1
|
Murugesan A, Doan P, Rajendran P, Thiyagarajan R, Chandrabose S, Abass KS, Kandhavelu M. Anti-GBM effects of polyphenolic extract from Avicennia marina. JOURNAL OF ETHNOPHARMACOLOGY 2025:120107. [PMID: 40490234 DOI: 10.1016/j.jep.2025.120107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2025] [Revised: 05/28/2025] [Accepted: 06/04/2025] [Indexed: 06/11/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Avicennia marina, (Forssk.) Vierh. the most common halo-tolerant plant, is rich in polyphenolic compounds such as phenols, flavonoids, saponins, alkaloids, and steroids. Avicennia marina exhibits significant therapeutic potential, including antioxidants, antiviral, immunosuppressive, and anticancer properties. Numerous halo-tolerant plant metabolites have been examined for their anti-cancer effects; nevertheless, their efficacy against Glioblastoma multiforme (GBM), an aggressive malignant neoplasm, remains poorly understood. AIM OF THE STUDY This study investigates the anticancer efficacy of Avicennia marina (AM) in GBM. The majority of GBM treatments are non-specific and provide limited survival benefits for people with GBM. This study aims to scientifically validate the traditional usage of AM by evaluating its cytotoxic effects and exploring its therapeutic potential. MATERIALS AND METHODS To evaluate the cytotoxic effect of the purified polyphenols of AM against GBM cells, various in-vitro analysis was performed cytotoxicity assay, apoptosis assay, wound healing assay, caspase 3/7, Reactive oxygen species (ROS) generation and Mitogen-Activated Protein Kinase (MAPKinase) proteomic array. Gas chromatography-mass spectrometry (GC-MS) was performed to profile the secondary metabolites present in the phenolic fraction of AM. RESULTS The half maximum inhibitory concentration (IC50) of AM was 0.80 μg/mL for LN229 cells and 1.10 μg/mL for SNB19 cells, both of which are lower than the known chemotherapeutic drug, Temozolomide (TMZ). GC-MS examination of the isolated polyphenols identified 14 secondary metabolites. Furthermore, AM elevated intracellular reactive oxygen species and activated caspase 3/7 in both cell lines, triggering apoptosis and leads to GBM cell mortality. Furthermore, AM inhibits the migration of GBM cells over time, demonstrating its anti-metastatic efficacy. Additionally, investigation of the MAPK signaling pathway demonstrated the phosphorylation of critical proteins associated with the ERK, JNK, and p38 protein families. The upregulation of p70S6 kinase and the downregulation of Akt2, Akt3, p38α, and RSK1 indicate that AM could impact the protein synthesis pathway and cell cycle progression. CONCLUSION The study indicates that polyphenols in Avicennia marina may serve as a promising candidate for the development of new anti-GBM pharmaceuticals.
Collapse
Affiliation(s)
- Akshaya Murugesan
- Molecular Signaling Group, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland; Department of Biotechnology, Lady Doak College, Thallakulam, Madurai, India.
| | - Phuong Doan
- Department of Biotechnology, Lady Doak College, Thallakulam, Madurai, India
| | | | - Ramesh Thiyagarajan
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sureka Chandrabose
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Kasim Sakran Abass
- Department of Physiology, Biochemistry, and Pharmacology, College of Veterinary Medicine, University of Kirkuk, Kirkuk-36001, Iraq
| | - Meenakshisundaram Kandhavelu
- Department of Biotechnology, Lady Doak College, Thallakulam, Madurai, India; BioMeditech and Tays Cancer Center, Tampere University, Hospital, P.O. Box 553, 33101 Tampere, Finland.
| |
Collapse
|
2
|
Durer ZA, İnce Hİ, Düvenci ZŞ, Timuçin E, Gräwert T, Orun O, Kan B, Sayers Z. Insights into the solution structure of the actin-binding tail domain of metavinculin by small angle X-ray scattering and molecular dynamics simulations. Int J Biol Macromol 2025:144376. [PMID: 40409637 DOI: 10.1016/j.ijbiomac.2025.144376] [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: 02/27/2025] [Revised: 05/07/2025] [Accepted: 05/17/2025] [Indexed: 05/25/2025]
Abstract
Vinculin is a ubiquitously expressed focal adhesion protein that plays an important role in cell-matrix and cell-to-cell junctions. Metavinculin, a muscle-specific splice variant of vinculin, contains a 68-amino acid disordered insert region in its actin binding tail domain (MVt). Mutations in this insert are linked to cardiomyopathies. This study investigates the solution structures and structural ensembles of wild-type (WT) and two mutant MVts, ΔLeu954 and R975W, which have been associated with cardiomyopathies, using small-angle X-ray scattering (SAXS) and molecular dynamics (MD) simulations. SAXS analyses revealed subtle differences in the estimated maximum dimensions and corroborated the elongated shape of the MVts. Quantitative comparisons of SAXS profiles indicated similarity between the WT and ΔLeu954, whereas R975W exhibited differences in the small-angle region. MD simulations demonstrated reduced conformational flexibility and greater packing of the insert in WT compared to mutants. Notably, a salt-bridge observed between R975 and D928 in a WT simulation provides a structural basis for the destabilization caused by the R975W mutation. These findings provide insights into the structure and dynamics of WT and mutant MVt, reflecting the promise of SAXS combined with MD simulations to elucidate the structural properties of proteins with structural disorder.
Collapse
Affiliation(s)
- Zeynep Aslıhan Durer
- School of Medicine, Department of Biophysics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey; Faculty of Pharmacy, Department of Biochemistry, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.
| | - Hande İpek İnce
- Institute of Health Sciences, Department of Biophysics, Marmara University, Istanbul, Turkey
| | - Zeynep Şevval Düvenci
- Institute of Health Sciences, Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Emel Timuçin
- Institute of Health Sciences, Department of Biostatistics and Bioinformatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey; School of Medicine, Department of Biostatistics and Medical Informatics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Tobias Gräwert
- European Molecular Biology Laboratory - Hamburg Unit, Biological Small Angle Scattering Group, Hamburg, Germany
| | - Oya Orun
- Institute of Health Sciences, Department of Biophysics, Marmara University, Istanbul, Turkey; School of Medicine, Department of Biophysics, Marmara University, Istanbul, Turkey
| | - Beki Kan
- School of Medicine, Department of Biophysics, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Zehra Sayers
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, İstanbul, Turkey
| |
Collapse
|
3
|
DePalma SJ, Jilberto J, Stis AE, Huang DD, Lo J, Davidson CD, Chowdhury A, Kent RN, Jewett ME, Kobeissi H, Chen CS, Lejeune E, Helms AS, Nordsletten DA, Baker BM. Matrix Architecture and Mechanics Regulate Myofibril Organization, Costamere Assembly, and Contractility in Engineered Myocardial Microtissues. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309740. [PMID: 39558513 DOI: 10.1002/advs.202309740] [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: 12/12/2023] [Revised: 07/09/2024] [Indexed: 11/20/2024]
Abstract
The mechanical function of the myocardium is defined by cardiomyocyte contractility and the biomechanics of the extracellular matrix (ECM). Understanding this relationship remains an important unmet challenge due to limitations in existing approaches for engineering myocardial tissue. Here, they established arrays of cardiac microtissues with tunable mechanics and architecture by integrating ECM-mimetic synthetic, fiber matrices, and induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), enabling real-time contractility readouts, in-depth structural assessment, and tissue-specific computational modeling. They found that the stiffness and alignment of matrix fibers distinctly affect the structural development and contractile function of pure iPSC-CM tissues. Further examination into the impact of fibrous matrix stiffness enabled by computational models and quantitative immunofluorescence implicates cell-ECM interactions in myofibril assembly, myofibril maturation, and notably costamere assembly, which correlates with improved contractile function of tissues. These results highlight how iPSC-CM tissue models with controllable architecture and mechanics can elucidate mechanisms of tissue maturation and disease.
Collapse
Affiliation(s)
- Samuel J DePalma
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Javiera Jilberto
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Austin E Stis
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Darcy D Huang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jason Lo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | | | - Aamilah Chowdhury
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Maggie E Jewett
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Hiba Kobeissi
- Department of Mechanical Engineering, Boston University, Boston, MA, 02215, USA
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, MA, 02215, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Emma Lejeune
- Department of Mechanical Engineering, Boston University, Boston, MA, 02215, USA
| | - Adam S Helms
- Division of Cardiovascular Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - David A Nordsletten
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Cardiac Surgery, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biomedical Engineering, School of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, London, SE1 7EH, UK
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
4
|
Katoh K. Signal Transduction Mechanisms of Focal Adhesions: Src and FAK-Mediated Cell Response. FRONT BIOSCI-LANDMRK 2024; 29:392. [PMID: 39614431 DOI: 10.31083/j.fbl2911392] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 12/01/2024]
Abstract
Cell-to-substrate adhesion sites, also known as focal adhesion sites (FAs), are complexes of different proteins on the cell surface. FAs play important roles in communication between cells and the extracellular matrix (ECM), leading to signal transduction involving different proteins that ultimately produce the cell response. This cell response involves cell adhesion, migration, motility, cell survival, and cell proliferation. The most important component of FAs are integrins. Integrins are transmembrane proteins that receive signals from the ECM and communicate them to the cytoplasm, thus activating several downstream proteins in a signaling cascade. Cellular Proto-oncogene tyrosine-protein kinase Src (c-Src) and focal adhesion kinase (FAK) are non-receptor tyrosine kinases that functionally interact to promote crucial roles in FAs. c-Src is a tyrosine kinase, activated by autophosphorylation and, in turn, activates another important protein, FAK. Activated FAK directly interacts with the cytoplasmic domain of integrin and activates other FA proteins by attaching to them. These proteins activated by FAK then activate other downstream pathways such as mitogen-activated protein kinase (MAPK) and Akt pathways involved in cell proliferation, migration, and cell survival. Src can induce detachment of FAK from the integrin to increase the focal adhesion turnover. As a result, the Src-FAK complex in FAs is critical for cell adhesion and survival mechanisms. Overexpression of FA proteins has been linked to a variety of pathological disorders, including cancers, growth retardation, and bone deformities. FAK and Src are overexpressed in various cancers. This review, which focuses on the roles of two important signaling proteins, c-Src and FAK, attempts to provide a thorough and up-to-date examination of the signal transduction mechanisms mediated by focal adhesions. The author also described that FAK and Src may serve as potential targets for future therapies against diseases associated with their overexpression, such as certain types of cancer.
Collapse
Affiliation(s)
- Kazuo Katoh
- Laboratory of Human Anatomy and Cell Biology, Faculty of Health Sciences, Tsukuba University of Technology, 305-8521 Tsukuba, Japan
| |
Collapse
|
5
|
Callaway DJE, Nicholl ID, Shi B, Reyes G, Farago B, Bu Z. Nanoscale dynamics of the cadherin-catenin complex bound to vinculin revealed by neutron spin echo spectroscopy. Proc Natl Acad Sci U S A 2024; 121:e2408459121. [PMID: 39298480 PMCID: PMC11441495 DOI: 10.1073/pnas.2408459121] [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: 04/28/2024] [Accepted: 08/12/2024] [Indexed: 09/21/2024] Open
Abstract
We report a neutron spin echo (NSE) study of the nanoscale dynamics of the cell-cell adhesion cadherin-catenin complex bound to vinculin. Our measurements and theoretical physics analyses of the NSE data reveal that the dynamics of full-length α-catenin, β-catenin, and vinculin residing in the cadherin-catenin-vinculin complex become activated, involving nanoscale motions in this complex. The cadherin-catenin complex is the central component of the cell-cell adherens junction (AJ) and is fundamental to embryogenesis, tissue wound healing, neuronal plasticity, cancer metastasis, and cardiovascular health and disease. A highly dynamic cadherin-catenin-vinculin complex provides the molecular dynamics basis for the flexibility and elasticity that are necessary for the AJs to function as force transducers. Our theoretical physics analysis provides a way to elucidate these driving nanoscale motions within the complex without requiring large-scale numerical simulations, providing insights not accessible by other techniques. We propose a three-way "motorman" entropic spring model for the dynamic cadherin-catenin-vinculin complex, which allows the complex to function as a flexible and elastic force transducer.
