1
|
Kashani N, Sabbaghian A, Ahmadi K, Aalikhani M. In silico drug repurposing for potential HPV-induced skin wart treatment - A comparative transcriptome analysis. J Genet Eng Biotechnol 2025; 23:100485. [PMID: 40390498 PMCID: PMC11997329 DOI: 10.1016/j.jgeb.2025.100485] [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: 11/24/2024] [Revised: 03/12/2025] [Accepted: 03/23/2025] [Indexed: 05/21/2025]
Abstract
INTRODUCTION Warts are dermal disorders resulting from HPV infection and can be transmitted by direct contact. Existing treatment approaches, such as topical treatment with salicylate, have low efficiency and demonstrate side effects. Thus, the discovery of potent drug treatments for skin warts is necessary. Here we propose the use of alternative medications for the possible treatment of skin warts with the help of comparative transcriptome analysis and drug repurposing approaches. METHODS Gene expression datasets related to HPV-induced warts and cervical cancer were extracted from the GEO database. Differentially expressed genes (DEGs) were identified using DESeq2 in the Galaxy database. Upregulated DEGs were assessed for druggability using the DGIdb tool. Gene ontology and enrichment analysis were performed to investigate the characteristics of druggable DEGs. A molecular docking virtual screening was conducted using PyRx software to identify potential therapeutic targets for skin warts. The interactions between selected drug candidates and the target protein were analyzed using the BIOVIA Discovery Studio. The physicochemical characteristics of potential pharmaceuticals were evaluated using the SwissADME database. Finally, the molecular dynamics (MD) simulation was performed to validate the stability and dynamic behavior of drug-protein interactions. RESULTS Based on the findings from gene expression profiling, Integrin Alpha-X (ITGAX, CD11c) has been identified as a candidate protein that is significantly upregulated in individuals afflicted with skin warts. Integrin Alpha-X plays a crucial role in mediating intercellular interactions during inflammatory processes and notably enhances the adhesion and chemotactic activity of monocytes. Through molecular docking, MD, and physicochemical analyses, it has been demonstrated that dihydroergotamine effectively inhibits the ITGAX protein, suggesting its potential as a therapeutic agent for the management of skin warts. CONCLUSION Dihydroergotamine can be repurposed as a potential drug in the treatment of skin warts by targeting Integrin Alpha-X protein.
Collapse
Affiliation(s)
- Navid Kashani
- Department of Biology, Faculty of Science, Azad University Gorgan Branch, Gorgan, Iran
| | - Amir Sabbaghian
- Guoke Ningbo Life Science and Health Industry Research Institute, University of Chinese Academy of Sciences, Ningbo, Zhejiang, People's Republic of China
| | - Khadijeh Ahmadi
- Department of Medical Biotechnology, School of Paramedicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Mahdi Aalikhani
- Department of Medical Biotechnology, School of Paramedicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| |
Collapse
|
2
|
Reuning U, D'Amore VM, Hodivala-Dilke K, Marinelli L, Kessler H. Importance of integrin transmembrane helical interactions for antagonistic versus agonistic ligand behavior: Consequences for medical applications. Bioorg Chem 2025; 156:108193. [PMID: 39842299 DOI: 10.1016/j.bioorg.2025.108193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/27/2024] [Accepted: 01/17/2025] [Indexed: 01/24/2025]
Abstract
Integrins are well-characterized receptors involved in cell adhesion and signaling. With six approved drugs, they are recognized as valuable therapeutic targets. Here, we explore potential activation mechanisms that may clarify the agonist versus antagonist behavior of integrin ligands. The reorganization of the transmembrane domain (TMD) in the integrin receptor, forming homooligomers within focal adhesions, could be key to the understanding of the agonistic properties of integrin ligands at substoichiometric concentrations. This has significant implications for medical applications. While we focus on the RGD peptide-recognizing integrin subfamily, we propose that these mechanistic insights may also apply to other integrin subtypes. For application of integrin ligands in medicine it is essential to consider this mechanism and its consequences for affinity and bioavailability.
Collapse
Affiliation(s)
- Ute Reuning
- TUM University Hospital, Klinikum Rechts der Isar, School of Medicine and Health, Technical University of Munich, Department of Gynecology and Obstetrics, Clinical Research Unit, Ismaninger Strasse 22, 81675 Munich, Germany.
| | - Vincenzo Maria D'Amore
- University of Naples Federico II, UNINA-Department of Pharmacy, C.so Umberto I, 40, 80138 Naples, Italy.
