1
|
褚 乔, 王 小, 续 佳, 彭 荟, 赵 裕, 张 静, 陆 国, 王 恺. Pulsatilla saponin D inhibits invasion and metastasis of triple-negative breast cancer cells through multiple targets and pathways. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:150-161. [PMID: 39819723 PMCID: PMC11744280 DOI: 10.12122/j.issn.1673-4254.2025.01.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVES To explore the mechanism by which Pulsatilla saponin D (PSD) inhibits invasion and metastasis of triple-negative breast cancer (TNBC). METHODS The public databases were used to identify the potential targets of PSD and the invasion and metastasis targets of TNBC to obtain the intersection targets between PSD and TNBC. The "PSD-target-disease" interaction network was constructed and protein-protein interaction (PPI) analysis was performed to obtain the core targets, which were analyzed for KEGG pathway and GO functional enrichment. Molecular docking study of the core targets and PSD was performed, and the therapeutic effect and mechanism of PSD were verified using Transwell assay and Western blotting in cultured TNBC cells. RESULTS Network pharmacology analysis identified a total of 285 potential PSD targets and 26 drug-disease intersection core targets. GO analysis yielded 175 entries related to the binding of biomolecules (protein, DNA and RNA), enzyme activities, and regulation of gene transcription. KEGG analysis yielded 46 entries involving pathways in cancer, chemical carcinogenesis-receptor activation, microRNAs in cancer, chemical carcinogenesis-reactive oxygen species, PD-L1 expression and PD-1 checkpoint pathway in cancer. Molecular docking showed high binding affinities of PSD to MTOR, HDAC2, ABL1, CDK1, TLR4, TERT, PIK3R1, NFE2L2 and PTPN1. In cultured TNBC cells, treatment with PSD significantly inhibited cell invasion and migration and lowered the expressions of MMP2, MMP9, N-cadherin and the core proteins p-mTOR, ABL1, TERT, PTPN1, HDAC2, PIK3R1, CDK1, TLR4 as well as NFE2L2 expressionin the cell nuclei. CONCLUSIONS The inhibitory effects of PSD on TNBC invasion and metastasis are mediated by multiple targets and pathways.
Collapse
|
2
|
Wu H, Mao Y, Wang Q, Yu H, Bouaziz M, Makrides N, Koleske AJ, Radice GL, Zhang X. Abl kinases regulate FGF signaling independent of Crk phosphorylation to prevent Peters anomaly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.24.619064. [PMID: 39484567 PMCID: PMC11526961 DOI: 10.1101/2024.10.24.619064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Peters anomaly, the most common cause of congenital corneal opacity, stems from corneal-lenticular adhesion. Despite numerous identified mutations, a cohesive molecular framework of the disease's etiology remains elusive. Here, we identified Abl kinases as pivotal regulators of FGF signaling, as genetic ablation of Abl kinases restores lens induction even in the absence of FGF signaling. Intriguingly, both Abl kinase deficiency and increased FGF-Ras activity result in Peters anomaly independent of ERK signaling, which can be rescued by allelic deletion of Abl substrate, Crk. However, contrary to the prevailing belief that Abl kinases regulate Crk proteins by direct phosphorylation, mutations at Abl kinase phosphorylation sites on Crk and CrkL did not yield any observable effects. Instead, our findings reveal that Abl kinases phosphorylate Ptpn12, which in turn inhibits p130Cas phosphorylation and Crk recruitment, crucial for Rho GTPases activation and cytoskeletal dynamics. Consequently, Abl kinase deficiency reduces actomyosin contractility within the lens vesicle and genetically interacts with RhoA inhibition. Conversely, Rac1 deletion mitigates Peters anomaly in models with aberrant FGF, Abl kinase and RhoA signaling. Our results demonstrate that Abl kinases regulate FGF signaling to balance RhoA and Rac1 activity via the Ptpn12-p130Cas pathway, suggesting that targeting tension-mediated lens vesicle separation could be a therapeutic strategy for Peters anomaly.
Collapse
Affiliation(s)
- Hao Wu
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Yingyu Mao
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Qian Wang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Honglian Yu
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Michael Bouaziz
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Neoklis Makrides
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| | - Anthony J. Koleske
- Departments of Molecular Biophysics and Biochemistry and Neuroscience, Yale University, New Haven, CT 06520, USA
| | - Glenn L. Radice
- Department of Medicine, The Warren Alpert Medical School of Brown University, Lifespan Cardiovascular Institute, Rhode Island Hospital, Providence, RI 02903, USA
| | - Xin Zhang
- Departments of Ophthalmology, Pathology and Cell Biology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
3
|
Mezentsev A, Durymanov M, Makarov VA. A Comprehensive Review of Protein Biomarkers for Invasive Lung Cancer. Curr Oncol 2024; 31:4818-4854. [PMID: 39329988 PMCID: PMC11431409 DOI: 10.3390/curroncol31090360] [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: 07/25/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
Invasion and metastasis are important hallmarks of lung cancer, and affect patients' survival. Early diagnostics of metastatic potential are important for treatment management. Recent findings suggest that the transition to an invasive phenotype causes changes in the expression of 700-800 genes. In this context, the biomarkers restricted to the specific type of cancer, like lung cancer, are often overlooked. Some well-known protein biomarkers correlate with the progression of the disease and the immunogenicity of the tumor. Most of these biomarkers are not exclusive to lung cancer because of their significant role in tumorigenesis. The dysregulation of others does not necessarily indicate cell invasiveness, as they play an active role in cell division. Clinical studies of lung cancer use protein biomarkers to assess the invasiveness of cancer cells for therapeutic purposes. However, there is still a need to discover new biomarkers for lung cancer. In the future, minimally invasive techniques, such as blood or saliva analyses, may be sufficient for this purpose. Many researchers suggest unconventional biomarkers, like circulating nucleic acids, exosomal proteins, and autoantibodies. This review paper aims to discuss the advantages and limitations of protein biomarkers of invasiveness in lung cancer, to assess their prognostic value, and propose novel biomarker candidates.
Collapse
Affiliation(s)
- Alexandre Mezentsev
- Medical Informatics Laboratory, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia; (M.D.); (V.A.M.)
- Center for Theoretical Problems of Physicochemical Pharmacology, 109029 Moscow, Russia
| | - Mikhail Durymanov
- Medical Informatics Laboratory, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia; (M.D.); (V.A.M.)
| | - Vladimir A. Makarov
- Medical Informatics Laboratory, Yaroslav-the-Wise Novgorod State University, 173003 Veliky Novgorod, Russia; (M.D.); (V.A.M.)
| |
Collapse
|
4
|
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
|
5
|
Bao Q, Wang A, Hong W, Wang Y, Li B, He L, Yuan X, Ma G. The c-Abl-RACK1-FAK signaling axis promotes renal fibrosis in mice through regulating fibroblast-myofibroblast transition. Cell Commun Signal 2024; 22:247. [PMID: 38689280 PMCID: PMC11059681 DOI: 10.1186/s12964-024-01603-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Renal fibrosis is a prevalent manifestation of chronic kidney disease (CKD), and effective treatments for this disease are currently lacking. Myofibroblasts, which originate from interstitial fibroblasts, aggregate in the renal interstitium, leading to significant accumulation of extracellular matrix and impairment of renal function. The nonreceptor tyrosine kinase c-Abl (encoded by the Abl1 gene) has been implicated in the development of renal fibrosis. However, the precise role of c-Abl in this process and its involvement in fibroblast-myofibroblast transition (FMT) remain poorly understood. METHODS To investigate the effect of c-Abl in FMT during renal fibrosis, we investigated the expression of c-Abl in fibrotic renal tissues of patients with CKD and mouse models. We studied the phenotypic changes in fibroblast or myofibroblast-specific c-Abl conditional knockout mice. We explored the potential targets of c-Abl in NRK-49F fibroblasts. RESULTS In this study, fibrotic mouse and cell models demonstrated that c-Abl deficiency in fibroblasts mitigated fibrosis by suppressing fibroblast activation, fibroblast-myofibroblast transition, and extracellular matrix deposition. Mechanistically, c-Abl maintains the stability of the RACK1 protein, which serves as a scaffold for proteins such as c-Abl and focal adhesion kinase at focal adhesions, driving fibroblast activation and differentiation during renal fibrosis. Moreover, specifically targeting c-Abl deletion in renal myofibroblasts could prove beneficial in established kidney fibrosis by reducing RACK1 expression and diminishing the extent of fibrosis. CONCLUSIONS Our findings suggest that c-Abl plays a pathogenic role in interstitial fibrosis through the regulation of RACK1 protein stabilization and myofibroblast differentiation, suggesting a promising strategy for the treatment of CKD.
Collapse
Affiliation(s)
- Qianyi Bao
- School of Medicine, Southeast University, 87 Ding Jiaqiao Rd, Nanjing, 210009, P.R. China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Anyu Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Wenxuan Hong
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Yushu Wang
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Baojie Li
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Lin He
- School of Medicine, Southeast University, 87 Ding Jiaqiao Rd, Nanjing, 210009, P.R. China
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Xiaodong Yuan
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, P.R. China.
| | - Gang Ma
- School of Medicine, Southeast University, 87 Ding Jiaqiao Rd, Nanjing, 210009, P.R. China.
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, P.R. China.
| |
Collapse
|
6
|
Ge G, Wen Y, Li P, Guo Z, Liu Z. Single-Cell Plasmonic Immunosandwich Assay Reveals the Modulation of Nucleocytoplasmic Localization Fluctuation of ABL1 on Cell Migration. Anal Chem 2023; 95:17502-17512. [PMID: 38050674 DOI: 10.1021/acs.analchem.3c02593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
Cell migration is an essential process of cancer metastasis. The spatiotemporal dynamics of signaling molecules influences cellular phenotypic outcomes. It has been increasingly documented that the Abelson (ABL) family kinases play critical roles in solid tumors. However, ABL1's shuttling dynamics in cell migration still remains unexplored. This is mainly because tools permitting the investigation of translocation dynamics of proteins in single living cells are lacking. Herein, to bridge this gap, we developed a unique multifunctional integrated single-cell analysis method that enables long-term observation of cell migration behavior and monitoring of signaling proteins and complexes at the subcellular level. We found that the shuttling of ABL1's to the cytoplasm results in a higher migration speed, while its trafficking back to the nucleus leads to a lower one. Furthermore, our results indicated that fluctuant protein-protein interactions between 14-3-3 and ABL1 modulate ABL1's nucleocytoplasmic fluctuation and eventually affect the cell speed. Importantly, based on these new insights, we demonstrated that disturbing ABL1's nuclear export traffic and 14-3-3-ABL1 complexes formation can effectively suppress cell migration. Thus, our method opens up a new possibility for simultaneous tracking of internal molecular mechanisms and cell behavior, providing a promising tool for the in-depth study of cancer.