Collapse
Affiliation(s)
- David J. E. Callaway
- Department of Chemistry and Biochemistry, City College of New York, City University of New York, New York, NY10031
| | - Iain D. Nicholl
- Department of Biomedical Science and Physiology, Faculty of Science and Engineering, University of Wolverhampton, WolverhamptonWV1 1LY, United Kingdom
| | - Bright Shi
- Department of Chemistry and Biochemistry, City College of New York, City University of New York, New York, NY10031
- Ph.D. Programs in Chemistry and Biochemistry, City University of New York Graduate Center, New York, NY10016
| | - Gilbert Reyes
- Department of Chemistry and Biochemistry, City College of New York, City University of New York, New York, NY10031
- Ph.D. Programs in Chemistry and Biochemistry, City University of New York Graduate Center, New York, NY10016
| | - Bela Farago
- High-Resolution Spectroscopy Group, Institut Laue-Langevin, F-38042 Grenoble Cedex 9, France
| | - Zimei Bu
- Department of Chemistry and Biochemistry, City College of New York, City University of New York, New York, NY10031
- Ph.D. Programs in Chemistry and Biochemistry, City University of New York Graduate Center, New York, NY10016
| |
Collapse
|
6
|
Micolonghi C, Perrone F, Fabiani M, Caroselli S, Savio C, Pizzuti A, Germani A, Visco V, Petrucci S, Rubattu S, Piane M. Unveiling the Spectrum of Minor Genes in Cardiomyopathies: A Narrative Review. Int J Mol Sci 2024; 25:9787. [PMID: 39337275 PMCID: PMC11431948 DOI: 10.3390/ijms25189787] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Hereditary cardiomyopathies (CMPs), including arrhythmogenic cardiomyopathy (ACM), dilated cardiomyopathy (DCM), and hypertrophic cardiomyopathy (HCM), represent a group of heart disorders that significantly contribute to cardiovascular morbidity and mortality and are often driven by genetic factors. Recent advances in next-generation sequencing (NGS) technology have enabled the identification of rare variants in both well-established and minor genes associated with CMPs. Nowadays, a set of core genes is included in diagnostic panels for ACM, DCM, and HCM. On the other hand, despite their lesser-known status, variants in the minor genes may contribute to disease mechanisms and influence prognosis. This review evaluates the current evidence supporting the involvement of the minor genes in CMPs, considering their potential pathogenicity and clinical significance. A comprehensive analysis of databases, such as ClinGen, ClinVar, and GeneReviews, along with recent literature and diagnostic guidelines provides a thorough overview of the genetic landscape of minor genes in CMPs and offers guidance in clinical practice, evaluating each case individually based on the clinical referral, and insights for future research. Given the increasing knowledge on these less understood genetic factors, future studies are essential to clearly assess their roles, ultimately leading to improved diagnostic precision and therapeutic strategies in hereditary CMPs.
Collapse
Affiliation(s)
- Caterina Micolonghi
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
| | - Federica Perrone
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Department of Neuroscience, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Marco Fabiani
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- ALTAMEDICA, Human Genetics, 00198 Rome, Italy
| | - Silvia Caroselli
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Juno Genetics, Reproductive Genetics, 00188 Rome, Italy
| | | | - Antonio Pizzuti
- Department of Experimental Medicine, Faculty of Medicine and Dentistry, Sapienza University of Rome, 00161 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Aldo Germani
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Vincenzo Visco
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Simona Petrucci
- S. Andrea University Hospital, 00189 Rome, Italy
- Medical Genetics Unit, IRCCS Mendel Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| | - Speranza Rubattu
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Maria Piane
- S. Andrea University Hospital, 00189 Rome, Italy
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
7
|
Turkki P, Chowdhury I, Öhman T, Azizi L, Varjosalo M, Hytönen VP. Tensin-2 interactomics reveals interaction with GAPDH and a phosphorylation-mediated regulatory role in glycolysis. Sci Rep 2024; 14:19862. [PMID: 39191795 PMCID: PMC11350193 DOI: 10.1038/s41598-024-65787-7] [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: 04/20/2023] [Accepted: 06/24/2024] [Indexed: 08/29/2024] Open
Abstract
Integrin adaptor proteins, like tensin-2, are crucial for cell adhesion and signaling. However, the function of tensin-2 beyond localizing to focal adhesions remain poorly understood. We utilized proximity-dependent biotinylation and Strep-tag affinity proteomics to identify interaction partners of tensin-2 in Flp-In 293 T-REx cells. Interactomics linked tensin-2 to known focal adhesion proteins and the dystrophin glycoprotein complex, while also uncovering novel interaction with the glycolytic enzyme GAPDH. We demonstrated that Y483-phosphorylation of tensin-2 regulates the glycolytic rate in Flp-In 293 T-REx and MEF cells and found that pY483 tensin-2 is enriched in adhesions in MEF cells. Our study unveils novel interaction partners for tensin-2 and further solidifies its speculated role in cell energy metabolism. These findings shed fresh insight on the functions of tensin-2, highlighting its potential as a therapeutic target for diseases associated with impaired cell adhesion and metabolism.
Collapse
Affiliation(s)
- Paula Turkki
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories, Tampere, Finland
| | | | - Tiina Öhman
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Latifeh Azizi
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.
- Fimlab Laboratories, Tampere, Finland.
| |
Collapse
|
8
|
Wilson ZS, Raya-Sandino A, Miranda J, Fan S, Brazil JC, Quiros M, Garcia-Hernandez V, Liu Q, Kim CH, Hankenson KD, Nusrat A, Parkos CA. Critical role of thrombospondin-1 in promoting intestinal mucosal wound repair. JCI Insight 2024; 9:e180608. [PMID: 39078701 PMCID: PMC11385097 DOI: 10.1172/jci.insight.180608] [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: 02/23/2024] [Accepted: 07/18/2024] [Indexed: 09/11/2024] Open
Abstract
Thrombospondin-1 (TSP1) is a matricellular protein associated with the regulation of cell migration through direct binding interactions with integrin proteins and by associating with other receptors known to regulate integrin function, including CD47 and CD36. We previously demonstrated that deletion of an epithelial TSP1 receptor, CD47, attenuates epithelial wound repair following intestinal mucosal injury. However, the mechanisms by which TSP1 contributes to intestinal mucosal repair remain poorly understood. Our results show upregulated TSP1 expression in colonic mucosal wounds and impaired intestinal mucosal wound healing in vivo upon intestinal epithelium-specific loss of TSP1 (VillinCre/+ Thbs1fl/fl or Thbs1ΔIEC mice). We report that exposure to exogenous TSP1 enhanced migration of intestinal epithelial cells in a CD47- and TGF-β1-dependent manner and that deficiency of TSP1 in primary murine colonic epithelial cells resulted in impaired wound healing. Mechanistically, TSP1 modulated epithelial actin cytoskeletal dynamics through suppression of RhoA activity, activation of Rho family small GTPase (Rac1), and changes in filamentous-actin bundling. Overall, TSP1 was found to regulate intestinal mucosal wound healing via CD47 and TGF-β1, coordinate integrin-containing cell-matrix adhesion dynamics, and remodel the actin cytoskeleton in migrating epithelial cells to enhance cell motility and promote wound repair.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Qingyang Liu
- Department of Pathology
- Mary H. Weiser Food Allergy Center, and
| | - Chang H. Kim
- Department of Pathology
- Mary H. Weiser Food Allergy Center, and
| | - Kurt D. Hankenson
- Department of Orthopedic Surgery, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
9
|
Aboonabi A, McCauley MD. Myofilament dysfunction in diastolic heart failure. Heart Fail Rev 2024; 29:79-93. [PMID: 37837495 PMCID: PMC10904515 DOI: 10.1007/s10741-023-10352-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/16/2023]
Abstract
Diastolic heart failure (DHF), in which impaired ventricular filling leads to typical heart failure symptoms, represents over 50% of all heart failure cases and is linked with risk factors, including metabolic syndrome, hypertension, diabetes, and aging. A substantial proportion of patients with this disorder maintain normal left ventricular systolic function, as assessed by ejection fraction. Despite the high prevalence of DHF, no effective therapeutic agents are available to treat this condition, partially because the molecular mechanisms of diastolic dysfunction remain poorly understood. As such, by focusing on the underlying molecular and cellular processes contributing to DHF can yield new insights that can represent an exciting new avenue and propose a novel therapeutic approach for DHF treatment. This review discusses new developments from basic and clinical/translational research to highlight current knowledge gaps, help define molecular determinants of diastolic dysfunction, and clarify new targets for treatment.
Collapse
Affiliation(s)
- Anahita Aboonabi
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago, 840 S. Wood St., 920S (MC 715), Chicago, IL, 60612, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
| | - Mark D McCauley
- Division of Cardiology, Department of Medicine, College of Medicine, University of Illinois at Chicago, 840 S. Wood St., 920S (MC 715), Chicago, IL, 60612, USA.
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- Department of Physiology and Biophysics and the Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
10
|
Chirasani VR, Khan MAI, Malavade JN, Dokholyan NV, Hoffman BD, Campbell SL. Molecular basis and cellular functions of vinculin-actin directional catch bonding. Nat Commun 2023; 14:8300. [PMID: 38097542 PMCID: PMC10721916 DOI: 10.1038/s41467-023-43779-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 11/20/2023] [Indexed: 12/17/2023] Open
Abstract
The ability of cells and tissues to respond differentially to mechanical forces applied in distinct directions is mediated by the ability of load-bearing proteins to preferentially maintain physical linkages in certain directions. However, the molecular basis and biological consequences of directional force-sensitive binding remain unclear. Vinculin (Vcn) is a load-bearing linker protein that exhibits directional catch bonding due to interactions between the Vcn tail domain (Vt) and filamentous (F)-actin. We developed a computational approach to predict Vcn residues involved in directional catch bonding and produced a set of associated Vcn variants with unaltered Vt structure, actin binding, or phospholipid interactions. Incorporation of the variants did not affect Vcn activation but reduced Vcn loading and altered exchange dynamics, consistent with the loss of directional catch bonding. Expression of Vcn variants perturbed the coordination of subcellular structures and cell migration, establishing key cellular functions for Vcn directional catch bonding.
Collapse
Affiliation(s)
- Venkat R Chirasani
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mohammad Ashhar I Khan
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Juilee N Malavade
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nikolay V Dokholyan
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.
- Department of Biochemistry & Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
- Department of Chemistry, Penn State College of Medicine, Hershey, PA, USA.
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Department of Cell Biology, Duke University, Durham, NC, USA.
| | - Sharon L Campbell
- Department of Biochemistry & Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
11
|
Shoyer TC, Gates EM, Cabe JI, Urs AN, Conway DE, Hoffman BD. Coupling during collective cell migration is controlled by a vinculin mechanochemical switch. Proc Natl Acad Sci U S A 2023; 120:e2316456120. [PMID: 38055737 PMCID: PMC10722971 DOI: 10.1073/pnas.2316456120] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/24/2023] [Indexed: 12/08/2023] Open
Abstract
The ability of cells to move in a mechanically coupled, coordinated manner, referred to as collective cell migration, is central to many developmental, physiological, and pathophysiological processes. Limited understanding of how mechanical forces and biochemical regulation interact to affect coupling has been a major obstacle to unravelling the underlying mechanisms. Focusing on the linker protein vinculin, we use a suite of Förster resonance energy transfer-based biosensors to probe its mechanical functions and biochemical regulation, revealing a switch that toggles vinculin between loadable and unloadable states. Perturbation of the switch causes covarying changes in cell speed and coordination, suggesting alteration of the friction within the system. Molecular scale modelling reveals that increasing levels of loadable vinculin increases friction, due to engagement of self-stabilizing catch bonds. Together, this work reveals a regulatory switch for controlling cell coupling and describes a paradigm for relating biochemical regulation, altered mechanical properties, and changes in cell behaviors.
Collapse
Affiliation(s)
- T. Curtis Shoyer
- Department of Biomedical Engineering, Duke University, Durham, NC27708
| | - Evan M. Gates
- Department of Biomedical Engineering, Duke University, Durham, NC27708
| | - Jolene I. Cabe
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA23284
| | - Aarti N. Urs
- Department of Cell Biology, Duke University, Durham, NC27710
| | - Daniel E. Conway
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH43210
| | - Brenton D. Hoffman
- Department of Biomedical Engineering, Duke University, Durham, NC27708
- Department of Cell Biology, Duke University, Durham, NC27710
| |
Collapse
|
12
|
Nguyen HT, Martin LJ. Classical cadherins in the testis: how are they regulated? Reprod Fertil Dev 2023; 35:641-660. [PMID: 37717581 DOI: 10.1071/rd23084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/31/2023] [Indexed: 09/19/2023] Open
Abstract
Cadherins (CDH) are crucial intercellular adhesion molecules, contributing to morphogenesis and creating tissue barriers by regulating cells' movement, clustering and differentiation. In the testis, classical cadherins such as CDH1, CDH2 and CDH3 are critical to gonadogenesis by promoting the migration and the subsequent clustering of primordial germ cells with somatic cells. While CDH2 is present in both Sertoli and germ cells in rodents, CDH1 is primarily detected in undifferentiated spermatogonia. As for CDH3, its expression is mainly found in germ and pre-Sertoli cells in developing gonads until the establishment of the blood-testis barrier (BTB). This barrier is made of Sertoli cells forming intercellular junctional complexes. The restructuring of the BTB allows the movement of early spermatocytes toward the apical compartment as they differentiate during a process called spermatogenesis. CDH2 is among many junctional proteins participating in this process and is regulated by several pathways. While cytokines promote the disassembly of the BTB by enhancing junctional protein endocytosis for degradation, testosterone facilitates the assembly of the BTB by increasing the recycling of endocytosed junctional proteins. Mitogen-activated protein kinases (MAPKs) are also mediators of the BTB kinetics in many chemically induced damages in the testis. In addition to regulating Sertoli cell functions, follicle stimulating hormone can also regulate the expression of CDH2. In this review, we discuss the current knowledge on regulatory mechanisms of cadherin localisation and expression in the testis.