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, United Kingdom.
| | - Luciana Marinelli
- University of Naples Federico II, UNINA-Department of Pharmacy, C.so Umberto I, 40, 80138 Naples, Italy.
| | - Horst Kessler
- Institute for Advanced Study, Department of Chemistry, School of Natural Sciences and Bavarian NMR Center (BNMRZ), Technical University Munich, Ernst-Otto-Fischer-Str. 2, 85748 Garching, Germany.
| |
Collapse
|
3
|
Miao MZ, Lee JS, Yamada KM, Loeser RF. Integrin signalling in joint development, homeostasis and osteoarthritis. Nat Rev Rheumatol 2024; 20:492-509. [PMID: 39014254 PMCID: PMC11886400 DOI: 10.1038/s41584-024-01130-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2024] [Indexed: 07/18/2024]
Abstract
Integrins are key regulators of cell-matrix interactions during joint development and joint tissue homeostasis, as well as in the development of osteoarthritis (OA). The signalling cascades initiated by the interactions of integrins with a complex network of extracellular matrix (ECM) components and intracellular adaptor proteins orchestrate cellular responses necessary for maintaining joint tissue integrity. Dysregulated integrin signalling, triggered by matrix degradation products such as matrikines, disrupts this delicate balance, tipping the scales towards an environment conducive to OA pathogenesis. The interplay between integrin signalling and growth factor pathways further underscores the multifaceted nature of OA. Moreover, emerging insights into the role of endocytic trafficking in regulating integrin signalling add a new layer of complexity to the understanding of OA development. To harness the therapeutic potential of targeting integrins for mitigation of OA, comprehensive understanding of their molecular mechanisms across joint tissues is imperative. Ultimately, deciphering the complexities of integrin signalling will advance the ability to treat OA and alleviate its global burden.
Collapse
Affiliation(s)
- Michael Z Miao
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Division of Rheumatology, Allergy, and Immunology and the Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Janice S Lee
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Office of the Clinical Director, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| | - Richard F Loeser
- Division of Rheumatology, Allergy, and Immunology and the Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
4
|
Lee DK, Oh J, Park HW, Gee HY. Anchorage Dependence and Cancer Metastasis. J Korean Med Sci 2024; 39:e156. [PMID: 38769921 PMCID: PMC11106561 DOI: 10.3346/jkms.2024.39.e156] [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] [Received: 04/03/2024] [Accepted: 04/19/2024] [Indexed: 05/22/2024] Open
Abstract
The process of cancer metastasis is dependent on the cancer cells' capacity to detach from the primary tumor, endure in a suspended state, and establish colonies in other locations. Anchorage dependence, which refers to the cells' reliance on attachment to the extracellular matrix (ECM), is a critical determinant of cellular shape, dynamics, behavior, and, ultimately, cell fate in nonmalignant and cancer cells. Anchorage-independent growth is a characteristic feature of cells resistant to anoikis, a programmed cell death process triggered by detachment from the ECM. This ability to grow and survive without attachment to a substrate is a crucial stage in the progression of metastasis. The recently discovered phenomenon named "adherent-to-suspension transition (AST)" alters the requirement for anchoring and enhances survival in a suspended state. AST is controlled by four transcription factors (IKAROS family zinc finger 1, nuclear factor erythroid 2, BTG anti-proliferation factor 2, and interferon regulatory factor 8) and can detach cells without undergoing the typical epithelial-mesenchymal transition. Notably, AST factors are highly expressed in circulating tumor cells compared to their attached counterparts, indicating their crucial role in the spread of cancer. Crucially, the suppression of AST substantially reduces metastasis while sparing primary tumors. These findings open up possibilities for developing targeted therapies that inhibit metastasis and emphasize the importance of AST, leading to a fundamental change in our comprehension of how cancer spreads.
Collapse
Affiliation(s)
- Dong Ki Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Korea
| | - Jongwook Oh
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Korea
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Brain Korea 21 Project, Yonsei University, Seoul, Korea.
| | - Heon Yung Gee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Korea
- Woo Choo Lee Institute for Precision Drug Development, Seoul, Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
5
|
Zhao Y, Zhang S, Cheng B, Feng F, Zhu Y, Liu Y, Wang J, Zou D, Ma H, Xu F, Zhang M. Mechanochemical coupling of MGF mediates periodontal regeneration. Bioeng Transl Med 2024; 9:e10603. [PMID: 38193124 PMCID: PMC10771565 DOI: 10.1002/btm2.10603] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 08/26/2023] [Accepted: 09/05/2023] [Indexed: 01/10/2024] Open
Abstract
Clinical evidence shows that the mechanical stimulation obtained from occlusion could enhance periodontal ligament (PDL) remodeling. Mechano-growth factor (MGF) is a growth factor produced specifically following mechanical stimulus Here, we aim to investigate the mechanical enhancement potential and mechanism of the MGF in PDL regeneration. In vivo study found that MGF produced from the PDL under occlusion force could strongly enhance PDL remodeling. In vitro experiments and mathematical modeling further confirmed the mechanical enhancement effect of MGF for PDLSC differentiation toward fibroblasts. A mechanochemical coupling effect of MGF mediated the enhancement of mechanical effect, which was modulated by Fyn-FAK kinases signaling and subsequent MAPK pathway. Finally, enhanced PDL regeneration under the mechanochemical coupling of MGF and occlusal force was verified in vivo. There exists an additive mechanical effect of MGF mediated by Fyn-FAK crosstalk and subsequent ERK1/2 and p38 phosphorylation, which could be developed as an MGF-centered adjuvant treatment to optimize PDL remodeling, especially for patients with weakened bite force or destroyed periodontium.
