1
|
Völkl M, Burgers LD, Zech TJ, Ciurus S, Dorovska S, Liu H, Zahler S, Fürst R. Homoharringtonine (omacetaxine mepesuccinate) limits the angiogenic capacity of endothelial cells and reorganises filamentous actin. Biomed Pharmacother 2025; 186:118025. [PMID: 40184838 DOI: 10.1016/j.biopha.2025.118025] [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: 11/11/2024] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025] Open
Abstract
Homoharringtonine (HHT), an alkaloid from the plant genus Cephalotaxus, disrupts the first elongation phase of protein synthesis by interacting with the 60S ribosomal subunit, making it effective in treating diseases such as myeloid leukaemia. Semi-synthetically produced as omacetaxine mepesuccinate, HHT has been approved in Europe and in the US for patients resistant to two or more tyrosine kinase inhibitors. Although recent studies assume an anti-angiogenic capacity, the actions of HHT have not yet been characterised in primary endothelial cells, the major cell type driving angiogenesis. Therefore, this study addresses this issue by investigating the anti-angiogenic effect of HHT ex vivo and in vitro. A concentration-dependent decrease in sprouting was observed in a mouse aortic ring assay and in spheroids generated from human umbilical vein endothelial cells (HUVECs). Other angiogenic key features such as migration, proliferation and tube formation were similarly decreased by HHT. Interestingly, we observed an accumulation of F-actin. Inhibition of the ROCK pathway restored the angiogenic effects. A specific inhibition of typical upstream or downstream proteins of the ROCK pathway like Rho, MLC-2 or LIMK only marginally restored the angiogenic capability. Further analyses revealed that the alteration of the actin network might relate to the p38 MAPK/HSP27 axis: A significant prolongation of p38 phosphorylation induced by HHT treatment resulted in a partial restoration of endothelial spheroid sprouting. This study demonstrates the anti-angiogenic capabilities of HHT in endothelial cells and opens a promising further research field for an already approved drug.
Collapse
Affiliation(s)
- Matthias Völkl
- Pharmaceutical Biology, Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Luisa D Burgers
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt, Germany
| | - Thomas Josef Zech
- Pharmaceutical Biology, Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Sarah Ciurus
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt, Germany
| | - Senta Dorovska
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University, Frankfurt, Germany
| | - Hong Liu
- Pharmaceutical Biology, Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Stefan Zahler
- Pharmaceutical Biology, Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Robert Fürst
- Pharmaceutical Biology, Department of Pharmacy - Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
2
|
Bankolé A, Srivastava A, Shihavuddin A, Tighanimine K, Faucourt M, Koka V, Weill S, Nemazanyy I, Nelson AJ, Stokes MP, Delgehyr N, Genovesio A, Meunier A, Fumagalli S, Pende M, Spassky N. mTOR controls ependymal cell differentiation by targeting the alternative cell cycle and centrosomal proteins. EMBO Rep 2025:10.1038/s44319-025-00460-2. [PMID: 40307619 DOI: 10.1038/s44319-025-00460-2] [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: 10/21/2024] [Revised: 03/18/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025] Open
Abstract
Ependymal cells are multiciliated glial cells lining the ventricles of the mammalian brain. Their differentiation from progenitor cells involves cell enlargement and progresses through centriole amplification phases and ciliogenesis. These phases are accompanied by the sharp up-regulation of mTOR Complex 1 activity (mTORC1), a master regulator of macromolecule biosynthesis and cell growth, whose function in ependymal cell differentiation is unknown. We demonstrate that mTORC1 inhibition by rapamycin preserves the progenitor pool by reinforcing quiescence and preventing alternative cell cycle progression for centriole amplification. Overexpressing E2F4 and MCIDAS circumvents mTORC1-regulated processes, enabling centriole amplification despite rapamycin, and enhancing mTORC1 activity through positive feedback. Acute rapamycin treatment in multicentriolar cells during the late phases of differentiation causes centriole regrouping, indicating a direct role of mTORC1 in centriole dynamics. By phosphoproteomic and phosphomutant analysis, we reveal that the mTORC1-mediated phosphorylation of GAS2L1, a centrosomal protein that links actin and microtubule cytoskeletons, participates in centriole disengagement. This multilayered and sequential control of ependymal development by mTORC1, from the progenitor pool to centriolar function, has implications for pathophysiological conditions like aging and hydrocephalus-prone genetic diseases.
Collapse
Affiliation(s)
- Alexia Bankolé
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Ayush Srivastava
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Asm Shihavuddin
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Computational bioimaging and bioinformatics, 75005, Paris, France
- Department of EEE, Presidency University, Dhaka, Bangladesh
| | - Khaled Tighanimine
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Marion Faucourt
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Vonda Koka
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Solene Weill
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Alissa J Nelson
- Cell Signaling Technology INC, 3 Trask Lane, Danvers, MA, 01923, USA
| | - Matthew P Stokes
- Cell Signaling Technology INC, 3 Trask Lane, Danvers, MA, 01923, USA
| | - Nathalie Delgehyr
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Auguste Genovesio
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Computational bioimaging and bioinformatics, 75005, Paris, France
| | - Alice Meunier
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France
| | - Stefano Fumagalli
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France
| | - Mario Pende
- Université Paris Cité, CNRS, INSERM, Institut Necker Enfants Malades-INEM, F-75015, Paris, France.
| | - Nathalie Spassky
- Institut de Biologie de l'Ecole Normale Superieure (IBENS), Ecole Normale Supérieure, CNRS, INSERM, Université PSL, Team Cilia Biology and neurogenesis, 75005, Paris, France.
| |
Collapse
|
3
|
Mekata M, Yoshida K, Takai A, Hiroshima Y, Ikuta A, Seyama M, Yoshida K, Ozaki K. Porphyromonas gingivalis outer membrane vesicles increase vascular permeability by inducing stress fiber formation and degrading vascular endothelial-cadherin in endothelial cells. FEBS J 2025; 292:1696-1709. [PMID: 39690116 PMCID: PMC11970716 DOI: 10.1111/febs.17349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 07/24/2024] [Accepted: 09/13/2024] [Indexed: 12/19/2024]
Abstract
Porphyromonas gingivalis (Pg) is a keystone bacterium associated with systemic diseases, such as diabetes mellitus and Alzheimer's disease. Outer membrane vesicles (OMVs) released from Pg have been implicated in systemic diseases by delivering Pg virulence factors to host cells in distant organs and inducing cellular dysfunction. Pg OMVs also have the potential to enter distant organs via the bloodstream. However, the effects of Pg OMVs on the vascular function are poorly understood. Here, we showed that Pg OMVs increase vascular permeability by promoting stress fiber formation and lysosome/endosome-mediated vascular endothelial-cadherin (VEc) degradation in human umbilical vein endothelial cells (HUVECs) and human pulmonary microvascular endothelial cells (HPMECs). F-actin, visualized via fluorescein isothiocyanate-phalloidin, became thicker and longer, leading to the formation of radical stress fibers in response to Pg OMVs in HUVECs and HPMECs. Western blotting and quantitative real-time polymerase chain reaction analyses revealed that Pg OMVs decreased VEc protein levels in a gene-independent manner. Pg OMVs enhanced vesicular VEc accumulation in the cytoplasm around lysosome-associated membrane protein 1-positive structures during pretreatment with the lysosomal inhibitor chloroquine. This suggests that Pg OMVs decrease VEc protein levels by accelerating their internalization and degradation via lysosomes and endosomes. A27632 inhibition of Rho kinases impaired the Pg OMV-induced stress fiber formation and VEc degradation, resulting in the recovery of hyperpermeability. These findings provide new insights into the pathogenesis of systemic diseases that are associated with periodontal diseases.
Collapse
Affiliation(s)
- Mana Mekata
- Department of Oral Healthcare Promotion, Graduate School of Biomedical SciencesTokushima UniversityJapan
| | - Kaya Yoshida
- Department of Oral Healthcare Promotion, Graduate School of Biomedical SciencesTokushima UniversityJapan
| | - Ayu Takai
- Department of Oral Healthcare Promotion, Graduate School of Biomedical SciencesTokushima UniversityJapan
| | - Yuka Hiroshima
- Department of Oral Microbiology, Graduate School of Biomedical SciencesTokushima UniversityJapan
| | - Ayu Ikuta
- Department of Oral Healthcare Promotion, Graduate School of Biomedical SciencesTokushima UniversityJapan
| | - Mariko Seyama
- Department of Oral Healthcare Promotion, Graduate School of Biomedical SciencesTokushima UniversityJapan
| | - Kayo Yoshida
- Department of Oral Healthcare Promotion, Graduate School of Biomedical SciencesTokushima UniversityJapan
| | - Kazumi Ozaki
- Department of Oral Healthcare Promotion, Graduate School of Biomedical SciencesTokushima UniversityJapan
| |
Collapse
|
4
|
Hemavathy N, Umashankar V, Jeyakanthan J. Unveiling novel type 1 inhibitors for targeting LIM kinase 2 (LIMK2) for cancer therapeutics: An integrative pharmacoinformatics approach. Comput Biol Chem 2025; 115:108289. [PMID: 39631222 DOI: 10.1016/j.compbiolchem.2024.108289] [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: 06/07/2024] [Revised: 10/18/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024]
Abstract
LIMK2 is crucial in regulating actin cytoskeleton dynamics, significantly contributing to cancer cell proliferation, invasion, and metastasis. Inhibitors like LIMKi3 effectively suppress LIMK2 kinase activity by directly affecting actin polymerization and preventing the formation of structures like filopodia and lamellipodia, which are typical of motile cancer cells. By modulating these actin dynamics, LIMKi3 inhibits cancer cell migration and invasion, reducing the potential for metastasis. Thus, this study aims to explore potential anti-cancer therapeutic LIMK2 inhibitors with properties resembling LIMKi3. Henceforth, molecular docking was utilized in this study to comprehend the ATP mimetic binding mode of LIMKi3, followed by Pharmacophore-based virtual screening to identify small molecules resembling LIMKi3. In addition, molecular dynamics simulations were performed to explore the dynamic behavior of LIMK2 and potential inhibitors. Further, network analysis and binding free energy calculations were implemented to comprehensively assess the interactions between the compounds and LIMK2. In molecular docking, LIMKi3 demonstrated an ATP mimetic hinge binding mode with hydrogen bonds at Ile408. Among the screened compounds (NCI300395, ChemDiv-8020-2508, and ChemDiv-7997-0024), three displayed "ADRH" pharmacophoric features like LIMKi3, with favorable ADMET properties, higher binding affinity, and significant hydrogen bond interactions at Ile408. LIMK2-inhibitor complexes showed lower RMSD than LIMK2-LIMKi3, indicating higher equilibrium by identified compounds. Protein-drug Complexes exhibited significant inter-domain correlation in N-lobe residues of LIMK2, including conserved β3, αC, and Hinge residues. Binding free energy analysis ranked LIMK2-NCI300395 highest, followed by LIMK2-ChemDiv-7997-0024 and LIMK2-ChemDiv-8020-2508, highlighting their potential as effective LIMK2-targeting compounds. Hence, this study emphasizes LIMKi3's significance and identifies potential candidates (NCI300395, ChemDiv-7997-0024, and ChemDiv-8020-2508) for developing cancer therapeutics targeting LIMK2. These findings open avenues for further investigations into the complex interplay between cytoskeletal dynamics and cancer progression.
Collapse
Affiliation(s)
- Nagarajan Hemavathy
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu 630 003, India
| | - Vetrivel Umashankar
- Virology & Biotechnology/Bioinformatics Division, ICMR-National Institute for Research in Tuberculosis, Chennai, Tamil Nadu 600 031, India
| | - Jeyaraman Jeyakanthan
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, Tamil Nadu 630 003, India.
| |
Collapse
|
5
|
Metkari AS, Witt RL, Cognetti DM, Dhong C, Jia X. Promoting Polarization and Differentiation of Primary Human Salivary Gland Stem/Progenitor Cells in Protease-Degradable Hydrogels via ROCK Inhibition. ACS APPLIED MATERIALS & INTERFACES 2025; 17:18083-18095. [PMID: 40095914 DOI: 10.1021/acsami.4c22507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Toward the goal of in vitro engineering of functional salivary gland tissues, we cultured primary human salivary stem/progenitor cells (hS/PCs) in hyaluronic acid-based matrices with varying percentages of proteolytically degradable crosslinks in the presence of Rho kinase (ROCK) inhibitor. Single cells encapsulated in the hydrogel grew into organized multicellular structures by day 15, and over 60% of the structures developed in the nondegradable and 50% degradable hydrogels contained a central lumen. Importantly, ROCK inhibition led to the establishment of multicellular structures that were correctly polarized, as evidenced by apical localization of a Golgi marker GM130, apical/lateral localization of tight junction protein zonula occludens-1 (ZO-1), and basal localization of integrin β1 and basement membrane proteins laminin α1 and collagen IV. Cultures maintained in 50% degradable gels with ROCK inhibition exhibited an increased expression of acinar markers aquaporin 5 (AQP5, AQP5) and sodium-potassium-chloride cotransporter 1 (SLC12A2, NKCC1) at the transcript and the protein levels, respectively, as compared to those without ROCK inhibition. Upon stimulation with isoproterenol, α-amylase secretion into the lumen was observed. Particle-tracking microrheology was employed to analyze the stiffness of cells using mitochondria as the passive tracer particles. Our results indicated that cells grown in 100% degradable gels were stiffer than those maintained in nondegradable gels, and cells cultured with the ROCK inhibitor were softer than those maintained without the inhibitor. We conclude that reducing cellular contractility via ROCK inhibition while retaining some degree of matrix confinement promotes the establishment of multicellular structures containing pro-acinar cells with correct apicobasal polarization.
