1
|
Yang LY, Tang DR, Luo SQ, Li WW, Jiang YH, Lin LB, Zhang QL. Time-dependent changes in genome-wide gene expression and post-transcriptional regulation across the post-death process in silkworm. DNA Res 2024; 31:dsae031. [PMID: 39546332 PMCID: PMC11605879 DOI: 10.1093/dnares/dsae031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/14/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024] Open
Abstract
Despite death marking the end of life, several gene expression and miRNA-mediated post-transcriptional regulation events may persist or be initiated. The silkworm (Bombyx mori) is a valuable model for exploring life processes, including death. In this study, we combined transcriptomics and miRNAomics analyses of young, old, and post-mortem silkworms across the entire process after death to unravel the dynamics of gene expression and miRNA-mediated post-transcriptional regulation. In total, 171 genes exhibited sustained differential expression in post-mortem silkworms compared to the pre-death state, which are primarily involved in nerve signalling, transport, and immune response. Post-mortem time-specific genes were associated with cell cycle regulation, thermogenesis, immunity, and zinc ion homeostasis. We found that the down-regulated expression of 36 genes related to transcription, epigenetic modification, and homeostasis resulted in a significant shift in global gene expression patterns at 2 h post-death. We also identified 5 mRNA-miRNA pairs (i.e. bmo-miR-2795-mhca, 2784-achi, 2762-oa1, 277-5p-creb, and 1000-tcb1) associated with stress hormone regulation, transcription activity, and signal transduction. The roles of these pairs were validated through in vivo experiments using miRNA mimics in silkworms. The findings provide valuable insights into the intricate mechanisms underlying the transcriptional and miRNA-mediated post-transcriptional regulation events in animals after death.
Collapse
Affiliation(s)
- Lin-Yu Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Da-Rui Tang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Shi-Qi Luo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Wei-Wei Li
- Kunming Institute of Zoology, The Chinese Academy of Sciences, Kunming 650201, China
| | - Yu-Hang Jiang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Lian-Bing Lin
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Qi-Lin Zhang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| |
Collapse
|
2
|
Venkateshwarlu A, Akshayveer, Singh S, Melnik R. Piezoelectricity and flexoelectricity in biological cells: the role of cell structure and organelles. Biomech Model Mechanobiol 2024:10.1007/s10237-024-01895-7. [PMID: 39455540 DOI: 10.1007/s10237-024-01895-7] [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: 04/22/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024]
Abstract
Living tissues experience various external forces on cells, influencing their behaviour, physiology, shape, gene expression, and destiny through interactions with their environment. Despite much research done in this area, challenges remain in our better understanding of the behaviour of the cell in response to external stimuli, including the arrangement, quantity, and shape of organelles within the cell. This study explores the electromechanical behaviour of biological cells, including organelles like microtubules, mitochondria, nuclei, and cell membranes. A two-dimensional bio-electromechanical model for two distinct cell structures has been developed to analyze the behavior of the biological cell to the external electrical and mechanical responses. The piezoelectric and flexoelectric effects have been included via multiphysics coupling for the biological cell. All the governing equations have been discretized and solved by the finite element method. It is found that the longitudinal stress is absent and only the transverse stress plays a crucial role when the mechanical load is imposed on the top side of the cell through compressive displacement. The impact of flexoelectricity is elucidated by introducing a new parameter called the maximum electric potential ratio ( V R , max ). It has been found that V R , max depends upon the orientation angle and shape of the microtubules. The magnitude of V R , max exhibit huge change when we change the shape and orientation of the organelles, which in some cases (boundary condition (BC)-3) can reach to three times of regular shape organelles. Further, the study reveals that the number of microtubules significantly impacts effective elastic and piezoelectric coefficients, affecting cell behavior based on structure, microtubule orientation, and mechanical stress direction. The insight obtained from the current study can assist in advancements in medical therapies such as tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Akepogu Venkateshwarlu
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, 75 University Avenue West, Waterloo, ON, N2L 3C5, Canada.
| | - Akshayveer
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, 75 University Avenue West, Waterloo, ON, N2L 3C5, Canada
| | - Sundeep Singh
- Faculty of Sustainable Design Engineering, University of Prince Edward Island, Charlottetown, PE, C1A 4P3, Canada
| | - Roderick Melnik
- MS2Discovery Interdisciplinary Research Institute, Wilfrid Laurier University, 75 University Avenue West, Waterloo, ON, N2L 3C5, Canada
| |
Collapse
|
3
|
Chu Z, Zhu M, Luo Y, Hu Y, Feng X, Shen J, Wang H, Sunagawa M, Liu Y. Terpene extract from the stem of Celastrus orbiculatus inhibits actin cytoskeleton remodelling in gastric cancer cells by regulating the protein interaction between PTBP1 and ACTN4. J Pharm Anal 2024; 14:101021. [PMID: 39263353 PMCID: PMC11388708 DOI: 10.1016/j.jpha.2024.101021] [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: 03/02/2024] [Revised: 06/02/2024] [Accepted: 06/11/2024] [Indexed: 09/13/2024] Open
Abstract
Adjuvant chemoradiotherapy, molecular targeted therapy, and immunotherapy are frequently employed to extend the survival of patients with advanced gastric cancer (GC). However, most of these treatments have toxic side effects, drug resistance, and limited improvements in survival and quality of life. Therefore, it is crucial to discover and develop new medications targeting GC that are highly effective and have minimal toxicity. In previous studies, the total terpene extract from the stem of Celastrus orbiculatus demonstrated anti-GC activity; however, the specific mechanism was unclear. Our research utilising co-immunoprecipitation-mass spectrometry (Co-IP-MS), polypyrimidine tract binding protein 1 (ptbp1) clustered regularly interspaced short palindromic repeat-associated protein 9 (Cas9)-knockout (KO) mouse model, tissue microarray, and functional experiments suggests that alpha actinin-4 (ACTN4) could be a significant biomarker of GC. PTBP1 influences actin cytoskeleton restructuring in GC cells by interacting with ACTN4. Celastrus orbiculatus stem extract (COE) may directly target ACTN4 and affect the interaction between PTBP1 and ACTN4, thereby exerting anti-GC effects.
Collapse
Affiliation(s)
- Zewen Chu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, Jiangsu, 225001, China
| | - Miao Zhu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, Jiangsu, 225001, China
| | - Yuanyuan Luo
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, Jiangsu, 225001, China
| | - Yaqi Hu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, Jiangsu, 225001, China
| | - Xinyi Feng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, Jiangsu, 225001, China
| | - Jiacheng Shen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, Jiangsu, 225001, China
| | - Haibo Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, Jiangsu, 225001, China
| | - Masataka Sunagawa
- Department of Physiology, School of Medicine, Showa University, Tokyo, 142-8555, Japan
| | - Yanqing Liu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, 225001, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, Jiangsu, 225001, China
| |
Collapse
|
4
|
Park J, Wu Y, Suk Kim J, Byun J, Lee J, Oh YK. Cytoskeleton-modulating nanomaterials and their therapeutic potentials. Adv Drug Deliv Rev 2024; 211:115362. [PMID: 38906478 DOI: 10.1016/j.addr.2024.115362] [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: 03/14/2024] [Revised: 05/25/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
The cytoskeleton, an intricate network of protein fibers within cells, plays a pivotal role in maintaining cell shape, enabling movement, and facilitating intracellular transport. Its involvement in various pathological states, ranging from cancer proliferation and metastasis to the progression of neurodegenerative disorders, underscores its potential as a target for therapeutic intervention. The exploration of nanotechnology in this realm, particularly the use of nanomaterials for cytoskeletal modulation, represents a cutting-edge approach with the promise of novel treatments. Inorganic nanomaterials, including those derived from gold, metal oxides, carbon, and black phosphorus, alongside organic variants such as peptides and proteins, are at the forefront of this research. These materials offer diverse mechanisms of action, either by directly interacting with cytoskeletal components or by influencing cellular signaling pathways that, in turn, modulate the cytoskeleton. Recent advancements have introduced magnetic field-responsive and light-responsive nanomaterials, which allow for targeted and controlled manipulation of the cytoskeleton. Such precision is crucial in minimizing off-target effects and enhancing therapeutic efficacy. This review explores the importance of research into cytoskeleton-targeting nanomaterials for developing therapeutic interventions for a range of diseases. It also addresses the progress made in this field, the challenges encountered, and future directions for using nanomaterials to modulate the cytoskeleton. The continued exploration of nanomaterials for cytoskeleton modulation holds great promise for advancing therapeutic strategies against a broad spectrum of diseases, marking a significant step forward in the intersection of nanotechnology and medicine.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jung Suk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junho Byun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
5
|
Li Z, Zhou T, Bao Z, Wu M, Mao Y. The Porous SilMA Hydrogel Scaffolds Carrying Dual-Sensitive Paclitaxel Nanoparticles Promote Neuronal Differentiation for Spinal Cord Injury Repair. Tissue Eng Regen Med 2024; 21:809-827. [PMID: 39004636 PMCID: PMC11286913 DOI: 10.1007/s13770-024-00659-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND In the intricate pathological milieu post-spinal cord injury (SCI), neural stem cells (NSCs) frequently differentiate into astrocytes rather than neurons, significantly limiting nerve repair. Hence, the utilization of biocompatible hydrogel scaffolds in conjunction with exogenous factors to foster the differentiation of NSCs into neurons has the potential for SCI repair. METHODS In this study, we engineered a 3D-printed porous SilMA hydrogel scaffold (SM) supplemented with pH-/temperature-responsive paclitaxel nanoparticles (PTX-NPs). We analyzed the biocompatibility of a specific concentration of PTX-NPs and its effect on NSC differentiation. We also established an SCI model to explore the ability of composite scaffolds for in vivo nerve repair. RESULTS The physical adsorption of an optimal PTX-NPs dosage can simultaneously achieve pH/temperature-responsive release and commendable biocompatibility, primarily reflected in cell viability, morphology, and proliferation. An appropriate PTX-NPs concentration can steer NSC differentiation towards neurons over astrocytes, a phenomenon that is also efficacious in simulated injury settings. Immunoblotting analysis confirmed that PTX-NPs-induced NSC differentiation occurred via the MAPK/ERK signaling cascade. The repair of hemisected SCI in rats demonstrated that the composite scaffold augmented neuronal regeneration at the injury site, curtailed astrocyte and fibrotic scar production, and enhanced motor function recovery in rat hind limbs. CONCLUSION The scaffold's porous architecture serves as a cellular and drug carrier, providing a favorable microenvironment for nerve regeneration. These findings corroborate that this strategy amplifies neuronal expression within the injury milieu, significantly aiding in SCI repair.
Collapse
Affiliation(s)
- Zhixiang Li
- School of Life Sciences, Bengbu Medical University, 2600 Donghai Road, Bengbu, 233030, China
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Tao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Zhengqi Bao
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China
| | - Min Wu
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China.
| | - Yingji Mao
- School of Life Sciences, Bengbu Medical University, 2600 Donghai Road, Bengbu, 233030, China.
- Department of Orthopedics, The First Affiliated Hospital of Bengbu Medical University, 287 Changhuai Road, Bengbu, 233004, China.
- Anhui Engineering Research Center of Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
6
|
Daly ML, Nishi K, Klawa SJ, Hinton KY, Gao Y, Freeman R. Designer peptide-DNA cytoskeletons regulate the function of synthetic cells. Nat Chem 2024; 16:1229-1239. [PMID: 38654104 PMCID: PMC11322001 DOI: 10.1038/s41557-024-01509-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 03/15/2024] [Indexed: 04/25/2024]
Abstract
The bottom-up engineering of artificial cells requires a reconfigurable cytoskeleton that can organize at distinct locations and dynamically modulate its structural and mechanical properties. Here, inspired by the vast array of actin-binding proteins and their ability to reversibly crosslink or bundle filaments, we have designed a library of peptide-DNA crosslinkers varying in length, valency and geometry. Peptide filaments conjoint through DNA hybridization give rise to tactoid-shaped bundles with tunable aspect ratios and mechanics. When confined in cell-sized water-in-oil droplets, the DNA crosslinker design guides the localization of cytoskeletal structures at the cortex or within the lumen of the synthetic cells. The tunable spatial arrangement regulates the passive diffusion of payloads within the droplets and complementary DNA handles allow for the reversible recruitment and release of payloads on and off the cytoskeleton. Heat-induced reconfiguration of peptide-DNA architectures triggers shape deformations of droplets, regulated by DNA melting temperatures. Altogether, the modular design of peptide-DNA architectures is a powerful strategy towards the bottom-up assembly of synthetic cells.