Collapse
Affiliation(s)
- Ge Ge
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Yanrong Wen
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Pengfei Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Zhanchen Guo
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| | - Zhen Liu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Nanjing University, 163 Xianlin Avenue, Nanjing 210023, China
| |
Collapse
|
7
|
Romeiro Motta M, Biswas S, Schaedel L. Beyond uniformity: Exploring the heterogeneous and dynamic nature of the microtubule lattice. Eur J Cell Biol 2023; 102:151370. [PMID: 37922811 DOI: 10.1016/j.ejcb.2023.151370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
A fair amount of research on microtubules since their discovery in 1963 has focused on their dynamic tips. In contrast, the microtubule lattice was long believed to be highly regular and static, and consequently received far less attention. Yet, as it turned out, the microtubule lattice is neither as regular, nor as static as previously believed: structural studies uncovered the remarkable wealth of different conformations the lattice can accommodate. In the last decade, the microtubule lattice was shown to be labile and to spontaneously undergo renovation, a phenomenon that is intimately linked to structural defects and was called "microtubule self-repair". Following this breakthrough discovery, further recent research provided a deeper understanding of the lattice self-repair mechanism, which we review here. Instrumental to these discoveries were in vitro microtubule reconstitution assays, in which microtubules are grown from the minimal components required for their dynamics. In this review, we propose a shift from the term "lattice self-repair" to "lattice dynamics", since this phenomenon is an inherent property of microtubules and can happen without microtubule damage. We focus on how in vitro microtubule reconstitution assays helped us learn (1) which types of structural variations microtubules display, (2) how these structural variations influence lattice dynamics and microtubule damage caused by mechanical stress, (3) how lattice dynamics impact tip dynamics, and (4) how microtubule-associated proteins (MAPs) can play a role in structuring the lattice. Finally, we discuss the unanswered questions about lattice dynamics and how technical advances will help us tackle these questions.
Collapse
Affiliation(s)
- Mariana Romeiro Motta
- Department of Physics, Center for Biophysics, Campus A2 4, Saarland University, 66123 Saarbrücken, Germany; Laboratoire Reproduction et Développement des Plantes, Université de Lyon, École normale supérieure de Lyon, Lyon 69364, France
| | - Subham Biswas
- Department of Physics, Center for Biophysics, Campus A2 4, Saarland University, 66123 Saarbrücken, Germany
| | - Laura Schaedel
- Department of Physics, Center for Biophysics, Campus A2 4, Saarland University, 66123 Saarbrücken, Germany.
| |
Collapse
|
8
|
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
|
9
|
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
|
10
|
Chen YC, Liao CC, Shui HA, Huang PH, Shih LJ. A Proteomics-Based Identification of the Biological Networks Mediating the Impact of Epigallocatechin-3-Gallate on Trophoblast Cell Migration and Invasion, with Potential Implications for Maternal and Fetal Health. Proteomes 2023; 11:31. [PMID: 37873873 PMCID: PMC10594419 DOI: 10.3390/proteomes11040031] [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: 07/13/2023] [Revised: 09/21/2023] [Accepted: 09/29/2023] [Indexed: 10/25/2023] Open
Abstract
Trophoblast migration and invasion play crucial roles in placental development. However, the effects of (-)-epigallocatechin-3-gallate (EGCG) on trophoblast cell functions remain largely unexplored. In this study, we investigated the impact of EGCG on the survival of trophoblast cells and employed a proteomics analysis to evaluate its influence on trophoblast cell migration and invasion. Be-Wo trophoblast cells were treated with EGCG, and a zone closure assay was conducted to assess the cell migration and invasion. Subsequently, a proteomics analysis was performed on the treated and control groups, followed by a bioinformatics analysis to evaluate the affected biological pathways and protein networks. A quantitative real-time PCR and Western blot analysis were carried out to validate the proteomics findings. Our results showed that EGCG significantly suppressed the trophoblast migration and invasion at a concentration not affecting cell survival. The proteomics analysis revealed notable differences in the protein expression between the EGCG-treated and control groups. Specifically, EGCG downregulated the signaling pathways related to EIF2, mTOR, and estrogen response, as well as the processes associated with the cytoskeleton, extracellular matrix, and protein translation. Conversely, EGCG upregulated the pathways linked to lipid degradation and oxidative metabolism. The quantitative PCR showed that EGCG modulated protein expression by regulating gene transcription, and the Western blot analysis confirmed its impact on cytoskeleton and extracellular matrix reorganization. These findings suggest EGCG may inhibit trophoblast migration and invasion through multiple signaling pathways, highlighting the potential risks associated with consuming EGCG-containing products during pregnancy. Future research should investigate the impact of EGCG intake on maternal and fetal proteoforms.
Collapse
Affiliation(s)
- Yueh-Chung Chen
- Department of Medicine, School of Medicine, National Defense Medical Center, Taipei 114201, Taiwan;
- Division of Cardiology, Department of Internal Medicine, Taipei City Hospital, Renai Branch, Taipei 106243, Taiwan
- Department of Health Promotion and Gerontological Care, Taipei University of Marine Technology, Taipei 111078, Taiwan
- Department of Special Education, University of Taipei, Taipei 100234, Taiwan
| | - Chen-Chung Liao
- Mass Spectrometry Facility, Instrumentation Resource Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (C.-C.L.)
- Cancer Progression Research Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hao-Ai Shui
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114201, Taiwan
| | - Pei-Hsuan Huang
- Mass Spectrometry Facility, Instrumentation Resource Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; (C.-C.L.)
| | - Li-Jane Shih
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114201, Taiwan
- Department of Medical Laboratory, Taoyuan Armed Forces General Hospital, Longtan, Taoyuan 325208, Taiwan
| |
Collapse
|
11
|
Fang HY, Forghani R, Clarke A, McQueen PG, Chandrasekaran A, O’Neill KM, Losert W, Papoian GA, Giniger E. Enabled primarily controls filopodial morphology, not actin organization, in the TSM1 growth cone in Drosophila. Mol Biol Cell 2023; 34:ar83. [PMID: 37223966 PMCID: PMC10398877 DOI: 10.1091/mbc.e23-01-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 05/25/2023] Open
Abstract
Ena/VASP proteins are processive actin polymerases that are required throughout animal phylogeny for many morphogenetic processes, including axon growth and guidance. Here we use in vivo live imaging of morphology and actin distribution to determine the role of Ena in promoting the growth of the TSM1 axon of the Drosophila wing. Altering Ena activity causes stalling and misrouting of TSM1. Our data show that Ena has a substantial impact on filopodial morphology in this growth cone but exerts only modest effects on actin distribution. This is in contrast to the main regulator of Ena, Abl tyrosine kinase, which was shown previously to have profound effects on actin and only mild effects on TSM1 growth cone morphology. We interpret these data as suggesting that the primary role of Ena in this axon may be to link actin to the morphogenetic processes of the plasma membrane, rather than to regulate actin organization itself. These data also suggest that a key role of Ena, acting downstream of Abl, may be to maintain consistent organization and reliable evolution of growth cone structure, even as Abl activity varies in response to guidance cues in the environment.
Collapse
Affiliation(s)
- Hsiao Yu Fang
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Rameen Forghani
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Akanni Clarke
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Philip G. McQueen
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| | - Aravind Chandrasekaran
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20752
| | - Kate M. O’Neill
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
- Institute for Physical Sciences and Department of Physics, University of Maryland, College Park, MD 20752
| | - Wolfgang Losert
- Institute for Physical Sciences and Department of Physics, University of Maryland, College Park, MD 20752
| | - Garegin A. Papoian
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20752
| | - Edward Giniger
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
12
|
Sprenger A, Carr HS, Ulu A, Frost JA. Src stimulates Abl-dependent phosphorylation of the guanine exchange factor Net1A to promote its cytosolic localization and cell motility. J Biol Chem 2023; 299:104887. [PMID: 37271338 PMCID: PMC10404680 DOI: 10.1016/j.jbc.2023.104887] [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: 08/24/2022] [Revised: 05/26/2023] [Accepted: 05/27/2023] [Indexed: 06/06/2023] Open
Abstract
The neuroepithelial cell transforming gene 1 (Net1) is a guanine nucleotide exchange factor for the small GTPase RhoA that promotes cancer cell motility and metastasis. Two isoforms of Net1 exist, Net1 and Net1A, both of which are sequestered in the nucleus in quiescent cells to prevent aberrant RhoA activation. Many cell motility stimuli drive cytosolic relocalization of Net1A, but mechanisms controlling this event are not fully understood. Here, we demonstrate that epithelial growth factor stimulates protein kinase Src- and Abl1-dependent phosphorylation of Net1A to promote its cytosolic localization. We show that Abl1 efficiently phosphorylates Net1A on Y373, and that phenylalanine substitution of Y373 prevents Net1A cytosolic localization. Furthermore, we found that Abl1-driven cytosolic localization of Net1A does not require S52, which is a phosphorylation site of a different kinase, c-Jun N-terminal kinase, that inhibits nuclear import of Net1A. However, we did find that MKK7-stimulated cytosolic localization of Net1A does require Y373. We also demonstrate that aspartate substitution at Y373 is sufficient to promote Net1A cytosolic accumulation, and expression of Net1A Y373D potentiates epithelial growth factor-stimulated RhoA activation, downstream myosin light chain 2 phosphorylation, and F-actin accumulation. Moreover, we show that expression of Net1A Y373D in breast cancer cells also significantly increases cell motility and Matrigel invasion. Finally, we show that Net1A is required for Abl1-stimulated cell motility, which is rescued by expression of Net1A Y373D, but not Net1A Y373F. Taken together, this work demonstrates a novel mechanism controlling Net1A subcellular localization to regulate RhoA-dependent cell motility and invasion.
Collapse
Affiliation(s)
- Ashabari Sprenger
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Heather S Carr
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Arzu Ulu
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Jeffrey A Frost
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, Texas, USA.
| |
Collapse
|
13
|
Escudero-Flórez M, Torres-Hoyos D, Miranda-Brand Y, Boudreau RL, Gallego-Gómez JC, Vicente-Manzanares M. Dengue Virus Infection Alters Inter-Endothelial Junctions and Promotes Endothelial-Mesenchymal-Transition-Like Changes in Human Microvascular Endothelial Cells. Viruses 2023; 15:1437. [PMID: 37515125 PMCID: PMC10386726 DOI: 10.3390/v15071437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
Dengue virus (DENV) is a pathogenic arbovirus that causes human disease. The most severe stage of the disease (severe dengue) is characterized by vascular leakage, hypovolemic shock, and organ failure. Endothelial dysfunction underlies these phenomena, but the causal mechanisms of endothelial dysfunction are poorly characterized. This study investigated the role of c-ABL kinase in DENV-induced endothelial dysfunction. Silencing c-ABL with artificial miRNA or targeting its catalytic activity with imatinib revealed that c-ABL is required for the early steps of DENV infection. DENV-2 infection and conditioned media from DENV-infected cells increased endothelial expression of c-ABL and CRKII phosphorylation, promoted expression of mesenchymal markers, e.g., vimentin and N-cadherin, and decreased the levels of endothelial-specific proteins, e.g., VE-cadherin and ZO-1. These effects were reverted by silencing or inhibiting c-ABL. As part of the acquisition of a mesenchymal phenotype, DENV infection and treatment with conditioned media from DENV-infected cells increased endothelial cell motility in a c-ABL-dependent manner. In conclusion, DENV infection promotes a c-ABL-dependent endothelial phenotypic change that leads to the loss of intercellular junctions and acquisition of motility.