Collapse
Affiliation(s)
- Apoorva S Metkari
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Robert L Witt
- Helen F. Graham Cancer Center and Research Institute, Christiana Care, Newark, Delaware 19713, United States
- Department of Otolaryngology, Thomas Jefferson University, Philadelphia, Pennsylvania 19130, United States
| | - David M Cognetti
- Department of Otolaryngology, Thomas Jefferson University, Philadelphia, Pennsylvania 19130, United States
| | - Charles Dhong
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
| | - Xinqiao Jia
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware 19716, United States
- Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, United States
- Delaware Biotechnology Institute, 590 Avenue 1743, Newark, Delaware 19713, United States
| |
Collapse
|
6
|
Jacobs T, Isasti Sanchez J, Reger S, Luschnig S. Rho/Rok-dependent regulation of actomyosin contractility at tricellular junctions restricts epithelial permeability in Drosophila. Curr Biol 2025; 35:1181-1196.e5. [PMID: 39965573 DOI: 10.1016/j.cub.2025.01.043] [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: 10/03/2024] [Revised: 01/02/2025] [Accepted: 01/21/2025] [Indexed: 02/20/2025]
Abstract
Cell contacts in epithelia are remodeled to regulate paracellular permeability and to control the passage of migrating cells, but how barrier function is modulated while preserving epithelial integrity is not clear. In the follicular epithelium of Drosophila ovaries, tricellular junctions (TCJs) open transiently in a process termed patency to allow passage of externally produced yolk proteins for uptake by the oocyte. Here, we show that modulation of actomyosin contractility at cell vertices controls TCJ permeability. Before patency, circumferential actomyosin bundles are anchored at apical follicle cell vertices, where tension-sensing junctional proteins, Rho-associated kinase (Rok), and active myosin II accumulate and maintain vertices closed. TCJ opening is initiated by redistribution of myosin II from circumferential bundles to the medial zone, accompanied by decreasing tension on vertices. This transition requires activation of Cofilin-dependent filamentous actin (F-actin) disassembly by the phosphatase Slingshot and myosin II inactivation by myosin light-chain phosphatase and is counteracted by Rok. Accordingly, constitutive activation of myosin or of Rho signaling prevents vertex opening, whereas reduced myosin II or Rok activity causes excessive vertex opening. Thus, the opening of intercellular gaps in the follicular epithelium relies on relaxation of actomyosin contractility rather than active actomyosin-based pulling forces. Conversely, F-actin assembly is required for closing intercellular gaps after patency. Our findings are consistent with a force transduction model in which TCJ integrity is maintained by vertex-anchored contractile actomyosin. We propose that the cell-type-specific organization of actomyosin at cell vertices determines the mode of contractility-dependent regulation of epithelial permeability.
Collapse
Affiliation(s)
- Thea Jacobs
- Institute of Integrative Cell Biology and Physiology, Cells in Motion (CiM) Interfaculty Center, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany
| | - Jone Isasti Sanchez
- Institute of Integrative Cell Biology and Physiology, Cells in Motion (CiM) Interfaculty Center, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany
| | - Steven Reger
- Institute of Integrative Cell Biology and Physiology, Cells in Motion (CiM) Interfaculty Center, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany
| | - Stefan Luschnig
- Institute of Integrative Cell Biology and Physiology, Cells in Motion (CiM) Interfaculty Center, University of Münster, Röntgenstrasse 16, 48149 Münster, Germany.
| |
Collapse
|
7
|
Roets B, Abrahamse H, Crous A. Biomaterial Properties and Differentiation Strategies for Tenogenic Differentiation of Mesenchymal Stem Cells. Cells 2025; 14:452. [PMID: 40136701 PMCID: PMC11940850 DOI: 10.3390/cells14060452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
Tendinopathy is a prevalent musculoskeletal condition that affects both aging populations and individuals involved in repetitive, high-intensity activities, such as athletes. Current treatment options primarily address symptom management or involve surgery, which carries a significant risk of complications and re-injury. This highlights the need for regenerative medicine approaches that combine stem cells, biomaterials, and growth factors. However, achieving effective tenogenic differentiation remains challenging due to the absence of standardized differentiation protocols. Consequently, a review of existing research has been conducted to identify optimal biomaterial properties and growth factor protocols. Findings suggest that the ideal biomaterial for tenogenic differentiation should feature a 3D structure to preserve tenogenic expression, incorporate a combination of aligned micro- and nanofibers to promote differentiation, and require further investigation into optimal stiffness. Additionally, growth factor protocols should include an induction phase to initiate tenogenic lineage commitment, followed by a maintenance phase to support matrix production and maturation.
Collapse
Affiliation(s)
| | | | - Anine Crous
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, P.O. Box 17011, Johannesburg 2028, South Africa; (B.R.); (H.A.)
| |
Collapse
|
8
|
Li Y, Fan C, Jiang F, Zhang J, Li Y, Jiang Y, Zhang R, Yu Z, Wang S. Identification of LIMK1 as a biomarker in clear cell renal cell carcinoma: from data mining to validation. J Cancer Res Clin Oncol 2025; 151:104. [PMID: 40056237 PMCID: PMC11890329 DOI: 10.1007/s00432-025-06146-5] [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: 10/24/2024] [Accepted: 02/20/2025] [Indexed: 03/10/2025]
Abstract
PURPOSE Clear cell renal cell carcinoma (ccRCC) is one of the most common types of renal cancer. LIM kinase 1 (LIMK1) reportedly plays an important role in tumorigenesis. However, the involvement of LIMK1 in the progression of ccRCC remains ambiguous. METHODS Based on the TCGA and CPTAC databases, the expression of LIMK1 in ccRCC was evaluated. In the TCGA-ccRCC cohort, the relationships between LIMK1 and immune cell infiltration as well as immune checkpoints were assessed. The high expression of LIMK1 in ccRCC was verified by qRT-PCR in four RCC cell lines. Immunohistochemistry was used to evaluate the expression of LIMK1 in clinical samples. The association between LIMK1 expression and survival prognosis was explored via Kaplan-Meier survival curve in the TCGA-ccRCC and local cohorts. The effects of LIMK1 knockdown on the proliferation, migration, and invasion abilities of RCC cells were evaluated via colony, CCK-8, wound healing, and Transwell assays. RESULTS Elevated expression level of LIMK1 was found in the TCGA-ccRCC cohort and was confirmed in RCC cell lines and clinical samples. Up-regulation of LIMK1 was found to be correlated with poor prognosis in TCGA-ccRCC and external cohorts. In addition, high-LIMK1 was associated with clinicopathological stage, immune cell infiltration and immune checkpoint in ccRCC. Importantly, knockdown of LIMK1 diminished the capability of proliferation, migration, and invasion in RCC cells. CONCLUSION LIMK1 may serve as a promising diagnostic and prognostic biomarker of ccRCC.
Collapse
Affiliation(s)
- Yifei Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Congcong Fan
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Feng Jiang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jingnan Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yanzhen Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yanjie Jiang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Rui Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhixian Yu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Siqi Wang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
9
|
Rangaswamy R, Hemavathy N, Subramaniyan S, Vetrivel U, Jeyakanthan J. Harnessing allosteric inhibition: prioritizing LIMK2 inhibitors for targeted cancer therapy through pharmacophore-based virtual screening and essential molecular dynamics. J Biomol Struct Dyn 2025; 43:1129-1146. [PMID: 38063080 DOI: 10.1080/07391102.2023.2291171] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/21/2023] [Indexed: 01/16/2025]
Abstract
The therapeutic potential of small molecule kinase inhibitors in cancer treatment is well recognized. However, achieving selectivity remains a formidable challenge, primarily due to the structural similarity of ATP binding pockets among kinases. Allosteric inhibition, which involves targeting binding pockets beyond the ATP-binding site, provides a promising alternative to overcome this challenge. In this study, a meticulous approach was implemented to prioritize type 3 inhibitors for LIMK2, employing a range of techniques including Molecular Dynamics (MD) simulations, e-pharmacophore-guided High Throughput Virtual Screening (HTVS), MM/GBSA and ADMETox analyses, Density Functional Theory (DFT) calculations, and MM/PBSA investigations. The e-pharmacophore model identifies a hypothesis featuring five essential pharmacophoric elements (RRRAH). Through virtual screening of the ZINC compound database, we identified only five compounds that align with all four pharmacophoric features: ZINC1044382792, ZINC1433610865, ZINC1044109145, ZINC952869440, and ZINC490621334. These compounds not only exhibit higher binding affinity but also demonstrate favorable ADME/Tox profiles. Molecular dynamics simulations underscore the stability of hydrogen bond interactions with critical cryptic LIMK2 pocket residues, Asp469 and Arg474, only for two compounds: ZINC143361086 and ZINC1044382792. These compounds also exhibit superior occupancy interactions, as indicated by HOMO-LUMO analysis. Additionally, binding free energy calculations highlight the significant affinities of these two compounds when complexed with LIMK2: -83.491 ± 1.230 kJ/mol and -90.122 ± 1.248 kJ/mol for ZINC1044382792 and ZINC1433610862, respectively. Hence, this comprehensive investigation identifies ZINC1433610862 and ZINC1044382792 as prospective hits, representing promising leads for targeting LIMK2 in cancer therapeutics.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Raghu Rangaswamy
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Nagarajan Hemavathy
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Sneha Subramaniyan
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| | - Umashankar Vetrivel
- Virology & Biotechnology/Bioinformatics Division, ICMR-National Institute for Research in Tuberculosis, Chennai, Tamil Nadu, India
| | - Jeyaraman Jeyakanthan
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Alagappa University, Karaikudi, Tamil Nadu, India
| |
Collapse
|
10
|
Teng H, Huang H, Lin C, Twu Y, Yang W, Lin W, Lan H, Lin Y, Hwang W. CT45A1-mediated MLC2 (MYL9) phosphorylation promotes natural killer cell resistance and outer cell fate in a cell-in-cell structure, potentiating the progression of microsatellite instability-high colorectal cancer. Mol Oncol 2025; 19:430-451. [PMID: 39322998 PMCID: PMC11793002 DOI: 10.1002/1878-0261.13736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 07/15/2024] [Accepted: 09/10/2024] [Indexed: 09/27/2024] Open
Abstract
Patients with microsatellite instability-high (MSI-H) colorectal cancer (CRC) have high tumor mutation burden and tumor immunogenicity, exhibiting a higher response rate to immunotherapy and better survival. However, a portion of MSI-H CRC patients still experience adverse disease outcomes. We aimed to identify the tumor-autonomous regulators determining these heterogeneous clinical outcomes. The Cancer Genome Atlas (TCGA) dataset was used to identify regulators in MSI-H CRC patients with unfavorable outcomes. Stable CRC tumor clones expressing targeted regulators were established to evaluate migratory and stemness properties, immune cell vulnerability, and cell-in-cell (CIC) structure formation. RNA-sequencing (RNA-seq) was used to identify enriched biological pathways in stable CRC tumor clones. Clinicopathological characterization of formalin-fixed paraffin-embedded (FFPE) MSI-H CRC specimens was performed to explore the underlying mechanisms involved. We showed that cancer/testis antigen family 45 member A1 (CT45A1) expression was upregulated in MSI-H CRC patients with poor survival outcomes. CT45A1-expressing microsatellite stable (MSS) CRC cells showed enhanced migratory ability. However, CT45A1-expressing MSI-H CRC cells, but not MSS CRC cells, showed higher resistance to natural killer (NK) cell cytotoxicity and served as outer cells in homotypic CIC structures, preventing exogenous or therapeutic antibody access to inner CRC cells. Inactivating RHO-ROCK/MLCK-MLC2 signaling with small-molecule inhibitors or short-hairpin RNAs (shRNAs) targeting myosin light chain kinase (MYLK) abolished NK cell resistance and reduced the outer cell fate of CT45A1-expressing MSI-H CRC cells. In MSI-H CRC patients, CT45A1-positive tumors exhibited increased MLC2 phosphorylation, increased outer cell fate, and decreased survival. We demonstrated that CT45A1 potentiates the advanced progression of MSI-H CRC, and targeting MLC2 phosphorylation may enhance immunotherapy efficacy in CT45A1-positive MSI-H CRC patients.
Collapse
Affiliation(s)
- Hao‐Wei Teng
- Division of Medical Oncology, Department of OncologyTaipei Veterans General HospitalTaiwan
- School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Hsiang‐Yueh Huang
- Department of Biotechnology and Laboratory Science in MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Chun‐Chi Lin
- Division of Colon and Rectum Surgery, Department of SurgeryTaipei Veterans General HospitalTaiwan
- Department of Surgery, Faculty of Medicine, School of MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yuh‐Ching Twu
- Department of Biotechnology and Laboratory Science in MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Wen‐Hao Yang
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular MedicineChina Medical UniversityTaichungTaiwan
| | - Wen‐Chun Lin
- Department of Biotechnology and Laboratory Science in MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Hsin‐Yi Lan
- Department of Biotechnology and Laboratory Science in MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yen‐Yu Lin
- Department of Pathology, Fu Jen Catholic University HospitalFu Jen Catholic UniversityNew Taipei CityTaiwan
- School of Medicine, College of MedicineFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Wei‐Lun Hwang
- Department of Biotechnology and Laboratory Science in MedicineNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
- Cancer and Immunology Research CenterNational Yang Ming Chiao Tung UniversityTaipeiTaiwan
| |
Collapse
|
11
|
Baldwin AG, Foley DW, Collins R, Lee H, Jones DH, Wahab B, Waters L, Pedder J, Paine M, Feng GJ, Privitera L, Ashall-Kelly A, Thomas C, Gillespie JA, Schino L, Belelli D, Rocha C, Maussion G, Krahn AI, Durcan TM, Elkins JM, Lambert JJ, Atack JR, Ward SE. Discovery of MDI-114215: A Potent and Selective LIMK Inhibitor To Treat Fragile X Syndrome. J Med Chem 2025; 68:719-752. [PMID: 39711116 PMCID: PMC11726654 DOI: 10.1021/acs.jmedchem.4c02694] [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: 11/04/2024] [Revised: 11/29/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024]
Abstract
LIMKs are serine/threonine and tyrosine kinases responsible for controlling cytoskeletal dynamics as key regulators of actin stability, ensuring synaptic health through normal synaptic bouton structure and function. However, LIMK1 overactivation results in abnormal dendritic synaptic development that characterizes the pathogenesis of Fragile X Syndrome (FXS). As a result, the development of LIMK inhibitors represents an emerging disease-modifying therapeutic approach for FXS. We report the discovery of MDI-114215 (85), a novel, potent allosteric dual-LIMK1/2 inhibitor that demonstrates exquisite kinome selectivity. 85 reduces phospho-cofilin in mouse brain slices and rescues impaired hippocampal long-term potentiation in brain slices from FXS mice. We also show that LIMK inhibitors are effective in reducing phospho-cofilin levels in iPSC neurons derived from FXS patients, demonstrating 85 to be a potential therapeutic candidate for FXS that could have broad application to neurological disorders or cancers caused by LIMK1/2 overactivation and actin instability.