Collapse
Affiliation(s)
- Ying Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and EmergencySchool of Stomatology, Fourth Military Medical UniversityXi’anPeople's Republic of China
- Department of Anesthesiology and Perioperative MedicineXi'an People's Hospital (Xi'an Fourth Hospital), Northwest UniversityXi'anPeople's Republic of China
| | - Songbai Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and EmergencySchool of Stomatology, Fourth Military Medical UniversityXi’anPeople's Republic of China
| | - Bo Cheng
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and Technology, Xi'an Jiaotong UniversityXi’anPeople's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong UniversityXi’anPeople's Republic of China
| | - Fan Feng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and EmergencySchool of Stomatology, Fourth Military Medical UniversityXi’anPeople's Republic of China
| | - Yue Zhu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and EmergencySchool of Stomatology, Fourth Military Medical UniversityXi’anPeople's Republic of China
| | - Yanli Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and EmergencySchool of Stomatology, Fourth Military Medical UniversityXi’anPeople's Republic of China
| | - Junjun Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and EmergencySchool of Stomatology, Fourth Military Medical UniversityXi’anPeople's Republic of China
| | - Dehui Zou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and EmergencySchool of Stomatology, Fourth Military Medical UniversityXi’anPeople's Republic of China
| | - Heng Ma
- Department of Physiology & Department of PathophysiologySchool of Basic Medical Sciences, Fourth Military Medical UniversityXi’anPeople's Republic of China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of EducationSchool of Life Science and Technology, Xi'an Jiaotong UniversityXi’anPeople's Republic of China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong UniversityXi’anPeople's Republic of China
| | - Min Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and EmergencySchool of Stomatology, Fourth Military Medical UniversityXi’anPeople's Republic of China
| |
Collapse
|
6
|
Zou X, Zhang H, Benson JM, Gao H, Burris DL, Fox JM, Jia X. Modeling the Maturation of the Vocal Fold Lamina Propria Using a Bioorthogonally Tunable Hydrogel Platform. Adv Healthc Mater 2023; 12:e2301701. [PMID: 37530909 PMCID: PMC10834846 DOI: 10.1002/adhm.202301701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 07/10/2023] [Indexed: 08/03/2023]
Abstract
Toward the goal of establishing an engineered model of the vocal fold lamina propria (LP), mesenchymal stem cells (MSCs) are encapsulated in hyaluronic acid (HA)-based hydrogels employing tetrazine ligation with strained alkenes. To mimic matrix stiffening during LP maturation, diffusion-controlled interfacial bioorthogonal crosslinking is carried out on the soft cellular construct using HA modified with a ferocious dienophile, trans-cyclooctene (TCO). Cultures are maintained in MSC growth media for 14 days to afford a model of a newborn LP that is homogeneously soft (nLP), a homogeneously stiffened construct zero (sLP0) or 7 days (sLP7) post cell encapsulation, and a mature LP model (mLP) with a stiff top layer and a soft bottom layer. Installation of additional HA crosslinks restricts cell spreading. Compared to the nLP controls, sLP7 conditions upregulate the expression of fibrous matrix proteins (Col I, DCN, and FN EDA), classic fibroblastic markers (TNC, FAP, and FSP1), and matrix remodeling enzymes (MMP2, TIMP1, and HAS3). Day 7 stiffening also upregulates the catabolic activities, enhances ECM turnover, and promotes YAP expression. Overall, in situ delayed matrix stiffening promotes a fibroblast transition from MSCs and enhances YAP-regulated mechanosensing.