Collapse
Affiliation(s)
- Manuela Escudero-Flórez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia; (M.E.-F.); (D.T.-H.); (Y.M.-B.)
| | - David Torres-Hoyos
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia; (M.E.-F.); (D.T.-H.); (Y.M.-B.)
| | - Yaneth Miranda-Brand
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia; (M.E.-F.); (D.T.-H.); (Y.M.-B.)
| | - Ryan L. Boudreau
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA;
| | - Juan Carlos Gallego-Gómez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia; (M.E.-F.); (D.T.-H.); (Y.M.-B.)
| | - 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), Universidad de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
14
|
Xue Y, Mei H, Chen Y, Griffin JD, Liu Q, Weisberg E, Yang J. Repurposing clinically available drugs and therapies for pathogenic targets to combat SARS-CoV-2. MedComm (Beijing) 2023; 4:e254. [PMID: 37193304 PMCID: PMC10183156 DOI: 10.1002/mco2.254] [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: 12/02/2022] [Revised: 02/11/2023] [Accepted: 03/07/2023] [Indexed: 05/18/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has affected a large portion of the global population, both physically and mentally. Current evidence suggests that the rapidly evolving coronavirus subvariants risk rendering vaccines and antibodies ineffective due to their potential to evade existing immunity, with enhanced transmission activity and higher reinfection rates that could lead to new outbreaks across the globe. The goal of viral management is to disrupt the viral life cycle as well as to relieve severe symptoms such as lung damage, cytokine storm, and organ failure. In the fight against viruses, the combination of viral genome sequencing, elucidation of the structure of viral proteins, and identifying proteins that are highly conserved across multiple coronaviruses has revealed many potential molecular targets. In addition, the time- and cost-effective repurposing of preexisting antiviral drugs or approved/clinical drugs for these targets offers considerable clinical advantages for COVID-19 patients. This review provides a comprehensive overview of various identified pathogenic targets and pathways as well as corresponding repurposed approved/clinical drugs and their potential against COVID-19. These findings provide new insight into the discovery of novel therapeutic strategies that could be applied to the control of disease symptoms emanating from evolving SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Yiying Xue
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Husheng Mei
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical ScienceChinese Academy of SciencesHefeiChina
- University of Science and Technology of ChinaHefeiAnhuiChina
| | - Yisa Chen
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - James D. Griffin
- Department of Medical Oncology, Dana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medicine, Harvard Medical SchoolBostonMassachusettsUSA
| | - Qingsong Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical ScienceChinese Academy of SciencesHefeiChina
- University of Science and Technology of ChinaHefeiAnhuiChina
- Hefei Cancer HospitalChinese Academy of SciencesHefeiChina
| | - Ellen Weisberg
- Department of Medical Oncology, Dana‐Farber Cancer InstituteBostonMassachusettsUSA
- Department of Medicine, Harvard Medical SchoolBostonMassachusettsUSA
| | - Jing Yang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and TechnologyTongji UniversityShanghaiChina
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical ScienceChinese Academy of SciencesHefeiChina
| |
Collapse
|
15
|
Roa-Linares VC, Escudero-Flórez M, Vicente-Manzanares M, Gallego-Gómez JC. Host Cell Targets for Unconventional Antivirals against RNA Viruses. Viruses 2023; 15:v15030776. [PMID: 36992484 PMCID: PMC10058429 DOI: 10.3390/v15030776] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/12/2023] [Accepted: 02/28/2023] [Indexed: 03/31/2023] Open
Abstract
The recent COVID-19 crisis has highlighted the importance of RNA-based viruses. The most prominent members of this group are SARS-CoV-2 (coronavirus), HIV (human immunodeficiency virus), EBOV (Ebola virus), DENV (dengue virus), HCV (hepatitis C virus), ZIKV (Zika virus), CHIKV (chikungunya virus), and influenza A virus. With the exception of retroviruses which produce reverse transcriptase, the majority of RNA viruses encode RNA-dependent RNA polymerases which do not include molecular proofreading tools, underlying the high mutation capacity of these viruses as they multiply in the host cells. Together with their ability to manipulate the immune system of the host in different ways, their high mutation frequency poses a challenge to develop effective and durable vaccination and/or treatments. Consequently, the use of antiviral targeting agents, while an important part of the therapeutic strategy against infection, may lead to the selection of drug-resistant variants. The crucial role of the host cell replicative and processing machinery is essential for the replicative cycle of the viruses and has driven attention to the potential use of drugs directed to the host machinery as therapeutic alternatives to treat viral infections. In this review, we discuss small molecules with antiviral effects that target cellular factors in different steps of the infectious cycle of many RNA viruses. We emphasize the repurposing of FDA-approved drugs with broad-spectrum antiviral activity. Finally, we postulate that the ferruginol analog (18-(phthalimide-2-yl) ferruginol) is a potential host-targeted antiviral.
Collapse
Affiliation(s)
- Vicky C Roa-Linares
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - Manuela Escudero-Flórez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| | - 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 C Gallego-Gómez
- Molecular and Translation Medicine Group, University of Antioquia, Medellin 050010, Colombia
| |
Collapse
|
16
|
c-Abl Tyrosine Kinase Is Required for BDNF-Induced Dendritic Branching and Growth. Int J Mol Sci 2023; 24:ijms24031944. [PMID: 36768268 PMCID: PMC9916151 DOI: 10.3390/ijms24031944] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 01/09/2023] [Accepted: 01/09/2023] [Indexed: 01/20/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) induces activation of the TrkB receptor and several downstream pathways (MAPK, PI3K, PLC-γ), leading to neuronal survival, growth, and plasticity. It has been well established that TrkB signaling regulation is required for neurite formation and dendritic arborization, but the specific mechanism is not fully understood. The non-receptor tyrosine kinase c-Abl is a possible candidate regulator of this process, as it has been implicated in tyrosine kinase receptors' signaling and trafficking, as well as regulation of neuronal morphogenesis. To assess the role of c-Abl in BDNF-induced dendritic arborization, wild-type and c-Abl-KO neurons were stimulated with BDNF, and diverse strategies were employed to probe the function of c-Abl, including the use of pharmacological inhibitors, an allosteric c-Abl activator, and shRNA to downregulates c-Abl expression. Surprisingly, BDNF promoted c-Abl activation and interaction with TrkB receptors. Furthermore, pharmacological c-Abl inhibition and genetic ablation abolished BDNF-induced dendritic arborization and increased the availability of TrkB in the cell membrane. Interestingly, inhibition or genetic ablation of c-Abl had no effect on the classic TrkB downstream pathways. Together, our results suggest that BDNF/TrkB-dependent c-Abl activation is a novel and essential mechanism in TrkB signaling.
Collapse
|
17
|
Cofre J, Saalfeld K. The first embryo, the origin of cancer and animal phylogeny. I. A presentation of the neoplastic process and its connection with cell fusion and germline formation. Front Cell Dev Biol 2023; 10:1067248. [PMID: 36684435 PMCID: PMC9846517 DOI: 10.3389/fcell.2022.1067248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/16/2022] [Indexed: 01/05/2023] Open
Abstract
The decisive role of Embryology in understanding the evolution of animal forms is founded and deeply rooted in the history of science. It is recognized that the emergence of multicellularity would not have been possible without the formation of the first embryo. We speculate that biophysical phenomena and the surrounding environment of the Ediacaran ocean were instrumental in co-opting a neoplastic functional module (NFM) within the nucleus of the first zygote. Thus, the neoplastic process, understood here as a biological phenomenon with profound embryologic implications, served as the evolutionary engine that favored the formation of the first embryo and cancerous diseases and allowed to coherently create and recreate body shapes in different animal groups during evolution. In this article, we provide a deep reflection on the Physics of the first embryogenesis and its contribution to the exaptation of additional NFM components, such as the extracellular matrix. Knowledge of NFM components, structure, dynamics, and origin advances our understanding of the numerous possibilities and different innovations that embryos have undergone to create animal forms via Neoplasia during evolutionary radiation. The developmental pathways of Neoplasia have their origins in ctenophores and were consolidated in mammals and other apical groups.
Collapse
Affiliation(s)
- Jaime Cofre
- Laboratório de Embriologia Molecular e Câncer, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil,*Correspondence: Jaime Cofre,
| | - Kay Saalfeld
- Laboratório de Filogenia Animal, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
18
|
Álvarez A, Gutiérrez D, Chandía-Cristi A, Yáñez M, Zanlungo S. c-Abl kinase at the crossroads of healthy synaptic remodeling and synaptic dysfunction in neurodegenerative diseases. Neural Regen Res 2023; 18:237-243. [PMID: 35900397 PMCID: PMC9396477 DOI: 10.4103/1673-5374.346540] [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] [Indexed: 11/28/2022] Open
Abstract
Our ability to learn and remember depends on the active formation, remodeling, and elimination of synapses. Thus, the development and growth of synapses as well as their weakening and elimination are essential for neuronal rewiring. The structural reorganization of synaptic complexes, changes in actin cytoskeleton and organelle dynamics, as well as modulation of gene expression, determine synaptic plasticity. It has been proposed that dysregulation of these key synaptic homeostatic processes underlies the synaptic dysfunction observed in many neurodegenerative diseases. Much is known about downstream signaling of activated N-methyl-D-aspartate and α-amino-3-hydroxy-5-methyl-4-isoazolepropionate receptors; however, other signaling pathways can also contribute to synaptic plasticity and long-lasting changes in learning and memory. The non-receptor tyrosine kinase c-Abl (ABL1) is a key signal transducer of intra and extracellular signals, and it shuttles between the cytoplasm and the nucleus. This review focuses on c-Abl and its synaptic and neuronal functions. Here, we discuss the evidence showing that the activation of c-Abl can be detrimental to neurons, promoting the development of neurodegenerative diseases. Nevertheless, c-Abl activity seems to be in a pivotal balance between healthy synaptic plasticity, regulating dendritic spines remodeling and gene expression after cognitive training, and synaptic dysfunction and loss in neurodegenerative diseases. Thus, c-Abl genetic ablation not only improves learning and memory and modulates the brain genetic program of trained mice, but its absence provides dendritic spines resiliency against damage. Therefore, the present review has been designed to elucidate the common links between c-Abl regulation of structural changes that involve the actin cytoskeleton and organelles dynamics, and the transcriptional program activated during synaptic plasticity. By summarizing the recent discoveries on c-Abl functions, we aim to provide an overview of how its inhibition could be a potentially fruitful treatment to improve degenerative outcomes and delay memory loss.
Collapse
|
19
|
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
|
20
|
Notch-dependent Abl signaling regulates cell motility during ommatidial rotation in Drosophila. Cell Rep 2022; 41:111788. [PMID: 36476875 PMCID: PMC9887719 DOI: 10.1016/j.celrep.2022.111788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/19/2022] [Accepted: 11/15/2022] [Indexed: 12/12/2022] Open
Abstract
A collective cell motility event that occurs during Drosophila eye development, ommatidial rotation (OR), serves as a paradigm for signaling-pathway-regulated directed movement of cell clusters. OR is instructed by the EGFR and Notch pathways and Frizzled/planar cell polarity (Fz/PCP) signaling, all of which are associated with photoreceptor R3 and R4 specification. Here, we show that Abl kinase negatively regulates OR through its activity in the R3/R4 pair. Abl is localized to apical junctional regions in R4, but not in R3, during OR, and this apical localization requires Notch signaling. We demonstrate that Abl and Notch interact genetically during OR, and Abl co-immunoprecipitates in complexes with Notch in eye discs. Perturbations of Abl interfere with adherens junctional organization of ommatidial preclusters, which mediate the OR process. Together, our data suggest that Abl kinase acts directly downstream of Notch in R4 to fine-tune OR via its effect on adherens junctions.