Collapse
Affiliation(s)
- Ha Tuyen Nguyen
- Biology Department, Université de Moncton, Moncton, NB E1A 3E9, Canada
| | - Luc J Martin
- Biology Department, Université de Moncton, Moncton, NB E1A 3E9, Canada
| |
Collapse
|
13
|
Ruijmbeek CW, Housley F, Idrees H, Housley MP, Pestel J, Keller L, Lai JK, van der Linde HC, Willemsen R, Piesker J, Al-Hassnan ZN, Almesned A, Dalinghaus M, van den Bersselaar LM, van Slegtenhorst MA, Tessadori F, Bakkers J, van Ham TJ, Stainier DY, Verhagen JM, Reischauer S. Biallelic variants in FLII cause pediatric cardiomyopathy by disrupting cardiomyocyte cell adhesion and myofibril organization. JCI Insight 2023; 8:e168247. [PMID: 37561591 PMCID: PMC10544232 DOI: 10.1172/jci.insight.168247] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 07/20/2023] [Indexed: 08/12/2023] Open
Abstract
Pediatric cardiomyopathy (CM) represents a group of rare, severe disorders that affect the myocardium. To date, the etiology and mechanisms underlying pediatric CM are incompletely understood, hampering accurate diagnosis and individualized therapy development. Here, we identified biallelic variants in the highly conserved flightless-I (FLII) gene in 3 families with idiopathic, early-onset dilated CM. We demonstrated that patient-specific FLII variants, when brought into the zebrafish genome using CRISPR/Cas9 genome editing, resulted in the manifestation of key aspects of morphological and functional abnormalities of the heart, as observed in our patients. Importantly, using these genetic animal models, complemented with in-depth loss-of-function studies, we provided insights into the function of Flii during ventricular chamber morphogenesis in vivo, including myofibril organization and cardiomyocyte cell adhesion, as well as trabeculation. In addition, we identified Flii function to be important for the regulation of Notch and Hippo signaling, crucial pathways associated with cardiac morphogenesis and function. Taken together, our data provide experimental evidence for a role for FLII in the pathogenesis of pediatric CM and report biallelic variants as a genetic cause of pediatric CM.
Collapse
Affiliation(s)
- Claudine W.B. Ruijmbeek
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Filomena Housley
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Hafiza Idrees
- Medical Clinic I (Cardiology/Angiology) and Campus Kerckhoff, Justus-Liebig-University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen/Bad Nauheim, Germany
| | - Michael P. Housley
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jenny Pestel
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Leonie Keller
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Jason K.H. Lai
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Herma C. van der Linde
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Janett Piesker
- Scientific Service Group Microscopy, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Zuhair N. Al-Hassnan
- Department of Medical Genetics, and
- Cardiovascular Genetics Program, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | | | - Michiel Dalinghaus
- Department of Pediatric Cardiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Lisa M. van den Bersselaar
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Marjon A. van Slegtenhorst
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Federico Tessadori
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, Netherlands
- Department of Pediatric Cardiology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tjakko J. van Ham
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Didier Y.R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen/Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), RheinMain partner site, Bad Nauheim, Germany
| | - Judith M.A. Verhagen
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Sven Reischauer
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Medical Clinic I (Cardiology/Angiology) and Campus Kerckhoff, Justus-Liebig-University Giessen, Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen/Bad Nauheim, Germany
- German Centre for Cardiovascular Research (DZHK), RheinMain partner site, Bad Nauheim, Germany
| |
Collapse
|
14
|
Zhao S, Gu T, Weng K, Zhang Y, Cao Z, Zhang Y, Zhao W, Chen G, Xu Q. Phosphoproteome Reveals Extracellular Regulated Protein Kinase Phosphorylation Mediated by Mitogen-Activated Protein Kinase Kinase-Regulating Granulosa Cell Apoptosis in Broody Geese. Int J Mol Sci 2023; 24:12278. [PMID: 37569653 PMCID: PMC10418642 DOI: 10.3390/ijms241512278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Geese have strong brooding abilities, which severely affect their egg-laying performance. Phosphorylation is widely involved in regulating reproductive activities, but its role in goose brooding behavior is unclear. In this study, we investigated differences in the phosphoprotein composition of ovarian tissue between laying and brooding geese. Brooding geese exhibited ovarian and follicular atrophy, as well as significant oxidative stress and granulosa cell apoptosis. We identified 578 highly phosphorylated proteins and 281 lowly phosphorylated proteins, and a KEGG pathway analysis showed that these differentially phosphorylated proteins were mainly involved in cell apoptosis, adhesion junctions, and other signaling pathways related to goose brooding behavior. The extracellular regulated protein kinase (ERK)-B-Cell Lymphoma 2(BCL2) signaling pathway was identified as playing an important role in regulating cell apoptosis. The phosphorylation levels of ERK proteins were significantly lower in brooding geese than in laying geese, and the expression of mitogen-activated protein kinase kinase (MEK) was downregulated. Overexpression of MEK led to a significant increase in ERK phosphorylation and BCL2 transcription in H2O2-induced granulosa cells (p < 0.05), partially rescuing cell death. Conversely, granulosa cells receiving MEK siRNA exhibited the opposite trend. In conclusion, geese experience significant oxidative stress and granulosa cell apoptosis during brooding, with downregulated MEK expression, decreased phosphorylation of ERK protein, and inhibited expression of BCL2.
Collapse
Affiliation(s)
- Shuai Zhao
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China; (S.Z.)
| | - Tiantian Gu
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China; (S.Z.)
| | - Kaiqi Weng
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China; (S.Z.)
| | - Yu Zhang
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China; (S.Z.)
| | - Zhengfeng Cao
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China; (S.Z.)
| | - Yang Zhang
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China; (S.Z.)
| | - Wenming Zhao
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China; (S.Z.)
| | - Guohong Chen
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China; (S.Z.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Qi Xu
- Jiangsu Key Laboratory for Animal Genetic, Breeding and Molecular Design, Yangzhou University, Yangzhou 225009, China; (S.Z.)
| |
Collapse
|
15
|
Franz F, Tapia-Rojo R, Winograd-Katz S, Boujemaa-Paterski R, Li W, Unger T, Albeck S, Aponte-Santamaria C, Garcia-Manyes S, Medalia O, Geiger B, Gräter F. Allosteric activation of vinculin by talin. Nat Commun 2023; 14:4311. [PMID: 37463895 DOI: 10.1038/s41467-023-39646-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 06/22/2023] [Indexed: 07/20/2023] Open
Abstract
The talin-vinculin axis is a key mechanosensing component of cellular focal adhesions. How talin and vinculin respond to forces and regulate one another remains unclear. By combining single-molecule magnetic tweezers experiments, Molecular Dynamics simulations, actin-bundling assays, and adhesion assembly experiments in live cells, we here describe a two-ways allosteric network within vinculin as a regulator of the talin-vinculin interaction. We directly observe a maturation process of vinculin upon talin binding, which reinforces the binding to talin at a rate of 0.03 s-1. This allosteric transition can compete with force-induced dissociation of vinculin from talin only at forces up to 10 pN. Mimicking the allosteric activation by mutation yields a vinculin molecule that bundles actin and localizes to focal adhesions in a force-independent manner. Hence, the allosteric switch confines talin-vinculin interactions and focal adhesion build-up to intermediate force levels. The 'allosteric vinculin mutant' is a valuable molecular tool to further dissect the mechanical and biochemical signalling circuits at focal adhesions and elsewhere.
Collapse
Affiliation(s)
- Florian Franz
- Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Mathematikon, INF 205, 69120, Heidelberg, Germany
| | - Rafael Tapia-Rojo
- Department of Physics, Randall Centre for Cell and Molecular Biophysics, Centre for the Physical Science of Life and London Centre for Nanotechnology, King's College London, Strand, WC2R 2LS London, UK.
- Single Molecule Mechanobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, London, UK.
| | - Sabina Winograd-Katz
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Wenhong Li
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Unger
- The Dana and Yossie Hollander Center for Structural Proteomics, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Albeck
- The Dana and Yossie Hollander Center for Structural Proteomics, Weizmann Institute of Science, Rehovot, Israel
| | - Camilo Aponte-Santamaria
- Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Mathematikon, INF 205, 69120, Heidelberg, Germany
| | - Sergi Garcia-Manyes
- Department of Physics, Randall Centre for Cell and Molecular Biophysics, Centre for the Physical Science of Life and London Centre for Nanotechnology, King's College London, Strand, WC2R 2LS London, UK
- Single Molecule Mechanobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, London, UK
| | - Ohad Medalia
- Department of Biochemistry, University of Zurich, 8057, Zurich, Switzerland.
| | - Benjamin Geiger
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Frauke Gräter
- Heidelberg Institute for Theoretical Studies (HITS), Schloß-Wolfsbrunnenweg 35, 69118, Heidelberg, Germany.
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, Mathematikon, INF 205, 69120, Heidelberg, Germany.
- IMSEAM, Heidelberg University, INF 225, 69120, Heidelberg, Germany.
| |
Collapse
|
16
|
Gaffke L, Rintz E, Pierzynowska K, Węgrzyn G. Actin Cytoskeleton Polymerization and Focal Adhesion as Important Factors in the Pathomechanism and Potential Targets of Mucopolysaccharidosis Treatment. Cells 2023; 12:1782. [PMID: 37443816 PMCID: PMC10341097 DOI: 10.3390/cells12131782] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/24/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
The main approach used in the current therapy of mucopolysaccharidosis (MPS) is to reduce the levels of glycosaminoglycans (GAGs) in cells, the deposits considered to be the main cause of the disease. Previous studies have revealed significant differences in the expression of genes encoding proteins involved in many processes, like those related to actin filaments, in MPS cells. Since the regulation of actin filaments is essential for the intracellular transport of specific molecules, the process which may affect the course of MPSs, the aim of this study was to evaluate the changes that occur in the actin cytoskeleton and focal adhesion in cells derived from patients with this disease, as well as in the MPS I mouse model, and to assess whether they could be potential therapeutic targets for different MPS types. Western-blotting, flow cytometry and transcriptomic analyses were employed to address these issues. The levels of the key proteins involved in the studied processes, before and after specific treatment, were assessed. We have also analyzed transcripts whose levels were significantly altered in MPS cells. We identified genes whose expressions were changed in the majority of MPS types and those with particularly highly altered expression. For the first time, significant changes in the expression of genes involved in the actin cytoskeleton structure/functions were revealed which may be considered as an additional element in the pathogenesis of MPSs. Our results suggest the possibility of using the actin cytoskeleton as a potential target in therapeutic approaches for this disease.
Collapse
Affiliation(s)
- Lidia Gaffke
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (E.R.); (K.P.); (G.W.)
| | | | | | | |
Collapse
|
17
|
Benz PM, Frömel T, Laban H, Zink J, Ulrich L, Groneberg D, Boon RA, Poley P, Renne T, de Wit C, Fleming I. Cardiovascular Functions of Ena/VASP Proteins: Past, Present and Beyond. Cells 2023; 12:1740. [PMID: 37443774 PMCID: PMC10340426 DOI: 10.3390/cells12131740] [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: 04/26/2023] [Revised: 06/18/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Actin binding proteins are of crucial importance for the spatiotemporal regulation of actin cytoskeletal dynamics, thereby mediating a tremendous range of cellular processes. Since their initial discovery more than 30 years ago, the enabled/vasodilator-stimulated phosphoprotein (Ena/VASP) family has evolved as one of the most fascinating and versatile family of actin regulating proteins. The proteins directly enhance actin filament assembly, but they also organize higher order actin networks and link kinase signaling pathways to actin filament assembly. Thereby, Ena/VASP proteins regulate dynamic cellular processes ranging from membrane protrusions and trafficking, and cell-cell and cell-matrix adhesions, to the generation of mechanical tension and contractile force. Important insights have been gained into the physiological functions of Ena/VASP proteins in platelets, leukocytes, endothelial cells, smooth muscle cells and cardiomyocytes. In this review, we summarize the unique and redundant functions of Ena/VASP proteins in cardiovascular cells and discuss the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Peter M. Benz
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
| | - Timo Frömel
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Hebatullah Laban
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Joana Zink
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Lea Ulrich
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Dieter Groneberg
- Institute of Physiology I, University of Würzburg, 97070 Würzburg, Germany
| | - Reinier A. Boon
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
- Centre of Molecular Medicine, Institute of Cardiovascular Regeneration, Goethe-University, 60596 Frankfurt am Main, Germany
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Centre, 1081 HZ Amsterdam, The Netherlands
| | - Philip Poley
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Thomas Renne
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, 55131 Mainz, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, D02 VN51 Dublin, Ireland
| | - Cor de Wit
- Institut für Physiologie, Universität zu Lübeck, 23562 Lübeck, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, 23562 Lübeck, Germany
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
- German Centre of Cardiovascular Research (DZHK), Partner Site Rhein-Main, 60596 Frankfurt am Main, Germany
- Cardiopulmonary Institute, 60596 Frankfurt am Main, Germany
| |
Collapse
|
18
|
Shi B, Matsui T, Qian S, Weiss TM, Nicholl ID, Callaway DJE, Bu Z. An ensemble of cadherin-catenin-vinculin complex employs vinculin as the major F-actin binding mode. Biophys J 2023; 122:2456-2474. [PMID: 37147801 PMCID: PMC10323030 DOI: 10.1016/j.bpj.2023.04.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 02/14/2023] [Accepted: 04/27/2023] [Indexed: 05/07/2023] Open
Abstract
The cell-cell adhesion cadherin-catenin complexes recruit vinculin to the adherens junction (AJ) to modulate the mechanical couplings between neighboring cells. However, it is unclear how vinculin influences the AJ structure and function. Here, we identified two patches of salt bridges that lock vinculin in the head-tail autoinhibited conformation and reconstituted the full-length vinculin activation mimetics bound to the cadherin-catenin complex. The cadherin-catenin-vinculin complex contains multiple disordered linkers and is highly dynamic, which poses a challenge for structural studies. We determined the ensemble conformation of this complex using small-angle x-ray and selective deuteration/contrast variation small-angle neutron scattering. In the complex, both α-catenin and vinculin adopt an ensemble of flexible conformations, but vinculin has fully open conformations with the vinculin head and actin-binding tail domains well separated from each other. F-actin binding experiments show that the cadherin-catenin-vinculin complex binds and bundles F-actin. However, when the vinculin actin-binding domain is removed from the complex, only a minor fraction of the complex binds to F-actin. The results show that the dynamic cadherin-catenin-vinculin complex employs vinculin as the primary F-actin binding mode to strengthen AJ-cytoskeleton interactions.