Collapse
Affiliation(s)
- Margaret L Daly
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Kengo Nishi
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Stephen J Klawa
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Kameryn Y Hinton
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Yuan Gao
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA
| | - Ronit Freeman
- Department of Applied Physical Sciences, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
7
|
Ferris E, Gonzalez Murcia JD, Cristina Rodriguez A, Steinwand S, Stacher Hörndli C, Traenkner D, Maldonado-Catala PJ, Gregg C. Genomic Convergence in Hibernating Mammals Elucidates the Genetics of Metabolic Regulation in the Hypothalamus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600891. [PMID: 38979381 PMCID: PMC11230405 DOI: 10.1101/2024.06.26.600891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Elucidating the genetic basis of mammalian metabolism could help define mechanisms central to health and disease. Here, we define conserved cis-regulatory elements (CREs) and programs for mammalian metabolic control. We delineate gene expression and chromatin responses in the mouse hypothalamus for 7 steps of the Fed-to-Fasted-to-Refed (FFR) response process. Comparative genomics of hibernating versus non-hibernating lineages then illuminates cis-elements showing convergent changes in hibernators. Hibernators accumulated loss-of-function effects for specific CREs regulating hypothalamic FFR responses. Multi-omics approaches pinpoint key CREs, genes, regulatory programs, and cell types in the divergence of hibernating and homeothermic lineages. The refeeding period after extended fasting is revealed as one critical period of chromatin remodeling with convergent genomic changes. This genetic framework is a step toward harnessing hibernator adaptations in medicine.
Collapse
Affiliation(s)
- Elliott Ferris
- Departments of Neurobiology, University of Utah; Salt Lake City, 84105, USA
| | | | | | - Susan Steinwand
- Departments of Neurobiology, University of Utah; Salt Lake City, 84105, USA
| | | | - Dimitri Traenkner
- Departments of Neurobiology, University of Utah; Salt Lake City, 84105, USA
| | - Pablo J Maldonado-Catala
- Departments of Neurobiology, University of Utah; Salt Lake City, 84105, USA
- Biomedical Informatics, University of Utah; Salt Lake City, 84105, USA
| | - Christopher Gregg
- Departments of Neurobiology, University of Utah; Salt Lake City, 84105, USA
- Human Genetics, University of Utah; Salt Lake City, 84105, USA
| |
Collapse
|
8
|
Latypova AA, Yaremenko AV, Pechnikova NA, Minin AS, Zubarev IV. Magnetogenetics as a promising tool for controlling cellular signaling pathways. J Nanobiotechnology 2024; 22:327. [PMID: 38858689 PMCID: PMC11163773 DOI: 10.1186/s12951-024-02616-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: 03/28/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024] Open
Abstract
Magnetogenetics emerges as a transformative approach for modulating cellular signaling pathways through the strategic application of magnetic fields and nanoparticles. This technique leverages the unique properties of magnetic nanoparticles (MNPs) to induce mechanical or thermal stimuli within cells, facilitating the activation of mechano- and thermosensitive proteins without the need for traditional ligand-receptor interactions. Unlike traditional modalities that often require invasive interventions and lack precision in targeting specific cellular functions, magnetogenetics offers a non-invasive alternative with the capacity for deep tissue penetration and the potential for targeting a broad spectrum of cellular processes. This review underscores magnetogenetics' broad applicability, from steering stem cell differentiation to manipulating neuronal activity and immune responses, highlighting its potential in regenerative medicine, neuroscience, and cancer therapy. Furthermore, the review explores the challenges and future directions of magnetogenetics, including the development of genetically programmed magnetic nanoparticles and the integration of magnetic field-sensitive cells for in vivo applications. Magnetogenetics stands at the forefront of cellular manipulation technologies, offering novel insights into cellular signaling and opening new avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Anastasiia A Latypova
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia
- Moscow Center for Advanced Studies, Moscow, 123592, Russia
| | - Alexey V Yaremenko
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece.
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, 117997, Russia.
| | - Nadezhda A Pechnikova
- Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
- Saint Petersburg Pasteur Institute, Saint Petersburg, 197101, Russia
| | - Artem S Minin
- M.N. Mikheev Institute of Metal Physics of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, 620108, Russia
| | - Ilya V Zubarev
- Institute of Future Biophysics, Dolgoprudny, 141701, Russia.
| |
Collapse
|
9
|
Sharafutdinov I, Harrer A, Müsken M, Rottner K, Sticht H, Täger C, Naumann M, Tegtmeyer N, Backert S. Cortactin-dependent control of Par1b-regulated epithelial cell polarity in Helicobacter infection. CELL INSIGHT 2024; 3:100161. [PMID: 38646547 PMCID: PMC11033139 DOI: 10.1016/j.cellin.2024.100161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 03/01/2024] [Accepted: 03/02/2024] [Indexed: 04/23/2024]
Abstract
Cell polarity is crucial for gastric mucosal barrier integrity and mainly regulated by polarity-regulating kinase partitioning-defective 1b (Par1b). During infection, the carcinogen Helicobacter pylori hijacks Par1b via the bacterial oncoprotein CagA leading to loss of cell polarity, but the precise molecular mechanism is not fully clear. Here we discovered a novel function of the actin-binding protein cortactin in regulating Par1b, which forms a complex with cortactin and the tight junction protein zona occludens-1 (ZO-1). We found that serine phosphorylation at S405/418 and the SH3 domain of cortactin are important for its interaction with both Par1b and ZO-1. Cortactin knockout cells displayed disturbed Par1b cellular localization and exhibited morphological abnormalities that largely compromised transepithelial electrical resistance, epithelial cell polarity, and apical microvilli. H. pylori infection promoted cortactin/Par1b/ZO-1 abnormal interactions in the tight junctions in a CagA-dependent manner. Infection of human gastric organoid-derived mucosoids supported these observations. We therefore hypothesize that CagA disrupts gastric epithelial cell polarity by hijacking cortactin, and thus Par1b and ZO-1, suggesting a new signaling pathway for the development of gastric cancer by Helicobacter.
Collapse
Affiliation(s)
- Irshad Sharafutdinov
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058, Erlangen, Germany
| | - Aileen Harrer
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058, Erlangen, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, D-38124, Braunschweig, Germany
| | - Klemens Rottner
- Department of Cell Biology, Helmholtz Centre for Infection Research, D-38124, Braunschweig, Germany
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, D-38106, Braunschweig, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054, Erlangen, Germany
| | - Christian Täger
- Otto von Guericke University, Institute of Experimental Internal Medicine, Medical Faculty, D-39120, Magdeburg, Germany
| | - Michael Naumann
- Otto von Guericke University, Institute of Experimental Internal Medicine, Medical Faculty, D-39120, Magdeburg, Germany
| | - Nicole Tegtmeyer
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058, Erlangen, Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91058, Erlangen, Germany
| |
Collapse
|
10
|
Vicente JJ, Khan K, Tillinghast G, McFaline-Figueroa JL, Sancak Y, Stella N. The microtubule targeting agent ST-401 triggers cell death in interphase and prevents the formation of polyploid giant cancer cells. J Transl Med 2024; 22:441. [PMID: 38730481 PMCID: PMC11084142 DOI: 10.1186/s12967-024-05234-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
Microtubule targeting agents (MTAs) are commonly prescribed to treat cancers and predominantly kill cancer cells in mitosis. Significantly, some MTA-treated cancer cells escape death in mitosis, exit mitosis and become malignant polyploid giant cancer cells (PGCC). Considering the low number of cancer cells undergoing mitosis in tumor tissues, killing them in interphase may represent a favored antitumor approach. We discovered that ST-401, a mild inhibitor of microtubule (MT) assembly, preferentially kills cancer cells in interphase as opposed to mitosis, a cell death mechanism that avoids the development of PGCC. Single cell RNA sequencing identified mRNA transcripts regulated by ST-401, including mRNAs involved in ribosome and mitochondrial functions. Accordingly, ST-401 induces a transient integrated stress response, reduces energy metabolism, and promotes mitochondria fission. This cell response may underly death in interphase and avoid the development of PGCC. Considering that ST-401 is a brain-penetrant MTA, we validated these results in glioblastoma cell lines and found that ST-401 also reduces energy metabolism and promotes mitochondria fission in GBM sensitive lines. Thus, brain-penetrant mild inhibitors of MT assembly, such as ST-401, that induce death in interphase through a previously unanticipated antitumor mechanism represent a potentially transformative new class of therapeutics for the treatment of GBM.
Collapse
Affiliation(s)
- Juan Jesus Vicente
- Department of Physiology and Biophysics, University of Washington, Health Sciences Building G424, 1705 NE Pacific Str., Seattle, WA, 98195-7280, USA.
| | - Kainat Khan
- Department of Pharmacology, University of Washington, Health Sciences Center F404A, 1959 NE Pacific Str., Seattle, WA, 98195-7280, USA
| | - Grant Tillinghast
- Department of Biomedical Engineering, Columbia University, New York, NY, 10025, USA
| | | | - Yasemin Sancak
- Department of Pharmacology, University of Washington, Health Sciences Center F404A, 1959 NE Pacific Str., Seattle, WA, 98195-7280, USA
| | - Nephi Stella
- Department of Pharmacology, University of Washington, Health Sciences Center F404A, 1959 NE Pacific Str., Seattle, WA, 98195-7280, USA.
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
11
|
Limonta G, Panti C, Fossi MC, Nardi F, Baini M. Exposure to virgin and marine incubated microparticles of biodegradable and conventional polymers modulates the hepatopancreas transcriptome of Mytilus galloprovincialis. JOURNAL OF HAZARDOUS MATERIALS 2024; 468:133819. [PMID: 38402680 DOI: 10.1016/j.jhazmat.2024.133819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/02/2024] [Accepted: 02/15/2024] [Indexed: 02/27/2024]
Abstract
Biodegradable polymers have been proposed as an alternative to conventional plastics to mitigate the impact of marine litter, but the research investigating their toxicity is still in its infancy. This study evaluates the potential ecotoxicological effects of both virgin and marine-incubated microparticles (MPs), at environmentally relevant concentration (0.1 mg/l), made of different biodegradable polymers (Polycaprolactone, Mater-Bi, cellulose) and conventional polymers (Polyethylene) on Mytilus galloprovincialis by using transcriptomics. This approach is increasingly being used to assess the effects of pollutants on organisms, obtaining data on numerous biological pathways simultaneously. Whole hepatopancreas de novo transcriptome sequencing was performed, individuating 972 genes differentially expressed across experimental groups compared to the control. Through the comparative transcriptomic profiling emerges that the preponderant effect is attributable to the marine incubation of MPs, especially for incubated polycaprolactone (731 DEGs). Mater-Bi and cellulose alter the smallest number of genes and biological processes in the mussel hepatopancreas. All microparticles, regardless of their polymeric composition, dysregulated innate immunity, and fatty acid metabolism biological processes. These findings highlight the necessity of considering the interactions of MPs with the environmental factors in the marine ecosystem when performing ecotoxicological evaluations. The results obtained contribute to fill current knowledge gaps regarding the potential environmental impacts of biodegradable polymers.
Collapse
Affiliation(s)
- Giacomo Limonta
- Department of Physical, Earth and Environmental Sciences, University of Siena, Via P.A. Mattioli, 4, Siena, Italy; National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Cristina Panti
- Department of Physical, Earth and Environmental Sciences, University of Siena, Via P.A. Mattioli, 4, Siena, Italy; National Biodiversity Future Center (NBFC), Palermo, Italy.
| | - Maria Cristina Fossi
- Department of Physical, Earth and Environmental Sciences, University of Siena, Via P.A. Mattioli, 4, Siena, Italy; National Biodiversity Future Center (NBFC), Palermo, Italy
| | - Francesco Nardi
- National Biodiversity Future Center (NBFC), Palermo, Italy; Department of Life Sciences, University of Siena, Via A. Moro, 2, Siena, Italy
| | - Matteo Baini
- Department of Physical, Earth and Environmental Sciences, University of Siena, Via P.A. Mattioli, 4, Siena, Italy; National Biodiversity Future Center (NBFC), Palermo, Italy
| |
Collapse
|
12
|
Maffeis V, Heuberger L, Nikoletić A, Schoenenberger C, Palivan CG. Synthetic Cells Revisited: Artificial Cells Construction Using Polymeric Building Blocks. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305837. [PMID: 37984885 PMCID: PMC10885666 DOI: 10.1002/advs.202305837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/06/2023] [Indexed: 11/22/2023]
Abstract
The exponential growth of research on artificial cells and organelles underscores their potential as tools to advance the understanding of fundamental biological processes. The bottom-up construction from a variety of building blocks at the micro- and nanoscale, in combination with biomolecules is key to developing artificial cells. In this review, artificial cells are focused upon based on compartments where polymers are the main constituent of the assembly. Polymers are of particular interest due to their incredible chemical variety and the advantage of tuning the properties and functionality of their assemblies. First, the architectures of micro- and nanoscale polymer assemblies are introduced and then their usage as building blocks is elaborated upon. Different membrane-bound and membrane-less compartments and supramolecular structures and how they combine into advanced synthetic cells are presented. Then, the functional aspects are explored, addressing how artificial organelles in giant compartments mimic cellular processes. Finally, how artificial cells communicate with their surrounding and each other such as to adapt to an ever-changing environment and achieve collective behavior as a steppingstone toward artificial tissues, is taken a look at. Engineering artificial cells with highly controllable and programmable features open new avenues for the development of sophisticated multifunctional systems.