Collapse
Affiliation(s)
- Xiaoyu Zou
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
| | - He Zhang
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
| | - Jamie M. Benson
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
| | - Hanyuan Gao
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
| | - David L. Burris
- Department of Mechanical Engineering, University of Delaware, Newark, Delaware, USA
| | - Joseph. M. Fox
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware, USA
| | - Xinqiao Jia
- Department of Materials Science and Engineering, University of Delaware, Newark, Delaware, USA
- Department of Biomedical Engineering, University of Delaware, Newark, Delaware, USA
- Delaware Biotechnology Institute, 590 Avenue 1743, Newark, Delaware, USA
| |
Collapse
|
7
|
Wu JL, Xu CF, Yang XH, Wang MS. Fibronectin promotes tumor progression through integrin αvβ3/PI3K/AKT/SOX2 signaling in non-small cell lung cancer. Heliyon 2023; 9:e20185. [PMID: 37809806 PMCID: PMC10559956 DOI: 10.1016/j.heliyon.2023.e20185] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 09/09/2023] [Accepted: 09/13/2023] [Indexed: 10/10/2023] Open
Abstract
The tumor microenvironment, especially the extracellular matrix (ECM), is strongly associated with tumor cell proliferation and metastasis. Numerous studies have provided evidence suggesting that fibronectin (FN) in ECM supports cancer cell escape and contributes to cell migration, resulting in distant cancer metastasis and poor outcomes in patients. In our study, it was demonstrated that FN expression was elevated in tumor tissues from highly malignant NSCLC patients, compared to those with low malignancy (p = 0.0076). Importantly, FN promoted proliferative phenotypes and strengthened tumorigenesis capabilities in NSCLC cells, including A549 and Lewis cells, leading to sustained tumor growth in vivo. Mechanistically, it was identified that FN facilitated the activation of the integrin αvβ3/PI3K/AKT signaling pathway, which subsequently upregulated tumor stemness through the downstream transcription factor SOX2. Blockade of integrin αvβ3 signal efficiently suppressed NSCLC proliferation and tumorigenesis both in vitro and in vivo. In conclusion, our study demonstrated that extracellular FN could facilitate NSCLC development through the integrin αvβ3/PI3K/AKT/SOX2 signaling pathway. Blockade of integrin αvβ3 could efficiently enhance the anticancer effects of chemotherapy, offering an innovative approach for clinical NSCLC therapy.
Collapse
Affiliation(s)
- Jin-Long Wu
- Department of Thoracic Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai City, 200011, China
| | - Cheng-Feng Xu
- Department of Pharmacy, Shidong Hospital of Shanghai Yangpu District, Shanghai City, 200438, China
| | - Xu-Hui Yang
- Department of Thoracic Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai City, 200011, China
| | - Ming-Song Wang
- Department of Thoracic Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai City, 200011, China
| |
Collapse
|
8
|
Kumar S, Stainer A, Dubrulle J, Simpkins C, Cooper JA. Cas phosphorylation regulates focal adhesion assembly. eLife 2023; 12:e90234. [PMID: 37489578 PMCID: PMC10435235 DOI: 10.7554/elife.90234] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 07/19/2023] [Indexed: 07/26/2023] Open
Abstract
Integrin-mediated cell attachment rapidly induces tyrosine kinase signaling. Despite years of research, the role of this signaling in integrin activation and focal adhesion assembly is unclear. We provide evidence that the Src-family kinase (SFK) substrate Cas (Crk-associated substrate, p130Cas, BCAR1) is phosphorylated and associated with its Crk/CrkL effectors in clusters that are precursors of focal adhesions. The initial phospho-Cas clusters contain integrin β1 in its inactive, bent closed, conformation. Later, phospho-Cas and total Cas levels decrease as integrin β1 is activated and core focal adhesion proteins including vinculin, talin, kindlin, and paxillin are recruited. Cas is required for cell spreading and focal adhesion assembly in epithelial and fibroblast cells on collagen and fibronectin. Cas cluster formation requires Cas, Crk/CrkL, SFKs, and Rac1 but not vinculin. Rac1 provides positive feedback onto Cas through reactive oxygen, opposed by negative feedback from the ubiquitin proteasome system. The results suggest a two-step model for focal adhesion assembly in which clusters of phospho-Cas, effectors and inactive integrin β1 grow through positive feedback prior to integrin activation and recruitment of core focal adhesion proteins.
Collapse
Affiliation(s)
- Saurav Kumar
- Fred Hutchinson Cancer CenterSeattleUnited States
| | | | | | | | | |
Collapse
|
9
|
Davis MJ, Earley S, Li YS, Chien S. Vascular mechanotransduction. Physiol Rev 2023; 103:1247-1421. [PMID: 36603156 PMCID: PMC9942936 DOI: 10.1152/physrev.00053.2021] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 09/26/2022] [Accepted: 10/04/2022] [Indexed: 01/07/2023] Open
Abstract
This review aims to survey the current state of mechanotransduction in vascular smooth muscle cells (VSMCs) and endothelial cells (ECs), including their sensing of mechanical stimuli and transduction of mechanical signals that result in the acute functional modulation and longer-term transcriptomic and epigenetic regulation of blood vessels. The mechanosensors discussed include ion channels, plasma membrane-associated structures and receptors, and junction proteins. The mechanosignaling pathways presented include the cytoskeleton, integrins, extracellular matrix, and intracellular signaling molecules. These are followed by discussions on mechanical regulation of transcriptome and epigenetics, relevance of mechanotransduction to health and disease, and interactions between VSMCs and ECs. Throughout this review, we offer suggestions for specific topics that require further understanding. In the closing section on conclusions and perspectives, we summarize what is known and point out the need to treat the vasculature as a system, including not only VSMCs and ECs but also the extracellular matrix and other types of cells such as resident macrophages and pericytes, so that we can fully understand the physiology and pathophysiology of the blood vessel as a whole, thus enhancing the comprehension, diagnosis, treatment, and prevention of vascular diseases.