Collapse
Affiliation(s)
- Alex G. Baldwin
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - David W. Foley
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Ross Collins
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Hyunah Lee
- Centre
for Medicines Discovery, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - D. Heulyn Jones
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Ben Wahab
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Loren Waters
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Josephine Pedder
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Marie Paine
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Gui Jie Feng
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Lucia Privitera
- Division
of Neuroscience, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1
5HL, U.K.
| | - Alexander Ashall-Kelly
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Carys Thomas
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Jason A. Gillespie
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Lauramariú Schino
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Delia Belelli
- Division
of Neuroscience, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1
5HL, U.K.
| | - Cecilia Rocha
- The
Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology
and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Gilles Maussion
- The
Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology
and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Andrea I. Krahn
- The
Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology
and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Thomas M. Durcan
- The
Neuro’s Early Drug Discovery Unit (EDDU), Department of Neurology
and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, 3801 University Street, Montreal, Quebec H3A 2B4, Canada
| | - Jonathan M. Elkins
- Centre
for Medicines Discovery, University of Oxford, Roosevelt Drive, Oxford OX3 7DQ, U.K.
| | - Jeremy J. Lambert
- Division
of Neuroscience, School of Medicine, Medical Sciences Institute, Dundee University, Dow Street, Dundee DD1
5HL, U.K.
| | - John R. Atack
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| | - Simon E. Ward
- Medicines
Discovery Institute, School of Biosciences, Cardiff University, Main Building, Park Place, Cardiff CF10 3AT, U.K.
| |
Collapse
|
12
|
McNicol GR, Dalby MJ, Stewart PS. A theoretical model for focal adhesion and cytoskeleton formation in non-motile cells. J Theor Biol 2025; 596:111965. [PMID: 39442686 DOI: 10.1016/j.jtbi.2024.111965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 07/25/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
To function and survive cells need to be able to sense and respond to their local environment through mechanotransduction. Crucially, mechanical and biochemical perturbations initiate cell signalling cascades, which can induce responses such as growth, apoptosis, proliferation and differentiation. At the heart of this process are actomyosin stress fibres (SFs), which form part of the cell cytoskeleton, and focal adhesions (FAs), which bind this cytoskeleton to the extra-cellular matrix (ECM). The formation and maturation of these structures (connected by a positive feedback loop) is pivotal in non-motile cells, where SFs are generally of ventral type, interconnecting FAs and producing isometric tension. In this study we formulate a one-dimensional bio-chemo-mechanical continuum model to describe the coupled formation and maturation of ventral SFs and FAs. We use a set of reaction-diffusion-advection equations to describe three sets of biochemical events: the polymerisation of actin and subsequent bundling into activated SFs; the formation and maturation of cell-substrate adhesions; and the activation of signalling proteins in response to FA and SF formation. The evolution of these key proteins is coupled to a Kelvin-Voigt viscoelastic description of the cell cytoplasm and the ECM. We employ this model to understand how cells respond to external and intracellular cues in vitro and are able to reproduce experimentally observed phenomena including non-uniform cell striation and cells forming weaker SFs and FAs on softer substrates.
Collapse
Affiliation(s)
- Gordon R McNicol
- School of Mathematics and Statistics, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Matthew J Dalby
- Centre for the Cellular Microenvironment, Mazumdar-Shaw Advanced Research Centre, University of Glasgow, Glasgow, G11 6EW, United Kingdom
| | - Peter S Stewart
- School of Mathematics and Statistics, University of Glasgow, Glasgow, G12 8QQ, United Kingdom.
| |
Collapse
|
13
|
Beck C, Kunze A. Parallelized Mechanical Stimulation of Neuronal Calcium Through Cell-Internal Nanomagnetic Forces Provokes Lasting Shifts in the Network Activity State. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406678. [PMID: 39460486 PMCID: PMC11812431 DOI: 10.1002/smll.202406678] [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: 08/14/2024] [Revised: 10/14/2024] [Indexed: 10/28/2024]
Abstract
Neurons differentiate mechanical stimuli force and rate to elicit unique functional responses, driving the need for further tools to generate various mechanical stimuli. Here, cell-internal nanomagnetic forces (iNMF) are introduced by manipulating internalized magnetic nanoparticles with an external magnetic field across cortical neuron networks in vitro. Under iNMF, cortical neurons exhibit calcium (Ca2+) influx, leading to modulation of activity observed through Ca2+ event rates. Inhibiting particle uptake or altering nanoparticle exposure time reduced the neuronal response to nanomagnetic forces, exposing the requirement of nanoparticle uptake to induce the Ca2+ response. In highly active cortical networks, iNMF robustly modulates synchronous network activity, which is lasting and repeatable. Using pharmacological blockers, it is shown that iNMF activates mechanosensitive ion channels to induce the Ca2+ influx. Then, in contrast to transient mechanically evoked neuronal activity, iNMF activates Ca2+-activated potassium (KCa) channels to stabilize the neuronal membrane potential and induce network activity shifts. The findings reveal the potential of magnetic nanoparticle-mediated mechanical stimulation to modulate neuronal circuit dynamics, providing insights into the biophysics of neuronal computation.
Collapse
Affiliation(s)
- Connor Beck
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, USA
| | - Anja Kunze
- Department of Electrical and Computer Engineering, Montana State University, Bozeman, Montana 59717, USA
- Montana Nanotechnology Facility, Montana State University, Bozeman, Montana 59717, USA
- Optical Technology Center, Montana State University, Bozeman, Montana, 59717, USA
| |
Collapse
|
14
|
Metkari AS, Witt RL, Cognetti DM, Dhong C, Jia X. Promoting Polarization and Differentiation of Primary Human Salivary Gland Stem/Progenitor Cells in Protease-Degradable Hydrogels via ROCK Inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.24.625065. [PMID: 39651209 PMCID: PMC11623551 DOI: 10.1101/2024.11.24.625065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Towards the goal of in vitro engineering of functional salivary gland tissues, we cultured primary human salivary stem/progenitor cells (hS/PCs) in hyaluronic acid-based matrices with varying percentages of proteolytically degradable crosslinks in the presence of Rho kinase (ROCK) inhibitor. Single cells encapsulated in the hydrogel grew into organized multicellular structures by day 15, and over 60% of the structures developed in the non-degradable and 50% degradable hydrogels contained a central lumen. Importantly, ROCK inhibition led to the establishment of multicellular structures that were correctly polarized, as evidenced by apical localization of a Golgi marker GM130, apical/lateral localization of tight junction protein zonula occludens-1 (ZO-1), and basal localization of integrin β1 and basement membrane proteins laminin α1 and collagen IV. Cultures maintained in 50% degradable gels with ROCK inhibition exhibited an increased expression of acinar markers AQP5 and SLC12A2 (at the transcript level) and AQP5 and NKCC1 (at the protein level) as compared to those without ROCK inhibition. Upon stimulation with isoproterenol, α-amylase secretion into the lumen was observed. Particle-tracking microrheology was employed to analyze the stiffness of cells using mitochondria as the passive tracer particles. Our results indicated that cells grown in 100% degradable gels were stiffer than those maintained in non-degradable gels, and cells cultured with the ROCK inhibitor were softer than those maintained without the inhibitor. We conclude that reducing cellular contractility via ROCK inhibition while retaining some degree of matrix confinement promotes the establishment of multicellular structures containing pro-acinar cells with correct apicobasal polarization.
Collapse
|
15
|
Li B, Ling Z, Wang Y, Xing Y. Receptor-Interacting Protein Kinase 3 Augments Neuroinflammation by Facilitating Neutrophil Infiltration during an Ischemic Stroke. J Vasc Res 2024; 62:51-62. [PMID: 39571563 DOI: 10.1159/000542571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 10/22/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION Neutrophil infiltration is responsible for the neuroinflammation during an ischemic stroke. Here, we explored the role of receptor-interacting protein kinase 3 (RIP3) in neutrophil infiltration during an ischemic stroke. METHODS The rat middle cerebral artery occlusion (MCAO) model was utilized to identify pivotal proteins involved in neutrophil infiltration during an ischemic stroke. Neutrophils were isolated from the peripheral blood of mice, and a co-immunoprecipitation (co-IP) assay was performed to identify the proteins that interact with RIP3. RESULTS The rat MCAO model was successfully established. Myeloperoxidase (MPO) was significantly upregulated in the MCAO group, indicating the presence of neutrophil infiltration. RIP3 protein level exhibited a similar trend to MPO protein level, suggesting that neuroinflammation might be partly activated by RIP3 through the promotion of neutrophil infiltration. Co-IP and mass spectrometry analyses suggested that RIP3 facilitated neutrophil infiltration partly by affecting protein kinases (Rock1 and Prkaca) downstream of RIP3, and the interaction between RIP3 and Rock1 or Prkaca was validated by IF and co-IP assays. CONCLUSION In this study, it was observed that RIP3 affects neutrophil infiltration, a critical phenomenon associated with neuronal injury during ischemic stroke, partly by the modulation of downstream proteins such as Rock1 and Prkaca.
Collapse
Affiliation(s)
- Baiyu Li
- Department of Neurology Cadre Ward, Gansu Provincial Hospital, Lanzhou, China
| | - Zexia Ling
- Department of Gastroenterology Cadre Ward, Gansu Provincial Hospital, Lanzhou, China
| | - Yanyan Wang
- Department of Neurology Cadre Ward, Gansu Provincial Hospital, Lanzhou, China
| | - Yinhua Xing
- Department of Gastroenterology Cadre Ward, Gansu Provincial Hospital, Lanzhou, China
| |
Collapse
|
16
|
Samsami Y, Akhlaghipour I, Taghehchian N, Palizkaran Yazdi M, Farrokhi S, Rahimi HR, Moghbeli M. MicroRNA-382 as a tumor suppressor during tumor progression. Bioorg Med Chem Lett 2024; 113:129967. [PMID: 39293533 DOI: 10.1016/j.bmcl.2024.129967] [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: 06/12/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Despite the recent progresses in therapeutic and diagnostic methods, there is still a significantly high rate of mortality among cancer patients. One of the main reasons for the high mortality rate in cancer patients is late diagnosis, which leads to the failure of therapeutic strategies. Therefore, investigation of cancer biology can lead to the introduction of early diagnostic markers in these patients. MicroRNAs (miRNAs) play an important role in regulation of cellular processes associated with tumor progression. Due to the high stability of miRNAs in body fluids, these factors can be considered as the non-invasive tumor markers. Deregulation of miR-382 has been widely reported in different cancers. Therefore, in this review, we investigated the role of miR-382 during tumor development. It has shown that miR-382 has mainly a tumor suppressive, which inhibits the growth of tumor cells through the regulation of signaling pathways, RNA-binding proteins, and transcription factors. Therefore, miR-382 can be suggested as a diagnostic and therapeutic marker in cancer patients.
Collapse
Affiliation(s)
- Yalda Samsami
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negin Taghehchian
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Saba Farrokhi
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
17
|
Dhimmar B, Modi U, Parihar SS, Makwana P, Boldrini CL, Vasita R. Fabrication of micropatterned PCL-collagen nanofibrous scaffold for cellular confinement induced early osteogenesis. BIOMATERIALS ADVANCES 2024; 164:213991. [PMID: 39146607 DOI: 10.1016/j.bioadv.2024.213991] [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: 04/23/2024] [Revised: 07/06/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024]
Abstract
The intricate interaction of the scaffold's architecture/geometry and with the cells is essential for tissue engineering and regenerative medicine. Cells sense their surrounding dynamic cues such as biophysical, biomechanical, and biochemical, and respond to them differently. Numerous studies have recently explored and reported the effect of contact guidance by culturing various types of cells on different types of micropatterned substrates such as microgrooves, geometric (square and triangle) micropattern, microstrips, micropatterned nanofibers. Amongst all of these micropatterned polymeric substrates; electrospun nanofibers have been regarded as a suitable substrate as it mimics the native ECM architectures. Therefore, in the present study; stencil-assisted electrospun Grid-lined micropatterned PCL-Collagen nanofibers (GLMPCnfs) were fabricated and its influence on the alignment and differentiation of pre-osteoblast cells (MC3T3-E1) was investigated. The randomly orientated Non-patterned PCL-Collagen nanofibers (NPPCnfs) were used as control. The patterns were characterized for their geometrical features such as area and thickness of deposition using surface profiler and scanning electron microscopy. A 61 % decrease in the overall area of GLMPCnfs as compared to the stencil area demonstrated the potential of electrofocusing phenomenon in the process of patterning electrospun nanofibers into various micron-scale structures. The MC3T3-E1 cells were confined and aligned in the direction of GLMPCnfs as confirmed by a high cellular aspect ratio (AR = 5.41), lower cellular shape index (CSI = 0.243), and cytoskeletal reorganization assessed through the F-actin filament immunocytochemistry (ICC) imaging. The aligned cells along the GLMPCnfs exhibited elevated alkaline phosphatase activity and enhanced mineralization. Furthermore, the gene expression profiling revealed upregulation of key osteogenic markers, such as ALP, OCN, OPN, COL1A1, and osteocyte markers DMP1, and SOST. Consequently, the research highlights the impact of GLMPCnfs on the cellular behaviour that results to the pre-osteoblast differentiation and the potential for stimulant-free early osteogenesis. These results offer an extensive understanding and mechanistic insight into how scaffold topography can be modified to influence cellular responses for effective bone regeneration strategies.