Collapse
Affiliation(s)
- Viviana Maffeis
- Department of ChemistryUniversity of BaselMattenstrasse 22BaselCH‐4002Switzerland
- NCCR‐Molecular Systems EngineeringBPR 1095, Mattenstrasse 24aBaselCH‐4058Switzerland
| | - Lukas Heuberger
- Department of ChemistryUniversity of BaselMattenstrasse 22BaselCH‐4002Switzerland
| | - Anamarija Nikoletić
- Department of ChemistryUniversity of BaselMattenstrasse 22BaselCH‐4002Switzerland
- Swiss Nanoscience InstituteUniversity of BaselKlingelbergstrasse 82BaselCH‐4056Switzerland
| | | | - Cornelia G. Palivan
- Department of ChemistryUniversity of BaselMattenstrasse 22BaselCH‐4002Switzerland
- NCCR‐Molecular Systems EngineeringBPR 1095, Mattenstrasse 24aBaselCH‐4058Switzerland
- Swiss Nanoscience InstituteUniversity of BaselKlingelbergstrasse 82BaselCH‐4056Switzerland
| |
Collapse
|
13
|
YunhongYang, Mao T, Ding Y, Ge L, Feng L, Cai M, Han C, Yang J. Variations in life history parameters, population dynamics, and transcriptome regulation of Brachionus plicatilis exposed to triclosan. MARINE POLLUTION BULLETIN 2024; 199:115918. [PMID: 38134871 DOI: 10.1016/j.marpolbul.2023.115918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/24/2023]
Abstract
Triclosan (TCS) poses an ecological health risk due to its lipophilic nature, long half-life, and bioconcentration. To evaluate the toxicity of TCS on aquatic organisms, the life history parameters, population dynamics, and transcriptome regulation of Brachionus plicatilis exposed to TCS were investigated. In this study, the fecundity of rotifers was promoted by 25 μg/L of TCS and inhibited by higher concentrations (100 μg/L, 200 μg/L). The reproductive period of rotifers was shortened by 46.24 % but the post-reproductive period was prolonged by 176.47 % in 200 μg/L TCS. Both population growth and life table parameters indicated that a high concentration of TCS (200 μg/L) had negative impacts on population growth. Transcriptomic analysis showed that the effects of TCS on the life history parameters and population dynamics of rotifers were determined by regulating the expression of functional genes in cilium organization and cilium assembly and involved in pathways of focal adhesion.
Collapse
Affiliation(s)
- YunhongYang
- Jiangsu Province Key Laboratory for Fisheries Live Food, School of Marine Science and Engineering, Nanjing Normal University, No. 2 Xuelin Rd, Nanjing 210023, People's Republic of China
| | - Tianyue Mao
- Jiangsu Province Key Laboratory for Fisheries Live Food, School of Marine Science and Engineering, Nanjing Normal University, No. 2 Xuelin Rd, Nanjing 210023, People's Republic of China
| | - Yifan Ding
- Jiangsu Province Key Laboratory for Fisheries Live Food, School of Marine Science and Engineering, Nanjing Normal University, No. 2 Xuelin Rd, Nanjing 210023, People's Republic of China
| | - Lingling Ge
- Jiangsu Province Key Laboratory for Fisheries Live Food, School of Marine Science and Engineering, Nanjing Normal University, No. 2 Xuelin Rd, Nanjing 210023, People's Republic of China
| | - Lei Feng
- Jiangsu Province Key Laboratory for Fisheries Live Food, School of Marine Science and Engineering, Nanjing Normal University, No. 2 Xuelin Rd, Nanjing 210023, People's Republic of China
| | - Meng Cai
- Jiangsu Province Key Laboratory for Fisheries Live Food, School of Marine Science and Engineering, Nanjing Normal University, No. 2 Xuelin Rd, Nanjing 210023, People's Republic of China
| | - Cui Han
- Jiangsu Province Key Laboratory for Fisheries Live Food, School of Marine Science and Engineering, Nanjing Normal University, No. 2 Xuelin Rd, Nanjing 210023, People's Republic of China
| | - Jiaxin Yang
- Jiangsu Province Key Laboratory for Fisheries Live Food, School of Marine Science and Engineering, Nanjing Normal University, No. 2 Xuelin Rd, Nanjing 210023, People's Republic of China.
| |
Collapse
|
14
|
Wang AC, Qi XM, Li QF, Feng YJ, Zhang YL, Wei HZ, Li JS, Qiao YB, Li QS. Methionine redox regulation of actin-interacting proteins primarily governs antioxidative signaling and response to the salvianolic acid B treatment in EA.hy926 cells. Toxicol Appl Pharmacol 2024; 483:116835. [PMID: 38272317 DOI: 10.1016/j.taap.2024.116835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
Actin-interacting proteins are important molecules for filament assembly and cytoskeletal signaling within vascular endothelium. Disruption in their interactions causes endothelial pathogenesis through redox imbalance. Actin filament redox regulation remains largely unexplored, in the context of pharmacological treatment. This work focused on the peptidyl methionine (M) redox regulation of actin-interacting proteins, aiming at elucidating its role on governing antioxidative signaling and response. Endothelial EA.hy926 cells were subjected to treatment with salvianolic acid B (Sal B) and tert-butyl-hydroperoxide (tBHP) stimulation. Mass spectrometry was employed to characterize redox status of proteins, including actin, myosin-9, kelch-like erythroid-derived cap-n-collar homology-associated protein 1 (Keap1), plastin-3, prelamin-A/C and vimentin. The protein redox landscape revealed distinct stoichiometric ratios or reaction site transitions mediated by M sulfoxide reductase and reactive oxygen species. In comparison with effects of tBHP stimulation, Sal B treatment prevented oxidation at actin M325, myosin-9 M1489/1565, Keap1 M120, plastin-3 M592, prelamin-A/C M187/371/540 and vimentin M344. For Keap1, reaction site was transitioned within its scaffolding region to the actin ring. These protein M oxidation regulations contributed to the Sal B cytoprotective effects on actin filament. Additionally, regarding the Keap1 homo-dimerization region, Sal B preventive roles against M120 oxidation acted as a primary signal driver to activate nuclear factor erythroid 2-related factor 2 (Nrf2). Transcriptional splicing of non-POU domain-containing octamer-binding protein was validated during the Sal B-mediated overexpression of NAD(P)H dehydrogenase [quinone] 1. This molecular redox regulation of actin-interacting proteins provided valuable insights into the phenolic structures of Sal B analogs, showing potential antioxidative effects on vascular endothelium.
Collapse
Affiliation(s)
- Ai-Cheng Wang
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Xiao-Ming Qi
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China.
| | - Qing-Fang Li
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Yi-Jia Feng
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Yuan-Lin Zhang
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China.
| | - Hui-Zhi Wei
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China; School of Public Health Science & Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, China.
| | - Jin-Shan Li
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China
| | - Yuan-Biao Qiao
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China.
| | - Qing-Shan Li
- Shanxi Key Laboratory of Innovative Drug for the Treatment of Serious Diseases Basing on the Chronic Inflammation, College of Traditional Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Taiyuan, Shanxi 030619, China; School of Public Health Science & Pharmaceutical Science, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
15
|
Gonzalez-Nolde S, Schweiger CJ, Davis EE, Manzoni TJ, Hussein SM, Schmidt TA, Cone SG, Jay GD, Parreno J. The Actin Cytoskeleton as a Regulator of Proteoglycan 4. Cartilage 2024:19476035231223455. [PMID: 38183234 PMCID: PMC11569590 DOI: 10.1177/19476035231223455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024] Open
Abstract
OBJECTIVE The superficial zone (SZ) of articular cartilage is responsible for distributing shear forces for optimal cartilage loading and contributes to joint lubrication through the production of proteoglycan 4 (PRG4). PRG4 plays a critical role in joint homeostasis and is chondroprotective. Normal PRG4 production is affected by inflammation and irregular mechanical loading in post-traumatic osteoarthritis (PTOA). THe SZ chondrocyte (SZC) phenotype, including PRG4 expression, is regulated by the actin cytoskeleton in vitro. There remains a limited understanding of the regulation of PRG4 by the actin cytoskeleton in native articular chondrocytes. The filamentous (F)-actin cytoskeleton is a potential node in crosstalk between mechanical stimulation and cytokine activation and the regulation of PRG4 in SZCs, therefore developing insights in the regulation of PRG4 by actin may identify molecular targets for novel PTOA therapies. MATERIALS AND METHODS A comprehensive literature search on PRG4 and the regulation of the SZC phenotype by actin organization was performed. RESULTS PRG4 is strongly regulated by the actin cytoskeleton in isolated SZCs in vitro. Biochemical and mechanical stimuli have been characterized to regulate PRG4 and may converge upon actin cytoskeleton signaling. CONCLUSION Actin-based regulation of PRG4 in native SZCs is not fully understood and requires further elucidation. Understanding the regulation of PRG4 by actin in SZCs requires an in vivo context to further potential of leveraging actin arrangement to arthritic therapeutics.
Collapse
|
16
|
Lee W. The Cytoskeleton and Its Binding Proteins as Mechanosensors, Transducers, and Functional Regulators of Cells. Int J Mol Sci 2023; 25:172. [PMID: 38203343 PMCID: PMC10779244 DOI: 10.3390/ijms25010172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 12/14/2023] [Indexed: 01/12/2024] Open
Abstract
Due to its complement of diverse proteins, such as actin filaments, intermediate filaments, and microtubules, the cytoskeleton is essential not only for structural stability but also for regulating cellular signaling, intracellular transportation, and cell division [...].
Collapse
Affiliation(s)
- Wan Lee
- Department of Biochemistry, Dongguk University College of Medicine, 123 Dongdae-ro, Gyeongju 38066, Republic of Korea; ; Tel.: +82-54-770-2409; Fax: +82-54-770-2447
- Channelopathy Research Center (CRC), Dongguk University College of Medicine, 32 Dongguk-ro, Ilsan Dong-gu, Goyang 10326, Republic of Korea
| |
Collapse
|
17
|
Zhang R, Chen S, Yang Z, Zhang N, Guo K, Lv K, Zhou Z, Gao M, Hu X, Su Y, He J, Wang F. Actin polymerization inhibition by targeting ARPC2 affects intestinal stem cell homeostasis. BURNS & TRAUMA 2023; 11:tkad038. [PMID: 37849945 PMCID: PMC10578047 DOI: 10.1093/burnst/tkad038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/28/2023] [Accepted: 05/25/2023] [Indexed: 10/19/2023]
Abstract
Background The rapid turnover of the intestinal epithelium is driven by the proliferation and differentiation of intestinal stem cells (ISCs). The dynamics of the F-actin cytoskeleton are critical for maintaining intercellular force and the signal transduction network. However, it remains unclear how direct interference with actin polymerization impacts ISC homeostasis. This study aims to reveal the regulatory effects of the F-actin cytoskeleton on the homeostasis of intestinal epithelium, as well as the potential risks of benproperine (BPP) as an anti-tumor drug. Methods Phalloidin fluorescence staining was utilized to test F-actin polymerization. Flow cytometry and IHC staining were employed to discriminate different types of intestinal epithelial cells. Cell proliferation was assessed through bromo-deoxyuridine (BrdU) and 5-ethynyl-2'-deoxyuridine (EdU) incorporation assays. The proliferation and differentiation of intestinal stem cells were replicated in vitro through organoid culture. Epithelial migration was evaluated through BrdU pulse labeling and chasing in mice. Results The F-actin content was observed to significantly increase as crypt cells migrated into the villus region. Additionally, actin polymerization in secretory cells, especially in Paneth cells (PCs), was much higher than that in neighboring ISCs. Treatment with the newly identified actin-related protein 2/3 complex subunit 2 (ARPC2) inhibitor BPP led to a dose-dependent increase or inhibition of intestinal organoid growth in vitro and crypt cell proliferation in vivo. Compared with the vehicle group, BPP treatment decreased the expression of Lgr5 ISC feature genes in vivo and in organoid culture. Meanwhile, PC differentiation derived from ISCs and progenitors was decreased by inhibition of F-actin polymerization. Mechanistically, BPP-induced actin polymerization inhibition may activate the Yes1-associated transcriptional regulator pathway, which affects ISC proliferation and differentiation. Accordingly, BPP treatment affected intestinal epithelial cell migration in a dose-dependent manner. Conclusion Our findings indicate that the regulation of cytoskeleton reorganization can affect ISC homeostasis. In addition, inhibiting ARPC2 with the Food and Drug Administration-approved drug BPP represents a novel approach to influencing the turnover of intestinal epithelial cells.
