple novel sites on the N-terminal half of Abl2, including Y116, Y139, and Y161 within the Src homology 3 domain, and Y299, Y303, and Y310 on the kinase domain. Y116, Y161, Y272, and Y310 are all located at or near the Src homology 3/Src homology 2-kinase linker interface, which helps maintain Abl family kinases in an autoinhibited conformation. We also found that PDGFRβ-mediated phosphorylation of Abl2 in vitro activates Abl2 kinase activity, but mutation of these four tyrosines (Y116, Y161, Y272, and Y310) to phenylalanine abrogated PDGFRβ-mediated activation of Abl2. These findings reveal how PDGFRβ engages and phosphorylates Abl2 leading to activation of the kinase, providing a framework to understand how growth factor receptors engage and activate Abl family kinases.
Collapse
Affiliation(s)
- Kuanlin Wu
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Hanzhi Wu
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Wanqing Lyu
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Youngjoo Kim
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Karen S Anderson
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA; Department of Pharmacology, Yale University, New Haven, Connecticut, USA
| | - Anthony J Koleske
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA; Department of Neuroscience, Yale University, New Haven, Connecticut, USA.
| |
Collapse
|
50
|
Yu Z, Seim I, Yin M, Tian R, Sun D, Ren W, Yang G, Xu S. Comparative analyses of aging-related genes in long-lived mammals provide insights into natural longevity. Innovation (N Y) 2021; 2:100108. [PMID: 34557758 PMCID: PMC8454735 DOI: 10.1016/j.xinn.2021.100108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 04/26/2021] [Indexed: 11/29/2022] Open
Abstract
Extreme longevity has evolved multiple times during the evolution of mammals, yet its underlying molecular mechanisms remain largely underexplored. Here, we compared the evolution of 115 aging-related genes in 11 long-lived species and 25 mammals with non-increased lifespan (control group) in the hopes of better understanding the common molecular mechanisms behind longevity. We identified 16 unique positively selected genes and 23 rapidly evolving genes in long-lived species, which included nine genes involved in regulating lifespan through the insulin/IGF-1 signaling (IIS) pathway and 11 genes highly enriched in immune-response-related pathways, suggesting that the IIS pathway and immune response play a particularly important role in exceptional mammalian longevity. Interestingly, 11 genes related to cancer progression, including four positively selected genes and seven genes with convergent amino acid changes, were shared by two or more long-lived lineages, indicating that long-lived mammals might have evolved convergent or similar mechanisms of cancer resistance that extended their lifespan. This suggestion was further corroborated by our identification of 12 robust candidates for longevity-related genes closely related to cancer. Evolution analyses of 115 aging-related genes exploring natural longevity in mammals Positively selected genes & rapidly evolved genes enriched in IIS and immune pathways Convergent mutations in genes associated with cancer in long-lived species Evolution of longevity through cancer resistance in long-lived mammals
Collapse
Affiliation(s)
- Zhenpeng Yu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Inge Seim
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.,Integrative Biology Laboratory, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China.,School of Biology and Environmental Science, Faculty of Science and Engineering, Queensland University of Technology, Brisbane, QLD, Australia
| | - Mengxin Yin
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Ran Tian
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Di Sun
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Wenhua Ren
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Guang Yang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Shixia Xu
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|