Collapse
Affiliation(s)
- Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno, Nevada
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, California
- Institute of Engineering in Medicine, University of California, San Diego, California
- Department of Medicine, University of California, San Diego, California
| |
Collapse
|
10
|
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
|
11
|
Wang F, Wang Q, Zhao Y, Tian Z, Chang S, Tong H, Liu N, Bai S, Li X, Fan J. Adipose-derived stem cells with miR-150-5p inhibition laden in hydroxyapatite/tricalcium phosphate ceramic powders promote osteogenesis via regulating Notch3 and activating FAK/ERK and RhoA. Acta Biomater 2023; 155:644-653. [PMID: 36206975 DOI: 10.1016/j.actbio.2022.09.070] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 02/02/2023]
Abstract
Adipose-derived mesenchymal stem cells (ADSCs) are multipotent stromal cells and play huge role in forming and repairing bone tissues. Emerging evidence shows that MicroRNAs (miRNAs) are involved in ADSCs differentiation. Here, we explored the role of miR-150-5p and its related mechanisms in ADSCs osteogenesis. Real-time PCR was used to determine miR-150-5p expression during ADSCs osteogenesis. miR-150-5p inhibitors, miR-150-5p ADV or short hairpin RNA (shRNA) of Notch3 were transfected to ADSCs for analyzing the effects on osteogenesis. The mixture of hydroxyapatite/tricalcium phosphate (HA/TCP) ceramic powders and transfected ADSCs was implanted into BALB/C nude mice. Micro-CT and histological methods were performed to evaluate the new bone formation. Compared with negative control (NC) and miR-150-5p overexpression, inhibition of miR-150-5p increased ADSCs osteogenesis by regulating Notch3. MiR-150-5p overexpression decreased the expression of pFAK, pERK1/2, and RhoA, while these were up-regulated when miR-150-5p was inhibited, or notch3 was silenced. Furthermore, miR-150-5p inhibition partially reversed the suppression effect of notch3 knockdown on osteogenesis in vitro and in vivo. This study demonstrated the critical function of miR-150-5p during osteogenesis. The combination of ADSCs with miR-150-5p inhibition and HA/TCP might be a promising strategy for bone damage repair. STATEMENT OF SIGNIFICANCE: Osteoporosis is a common chronic metabolic bone disease in humans. Bone tissue engineering based on mesenchymal stem cells, biomaterials, and growth factors, provides a promising way to treat osteoporosis and bone defects. ADSCs commonly differentiate into adipose cells, they can also differentiate into osteogenic cell lineages. Nucleic acids and protein have usually been considered as regulators of ADSCs osteogenic differentiation. In the current study, we demonstrated the combination of ADSCs with miR-150-5p inhibition and hydroxyapatite/tricalcium phosphate ceramic powders enhanced bone regeneration. Furthermore, miR-150-5p/Notch3 axis regulating osteogenesis via the FAK/ERK1/2 and RhoA pathway was assessed. The current study showed the application of ADSCs in bone regeneration might be a promising strategy for osteoporosis and bone damage repairing.
Collapse
Affiliation(s)
- Fanglin Wang
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Qiao Wang
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Yu Zhao
- Department of Plastic Surgery, Shengjing Hospital, Affiliated Hospital of China Medical University, No.36 Sanhao Street, Heping area, Shenyang, Liaoning 110004, PR China
| | - Zhiyu Tian
- Clinical Primary Department 105K, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Shijie Chang
- Division of Biomedical Engineering, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Hao Tong
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Ningwei Liu
- 5+3 Integration of Clinical Medicine 106K, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Shuling Bai
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China
| | - Xiang Li
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China; Department of Cell Biology, School of Life Sciences, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China.
| | - Jun Fan
- Department of Tissue Engineering, School of Intelligent Medicine, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning 110122, PR China.