Collapse
Affiliation(s)
- Ruzhen Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
- College of Life Sciences, Chongqing Normal University,Chongqing, 401331China
| | - Sheng Chen
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Zhifan Yang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Ning Zhang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Kenan Guo
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
- Department of Laboratory Animal Science, College of Basic Medical Sciences, Army Medical University, Third Military Medical University, Chongqing 400038, China
| | - Keyi Lv
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University, Chongqing, 400038, China
| | - Zimo Zhou
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Meijiao Gao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Xiancheng Hu
- College of Life Sciences, Chongqing Normal University,Chongqing, 401331China
| | - Yongping Su
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| | - Jianming He
- Department of Radiotherapy, Hebei Province Hospital of Chinese Medicine, Hebei University of Chinese Medicine, Key Laboratory of Integrated Chinese and Western Medicine for Gastroenterology Research (Hebei), Shijiazhuang, 050011
| | - Fengchao Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
18
|
Huang D, Chen S, Xiong D, Wang H, Zhu L, Wei Y, Li Y, Zou S. Mitochondrial Dynamics: Working with the Cytoskeleton and Intracellular Organelles to Mediate Mechanotransduction. Aging Dis 2023; 14:1511-1532. [PMID: 37196113 PMCID: PMC10529762 DOI: 10.14336/ad.2023.0201] [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: 10/21/2022] [Accepted: 02/01/2023] [Indexed: 05/19/2023] Open
Abstract
Cells are constantly exposed to various mechanical environments; therefore, it is important that they are able to sense and adapt to changes. It is known that the cytoskeleton plays a critical role in mediating and generating extra- and intracellular forces and that mitochondrial dynamics are crucial for maintaining energy homeostasis. Nevertheless, the mechanisms by which cells integrate mechanosensing, mechanotransduction, and metabolic reprogramming remain poorly understood. In this review, we first discuss the interaction between mitochondrial dynamics and cytoskeletal components, followed by the annotation of membranous organelles intimately related to mitochondrial dynamic events. Finally, we discuss the evidence supporting the participation of mitochondria in mechanotransduction and corresponding alterations in cellular energy conditions. Notable advances in bioenergetics and biomechanics suggest that the mechanotransduction system composed of mitochondria, the cytoskeletal system, and membranous organelles is regulated through mitochondrial dynamics, which may be a promising target for further investigation and precision therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuyu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Chu Z, Zhu M, Luo Y, Hu Y, Feng X, Wang H, Sunagawa M, Liu Y. PTBP1 plays an important role in the development of gastric cancer. Cancer Cell Int 2023; 23:195. [PMID: 37670313 PMCID: PMC10478210 DOI: 10.1186/s12935-023-03043-0] [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: 03/23/2023] [Accepted: 08/25/2023] [Indexed: 09/07/2023] Open
Abstract
BACKGROUND Polypyrimidine tract binding protein 1 (PTBP1) has been found to play an important role in the occurrence and development of various tumors. At present, the role of PTBP1 in gastric cancer (GC) is still unknown and worthy of further investigation. METHODS We used bioinformatics to analyze the expression of PTBP1 in patients with GC. Cell proliferation related experiments were used to detect cell proliferation after PTBP1 knockdown. Skeleton staining, scanning electron microscopy and transmission electron microscopy were used to observe the changes of actin skeleton. Proliferation and actin skeleton remodeling signaling pathways were detected by Western Blots. The relationship between PTBP1 and proliferation of gastric cancer cells was further detected by subcutaneous tumor transplantation. Finally, tissue microarray data from clinical samples were used to further explore the expression of PTBP1 in patients with gastric cancer and its correlation with prognosis. RESULTS Through bioinformatics studies, we found that PTBP1 was highly expressed in GC patients and correlated with poor prognosis. Cell proliferation and cycle analysis showed that PTBP1 down-regulation could significantly inhibit cell proliferation. The results of cell proliferation detection related experiments showed that PTBP1 down-regulation could inhibit the division and proliferation of GC cells. Furthermore, changes in the morphology of the actin skeleton of cells showed that PTBP1 down-regulation inhibited actin skeletal remodeling in GC cells. Western Blots showed that PTBP1 could regulate proliferation and actin skeleton remodeling signaling pathways. In addition, we constructed PTBP1 Cas9-KO mouse model and performed xenograft assays to further confirm that down-regulation of PTBP1 could inhibit the proliferation of GC cells. Finally, tissue microarray was used to further verify the close correlation between PTBP1 and poor prognosis in patients with GC. CONCLUSIONS Our study demonstrates for the first time that PTBP1 may affect the proliferation of GC cells by regulating actin skeleton remodeling. In addition, PTBP1 is closely related to actin skeleton remodeling and proliferation signaling pathways. We suppose that PTBP1 might be a potential target for the treatment of GC.
Collapse
Affiliation(s)
- Zewen Chu
- The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Miao Zhu
- The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Yuanyuan Luo
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Yaqi Hu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Xinyi Feng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China
| | - Haibo Wang
- The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China.
| | - Masataka Sunagawa
- Department of physiology, School of Medicine, Showa University, Tokyo, Japan.
| | - Yanqing Liu
- The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China.
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.
- The Key Laboratory of Syndrome Differentiation and Treatment of Gastric Cancer of the State Administration of Traditional Chinese Medicine, Yangzhou, China.
| |
Collapse
|
20
|
He S, Liu R, Luo Q, Song G. Tensile Overload Injures Human Alveolar Epithelial Cells through YAP/F-Actin/MAPK Signaling. Biomedicines 2023; 11:1833. [PMID: 37509472 PMCID: PMC10376431 DOI: 10.3390/biomedicines11071833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/18/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Explosion shockwaves can generate overloaded mechanical forces and induce lung injuries. However, the mechanism of lung injuries caused by tensile overload is still unclear. METHODS Flow cytometry was used to detect the apoptosis of human alveolar epithelial cells (BEAS-2B) induced by tensile overload, and cell proliferation was detected using 5-ethynyl-2'-deoxyuridine (EdU). Immunofluorescence and Western blot analysis were used to identify the tensile overload on the actin cytoskeleton, proteins related to the mitogen-activated protein kinase (MAPK) signal pathway, and the Yes-associated protein (YAP). RESULTS Tensile overload reduced BEAS-2B cell proliferation and increased apoptosis. In terms of the mechanism, we found that tensile overload led to the depolymerization of the actin cytoskeleton, the activation of c-Jun N-terminal kinase (JNK) and extracellular-signal-regulated kinase 1/2 (ERK1/2), and the upregulation of YAP expression. Jasplakinolide (Jasp) treatment promoted the polymerization of the actin cytoskeleton and reduced the phosphorylation of tension-overload-activated JNK and ERK1/2 and the apoptosis of BEAS-2B cells. Moreover, the inhibition of the JNK and ERK1/2 signaling pathways, as well as the expression of YAP, also reduced apoptosis caused by tensile overload. CONCLUSION Our study establishes the role of the YAP/F-actin/MAPK axis in tensile-induced BEAS-2B cell injury and proposes new strategies for the treatment and repair of future lung injuries.
Collapse
Affiliation(s)
- Shan He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Ruihan Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| |
Collapse
|
21
|
Paukner D, Eichinger JF, Cyron CJ. What are the key mechanical mechanisms governing integrin-mediated cell migration in three-dimensional fiber networks? Biomech Model Mechanobiol 2023:10.1007/s10237-023-01709-2. [PMID: 37318643 PMCID: PMC10366304 DOI: 10.1007/s10237-023-01709-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 03/01/2023] [Indexed: 06/16/2023]
Abstract
Cell migration plays a vital role in numerous processes such as development, wound healing, or cancer. It is well known that numerous complex mechanisms are involved in cell migration. However, so far it remains poorly understood what are the key mechanisms required to produce the main characteristics of this behavior. The reason is a methodological one. In experimental studies, specific factors and mechanisms can be promoted or inhibited. However, while doing so, there can always be others in the background which play key roles but which have simply remained unattended so far. This makes it very difficult to validate any hypothesis about a minimal set of factors and mechanisms required to produce cell migration. To overcome this natural limitation of experimental studies, we developed a computational model where cells and extracellular matrix fibers are represented by discrete mechanical objects on the micrometer scale. In this model, we had exact control of the mechanisms by which cells and matrix fibers interacted with each other. This enabled us to identify the key mechanisms required to produce physiologically realistic cell migration (including advanced phenomena such as durotaxis and a biphasic relation between migration efficiency and matrix stiffness). We found that two main mechanisms are required to this end: a catch-slip bond of individual integrins and cytoskeletal actin-myosin contraction. Notably, more advanced phenomena such as cell polarization or details of mechanosensing were not necessary to qualitatively reproduce the main characteristics of cell migration observed in experiments.
Collapse
Affiliation(s)
- Daniel Paukner
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Eißendorfer Straße 42 (M), 21073, Hamburg, Hamburg, Germany
- Institute of Material Systems Modeling, Helmholtz-Zentrum Hereon, Max-Planck Straße 1, 21502, Geesthacht, Schleswig-Holstein, Germany
| | - Jonas F Eichinger
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Eißendorfer Straße 42 (M), 21073, Hamburg, Hamburg, Germany
- Institute for Computational Mechanics, Technical University of Munich, Boltzmannstraße 15, Garching b., 85748, München, Bavaria, Germany
| | - Christian J Cyron
- Institute for Continuum and Material Mechanics, Hamburg University of Technology, Eißendorfer Straße 42 (M), 21073, Hamburg, Hamburg, Germany.
- Institute of Material Systems Modeling, Helmholtz-Zentrum Hereon, Max-Planck Straße 1, 21502, Geesthacht, Schleswig-Holstein, Germany.
| |
Collapse
|
22
|
Lu X, Yan G, Fu L, Cui B, Wang J, Zhou D. A review of filamentous sludge bulking controls from conventional methods to emerging quorum quenching strategies. WATER RESEARCH 2023; 236:119922. [PMID: 37098319 DOI: 10.1016/j.watres.2023.119922] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/16/2023] [Accepted: 03/26/2023] [Indexed: 06/19/2023]
Abstract
Filamentous bulking, which results from the overgrowth of filamentous microorganisms, is a common issue that frequently disrupts the stable operation of activated sludge processes. Recent literature has paid attention to the relationship between quorum sensing (QS) and filamentous bulking highlighting that the morphological transformations of filamentous microbes are regulated by functional signal molecules in the bulking sludge system. In response to this, a novel quorum quenching (QQ) technology has been developed to control sludge bulking effectively and precisely by disturbing QS-mediated filamentation behaviors. This paper presents a critical review on the limitations of classical bulking hypotheses and traditional control methods, and provides an overview of recent QS/QQ studies that aim to elucidate and control filamentous bulking, including the characterization of molecule structures, the elaboration of QS pathways, and the precise design of QQ molecules to mitigate filamentous bulking. Finally, suggestions for further research and development of QQ strategies for precise bulking control are put forward.
Collapse
Affiliation(s)
- Xin Lu
- Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun 130117, PR China
| | - Ge Yan
- Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun 130117, PR China
| | - Liang Fu
- Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun 130117, PR China
| | - Bin Cui
- Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun 130117, PR China
| | - Jinfeng Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, Jiangsu, PR China
| | - Dandan Zhou
- Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun 130117, PR China.
| |
Collapse
|
23
|
Villablanca C, Vidal R, Gonzalez-Billault C. Are cytoskeleton changes observed in astrocytes functionally linked to aging? Brain Res Bull 2023; 196:59-67. [PMID: 36935053 DOI: 10.1016/j.brainresbull.2023.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/22/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
Astrocytes are active participants in the performance of the Central Nervous System (CNS) in both health and disease. During aging, astrocytes are susceptible to reactive astrogliosis, a molecular state characterized by functional changes in response to pathological situations, and cellular senescence, characterized by loss of cell division, apoptosis resistance, and gain of proinflammatory functions. This results in two different states of astrocytes, which can produce proinflammatory phenotypes with harmful consequences in chronic conditions. Reactive astrocytes and senescent astrocytes share morpho-functional features that are dependent on the organization of the cytoskeleton. However, such changes in the cytoskeleton have yet to receive the necessary attention to explain their role in the alterations of astrocytes that are associated with aging and pathologies. In this review, we summarize all the available findings that connect changes in the cytoskeleton of the astrocytes with aging. In addition, we discuss future avenues that we believe will guide such a novel topic.
Collapse
Affiliation(s)
- Cristopher Villablanca
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - René Vidal
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile
| | - Christian Gonzalez-Billault
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile, Santiago, Chile; Geroscience Center for Brain Health and Metabolism (GERO), Santiago, Chile; Department of Neurosciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile; Institute for Nutrition and Food Technologies, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
24
|
Khan I, Siraj M. An updated review on cell signaling pathways regulated by candidate miRNAs in coronary artery disease. Noncoding RNA Res 2023; 8:326-334. [PMID: 37077752 PMCID: PMC10106733 DOI: 10.1016/j.ncrna.2023.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 03/31/2023] Open
Abstract
MicroRNAs (miRNAs) are small endogenous non-coding RNA, size range from 17 to 25 nucleotides that regulate gene expression at the post-transcriptional level. More than 2000 different types of miRNAs have been identified in humans which regulate about 60% of gene expression, since the discovery of the first miRNA in 1993. MicroRNA performs many functions such as being involved in the regulation of various biological pathways for example cell migration, cell proliferation, cell differentiation, disease progression, and initiation. miRNAs also play an important role in the development of atherosclerosis lesions, cardiac fibroblast, cardiac hypertrophy, cancer, and neurological disorders. Abnormal activation of many cell signaling pathways has been observed in the development of coronary artery disease. Abnormal expression of these candidate miRNA genes leads to up or downregulation of specific genes, these specific genes play an important role in the regulation of cell signaling pathways involved in coronary artery disease. Many studies have found that miRNAs play a key role in the regulation of crucial signaling pathways that are involved in the pathophysiology of coronary artery disease. This review is designed to investigate the role of cell signaling pathways regulated by candidate miRNAs in Coronary artery disease.