Collapse
Affiliation(s)
- Bindiya Dhimmar
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Unnati Modi
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Shayan Singh Parihar
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Pooja Makwana
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India
| | - Chiara Liliana Boldrini
- Department of Materials Science and Solar Energy Research Center MIBSOLAR University of Milano-Biococca, and INSTM Milano-Biococca Research Unit Via Cozzi 55, I-20125 Milano, Italy
| | - Rajesh Vasita
- Biomaterials & Biomimetics Laboratory, School of Life Sciences, Central University of Gujarat, Gandhinagar 382030, Gujarat, India; Terasaki Institute of Biomedical Innovation, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Li Q, Chen X, Li X, Jiang X, Li X, Men X, Li Y, Chen S. Plexin-B2 Mediates Orthodontic Tension-Induced Osteogenesis via the RhoA/F-Actin/YAP Pathway. J Periodontal Res 2024. [PMID: 39485327 DOI: 10.1111/jre.13358] [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: 08/11/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 11/03/2024]
Abstract
AIMS This study aims to investigate the role of Plexin-B2 in tension-induced osteogenesis of periodontal ligament stem cells (PDLSCs) and its biomechanical mechanism. METHODS In vitro, cyclic tension simulated orthodontic forces to assess Plexin-B2 expression in PDLSCs. We then knocked out Plexin-B2 using lentivirus to explore its role in tension-induced osteogenesis. In vivo, we used nickel-titanium springs to establish orthodontic tooth movement (OTM) models in mice. Local periodontal Plexin-B2 expression was knocked down using adeno-associated viruses (AAVs) to study its influence on new bone formation under mechanical tension in OTM models. Molecular mechanisms were elucidated by manipulating Plexin-B2 and RhoA expression, assessing related proteins, and observing F-actin and Yes-associated protein (YAP) through immunofluorescence. RESULTS Plexin-B2 expression in PDLSCs increased under cyclic tension. Decrease of Plexin-B2 reduced the expression of osteogenic protein in PDLSCs and negatively affected new bone formation during OTM. RhoA expression and phosphorylation of ROCK2/LIMK2/Cofilin decreased in Plexin-B2 knockout PDLSCs but were reversed by RhoA overexpression. The level of F-actin decreased in Plexin-B2 knockout PDLSCs but increased after RhoA rescue. Nuclear YAP was reduced in Plexin-B2 knockout PDLSCs but increased after RhoA overexpression. CONCLUSIONS Plexin-B2 is involved in tension-induced osteogenesis. Mechanistically, the RhoA signaling pathway, the F-actin arrangement, and the nuclear translocation of YAP are involved in the mechanotransduction of Plexin-B2.
Collapse
Affiliation(s)
- Qiming Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyi Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyi Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaoge Jiang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xingjian Li
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinrui Men
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yan Li
- Department of Stomatology, Inner Mongolia Hulunbuir Yakeshi Municipal People's Hospital, Hulunbuir, China
| | - Song Chen
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
19
|
Koike S, Tachikawa M, Tsutsumi M, Okada T, Nemoto T, Keino-Masu K, Masu M. Actin dynamics switches two distinct modes of endosomal fusion in yolk sac visceral endoderm cells. eLife 2024; 13:RP95999. [PMID: 39441732 PMCID: PMC11498936 DOI: 10.7554/elife.95999] [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] [Indexed: 10/25/2024] Open
Abstract
Membranes undergo various patterns of deformation during vesicle fusion, but how this membrane deformation is regulated and contributes to fusion remains unknown. In this study, we developed a new method of observing the fusion of individual late endosomes and lysosomes by using mouse yolk sac visceral endoderm cells that have huge endocytic vesicles. We found that there were two distinct fusion modes that were differently regulated. In homotypic fusion, two late endosomes fused quickly, whereas in heterotypic fusion they fused to lysosomes slowly. Mathematical modeling showed that vesicle size is a critical determinant of these fusion types and that membrane fluctuation forces can overcome the vesicle size effects. We found that actin filaments were bound to late endosomes and forces derived from dynamic actin remodeling were necessary for quick fusion during homotypic fusion. Furthermore, cofilin played a role in endocytic fusion by regulating actin turnover. These data suggest that actin promotes vesicle fusion for efficient membrane trafficking in visceral endoderm cells.
Collapse
Affiliation(s)
- Seiichi Koike
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
- Laboratory of Molecular and Cellular Biology, Graduate School of Science and Engineering for Research, University of ToyamaToyamaJapan
| | - Masashi Tachikawa
- Graduate School of Nanobioscience, Yokohama City UniversityYokohamaJapan
| | - Motosuke Tsutsumi
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural SciencesOkazakiJapan
- National Institute for Physiological Sciences, National Institutes of Natural SciencesOkazakiJapan
| | - Takuya Okada
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
| | - Tomomi Nemoto
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural SciencesOkazakiJapan
- National Institute for Physiological Sciences, National Institutes of Natural SciencesOkazakiJapan
| | - Kazuko Keino-Masu
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
| | - Masayuki Masu
- Graduate School of Comprehensive Human Sciences, University of TsukubaTsukubaJapan
- Department of Molecular Neurobiology, Institute of Medicine, University of TsukubaTsukubaJapan
| |
Collapse
|
20
|
Ning Y, Zheng M, Zhang Y, Jiao Y, Wang J, Zhang S. RhoA-ROCK2 signaling possesses complex pathophysiological functions in cancer progression and shows promising therapeutic potential. Cancer Cell Int 2024; 24:339. [PMID: 39402585 PMCID: PMC11475559 DOI: 10.1186/s12935-024-03519-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024] Open
Abstract
The Rho GTPase signaling pathway is responsible for cell-specific processes, including actin cytoskeleton organization, cell motility, cell division, and the transcription of specific genes. The implications of RhoA and the downstream effector ROCK2 in cancer epithelial-mesenchymal transition, migration, invasion, and therapy resistance associated with stem cells highlight the potential of targeting RhoA/ROCK2 signaling in therapy. Tumor relapse can occur due to cancer cells that do not fully respond to adjuvant chemoradiotherapy, targeted therapy, or immunotherapy. Rho signaling-mediated mitotic defects and cytokinesis failure lead to asymmetric cell division, allowing cells to form polyploids to escape cytotoxicity and promote tumor recurrence and metastasis. In this review, we elucidate the significance of RhoA/ROCK2 in the mechanisms of cancer progression and summarize their inhibitors that may improve treatment strategies.
Collapse
Affiliation(s)
- Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, P.R. China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China
| | - Yue Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Yuqi Jiao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Jiangping Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China.
| |
Collapse
|
21
|
Liu Y, Zhao C, Zhang R, Pang Y, Li L, Feng S. Progression of mesenchymal stem cell regulation on imbalanced microenvironment after spinal cord injury. Stem Cell Res Ther 2024; 15:343. [PMID: 39354635 PMCID: PMC11446099 DOI: 10.1186/s13287-024-03914-x] [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: 04/27/2024] [Accepted: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
Spinal cord injury (SCI) results in significant neural damage and inhibition of axonal regeneration due to an imbalanced microenvironment. Extensive evidence supports the efficacy of mesenchymal stem cell (MSC) transplantation as a therapeutic approach for SCI. This review aims to present an overview of MSC regulation on the imbalanced microenvironment following SCI, specifically focusing on inflammation, neurotrophy and axonal regeneration. The application, limitations and future prospects of MSC transplantation are discussed as well. Generally, a comprehensive perspective is provided for the clinical translation of MSC transplantation for SCI.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Chenxi Zhao
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Rong Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Yilin Pang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Linquan Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Shiqing Feng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China.
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China.
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
22
|
Beljkas M, Petkovic M, Vuletic A, Djuric A, Santibanez JF, Srdic-Rajic T, Nikolic K, Oljacic S. Development of Novel ROCK Inhibitors via 3D-QSAR and Molecular Docking Studies: A Framework for Multi-Target Drug Design. Pharmaceutics 2024; 16:1250. [PMID: 39458584 PMCID: PMC11514586 DOI: 10.3390/pharmaceutics16101250] [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: 08/30/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Alterations in the actin cytoskeleton correlates to tumor progression and affect critical cellular processes such as adhesion, migration and invasion. Rho-associated coiled-coil-containing protein kinases (ROCK1 and ROCK2), important regulators of the actin cytoskeleton, are frequently overexpressed in various malignancies. The aim of this study was therefore to identify the key structural features of ROCK1/ROCK2 inhibitors using computer-aided drug design (CADD) approaches. In addition, new developed ROCK inhibitors provided a significant framework for the development of multitarget therapeutics-ROCK/HDAC (histone deacetylases) multitarget inhibitors. Methods: 3D-QSAR (Quantitative structure-activity relationship study) and molecular docking study were employed in order to identify key structural features that positively correlate with ROCK inhibition. MDA-MB-231, HCC1937, Panc-1 and Mia PaCa-2 cells were used for evaluation of anticancer properties of synthesized compounds. Results: C-19 showed potent anti-cancer properties, especially enhancement of apoptosis and cell cycle modulation in pancreatic cancer cell lines. In addition, C-19 and C-22 showed potent anti-migratory and anti-invasive effects comparable to the well-known ROCK inhibitor fasudil. Conclusions: In light of the results of this study, we propose a novel multi-target approach focusing on developing dual HDAC/ROCK inhibitors based on the structure of both C-19 and C-22, exploiting the synergistic potential of these two signaling pathways to improve therapeutic efficacy in metastatic tumors. Our results emphasize the potential of multi-target ROCK inhibitors as a basis for future cancer therapies.
Collapse
Affiliation(s)
- Milan Beljkas
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (S.O.)
| | - Milos Petkovic
- Department of Organic Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia;
| | - Ana Vuletic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia; (A.V.); (A.D.)
| | - Ana Djuric
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia; (A.V.); (A.D.)
| | - Juan Francisco Santibanez
- Group for Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotica 4, 11129 Belgrade, Serbia;
| | - Tatjana Srdic-Rajic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia; (A.V.); (A.D.)
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (S.O.)
| | - Slavica Oljacic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (M.B.); (S.O.)
| |
Collapse
|
23
|
Sajib MS, Zahra FT, Lamprou M, Akwii RG, Park JH, Osorio M, Tullar P, Doci CL, Zhang C, Huveneers S, Van Buul JD, Wang MH, Markiewski MM, Srivastava SK, Zheng Y, Gutkind JS, Hu J, Bickel U, Maeda DY, Zebala JA, Lionakis MS, Trasti S, Mikelis CM. Tumor-induced endothelial RhoA activation mediates tumor cell transendothelial migration and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614304. [PMID: 39372784 PMCID: PMC11451620 DOI: 10.1101/2024.09.22.614304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The endothelial barrier plays an active role in transendothelial tumor cell migration during metastasis, however, the endothelial regulatory elements of this step remain obscure. Here we show that endothelial RhoA activation is a determining factor during this process. Breast tumor cell-induced endothelial RhoA activation is the combined outcome of paracrine IL-8-dependent and cell-to-cell contact β 1 integrin-mediated mechanisms, with elements of this pathway correlating with clinical data. Endothelial-specific RhoA blockade or in vivo deficiency inhibited the transendothelial migration and metastatic potential of human breast tumor and three murine syngeneic tumor cell lines, similar to the pharmacological blockade of the downstream RhoA pathway. These findings highlight endothelial RhoA as a potent, universal target in the tumor microenvironment for anti-metastatic treatment of solid tumors.
Collapse
|
24
|
Funahashi Y, Ahammad RU, Zhang X, Hossen E, Kawatani M, Nakamuta S, Yoshimi A, Wu M, Wang H, Wu M, Li X, Faruk MO, Shohag MH, Lin YH, Tsuboi D, Nishioka T, Kuroda K, Amano M, Noda Y, Yamada K, Sakimura K, Nagai T, Yamashita T, Uchino S, Kaibuchi K. Signal flow in the NMDA receptor-dependent phosphoproteome regulates postsynaptic plasticity for aversive learning. Sci Signal 2024; 17:eado9852. [PMID: 39255336 DOI: 10.1126/scisignal.ado9852] [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: 03/01/2024] [Accepted: 08/21/2024] [Indexed: 09/12/2024]
Abstract
Structural plasticity of dendritic spines in the nucleus accumbens (NAc) is crucial for learning from aversive experiences. Activation of NMDA receptors (NMDARs) stimulates Ca2+-dependent signaling that leads to changes in the actin cytoskeleton, mediated by the Rho family of GTPases, resulting in postsynaptic remodeling essential for learning. We investigated how phosphorylation events downstream of NMDAR activation drive the changes in synaptic morphology that underlie aversive learning. Large-scale phosphoproteomic analyses of protein kinase targets in mouse striatal/accumbal slices revealed that NMDAR activation resulted in the phosphorylation of 194 proteins, including RhoA regulators such as ARHGEF2 and ARHGAP21. Phosphorylation of ARHGEF2 by the Ca2+-dependent protein kinase CaMKII enhanced its RhoGEF activity, thereby activating RhoA and its downstream effector Rho-associated kinase (ROCK/Rho-kinase). Further phosphoproteomic analysis identified 221 ROCK targets, including the postsynaptic scaffolding protein SHANK3, which is crucial for its interaction with NMDARs and other postsynaptic scaffolding proteins. ROCK-mediated phosphorylation of SHANK3 in the NAc was essential for spine growth and aversive learning. These findings demonstrate that NMDAR activation initiates a phosphorylation cascade crucial for learning and memory.
Collapse
Affiliation(s)
- Yasuhiro Funahashi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Rijwan Uddin Ahammad
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Alzheimer's Therapeutic Research Institute, Keck School of Medicine of the University of Southern California, San Diego, CA 92121, USA
| | - Xinjian Zhang
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Emran Hossen
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Masahiro Kawatani
- Department of Physiology, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
| | - Shinichi Nakamuta
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Akira Yoshimi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
- Division of Clinical Sciences and Neuropsychopharmacology, Faculty and Graduate School of Pharmacy, Meijo University, Nagoya, Aichi 468-8503, Japan
| | - Minhua Wu
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Huanhuan Wang
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Mengya Wu
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Xu Li
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Md Omar Faruk
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Md Hasanuzzaman Shohag
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - You-Hsin Lin
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Daisuke Tsuboi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Tomoki Nishioka
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Keisuke Kuroda
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Mutsuki Amano
- Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Yukihiko Noda
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
- Division of Clinical Sciences and Neuropsychopharmacology, Faculty and Graduate School of Pharmacy, Meijo University, Nagoya, Aichi 468-8503, Japan
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Aichi 466-8550, Japan
| | - Kenji Sakimura
- Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Taku Nagai
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Takayuki Yamashita
- Department of Physiology, Fujita Health University School of Medicine, Toyoake, Aichi 470-1192, Japan
- Division of Neurophysiology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Shigeo Uchino
- Department of Biosciences, School of Science and Engineering, Teikyo University, Utsunomiya, Tochigi 320-8551, Japan
| | - Kozo Kaibuchi
- Division of Cell Biology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
- Center for Medical Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
25
|
Li WJ, Li RY, Wang DY, Shen M, Liu HL. CXCR3 participates in asymmetric division of mouse oocytes by modulating actin dynamics. Theriogenology 2024; 225:43-54. [PMID: 38788628 DOI: 10.1016/j.theriogenology.2024.05.028] [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: 12/29/2023] [Revised: 04/24/2024] [Accepted: 05/18/2024] [Indexed: 05/26/2024]
Abstract
Extensive research has been conducted on the role of CXCR3 in immune responses and inflammation. However, the role of CXCR3 in the reproductive system, particularly in oocyte development, remains unknown. In this study, we present findings on the involvement of CXCR3 in the meiotic division process of mouse oocytes. We found CXCR3 was expressed consistently throughout the entire maturation process of mouse oocyte. Inhibition of CXCR3 impaired the asymmetric division of oocyte, while the injection of Cxcr3 mRNA was capable of restoring these defects. Further study showed that inhibition of CXCR3 perturbed spindle migration by affecting LIMK/cofilin pathway-mediated actin remodeling. Knockout of CXCR3 led to an upregulation of actin-binding protein and an increased ATP level in GV-stage oocytes, while maintaining normal actin dynamics during the process of meiosis. Additionally, we noticed the expression level of DYNLT1 is markedly elevated in CXCR3-null oocytes. DYNLT1 bound with the Arp2/3 complex, and knockdown of DYNLT1 in CXCR3-null oocytes impaired the organization of cytoplasmic actin, suggesting the regulatory role of DYNLT1 in actin organization, and the compensatory expression of DYNLT1 may contribute to maintain normal actin dynamics in CXCR3-knockout oocytes. In summary, our findings provide insights into the intricate network of actin dynamics associated with CXCR3 during oocyte meiosis.