| |
Collapse
|
12
|
Ain U, Firdaus H. Parvin: A hub of intracellular signalling pathways regulating cellular behaviour and disease progression. Acta Histochem 2022; 124:151935. [PMID: 35932544 DOI: 10.1016/j.acthis.2022.151935] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 11/15/2022]
Abstract
α-actinin superfamily houses the family of parvins, comprising α, β and γ isoforms in the vertebrates and a single orthologue in the invertebrates. Parvin as an adaptor protein is a member of the ternary IPP-complex including Integrin Linked Kinase (ILK) and particularly-interesting-Cys-His-rich protein (PINCH). Each of the complex proteins showed a conserved lineage and was principally used by the evolutionarily primitive integrin-adhesome machinery to regulate cellular behaviour and signalling pathways. Parvin facilitated integrin mediated integration of the extracellular matrix with cytoskeletal framework culminating in regulation of cellular adhesion and spreading, cytoskeleton reorganisation and cell survival. Studies have established role of parvin in pregnancy, lactation, matrix degradation, blood vessel formation and in several diseases such as cancer, NAFLD and cardiac diseases etc. This review narrates the history of parvin discovery, its elaborate gene structure and conservation across phyla including cellular expression, localisation and interacting partners in vertebrates as well as invertebrates. The review further discusses how parvin acts as an epicentre of signalling pathways, its associated mutants and diseased conditions.
Collapse
Affiliation(s)
- Ushashi Ain
- Department of Life Sciences, Central University of Jharkhand, CTI Campus, Ratu-Lohardaga Road, Ranchi 835205, India
| | - Hena Firdaus
- Department of Life Sciences, Central University of Jharkhand, CTI Campus, Ratu-Lohardaga Road, Ranchi 835205, India.
| |
Collapse
|
13
|
Luthold C, Hallal T, Labbé DP, Bordeleau F. The Extracellular Matrix Stiffening: A Trigger of Prostate Cancer Progression and Castration Resistance? Cancers (Basel) 2022; 14:cancers14122887. [PMID: 35740556 PMCID: PMC9221142 DOI: 10.3390/cancers14122887] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023] Open
Abstract
Despite advancements made in diagnosis and treatment, prostate cancer remains the second most diagnosed cancer among men worldwide in 2020, and the first in North America and Europe. Patients with localized disease usually respond well to first-line treatments, however, up to 30% develop castration-resistant prostate cancer (CRPC), which is often metastatic, making this stage of the disease incurable and ultimately fatal. Over the last years, interest has grown into the extracellular matrix (ECM) stiffening as an important mediator of diseases, including cancers. While this process is increasingly well-characterized in breast cancer, a similar in-depth look at ECM stiffening remains lacking for prostate cancer. In this review, we scrutinize the current state of literature regarding ECM stiffening in prostate cancer and its potential association with disease progression and castration resistance.
Collapse
Affiliation(s)
- Carole Luthold
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada;
- Division of Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
| | - Tarek Hallal
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
| | - David P. Labbé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
- Division of Urology, Department of Surgery, McGill University, Montréal, QC H4A 3J1, Canada
- Correspondence: (D.P.L.); (F.B.)
| | - François Bordeleau
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada;
- Division of Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
- Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: (D.P.L.); (F.B.)
| |
Collapse
|
14
|
Haroon M, Boers HE, Bakker AD, Bloks NGC, Hoogaars WMH, Giordani L, Musters RJP, Deldicque L, Koppo K, Le Grand F, Klein-Nulend J, Jaspers RT. Reduced growth rate of aged muscle stem cells is associated with impaired mechanosensitivity. Aging (Albany NY) 2022; 14:28-53. [PMID: 35023852 PMCID: PMC8791224 DOI: 10.18632/aging.203830] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 01/03/2022] [Indexed: 11/25/2022]
Abstract
Aging-associated muscle wasting and impaired regeneration are caused by deficiencies in muscle stem cell (MuSC) number and function. We postulated that aged MuSCs are intrinsically impaired in their responsiveness to omnipresent mechanical cues through alterations in MuSC morphology, mechanical properties, and number of integrins, culminating in impaired proliferative capacity. Here we show that aged MuSCs exhibited significantly lower growth rate and reduced integrin-α7 expression as well as lower number of phospho-paxillin clusters than young MuSCs. Moreover, aged MuSCs were less firmly attached to matrigel-coated glass substrates compared to young MuSCs, as 43% of the cells detached in response to pulsating fluid shear stress (1 Pa). YAP nuclear localization was 59% higher than in young MuSCs, yet YAP target genes Cyr61 and Ctgf were substantially downregulated. When subjected to pulsating fluid shear stress, aged MuSCs exhibited reduced upregulation of proliferation-related genes. Together these results indicate that aged MuSCs exhibit impaired mechanosensitivity and growth potential, accompanied by altered morphology and mechanical properties as well as reduced integrin-α7 expression. Aging-associated impaired muscle regenerative capacity and muscle wasting is likely due to aging-induced intrinsic MuSC alterations and dysfunctional mechanosensitivity.