Collapse
|
25
|
Li M, Peng L, Wang Z, Liu L, Cao M, Cui J, Wu F, Yang J. Roles of the cytoskeleton in human diseases. Mol Biol Rep 2023; 50:2847-2856. [PMID: 36609753 DOI: 10.1007/s11033-022-08025-5] [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: 02/07/2022] [Accepted: 10/12/2022] [Indexed: 01/08/2023]
Abstract
Recently, researches have revealed the key roles of the cytoskeleton in the occurrence and development of multiple diseases, suggesting that targeting the cytoskeleton is a viable approach for treating numerous refractory diseases. The cytoskeleton is a highly structured and complex network composed of actin filaments, microtubules, and intermediate filaments. In normal cells, these three cytoskeleton components are highly integrated and coordinated. However, the cytoskeleton undergoes drastic remodeling in cytoskeleton-related diseases, causing changes in cell polarity, affecting the cell cycle, leading to senescent diseases, and influencing cell migration to accelerate cancer metastasis. Additionally, mutations or abnormalities in cytoskeletal proteins and their related proteins are closely associated with several congenital diseases. Therefore, this review summarizes the roles of the cytoskeleton in cytoskeleton-related diseases as well as its potential roles in disease treatment to provide insights regarding the physiological functions and pathological roles of the cytoskeleton.
Collapse
Affiliation(s)
- Mengxin Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Li Peng
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, 610065, Chengdu, China
| | - Zhenming Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Lijia Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Mengjiao Cao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Jingyao Cui
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Fanzi Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China
| | - Jing Yang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cardiology and Endodontics, West China Hospital of Stomatology, Sichuan University, 610021, Chengdu, China.
| |
Collapse
|
26
|
Faria L, Canato S, Jesus TT, Gonçalves M, Guerreiro PS, Lopes CS, Meireles I, Morais-de-Sá E, Paredes J, Janody F. Activation of an actin signaling pathway in pre-malignant mammary epithelial cells by P-cadherin is essential for transformation. Dis Model Mech 2023; 16:dmm049652. [PMID: 36808468 PMCID: PMC9983776 DOI: 10.1242/dmm.049652] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 01/19/2023] [Indexed: 02/23/2023] Open
Abstract
Alterations in the expression or function of cell adhesion molecules have been implicated in all steps of tumor progression. Among those, P-cadherin is highly enriched in basal-like breast carcinomas, playing a central role in cancer cell self-renewal, collective cell migration and invasion. To establish a clinically relevant platform for functional exploration of P-cadherin effectors in vivo, we generated a humanized P-cadherin Drosophila model. We report that actin nucleators, Mrtf and Srf, are main P-cadherin effectors in fly. We validated these findings in a human mammary epithelial cell line with conditional activation of the SRC oncogene. We show that, prior to promoting malignant phenotypes, SRC induces a transient increase in P-cadherin expression, which correlates with MRTF-A accumulation, its nuclear translocation and the upregulation of SRF target genes. Moreover, knocking down P-cadherin, or preventing F-actin polymerization, impairs SRF transcriptional activity. Furthermore, blocking MRTF-A nuclear translocation hampers proliferation, self-renewal and invasion. Thus, in addition to sustaining malignant phenotypes, P-cadherin can also play a major role in the early stages of breast carcinogenesis by promoting a transient boost of MRTF-A-SRF signaling through actin regulation.
Collapse
Affiliation(s)
- Lídia Faria
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Master Programme in Oncology, School of Medicine and Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Sara Canato
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Physiology and Cancer Program, Champalimaud Foundation, Avenida de Brasília, 1400-038 Lisboa, Portugal
| | - Tito T. Jesus
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
| | - Margarida Gonçalves
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Patrícia S. Guerreiro
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Vector B2B - Drug Developing - Associação Para Investigação em Biotecnologia, Av. Prof. Egas Moniz, Edifício Egas Moniz, 1649-028 Lisboa, Portugal
| | - Carla S. Lopes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Isabel Meireles
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
| | - Eurico Morais-de-Sá
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Joana Paredes
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- FMUP, Medical Faculty of University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Florence Janody
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, 4200-135 Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (Ipatimup), Rua Júlio Amaral de Carvalho, n 45, 4200-135 Porto, Portugal
- Instituto Gulbenkian de Ciência, Rua da Quinta Grande 6, P-2780-156 Oeiras, Portugal
| |
Collapse
|
27
|
Microtubules as a potential platform for energy transfer in biological systems: a target for implementing individualized, dynamic variability patterns to improve organ function. Mol Cell Biochem 2023; 478:375-392. [PMID: 35829870 DOI: 10.1007/s11010-022-04513-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/24/2022] [Indexed: 02/07/2023]
Abstract
Variability characterizes the complexity of biological systems and is essential for their function. Microtubules (MTs) play a role in structural integrity, cell motility, material transport, and force generation during mitosis, and dynamic instability exemplifies the variability in the proper function of MTs. MTs are a platform for energy transfer in cells. The dynamic instability of MTs manifests itself by the coexistence of growth and shortening, or polymerization and depolymerization. It results from a balance between attractive and repulsive forces between tubulin dimers. The paper reviews the current data on MTs and their potential roles as energy-transfer cellular structures and presents how variability can improve the function of biological systems in an individualized manner. The paper presents the option for targeting MTs to trigger dynamic improvement in cell plasticity, regulate energy transfer, and possibly control quantum effects in biological systems. The described system quantifies MT-dependent variability patterns combined with additional personalized signatures to improve organ function in a subject-tailored manner. The platform can regulate the use of MT-targeting drugs to improve the response to chronic therapies. Ongoing trials test the effects of this platform on various disorders.
Collapse
|
28
|
Wang Z, Wu S, Wang G, Yang Z, Zhang Y, Zhu C, Qin X. ARHGAP21 Is Involved in the Carcinogenic Mechanism of Cholangiocarcinoma: A Study Based on Bioinformatic Analyses and Experimental Validation. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:medicina59010139. [PMID: 36676763 PMCID: PMC9867224 DOI: 10.3390/medicina59010139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/12/2023]
Abstract
Background and Objectives: Rho GTPase-activating protein (RhoGAP) is a negative regulatory element of Rho GTPases and participates in tumorigenesis. Rho GTPase-activating protein 21 (ARHGAP21) is one of the RhoGAPs and its role in cholangiocarcinoma (CCA) has never been disclosed in any publications. Materials and Methods: The bioinformatics public datasets were utilized to investigate the expression patterns and mutations of ARHGAP21 as well as its prognostic significance in CCA. The biological functions of ARHGAP21 in CCA cells (RBE and Hccc9810 cell) were evaluated by scratch assay, cell counting kit-8 assay (CCK8) assay, and transwell migration assay. In addition, the underlying mechanism of ARHGAP21 involved in CCA was investigated by the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, and the most significant signaling pathway was identified through gene set enrichment analysis (GSEA) and the Western blot method. The ssGSEA algorithm was further used to explore the immune-related mechanism of ARHGAP21 in CCA. Results: The ARHGAP21 expression in CCA tissue was higher than it was in normal tissue, and missense mutation was the main alteration of ARHGAP21 in CCA. Moreover, the expression of ARHGAP21 had obvious differences in patients with different clinical characteristics and it had great prognostic significance. Based on cell experiments, we further observed that the proliferation ability and migration ability of the ARHGAP21-knockdown group was reduced in CCA cells. Several pathological signaling pathways correlated with proliferation and migration were determined by GO and KEGG analysis. Furthermore, the PI3K/Akt signaling pathway was the most significant one. GSEA analysis further verified that ARHGAP21 was highly enriched in PI3K/Akt signaling pathway, and the results of Western blot suggested that the phosphorylated PI3K and Akt were decreased in the ARHGAP21-knockdown group. The drug susceptibility of the PI3K/Akt signaling pathway targeted drugs were positively correlated with ARHGAP21 expression. Moreover, we also discovered that ARHGAP21 was correlated with neutrophil, pDC, and mast cell infiltration as well as immune-related genes in CCA. Conclusions: ARHGAP21 could promote the proliferation and migration of CCA cells by activating the PI3K/Akt signaling pathway, and ARHGAP21 may participate in the immune modulating function of the tumor microenvironment.
Collapse
Affiliation(s)
- Zhihuai Wang
- Department of General Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Graduate School, Nanjing Medical University, Nanjing 211166, China
| | - Siyuan Wu
- Department of General Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Graduate School, Nanjing Medical University, Nanjing 211166, China
| | - Gaochao Wang
- Graduate School, Nanjing Medical University, Nanjing 211166, China
| | - Zhen Yang
- Graduate School, Nanjing Medical University, Nanjing 211166, China
| | - Yinjie Zhang
- Department of General Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Graduate School, Nanjing Medical University, Nanjing 211166, China
| | - Chunfu Zhu
- Department of General Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Graduate School, Nanjing Medical University, Nanjing 211166, China
- Correspondence: (C.Z.); (X.Q.)
| | - Xihu Qin
- Department of General Surgery, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou 213000, China
- Graduate School, Nanjing Medical University, Nanjing 211166, China
- Correspondence: (C.Z.); (X.Q.)
| |
Collapse
|
29
|
Fang H, Hu L, Chen Q, Geng S, Qiu K, Wang C, Hao M, Tian Z, Chen H, Liu L, Guan JL, Chen Y, Dong L, Guo Z, He W, Diao J. An ER-targeted "reserve-release" fluorogen for topological quantification of reticulophagy. Biomaterials 2023; 292:121929. [PMID: 36455487 DOI: 10.1016/j.biomaterials.2022.121929] [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: 03/27/2022] [Revised: 11/09/2022] [Accepted: 11/21/2022] [Indexed: 11/23/2022]
Abstract
The endoplasmic reticulum's (ER) dynamic nature, essential for maintaining cellular homeostasis, can be influenced by stress-induced damage, which can be assessed by examining the morphology of ER dynamics and, more locally, ER properties such as hydrophobicity, viscosity, and polarity. Although numerous ER-specific chemical probes have been developed to monitor the ER's physical and chemical parameters, the quantitative detection and super-resolution imaging of its local hydrophobicity have yet to be explored. Here, we describe a photostable ER-targeted probe with high signal-to-noise ratio for super-resolution imaging that can specifically respond to changes in ER hydrophobicity under stress based on a "reserve-release" mechanism. The probe shows an excellent ability to target ER over commercial ER dyes and can be used to track local changes of hydrophobicity by fluorescence intensity and morphology during the selective autophagy of ER (i.e., reticulophagy). By correlating the level and location of ER damage with the distribution of fluorescence intensity, we were able to assess reticulophagy at the subcellular level. Beyond that, we developed a topological analytical tool adaptable to any ER probe for detecting structural changes in ER and thus quantitatively identifying reticulophagy. The algorithm-assisted tool can also be adapted to a wide range of molecular probes and organelles. Altogether, the new probe and analytical strategy described here show promise for the quantitative detection and analysis of subtle ER damage and stress.
Collapse
Affiliation(s)
- Hongbao Fang
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China; Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Lianting Hu
- Medical Big Data Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China; Guangdong Cardiovascular Institute, Guangzhou, 510080, China; School of Information Management, Wuhan University, Wuhan 430072, China
| | - Qixin Chen
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA; Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Shanshan Geng
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Kangqiang Qiu
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Chengjun Wang
- Sinopec Shengli Petroleum Engineering Limited Company, Dongying, 257000, China
| | - Mingang Hao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Zhiqi Tian
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Huimin Chen
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Lei Liu
- Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Jun-Lin Guan
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
| | - Yuncong Chen
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Lei Dong
- School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Weijiang He
- State Key Laboratory of Coordination Chemistry, Chemistry and Biomedicine Innovation Center (ChemBIC), School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, China.
| | - Jiajie Diao
- Department of Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA.
| |
Collapse
|
30
|
Mazzei A, Pagliara P, Del Vecchio G, Giampetruzzi L, Croce F, Schiavone R, Verri T, Barca A. Cytoskeletal Responses and Aif-1 Expression in Caco-2 Monolayers Exposed to Phorbol-12-Myristate-13-Acetate and Carnosine. BIOLOGY 2022; 12:biology12010036. [PMID: 36671729 PMCID: PMC9855102 DOI: 10.3390/biology12010036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/01/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022]
Abstract
The dis(re)organization of the cytoskeletal actin in enterocytes mediates epithelial barrier dys(re)function, playing a key role in modulating epithelial monolayer's integrity and remodeling under transition from physiological to pathological states. Here, by fluorescence-based morphological and morphometric analyses, we detected differential responses of cytoskeletal actin in intestinal epithelial Caco-2 cell monolayers at two different stages of their spontaneous differentiation, i.e., undifferentiated cells at 7 days post-seeding (dps) and differentiated enterocyte-like cells at 21 dps, upon challenge in vitro with the inflammation-mimicking stimulus of phorbol-12-myristate-13-acetate (PMA). In addition, specific responses were found in the presence of the natural dipeptide carnosine detecting its potential counteraction against PMA-induced cytoskeletal alterations and remodeling in differentiated Caco-2 monolayers. In such an experimental context, by both immunocytochemistry and Western blot assays in Caco-2 monolayers, we identified the expression of the allograft inflammatory factor 1 (AIF-1) as protein functionally related to both inflammatory and cytoskeletal pathways. In 21 dps monolayers, particularly, we detected variations of its intracellular localization associated with the inflammatory stimulus and its mRNA/protein increase associated with the differentiated 21 dps enterocyte-like monolayer compared to the undifferentiated cells.