Collapse
Affiliation(s)
- Wei-Jian Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
| | - Rong-Yang Li
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| | - Da-Yu Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
| | - Ming Shen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
| | - Hong-Lin Liu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
26
|
Archakov AI, Vavilov NE, Zgoda VG. Detection of low-copy proteins in proteomic studies: issues and solutions. BIOMEDITSINSKAIA KHIMIIA 2024; 70:342-348. [PMID: 39324198 DOI: 10.18097/pbmc20247005342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Detection of low-copy proteins in complex biological samples is one of the most important issues of modern proteomics. The main reason for inefficient detection of low protein concentrations is the insufficient sensitivity of mass spectrometric detectors and the high dynamic range of protein concentrations. In this study we have investigated the possibilities and limitations of a targeted mass spectrometric analysis using the reconstructed system of standard proteins UPS1 (Universal Proteomic Standard 1) as an example. The study has shown that the sensitivity of the method is affected by the concentration of target proteins of the UPS1 system, as well as by a high level of biological noise modelled by proteins of whole E. coli cell lysate. The limitations of the method have been overcome by concentrating and pre-fractionating the sample peptides in a reversed phase chromatographic system under alkaline elution conditions. Proteomic analysis of the biological sample (proteins of the human hepatocellular carcinoma cell line HepG2 encoded by genes of human chromosome 18) showed an increase in the sensitivity of the method as compared to the standard targeted mass spectrometric analysis. This culminated in registration of 94 proteins encoded by genes located on human chromosome18.
Collapse
Affiliation(s)
- A I Archakov
- Institute of Biomedical Chemistry, Moscow, Russia
| | - N E Vavilov
- Institute of Biomedical Chemistry, Moscow, Russia
| | - V G Zgoda
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
27
|
Harrison A, Evans G, Blanco G. Expanding science skills: teaching tissue culture, data analysis, and reporting through imaging the actin cytoskeleton. JOURNAL OF MICROBIOLOGY & BIOLOGY EDUCATION 2024; 25:e0019023. [PMID: 38722163 PMCID: PMC11360407 DOI: 10.1128/jmbe.00190-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/13/2024] [Indexed: 08/30/2024]
Abstract
Within the eukaryotic cell, the actin cytoskeleton is a crucial structural framework that maintains cellular form, regulates cell movement and division, and facilitates the internal transportation of proteins and organelles. External cues induce alterations in the actin cytoskeleton primarily through the activation of Rho GTPases, which then bind to a diverse array of effector proteins to promote the local assembly or disassembly of actin. We have harnessed the extensively studied functions of RhoA in the dynamics of the actin cytoskeleton to craft a practical series for Stage 2 Biology students. This series not only imparts essential tissue culture laboratory skills but also reinforces them through repetition. These activities are presented in a scenario designed for students to explore the function of a hypothetical RhoA family member. Students produce slides from transfected cells, undertake fluorescence microscopy, process the images using ImageJ, and compile their findings in a comprehensive scientific report. The composition of the report requires independent acquisition of new knowledge and synoptic learning. According to student feedback, this early experience greatly aids in solidifying and honing the skills required to report on more extensive and intricate research projects, such as capstone projects.
Collapse
Affiliation(s)
- Adrian Harrison
- Department of Biology, University of York, York, United Kingdom
| | - Gareth Evans
- Department of Biology, University of York, York, United Kingdom
| | - Gonzalo Blanco
- Department of Biology, University of York, York, United Kingdom
| |
Collapse
|
28
|
Mercier AE, Joubert AM, Prudent R, Viallet J, Desroches-Castan A, De Koning L, Mabeta P, Helena J, Pepper MS, Lafanechère L. Sulfamoylated Estradiol Analogs Targeting the Actin and Microtubule Cytoskeletons Demonstrate Anti-Cancer Properties In Vitro and In Ovo. Cancers (Basel) 2024; 16:2941. [PMID: 39272798 PMCID: PMC11394244 DOI: 10.3390/cancers16172941] [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: 06/25/2024] [Revised: 07/30/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024] Open
Abstract
The microtubule-disrupting agent 2-methoxyestradiol (2-ME) displays anti-tumor and anti-angiogenic properties, but its clinical development is halted due to poor pharmacokinetics. We therefore designed two 2-ME analogs in silico-an ESE-15-one and an ESE-16 one-with improved pharmacological properties. We investigated the effects of these compounds on the cytoskeleton in vitro, and their anti-angiogenic and anti-metastatic properties in ovo. Time-lapse fluorescent microscopy revealed that sub-lethal doses of the compounds disrupted microtubule dynamics. Phalloidin fluorescent staining of treated cervical (HeLa), metastatic breast (MDA-MB-231) cancer, and human umbilical vein endothelial cells (HUVECs) displayed thickened, stabilized actin stress fibers after 2 h, which rearranged into a peripheral radial pattern by 24 h. Cofilin phosphorylation and phosphorylated ezrin/radixin/moesin complexes appeared to regulate this actin response. These signaling pathways overlap with anti-angiogenic, extra-cellular communication and adhesion pathways. Sub-lethal concentrations of the compounds retarded both cellular migration and invasion. Anti-angiogenic and extra-cellular matrix signaling was evident with TIMP2 and P-VEGF receptor-2 upregulation. ESE-15-one and ESE-16 exhibited anti-tumor and anti-metastatic properties in vivo, using the chick chorioallantoic membrane assay. In conclusion, the sulfamoylated 2-ME analogs displayed promising anti-tumor, anti-metastatic, and anti-angiogenic properties. Future studies will assess the compounds for myeloproliferative effects, as seen in clinical applications of other drugs in this class.
Collapse
Affiliation(s)
- Anne Elisabeth Mercier
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Anna Margaretha Joubert
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Renaud Prudent
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Jean Viallet
- Inovotion SAS France, Biopolis, 38700 La Tronche, France
| | - Agnes Desroches-Castan
- Laboratoire Biosanté U1292, Université Grenoble Alpes, Inserm, CEA, 38000 Grenoble, France
| | - Leanne De Koning
- Institut Curie Centre de Recherche, PSL Research University, 75248 Paris Cedex 05, France
| | - Peace Mabeta
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Jolene Helena
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Michael Sean Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology, and South African Medical Research Council Extramural Unit for Stem Cell Research and Therapy, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
| | - Laurence Lafanechère
- Department of Physiology, School of Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria 0028, South Africa
- Team Cytoskeleton Dynamics and Nuclear Functions, Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
| |
Collapse
|
29
|
Miyamoto S. Untangling the role of RhoA in the heart: protective effect and mechanism. Cell Death Dis 2024; 15:579. [PMID: 39122698 PMCID: PMC11315981 DOI: 10.1038/s41419-024-06928-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
RhoA (ras homolog family member A) is a small G-protein that transduces intracellular signaling to regulate a broad range of cellular functions such as cell growth, proliferation, migration, and survival. RhoA serves as a proximal downstream effector of numerous G protein-coupled receptors (GPCRs) and is also responsive to various stresses in the heart. Upon its activation, RhoA engages multiple downstream signaling pathways. Rho-associated coiled-coil-containing protein kinase (ROCK) is the first discovered and best characterized effector or RhoA, playing a major role in cytoskeletal arrangement. Many other RhoA effectors have been identified, including myocardin-related transcription factor A (MRTF-A), Yes-associated Protein (YAP) and phospholipase Cε (PLCε) to regulate transcriptional and post-transcriptional processes. The role of RhoA signaling in the heart has been increasingly studied in last decades. It was initially suggested that RhoA signaling pathway is maladaptive in the heart, but more recent studies using cardiac-specific expression or deletion of RhoA have revealed that RhoA activation provides cardioprotection against stress through various mechanisms including the novel role of RhoA in mitochondrial quality control. This review summarizes recent advances in understanding the role of RhoA in the heart and its signaling pathways to prevent progression of heart disease.
Collapse
Affiliation(s)
- Shigeki Miyamoto
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093-0636, USA.
| |
Collapse
|
30
|
McDuffie EL, Panettieri RA, Scott CP. G 12/13 signaling in asthma. Respir Res 2024; 25:295. [PMID: 39095798 PMCID: PMC11297630 DOI: 10.1186/s12931-024-02920-0] [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: 06/14/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
Shortening of airway smooth muscle and bronchoconstriction are pathognomonic for asthma. Airway shortening occurs through calcium-dependent activation of myosin light chain kinase, and RhoA-dependent calcium sensitization, which inhibits myosin light chain phosphatase. The mechanism through which pro-contractile stimuli activate calcium sensitization is poorly understood. Our review of the literature suggests that pro-contractile G protein coupled receptors likely signal through G12/13 to activate RhoA and mediate calcium sensitization. This hypothesis is consistent with the effects of pro-contractile agonists on RhoA and Rho kinase activation, actin polymerization and myosin light chain phosphorylation. Recognizing the likely role of G12/13 signaling in the pathophysiology of asthma rationalizes the effects of pro-contractile stimuli on airway hyperresponsiveness, immune activation and airway remodeling, and suggests new approaches for asthma treatment.
Collapse
Affiliation(s)
- Elizabeth L McDuffie
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Reynold A Panettieri
- Rutgers Institute for Translational Medicine and Science, Child Health Institute, Rutgers University, New Brunswick, NJ, USA
| | - Charles P Scott
- Department of Biochemistry and Molecular Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
31
|
Sidibé A, Mykuliak VV, Zhang P, Hytönen VP, Wu J, Wehrle-Haller B. Acetyl-NPKY of integrin-β1 binds KINDLIN2 to control endothelial cell proliferation and junctional integrity. iScience 2024; 27:110129. [PMID: 38904068 PMCID: PMC11187247 DOI: 10.1016/j.isci.2024.110129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 02/09/2024] [Accepted: 05/24/2024] [Indexed: 06/22/2024] Open
Abstract
Integrin-dependent crosstalk between cell-matrix adhesions and cell-cell junctions is critical for controlling endothelial permeability and proliferation in cancer and inflammatory diseases but remains poorly understood. Here, we investigated how acetylation of the distal NPKY-motif of Integrin-β1 influences endothelial cell physiology and barrier function. Expression of an acetylation-mimetic β1-K794Q-GFP mutant led to the accumulation of immature cell-matrix adhesions accompanied by a transcriptomic reprograming of endothelial cells, involving genes associated with cell adhesion, proliferation, polarity, and barrier function. β1-K794Q-GFP induced constitutive MAPK signaling, junctional impairment, proliferation, and reduced contact inhibition at confluence. Structural analysis of Integrin-β1 interaction with KINDLIN2, biochemical pulldown assay, and binding energy determination by using molecular dynamics simulation showed that acetylation of K794 and the K794Q-mutant increased KINDLIN2 binding affinity to the Integrin-β1. Thus, enhanced recruitment of KINDLIN2 to Lysine-acetylated Integrin-β1 and resulting modulation of barrier function, offers new therapeutic possibilities for controlling vascular permeability and disease conditions.
Collapse
Affiliation(s)
- Adama Sidibé
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| | - Vasyl V. Mykuliak
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Pingfeng Zhang
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
| | - Vesa P. Hytönen
- Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, FI-33520 Tampere, Finland
- Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Jinhua Wu
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA
| | - Bernhard Wehrle-Haller
- Department of Cell Physiology and Metabolism, University of Geneva, Centre Médical Universitaire, Rue Michel-Servet 1, CH-1211 Geneva, Switzerland
| |
Collapse
|
32
|
Nguyen TP, Otani T, Tsutsumi M, Kinoshita N, Fujiwara S, Nemoto T, Fujimori T, Furuse M. Tight junction membrane proteins regulate the mechanical resistance of the apical junctional complex. J Cell Biol 2024; 223:e202307104. [PMID: 38517380 PMCID: PMC10959758 DOI: 10.1083/jcb.202307104] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/29/2024] [Accepted: 02/16/2024] [Indexed: 03/23/2024] Open
Abstract
Epithelia must be able to resist mechanical force to preserve tissue integrity. While intercellular junctions are known to be important for the mechanical resistance of epithelia, the roles of tight junctions (TJs) remain to be established. We previously demonstrated that epithelial cells devoid of the TJ membrane proteins claudins and JAM-A completely lack TJs and exhibit focal breakages of their apical junctions. Here, we demonstrate that apical junctions fracture when claudin/JAM-A-deficient cells undergo spontaneous cell stretching. The junction fracture was accompanied by actin disorganization, and actin polymerization was required for apical junction integrity in the claudin/JAM-A-deficient cells. Further deletion of CAR resulted in the disruption of ZO-1 molecule ordering at cell junctions, accompanied by severe defects in apical junction integrity. These results demonstrate that TJ membrane proteins regulate the mechanical resistance of the apical junctional complex in epithelial cells.