Collapse
Affiliation(s)
- Mohammad Haroon
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 HZ, The Netherlands
| | - Heleen E Boers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 HZ, The Netherlands
| | - Astrid D Bakker
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 LA, The Netherlands
| | - Niek G C Bloks
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 HZ, The Netherlands
| | - Willem M H Hoogaars
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 HZ, The Netherlands
| | - Lorenzo Giordani
- Sorbonne Université, INSERM UMRS974, Center for Research in Myology, Paris 75013, France
| | - René J P Musters
- Department of Physiology, Amsterdam University Medical Center VUmc, Amsterdam Cardiovascular Sciences, Amsterdam 1081 HZ, The Netherlands
| | - Louise Deldicque
- Institute of Neuroscience, Université Catholique de Louvain, Louvain-la-Neuve 1348, Belgium
| | - Katrien Koppo
- Exercise Physiology Research Group, Department of Movement Sciences, KU Leuven, Leuven 3001, Belgium
| | - Fabien Le Grand
- Faculty of Medicine and Pharmacy, NeuroMyoGène UCBL-CNRS UMR 5310, INSERM U1217, Lyon 69008, France
| | - Jenneke Klein-Nulend
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 LA, The Netherlands
| | - Richard T Jaspers
- Laboratory for Myology, Department of Human Movement Sciences, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam 1081 HZ, The Netherlands
| |
Collapse
|
15
|
Zarrintaj P, Ghorbani S, Barani M, Singh Chauhan NP, Khodadadi Yazdi M, Saeb MR, Ramsey JD, Hamblin MR, Mozafari M, Mostafavi E. Polylysine for skin regeneration: A review of recent advances and future perspectives. Bioeng Transl Med 2022; 7:e10261. [PMID: 35111953 PMCID: PMC8780928 DOI: 10.1002/btm2.10261] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/28/2021] [Accepted: 10/03/2021] [Indexed: 12/19/2022] Open
Abstract
There have been several attempts to find promising biomaterials for skin regeneration, among which polylysine (a homopolypeptide) has shown benefits in the regeneration and treatment of skin disorders. This class of biomaterials has shown exceptional abilities due to their macromolecular structure. Polylysine-based biomaterials can be used as tissue engineering scaffolds for skin regeneration, and as drug carriers or even gene delivery vectors for the treatment of skin diseases. In addition, polylysine can play a preservative role in extending the lifetime of skin tissue by minimizing the appearance of photodamaged skin. Research on polylysine is growing today, opening new scenarios that expand the potential of these biomaterials from traditional treatments to a new era of tissue regeneration. This review aims to address the basic concepts, recent trends, and prospects of polylysine-based biomaterials for skin regeneration. Undoubtedly, this class of biomaterials needs further evaluations and explorations, and many critical questions have yet to be answered.
Collapse
Affiliation(s)
- Payam Zarrintaj
- School of Chemical EngineeringOklahoma State UniversityStillwaterOklahomaUSA
| | - Sadegh Ghorbani
- Interdisciplinary Nanoscience Center (iNANO)Aarhus UniversityAarhusDenmark
| | - Mahmood Barani
- Medical Mycology and Bacteriology Research CenterKerman University of Medical SciencesKermanIran
| | | | | | - Mohammad Reza Saeb
- Department of Polymer Technology, Faculty of ChemistryGdańsk University of TechnologyGdańskPoland
| | - Joshua D. Ramsey
- School of Chemical EngineeringOklahoma State UniversityStillwaterOklahomaUSA
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health ScienceUniversity of JohannesburgSouth Africa
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in MedicineIran University of Medical SciencesTehranIran
- Present address:
Lunenfeld‐Tanenbaum Research InstituteMount Sinai Hospital, University of TorontoTorontoONCanada.
| | - Ebrahim Mostafavi
- Stanford Cardiovascular InstituteStanford University School of MedicineStanfordCaliforniaUSA
- Department of MedicineStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
16
|
Stanislovas J, Kermorgant S. c-Met-integrin cooperation: Mechanisms, tumorigenic effects, and therapeutic relevance. Front Cell Dev Biol 2022; 10:994528. [PMID: 36330337 PMCID: PMC9624249 DOI: 10.3389/fcell.2022.994528] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
c-Met is a receptor tyrosine kinase which upon activation by its ligand, the hepatocyte growth factor, mediates many important signalling pathways that regulate cellular functions such as survival, proliferation, and migration. Its oncogenic and tumorigenic signalling mechanisms, greatly contributing to cancer development and progression, are well documented. Integrins, heterogeneous adhesion receptors which facilitate cell-extracellular matrix interactions, are important in biomechanically sensitive cell adhesion and motility but also modulate diverse cell behaviour. Here we review the studies which reported cooperation between c-Met and several integrins, particularly β1 and β4, in various cell models including many tumour cell types. From the various experimental models and results analysed, we propose that c-Met-integrin cooperation occurs via inside-out or outside-in signalling. Thus, either c-Met activation triggers integrin activation and cell adhesion or integrin adhesion to its extracellular ligand triggers c-Met activation. These two modes of cooperation require the adhesive function of integrins and mostly lead to cell migration and invasion. In a third, less conventional, mode of cooperation, the integrin plays the role of a signalling adaptor for c-Met, independently from its adhesive property, leading to anchorage independent survival. Recent studies have revealed the influence of endocytic trafficking in c-Met-integrin cooperation including the adaptor function of integrin occurring on endomembranes, triggering an inside-in signalling, believed to promote survival of metastatic cells. We present the evidence of the cooperation in vivo and in human tissues and highlight its therapeutic relevance. A better understanding of the mechanisms regulating c-Met-integrin cooperation in cancer progression could lead to the design of new therapies targeting this cooperation, providing more effective therapeutic approaches than c-Met or integrin inhibitors as monotherapies used in the clinic.