Collapse
|
21
|
Marín T, Valls C, Jerez C, Huerta T, Elgueta D, Vidal RL, Alvarez AR, Cancino GI. The c-Abl/p73 pathway induces neurodegeneration in a Parkinson's disease model. IBRO Neurosci Rep 2022; 13:378-387. [PMID: 36590096 PMCID: PMC9795287 DOI: 10.1016/j.ibneur.2022.10.006] [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: 01/31/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/08/2022] Open
Abstract
Parkinson's disease is the second most common neurodegenerative disorder. Although it is clear that dopaminergic neurons degenerate, the underlying molecular mechanisms are still unknown, and thus, successful treatment is still elusive. One pro-apoptotic pathway associated with several neurodegenerative diseases is the tyrosine kinase c-Abl and its target p73. Here, we evaluated the contribution of c-Abl and p73 in the degeneration of dopaminergic neurons induced by the neurotoxin 6-hydroxydopamine as a model for Parkinson's disease. First, we found that in SH-SY5Y cells treated with 6-hydroxydopamine, c-Abl and p73 phosphorylation levels were up-regulated. Also, we found that the pro-apoptotic p73 isoform TAp73 was up-regulated. Then, to evaluate whether c-Abl tyrosine kinase activity is necessary for 6-hydroxydopamine-induced apoptosis, we co-treated SH-SY5Y cells with 6-hydroxydopamine and Imatinib, a c-Abl specific inhibitor, observing that Imatinib prevented p73 phosphorylation, TAp73 up-regulation, and protected SH-SY5Y cells treated with 6-hydroxydopamine from apoptosis. Interestingly, this observation was confirmed in the c-Abl conditional null mice, where 6-hydroxydopamine stereotaxic injections induced a lesser reduction of dopaminergic neurons than in the wild-type mice significantly. Finally, we found that the intraperitoneal administration of Imatinib prevented the death of dopaminergic neurons induced by injecting 6-hydroxydopamine stereotaxically in the mice striatum. Thus, our findings support the idea that the c-Abl/p73 pathway is involved in 6-hydroxydopamine degeneration and suggest that inhibition of its kinase activity might be used as a therapeutical drug in Parkinson's disease.
Collapse
Affiliation(s)
- Tamara Marín
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Cristian Valls
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
| | - Carolina Jerez
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
| | - Tomás Huerta
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
| | - Daniela Elgueta
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
| | - René L. Vidal
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago 8580745, Chile
| | - Alejandra R. Alvarez
- Cell Signaling Laboratory, Department of Cellular and Molecular Biology, Millennium Institute on Immunology and Immunotherapy, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
- Corresponding author.
| | - Gonzalo I. Cancino
- Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago 8331010, Chile
- Corresponding author at: Center for Integrative Biology, Facultad de Ciencias, Universidad Mayor, Santiago 8580745, Chile.
| |
Collapse
|
22
|
Lan F, Chen X, Xiong Z, Cao Z, Lu L, Zhong Y, Zhan X, Yang Y, Shao Y, Li M, Han Z, Zhu X. Comprehensive transcriptomic and co-expression analysis of ABL1 gene and molecularly targeted drugs in hepatocellular carcinoma based on multi-database mining. MEDICAL ONCOLOGY (NORTHWOOD, LONDON, ENGLAND) 2022; 39:146. [PMID: 35834027 DOI: 10.1007/s12032-022-01730-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 04/05/2022] [Indexed: 11/28/2022]
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer death worldwide. Consequently, it is essential to identify biomarkers for treatment response and the prognosis prediction. We investigated whether ABL1 can function as a biomarker or a drug target for HCC. We assessed the ABL1 expression, genetic alterations and patients' survival from LinkedOmics, GEO, TCGA and Human Protein Atlas. We analyzed PPI, GO and KEGG pathways. GSEA was analyzed for functional comparison. The current drugs targeting ABL1 were statistically analyzed using DRUGSURV and DGIdb database. We found ABL1 is overexpressed in HCC and its higher expression reduces survival probability. Genetic changes of ABL1 are not frequent. We screened out 25 differentially expressed genes correlated with ABL1. The top functions of ABL1 are biological regulation, metabolic process, protein-containing, and protein binding. KEGG pathways showed that ABL1 and correlated with ABL1 significantly genes markedly enriched in the ErbB signaling pathway, and pathways in cancer. We counted the existing drugs targeting ABL1, which indicates that inhibiting ABL1 expression may improve the survival probability of HCC. In conclusion, ABL1 plays a crucial role in the development and progression of this cancerization and is a potential drug target.
Collapse
Affiliation(s)
- Feifei Lan
- Medical Genetics Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Xinqia Chen
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Zhuolong Xiong
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Zitong Cao
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Liangzong Lu
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Yueyuan Zhong
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Xuliang Zhan
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Yue Yang
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Yingqi Shao
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Minhua Li
- Zhu's Team, Guangdong Medical University, Zhanjiang, China
| | - Zenglei Han
- Department of Pathology, Qingdao Municipal Hospital, Qingdao, China.
| | - Xiao Zhu
- Zhu's Team, Guangdong Medical University, Zhanjiang, China. .,School of Laboratory Medicine and Biomedical Engineering, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
23
|
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
|
24
|
Bai H, Yang J, Meng S, Liu C. Oral Microbiota-Driven Cell Migration in Carcinogenesis and Metastasis. Front Cell Infect Microbiol 2022; 12:864479. [PMID: 35573798 PMCID: PMC9103474 DOI: 10.3389/fcimb.2022.864479] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/04/2022] [Indexed: 12/20/2022] Open
Abstract
The oral cavity harbors approximately 1,000 microbial species, and both pathogenic and commensal strains are involved in the development of carcinogenesis by stimulating chronic inflammation, affecting cell proliferation, and inhibiting cell apoptosis. Moreover, some substances produced by oral bacteria can also act in a carcinogenic manner. The link between oral microbiota and chronic inflammation as well as cell proliferation has been well established. Recently, increasing evidence has indicated the association of the oral microbiota with cell migration, which is crucial in regulating devastating diseases such as cancer. For instance, increased cell migration induced the spread of highly malignant cancer cells. Due to advanced technologies, the mechanistic understanding of cell migration in carcinogenesis and cancer metastasis is undergoing rapid progress. Thus, this review addressed the complexities of cell migration in carcinogenesis and cancer metastasis. We also integrate recent findings on the molecular mechanisms by which the oral microbiota regulates cell migration, with emphasis on the effect of the oral microbiota on adhesion, polarization, and guidance. Finally, we also highlight critical techniques, such as intravital microscopy and superresolution microscopy, for studies in this field.
Collapse
Affiliation(s)
- Huimin Bai
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shu Meng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Shu Meng, ; Chengcheng Liu,
| | - Chengcheng Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Periodontics, West China School & Hospital of Stomatology, Sichuan University, Chengdu, China
- *Correspondence: Shu Meng, ; Chengcheng Liu,
| |
Collapse
|
25
|
Naik RR, Shakya AK, Aladwan SM, El-Tanani M. Kinase Inhibitors as Potential Therapeutic Agents in the Treatment of COVID-19. Front Pharmacol 2022; 13:806568. [PMID: 35444538 PMCID: PMC9014181 DOI: 10.3389/fphar.2022.806568] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
Corona virus is quickly spreading around the world. The goal of viral management is to disrupt the virus's life cycle, minimize lung damage, and alleviate severe symptoms. Numerous strategies have been used, including repurposing existing antivirals or drugs used in previous viral outbreaks. One such strategy is to repurpose FDA-approved kinase inhibitors that are potential chemotherapeutic agents and have demonstrated antiviral activity against a variety of viruses, including MERS, SARS-CoV-1, and others, by inhibiting the viral life cycle and the inflammatory response associated with COVID-19. The purpose of this article is to identify licensed kinase inhibitors that have the ability to reduce the virus's life cycle, from entrance through viral propagation from cell to cell. Several of these inhibitors, including imatinib, ruxolitinib, silmitasertib, and tofacitinib (alone and in conjunction with hydroxychloroquine), are now undergoing clinical studies to determine their efficacy as a possible treatment drug. The FDA approved baricitinib (a Janus kinase inhibitor) in combination with remdesivir for the treatment of COVID-19 patients receiving hospital care in November 2020. While in vitro trials with gilteritinib, fedratinib, and osimertinib are encouraging, further research is necessary before these inhibitors may be used to treat COVID-19 patients.
Collapse
Affiliation(s)
- Rajashri R. Naik
- Department of Biopharmaceutics and Clinical Pharmacy, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Ashok K. Shakya
- Faculty of Pharmacy, Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman, Jordan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Safwan M. Aladwan
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| | - Mohamed El-Tanani
- Department of Biopharmaceutics and Clinical Pharmacy, Al-Ahliyya Amman University, Faculty of Pharmacy, Amman, Jordan
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
- Faculty of Pharmacy, Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman, Jordan
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| |
Collapse
|
26
|
Abstract
The global coronavirus disease-19 (COVID-19) has affected more than 140 million and killed more than 3 million people worldwide as of April 20, 2021. The novel human severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has been identified as an etiological agent for COVID-19. Several kinases have been proposed as possible mediators of multiple viral infections, including life-threatening coronaviruses like SARS-CoV-1, Middle East syndrome coronavirus (MERS-CoV), and SARS-CoV-2. Viral infections hijack abundant cell signaling pathways, resulting in drastic phosphorylation rewiring in the host and viral proteins. Some kinases play a significant role throughout the viral infection cycle (entry, replication, assembly, and egress), and several of them are involved in the virus-induced hyperinflammatory response that leads to cytokine storm, acute respiratory distress syndrome (ARDS), organ injury, and death. Here, we highlight kinases that are associated with coronavirus infections and their inhibitors with antiviral and potentially anti-inflammatory, cytokine-suppressive, or antifibrotic activity.