Collapse
Affiliation(s)
- Aurora Mazzei
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
| | - Patrizia Pagliara
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
- Correspondence: (P.P.); (A.B.); Tel.: +39-0832-298662 (A.B.)
| | - Gianmarco Del Vecchio
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
| | - Lucia Giampetruzzi
- Institute for Microelectronics and Microsystems IMM-CNR, Via per Monteroni “Campus Ecotekne”, 73100 Lecce, Italy
| | - Francesca Croce
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
| | - Roberta Schiavone
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
| | - Tiziano Verri
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
| | - Amilcare Barca
- Department of Biological and Environmental Sciences and Technologies (DeBEST), University of Salento, 73100 Lecce, Italy
- Correspondence: (P.P.); (A.B.); Tel.: +39-0832-298662 (A.B.)
| |
Collapse
|
31
|
Chu S, Moujaber O, Lemay S, Stochaj U. Multiple pathways promote microtubule stabilization in senescent intestinal epithelial cells. NPJ AGING 2022; 8:16. [PMID: 36526654 PMCID: PMC9758230 DOI: 10.1038/s41514-022-00097-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 11/25/2022] [Indexed: 12/23/2022]
Abstract
Intestinal epithelial cells are critical for gastrointestinal homeostasis. However, their function declines during aging. The aging-related loss of organ performance is largely driven by the increase in senescent cells. To date, the hallmarks and molecular mechanisms related to cellular senescence are not fully understood. Microtubules control epithelial functions, and we identified microtubule stabilization as a phenotypic marker of senescent intestinal epithelial cells. The senescence inducer determined the pathway to microtubule stabilization. Specifically, enhanced microtubule stability was associated with α-tubulin hyperacetylation or increased abundance of the microtubule-binding protein tau. We show further that overexpression of MAPT, which encodes tau, augmented microtubule stability in intestinal epithelial cells. Notably, pharmacological microtubule stabilization was sufficient to induce cellular senescence. Taken together, this study provides new insights into the molecular mechanisms that control epithelial cell homeostasis. Our results support the concept that microtubule stability serves as a critical cue to trigger intestinal epithelial cell senescence.
Collapse
Affiliation(s)
- Siwei Chu
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6 Canada
| | - Ossama Moujaber
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6 Canada
| | - Serge Lemay
- grid.63984.300000 0000 9064 4811Department of Medicine, Division of Nephrology, McGill University Health Centre, Montreal, QC Canada
| | - Ursula Stochaj
- grid.14709.3b0000 0004 1936 8649Department of Physiology, McGill University, Montreal, Quebec H3G 1Y6 Canada
| |
Collapse
|
32
|
Duwe L, Fouassier L, Lafuente-Barquero J, Andersen JB. Unraveling the actin cytoskeleton in the malignant transformation of cholangiocyte biology. Transl Oncol 2022; 26:101531. [PMID: 36113344 PMCID: PMC9483793 DOI: 10.1016/j.tranon.2022.101531] [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: 07/16/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/13/2022] Open
Abstract
Correct actin cytoskeleton organization is vital in the liver organ homeostasis and disease control. Rearrangements of the actin cytoskeleton may play a vital role in the bile duct cells cholangiocytes. An abnormal actin network leads to aberrant cell morphology, deregulated signaling networks and ultimately triggering the development of cholangiocarcinoma (CCA) and paving the route for cancer cell dissemination (metastasis). In this review, we will outline alterations of the actin cytoskeleton and the potential role of this dynamic network in initiating CCA, as well as regulating the course of this malignancy. Actin rearrangements not only occur because of signaling pathways, but also regulate and modify cellular signaling. This emphasizes the importance of the actin cytoskeleton itself as cause for aberrant signaling and in promoting tumorigenic phenotypes. We will highlight the impact of aberrant signaling networks on the actin cytoskeleton and its rearrangement as potential cause for CCA. Often, these exact mechanisms in CCA are limited understood and still must be elucidated. Indeed, focusing future research on how actin affects and regulates other signaling pathways may provide more insights into the mechanisms of CCA development, progression, and metastasis. Moreover, manipulation of the actin cytoskeleton organization highlights the potential for a novel therapeutic area.
Collapse
Affiliation(s)
- Lea Duwe
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK2200, Denmark
| | - Laura Fouassier
- Sorbonne Université, Inserm, Centre de Recherche Saint-Antoine, CRSA, Paris, France
| | - Juan Lafuente-Barquero
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK2200, Denmark
| | - Jesper B Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen N DK2200, Denmark.
| |
Collapse
|
33
|
Merino-Casallo F, Gomez-Benito MJ, Hervas-Raluy S, Garcia-Aznar JM. Unravelling cell migration: defining movement from the cell surface. Cell Adh Migr 2022; 16:25-64. [PMID: 35499121 PMCID: PMC9067518 DOI: 10.1080/19336918.2022.2055520] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/10/2022] [Indexed: 12/13/2022] Open
Abstract
Cell motility is essential for life and development. Unfortunately, cell migration is also linked to several pathological processes, such as cancer metastasis. Cells' ability to migrate relies on many actors. Cells change their migratory strategy based on their phenotype and the properties of the surrounding microenvironment. Cell migration is, therefore, an extremely complex phenomenon. Researchers have investigated cell motility for more than a century. Recent discoveries have uncovered some of the mysteries associated with the mechanisms involved in cell migration, such as intracellular signaling and cell mechanics. These findings involve different players, including transmembrane receptors, adhesive complexes, cytoskeletal components , the nucleus, and the extracellular matrix. This review aims to give a global overview of our current understanding of cell migration.
Collapse
Affiliation(s)
- Francisco Merino-Casallo
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Maria Jose Gomez-Benito
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Silvia Hervas-Raluy
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Jose Manuel Garcia-Aznar
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
34
|
Nevarez AJ, Hao N. Quantitative cell imaging approaches to metastatic state profiling. Front Cell Dev Biol 2022; 10:1048630. [PMID: 36393865 PMCID: PMC9640958 DOI: 10.3389/fcell.2022.1048630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
Genetic heterogeneity of metastatic dissemination has proven challenging to identify exploitable markers of metastasis; this bottom-up approach has caused a stalemate between advances in metastasis and the late stage of the disease. Advancements in quantitative cellular imaging have allowed the detection of morphological phenotype changes specific to metastasis, the morphological changes connected to the underlying complex signaling pathways, and a robust readout of metastatic cell state. This review focuses on the recent machine and deep learning developments to gain detailed information about the metastatic cell state using light microscopy. We describe the latest studies using quantitative cell imaging approaches to identify cell appearance-based metastatic patterns. We discuss how quantitative cancer biologists can use these frameworks to work backward toward exploitable hidden drivers in the metastatic cascade and pioneering new Frontier drug discoveries specific for metastasis.
Collapse
Affiliation(s)
| | - Nan Hao
- *Correspondence: Andres J. Nevarez, ; Nan Hao,
| |
Collapse
|
35
|
Lestrell E, Chen Y, Aslanoglou S, O'Brien CM, Elnathan R, Voelcker NH. Silicon Nanoneedle-Induced Nuclear Deformation: Implications for Human Somatic and Stem Cell Nuclear Mechanics. ACS APPLIED MATERIALS & INTERFACES 2022; 14:45124-45136. [PMID: 36173149 DOI: 10.1021/acsami.2c10583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Cell nuclear size and shape are strictly regulated, with aberrations often leading to or being indicative of disease. Nuclear mechanics are critically responsible for intracellular responses to extracellular cues, such as the nanotopography of the external environment. Silicon nanoneedle (SiNN) arrays are tunable, engineered cell culture substrates that permit precise, nanoscale modifications to a cell's external environment to probe mechanotransduction and intracellular signaling. We use a library of four different SiNN arrays to investigate the immediate and downstream effects of controlled geometries of nanotopographical cues on the nuclear integrity/dynamics of human immortalized somatic and renewing stem cell types. We quantify the significant, albeit different, nuclear shape changes that both cell types undergo, which suggest that cellular responses to SiNN arrays are more comparable to three-dimensional (3D) environments than traditional flat cultureware. We show that nanotopography-induced effects on nuclear envelope integrity, protein localization, and focal adhesion complex formation are cell-dependent. Migration is shown to be dramatically impeded for human neural progenitor cells (hNPCs) on nanotopographies compared to flat substrates but not for somatic cells. Our results indicate an additional layer of complexity in cellular mechanotransduction, which warrants closer attention in the context of engineered substrates and scaffolds for clinical applications.
Collapse
Affiliation(s)
- Esther Lestrell
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, Victoria 3168, Australia
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| | - Yaping Chen
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, Victoria 3168, Australia
| | - Stella Aslanoglou
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, Victoria 3168, Australia
| | - Carmel M O'Brien
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3168, Australia
| | - Roey Elnathan
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, Victoria 3168, Australia
| | - Nicolas H Voelcker
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, 151 Wellington Road, Clayton, Victoria 3168, Australia
- CSIRO Manufacturing, Clayton, Victoria 3168, Australia
| |
Collapse
|
36
|
Wong CW, Han HW, Hsu SH. Changes of cell membrane fluidity for mesenchymal stem cell spheroids on biomaterial surfaces. World J Stem Cells 2022; 14:616-632. [PMID: 36157913 PMCID: PMC9453270 DOI: 10.4252/wjsc.v14.i8.616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/02/2022] [Accepted: 07/11/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The therapeutic potential of mesenchymal stem cells (MSCs) in the form of three-dimensional spheroids has been extensively demonstrated. The underlying mechanisms for the altered cellular behavior of spheroids have also been investigated. Cell membrane fluidity is a critically important physical property for the regulation of cell behavior, but it has not been studied for the spheroid-forming cells to date.
AIM To explore the association between cell membrane fluidity and the morphological changes of MSC spheroids on the surface of biomaterials to elucidate the role of membrane fluidity during the spheroid-forming process of MSCs.
METHODS We generated three-dimensional (3D) MSC spheroids on the surface of various culture substrates including chitosan (CS), CS-hyaluronan (CS-HA), and polyvinyl alcohol (PVA) substrates. The cell membrane fluidity and cell morphological change were examined by a time-lapse recording system as well as a high-resolution 3D cellular image explorer. MSCs and normal/cancer cells were pre-stained with fluorescent dyes and co-cultured on the biomaterials to investigate the exchange of cell membrane during the formation of heterogeneous cellular spheroids.
RESULTS We discovered that vesicle-like bubbles randomly appeared on the outer layer of MSC spheroids cultured on different biomaterial surfaces. The average diameter of the vesicle-like bubbles of MSC spheroids on CS-HA at 37 °C was approximately 10 μm, smaller than that on PVA substrates (approximately 27 μm). Based on time-lapse images, these unique bubbles originated from the dynamic movement of the cell membrane during spheroid formation, which indicated an increment of membrane fluidity for MSCs cultured on these substrates. Moreover, the membrane interaction in two different types of cells with similar membrane fluidity may further induce a higher level of membrane translocation during the formation of heterogeneous spheroids.
CONCLUSION Changes in cell membrane fluidity may be a novel path to elucidate the complicated physiological alterations in 3D spheroid-forming cells.
Collapse
Affiliation(s)
- Chui-Wei Wong
- National Taiwan University, Institute of Polymer Science and Engineering, Taipei 10617, Taiwan
| | - Hao-Wei Han
- National Taiwan University, Institute of Polymer Science and Engineering, Taipei 10617, Taiwan
| | - Shan-hui Hsu
- National Taiwan University, Institute of Polymer Science and Engineering, Taipei 10617, Taiwan
- National Health Research Institutes, Institute of Cellular and System Medicine, Miaoli 350, Taiwan
- National Taiwan University, Research and Development Center for Medical Devices, Taipei 10617, Taiwan
| |
Collapse
|
37
|
Svadlakova T, Holmannova D, Kolackova M, Malkova A, Krejsek J, Fiala Z. Immunotoxicity of Carbon-Based Nanomaterials, Starring Phagocytes. Int J Mol Sci 2022; 23:ijms23168889. [PMID: 36012161 PMCID: PMC9408998 DOI: 10.3390/ijms23168889] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
In the field of science, technology and medicine, carbon-based nanomaterials and nanoparticles (CNMs) are becoming attractive nanomaterials that are increasingly used. However, it is important to acknowledge the risk of nanotoxicity that comes with the widespread use of CNMs. CNMs can enter the body via inhalation, ingestion, intravenously or by any other route, spread through the bloodstream and penetrate tissues where (in both compartments) they interact with components of the immune system. Like invading pathogens, CNMs can be recognized by large numbers of receptors that are present on the surface of innate immune cells, notably monocytes and macrophages. Depending on the physicochemical properties of CNMs, i.e., shape, size, or adsorbed contamination, phagocytes try to engulf and process CNMs, which might induce pro/anti-inflammatory response or lead to modulation and disruption of basic immune activity. This review focuses on existing data on the immunotoxic potential of CNMs, particularly in professional phagocytes, as they play a central role in processing and eliminating foreign particles. The results of immunotoxic studies are also described in the context of the entry routes, impacts of contamination and means of possible elimination. Mechanisms of proinflammatory effect depending on endocytosis and intracellular distribution of CNMs are highlighted as well.