Collapse
Affiliation(s)
- Thanh Phuong Nguyen
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Tetsuhisa Otani
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Japan Science and Technology Agency, Precursory Research for Embryonic Science and Technology, Kawaguchi, Japan
| | - Motosuke Tsutsumi
- Division of Biophotonics, National Institute for Physiological Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Noriyuki Kinoshita
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
- Basic Biology Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Sachiko Fujiwara
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Tomomi Nemoto
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Division of Biophotonics, National Institute for Physiological Sciences, Okazaki, Japan
- Biophotonics Research Group, Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, Okazaki, Japan
| | - Toshihiko Fujimori
- Division of Embryology, National Institute for Basic Biology, Okazaki, Japan
- Basic Biology Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, Japan
- Physiological Sciences Program, Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Japan
- Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
33
|
Yang C, Wang D. Antibiotic bone cement accelerates diabetic foot wound healing: Elucidating the role of ROCK1 protein expression. Int Wound J 2024; 21:e14590. [PMID: 38531354 PMCID: PMC10965272 DOI: 10.1111/iwj.14590] [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: 10/16/2023] [Accepted: 12/08/2023] [Indexed: 03/28/2024] Open
Abstract
Clinical studies indicate antibiotic bone cement with propeller flaps improves diabetic foot wound repair and reduces amputation rates, but the molecular mechanisms, particularly key proteins' role remain largely unexplored. This study assessed the efficacy of antibiotic bone cement for treating diabetic foot wounds, focusing on molecular impact on ROCK1. Sixty patients were randomized into experimental (EXP, n = 40) and control (CON, n = 20) groups, treated with antibiotic bone cement and negative pressure. Wound healing rate, amputation rate, wound secretion culture and C-reactive protein (CRP) changes, were monitored. Comprehensive molecular investigations were conducted and animal experiments were performed to further validate the findings. Statistical methods were employed to verify significant differences between the groups and treatment outcomes. The EXP group showed significant improvements in wound healing (χ 2 $$ {\chi}^2 $$ = 11.265, p = 0.004) and reduced amputation rates. Elevated levels of ROCK1, fibroblasts and VGF were observed in the trauma tissue post-treatment in the experimental group compared to pre-treatment and the control group (all p < 0.05). Improved trauma secretion culture and CRP were also noted in the EXP group (all p < 0.05). The study suggests that antibiotic bone cement enhances diabetic foot wound healing, possibly via upregulation of ROCK1. Further research is needed to elucidate the underlying molecular mechanisms and broader clinical implications.
Collapse
Affiliation(s)
- Chenglan Yang
- Soochow University School of MedicineSoochow UniversitySuzhouJiangsuChina
| | - Dali Wang
- Department of Burn Plastic SurgeryAffiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| |
Collapse
|
34
|
Liu J, Wu J. The Pathogenesis and Impact of Arterial Stiffening in Hypertension: The 2023 John H. Laragh Research Award. Am J Hypertens 2024; 37:241-247. [PMID: 38214376 PMCID: PMC11484606 DOI: 10.1093/ajh/hpae006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/13/2024] Open
Abstract
Fifty years ago, Dr. John Laragh brought forward the "vasoconstriction-volume hypothesis" of hypertension. This is Ohm's Law in blood pressure regulation, explicating hypertension as a consequence of increased peripheral vascular resistance, cardiac output, or both. Resistance vessels, those of a diameter less than 200 μm, determines mean arterial pressure by controlling peripheral vascular resistance. In comparison, large capacitance arteries, particularly the aorta, confines the systolic and diastolic blood pressure in physiological range through the "windkessel effect." Loss of this cushioning function results in aortic stiffening and isolated systolic hypertension, both of which are independently associated with increased risk for coronary, cerebral, and renal diseases. Aortic stiffening is both a cause and a consequence of hypertension. On one hand, aortic stiffness precedes the onset of hypertension in populations and experimental models, and hemodynamic derangements related to aortic stiffening contributes to the development of hypertension by promoting renal dysfunction. On the other hand, the vasculature itself is a hypertensive target organ and hypertensive mechanical stretch directly induces the pathogenesis of aortic adventitial remodeling. Various cell types, including bone marrow-derived circulating fibrocytes, vascular stem cell antigen-1 positive progenitors, and endothelial to mesenchymal transition, and to a lesser extent resident fibroblasts, contribute to adventitial matrix deposition and aortic stiffening in hypertension. Vascular smooth muscle stiffness is another important contributor of aortic stiffening. Understanding the roles of immune components and specific signal pathways in the pathogenesis aortic stiffening paves the path to novel antihypertensive and anti-fibrosis therapies.
Collapse
Affiliation(s)
- Jing Liu
- Division of Nephrology, Department of Medicine, School of Medicine & Dentistry, University of Rochester, Rochester, NY 14642, USA
| | - Jing Wu
- Division of Nephrology, Department of Medicine, School of Medicine & Dentistry, University of Rochester, Rochester, NY 14642, USA
- Department of Pharmacology & Physiology, School of Medicine & Dentistry, University of Rochester, Rochester, NY 14642, USA
- Environmental Health Science Center, Institute of Human Health and the Environment, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
35
|
Shi J, Wei L. ROCK1 deficiency preserves caveolar compartmentalization of signaling molecules and cell membrane integrity. FASEB Bioadv 2024; 6:85-102. [PMID: 38463696 PMCID: PMC10918988 DOI: 10.1096/fba.2024-00015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/05/2024] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
In this study, we investigated the roles of ROCK1 in regulating structural and functional features of caveolae located at the cell membrane of cardiomyocytes, adipocytes, and mouse embryonic fibroblasts (MEFs) as well as related physiopathological effects. Caveolae are small bulb-shaped cell membrane invaginations, and their roles have been associated with disease conditions. One of the unique features of caveolae is that they are physically linked to the actin cytoskeleton that is well known to be regulated by RhoA/ROCKs pathway. In cardiomyocytes, we observed that ROCK1 deficiency is coincident with an increased caveolar density, clusters, and caveolar proteins including caveolin-1 and -3. In the mouse cardiomyopathy model with transgenic overexpressing Gαq in myocardium, we demonstrated the reduced caveolar density at cell membrane and reduced caveolar protein contents. Interestingly, coexisting ROCK1 deficiency in cardiomyocytes can rescue these defects and preserve caveolar compartmentalization of β-adrenergic signaling molecules including β1-adrenergic receptor and type V/VI adenylyl cyclase. In cardiomyocytes and adipocytes, we detected that ROCK1 deficiency increased insulin signaling with increased insulin receptor activation in caveolae. In MEFs, we identified that ROCK1 deficiency increased caveolar and total levels of caveolin-1 and cell membrane repair ability after mechanical or chemical disruptions. Together, these results demonstrate that ROCK1 can regulate caveolae plasticity and multiple functions including compartmentalization of signaling molecules and cell membrane repair following membrane disruption by mechanical force and oxidative damage. These findings provide possible molecular insights into the beneficial effects of ROCK1 deletion/inhibition in cardiomyocytes, adipocytes, and MEFs under certain diseased conditions.
Collapse
Affiliation(s)
- Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of MedicineIndiana UniversityIndianapolisIndianaUSA
| | - Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, School of MedicineIndiana UniversityIndianapolisIndianaUSA
| |
Collapse
|
36
|
Yasuda Y, Wang L, Chitano P, Seow CY. Rho-Kinase Inhibition of Active Force and Passive Tension in Airway Smooth Muscle: A Strategy for Treating Airway Hyperresponsiveness in Asthma. BIOLOGY 2024; 13:115. [PMID: 38392332 PMCID: PMC10886476 DOI: 10.3390/biology13020115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/05/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
Rho-kinase inhibitors have been identified as a class of potential drugs for treating asthma because of their ability to reduce airway inflammation and active force in airway smooth muscle (ASM). Past research has revealed that, besides the effect on the ASM's force generation, rho-kinase (ROCK) also regulates actin filament formation and filament network architecture and integrity, thus affecting ASM's cytoskeletal stiffness. The present review is not a comprehensive examination of the roles played by ROCK in regulating ASM function but is specifically focused on passive tension, which is partially determined by the cytoskeletal stiffness of ASM. Understanding the molecular basis for maintaining active force and passive tension in ASM by ROCK will allow us to determine the suitability of ROCK inhibitors and its downstream enzymes as a class of drugs in treating airway hyperresponsiveness seen in asthma. Because clinical trials using ROCK inhibitors in the treatment of asthma have yet to be conducted, the present review focuses on the in vitro effects of ROCK inhibitors on ASM's mechanical properties which include active force generation, relaxation, and passive stiffness. The review provides justification for future clinical trials in the treatment of asthma using ROCK inhibitors alone and in combination with other pharmacological and mechanical interventions.
Collapse
Affiliation(s)
- Yuto Yasuda
- Centre for Heart Lung Innovation, St. Paul's Hospital, Providence Health Care, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Lu Wang
- Centre for Heart Lung Innovation, St. Paul's Hospital, Providence Health Care, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Pasquale Chitano
- Centre for Heart Lung Innovation, St. Paul's Hospital, Providence Health Care, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Chun Y Seow
- Centre for Heart Lung Innovation, St. Paul's Hospital, Providence Health Care, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
37
|
Ayata C, Kim H, Morrison L, Liao JK, Gutierrez J, Lopez-Toledano M, Carrazana E, Rabinowicz AL, Awad IA. Role of Rho-Associated Kinase in the Pathophysiology of Cerebral Cavernous Malformations. Neurol Genet 2024; 10:e200121. [PMID: 38179414 PMCID: PMC10766084 DOI: 10.1212/nxg.0000000000200121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 11/20/2023] [Indexed: 01/06/2024]
Abstract
Cerebral cavernous malformations (CCMs) are vascular lesions characterized by a porous endothelium. The lack of a sufficient endothelial barrier can result in microbleeds and frank intracerebral hemorrhage. A primary mechanism for lesion development is a sequence variant in at least 1 of the 3 CCM genes (CCM1, CCM2, and CCM3), which influence various signaling pathways that lead to the CCM phenotype. A common downstream process associated with CCM gene loss of function involves overactivation of RhoA and its effector Rho-associated kinase (ROCK). In this study, we review RhoA/ROCK-related mechanisms involved in CCM pathophysiology as potential therapeutic targets. Literature searches were conducted in PubMed using combinations of search terms related to RhoA/ROCK and CCMs. In endothelial cells, CCM1, CCM2, and CCM3 proteins normally associate to form the CCM protein complex, which regulates the functions of a wide variety of protein targets (e.g., MAP3K3, SMURF1, SOK-1, and ICAP-1) that directly or indirectly increase RhoA/ROCK activity. Loss of CCM complex function and increased RhoA/ROCK activity can lead to the formation of stress fibers that contribute to endothelial junction instability. Other RhoA/ROCK-mediated pathophysiologic outcomes include a shift to a senescence-associated secretory phenotype (primarily mediated by ROCK2), which is characterized by endothelial cell migration, cell cycle arrest, extracellular matrix degradation, leukocyte chemotaxis, and inflammation. ROCK represents a potential therapeutic target, and direct (fasudil, NRL-1049) and indirect (statins) ROCK inhibitors have demonstrated various levels of efficacy in reducing lesion burden in preclinical models of CCM. Current (atorvastatin) and planned (NRL-1049) clinical studies will determine the efficacy of ROCK inhibitors for CCM in humans, for which no US Food and Drug Administration-approved or EU-approved pharmacologic treatment exists.
Collapse
Affiliation(s)
- Cenk Ayata
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Helen Kim
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Leslie Morrison
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - James K Liao
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Juan Gutierrez
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Miguel Lopez-Toledano
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Enrique Carrazana
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Adrian L Rabinowicz
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| | - Issam A Awad
- From the Neurovascular Research Unit (C.A.), Department of Radiology; Stroke Service, Department of Neurology (C.A.), Massachusetts General Hospital, Harvard Medical School, Boston; Center for Cerebrovascular Research (H.K.), Department of Anesthesia and Perioperative Care, University of California, San Francisco; University of New Mexico Health Sciences Center (L.M.), Albuquerque; University of Arizona (J.K.L.), College of Medicine, Tucson; Neurelis, Inc. (J.G., M.L.-T., E.C., A.L.R.), San Diego, CA; University of Hawaii John A. Burns School of Medicine (E.C.), Honolulu, HI; and University of Chicago Medicine and Biological Sciences (I.A.A.), Chicago, IL
| |
Collapse
|
38
|
Stinson MW, Liu S, Laurenson AJ, Rotty JD. Macrophage migration is differentially regulated by fibronectin and laminin through altered adhesion and myosin II localization. Mol Biol Cell 2024; 35:ar22. [PMID: 38088893 PMCID: PMC10881148 DOI: 10.1091/mbc.e23-04-0137] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 10/30/2023] [Accepted: 11/28/2023] [Indexed: 12/26/2023] Open
Abstract
Macrophages are indispensable for proper immune surveillance and inflammatory regulation. They also exhibit dramatic phenotypic plasticity and are highly responsive to their local microenvironment, which includes the extracellular matrix (ECM). This work demonstrates that two fibrous ECM glycoproteins, fibronectin (FN) and laminin (LAM), elicit distinct morphological and migratory responses from macrophages in two-dimensional environments. LAM 111 inhibits macrophage cell spreading, but drives them to migrate rapidly and less persistently compared with cells on FN. Differential integrin engagement and ROCK/myosin II organization helps explain why macrophages alter their morphology and migration character on these two ECM components. This study also demonstrates that LAM 111 exerts a suppressive effect toward FN, as macrophages plated on a LAM/FN mixture adopt a morphology and migratory character almost identical to LAM alone. This suggests that distinct responses can be initiated downstream of receptor-ECM engagement, and that one component of the microenvironment may affect the cell's ability to sense another. Overall, macrophages appear intrinsically poised to rapidly switch between distinct migratory characters based on their ECM environments. The role of ECM composition in dictating motile and inflammatory responses in three-dimensional and in vivo contexts warrants further study.