Collapse
Affiliation(s)
- Bright Shi
- Department of Chemistry and Biochemistry, City College of New York, City University of New York (CUNY), New York; PhD Programs in Chemistry and Biochemistry, CUNY Graduate Center, New York
| | - Tsutomu Matsui
- Stanford Synchrotron Radiation Light Source, Menlo Park, California
| | - Shuo Qian
- Second Target Station Project, Oak Ridge National Laboratory, Oak Ridge, Tennessee
| | - Thomas M Weiss
- Stanford Synchrotron Radiation Light Source, Menlo Park, California
| | - Iain D Nicholl
- Department of Biomedical Science and Physiology, Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, United Kingdom
| | - David J E Callaway
- Department of Chemistry and Biochemistry, City College of New York, City University of New York (CUNY), New York.
| | - Zimei Bu
- Department of Chemistry and Biochemistry, City College of New York, City University of New York (CUNY), New York; PhD Programs in Chemistry and Biochemistry, CUNY Graduate Center, New York.
| |
Collapse
|
19
|
Li X, Goult BT, Ballestrem C, Zacharchenko T. The structural basis of the talin-KANK1 interaction that coordinates the actin and microtubule cytoskeletons at focal adhesions. Open Biol 2023; 13:230058. [PMID: 37339751 DOI: 10.1098/rsob.230058] [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: 02/22/2023] [Accepted: 05/26/2023] [Indexed: 06/22/2023] Open
Abstract
Adhesion between cells and the extracellular matrix is mediated by heterodimeric (αβ) integrin receptors that are intracellularly linked to the contractile actomyosin machinery. One of the proteins that control this link is talin, which organizes cytosolic signalling proteins into discrete complexes on β-integrin tails referred to as focal adhesions (FAs). The adapter protein KANK1 binds to talin in the region of FAs known as the adhesion belt. Here, we adapted a non-covalent crystallographic chaperone to resolve the talin-KANK1 complex. This structure revealed that the talin binding KN region of KANK1 contains a novel motif where a β-hairpin stabilizes the α-helical region, explaining both its specific interaction with talin R7 and high affinity. Single point mutants in KANK1 identified from the structure abolished the interaction and enabled us to examine KANK1 enrichment in the adhesion belt. Strikingly, in cells expressing a constitutively active form of vinculin that keeps the FA structure intact even in the presence of myosin inhibitors, KANK1 localizes throughout the entire FA structure even when actomyosin tension is released. We propose a model whereby actomyosin forces on talin eliminate KANK1 from talin binding in the centre of FAs while retaining it at the adhesion periphery.
Collapse
Affiliation(s)
- Xingchen Li
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Dover Street, Manchester M13 9PT, UK
| | | | - Christoph Ballestrem
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Dover Street, Manchester M13 9PT, UK
| | - Thomas Zacharchenko
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Dover Street, Manchester M13 9PT, UK
| |
Collapse
|
20
|
Osuka K, Ohmichi Y, Ohmichi M, Honma S, Suzuki C, Aoyama M, Iwami K, Watanabe Y, Miyachi S. Angiogenesis in the Outer Membrane of Chronic Subdural Hematomas through Thrombin-Cleaved Osteopontin and the Integrin α9 and Integrin β1 Signaling Pathways. Biomedicines 2023; 11:biomedicines11051440. [PMID: 37239111 DOI: 10.3390/biomedicines11051440] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND A chronic subdural hematoma (CSDH) is considered to be an inflammatory and angiogenic disease. The CSDH outer membrane, which contains inflammatory cells, plays an important role in CSDH development. Osteopontin (OPN) is an extracellular matrix protein that is cleaved by thrombin, generating the N-terminal half of OPN, which is prominently involved in integrin signal transduction. We explored the expression of the N-terminal half of OPN in CSDH fluid and the expression of integrins α9 and β1 and the downstream components of the angiogenic signaling pathways in the outer membrane of CSDHs. METHODS Twenty samples of CSDH fluid and eight samples of CSDH outer membrane were collected from patients suffering from CSDHs. The concentrations of the N-terminal half of OPN in CSDH fluid samples were measured using ELISA kits. The expression levels of integrins α9 and β1, vinculin, talin-1, focal adhesion kinase (FAK), paxillin, α-actin, Src and β-actin were examined by Western blot analysis. The expression levels of integrins α9 and β1, FAK and paxillin were also examined by immunohistochemistry. We investigated whether CSDH fluid could activate FAK in cultured endothelial cells in vitro. RESULTS The concentration of the N-terminal half of OPN in CSDH fluid was significantly higher than that in the serum. Western blot analysis confirmed the presence of these molecules. In addition, integrins α9 and β1, FAK and paxillin were localized in the endothelial cells of vessels within the CSDH outer membrane. FAK was significantly phosphorylated immediately after treatment with CSDH fluid. CONCLUSION Our data suggest that the N-terminal half of OPN in CSDH fluid promotes neovascularization in endothelial cells through integrins α9 and β1. The N-terminal half of OPN, which is part of the extracellular matrix, plays a critical role in the promotion of CSDHs.
Collapse
Affiliation(s)
- Koji Osuka
- Department of Neurological Surgery, Aichi Medical University, 1-1 Yazakokarimata, Nagakute 480-1195, Japan
| | - Yusuke Ohmichi
- Department of Anatomy II, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku 920-0293, Japan
| | - Mika Ohmichi
- Department of Anatomy II, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku 920-0293, Japan
| | - Satoru Honma
- Department of Anatomy II, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Kahoku 920-0293, Japan
| | - Chiharu Suzuki
- Department of Neurological Surgery, Aichi Medical University, 1-1 Yazakokarimata, Nagakute 480-1195, Japan
| | - Masahiro Aoyama
- Department of Neurological Surgery, Aichi Medical University, 1-1 Yazakokarimata, Nagakute 480-1195, Japan
| | - Kenichiro Iwami
- Department of Neurological Surgery, Aichi Medical University, 1-1 Yazakokarimata, Nagakute 480-1195, Japan
| | - Yasuo Watanabe
- High Technology Research Center, Pharmacology, Showa Pharmaceutical University, 3-3165 Higashi-Tamagawa Gakuen, Machida 194-8543, Japan
| | - Shigeru Miyachi
- Department of Neurological Surgery, Aichi Medical University, 1-1 Yazakokarimata, Nagakute 480-1195, Japan
| |
Collapse
|
21
|
Cao B, Zhao Y, Luo Q, Chen Y, Xu T, Sun Y. Vinculin B inhibits NF-κB signaling pathway by targeting MyD88 in miiuy croaker, Miichthys miiuy. FISH & SHELLFISH IMMUNOLOGY 2023; 135:108683. [PMID: 36931481 DOI: 10.1016/j.fsi.2023.108683] [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: 11/04/2022] [Revised: 02/14/2023] [Accepted: 03/11/2023] [Indexed: 06/18/2023]
Abstract
Myeloid differentiation factor 88 (MyD88) is the canonical adaptor for inflammatory signaling pathways downstream from members of the Toll-like receptor (TLR) and interleukin-1 (IL-1) receptor families, which activates the NF-κB signaling pathway and regulates immune and inflammatory responses. In this study, we found that Vinculin B (Vclb) is an inhibitor in the NF-κB signaling pathway, and its inhibitory effect was enhanced by LPS induction. Furthermore, Vclb inhibits NF-κB activation by targeting MyD88, thereby suppressing the production of inflammatory cytokines. Mechanistically, Vclb inhibits the NF-κB signaling pathway by targeting MyD88 ubiquitin-proteasome pathway. In summary, our study reveals that Vclb inhibits NF-κB signaling activation and mediates innate immunity in teleosts via the ubiquitin-proteasome pathway of MyD88.
Collapse
Affiliation(s)
- Baolan Cao
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Yan Zhao
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Qiang Luo
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Ya Chen
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Tianjun Xu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.
| | - Yuena Sun
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China; National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, China.
| |
Collapse
|
22
|
Estep JA, Sun LO, Riccomagno MM. A luciferase fragment complementation assay to detect focal adhesion kinase (FAK) signaling events. Heliyon 2023; 9:e15282. [PMID: 37089315 PMCID: PMC10119766 DOI: 10.1016/j.heliyon.2023.e15282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 03/29/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023] Open
Abstract
Integrin Adhesion Complexes (IACs) serve as links between the cytoskeleton and extracellular environment, acting as mechanosensing and signaling hubs. As such, IACs participate in many aspects of cellular motility, tissue morphogenesis, anchorage-dependent growth and cell survival. Focal Adhesion Kinase (FAK) has emerged as a critical organizer of IAC signaling events due to its early recruitment and diverse substrates, and thus has become a genetic and therapeutic target. Here we present the design and characterization of simple, reversible, and scalable Bimolecular Complementation sensors to monitor FAK phosphorylation in living cells. These probes provide novel means to quantify IAC signaling, expanding on the currently available toolkit for interrogating FAK phosphorylation during diverse cellular processes.
Collapse
Affiliation(s)
- Jason A. Estep
- Cell, Molecular and Developmental Biology Program, Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA 92521, USA
| | - Lu O. Sun
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Martin M. Riccomagno
- Cell, Molecular and Developmental Biology Program, Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA 92521, USA
- Neuroscience Program, Department of Molecular, Cell and Systems Biology, University of California, Riverside, CA 92521, USA
| |
Collapse
|
23
|
Murugesan M, Kandhavelu M, Thiyagarajan R, Natesan S, Rajendran P, Murugesan A. Marine halophyte derived polyphenols inhibit glioma cell growth through mitogen-activated protein kinase signaling pathway. Biomed Pharmacother 2023; 159:114288. [PMID: 36682245 DOI: 10.1016/j.biopha.2023.114288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/18/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Plants that are pharmacologically significant require intensive phytochemical characterization for bioactive profiling of the compounds, which has enabled their safe use in ayurvedic medicine. The present study is focused on the phytochemical analyses, quantitative estimation and profiling of secondary metabolites of leaf extract, as well as the antioxidant and cytotoxic activity of the potent halophytes such as Avicennia marina, Ceriops tagal, Ipomoea pes-caprae, and Sonneratia apetala. The in vitro antioxidant property was investigated using DPPH, ferric reducing antioxidant capacity (FRAP) assay. Bioactive compounds such as phenols, flavonoids, saponin and alkaloids were quantitatively estimated from the extracts of A.marina, C.tagal, I.pes-capra and S.apetala, which possessed higher phenol content than the other studied halophytes. The extracts at 200 µg/ml revealed higher antioxidant activity than the standard ascorbic acid and it functions as a powerful oxygen free radical scavenger with 77.37%, 75.35% and 72.84% for S.apetala, I.pes-caprae and C.tagal respectively and with least IC50 for I.pes-caprae (11.95 µg/ml) followed by C.tagal (49.94 µg/ml). Cell viability and anti-proliferative activity of different polyphenolic fractions of C.tagal (CT1 and CT2) and I.pes-caprae fraction (IP) against LN229, SNB19 revealed Ipomoea as the promising anti-cytotoxic fraction. IP-derived polyphenols was further subjected to apoptosis, migration assay, ROS and caspase - 3 and - 7 to elucidate its potentiality as a therapeutic drug. IP-polyphenols was found to have higher percentage of inhibition than the CT1 and CT2 polyphenols of C.tagal on comparison with TMZ. All the above-mentioned in-vitro analysis further validated the ability of IP-polyphenols inducing cell death via ROS-mediated caspase dependent pathway. Further, proteomic and phospho-proteomic analysis revealed the potential role of IP-polyphenols in the regulation of cell proliferation through MMK3, p53, p70 S6 kinase and RSK1 proteins involved in mitogen-activated protein kinase signaling pathway. Our analysis confirmed the promising role of I.pes-caprae derived polyphenols as an anti-metastatic compound against GBM cells.