Collapse
Affiliation(s)
- Thanigaimalai Pillaiyar
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry
and Tuebingen Center for Academic Drug Discovery, Eberhard Karls University
Tübingen, Auf der Morgenstelle 8, 72076 Tübingen,
Germany
| | - Stefan Laufer
- Institute of Pharmacy, Pharmaceutical/Medicinal Chemistry
and Tuebingen Center for Academic Drug Discovery, Eberhard Karls University
Tübingen, Auf der Morgenstelle 8, 72076 Tübingen,
Germany
| |
Collapse
|
27
|
Werner MH, Olanow CW. Parkinson's Disease Modification through Abl Kinase Inhibition: An Opportunity. Mov Disord 2021; 37:6-15. [PMID: 34816484 PMCID: PMC8770606 DOI: 10.1002/mds.28858] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/07/2021] [Accepted: 10/29/2021] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease of the central nervous system, with an estimated 5 000 000 cases worldwide. Historically characterized by the progressive loss of dopaminergic neurons in the substantia nigra pars compacta, PD pathology is now known to be widespread and to affect serotonin, cholinergic and norepinephrine neurons as well as nerve cells in the olfactory system, cerebral hemisphere, brain stem, spinal cord, and peripheral autonomic nervous system. PD pathology is characterized by the accumulation of misfolded α-synuclein, which is thought to play a critical role in the etiopathogenesis of the disease. Animal models of PD suggest that activation of the Abelson tyrosine kinase (c-Abl) plays an essential role in the initiation and progression of α-synuclein pathology and neurodegeneration. These studies demonstrate that internalization of misfolded α-synuclein activates c-Abl, which phosphorylates α-synuclein and promotes α-synuclein pathology within the affected neurons. Additionally, c-Abl inactivates parkin, disrupting mitochondrial quality control and biogenesis, promoting neurodegeneration. Post-mortem studies of PD patients demonstrate increased levels of tyrosine phosphorylated α-synuclein, consistent with the activation of c-Abl in human disease. Although the c-Abl inhibitor nilotinib failed to demonstrate clinical benefit in two double-blind trials, novel c-Abl inhibitors have been developed that accumulate in the brain and may inhibit c-Abl at saturating levels. These novel inhibitors have demonstrated benefits in animal models of PD and have now entered clinical development. Here, we review the role of c-Abl in the neurodegenerative disease process and consider the translational potential of c-Abl inhibitors from model studies to disease-modifying therapies for Parkinson's disease. © 2021 Inhibikase Therapeutics, Inc. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson Movement Disorder Society.
Collapse
Affiliation(s)
| | - C Warren Olanow
- Department of Neurology and Department of Neuroscience, Mount Sinai School of Medicine, New York, New York, USA.,Clintrex Research Corporation, Sarasota, Florida, USA
| |
Collapse
|
28
|
Saha T, Gil-Henn H. Invadopodia, a Kingdom of Non-Receptor Tyrosine Kinases. Cells 2021; 10:cells10082037. [PMID: 34440806 PMCID: PMC8391121 DOI: 10.3390/cells10082037] [Citation(s) in RCA: 4] [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: 06/29/2021] [Revised: 08/05/2021] [Accepted: 08/06/2021] [Indexed: 01/01/2023] Open
Abstract
Non-receptor tyrosine kinases (NRTKs) are crucial mediators of intracellular signaling and control a wide variety of processes such as cell division, morphogenesis, and motility. Aberrant NRTK-mediated tyrosine phosphorylation has been linked to various human disorders and diseases, among them cancer metastasis, to which no treatment presently exists. Invasive cancer cells leaving the primary tumor use invadopodia, feet-like structures which facilitate extracellular matrix (ECM) degradation and intravasation, to escape the primary tumor and disseminate into distant tissues and organs during metastasis. A major challenge in metastasis research is to elucidate the molecular mechanisms and signaling pathways underlying invadopodia regulation, as the general belief is that targeting these structures can potentially lead to the eradication of cancer metastasis. Non-receptor tyrosine kinases (NRTKs) play a central role in regulating invadopodia formation and function, but how they coordinate the signaling leading to these processes was not clear until recently. Here, we describe the major NRTKs that rule invadopodia and how they work in concert while keeping an accurate hierarchy to control tumor cell invasiveness and dissemination.
Collapse
|
29
|
González-Martín A, Moyano T, Gutiérrez DA, Carvajal FJ, Cerpa W, Hanley JG, Gutiérrez RA, Álvarez AR. c-Abl regulates a synaptic plasticity-related transcriptional program involved in memory and learning. Prog Neurobiol 2021; 205:102122. [PMID: 34284000 DOI: 10.1016/j.pneurobio.2021.102122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 11/20/2022]
Abstract
Memory consolidation requires activation of a gene expression program that allows de novo protein synthesis. But the molecular mechanisms that favour or restrict that program are poorly understood. The kinase c-Abl can modulate gene expression through transcription factors and chromatin modifiers. Here, we show that c-Abl ablation in the brain improves learning acquisition and memory consolidation in mice. Its absence also affects gene expression profiles in the mouse hippocampus. We found that genes involved in synaptic plasticity and actin cytoskeleton dynamics, such as Arp2 and Thorase, are up-regulated at the mRNA and protein levels in trained c-Abl KO mice and by a chemical-LTP stimulus. Trained c-Abl KO mice also show that dendritic spines are larger than in wild-type mice and present at a higher density. These results indicate that c-Abl kinase is an important part of the mechanism that limits or restricts signalling of relevant gene programs involved in morphological and functional spine changes upon neuronal stimulation.
Collapse
Affiliation(s)
- Adrián González-Martín
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Centre for Aging and Regeneration (CARE-UC), Chile
| | - Tomás Moyano
- FONDAP Center for Genome Regulation, Millennium Institute for Integrative Biology (iBio), Department of Molecular Genetics and Microbiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Avda. Libertador Bernardo O'Higgins 340, Santiago, 8331150, Chile
| | - Daniela A Gutiérrez
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Centre for Aging and Regeneration (CARE-UC), Chile
| | - Franciso J Carvajal
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Center of Excellence in Biomedicine of Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Waldo Cerpa
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Centre for Aging and Regeneration (CARE-UC), Chile; Center of Excellence in Biomedicine of Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Jonathan G Hanley
- Centre for Synaptic Plasticity and School of Biochemistry, Biomedical Sciences Building, University of Bristol, University Walk, Bristol, BS8 1TD, UK
| | - Rodrigo A Gutiérrez
- FONDAP Center for Genome Regulation, Millennium Institute for Integrative Biology (iBio), Department of Molecular Genetics and Microbiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Avda. Libertador Bernardo O'Higgins 340, Santiago, 8331150, Chile
| | - Alejandra R Álvarez
- Department of Cell & Molecular Biology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile; Centre for Aging and Regeneration (CARE-UC), Chile.
| |
Collapse
|
30
|
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
|
31
|
Luttman JH, Colemon A, Mayro B, Pendergast AM. Role of the ABL tyrosine kinases in the epithelial-mesenchymal transition and the metastatic cascade. Cell Commun Signal 2021; 19:59. [PMID: 34022881 PMCID: PMC8140471 DOI: 10.1186/s12964-021-00739-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/16/2021] [Indexed: 12/20/2022] Open
Abstract
The ABL kinases, ABL1 and ABL2, promote tumor progression and metastasis in various solid tumors. Recent reports have shown that ABL kinases have increased expression and/or activity in solid tumors and that ABL inactivation impairs metastasis. The therapeutic effects of ABL inactivation are due in part to ABL-dependent regulation of diverse cellular processes related to the epithelial to mesenchymal transition and subsequent steps in the metastatic cascade. ABL kinases target multiple signaling pathways required for promoting one or more steps in the metastatic cascade. These findings highlight the potential utility of specific ABL kinase inhibitors as a novel treatment paradigm for patients with advanced metastatic disease. Video abstract.
Collapse
Affiliation(s)
- Jillian Hattaway Luttman
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Ashley Colemon
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Benjamin Mayro
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, 308 Research Drive, C-233A LSRC Bldg., P.O. Box 3813, Durham, NC 27710 USA
| |
Collapse
|
32
|
Novel Roles of SH2 and SH3 Domains in Lipid Binding. Cells 2021; 10:cells10051191. [PMID: 34068055 PMCID: PMC8152464 DOI: 10.3390/cells10051191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 01/07/2023] Open
Abstract
Signal transduction, the ability of cells to perceive information from the surroundings and alter behavior in response, is an essential property of life. Studies on tyrosine kinase action fundamentally changed our concept of cellular regulation. The induced assembly of subcellular hubs via the recognition of local protein or lipid modifications by modular protein interactions is now a central paradigm in signaling. Such molecular interactions are mediated by specific protein interaction domains. The first such domain identified was the SH2 domain, which was postulated to be a reader capable of finding and binding protein partners displaying phosphorylated tyrosine side chains. The SH3 domain was found to be involved in the formation of stable protein sub-complexes by constitutively attaching to proline-rich surfaces on its binding partners. The SH2 and SH3 domains have thus served as the prototypes for a diverse collection of interaction domains that recognize not only proteins but also lipids, nucleic acids, and small molecules. It has also been found that particular SH2 and SH3 domains themselves might also bind to and rely on lipids to modulate complex assembly. Some lipid-binding properties of SH2 and SH3 domains are reviewed here.
Collapse
|
33
|
Rogers EM, Allred SC, Peifer M. Abelson kinase's intrinsically disordered region plays essential roles in protein function and protein stability. Cell Commun Signal 2021; 19:27. [PMID: 33627133 PMCID: PMC7905622 DOI: 10.1186/s12964-020-00703-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 12/29/2020] [Indexed: 11/29/2022] Open
Abstract
Background The non-receptor tyrosine kinase Abelson (Abl) is a key player in oncogenesis, with kinase inhibitors serving as paradigms of targeted therapy. Abl also is a critical regulator of normal development, playing conserved roles in regulating cell behavior, brain development and morphogenesis. Drosophila offers a superb model for studying Abl’s normal function, because, unlike mammals, there is only a single fly Abl family member. In exploring the mechanism of action of multi-domain scaffolding proteins like Abl, one route is to define the roles of their individual domains. Research into Abl’s diverse roles in embryonic morphogenesis revealed many surprises. For instance, kinase activity, while important, is not crucial for all Abl activities, and the C-terminal F-actin binding domain plays a very modest role. This turned our attention to one of Abl’s least understood features—the long intrinsically-disordered region (IDR) linking Abl’s kinase and F-actin binding domains. The past decade revealed unexpected, important roles for IDRs in diverse cell functions, as sites of posttranslational modifications, mediating multivalent interactions and enabling assembly of biomolecular condensates via phase separation. Previous work deleting conserved regions in Abl’s IDR revealed an important role for a PXXP motif, but did not identify any other essential regions. Methods Here we extend this analysis by deleting the entire IDR, and asking whether Abl∆IDR rescues the diverse roles of Abl in viability and embryonic morphogenesis in Drosophila. Results This revealed that the IDR is essential for embryonic and adult viability, and for cell shape changes and cytoskeletal regulation during embryonic morphogenesis, and, most surprisingly, revealed a role in modulating protein stability. Conclusion Our data provide new insights into the role of the IDR in an important signaling protein, the non-receptor kinase Abl, suggesting that it is essential for all aspects of protein function during embryogenesis, and revealing a role in protein stability. These data will stimulate new explorations of the mechanisms by which the IDR regulates Abl stability and function, both in Drosophila and also in mammals. They also will stimulate further interest in the broader roles IDRs play in diverse signaling proteins. Video Abstract
Collapse
Affiliation(s)
- Edward M Rogers
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - S Colby Allred
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Mark Peifer
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
34
|
Liu H, Cui Y, Bai Y, Fang Y, Gao T, Wang G, Zhu L, Dong Q, Zhang S, Yao Y, Song C, Niu X, Jin Y, Li P, Cao C, Liu X. The tyrosine kinase c-Abl potentiates interferon-mediated antiviral immunity by STAT1 phosphorylation. iScience 2021; 24:102078. [PMID: 33644712 PMCID: PMC7887405 DOI: 10.1016/j.isci.2021.102078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/28/2020] [Accepted: 01/15/2021] [Indexed: 01/02/2023] Open
Abstract
Interferon (IFN)-induced activation of the signal transducer and activator of transcription (STAT) family is an important event in antiviral immunity. Here, we show that the nonreceptor kinases c-Abl and Arg directly interact with STAT1 and potentiate the phosphorylation of STAT1 on Y701. c-Abl/Arg could mediate STAT1 phosphorylation independent of Janus kinases in the absence of IFNγ and potentiate IFNγ-mediated STAT1 phosphorylation. Moreover, STAT1 dimerization, nuclear translocation, and downstream gene transcription are regulated by c-Abl/Arg. c-Abl/Arg (abl1/abl2) deficiency significantly suppresses antiviral responses in vesicular stomatitis virus-infected cells. Compared to vehicle, administration of the c-Abl/Arg selective inhibitor AMN107 resulted in significantly increased mortality in mice infected with human influenza virus. Our study demonstrates that c-Abl plays an essential role in the STAT1 activation signaling pathway and provides an important approach for antiviral immunity regulation.