Collapse
Affiliation(s)
- Matthew W. Stinson
- Uniformed Services University of the Health Sciences, Department of Biochemistry, Bethesda, MD 20814
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | - Sophia Liu
- Uniformed Services University of the Health Sciences, Department of Biochemistry, Bethesda, MD 20814
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | - Alexander J. Laurenson
- Uniformed Services University of the Health Sciences, Department of Biochemistry, Bethesda, MD 20814
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817
| | - Jeremy D. Rotty
- Uniformed Services University of the Health Sciences, Department of Biochemistry, Bethesda, MD 20814
| |
Collapse
|
39
|
Wan S, Wang X, Chen W, Xu Z, Zhao J, Huang W, Wang M, Zhang H. Polystyrene Nanoplastics Activate Autophagy and Suppress Trophoblast Cell Migration/Invasion and Migrasome Formation to Induce Miscarriage. ACS NANO 2024; 18:3733-3751. [PMID: 38252510 DOI: 10.1021/acsnano.3c11734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Nanoplastics (NPs), as emerging pollutants, have attracted global attention. Nevertheless, the adverse effects of NPs on female reproductive health, especially unexplained miscarriage, are poorly understood. Defects of trophoblast cell migration and invasion are associated with miscarriage. Migrasomes were identified as cellular organelles with largely unidentified functions. Whether NPs might affect migration, invasion, and migrasome formation and induce miscarriage has been completely unexplored. In this study, we selected polystyrene nanoplastics (PS-NPs, 50 nm) as a model of plastic particles and treated human trophoblast cells and pregnant mice with PS-NPs at doses near the actual environmental exposure doses of plastic particles in humans. We found that exposure to PS-NPs induced a pregnant mouse miscarriage. PS-NPs suppressed ROCK1-mediated migration/invasion and migrasome formation. SOX2 was identified as the transcription factor of ROCK1. PS-NPs activated autophagy and promoted the autophagy degradation of SOX2, thus suppressing SOX2-mediated ROCK1 transcription. Supplementing with murine SOX2 or ROCK1 could efficiently rescue migration/invasion and migrasome formation and alleviate miscarriage. Analysis of the protein levels of SOX2, ROCK1, TSPAN4, NDST1, P62, and LC-3BII/I in PS-NP-exposed trophoblast cells, villous tissues of unexplained miscarriage patients, and placental tissues of PS-NP-exposed mice gave consistent results. Collectively, this study revealed the reproductive toxicity of nanoplastics and their potential regulatory mechanism, indicating that NP exposure is a risk factor for female reproductive health.
Collapse
Affiliation(s)
- Shukun Wan
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoqing Wang
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Weina Chen
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
- Key Laboratory of Environment and Female Reproductive Health, West China School of Public Health & West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - Zhongyan Xu
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Jingsong Zhao
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Wenxin Huang
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Manli Wang
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| | - Huidong Zhang
- Research Center for Environment and Female Reproductive Health, the Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen 518033, China
| |
Collapse
|
40
|
Qian W, Yamaguchi N, Lis P, Cammer M, Knaut H. Pulses of RhoA signaling stimulate actin polymerization and flow in protrusions to drive collective cell migration. Curr Biol 2024; 34:245-259.e8. [PMID: 38096821 PMCID: PMC10872453 DOI: 10.1016/j.cub.2023.11.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 10/03/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023]
Abstract
In animals, cells often move as collectives to shape organs, close wounds, or-in the case of disease-metastasize. To accomplish this, cells need to generate force to propel themselves forward. The motility of singly migrating cells is driven largely by an interplay between Rho GTPase signaling and the actin network. Whether cells migrating as collectives use the same machinery for motility is unclear. Using the zebrafish posterior lateral line primordium as a model for collective cell migration, we find that active RhoA and myosin II cluster on the basal sides of the primordium cells and are required for primordium motility. Positive and negative feedbacks cause RhoA and myosin II activities to pulse. These pulses of RhoA signaling stimulate actin polymerization at the tip of the protrusions and myosin-II-dependent actin flow and protrusion retraction at the base of the protrusions and deform the basement membrane underneath the migrating primordium. This suggests that RhoA-induced actin flow on the basal sides of the cells constitutes the motor that pulls the primordium forward, a scenario that likely underlies collective migration in other contexts.
Collapse
Affiliation(s)
- Weiyi Qian
- Skirball Institute of Biomolecular Medicine and Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA.
| | - Naoya Yamaguchi
- Skirball Institute of Biomolecular Medicine and Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Patrycja Lis
- Skirball Institute of Biomolecular Medicine and Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Michael Cammer
- Microscopy Laboratory, Division of Advanced Research Technologies, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Holger Knaut
- Skirball Institute of Biomolecular Medicine and Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
41
|
Futterknecht S, Chatzimichail E, Gugleta K, Panos GD, Gatzioufas Z. The Role of Rho Kinase Inhibitors in Corneal Diseases. Drug Des Devel Ther 2024; 18:97-108. [PMID: 38264539 PMCID: PMC10804875 DOI: 10.2147/dddt.s435522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/10/2024] [Indexed: 01/25/2024] Open
Abstract
The cornea, as the outermost layer of the eye, plays a crucial role in vision by focusing light onto the retina. Various diseases and injuries can compromise its clarity, leading to impaired vision. This review aims to provide a thorough overview of the pharmacological properties, therapeutic potential and associated risks of Rho-associated protein kinase (ROCK) inhibitors in the management of corneal diseases. The article focuses on four key ROCK inhibitors: Y-27632, fasudil, ripasudil, and netarsudil, providing a comparative examination. Studies supporting the use of ROCK inhibitors highlight their efficacy across diverse corneal conditions. In Fuchs' endothelial corneal dystrophy, studies on the application of Y-27632, ripasudil, and netarsudil demonstrated noteworthy enhancements in corneal clarity, endothelial cell density, and visual acuity. In pseudophakic bullous keratopathy, the injection of Y-27632 together with cultured corneal endothelial cells into the anterior chamber lead to enhanced corneal endothelial cell density and improved visual acuity. Animal models simulating chemical injury to the cornea showed a reduction of neovascularization and epithelial defects after application of fasudil and in a case of iridocorneal endothelial syndrome netarsudil improved corneal edema. Addressing safety considerations, netarsudil and ripasudil, both clinically approved, exhibit adverse events such as conjunctival hyperemia, conjunctival hemorrhage, cornea verticillata, conjunctivitis, and blepharitis. Monitoring patients during treatment becomes crucial to balancing the potential therapeutic benefits with these associated risks. In conclusion, ROCK inhibitors, particularly netarsudil and ripasudil, offer promise in managing corneal diseases. The comparative analysis of their pharmacological properties and studies supporting their efficacy underscore their potential therapeutic significance. However, ongoing research is paramount to comprehensively understand their safety profiles and long-term outcomes in diverse corneal conditions, guiding their optimal application in clinical practice.
Collapse
Affiliation(s)
- Stefan Futterknecht
- Department of Ophthalmology, University Hospital of Basel, Basel, Switzerland
- Institute of Molecular and Clinical Ophthalmology Basel, Basel, Switzerland
| | | | - Konstantin Gugleta
- Department of Ophthalmology, University Hospital of Basel, Basel, Switzerland
- Department of Ophthalmology, School of Medicine, University of Basel, Basel, Switzerland
| | - Georgios D Panos
- Department of Ophthalmology, Queen’s Medical Centre, Nottingham University Hospitals, Nottingham, UK
- Division of Ophthalmology and Visual Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Zisis Gatzioufas
- Department of Ophthalmology, University Hospital of Basel, Basel, Switzerland
- Department of Ophthalmology, School of Medicine, University of Basel, Basel, Switzerland
| |
Collapse
|
42
|
Kim J, Kwon EJ, Kim YJ, Kim D, Shin YZ, Gil D, Kim JH, Shin HD, Kim LH, Lee MO, Go YH, Cha HJ. Epigenetic repression of CHCHD2 enhances survival from single cell dissociation through attenuated Rho A kinase activity. Cell Mol Life Sci 2024; 81:38. [PMID: 38214772 PMCID: PMC10787008 DOI: 10.1007/s00018-023-05060-8] [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: 06/14/2023] [Revised: 10/18/2023] [Accepted: 11/17/2023] [Indexed: 01/13/2024]
Abstract
During in vitro culture, human pluripotent stem cells (hPSCs) often acquire survival advantages characterized by decreased susceptibility to mitochondrial cell death, known as "culture adaptation." This adaptation is associated with genetic and epigenetic abnormalities, including TP53 mutations, copy number variations, trisomy, and methylation changes. Understanding the molecular mechanisms underlying this acquired survival advantage is crucial for safe hPSC-based cell therapies. Through transcriptome and methylome analysis, we discovered that the epigenetic repression of CHCHD2, a mitochondrial protein, is a common occurrence during in vitro culture using enzymatic dissociation. We confirmed this finding through genetic perturbation and reconstitution experiments in normal human embryonic stem cells (hESCs). Loss of CHCHD2 expression conferred resistance to single cell dissociation-induced cell death, a common stress encountered during in vitro culture. Importantly, we found that the downregulation of CHCHD2 significantly attenuates the activity of Rho-associated protein kinase (ROCK), which is responsible for inducing single cell death in hESCs. This suggests that hESCs may survive routine enzyme-based cell dissociation by downregulating CHCHD2 and thereby attenuating ROCK activity. These findings provide insights into the mechanisms by which hPSCs acquire survival advantages and adapt to in vitro culture conditions.
Collapse
Affiliation(s)
- Jumee Kim
- College of Pharmacy, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Eun-Ji Kwon
- College of Pharmacy, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Yun-Jeong Kim
- College of Pharmacy, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Dayeon Kim
- College of Pharmacy, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Yoon-Ze Shin
- College of Pharmacy, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Dayeon Gil
- Korea National Stem Cell Bank, Osong, Republic of Korea
- Division of Intractable Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Osong Health Technology Administration Complex 202, Osong, Republic of Korea
| | - Jung-Hyun Kim
- Korea National Stem Cell Bank, Osong, Republic of Korea
- Division of Intractable Disease Research, Department of Chronic Disease Convergence Research, Korea National Institute of Health, Osong Health Technology Administration Complex 202, Osong, Republic of Korea
| | - Hyoung Doo Shin
- Department of Life Science, Sogang University, Seoul, Republic of Korea
- Research Institute for Basic Science, Sogang University, Seoul, Republic of Korea
| | - Lyoung Hyo Kim
- Research Institute for Life Science, GW Vitek, Inc., Seoul, Republic of Korea
| | - Mi-Ok Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea
| | - Young-Hyun Go
- College of Pharmacy, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea.
- Research Institute of Pharmaceutical Science, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea.
| | - Hyuk-Jin Cha
- College of Pharmacy, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea.
- Research Institute of Pharmaceutical Science, Seoul National University, 1 Gwanak-ro Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
43
|
Maas-Bauer K, Stell AV, Yan KL, de Vega E, Vinnakota JM, Unger S, Núñez N, Norona J, Talvard-Balland N, Koßmann S, Schwan C, Miething C, Martens US, Shoumariyeh K, Nestor RP, Duquesne S, Hanke K, Rackiewicz M, Hu Z, El Khawanky N, Taromi S, Andrlova H, Faraidun H, Walter S, Pfeifer D, Follo M, Waldschmidt J, Melchinger W, Rassner M, Wehr C, Schmitt-Graeff A, Halbach S, Liao J, Häcker G, Brummer T, Dengjel J, Andrieux G, Grosse R, Tugues S, Blazar BR, Becher B, Boerries M, Zeiser R. ROCK1/2 signaling contributes to corticosteroid-refractory acute graft-versus-host disease. Nat Commun 2024; 15:446. [PMID: 38199985 PMCID: PMC10781952 DOI: 10.1038/s41467-024-44703-7] [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: 04/18/2020] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Patients with corticosteroid-refractory acute graft-versus-host disease (aGVHD) have a low one-year survival rate. Identification and validation of novel targetable kinases in patients who experience corticosteroid-refractory-aGVHD may help improve outcomes. Kinase-specific proteomics of leukocytes from patients with corticosteroid-refractory-GVHD identified rho kinase type 1 (ROCK1) as the most significantly upregulated kinase. ROCK1/2 inhibition improved survival and histological GVHD severity in mice and was synergistic with JAK1/2 inhibition, without compromising graft-versus-leukemia-effects. ROCK1/2-inhibition in macrophages or dendritic cells prior to transfer reduced GVHD severity. Mechanistically, ROCK1/2 inhibition or ROCK1 knockdown interfered with CD80, CD86, MHC-II expression and IL-6, IL-1β, iNOS and TNF production in myeloid cells. This was accompanied by impaired T cell activation by dendritic cells and inhibition of cytoskeletal rearrangements, thereby reducing macrophage and DC migration. NF-κB signaling was reduced in myeloid cells following ROCK1/2 inhibition. In conclusion, ROCK1/2 inhibition interferes with immune activation at multiple levels and reduces acute GVHD while maintaining GVL-effects, including in corticosteroid-refractory settings.
Collapse
Affiliation(s)
- Kristina Maas-Bauer
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna-Verena Stell
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kai-Li Yan
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Enrique de Vega
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Janaki Manoja Vinnakota
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Susanne Unger
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Nicolas Núñez
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Johana Norona
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nana Talvard-Balland
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefanie Koßmann
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carsten Schwan
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Cornelius Miething
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Uta S Martens
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Khalid Shoumariyeh
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, a partnership between German Cancer Research Center (DKFZ) and Medical Center - University of Freiburg, Freiburg, Germany
| | - Rosa P Nestor
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sandra Duquesne
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kathrin Hanke
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michal Rackiewicz
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Department of Dermatology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Zehan Hu
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Department of Dermatology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Nadia El Khawanky
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sanaz Taromi
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hana Andrlova
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hemin Faraidun
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Stefanie Walter
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marie Follo
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Johannes Waldschmidt
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Wolfgang Melchinger
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael Rassner
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claudia Wehr
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Sebastian Halbach
- German Cancer Consortium (DKTK), Partner Site Freiburg, a partnership between German Cancer Research Center (DKFZ) and Medical Center - University of Freiburg, Freiburg, Germany
- IMMZ, University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - James Liao
- Department of Medicine, University of Arizona, Tucson, USA
| | - Georg Häcker
- IMMH, University Hospital Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Tilman Brummer
- German Cancer Consortium (DKTK), Partner Site Freiburg, a partnership between German Cancer Research Center (DKFZ) and Medical Center - University of Freiburg, Freiburg, Germany
- IMMZ, University of Freiburg, Faculty of Medicine, Freiburg, Germany
- Signaling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Joern Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Department of Dermatology, Medical Center, University of Freiburg, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Grosse
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty, University of Freiburg, Freiburg, Germany
- CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Sonia Tugues
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN, USA
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Melanie Boerries
- German Cancer Consortium (DKTK), Partner Site Freiburg, a partnership between German Cancer Research Center (DKFZ) and Medical Center - University of Freiburg, Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- German Cancer Consortium (DKTK), Partner Site Freiburg, a partnership between German Cancer Research Center (DKFZ) and Medical Center - University of Freiburg, Freiburg, Germany.