Collapse
Affiliation(s)
- Monica Murugesan
- Department of Zoology, Lady Doak College, Madurai Kamaraj University, Thallakulam, Madurai 625002, India
| | - Meenakshisundaram Kandhavelu
- BioMeditech and Tays Cancer Center, Tampere University Hospital, P.O. Box 553, 33101 Tampere, Finland; Molecular Signaling Group, Faculty of Medicine and Health Technology, Tampere University, P.O. Box 553, 33101 Tampere, Finland.
| | - Ramesh Thiyagarajan
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Kingdom of Saudi Arabia
| | - Sankar Natesan
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai 625021, India
| | - Priyatharsini Rajendran
- Department of Zoology, Lady Doak College, Madurai Kamaraj University, Thallakulam, Madurai 625002, India
| | - Akshaya Murugesan
- Department of Biotechnology, Lady Doak College, Madurai Kamaraj University, Thallakulam, Madurai 625002, India.
| |
Collapse
|
24
|
Essa A, Essa ES, El-deeb SM, Seleem HEM, Al Sahlawi M, Al-Omair OA, Shehab-Eldeen S. Elevated Serum Vinculin in Patients with HBV/HCV-Associated Liver Cirrhosis and Hepatocellular Carcinoma: A Pilot Study. Biologics 2023; 17:23-32. [PMID: 36969330 PMCID: PMC10035354 DOI: 10.2147/btt.s405500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/04/2023] [Indexed: 03/20/2023]
Abstract
Background The stiffness of the extracellular matrix (ECM) controls many cellular processes, such as migration and differentiation. Cells detect stiffness through adhesion structures termed focal adhesions (FAs). Vinculin, an actin-binding FA protein, plays a pivotal role in FA-mediated mechanotransduction. Aim This study aimed to explore the role of vinculin in the development of HBV/HCV-induced hepatocellular carcinoma (HCC). Methods Vinculin levels in a total number of 100 serum samples from patients with HBV/HCV-induced liver cirrhosis and HCC, as well as healthy controls, were analyzed using an enzyme-linked immunosorbent assay (ELISA). Results In patients with HCC and liver cirrhosis, the serum vinculin levels were significantly greater than in controls (503.8±242.2 and 728.4±1044.8 vs 77.7±36.1 respectively, p<0.001). However, results showed no link between serum vinculin and the clinicopathological features of HCC. Conclusion Patients with HBVor HCV-induced liver cirrhosis and HCC have significantly higher serum levels of vinculin than do controls. This might point to a potential role for vinculin in the development of HCC. More research into how this protein affects the development of HCC at the molecular level could lead to better clinical treatments and the development of new molecular therapies.
Collapse
Affiliation(s)
- Abdallah Essa
- Tropical Medicine Department, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
- Internal Medicine Department, College of Medicine, King Faisal University, Al-Ahsa, Kingdom of Saudi Arabia
| | - Enas Said Essa
- Clinical Pathology Department, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
| | - Sara Mahmoud El-deeb
- Clinical Pathology Department, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
| | | | - Muthana Al Sahlawi
- Internal Medicine Department, College of Medicine, King Faisal University, Al-Ahsa, Kingdom of Saudi Arabia
| | - Omar Ahmed Al-Omair
- Internal Medicine Department, College of Medicine, King Faisal University, Al-Ahsa, Kingdom of Saudi Arabia
| | - Somaia Shehab-Eldeen
- Tropical Medicine Department, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
- Internal Medicine Department, College of Medicine, King Faisal University, Al-Ahsa, Kingdom of Saudi Arabia
- Correspondence: Somaia Shehab-Eldeen, Tropical Medicine Department, Faculty of Medicine, Menoufia University, Yassen Abd Al Ghafar Street, Shebin Elkom, Menoufia Governorate, 32511, Egypt, Tel +201117251523, Email
| |
Collapse
|
25
|
Sheppard L, Green DG, Lerchbaumer G, Rothenberg KE, Fernandez-Gonzalez R, Tepass U. The α-Catenin mechanosensing M region is required for cell adhesion during tissue morphogenesis. J Cell Biol 2023; 222:e202108091. [PMID: 36520419 PMCID: PMC9757846 DOI: 10.1083/jcb.202108091] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/08/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
α-Catenin couples the cadherin-catenin complex to the actin cytoskeleton. The mechanosensitive α-Catenin M region undergoes conformational changes upon application of force to recruit interaction partners. Here, we took advantage of the tension landscape in the Drosophila embryo to define three different states of α-Catenin mechanosensing in support of cell adhesion. Low-, medium-, and high-tension contacts showed a corresponding recruitment of Vinculin and Ajuba, which was dependent on the α-Catenin M region. In contrast, the Afadin homolog Canoe acts in parallel to α-Catenin at bicellular low- and medium-tension junctions but requires an interaction with α-Catenin for its tension-sensitive enrichment at high-tension tricellular junctions. Individual M region domains make complex contributions to cell adhesion through their impact on interaction partner recruitment, and redundancies with the function of Canoe. Our data argue that α-Catenin and its interaction partners are part of a cooperative and partially redundant mechanoresponsive network that supports AJs remodeling during morphogenesis.
Collapse
Affiliation(s)
- Luka Sheppard
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - David G. Green
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Gerald Lerchbaumer
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| | - Katheryn E. Rothenberg
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, Canada
- Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Ulrich Tepass
- Department of Cell and Systems Biology, University of Toronto, Toronto, Canada
| |
Collapse
|
26
|
Cardiac Differentiation Promotes Focal Adhesions Assembly through Vinculin Recruitment. Int J Mol Sci 2023; 24:ijms24032444. [PMID: 36768766 PMCID: PMC9916732 DOI: 10.3390/ijms24032444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/19/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Cells of the cardiovascular system are physiologically exposed to a variety of mechanical forces fundamental for both cardiac development and functions. In this context, forces generated by actomyosin networks and those transmitted through focal adhesion (FA) complexes represent the key regulators of cellular behaviors in terms of cytoskeleton dynamism, cell adhesion, migration, differentiation, and tissue organization. In this study, we investigated the involvement of FAs on cardiomyocyte differentiation. In particular, vinculin and focal adhesion kinase (FAK) family, which are known to be involved in cardiac differentiation, were studied. Results revealed that differentiation conditions induce an upregulation of both FAK-Tyr397 and vinculin, resulting also in the translocation to the cell membrane. Moreover, the role of mechanical stress in contractile phenotype expression was investigated by applying a uniaxial mechanical stretching (5% substrate deformation, 1 Hz frequency). Morphological evaluation revealed that the cell shape showed a spindle shape and reoriented following the stretching direction. Substrate deformation resulted also in modification of the length and the number of vinculin-positive FAs. We can, therefore, suggest that mechanotransductive pathways, activated through FAs, are highly involved in cardiomyocyte differentiation, thus confirming their role during cytoskeleton rearrangement and cardiac myofilament maturation.
Collapse
|
27
|
Manso AM, Romaine A, Christensen G, Ross RS. Integrins in Cardiac Form, Function, and Disease. BIOLOGY OF EXTRACELLULAR MATRIX 2023:135-183. [DOI: 10.1007/978-3-031-23781-2_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
28
|
Tu Y, Pal K, Austin J, Wang X. Filopodial adhesive force in discrete nodes revealed by integrin molecular tension imaging. Curr Biol 2022; 32:4386-4396.e3. [PMID: 36084647 PMCID: PMC9613586 DOI: 10.1016/j.cub.2022.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/30/2022] [Accepted: 08/15/2022] [Indexed: 10/14/2022]
Abstract
Filopodia are narrow cell extensions involved in various physiological processes. Integrins mediate filopodia adhesion and likely transmit adhesive force to regulate filopodia formation and functions, but the force is extremely weak to study and remains poorly understood. Using integrative tension sensor (ITS), we imaged filopodia adhesive force at the single molecular tension level and investigated the force dynamics and sources. Results show that filopodia integrin tension (FIT) is generated in discrete foci (force nodes) along single filopodia with a spacing of ∼1 μm. Inhibitions of actin polymerization or myosin II activity markedly reduced FIT signals in force nodes at filopodia tips and at filopodia bases, respectively, suggesting differential force sources of FIT in the distal force nodes and proximal ones in filopodia. Using two ITS constructs with different force thresholds for activation, we showed that the molecular force level of FIT is greater at filopodia bases than that at filopodia tips. We also tested the role of vinculin and myosin X in the FIT transmission. With vinculin knockout in cells, filopodia and associated force nodes were still formed normally, suggesting that vinculin is dispensable for the formation of filopodia and force nodes. However, vinculin is indeed required for the transmission of strong FIT (capable of rupturing DNA in a shear conformation), as the strong FIT vanished in filopodia with vinculin knockout. Co-imaging of FIT and myosin X shows no apparent co-localization, demonstrating that myosin X is not directly responsible for generating FIT, despite its prominent role in filopodium elongation.
Collapse
Affiliation(s)
- Ying Tu
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA
| | - Kaushik Pal
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA
| | - Jacob Austin
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA
| | - Xuefeng Wang
- Department of Physics and Astronomy, Iowa State University, Ames, IA 50011, USA; Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA 50011, USA.
| |
Collapse
|
29
|
Bohere J, Eldridge-Thomas BL, Kolahgar G. Vinculin recruitment to α-catenin halts the differentiation and maturation of enterocyte progenitors to maintain homeostasis of the Drosophila intestine. eLife 2022; 11:e72836. [PMID: 36269226 PMCID: PMC9586559 DOI: 10.7554/elife.72836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/11/2022] [Indexed: 11/23/2022] Open
Abstract
Mechanisms communicating changes in tissue stiffness and size are particularly relevant in the intestine because it is subject to constant mechanical stresses caused by peristalsis of its variable content. Using the Drosophila intestinal epithelium, we investigate the role of vinculin, one of the best characterised mechanoeffectors, which functions in both cadherin and integrin adhesion complexes. We discovered that vinculin regulates cell fate decisions, by preventing precocious activation and differentiation of intestinal progenitors into absorptive cells. It achieves this in concert with α-catenin at sites of cadherin adhesion, rather than as part of integrin function. Following asymmetric division of the stem cell into a stem cell and an enteroblast (EB), the two cells initially remain connected by adherens junctions, where vinculin is required, only on the EB side, to maintain the EB in a quiescent state and inhibit further divisions of the stem cell. By manipulating cell tension, we show that vinculin recruitment to adherens junction regulates EB activation and numbers. Consequently, removing vinculin results in an enlarged gut with improved resistance to starvation. Thus, mechanical regulation at the contact between stem cells and their progeny is used to control tissue cell number.
Collapse
Affiliation(s)
- Jerome Bohere
- Department of Physiology, Development and Neuroscience, Downing St, University of CambridgeCambridgeUnited Kingdom
| | - Buffy L Eldridge-Thomas
- Department of Physiology, Development and Neuroscience, Downing St, University of CambridgeCambridgeUnited Kingdom
| | - Golnar Kolahgar
- Department of Physiology, Development and Neuroscience, Downing St, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
30
|
Atherton P, Konstantinou R, Neo SP, Wang E, Balloi E, Ptushkina M, Bennett H, Clark K, Gunaratne J, Critchley D, Barsukov I, Manser E, Ballestrem C. Tensin3 interaction with talin drives the formation of fibronectin-associated fibrillar adhesions. J Biophys Biochem Cytol 2022; 221:213452. [PMID: 36074065 PMCID: PMC9462884 DOI: 10.1083/jcb.202107022] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 05/30/2022] [Accepted: 08/05/2022] [Indexed: 12/11/2022] Open
Abstract
The formation of healthy tissue involves continuous remodeling of the extracellular matrix (ECM). Whilst it is known that this requires integrin-associated cell-ECM adhesion sites (CMAs) and actomyosin-mediated forces, the underlying mechanisms remain unclear. Here, we examine how tensin3 contributes to the formation of fibrillar adhesions (FBs) and fibronectin fibrillogenesis. Using BioID mass spectrometry and a mitochondrial targeting assay, we establish that tensin3 associates with the mechanosensors such as talin and vinculin. We show that the talin R11 rod domain binds directly to a helical motif within the central intrinsically disordered region (IDR) of tensin3, whilst vinculin binds indirectly to tensin3 via talin. Using CRISPR knock-out cells in combination with defined tensin3 mutations, we show (i) that tensin3 is critical for the formation of α5β1-integrin FBs and for fibronectin fibrillogenesis, and (ii) the talin/tensin3 interaction drives this process, with vinculin acting to potentiate it.