Collapse
Affiliation(s)
- Tereza Svadlakova
- Department of Clinical Immunology and Allergology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic
- Department of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50003 Hradec Kralove, Czech Republic
- Correspondence:
| | - Drahomira Holmannova
- Department of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50003 Hradec Kralove, Czech Republic
| | - Martina Kolackova
- Department of Clinical Immunology and Allergology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic
| | - Andrea Malkova
- Department of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50003 Hradec Kralove, Czech Republic
- Department of Pathological Physiology, Faculty of Medicine in Hradec Kralove, Charles University, 50003 Hradec Kralove, Czech Republic
| | - Jan Krejsek
- Department of Clinical Immunology and Allergology, University Hospital Hradec Kralove and Faculty of Medicine in Hradec Kralove, Charles University, 50005 Hradec Kralove, Czech Republic
| | - Zdenek Fiala
- Department of Preventive Medicine, Faculty of Medicine in Hradec Kralove, Charles University, 50003 Hradec Kralove, Czech Republic
| |
Collapse
|
38
|
Khalil K, Eon A, Janody F. Cell Architecture-Dependent Constraints: Critical Safeguards to Carcinogenesis. Int J Mol Sci 2022; 23:8622. [PMID: 35955754 PMCID: PMC9369145 DOI: 10.3390/ijms23158622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/29/2022] [Accepted: 07/30/2022] [Indexed: 02/04/2023] Open
Abstract
Animal cells display great diversity in their shape. These morphological characteristics result from crosstalk between the plasma membrane and the force-generating capacities of the cytoskeleton macromolecules. Changes in cell shape are not merely byproducts of cell fate determinants, they also actively drive cell fate decisions, including proliferation and differentiation. Global and local changes in cell shape alter the transcriptional program by a multitude of mechanisms, including the regulation of physical links between the plasma membrane and the nuclear envelope and the mechanical modulation of cation channels and signalling molecules. It is therefore not surprising that anomalies in cell shape contribute to several diseases, including cancer. In this review, we discuss the possibility that the constraints imposed by cell shape determine the behaviour of normal and pro-tumour cells by organizing the whole interconnected regulatory network. In turn, cell behaviour might stabilize cells into discrete shapes. However, to progress towards a fully transformed phenotype and to acquire plasticity properties, pro-tumour cells might need to escape these cell shape restrictions. Thus, robust controls of the cell shape machinery may represent a critical safeguard against carcinogenesis.
Collapse
Affiliation(s)
- Komal Khalil
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Master Programme in Oncology, School of Medicine & Biomedical Sciences, University of Porto (ICBAS-UP), Rua Jorge Viterbo Ferreira 228, 4050-513 Porto, Portugal
| | - Alice Eon
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
- Magistère Européen de Génétique, Université Paris Cité, 5 Rue Thomas Mann, 75013 Paris, France
| | - Florence Janody
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; (K.K.); (A.E.)
- IPATIMUP—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| |
Collapse
|
39
|
Jung J, Khan MM, Landry J, Halavatyi A, Machado P, Reiss M, Pepperkok R. Regulation of the COPII secretory machinery via focal adhesions and extracellular matrix signaling. J Cell Biol 2022; 221:213351. [PMID: 35829701 PMCID: PMC9284426 DOI: 10.1083/jcb.202110081] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 05/10/2022] [Accepted: 06/24/2022] [Indexed: 12/19/2022] Open
Abstract
Proteins that enter the secretory pathway are transported from their place of synthesis in the endoplasmic reticulum to the Golgi complex by COPII-coated carriers. The networks of proteins that regulate these components in response to extracellular cues have remained largely elusive. Using high-throughput microscopy, we comprehensively screened 378 cytoskeleton-associated and related proteins for their functional interaction with the coat protein complex II (COPII) components SEC23A and SEC23B. Among these, we identified a group of proteins associated with focal adhesions (FERMT2, MACF1, MAPK8IP2, NGEF, PIK3CA, and ROCK1) that led to the downregulation of SEC23A when depleted by siRNA. Changes in focal adhesions induced by plating cells on ECM also led to the downregulation of SEC23A and decreases in VSVG transport from ER to Golgi. Both the expression of SEC23A and the transport defect could be rescued by treatment with a focal adhesion kinase inhibitor. Altogether, our results identify a network of cytoskeleton-associated proteins connecting focal adhesions and ECM-related signaling with the gene expression of the COPII secretory machinery and trafficking.
Collapse
Affiliation(s)
- Juan Jung
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Muzamil Majid Khan
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| | - Jonathan Landry
- Core Facilities Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Aliaksandr Halavatyi
- Core Facilities Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Pedro Machado
- Core Facilities Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Miriam Reiss
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Rainer Pepperkok
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Core Facilities Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Translational Lung Research Center Heidelberg, German Center for Lung Research, Heidelberg, Germany
| |
Collapse
|
40
|
Cook S, Lenardo MJ, Freeman AF. HEM1 Actin Immunodysregulatory Disorder: Genotypes, Phenotypes, and Future Directions. J Clin Immunol 2022; 42:1583-1592. [DOI: 10.1007/s10875-022-01327-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/01/2022] [Indexed: 11/30/2022]
Abstract
AbstractCells of the innate and adaptive immune systems depend on proper actin dynamics to control cell behavior for effective immune responses. Dysregulated actin networks are known to play a pathogenic role in an increasing number of inborn errors of immunity. The WAVE regulatory complex (WRC) mediates branched actin polymerization, a process required for key cellular functions including migration, phagocytosis, vesicular transport, and immune synapse formation. Recent reports of pathogenic variants in NCKAP1L, a hematopoietically restricted gene encoding the HEM1 protein component of the WRC, defined a novel disease involving recurrent bacterial and viral infections, autoimmunity, and excessive inflammation (OMIM 141180). This review summarizes the diverse clinical presentations and immunological phenotypes observed in HEM1-deficient patients. In addition, we integrate the pathophysiological mechanisms described in current literature and highlight the outstanding questions for diagnosis and management of the HEM1 actin immunodysregulatory disorder.
Collapse
|
41
|
Gill MR, Jarman PJ, Hearnden V, Fairbanks SD, Bassetto M, Maib H, Palmer J, Ayscough KR, Thomas JA, Smythe C. A Ruthenium(II) Polypyridyl Complex Disrupts Actin Cytoskeleton Assembly and Blocks Cytokinesis. Angew Chem Int Ed Engl 2022; 61:e202117449. [PMID: 35416386 PMCID: PMC9323417 DOI: 10.1002/anie.202117449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Indexed: 11/05/2022]
Abstract
The dinuclear RuII complex [(Ru(phen)2)2(tpphz)]4+ (phen=1,10‐phenanthroline, tpphz=tetrapyridophenazine) “RuRuPhen” blocks the transformation of G‐actin monomers to F‐actin filaments with no disassembly of pre‐formed F‐actin. Molecular docking studies indicate multiple RuRuPhen molecules bind to the surface of G‐actin but not the binding pockets of established actin polymerisation inhibitors. In cells, addition of RuRuPhen causes rapid disruption to actin stress fibre organisation, compromising actomyosin contractility and cell motility; due to this effect RuRuPhen interferes with late‐stage cytokinesis. Immunofluorescent microscopy reveals that RuRuPhen causes cytokinetic abscission failure by interfering with endosomal sorting complexes required for transport (ESCRT) complex recruitment.
Collapse
Affiliation(s)
- Martin R. Gill
- Department of Chemistry Faculty of Science and Engineering Swansea University UK
| | - Paul J. Jarman
- Department of Biomedical Science University of Sheffield UK
| | - Vanessa Hearnden
- Department of Materials Science and Engineering University of Sheffield UK
| | | | - Marcella Bassetto
- Department of Chemistry Faculty of Science and Engineering Swansea University UK
| | - Hannes Maib
- Department of Biomedical Science University of Sheffield UK
| | - John Palmer
- Department of Biomedical Science University of Sheffield UK
| | | | | | - Carl Smythe
- Department of Biomedical Science University of Sheffield UK
| |
Collapse
|
42
|
Gill MR, Jarman PJ, Hearnden V, Fairbanks SD, Bassetto M, Maib H, Palmer J, Ayscough KR, Thomas JA, Smythe C. A Ruthenium(II) Polypyridyl Complex Disrupts Actin Cytoskeleton Assembly and Blocks Cytokinesis. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 134:e202117449. [PMID: 38505667 PMCID: PMC10947085 DOI: 10.1002/ange.202117449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Indexed: 11/10/2022]
Abstract
The dinuclear RuII complex [(Ru(phen)2)2(tpphz)]4+ (phen=1,10-phenanthroline, tpphz=tetrapyridophenazine) "RuRuPhen" blocks the transformation of G-actin monomers to F-actin filaments with no disassembly of pre-formed F-actin. Molecular docking studies indicate multiple RuRuPhen molecules bind to the surface of G-actin but not the binding pockets of established actin polymerisation inhibitors. In cells, addition of RuRuPhen causes rapid disruption to actin stress fibre organisation, compromising actomyosin contractility and cell motility; due to this effect RuRuPhen interferes with late-stage cytokinesis. Immunofluorescent microscopy reveals that RuRuPhen causes cytokinetic abscission failure by interfering with endosomal sorting complexes required for transport (ESCRT) complex recruitment.
Collapse
Affiliation(s)
- Martin R. Gill
- Department of ChemistryFaculty of Science and EngineeringSwansea UniversityUK
| | - Paul J. Jarman
- Department of Biomedical ScienceUniversity of SheffieldUK
| | - Vanessa Hearnden
- Department of Materials Science and EngineeringUniversity of SheffieldUK
| | | | - Marcella Bassetto
- Department of ChemistryFaculty of Science and EngineeringSwansea UniversityUK
| | - Hannes Maib
- Department of Biomedical ScienceUniversity of SheffieldUK
| | - John Palmer
- Department of Biomedical ScienceUniversity of SheffieldUK
| | | | | | - Carl Smythe
- Department of Biomedical ScienceUniversity of SheffieldUK
| |
Collapse
|
43
|
Xu W, Liu X, Liu X. Modeling the dynamic growth and branching of actin filaments. SOFT MATTER 2022; 18:3649-3659. [PMID: 35438124 DOI: 10.1039/d2sm00283c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
As an essential component of the cytoskeleton, actin filaments play a key role in a variety of cellular physiological activities. To better understand the function of actin filaments, which are a special kind of polymer chain, researchers have started to focus on the Brownian dynamics of polymers. Currently, to study the dynamics of polymers, classical explicit bead-spring models and finite-element methods (FEMs) have both been broadly used. However, compared to bead-spring models, FEMs can address the mechanical properties of actin filaments and actin networks with more detail and better accuracy. However, current FEMs do not consider the dynamic assembly of actin into an actin filament network. Here, we extend the traditional FEM and present a new framework of the FEM based on the co-rotational grid method, which allows us to simulate the dynamic growth and branching of actin filaments. Several examples are studied. The proposed numerical model is capable of capturing the dynamic assembly of actin filaments.
Collapse
Affiliation(s)
- Wu Xu
- Department of Mechanics, Huazhong University of Science and Technology, Luoyu Road 1037, 430074, Wuhan, China.
- Hubei Key Laboratory of Engineering Structural Analysis and Safety Assessment, Luoyu Road 1037, 430074, Wuhan, China
| | - Xuheng Liu
- School of Civil Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaohu Liu
- Department of Mechanics, Huazhong University of Science and Technology, Luoyu Road 1037, 430074, Wuhan, China.
- Hubei Key Laboratory of Engineering Structural Analysis and Safety Assessment, Luoyu Road 1037, 430074, Wuhan, China
| |
Collapse
|
44
|
Huang Z, Zhang Z, Zhou C, Liu L, Huang C. Epithelial–mesenchymal transition: The history, regulatory mechanism, and cancer therapeutic opportunities. MedComm (Beijing) 2022; 3:e144. [PMID: 35601657 PMCID: PMC9115588 DOI: 10.1002/mco2.144] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 02/05/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a program wherein epithelial cells lose their junctions and polarity while acquiring mesenchymal properties and invasive ability. Originally defined as an embryogenesis event, EMT has been recognized as a crucial process in tumor progression. During EMT, cell–cell junctions and cell–matrix attachments are disrupted, and the cytoskeleton is remodeled to enhance mobility of cells. This transition of phenotype is largely driven by a group of key transcription factors, typically Snail, Twist, and ZEB, through epigenetic repression of epithelial markers, transcriptional activation of matrix metalloproteinases, and reorganization of cytoskeleton. Mechanistically, EMT is orchestrated by multiple pathways, especially those involved in embryogenesis such as TGFβ, Wnt, Hedgehog, and Hippo, suggesting EMT as an intrinsic link between embryonic development and cancer progression. In addition, redox signaling has also emerged as critical EMT modulator. EMT confers cancer cells with increased metastatic potential and drug resistant capacity, which accounts for tumor recurrence in most clinic cases. Thus, targeting EMT can be a therapeutic option providing a chance of cure for cancer patients. Here, we introduce a brief history of EMT and summarize recent advances in understanding EMT mechanisms, as well as highlighting the therapeutic opportunities by targeting EMT in cancer treatment.