Collapse
Affiliation(s)
- Hainan Liu
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yan Cui
- Beijing Institute of Biotechnology, Beijing 100850, China.,Staidson Bio-pharmaceutics (Beijing) Co. Ltd, Beijing 100176, China
| | - Yu Bai
- Anhui University, Hefei 230601, China
| | - Yi Fang
- The Fifth MedicaI Centre, Chinese PLA GeneraI HospitaI, Beijing 100071, China
| | - Ting Gao
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Guangfei Wang
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Lin Zhu
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Qincai Dong
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Shuwei Zhang
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yi Yao
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Caiwei Song
- Beijing Institute of Biotechnology, Beijing 100850, China
| | | | - Yanwen Jin
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Ping Li
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Cheng Cao
- Beijing Institute of Biotechnology, Beijing 100850, China
| | - Xuan Liu
- Beijing Institute of Biotechnology, Beijing 100850, China
| |
Collapse
|
35
|
Catozzi S, Halasz M, Kiel C. Predicted 'wiring landscape' of Ras-effector interactions in 29 human tissues. NPJ Syst Biol Appl 2021; 7:10. [PMID: 33580066 PMCID: PMC7881153 DOI: 10.1038/s41540-021-00170-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023] Open
Abstract
Ras is a plasma membrane (PM)-associated signaling hub protein that interacts with its partners (effectors) in a mutually exclusive fashion. We have shown earlier that competition for binding and hence the occurrence of specific binding events at a hub protein can modulate the activation of downstream pathways. Here, using a mechanistic modeling approach that incorporates high-quality proteomic data of Ras and 56 effectors in 29 (healthy) human tissues, we quantified the amount of individual Ras-effector complexes, and characterized the (stationary) Ras "wiring landscape" specific to each tissue. We identified nine effectors that are in significant amount in complex with Ras in at least one of the 29 tissues. We simulated both mutant- and stimulus-induced network re-configurations, and assessed their divergence from the reference scenario, specifically discussing a case study for two stimuli in three epithelial tissues. These analyses pointed to 32 effectors that are in significant amount in complex with Ras only if they are additionally recruited to the PM, e.g. via membrane-binding domains or domains binding to activated receptors at the PM. Altogether, our data emphasize the importance of tissue context for binding events at the Ras signaling hub.
Collapse
Affiliation(s)
- Simona Catozzi
- UCD Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, Dublin, 4, Ireland
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, 4, Ireland
| | - Melinda Halasz
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, 4, Ireland
| | - Christina Kiel
- UCD Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield, Dublin, 4, Ireland.
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, 4, Ireland.
| |
Collapse
|
36
|
Luo Y, Hu J, Liu Y, Li L, Li Y, Sun B, Kong R. Invadopodia: A potential target for pancreatic cancer therapy. Crit Rev Oncol Hematol 2021; 159:103236. [PMID: 33482351 DOI: 10.1016/j.critrevonc.2021.103236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 01/05/2021] [Accepted: 01/16/2021] [Indexed: 02/08/2023] Open
Abstract
Dissemination of cancer cells is an intricate multistep process that represents the most deadly aspect of cancer. Cancer cells form F-actin-rich protrusions known as invadopodia to invade surrounding tissues, blood vessels and lymphatics. A number of studies have demonstrated the significant roles of invadopodia in cancer. Therefore, the specific cells and molecules involved in invadopodia activity can provide as therapeutic targets. In this review, we included a thorough overview of studies in invadopodia and discussed their functions in cancer metastasis. We then presented the specific cells and molecules involved in invadopodia activity in pancreatic cancer and analyzed their suitability to be effective therapeutic targets. Currently, drugs targeting invadopodia and relevant clinical trials are negligible. Here, we highlighted the significance of potential drugs and discussed future obstacles in implementing clinical trials. This review presents a new perspective on invadopodia-induced pancreatic cancer metastasis and may prosper the development of targeted therapeutics against pancreatic cancer.
Collapse
Affiliation(s)
- Yan Luo
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jisheng Hu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yong Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Le Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilong Li
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Rui Kong
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China; Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
37
|
Yu HH, Zallen JA. Abl and Canoe/Afadin mediate mechanotransduction at tricellular junctions. Science 2021; 370:370/6520/eaba5528. [PMID: 33243859 DOI: 10.1126/science.aba5528] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 10/11/2020] [Indexed: 12/12/2022]
Abstract
Epithelial structure is generated by the dynamic reorganization of cells in response to mechanical forces. Adherens junctions transmit forces between cells, but how cells sense and respond to these forces in vivo is not well understood. We identify a mechanotransduction pathway involving the Abl tyrosine kinase and Canoe/Afadin that stabilizes cell adhesion under tension at tricellular junctions in the Drosophila embryo. Canoe is recruited to tricellular junctions in response to actomyosin contractility, and this mechanosensitivity requires Abl-dependent phosphorylation of a conserved tyrosine in the Canoe actin-binding domain. Preventing Canoe tyrosine phosphorylation destabilizes tricellular adhesion, and anchoring Canoe at tricellular junctions independently of mechanical inputs aberrantly stabilizes adhesion, arresting cell rearrangement. These results identify a force-responsive mechanism that stabilizes tricellular adhesion under tension during epithelial remodeling.
Collapse
Affiliation(s)
- Huapeng H Yu
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA
| | - Jennifer A Zallen
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering Institute, New York, NY, USA.
| |
Collapse
|
38
|
The NSs Protein Encoded by the Virulent Strain of Rift Valley Fever Virus Targets the Expression of Abl2 and the Actin Cytoskeleton of the Host, Affecting Cell Mobility, Cell Shape, and Cell-Cell Adhesion. J Virol 2020; 95:JVI.01768-20. [PMID: 33087469 DOI: 10.1128/jvi.01768-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/04/2020] [Indexed: 12/18/2022] Open
Abstract
Rift Valley fever virus (RVFV) is a highly pathogenic zoonotic arbovirus endemic in many African countries and the Arabian Peninsula. Animal infections cause high rates of mortality and abortion among sheep, goats, and cattle. In humans, an estimated 1 to 2% of RVFV infections result in severe disease (encephalitis, hepatitis, or retinitis) with a high rate of lethality when associated with hemorrhagic fever. The RVFV NSs protein, which is the main virulence factor, counteracts the host innate antiviral response to favor viral replication and spread. However, the mechanisms underlying RVFV-induced cytopathic effects and the role of NSs in these alterations remain for the most part unknown. In this work, we have analyzed the effects of NSs expression on the actin cytoskeleton while conducting infections with the NSs-expressing virulent (ZH548) and attenuated (MP12) strains of RVFV and the non-NSs-expressing avirulent (ZH548ΔNSs) strain, as well as after the ectopic expression of NSs. In macrophages, fibroblasts, and hepatocytes, NSs expression prevented the upregulation of Abl2 (a major regulator of the actin cytoskeleton) expression otherwise induced by avirulent infections and identified here as part of the antiviral response. The presence of NSs was also linked to an increased mobility of ZH548-infected cells compared to ZH548ΔNSs-infected fibroblasts and to strong changes in cell morphology in nonmigrating hepatocytes, with reduction of lamellipodia, cell spreading, and dissolution of adherens junctions reminiscent of the ZH548-induced cytopathic effects observed in vivo Finally, we show evidence of the presence of NSs within long actin-rich structures associated with NSs dissemination from NSs-expressing toward non-NSs-expressing cells.IMPORTANCE Rift Valley fever virus (RVFV) is a dangerous human and animal pathogen that was ranked by the World Health Organization in 2018 as among the eight pathogens of most concern for being likely to cause wide epidemics in the near future and for which there are no, or insufficient, countermeasures. The focus of this work is to address the question of the mechanisms underlying RVFV-induced cytopathic effects that participate in RVFV pathogenicity. We demonstrate here that RVFV targets cell adhesion and the actin cytoskeleton at the transcriptional and cellular level, affecting cell mobility and inducing cell shape collapse, along with distortion of cell-cell adhesion. All these effects may participate in RVFV-induced pathogenicity, facilitate virulent RVFV dissemination, and thus constitute interesting potential targets for future development of antiviral therapeutic strategies that, in the case of RVFV, as with several other emerging arboviruses, are presently lacking.
Collapse
|
39
|
Deville SS, Delgadillo Silva LF, Vehlow A, Cordes N. c-Abl Tyrosine Kinase Is Regulated Downstream of the Cytoskeletal Protein Synemin in Head and Neck Squamous Cell Carcinoma Radioresistance and DNA Repair. Int J Mol Sci 2020; 21:ijms21197277. [PMID: 33019757 PMCID: PMC7583921 DOI: 10.3390/ijms21197277] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/21/2020] [Accepted: 09/29/2020] [Indexed: 12/24/2022] Open
Abstract
The intermediate filament synemin has been previously identified as novel regulator of cancer cell therapy resistance and DNA double strand break (DSB) repair. c-Abl tyrosine kinase is involved in both of these processes. Using PamGene technology, we performed a broad-spectrum kinase activity profiling in three-dimensionally, extracellular matrix grown head and neck cancer cell cultures. Upon synemin silencing, we identified 86 deactivated tyrosine kinases, including c-Abl, in irradiated HNSCC cells. Upon irradiation and synemin inhibition, c-Abl hyperphosphorylation on tyrosine (Y) 412 and threonine (T) 735 was significantly reduced, prompting us to hypothesize that c-Abl tyrosine kinase is an important signaling component of the synemin-mediated radioresistance pathway. Simultaneous targeting of synemin and c-Abl resulted in similar radiosensitization and DSB repair compared with single synemin depletion, suggesting synemin as an upstream regulator of c-Abl. Immunoprecipitation assays revealed a protein complex formation between synemin and c-Abl pre- and post-irradiation. Upon pharmacological inhibition of ATM, synemin/c-Abl protein-protein interactions were disrupted implying synemin function to depend on ATM kinase activity. Moreover, deletion of the SH2 domain of c-Abl demonstrated a decrease in interaction, indicating the dependency of the protein-protein interaction on this domain. Mechanistically, radiosensitization upon synemin knockdown seems to be associated with an impairment of DNA repair via regulation of non-homologous end joining independent of c-Abl function. Our data generated in more physiological 3D cancer cell culture models suggest c-Abl as further key determinant of radioresistance downstream of synemin.