Collapse
Affiliation(s)
- Paul Atherton
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.,Division of Cell Biology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rafaella Konstantinou
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK.,sGSK Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Suat Peng Neo
- Quantitative Proteomics Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Emily Wang
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Eleonora Balloi
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Marina Ptushkina
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| | - Hayley Bennett
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Kath Clark
- Department of Biochemistry, University of Leicester, Leicester, UK
| | - Jayantha Gunaratne
- Quantitative Proteomics Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - David Critchley
- Department of Biochemistry, University of Leicester, Leicester, UK
| | - Igor Barsukov
- Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Edward Manser
- sGSK Group, Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | - Christoph Ballestrem
- Wellcome Trust Centre for Cell-Matrix Research, University of Manchester, Manchester, UK
| |
Collapse
|
31
|
Ibrahim NH, Fawzy IM, Gouda TM, El Sayed RAH, Morsi MH, Sabry ASM, Hashaad NI. Anti-vinculin antibodies as a novel biomarker in Egyptian patients with systemic sclerosis. Clin Rheumatol 2022; 41:3401-3409. [PMID: 35876914 PMCID: PMC9568454 DOI: 10.1007/s10067-022-06301-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 11/27/2022]
Abstract
INTRODUCTION Systemic sclerosis (SSc) is an autoimmune disorder that causes vasculopathy and scarring, most commonly in the lungs and skin, but it can also affect other organs. Endothelial vinculin plays a critical role in angiogenesis regulation. Therefore, vinculin overexpression in SSc may give rise to anti-vinculin antibodies, which may contribute to the development of SSc vasculopathy. The current research aims to (1) determine whether anti-vinculin autoantibodies play a significant role in the diagnosis of SSc and (2) compare anti-vinculin serum levels between two scleroderma patient populations, namely, pulmonary artery hypertension (PAH)-predominant and interstitial pulmonary fibrosis (IPF)-predominant groups. METHODS This research included 140 participants categorized into three groups: group I-patients with PAH-predominant; group II-patients with ILD-predominant; group III-the control group. Anti-vinculin antibodies were detected in serum samples collected from all participants using ELISA. All subjects underwent high-resolution computed tomography (CT), diffusing capacity for carbon monoxide, and pulmonary function tests. RESULTS Patients in group I (PAH-predominant group, N = 35) were 41.3 [± 11.4] years old, with 80% being women. Patients in group II (ILD-predominant group, N = 35) were 41.0 [± 11.5] years old. The SSc group showed significantly higher anti-vinculin antibody levels than the control group (P < 0.001). The PAH-predominant group demonstrated significantly higher anti-vinculin antibody levels and anti-vinculin positivity than the ILD-predominant group. CONCLUSION Anti-vinculin antibodies in the blood appear to be diagnostic biomarkers for scleroderma. Furthermore, they shed light on some novel perspectives on the pathophysiology of specific lung fibrotic changes. Key Points • This study included two groups of systemic sclerosis patients (PAH-predominant group, ILD-predominant group) as well as a control group to investigate the significance of anti-vinculin antibodies in such cases. • Our results have demonstrated that anti-vinculin antibodies can play a significant role in diagnosing and monitoring systemic sclerosis disease.
Collapse
Affiliation(s)
- Noha Hosni Ibrahim
- Faculty of Medicine, Benha University, Benha, Al-Qalyubia Governorate, Egypt.
| | | | | | | | - Maha Hosni Morsi
- Faculty of Applied Health Sciences Technology, Misr University for Sciences and Technology, 6Th of October City, Giza, Egypt
| | | | | |
Collapse
|
32
|
Focal Adhesion Protein Vinculin Is Required for Proper Meiotic Progression during Mouse Spermatogenesis. Cells 2022; 11:cells11132013. [PMID: 35805097 PMCID: PMC9265697 DOI: 10.3390/cells11132013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/17/2022] Open
Abstract
The focal adhesion protein Vinculin (VCL) is ascribed to various cytoplasmic functions; however, its nuclear role has so far been ambiguous. We observed that VCL localizes to the nuclei of mouse primary spermatocytes undergoing first meiotic division. Specifically, VCL localizes along the meiosis-specific structure synaptonemal complex (SC) during prophase I and the centromeric regions, where it remains until metaphase I. To study the role of VCL in meiotic division, we prepared a conditional knock-out mouse (VCLcKO). We found that the VCLcKO male mice were semi-fertile, with a decreased number of offspring compared to wild-type animals. This study of events in late prophase I indicated premature splitting of homologous chromosomes, accompanied by an untimely loss of SCP1. This caused erroneous kinetochore formation, followed by failure of the meiotic spindle assembly and metaphase I arrest. To assess the mechanism of VCL involvement in meiosis, we searched for its possible interacting partners. A mass spectrometry approach identified several putative interactors which belong to the ubiquitin–proteasome pathway (UPS). The depletion of VLC leads to the dysregulation of a key subunit of the proteasome complex in the meiotic nuclei and an altered nuclear SUMOylation level. Taken together, we show for the first time the presence of VCL in the nucleus of spermatocytes and its involvement in proper meiotic progress. It also suggests the direction for future studies regarding the role of VCL in spermatogenesis through regulation of UPS.
Collapse
|
33
|
Mukherjee A, Melamed S, Damouny-Khoury H, Amer M, Feld L, Nadjar-Boger E, Sheetz MP, Wolfenson H. α-Catenin links integrin adhesions to F-actin to regulate ECM mechanosensing and rigidity dependence. J Cell Biol 2022; 221:213257. [PMID: 35652786 PMCID: PMC9166284 DOI: 10.1083/jcb.202102121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 12/22/2021] [Accepted: 05/16/2022] [Indexed: 02/03/2023] Open
Abstract
Both cell-cell and cell-matrix adhesions are regulated by mechanical signals, but the mechanobiological processes that mediate the cross talk between these structures are poorly understood. Here we show that α-catenin, a mechanosensitive protein that is classically linked with cadherin-based adhesions, associates with and regulates integrin adhesions. α-Catenin is recruited to the edges of mesenchymal cells, where it interacts with F-actin. This is followed by mutual retrograde flow of α-catenin and F-actin from the cell edge, during which α-catenin interacts with vinculin within integrin adhesions. This interaction affects adhesion maturation, stress-fiber assembly, and force transmission to the matrix. In epithelial cells, α-catenin is present in cell-cell adhesions and absent from cell-matrix adhesions. However, when these cells undergo epithelial-to-mesenchymal transition, α-catenin transitions to the cell edge, where it facilitates proper mechanosensing. This is highlighted by the ability of α-catenin-depleted cells to grow on soft matrices. These results suggest a dual role of α-catenin in mechanosensing, through both cell-cell and cell-matrix adhesions.
Collapse
Affiliation(s)
- Abhishek Mukherjee
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Shay Melamed
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Hana Damouny-Khoury
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Malak Amer
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Lea Feld
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Elisabeth Nadjar-Boger
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel
| | - Michael P. Sheetz
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion—Israel Institute of Technology, Haifa, Israel,Correspondence to Haguy Wolfenson:
| |
Collapse
|
34
|
Omelchenko T. Cellular protrusions in 3D: Orchestrating early mouse embryogenesis. Semin Cell Dev Biol 2022; 129:63-74. [PMID: 35577698 DOI: 10.1016/j.semcdb.2022.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/04/2022] [Accepted: 05/04/2022] [Indexed: 12/26/2022]
Abstract
Cellular protrusions generated by the actin cytoskeleton are central to the process of building the body of the embryo. Problems with cellular protrusions underlie human diseases and syndromes, including implantation defects and pregnancy loss, congenital birth defects, and cancer. Cells use protrusive activity together with actin-myosin contractility to create an ordered body shape of the embryo. Here, I review how actin-rich protrusions are used by two major morphological cell types, epithelial and mesenchymal cells, during collective cell migration to sculpt the mouse embryo body. Pre-gastrulation epithelial collective migration of the anterior visceral endoderm is essential for establishing the anterior-posterior body axis. Gastrulation mesenchymal collective migration of the mesoderm wings is crucial for body elongation, and somite and heart formation. Analysis of mouse mutants with disrupted cellular protrusions revealed the key role of protrusions in embryonic morphogenesis and embryo survival. Recent technical approaches have allowed examination of the mechanisms that control cell and tissue movements in vivo in the complex 3D microenvironment of living mouse embryos. Advancing our understanding of protrusion-driven morphogenesis should provide novel insights into human developmental disorders and cancer metastasis.
Collapse
Affiliation(s)
- Tatiana Omelchenko
- Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, 1230 York Avenue, New York 10065, USA.
| |
Collapse
|
35
|
Yao L, Brice R, Shippy T. A Protein Composite Neural Scaffold Modulates Astrocyte Migration and Transcriptome Profile. Macromol Biosci 2022; 22:e2100406. [PMID: 35014754 PMCID: PMC9012687 DOI: 10.1002/mabi.202100406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/23/2021] [Indexed: 11/09/2022]
Abstract
Bioscaffold implantation is a promising approach to facilitate the repair and regeneration of wounded neural tissue after injury to the spinal cord or peripheral nerves. However, such bioscaffold grafts currently result in only limited functional recovery. The generation of a neural scaffold using a combination of collagen and glutenin is reported. The conduit material and mechanical properties, as well as its effect on astrocyte behavior is tested. After neural injuries, astrocytes move into the lesion and participate in the process of remodeling the micro-architecture of the wounded neural tissue. In this study, human astrocytes grown on glutenin-collagen scaffolds show higher motility and a lower proliferation rate compared with those grown on collagen scaffolds. RNA sequencing reveals that astrocytes grown on the two types of scaffolds show differentially expressed genes in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways such as actin cytoskeleton and focal adhesion that regulate astrocyte migration on scaffolds. The gene expression of aggrecan and versican, chondroitin sulfate proteoglycans that inhibit axonal growth, is down-regulated in astrocytes grown on glutenin-collagen scaffolds. These outcomes indicate that the implantation of glutenin-collagen scaffolds may promote astrocyte function in the neural regeneration process by enhanced cell migration and reduced glial scar formation.
Collapse
Affiliation(s)
- Li Yao
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, KS, 67260, USA
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Ryan Brice
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, KS, 67260, USA
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Teresa Shippy
- Department of Biological Sciences, Wichita State University, 1845 Fairmount Street, Wichita, KS, 67260, USA
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| |
Collapse
|
36
|
Fedeli V, Cucina A, Dinicola S, Fabrizi G, Catizone A, Gesualdi L, Ceccarelli S, Harrath AH, Alwasel SH, Ricci G, Pedata P, Bizzarri M, Monti N. Microgravity Modifies the Phenotype of Fibroblast and Promotes Remodeling of the Fibroblast-Keratinocyte Interaction in a 3D Co-Culture Model. Int J Mol Sci 2022; 23:ijms23042163. [PMID: 35216279 PMCID: PMC8879576 DOI: 10.3390/ijms23042163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/11/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
Microgravity impairs tissue organization and critical pathways involved in the cell–microenvironment interplay, where fibroblasts have a critical role. We exposed dermal fibroblasts to simulated microgravity by means of a Random Positioning Machine (RPM), a device that reproduces conditions of weightlessness. Molecular and structural changes were analyzed and compared to control samples growing in a normal gravity field. Simulated microgravity impairs fibroblast conversion into myofibroblast and inhibits their migratory properties. Consequently, the normal interplay between fibroblasts and keratinocytes were remarkably altered in 3D co-culture experiments, giving rise to several ultra-structural abnormalities. Such phenotypic changes are associated with down-regulation of α-SMA that translocate in the nucleoplasm, altogether with the concomitant modification of the actin-vinculin apparatus. Noticeably, the stress associated with weightlessness induced oxidative damage, which seemed to concur with such modifications. These findings disclose new opportunities to establish antioxidant strategies that counteract the microgravity-induced disruptive effects on fibroblasts and tissue organization.
Collapse
Affiliation(s)
- Valeria Fedeli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (V.F.); (S.C.); (M.B.)
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy; (S.D.); (G.F.)
| | - Alessandra Cucina
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, Via Antonio Scarpa 14, 00161 Rome, Italy;
- Policlinico Umberto I, Viale del Policlinico 155, 00161 Rome, Italy
| | - Simona Dinicola
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy; (S.D.); (G.F.)
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, Via Antonio Scarpa 14, 00161 Rome, Italy;
| | - Gianmarco Fabrizi
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy; (S.D.); (G.F.)
- Department of Surgery “Pietro Valdoni”, Sapienza University of Rome, Via Antonio Scarpa 14, 00161 Rome, Italy;
| | - Angela Catizone
- Section of Histology and Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (A.C.); (L.G.)
| | - Luisa Gesualdi
- Section of Histology and Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Viale Regina Elena 336, 00161 Rome, Italy; (A.C.); (L.G.)
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (V.F.); (S.C.); (M.B.)
| | - Abdel Halim Harrath
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.H.H.); (S.H.A.)
| | - Saleh H. Alwasel
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.H.H.); (S.H.A.)
| | - Giulia Ricci
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy;
| | - Paola Pedata
- Department of Medicine, University of Salerno, Via Giovanni Paolo II 132, 84084 Salerno, Italy;
| | - Mariano Bizzarri
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (V.F.); (S.C.); (M.B.)
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy; (S.D.); (G.F.)
| | - Noemi Monti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (V.F.); (S.C.); (M.B.)
- Systems Biology Group Laboratory, Sapienza University, 00161 Rome, Italy; (S.D.); (G.F.)
- Correspondence:
| |
Collapse
|
37
|
Bang ML, Bogomolovas J, Chen J. Understanding the molecular basis of cardiomyopathy. Am J Physiol Heart Circ Physiol 2022; 322:H181-H233. [PMID: 34797172 PMCID: PMC8759964 DOI: 10.1152/ajpheart.00562.2021] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 02/03/2023]
Abstract
Inherited cardiomyopathies are a major cause of mortality and morbidity worldwide and can be caused by mutations in a wide range of proteins located in different cellular compartments. The present review is based on Dr. Ju Chen's 2021 Robert M. Berne Distinguished Lectureship of the American Physiological Society Cardiovascular Section, in which he provided an overview of the current knowledge on the cardiomyopathy-associated proteins that have been studied in his laboratory. The review provides a general summary of the proteins in different compartments of cardiomyocytes associated with cardiomyopathies, with specific focus on the proteins that have been studied in Dr. Chen's laboratory.