Collapse
Affiliation(s)
- Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Zhe Zhang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| | - Chengwei Zhou
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Lin Liu
- Department of Thoracic Surgery the Affiliated Hospital of Medical School of Ningbo University Ningbo China
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine Sichuan University, and Collaborative Innovation Center for Biotherapy Chengdu 610041 China
| |
Collapse
|
45
|
Che H, Selig M, Rolauffs B. Micro-patterned cell populations as advanced pharmaceutical drugs with precise functional control. Adv Drug Deliv Rev 2022; 184:114169. [PMID: 35217114 DOI: 10.1016/j.addr.2022.114169] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/29/2022]
Abstract
Human cells are both advanced pharmaceutical drugs and 'drug deliverers'. However, functional control prior to or after cell implantation remains challenging. Micro-patterning cells through geometrically defined adhesion sites allows controlling morphogenesis, polarity, cellular mechanics, proliferation, migration, differentiation, stemness, cell-cell interactions, collective cell behavior, and likely immuno-modulatory properties. Consequently, generating micro-patterned therapeutic cells is a promising idea that has not yet been realized and few if any steps have been undertaken in this direction. This review highlights potential therapeutic applications, summarizes comprehensively the many cell functions that have been successfully controlled through micro-patterning, details the established micro-pattern designs, introduces the available fabrication technologies to the non-specialized reader, and suggests a quality evaluation score. Such a broad review is not yet available but would facilitate the manufacturing of therapeutically patterned cell populations using micro-patterned cell-instructive biomaterials for improved functional control as drug delivery systems in the context of cells as pharmaceutical products.
Collapse
Affiliation(s)
- Hui Che
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; Orthopedics and Sports Medicine Center, Suzhou Municipal Hospital (North District), Nanjing Medical University Affiliated Suzhou Hospital, Suzhou 215006, China
| | - Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany; Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, D-79104 Freiburg, Germany
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, 79085 Freiburg im Breisgau, Germany.
| |
Collapse
|
46
|
Huang Z, Khalifa MO, Gu W, Li TS. Hydrostatic pressure induces pro-fibrotic properties in hepatic stellate cells via the RhoA/ROCK signaling pathway. FEBS Open Bio 2022; 12:1230-1240. [PMID: 35357779 PMCID: PMC9157409 DOI: 10.1002/2211-5463.13405] [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: 10/31/2021] [Revised: 02/28/2022] [Accepted: 03/29/2022] [Indexed: 11/11/2022] Open
Abstract
Elevated interstitial fluid hydrostatic pressure is commonly observed in diseased livers. We herein examined the hypothesis that hydrostatic pressure induces hepatic stellate cells to acquire pro-fibrotic properties under pathological conditions. Human hepatic stellate cells were exposed to 50 mmHg pressure for 24 hours. Although we observed few changes of cell growth and morphology, PCR array data on the expression of fibrosis-associated genes suggested the acquisition of pro-fibrotic properties. The exposure of hepatic stellate cells to 50 mmHg pressure for 24 hours also significantly enhanced the expression of RhoA, ROCK1, α-SMA, TGF-β1 , p-MLC and p-Smad2, and this was effectively attenuated by ROCK inhibitor Y-27632. Our ex vivo experimental data suggests that elevated interstitial fluid hydrostatic pressure under pathological conditions may promote liver fibrosis by inducing acquisition of pro-fibrotic properties of hepatic stellate cells through the RhoA/ROCK signaling pathway.
Collapse
Affiliation(s)
- Zisheng Huang
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Mahmoud Osman Khalifa
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Weili Gu
- Department of Hepatopancreatobiliary Surgery, Guangzhou First People's Hospital, Guangzhou, 510180, China
| | - Tao-Sheng Li
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan.,Department of Stem Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| |
Collapse
|
47
|
Lu X, Wang Y, Chen C, Feng Z, Huo Y, Zhou D. C12-HSL is an across-boundary signal molecule that could alleviate fungi Galactomyces's filamentation: A new mechanism on activated sludge bulking. ENVIRONMENTAL RESEARCH 2022; 204:111823. [PMID: 34400160 DOI: 10.1016/j.envres.2021.111823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 06/13/2023]
Abstract
Fungal bulking is caused by fungi excessive growth and morphological changes, resulting from the evolution toward fungi dominant activated sludge. Communication across fungi and bacteria boundary that mediated by bacterial signal molecules (SMs) probably is the central induce caused fungal bulking occurrence. In this work, it intended to identify the bacterial SM that affected fungal bulking, and verified its roles in regulate the spore germination and hyphal growth. We found C12-HSL concentration decreased significantly from 12.36 to 3.38 ng/g-VSS (P < 0.05) when fungal sludge bulking happened, and filamentous Galactomyces's relatively abundant was correlatively enriched. To test the effects of this SM, trace commercial C12-HSL was added to pure cultured Galactomyces, in which spore germination rates decreased by 20 % and hyphal extension inhibited by 15 %. Ras1-cAMP-PKA and mitogen-activated protein kinase (MAPK) pathways of Galactomyces were responsible for signal C12-HSL transduction, which inhibited peroxisome biosynthesis, suppressed the biological activity of the actin cytoskeleton, and disrupted intercellular organelle transport. All these results showed C12-HSL was the functional SM that could suppress the development of fungal filamentous. This study provided a new insight into the sludge bulking mechanism from view of cross-kingdom communication.
Collapse
Affiliation(s)
- Xin Lu
- Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun, 130117, China
| | - Yue Wang
- Quality, Safety & Environmental Protection Department, Shanxi Road & Bridge Construction Group Co., Ltd, Taiyuan, 030000, China
| | - Congli Chen
- Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun, 130117, China
| | - Zhixuan Feng
- Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun, 130117, China
| | - Yang Huo
- School of Physics, Northeast Normal University, Changchun, 130117, China.
| | - Dandan Zhou
- Jilin Engineering Lab for Water Pollution Control and Resources Recovery, Northeast Normal University, Changchun, 130117, China.
| |
Collapse
|
48
|
Abstract
Apoptosis plays a key role in removing abnormal or senescent cells, maintaining the overall health of the tissue, and coordinating individual development. Recently, it has been discovered that the intracellular cytoskeleton plays a role in the apoptotic process. In addition, the regulatory role of extracellular matrix (ECM) fibrous proteins, which can be considered as the extracellular skeleton, in the process of apoptosis is rarely summarized. In this review, we collect the latest knowledge about how fibrous proteins inside and outside cells regulate apoptosis. We describe how ECM fibrous proteins participate in the regulation of death receptor and mitochondrial pathways through various signaling cascades mediated by integrins. We then explore the molecular mechanisms by which intracellular intermediate filaments regulate cell apoptosis by regulating death receptors on the cell membrane surface. Similarly, we report on novel supporting functions of microtubules in the execution phase of apoptosis and discuss their formation mechanisms. Finally, we discuss that the polypeptide fragments formed by caspase degradation of ECM fibrous proteins and intracellular intermediate filament act as local regulatory signals to play different regulatory roles in apoptosis.
Collapse
Affiliation(s)
- Jia-Hao Ni
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wan-Xi Yang
- The Sperm Laboratory, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
49
|
Lu H, Aida H, Kurokawa M, Chen F, Xia Y, Xu J, Li K, Ying BW, Yomo T. Primordial mimicry induces morphological change in Escherichia coli. Commun Biol 2022; 5:24. [PMID: 35017623 PMCID: PMC8752768 DOI: 10.1038/s42003-021-02954-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 12/07/2021] [Indexed: 11/09/2022] Open
Abstract
The morphology of primitive cells has been the subject of extensive research. A spherical form was commonly presumed in prebiotic studies but lacked experimental evidence in living cells. Whether and how the shape of living cells changed are unclear. Here we exposed the rod-shaped bacterium Escherichia coli to a resource utilization regime mimicking a primordial environment. Oleate was given as an easy-to-use model prebiotic nutrient, as fatty acid vesicles were likely present on the prebiotic Earth and might have been used as an energy resource. Six evolutionary lineages were generated under glucose-free but oleic acid vesicle (OAV)-rich conditions. Intriguingly, fitness increase was commonly associated with the morphological change from rod to sphere and the decreases in both the size and the area-to-volume ratio of the cell. The changed cell shape was conserved in either OAVs or glucose, regardless of the trade-offs in carbon utilization and protein abundance. Highly differentiated mutations present in the genome revealed two distinct strategies of adaption to OAV-rich conditions, i.e., either directly targeting the cell wall or not. The change in cell morphology of Escherichia coli for adapting to fatty acid availability supports the assumption of the primitive spherical form. Lu et al. investigate the evolution of the shape of living cells by generating six experimental lineages of the rod-shaped E. coli under glucose-free conditions in the presence of oleic acid mimicking a primordial environment. The authors show that the morphological changes from rod to sphere accompanied fitness increases and adaptation amongst fatty acid availability supports the assumption of a primitive spherical form.
Collapse
Affiliation(s)
- Hui Lu
- Biomedical Synthetic Biology Research Center, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, PR China
| | - Honoka Aida
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Masaomi Kurokawa
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8572, Japan
| | - Feng Chen
- School of Software Engineering, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, PR China
| | - Yang Xia
- Biomedical Synthetic Biology Research Center, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, PR China
| | - Jian Xu
- Biomedical Synthetic Biology Research Center, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, PR China
| | - Kai Li
- Biomedical Synthetic Biology Research Center, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, PR China
| | - Bei-Wen Ying
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennoudai, Tsukuba, Ibaraki, 305-8572, Japan.
| | - Tetsuya Yomo
- Biomedical Synthetic Biology Research Center, School of Life Sciences, East China Normal University, 3663 North Zhongshan Road, Shanghai, 200062, PR China.
| |
Collapse
|
50
|
Moreira V, Leiguez E, Janovits PM, Maia-Marques R, Fernandes CM, Teixeira C. Inflammatory Effects of Bothrops Phospholipases A 2: Mechanisms Involved in Biosynthesis of Lipid Mediators and Lipid Accumulation. Toxins (Basel) 2021; 13:toxins13120868. [PMID: 34941706 PMCID: PMC8709003 DOI: 10.3390/toxins13120868] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/19/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
Phospholipases A2s (PLA2s) constitute one of the major protein groups present in the venoms of viperid and crotalid snakes. Snake venom PLA2s (svPLA2s) exhibit a remarkable functional diversity, as they have been described to induce a myriad of toxic effects. Local inflammation is an important characteristic of snakebite envenomation inflicted by viperid and crotalid species and diverse svPLA2s have been studied for their proinflammatory properties. Moreover, based on their molecular, structural, and functional properties, the viperid svPLA2s are classified into the group IIA secreted PLA2s, which encompasses mammalian inflammatory sPLA2s. Thus, research on svPLA2s has attained paramount importance for better understanding the role of this class of enzymes in snake envenomation and the participation of GIIA sPLA2s in pathophysiological conditions and for the development of new therapeutic agents. In this review, we highlight studies that have identified the inflammatory activities of svPLA2s, in particular, those from Bothrops genus snakes, which are major medically important snakes in Latin America, and we describe recent advances in our collective understanding of the mechanisms underlying their inflammatory effects. We also discuss studies that dissect the action of these venom enzymes in inflammatory cells focusing on molecular mechanisms and signaling pathways involved in the biosynthesis of lipid mediators and lipid accumulation in immunocompetent cells.
Collapse
Affiliation(s)
- Vanessa Moreira
- Departamento de Farmacologia, Escola Paulista de Medicina, Universidade Federal de Sao Paulo, Sao Paulo 04044-020, Brazil;
| | - Elbio Leiguez
- Laboratório de Farmacologia, Instituto Butantan, Sao Paulo 05503-900, Brazil; (E.L.); (P.M.J.); (R.M.-M.); (C.M.F.)
| | - Priscila Motta Janovits
- Laboratório de Farmacologia, Instituto Butantan, Sao Paulo 05503-900, Brazil; (E.L.); (P.M.J.); (R.M.-M.); (C.M.F.)
| | - Rodrigo Maia-Marques
- Laboratório de Farmacologia, Instituto Butantan, Sao Paulo 05503-900, Brazil; (E.L.); (P.M.J.); (R.M.-M.); (C.M.F.)
| | - Cristina Maria Fernandes
- Laboratório de Farmacologia, Instituto Butantan, Sao Paulo 05503-900, Brazil; (E.L.); (P.M.J.); (R.M.-M.); (C.M.F.)
| | - Catarina Teixeira
- Laboratório de Farmacologia, Instituto Butantan, Sao Paulo 05503-900, Brazil; (E.L.); (P.M.J.); (R.M.-M.); (C.M.F.)
- Correspondence:
| |
Collapse
|