- Signaling Research Centres BIOSS and CIBSS - Centre for Integrative Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
44
|
Costigan A, Hollville E, Martin SJ. Discriminating Between Apoptosis, Necrosis, Necroptosis, and Ferroptosis by Microscopy and Flow Cytometry. Curr Protoc 2023; 3:e951. [PMID: 38112058 DOI: 10.1002/cpz1.951] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Apoptosis is a mode of programmed cell death that plays important roles in tissue sculpting during development, in the maintenance of tissue homeostasis in the adult, and in the eradication of injured or infected cells during pathological processes. Numerous physiological as well as pathological stimuli trigger apoptosis, such as engagement of plasma-membrane-associated Fas, TRAIL, or TNF receptors, growth factor deprivation, hypoxia, radiation, and exposure to diverse cytotoxic drugs. Apoptosis is coordinated by members of the caspase family of cysteine proteases, which, upon activation, trigger a series of dramatic morphological and biochemical changes including retraction from the substratum, cell shrinkage, extensive and protracted plasma membrane blebbing, chromatin condensation, DNA hydrolysis, nuclear fragmentation, and proteolytic cleavage of numerous caspase substrates. These dramatic structural and biochemical alterations result not only in the controlled dismantling of the cell, but also in the rapid recognition and removal of apoptotic cells by phagocytes through the cell surface display of phagocytotic triggers such as phosphatidylserine. Necrosis, which is typically nonprogrammed or imposed upon the cell by overwhelming membrane or organelle damage, is characterized by high-amplitude cell swelling, followed by rapid plasma membrane rupture and release of cellular contents into the extracellular space. Necrosis is often provoked by infectious agents or severe departure from physiological conditions due to toxins, temperature extremes, or physical injury. However, forms of programmed necrosis (necroptosis, pyroptosis, ferroptosis) can also occur in specific circumstances. Nonprogrammed and programmed necrosis can be distinguished from apoptosis by morphological features, based on the rapid uptake of vital dyes, and through the application of specific inhibitors of key molecules associated with the latter modes of cell death. This unit describes protocols for the measurement of apoptosis and necrosis and for distinguishing apoptosis from programmed as well as conventional necrosis. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Analysis of cell morphology by phase-contrast microscopy Alternative Protocol 1: Assessment of morphological changes using eosin-methylene blue staining Alternative Protocol 2: Analysis of nuclear morphology by fluorescence microscopy Support Protocol: Preparation of cytospins Basic Protocol 2: Measurement of plasma membrane composition with annexin V and propidium iodide Basic Protocol 3: Measurement of DNA fragmentation by flow cytometry Alternative Protocol 3: Analysis of DNA fragmentation by the TUNEL assay Basic Protocol 4: Measurement of caspase activation by flow cytometry Basic Protocol 5: Discriminating between apoptosis, necrosis, necroptosis, and ferroptosis.
Collapse
Affiliation(s)
- Aoife Costigan
- Molecular Cell Biology Laboratory, Department of Genetics, Trinity College, Dublin, Ireland
| | - Emilie Hollville
- Institute of Medical Sciences, University of Aberdeen Foresterhill, Aberdeen, Scotland, United Kingdom
| | - Seamus J Martin
- Molecular Cell Biology Laboratory, Department of Genetics, Trinity College, Dublin, Ireland
| |
Collapse
|
45
|
Soh JEC, Shimizu A, Sato A, Ogita H. Novel cardiovascular protective effects of RhoA signaling and its therapeutic implications. Biochem Pharmacol 2023; 218:115899. [PMID: 37907138 DOI: 10.1016/j.bcp.2023.115899] [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: 09/19/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/02/2023]
Abstract
Ras homolog gene family member A (RhoA) belongs to the Rho GTPase superfamily, which was first studied in cancers as one of the essential regulators controlling cellular function. RhoA has long attracted attention as a key molecule involved in cell signaling and gene transcription, through which it affects cellular processes. A series of studies have demonstrated that RhoA plays crucial roles under both physiological states and pathological conditions in cardiovascular diseases. RhoA has been identified as an important regulator in cardiac remodeling by regulating actin stress fiber dynamics and cytoskeleton formation. However, its underlying mechanisms remain poorly understood, preventing definitive conclusions being drawn about its protective role in the cardiovascular system. In this review, we outline the characteristics of RhoA and its related signaling molecules, and present an overview of RhoA classical function and the corresponding cellular responses of RhoA under physiological and pathological conditions. Overall, we provide an update on the novel signaling under RhoA in the cardiovascular system and its potential clinical and therapeutic targets in cardiovascular medicine.
Collapse
Affiliation(s)
- Joanne Ern Chi Soh
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan.
| |
Collapse
|
46
|
Chambel SS, Cruz CD. Axonal growth inhibitors and their receptors in spinal cord injury: from biology to clinical translation. Neural Regen Res 2023; 18:2573-2581. [PMID: 37449592 DOI: 10.4103/1673-5374.373674] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Abstract
Axonal growth inhibitors are released during traumatic injuries to the adult mammalian central nervous system, including after spinal cord injury. These molecules accumulate at the injury site and form a highly inhibitory environment for axonal regeneration. Among these inhibitory molecules, myelin-associated inhibitors, including neurite outgrowth inhibitor A, oligodendrocyte myelin glycoprotein, myelin-associated glycoprotein, chondroitin sulfate proteoglycans and repulsive guidance molecule A are of particular importance. Due to their inhibitory nature, they represent exciting molecular targets to study axonal inhibition and regeneration after central injuries. These molecules are mainly produced by neurons, oligodendrocytes, and astrocytes within the scar and in its immediate vicinity. They exert their effects by binding to specific receptors, localized in the membranes of neurons. Receptors for these inhibitory cues include Nogo receptor 1, leucine-rich repeat, and Ig domain containing 1 and p75 neurotrophin receptor/tumor necrosis factor receptor superfamily member 19 (that form a receptor complex that binds all myelin-associated inhibitors), and also paired immunoglobulin-like receptor B. Chondroitin sulfate proteoglycans and repulsive guidance molecule A bind to Nogo receptor 1, Nogo receptor 3, receptor protein tyrosine phosphatase σ and leucocyte common antigen related phosphatase, and neogenin, respectively. Once activated, these receptors initiate downstream signaling pathways, the most common amongst them being the RhoA/ROCK signaling pathway. These signaling cascades result in actin depolymerization, neurite outgrowth inhibition, and failure to regenerate after spinal cord injury. Currently, there are no approved pharmacological treatments to overcome spinal cord injuries other than physical rehabilitation and management of the array of symptoms brought on by spinal cord injuries. However, several novel therapies aiming to modulate these inhibitory proteins and/or their receptors are under investigation in ongoing clinical trials. Investigation has also been demonstrating that combinatorial therapies of growth inhibitors with other therapies, such as growth factors or stem-cell therapies, produce stronger results and their potential application in the clinics opens new venues in spinal cord injury treatment.
Collapse
Affiliation(s)
- Sílvia Sousa Chambel
- Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine of Porto; Translational NeuroUrology, Instituto de Investigação e Inovação em Saúde-i3S and IBMC, Universidade do Porto, Porto, Portugal
| | - Célia Duarte Cruz
- Experimental Biology Unit, Department of Biomedicine, Faculty of Medicine of Porto; Translational NeuroUrology, Instituto de Investigação e Inovação em Saúde-i3S and IBMC, Universidade do Porto, Porto, Portugal
| |
Collapse
|
47
|
Barroso-Gomila O, Merino-Cacho L, Muratore V, Perez C, Taibi V, Maspero E, Azkargorta M, Iloro I, Trulsson F, Vertegaal ACO, Mayor U, Elortza F, Polo S, Barrio R, Sutherland JD. BioE3 identifies specific substrates of ubiquitin E3 ligases. Nat Commun 2023; 14:7656. [PMID: 37996419 PMCID: PMC10667490 DOI: 10.1038/s41467-023-43326-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 11/07/2023] [Indexed: 11/25/2023] Open
Abstract
Hundreds of E3 ligases play a critical role in recognizing specific substrates for modification by ubiquitin (Ub). Separating genuine targets of E3s from E3-interactors remains a challenge. We present BioE3, a powerful approach for matching substrates to Ub E3 ligases of interest. Using BirA-E3 ligase fusions and bioUb, site-specific biotinylation of Ub-modified substrates of particular E3s facilitates proteomic identification. We show that BioE3 identifies both known and new targets of two RING-type E3 ligases: RNF4 (DNA damage response, PML bodies), and MIB1 (endocytosis, autophagy, centrosome dynamics). Versatile BioE3 identifies targets of an organelle-specific E3 (MARCH5) and a relatively uncharacterized E3 (RNF214). Furthermore, BioE3 works with NEDD4, a HECT-type E3, identifying new targets linked to vesicular trafficking. BioE3 detects altered specificity in response to chemicals, opening avenues for targeted protein degradation, and may be applicable for other Ub-likes (UbLs, e.g., SUMO) and E3 types. BioE3 applications shed light on cellular regulation by the complex UbL network.
Collapse
Affiliation(s)
- Orhi Barroso-Gomila
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
| | - Laura Merino-Cacho
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
| | - Veronica Muratore
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
| | - Coralia Perez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
| | - Vincenzo Taibi
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Elena Maspero
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Mikel Azkargorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
- CIBERehd, Instituto de Salud Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Ibon Iloro
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
- CIBERehd, Instituto de Salud Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Fredrik Trulsson
- Cell and Chemical Biology, Leiden University Medical Center (LUMC), 2333, ZA, Leiden, The Netherlands
| | - Alfred C O Vertegaal
- Cell and Chemical Biology, Leiden University Medical Center (LUMC), 2333, ZA, Leiden, The Netherlands
| | - Ugo Mayor
- Ikerbasque, Basque Foundation for Science, 48011, Bilbao, Spain
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), E-48940, Leioa, Spain
| | - Felix Elortza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain
- CIBERehd, Instituto de Salud Carlos III, C/ Monforte de Lemos 3-5, Pabellón 11, Planta 0, 28029, Madrid, Spain
| | - Simona Polo
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
- Dipartimento di oncologia ed emato-oncologia, Università degli Studi di Milano, Milan, Italy
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain.
| | - James D Sutherland
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Building 801A, 48160, Derio, Spain.
| |
Collapse
|
48
|
Ripoli C, Dagliyan O, Renna P, Pastore F, Paciello F, Sollazzo R, Rinaudo M, Battistoni M, Martini S, Tramutola A, Sattin A, Barone E, Saneyoshi T, Fellin T, Hayashi Y, Grassi C. Engineering memory with an extrinsically disordered kinase. SCIENCE ADVANCES 2023; 9:eadh1110. [PMID: 37967196 PMCID: PMC10651130 DOI: 10.1126/sciadv.adh1110] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 10/13/2023] [Indexed: 11/17/2023]
Abstract
Synaptic plasticity plays a crucial role in memory formation by regulating the communication between neurons. Although actin polymerization has been linked to synaptic plasticity and dendritic spine stability, the causal link between actin polymerization and memory encoding has not been identified yet. It is not clear whether actin polymerization and structural changes in dendritic spines are a driver or a consequence of learning and memory. Using an extrinsically disordered form of the protein kinase LIMK1, which rapidly and precisely acts on ADF/cofilin, a direct modifier of actin, we induced long-term enlargement of dendritic spines and enhancement of synaptic transmission in the hippocampus on command. The activation of extrinsically disordered LIMK1 in vivo improved memory encoding and slowed cognitive decline in aged mice exhibiting reduced cofilin phosphorylation. The engineered memory by an extrinsically disordered LIMK1 supports a direct causal link between actin-mediated synaptic transmission and memory.
Collapse
Affiliation(s)
- Cristian Ripoli
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Onur Dagliyan
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17165 Stockholm, Sweden
| | - Pietro Renna
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Pastore
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Fabiola Paciello
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Raimondo Sollazzo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marco Rinaudo
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Martina Battistoni
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Sara Martini
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy
| | - Andrea Sattin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences “A. Rossi-Fanelli”, Sapienza University of Rome, 00185 Rome, Italy
| | - Takeo Saneyoshi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Tommaso Fellin
- Optical Approaches to Brain Function Laboratory, Istituto Italiano di Tecnologia, 16163 Genoa, Italy
| | - Yasunori Hayashi
- Department of Pharmacology, Kyoto University Graduate School of Medicine, Kyoto 606-8501, Japan
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| |
Collapse
|
49
|
Viola JM, Liu J, Huang A, Grindel SH, Prahl LS, Hughes AJ. Rho/ROCK activity tunes cell compartment segregation and differentiation in nephron-forming niches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566308. [PMID: 37986773 PMCID: PMC10659296 DOI: 10.1101/2023.11.08.566308] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Controlling the time and place of nephron formation in vitro would improve nephron density and connectivity in next-generation kidney replacement tissues. Recent developments in kidney organoid technology have paved the way to achieving self-sustaining nephrogenic niches in vitro. The physical and geometric structure of the niche are key control parameters in tissue engineering approaches. However, their relationship to nephron differentiation is unclear. Here we investigate the relationship between niche geometry, cell compartment mixing, and nephron differentiation by targeting the Rho/ROCK pathway, a master regulator of the actin cytoskeleton. We find that the ROCK inhibitor Y-27632 increases mixing between nephron progenitor and stromal compartments in native mouse embryonic kidney niches, and also increases nephrogenesis. Similar increases are also seen in reductionist mouse primary cell and human induced pluripotent stem cell (iPSC)-derived organoids perturbed by Y-27632, dependent on the presence of stromal cells. Our data indicate that niche organization is a determinant of nephron formation rate, bringing renewed focus to the spatial context of cell-cell interactions in kidney tissue engineering efforts.
Collapse
Affiliation(s)
- John M. Viola
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Jiageng Liu
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Aria Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Samuel H. Grindel
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Louis S. Prahl
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
| | - Alex J. Hughes
- Department of Bioengineering, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Bioengineering Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Center for Soft and Living Matter, University of Pennsylvania, Philadelphia, 19104, PA, USA
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, 19104, PA, USA
| |
Collapse
|
50
|
Della Rosa G, Di Buduo CA, Balduini A. Unraveling the hormonal pathway to megakaryocyte well-being. J Thromb Haemost 2023; 21:3078-3081. [PMID: 37858524 DOI: 10.1016/j.jtha.2023.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 07/26/2023] [Accepted: 08/03/2023] [Indexed: 10/21/2023]
Affiliation(s)
| | | | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Pavia, Italy; Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA.
| |
Collapse
|