Collapse
Affiliation(s)
- Marie-Louise Bang
- Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Milan Unit, Milan, Italy
- IRCCS Humanitas Research Hospital, Rozzano (Milan), Italy
| | - Julius Bogomolovas
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| | - Ju Chen
- Division of Cardiovascular Medicine, Department of Medicine Cardiology, University of California, San Diego, La Jolla, California
| |
Collapse
|
38
|
Complete Model of Vinculin Suggests the Mechanism of Activation by Helical Super-Bundle Unfurling. Protein J 2022; 41:55-70. [PMID: 35006498 DOI: 10.1007/s10930-022-10040-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2022] [Indexed: 12/24/2022]
Abstract
To shed light onto the activation mechanism of vinculin, we carried out a detailed refinement of chicken vinculin and compared it to the human protein which is greater than 95% identical. Refinement resulted in a complete and significantly improved model. This model includes important elements such as a pro-rich strap region (PRR) and C-terminus. The conformation of the PRR stabilized by its inter- and intra-molecular contacts shows a dynamic, but relatively stable motif that constitutes a docking platform for multiple molecules. The contact of the C-terminus with the PRR suggests that phosphorylation of Tyr1065 might control activation and membrane binding. Improved electron densities showed the presence of large solvent molecules such as phosphates/sulfates and a head-group of PIP2. The improved model allowed for a computational stability analysis to be performed by the program Corex/Best which located numerous hot-spots of increased and decreased stability. Proximity of the identified binding sites for regulatory partners involved in inducing or suppressing the activation of vinculin to the unstable elements sheds new light onto the activation pathway and differential activation. This stability analysis suggests that the activation pathway proceeds by unfurling of the super-bundle built from four bundles of helices without separation of the Vt region (840-1066) from the head. According to our mechanism, when activating proteins bind at the strap region a separation of N and C terminal bundles occurs, followed by unfurling of the super-bundle and flattening of the general shape of the molecule, which exposes the interaction sites for binding of auxiliary proteins.
Collapse
|
39
|
Petzold J, Gentleman E. Intrinsic Mechanical Cues and Their Impact on Stem Cells and Embryogenesis. Front Cell Dev Biol 2021; 9:761871. [PMID: 34820380 PMCID: PMC8606660 DOI: 10.3389/fcell.2021.761871] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 10/14/2021] [Indexed: 12/25/2022] Open
Abstract
Although understanding how soluble cues direct cellular processes revolutionised the study of cell biology in the second half of the 20th century, over the last two decades, new insights into how mechanical cues similarly impact cell fate decisions has gained momentum. During development, extrinsic cues such as fluid flow, shear stress and compressive forces are essential for normal embryogenesis to proceed. Indeed, both adult and embryonic stem cells can respond to applied forces, but they can also detect intrinsic mechanical cues from their surrounding environment, such as the stiffness of the extracellular matrix, which impacts differentiation and morphogenesis. Cells can detect changes in their mechanical environment using cell surface receptors such as integrins and focal adhesions. Moreover, dynamic rearrangements of the cytoskeleton have been identified as a key means by which forces are transmitted from the extracellular matrix to the cell and vice versa. Although we have some understanding of the downstream mechanisms whereby mechanical cues are translated into changes in cell behaviour, many of the signalling pathways remain to be defined. This review discusses the importance of intrinsic mechanical cues on adult cell fate decisions, the emerging roles of cell surface mechano-sensors and the cytoskeleton in enabling cells to sense its microenvironment, and the role of intracellular signalling in translating mechanical cues into transcriptional outputs. In addition, the contribution of mechanical cues to fundamental processes during embryogenesis such as apical constriction and convergent extension is discussed. The continued development of tools to measure the biomechanical properties of soft tissues in vivo is likely to uncover currently underestimated contributions of these cues to adult stem cell fate decisions and embryogenesis, and may inform on regenerative strategies for tissue repair.
Collapse
Affiliation(s)
- Jonna Petzold
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London, United Kingdom
| |
Collapse
|
40
|
Legerstee K, Houtsmuller AB. A Layered View on Focal Adhesions. BIOLOGY 2021; 10:biology10111189. [PMID: 34827182 PMCID: PMC8614905 DOI: 10.3390/biology10111189] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/06/2021] [Accepted: 11/08/2021] [Indexed: 12/31/2022]
Abstract
Simple Summary The cytoskeleton is a network of protein fibres within cells that provide structure and support intracellular transport. Focal adhesions are protein complexes associated with the outer cell membrane that are found at the ends of specialised actin fibres of this cytoskeleton. They mediate cell adhesion by connecting the cytoskeleton to the extracellular matrix, a protein and sugar network that surrounds cells in tissues. Focal adhesions also translate forces on actin fibres into forces contributing to cell migration. Cell adhesion and migration are crucial to diverse biological processes such as embryonic development, proper functioning of the immune system or the metastasis of cancer cells. Advances in fluorescence microscopy and data analysis methods provided a more detailed understanding of the dynamic ways in which proteins bind and dissociate from focal adhesions and how they are organised within these protein complexes. In this review, we provide an overview of the advances in the current scientific understanding of focal adhesions and summarize relevant imaging techniques. One of the key insights is that focal adhesion proteins are organised into three layers parallel to the cell membrane. We discuss the relevance of this layered nature for the functioning of focal adhesion. Abstract The cytoskeleton provides structure to cells and supports intracellular transport. Actin fibres are crucial to both functions. Focal Adhesions (FAs) are large macromolecular multiprotein assemblies at the ends of specialised actin fibres linking these to the extracellular matrix. FAs translate forces on actin fibres into forces contributing to cell migration. This review will discuss recent insights into FA protein dynamics and their organisation within FAs, made possible by advances in fluorescence imaging techniques and data analysis methods. Over the last decade, evidence has accumulated that FAs are composed of three layers parallel to the plasma membrane. We focus on some of the most frequently investigated proteins, two from each layer, paxillin and FAK (bottom, integrin signalling layer), vinculin and talin (middle, force transduction layer) and zyxin and VASP (top, actin regulatory layer). Finally, we discuss the potential impact of this layered nature on different aspects of FA behaviour.
Collapse
|
41
|
Leppiniemi J, Mutahir Z, Dulebo A, Mikkonen P, Nuopponen M, Turkki P, Hytönen VP. Avidin-Conjugated Nanofibrillar Cellulose Hydrogel Functionalized with Biotinylated Fibronectin and Vitronectin Promotes 3D Culture of Fibroblasts. Biomacromolecules 2021; 22:4122-4137. [PMID: 34542997 DOI: 10.1021/acs.biomac.1c00579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The future success of physiologically relevant three-dimensional (3D) cell/tissue models is dependent on the development of functional biomaterials, which can provide a well-defined 3D environment instructing cellular behavior. To establish a platform to produce tailored hydrogels, we conjugated avidin (Avd) to anionic nanofibrillar cellulose (aNFC) and demonstrated the use of the resulting Avd-NFC hydrogel for 3D cell culture, where Avd-NFC allows easy functionalization via biotinylated molecules. Avidin was successfully conjugated to nanocellulose and remained functional, as demonstrated by electrophoresis and titration with fluorescent biotin. Rheological analysis indicated that Avd-NFC retained shear-thinning and gel-forming properties. Topological characterization using AFM revealed the preserved fiber structure and confirmed the binding of biotinylated vitronectin (B-VN) on the fiber surface. The 3D cell culture experiments with mouse embryonic fibroblasts demonstrated the performance of Avd-NFC hydrogels functionalized with biotinylated fibronectin (B-FN) and B-VN. Cells cultured in Avd-NFC hydrogels functionalized with B-FN or B-VN formed matured integrin-mediated adhesions, indicated by phosphorylated focal adhesion kinase. We observed significantly higher cell proliferation rates when biotinylated proteins were bound to the Avd-NFC hydrogel compared to cells cultured in Avd-NFC alone, indicating the importance of the presence of adhesive sites for fibroblasts. The versatile Avd-NFC allows the easy functionalization of hydrogels with virtually any biotinylated molecule and may become widely utilized in 3D cell/tissue culture applications.
Collapse
Affiliation(s)
- Jenni Leppiniemi
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, FI-33014 Tampere, Finland
| | - Zeeshan Mutahir
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, FI-33014 Tampere, Finland.,School of Biochemistry and Biotechnology, University of the Punjab, 54590 Lahore, Pakistan
| | - Alexander Dulebo
- JPK BioAFM Business, Bruker Nano GmbH, Am Studio 2D, 12489 Berlin, Germany
| | - Piia Mikkonen
- UPM-Kymmene Corporation, Alvar Aallon Katu 1, 00101 Helsinki, Finland
| | - Markus Nuopponen
- UPM-Kymmene Corporation, Alvar Aallon Katu 1, 00101 Helsinki, Finland
| | - Paula Turkki
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, FI-33014 Tampere, Finland.,Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| | - Vesa P Hytönen
- Faculty of Medicine and Health Technology and BioMediTech, Tampere University, FI-33014 Tampere, Finland.,Fimlab Laboratories, Biokatu 4, FI-33520 Tampere, Finland
| |
Collapse
|
42
|
Shewale B, Dubois N. Of form and function: Early cardiac morphogenesis across classical and emerging model systems. Semin Cell Dev Biol 2021; 118:107-118. [PMID: 33994301 PMCID: PMC8434962 DOI: 10.1016/j.semcdb.2021.04.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/31/2022]
Abstract
The heart is the earliest organ to develop during embryogenesis and is remarkable in its ability to function efficiently as it is being sculpted. Cardiac heart defects account for a high burden of childhood developmental disorders with many remaining poorly understood mechanistically. Decades of work across a multitude of model organisms has informed our understanding of early cardiac differentiation and morphogenesis and has simultaneously opened new and unanswered questions. Here we have synthesized current knowledge in the field and reviewed recent developments in the realm of imaging, bioengineering and genetic technology and ex vivo cardiac modeling that may be deployed to generate more holistic models of early cardiac morphogenesis, and by extension, new platforms to study congenital heart defects.
Collapse
Affiliation(s)
- Bhavana Shewale
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicole Dubois
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
43
|
Azizi L, Turkki P, Huynh N, Massera JM, Hytönen VP. Surface Modification of Bioactive Glass Promotes Cell Attachment and Spreading. ACS OMEGA 2021; 6:22635-22642. [PMID: 34514235 PMCID: PMC8427643 DOI: 10.1021/acsomega.1c02669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 08/17/2021] [Indexed: 06/13/2023]
Abstract
Phosphate glasses have several advantages over traditional silicate-based bioglasses but are inferior in the crucial step of cell attachment to their surface. Here, as a proof of concept, we analyze fibroblast attachment to the phosphate glass surface subjected to basic treatment and silanization. Silicate (S53P4)- and phosphate (Sr50)-based bioactive glasses were either untreated or surface-treated with basic buffer and functionalized with silane. The surface-treated samples were studied as such and after fibronectin was adsorbed on to their surface. With both glass types, surface treatment enhanced fibroblast adhesion and spreading in comparison to the untreated glass. The surface-treated Sr50 glass allowed for cell adhesion, proliferation, and spreading to a similar extent as seen with S53P4 and borosilicate control glasses. Here, we show that surface treatment of bioactive glass can be used to attract cell adhesion factors found in the serum and promote cell-material adhesion, both important for efficient tissue integration.
Collapse
Affiliation(s)
- Latifeh Azizi
- BioMediTech,
Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Ylpön katu 34, 33520 Tampere, Finland
| | - Paula Turkki
- BioMediTech,
Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Ylpön katu 34, 33520 Tampere, Finland
- Fimlab
Laboratories, Biokatu
4, 33520 Tampere, Finland
| | - Ngoc Huynh
- Laboratory
of Biomaterials and Tissue Engineering, Faculty of Medicine and Health
Technology, Tampere University, Hervanta Campus, Korkeakoulunkatu
3, 33720 Tampere, Finland
| | - Jonathan M. Massera
- Laboratory
of Biomaterials and Tissue Engineering, Faculty of Medicine and Health
Technology, Tampere University, Hervanta Campus, Korkeakoulunkatu
3, 33720 Tampere, Finland
| | - Vesa P. Hytönen
- BioMediTech,
Faculty of Medicine and Health Technology, Tampere University, Kauppi Campus, Arvo Ylpön katu 34, 33520 Tampere, Finland
- Fimlab
Laboratories, Biokatu
4, 33520 Tampere, Finland
| |
Collapse
|
44
|
Abe K, Baba K, Huang L, Wei KT, Okano K, Hosokawa Y, Inagaki N. Mechanosensitive axon outgrowth mediated by L1-laminin clutch interface. Biophys J 2021; 120:3566-3576. [PMID: 34384760 PMCID: PMC8456307 DOI: 10.1016/j.bpj.2021.08.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/28/2021] [Accepted: 08/04/2021] [Indexed: 11/11/2022] Open
Abstract
Mechanical properties of the extracellular environment modulate axon outgrowth. Growth cones at the tip of extending axons generate traction force for axon outgrowth by transmitting the force of actin filament retrograde flow, produced by actomyosin contraction and F-actin polymerization, to adhesive substrates through clutch and cell adhesion molecules. A molecular clutch between the actin filament flow and substrate is proposed to contribute to cellular mechanosensing. However, the molecular identity of the clutch interface responsible for mechanosensitive growth cone advance is unknown. We previously reported that mechanical coupling between actin filament retrograde flow and adhesive substrates through the clutch molecule shootin1a and the cell adhesion molecule L1 generates traction force for axon outgrowth and guidance. Here, we show that cultured mouse hippocampal neurons extend longer axons on stiffer substrates under elastic conditions that correspond to the soft brain environments. We demonstrate that this stiffness-dependent axon outgrowth requires actin-adhesion coupling mediated by shootin1a, L1, and laminin on the substrate. Speckle imaging analyses showed that L1 at the growth cone membrane switches between two adhesive states: L1 that is immobilized and that undergoes retrograde movement on the substrate. The duration of the immobilized phase was longer on stiffer substrates; this was accompanied by increases in actin-adhesion coupling and in the traction force exerted on the substrate. These data suggest that the interaction between L1 and laminin is enhanced on stiffer substrates, thereby promoting force generation for axon outgrowth.