Collapse
Affiliation(s)
- Sara Sofia Deville
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.S.D.); (A.V.)
- Helmholtz-Zentrum Dresden—Rossendorf (HZDR), Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
| | | | - Anne Vehlow
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.S.D.); (A.V.)
- National Center for Tumor Diseases, Partner Site Dresden, German Cancer Research Center, 69120 Heidelberg, Germany
- German Cancer Consortium, Partner Site Dresden, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Nils Cordes
- OncoRay—National Center for Radiation Research in Oncology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (S.S.D.); (A.V.)
- Helmholtz-Zentrum Dresden—Rossendorf (HZDR), Institute of Radiooncology—OncoRay, 01328 Dresden, Germany
- German Cancer Consortium, Partner Site Dresden, German Cancer Research Center, 69120 Heidelberg, Germany
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Correspondence: ; Tel.: +49-(0)351–458–7401; Fax: +49-(0)351–458–7311
| |
Collapse
|
40
|
Tiwari V, Beer JC, Sankaranarayanan NV, Swanson-Mungerson M, Desai UR. Discovering small-molecule therapeutics against SARS-CoV-2. Drug Discov Today 2020; 25:1535-1544. [PMID: 32574699 PMCID: PMC7305878 DOI: 10.1016/j.drudis.2020.06.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/19/2020] [Accepted: 06/16/2020] [Indexed: 02/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly become a global health pandemic. The lack of effective treatments, coupled with its etiology, has resulted in more than 400,000 deaths at the time of writing. The SARS-CoV-2 genome is highly homologous to that of SARS-CoV, the causative agent behind the 2003 SARS outbreak. Based on prior reports, clinicians have pursued the off-label use of several antiviral drugs, while the scientific community has responded by seeking agents against traditional targets, especially viral proteases. However, several avenues remain unexplored, including disrupting E and M protein oligomerization, outcompeting host glycan-virus interactions, interfering with the heparan sulfate proteoglycans-virus interaction, and others. In this review, we highlight some of these opportunities while summarizing the drugs currently in use against coronavirus 2019 (COVID-19).
Collapse
Affiliation(s)
- Vaibhav Tiwari
- Department of Microbiology and Immunology, College of Graduate Studies, Midwestern University Downers Grove, IL 6051, USA; Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA.
| | - Jacob C Beer
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Nehru Viji Sankaranarayanan
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 2321, USA
| | - Michelle Swanson-Mungerson
- Department of Microbiology and Immunology, College of Graduate Studies, Midwestern University Downers Grove, IL 6051, USA; Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL 60515, USA
| | - Umesh R Desai
- Department of Medicinal Chemistry, School of Pharmacy, Virginia Commonwealth University, Richmond, VA 23298, USA; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, Richmond, VA 2321, USA.
| |
Collapse
|
41
|
Cheong HSJ, Nona M, Guerra SB, VanBerkum MF. The first quarter of the C-terminal domain of Abelson regulates the WAVE regulatory complex and Enabled in axon guidance. Neural Dev 2020; 15:7. [PMID: 32359359 PMCID: PMC7196227 DOI: 10.1186/s13064-020-00144-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/17/2020] [Indexed: 12/16/2022] Open
Abstract
Background Abelson tyrosine kinase (Abl) plays a key role in axon guidance in linking guidance receptors to actin dynamics. The long C-terminal domain (CTD) of Drosophila Abl is important for this role, and previous work identified the ‘first quarter’ (1Q) of the CTD as essential. Here, we link the physical interactions of 1Q binding partners to Abl’s function in axon guidance. Methods Protein binding partners of 1Q were identified by GST pulldown and mass spectrometry, and validated using axon guidance assays in the embryonic nerve cord and motoneurons. The role of 1Q was assessed genetically, utilizing a battery of Abl transgenes in combination with mutation or overexpression of the genes of pulled down proteins, and their partners in actin dynamics. The set of Abl transgenes had the following regions deleted: all of 1Q, each half of 1Q (‘eighths’, 1E and 2E) or a PxxP motif in 2E, which may bind SH3 domains. Results GST pulldown identified Hem and Sra-1 as binding partners of 1Q, and our genetic analyses show that both proteins function with Abl in axon guidance, with Sra-1 likely interacting with 1Q. As Hem and Sra-1 are part of the actin-polymerizing WAVE regulatory complex (WRC), we extended our analyses to Abi and Trio, which interact with Abl and WRC members. Overall, the 1Q region (and especially 2E and its PxxP motif) are important for Abl’s ability to work with WRC in axon guidance. These areas are also important for Abl’s ability to function with the actin regulator Enabled. In comparison, 1E contributes to Abl function with the WRC at the midline, but less so with Enabled. Conclusions The 1Q region, and especially the 2E region with its PxxP motif, links Abl with the WRC, its regulators Trio and Abi, and the actin regulator Ena. Removing 1E has specific effects suggesting it may help modulate Abl’s interaction with the WRC or Ena. Thus, the 1Q region of Abl plays a key role in regulating actin dynamics during axon guidance.
Collapse
Affiliation(s)
| | - Mark Nona
- Department of Biological Sciences, Wayne State University, Detroit, MI, 48202, USA
| | | | | |
Collapse
|
42
|
A case of imatinib-related obstructive bronchiolitis followed long term. Respir Med Case Rep 2020; 30:101052. [PMID: 32300523 PMCID: PMC7152670 DOI: 10.1016/j.rmcr.2020.101052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 03/26/2020] [Accepted: 04/02/2020] [Indexed: 11/21/2022] Open
Abstract
Obstructive bronchiolitis (OB) is an intractable disease causing stenosis in the surrounding bronchiolar region and bronchiolar lumen obstruction. Causes of OB are lung and hematopoietic stem-cell transplantation, collagen diseases, infections, and foods, but there are very few reports of drug-induced OB [1]. Imatinib is a drug used for the treatment of leukemia, gastrointestinal stromal tumors, etc. Although there are some reports of imatinib-induced lung injury as a complication (Ohnishi et al., 2006; Ma et al., 2003; Yamasawa et al., 2008; Koide et al., 2011) [[2], [3], [4], [5]], OB has not been reported. We have encountered a patient with OB related to imatinib administered for chronic myelogenous leukemia, who we have followed for 10 years. Drug-induced OB is very rare, but our case demonstrates the importance of considering the possibility of airway lesions by evaluating pulmonary function and expiratory computed tomography in patients with respiratory symptoms despite no shading on imaging.
Collapse
|
43
|
Gutierrez DA, Vargas LM, Chandia-Cristi A, de la Fuente C, Leal N, Alvarez AR. c-Abl Deficiency Provides Synaptic Resiliency Against Aβ-Oligomers. Front Cell Neurosci 2019; 13:526. [PMID: 31849613 PMCID: PMC6902026 DOI: 10.3389/fncel.2019.00526] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
Spine pathology has been implicated in the early onset of Alzheimer’s disease (AD), where Aβ-Oligomers (AβOs) cause synaptic dysfunction and loss. Previously, we described that pharmacological inhibition of c-Abl prevents AβOs-induced synaptic alterations. Hence, this kinase seems to be a key element in AD progression. Here, we studied the role of c-Abl on dendritic spine morphological changes induced by AβOs using c-Abl null neurons (c-Abl-KO). First, we characterized the effect of c-Abl deficiency on dendritic spine density and found that its absence increases dendritic spine density. While AβOs-treatment reduces the spine number in both wild-type (WT) and c-Abl-KO neurons, AβOs-driven spine density loss was not affected by c-Abl. We then characterized AβOs-induced morphological changes in dendritic spines of c-Abl-KO neurons. AβOs induced a decrease in the number of mushroom spines in c-Abl-KO neurons while preserving the populations of immature stubby, thin, and filopodia spines. Furthermore, synaptic contacts evaluated by PSD95/Piccolo clustering and cell viability were preserved in AβOs-exposed c-Abl-KO neurons. In conclusion, our results indicate that in the presence of AβOs c-Abl participates in synaptic contact removal, increasing susceptibility to AβOs damage. Its deficiency increases the immature spine population reducing AβOs-induced synapse elimination. Therefore, c-Abl signaling could be a relevant actor in the early stages of AD.
Collapse
Affiliation(s)
- Daniela A Gutierrez
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Lina M Vargas
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - América Chandia-Cristi
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina de la Fuente
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nancy Leal
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra R Alvarez
- Cell Signaling Laboratory, Faculty of Biological Science, Department of Cell and Molecular Biology, Center for Aging and Regeneration (CARE), Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
44
|
Pharmaco-Genetic Screen To Uncover Actin Regulators Targeted by Prostaglandins During Drosophila Oogenesis. G3-GENES GENOMES GENETICS 2019; 9:3555-3565. [PMID: 31506320 PMCID: PMC6829128 DOI: 10.1534/g3.119.400704] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Prostaglandins (PGs) are lipid signaling molecules with numerous physiologic functions, including pain/inflammation, fertility, and cancer. PGs are produced downstream of cyclooxygenase (COX) enzymes, the targets of non-steroidal anti-inflammatory drugs (NSAIDs). In numerous systems, PGs regulate actin cytoskeletal remodeling, however, their mechanisms of action remain largely unknown. To address this deficiency, we undertook a pharmaco-genetic interaction screen during late-stage Drosophila oogenesis. Drosophila oogenesis is as an established model for studying both actin dynamics and PGs. Indeed, during Stage 10B, cage-like arrays of actin bundles surround each nurse cell nucleus, and during Stage 11, the cortical actin contracts, squeezing the cytoplasmic contents into the oocyte. Both of these cytoskeletal properties are required for follicle development and fertility, and are regulated by PGs. Here we describe a pharmaco-genetic interaction screen that takes advantage of the fact that Stage 10B follicles will mature in culture and COX inhibitors, such as aspirin, block this in vitro follicle maturation. In the screen, aspirin was used at a concentration that blocks 50% of the wild-type follicles from maturing in culture. By combining this aspirin treatment with heterozygosity for mutations in actin regulators, we quantitatively identified enhancers and suppressors of COX inhibition. Here we present the screen results and initial follow-up studies on three strong enhancers – Enabled, Capping protein, and non-muscle Myosin II Regulatory Light Chain. Overall, these studies provide new insight into how PGs regulate both actin bundle formation and cellular contraction, properties that are not only essential for development, but are misregulated in disease.
Collapse
|
45
|
Ng CL, Qian Y, Schulz C. Notch and Delta are required for survival of the germline stem cell lineage in testes of Drosophila melanogaster. PLoS One 2019; 14:e0222471. [PMID: 31513679 PMCID: PMC6742463 DOI: 10.1371/journal.pone.0222471] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 08/29/2019] [Indexed: 01/27/2023] Open
Abstract
In all metazoan species, sperm is produced from germline stem cells. These self-renew and produce daughter cells that amplify and differentiate dependent on interactions with somatic support cells. In the male gonad of Drosophila melanogaster, the germline and somatic cyst cells co-differentiate as cysts, an arrangement in which the germline is completely enclosed by cytoplasmic extensions from the cyst cells. Notch is a developmentally relevant receptor in a pathway requiring immediate proximity with the signal sending cell. Here, we show that Notch is expressed in the cyst cells of wild-type testes. Notch becomes activated in the transition zone, an apical area of the testes in which the cyst cells express stage-specific transcription factors and the enclosed germline finalizes transit-amplifying divisions. Reducing the ligand Delta from the germline cells via RNA-Interference or reducing the receptor Notch from the cyst cells via CRISPR resulted in cell death concomitant with loss of germline cells from the transition zone. This shows that Notch signaling is essential for the survival of the germline stem cell lineage.
Collapse
Affiliation(s)
- Chun L. Ng
- University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Yue Qian
- University of North Georgia, Department of Biology, Oakwood, Georgia, United States of America
| | - Cordula Schulz
- University of Georgia, Department of Cellular Biology, Athens, Georgia, United States of America
| |
Collapse
|
46
|
Nishikawa M, Nakano S, Nakao H, Sato K, Sugiyama T, Akao Y, Nagaoka H, Yamakawa H, Nagase T, Ueda H. The interaction between PLEKHG2 and ABL1 suppresses cell growth via the NF-κB signaling pathway in HEK293 cells. Cell Signal 2019; 61:93-107. [PMID: 31100317 DOI: 10.1016/j.cellsig.2019.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 03/31/2019] [Accepted: 04/01/2019] [Indexed: 01/30/2023]
Abstract
The Rho family small GTPases mediate cell responses through actin cytoskeletal rearrangement. We previously reported that PLEKHG2, a Rho-specific guanine nucleotide exchange factor, is regulated via interaction with several proteins. We found that PLEKHG2 interacted with non-receptor tyrosine kinase ABL1, but the cellular function remains unclear. Here, we show that the interaction between PLEKHG2 and ABL1 attenuated the PLEKHG2-induced serum response element-dependent gene transcription in a tyrosine phosphorylation-independent manner. PLEKHG2 and ABL1 were co-localized and accumulated within cells co-expressing PLEKHG2 and ABL1. The cellular fractionation analysis suggested that the accumulation involved actin cytoskeletal reorganization. We also revealed that the co-expression of PLEKHG2 with ABL1, but not BCR-ABL, suppressed cell growth and synergistically enhanced NF-κB-dependent gene transcription. The cell growth suppression was canceled by co-expression with IκBα, a member of the NF-κB inhibitor protein family. This study suggests that the interaction between PLEKHG2 and ABL1 suppresses cell growth through intracellular protein accumulation via the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Masashi Nishikawa
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Yanagido 1-1, Gifu 501-1193, Japan
| | - Shun Nakano
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Yanagido 1-1, Gifu 501-1193, Japan
| | - Hiromu Nakao
- Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Yanagido 1-1, Gifu 501-1193, Japan
| | - Katsuya Sato
- Department of Molecular Pathobiochemistry, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu 501-1193, Japan
| | - Tsuyoshi Sugiyama
- Department of Medical Technology, School of Health Sciences, Gifu University of Medical Science, Nagamine Ichihiraga 795-1, Seki, Gifu 501-3892, Japan
| | - Yukihiro Akao
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Yanagido 1-1, Gifu 501-1193, Japan
| | - Hitoshi Nagaoka
- Department of Molecular Pathobiochemistry, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu 501-1193, Japan
| | | | | | - Hiroshi Ueda
- United Graduate School of Drug Discovery and Medical Information Sciences, Gifu University, Yanagido 1-1, Gifu 501-1193, Japan; Department of Chemistry and Biomolecular Science, Faculty of Engineering, Gifu University, Yanagido 1-1, Gifu 501-1193, Japan.
| |
Collapse
|
47
|
Kui F, Ye H, Chen XL, Zhang J. Microarray meta-analysis identifies candidate genes for human spermatogenic arrest. Andrologia 2019; 51:e13301. [PMID: 31037746 DOI: 10.1111/and.13301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 02/06/2023] Open
Abstract
Male infertility affects approximately half of couples who have difficulty becoming pregnant, and its prevalence is continuously rising. Many studies have been performed using animal testes to reveal the mechanisms of male infertility, but few studies have investigated human testes due to various limitations. The aim of this study was to investigate the gene expression profile of impaired human testes through a meta-analysis of microarray data sets, which was accomplished by using 178 human testis samples and 7 microarray data sets. Impaired testes were categorised into four pathological phenotypes or the normal phenotype based on their Johnsen score. Then, a meta-analysis was performed to screen out the differentially expressed genes (DEGs) in each phenotype. The DEGs were used in a subsequent bioinformatics analysis. Our results identified several novel hub genes and pathways and suggested that G1 mitotic cell cycle arrest was a remarkable feature in pre-meiotic arrest. Furthermore, fifteen p53-interacting proteins, such as ABL1 and HDAC2, whose roles in spermatogenesis have not been well characterised, were selected from the DEGs through a strict screening procedure.
Collapse
Affiliation(s)
- Fang Kui
- Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui Ye
- Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi-Ling Chen
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Jun Zhang
- Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
48
|
Ren WB, Xia XJ, Huang J, Guo WF, Che YY, Huang TH, Lei LC. Interferon-γ regulates cell malignant growth via the c-Abl/HDAC2 signaling pathway in mammary epithelial cells. J Zhejiang Univ Sci B 2019; 20:39-48. [PMID: 30614229 DOI: 10.1631/jzus.b1800211] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Interferon-γ (IFN-γ) has been used to control cancers in clinical treatment. However, an increasing number of reports have suggested that in some cases effectiveness declines after a long treatment period, the reason being unclear. We have reported previously that long-term IFN-γ treatment induces malignant transformation of healthy lactating bovine mammary epithelial cells (BMECs) in vitro. In this study, we investigated the mechanisms underlying the malignant proliferation of BMECs under IFN-γ treatment. The primary BMECs used in this study were stimulated by IFN-γ (10 ng/mL) for a long term to promote malignancy. We observed that IFN-γ could promote malignant cell proliferation, increase the expression of cyclin D1/cyclin-dependent kinase 4 (CDK4), decrease the expression of p21, and upregulate the expression of cellular-abelsongene (c-Abl) and histone deacetylase 2 (HDAC2). The HDAC2 inhibitor, valproate (VPA) and the c-Abl inhibitor, imatinib, lowered the expression level of cyclin D1/CDK4, and increased the expression level of p21, leading to an inhibitory effect on IFN-γ-induced malignant cell growth. When c-Abl was downregulated, the HDAC2 level was also decreased by promoted proteasome degradation. These data suggest that IFN-γ promotes the growth of malignant BMECs through the c-Abl/HDAC2 signaling pathway. Our findings suggest that long-term application of IFN-γ may be closely associated with the promotion of cell growth and even the carcinogenesis of breast cancer.
Collapse
Affiliation(s)
- Wen-Bo Ren
- College of Veterinary Medicine, Jilin University, Changchun 130062, China.,The First Hospital, Jilin University, Changchun 130021, China
| | - Xiao-Jing Xia
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang 453099, China
| | - Jing Huang
- The First Hospital, Jilin University, Changchun 130021, China
| | - Wen-Fei Guo
- The First Hospital, Jilin University, Changchun 130021, China
| | - Yan-Yi Che
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Ting-Hao Huang
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Lian-Cheng Lei
- College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
49
|
Torsello B, De Marco S, Bombelli S, Chisci E, Cassina V, Corti R, Bernasconi D, Giovannoni R, Bianchi C, Perego RA. The 1ALCTL and 1BLCTL isoforms of Arg/Abl2 induce fibroblast activation and extra cellular matrix remodelling differently. Biol Open 2019; 8:bio.038554. [PMID: 30837227 PMCID: PMC6451347 DOI: 10.1242/bio.038554] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The fibrotic tissue and the stroma adjacent to cancer cells are characterised by the presence of activated fibroblasts (myofibroblasts) which play a role in creating a supportive tissue characterised by abundant extracellular matrix (ECM) secretion. The myofibroblasts remodel this tissue through secreted molecules and modulation of their cytoskeleton and specialized contractile structures. The non-receptor protein tyrosine kinase Arg (also called Abl2) has the unique ability to bind directly to the actin cytoskeleton, transducing diverse extracellular signals into cytoskeletal rearrangements. In this study we analysed the 1ALCTL and 1BLCTL Arg isoforms in Arg−/− murine embryonal fibroblasts (MEF) cell line, focusing on their capacity to activate fibroblasts and to remodel ECM. The results obtained showed that Arg isoform 1BLCTL has a major role in proliferation, migration/invasion of MEF and in inducing a milieu able to modulate tumour cell morphology, while 1ALCTL isoform has a role in MEF adhesion maintaining active focal adhesions. On the whole, the presence of Arg in MEF supports the proliferation, activation, adhesion, ECM contraction and stiffness, while the absence of Arg affected these myofibroblast features. This article has an associated First Person interview with the first author of the paper. Summary: The non-receptor tyrosine kinase Arg and its isoforms modulate the extra cellular matrix production that is relevant in fibrosis and tumour growth, this may open future novel therapeutic approaches.
Collapse
Affiliation(s)
- Barbara Torsello
- School of Medicine & Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Sofia De Marco
- School of Medicine & Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Silvia Bombelli
- School of Medicine & Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Elisa Chisci
- School of Medicine & Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Valeria Cassina
- School of Medicine & Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Roberta Corti
- School of Medicine & Surgery, University of Milano-Bicocca, 20900 Monza, Italy.,Department of Materials Science, University of Milano-Bicocca, 20125 Milan, Italy
| | - Davide Bernasconi
- School of Medicine & Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Roberto Giovannoni
- School of Medicine & Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Cristina Bianchi
- School of Medicine & Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Roberto A Perego
- School of Medicine & Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
50
|
Host-directed kinase inhibitors act as novel therapies against intracellular Staphylococcus aureus. Sci Rep 2019; 9:4876. [PMID: 30890742 PMCID: PMC6425000 DOI: 10.1038/s41598-019-41260-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 03/05/2019] [Indexed: 01/09/2023] Open
Abstract
Host-directed therapeutics are a promising anti-infective strategy against intracellular bacterial pathogens. Repurposing host-targeted drugs approved by the FDA in the US, the MHRA in the UK and/or regulatory equivalents in other countries, is particularly interesting because these drugs are commercially available, safe doses are documented and they have been already approved for other clinical purposes. In this study, we aimed to identify novel therapies against intracellular Staphylococcus aureus, an opportunistic pathogen that is able to exploit host molecular and metabolic pathways to support its own intracellular survival. We screened 133 host-targeting drugs and found three host-directed tyrosine kinase inhibitors (Ibrutinib, Dasatinib and Crizotinib) that substantially impaired intracellular bacterial survival. We found that Ibrutinib significantly increased host cell viability after S. aureus infection via inhibition of cell invasion and intracellular bacterial proliferation. Using phosphoproteomics data, we propose a putative mechanism of action of Ibrutinib involving several host factors, including EPHA2, C-JUN and NWASP. We confirmed the importance of EPHA2 for staphylococcal infection in an EPHA2-knock-out cell line. Our study serves as an important example of feasibility for identifying host-directed therapeutics as candidates for repurposing.
Collapse
|