Collapse
Affiliation(s)
- Kouki Abe
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Kentarou Baba
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Liguo Huang
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Koay Teng Wei
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Kazunori Okano
- Bio-processing Engineering Laboratory, Division of Materials Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Yoichiroh Hosokawa
- Bio-processing Engineering Laboratory, Division of Materials Science, Nara Institute of Science and Technology, Ikoma, Japan
| | - Naoyuki Inagaki
- Laboratory of Systems Neurobiology and Medicine, Division of Biological Science, Nara Institute of Science and Technology, Ikoma, Japan.
| |
Collapse
|
45
|
Mandal P, Belapurkar V, Nair D, Ramanan N. Vinculin-mediated axon growth requires interaction with actin but not talin in mouse neocortical neurons. Cell Mol Life Sci 2021; 78:5807-5826. [PMID: 34148098 PMCID: PMC11071915 DOI: 10.1007/s00018-021-03879-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 05/12/2021] [Accepted: 06/11/2021] [Indexed: 12/23/2022]
Abstract
The actin-binding protein vinculin is a major constituent of focal adhesion, but its role in neuronal development is poorly understood. We found that vinculin deletion in mouse neocortical neurons attenuated axon growth both in vitro and in vivo. Using functional mutants, we found that expression of a constitutively active vinculin significantly enhanced axon growth while the head-neck domain had an inhibitory effect. Interestingly, we found that vinculin-talin interaction was dispensable for axon growth and neuronal migration. Strikingly, expression of the tail domain delayed migration, increased branching, and stunted axon. Inhibition of the Arp2/3 complex or abolishing the tail domain interaction with actin completely reversed the branching phenotype caused by tail domain expression without affecting axon length. Super-resolution microscopy showed increased mobility of actin in tail domain expressing neurons. Our results provide novel insights into the role of vinculin and its functional domains in regulating neuronal migration and axon growth.
Collapse
Affiliation(s)
- Pranay Mandal
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Vivek Belapurkar
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Deepak Nair
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | - Narendrakumar Ramanan
- Centre for Neuroscience, Indian Institute of Science, Bangalore, 560012, Karnataka, India.
| |
Collapse
|
46
|
Alshehri S, Susapto HH, Hauser CAE. Scaffolds from Self-Assembling Tetrapeptides Support 3D Spreading, Osteogenic Differentiation, and Angiogenesis of Mesenchymal Stem Cells. Biomacromolecules 2021; 22:2094-2106. [PMID: 33908763 PMCID: PMC8382244 DOI: 10.1021/acs.biomac.1c00205] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/15/2021] [Indexed: 01/01/2023]
Abstract
The apparent rise of bone disorders demands advanced treatment protocols involving tissue engineering. Here, we describe self-assembling tetrapeptide scaffolds for the growth and osteogenic differentiation of human mesenchymal stem cells (hMSCs). The rationally designed peptides are synthetic amphiphilic self-assembling peptides composed of four amino acids that are nontoxic. These tetrapeptides can quickly solidify to nanofibrous hydrogels that resemble the extracellular matrix and provide a three-dimensional (3D) environment for cells with suitable mechanical properties. Furthermore, we can easily tune the stiffness of these peptide hydrogels by just increasing the peptide concentration, thus providing a wide range of peptide hydrogels with different stiffnesses for 3D cell culture applications. Since successful bone regeneration requires both osteogenesis and vascularization, our scaffold was found to be able to promote angiogenesis of human umbilical vein endothelial cells (HUVECs) in vitro. The results presented suggest that ultrashort peptide hydrogels are promising candidates for applications in bone tissue engineering.
Collapse
Affiliation(s)
- Salwa Alshehri
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Hepi H. Susapto
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Charlotte A. E. Hauser
- Laboratory
for Nanomedicine, Division of Biological and Environmental
Science and Engineering and Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology, Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
47
|
Biswas R, Banerjee A, Lembo S, Zhao Z, Lakshmanan V, Lim R, Le S, Nakasaki M, Kutyavin V, Wright G, Palakodeti D, Ross RS, Jamora C, Vasioukhin V, Jie Y, Raghavan S. Mechanical instability of adherens junctions overrides intrinsic quiescence of hair follicle stem cells. Dev Cell 2021; 56:761-780.e7. [PMID: 33725480 DOI: 10.1016/j.devcel.2021.02.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 11/24/2020] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Vinculin, a mechanotransducer associated with both adherens junctions (AJs) and focal adhesions (FAs), plays a central role in force transmission through cell-cell and cell-substratum contacts. We generated the conditional knockout (cKO) of vinculin in murine skin that results in the loss of bulge stem cell (BuSC) quiescence and promotes continual cycling of the hair follicles. Surprisingly, we find that the AJs in vinculin cKO cells are mechanically weak and impaired in force generation despite increased junctional expression of E-cadherin and α-catenin. Mechanistically, we demonstrate that vinculin functions by keeping α-catenin in a stretched/open conformation, which in turn regulates the retention of YAP1, another potent mechanotransducer and regulator of cell proliferation, at the AJs. Altogether, our data provide mechanistic insights into the hitherto-unexplored regulatory link between the mechanical stability of cell junctions and contact-inhibition-mediated maintenance of BuSC quiescence.
Collapse
Affiliation(s)
- Ritusree Biswas
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore 560065, India; SASTRA University, Thanjavur, Tamil Nadu 613401, India
| | - Avinanda Banerjee
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore 560065, India; Skin Research Institute of Singapore (A∗STAR), Singapore 138648, Singapore
| | - Sergio Lembo
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore 560065, India
| | - Zhihai Zhao
- Department of Physics, National University of Singapore, Singapore 117542, Singapore
| | - Vairavan Lakshmanan
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore 560065, India; SASTRA University, Thanjavur, Tamil Nadu 613401, India
| | - Ryan Lim
- Skin Research Institute of Singapore (A∗STAR), Singapore 138648, Singapore
| | - Shimin Le
- Department of Physics, National University of Singapore, Singapore 117542, Singapore
| | | | | | - Graham Wright
- A∗STAR Microscopy Platform, Skin Research Institute of Singapore (A∗STAR), Singapore 138648, Singapore
| | - Dasaradhi Palakodeti
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore 560065, India
| | - Robert S Ross
- University of California, San Diego, La Jolla, CA 92093, USA
| | - Colin Jamora
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore 560065, India
| | | | - Yan Jie
- Department of Physics, National University of Singapore, Singapore 117542, Singapore; Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
| | - Srikala Raghavan
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore 560065, India; Skin Research Institute of Singapore (A∗STAR), Singapore 138648, Singapore.
| |
Collapse
|
48
|
Wang Y, Qin Y, Peng R, Wang H. Loss-of-function or gain-of-function variations in VINCULIN (VCL) are risk factors of human neural tube defects. Mol Genet Genomic Med 2021; 9:e1563. [PMID: 33491343 PMCID: PMC8077129 DOI: 10.1002/mgg3.1563] [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: 08/21/2020] [Revised: 10/28/2020] [Accepted: 11/05/2020] [Indexed: 11/12/2022] Open
Abstract
Background Neural tube defects (NTDs) are severe birth defects resulting from the failure of neural tube closure during embryogenesis. Both genetic and environmental factors contribute to the occurrence of NTDs and the heritability of NTDs is approximately 70%. As a key component of focal adhesions, Vinculin (VCL) plays pivotal roles in cell skeleton remodeling and signal transduction. Vcl deficient mice displayed NTD, but how VCL variants contribute to human NTDs has not been addressed yet. Methods We screened VCL variants in a Chinese cohort of 387 NTDs and 244 controls by targeted next‐generation sequencing. Results We identified four case‐specific VCL variations (p.M209L, p.D256fs, p.L555V and p.R586Q). VCL p.D256fs and p.L555V are novel variations that have never been reported. Our analysis revealed that p.D256fs is a loss‐of‐function variant, while p.L555V showed a gain of function in planner cell polarity (PCP) pathway regulation and cell migration, probably due to its enhanced protein stability. Conclusion Our study reports human NTD specific novel variations in VCL and provides the functional evaluation of VCL variants related to the etiology of human NTDs.
Collapse
Affiliation(s)
- Yalan Wang
- Obstetrics & Gynecology Hospital, Institute of Reproduction & Development, Fudan University, Shanghai, China
| | - Yue Qin
- State Key Laboratory of Genetic, Engineering at School of Life Sciences, Fudan University, Shanghai, China
| | - Rui Peng
- Obstetrics & Gynecology Hospital, Institute of Reproduction & Development, Fudan University, Shanghai, China
| | - Hongyan Wang
- Obstetrics & Gynecology Hospital, Institute of Reproduction & Development, Fudan University, Shanghai, China.,State Key Laboratory of Genetic, Engineering at School of Life Sciences, Fudan University, Shanghai, China.,Key Laboratory of Reproduction Regulation of NPFPC, Collaborative Innovation Center of Genetics and Development, Fudan University, Shanghai, China.,Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
49
|
Design and synthesis of novel quinic acid derivatives: in vitro cytotoxicity and anticancer effect on glioblastoma. Future Med Chem 2020; 12:1891-1910. [PMID: 33124467 DOI: 10.4155/fmc-2020-0194] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Aim: Quinic acid (QA) is a cyclic polyol exhibiting anticancer properties on several cancers. However, potential role of QA derivatives against glioblastoma is not well established. Methodology & results: Sixteen novel QA derivatives and QA-16 encapsulated poly (lactic-co-glycolic acid) nanoparticles (QA-16-NPs) were screened for their anti-glioblastoma effect using standard cell and molecular biology methods. Presence of a tertiary hydroxy and silylether groups in the lead compound were identified for the antitumor activity. QA-16 have 90% inhibition with the IC50 of 10.66 μM and 28.22 μM for LN229 and SNB19, respectively. The induction of apoptosis is faster with the increased fold change of caspase 3/7 and reactive oxygen species. Conclusion: QA-16 and QA-16-NPs shows similar cytotoxicity effect, providing the opportunity to use QA-16 as a potential chemotherapeutic agent.
Collapse
|
50
|
Morales W, Triantafyllou K, Parodi G, Weitsman S, Park SC, Rezaie A, Pichetshote N, Lin E, Pimentel M. Immunization with cytolethal distending toxin B produces autoantibodies to vinculin and small bowel bacterial changes in a rat model of postinfectious irritable bowel syndrome. Neurogastroenterol Motil 2020; 32:e13875. [PMID: 32436301 DOI: 10.1111/nmo.13875] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 03/25/2020] [Accepted: 04/17/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Recent data substantiate the importance of acute gastroenteritis in the development of irritable bowel syndrome (IBS). An animal model of postinfectious IBS determined the importance of cytolethal distending toxin B (CdtB) during live Campylobacter jejuni infection and its development of autoimmunity to vinculin. In this study, we examine whether subcutaneous exposure to CdtB alone is sufficient to produce the postinfectious IBS effect and autoimmunity. METHODS Sixty adult Sprague Dawley rats were randomized into 2 groups to receive subcutaneous injection of either CdtB or vehicle and administered a booster injection of the same product 3 weeks later. Serum was collected for anti-CdtB and anti-vinculin titers. Duodenal and ileal luminal contents for total eubacterial qPCR, and ileal bowel segments were harvested for vinculin and ileal expression. In a second experiment, 4 adult, Sprague Dawley rats were injected with either Cy7-labeled anti-CdtB and anti-vinculin antibodies were injected into the tail vein and imaged to determine organ localization of the antibodies. KEY RESULTS Rats that received CdtB increased in serum anti-CdtB after injection. CdtB exposure also precipitated significant elevation in anti-vinculin antibodies (P < .001). This was associated with a reduction in intestinal vinculin expression (P < .001) that negatively correlated with serum anti-CdtB levels. CdtB exposure was also associated with greater levels of duodenal (P < .001) and ileal (P < .01) bacteria by qPCR that positively correlated with anti-CdtB levels. CONCLUSIONS AND INFERENCES Rats injected with CdtB developed a postinfectious IBS-like phenotype and autoimmunity to vinculin with corresponding reduction in intestinal vinculin expression.
Collapse
Affiliation(s)
- Walter Morales
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Konstantinos Triantafyllou
- Hepatogastroenterology Unit, 2nd Department of Internal Medicine - Propaedeutic, Research Institute and Diabetes Center, Medical School, Attikon University General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Gonzalo Parodi
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stacy Weitsman
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sung Chul Park
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Ali Rezaie
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nipaporn Pichetshote
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eugenia Lin
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mark Pimentel
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|