Collapse
Affiliation(s)
- Justas Stanislovas
- Spatial Signalling Group, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Stéphanie Kermorgant
- Spatial Signalling Group, John Vane Science Centre, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
17
|
Tang S, Davoudi Z, Wang G, Xu Z, Rehman T, Prominski A, Tian B, Bratlie KM, Peng H, Wang Q. Soft materials as biological and artificial membranes. Chem Soc Rev 2021; 50:12679-12701. [PMID: 34636824 DOI: 10.1039/d1cs00029b] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The past few decades have seen emerging growth in the field of soft materials for synthetic biology. This review focuses on soft materials involved in biological and artificial membranes. The biological membranes discussed here are mainly those involved in the structure and function of cells and organelles. As building blocks in medicine, non-native membranes including nanocarriers (NCs), especially liposomes and DQAsomes, and polymeric membranes for scaffolds are constructed from amphiphilic combinations of lipids, proteins, and carbohydrates. Artificial membranes can be prepared using synthetic, soft materials and molecules and then incorporated into structures through self-organization to form micelles or niosomes. The modification of artificial membranes can be realized using traditional chemical methods such as click reactions to target the delivery of NCs and control the release of therapeutics. The biomembrane, a lamellar structure inlaid with ion channels, receptors, lipid rafts, enzymes, and other functional units, separates cells and organelles from the environment. An active domain inserted into the membrane and organelles for energy conversion and cellular communication can target disease by changing the membrane's composition, structure, and fluidity and affecting the on/off status of the membrane gates. The biological membrane targets analyzing pathological mechanisms and curing complex diseases, which inspires us to create NCs with artificial membranes.
Collapse
Affiliation(s)
- Shukun Tang
- Department of Pharmaceutics, Daqing Branch, Harbin Medical University, Research and Development of Natural Products Key Laboratory of Harbin Medical University, 39 Xin Yang Road, Daqing, 163319, China.
| | - Zahra Davoudi
- Department of Chemical and Biological Engineering, Iowa State University, 1014 Sweeney Hall, Ames, IA 50011, USA.
| | - Guangtian Wang
- Department of Pharmaceutics, Daqing Branch, Harbin Medical University, Research and Development of Natural Products Key Laboratory of Harbin Medical University, 39 Xin Yang Road, Daqing, 163319, China.
| | - Zihao Xu
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011, USA
| | - Tanzeel Rehman
- Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011, USA
| | - Aleksander Prominski
- The James Franck Institute, Department of Chemistry, The Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, USA
| | - Bozhi Tian
- The James Franck Institute, Department of Chemistry, The Institute for Biophysical Dynamics, The University of Chicago, Chicago, Illinois 60637, USA
| | - Kaitlin M Bratlie
- Department of Chemical and Biological Engineering, Iowa State University, 1014 Sweeney Hall, Ames, IA 50011, USA. .,Department of Materials Science and Engineering, Iowa State University, Ames, IA 50011, USA
| | - Haisheng Peng
- Department of Pharmaceutics, Daqing Branch, Harbin Medical University, Research and Development of Natural Products Key Laboratory of Harbin Medical University, 39 Xin Yang Road, Daqing, 163319, China.
| | - Qun Wang
- Department of Chemical and Biological Engineering, Iowa State University, 1014 Sweeney Hall, Ames, IA 50011, USA.
| |
Collapse
|
18
|
Justo BL, Jasiulionis MG. Characteristics of TIMP1, CD63, and β1-Integrin and the Functional Impact of Their Interaction in Cancer. Int J Mol Sci 2021; 22:9319. [PMID: 34502227 PMCID: PMC8431149 DOI: 10.3390/ijms22179319] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 01/03/2023] Open
Abstract
Tissue Inhibitor of Metalloproteases 1, also known as TIMP-1, is named for its well-established function of inhibiting the proteolytic activity of matrix metalloproteases. Given this function, many studies were carried out to verify if TIMP-1 was able to interrupt processes such as tumor cell invasion and metastasis. In contrast, many studies have shown that TIMP-1 expression is increased in several types of tumors, and this increase was correlated with a poor prognosis and lower survival in cancer patients. Later, it was shown that TIMP-1 is also able to modulate cell behavior through the induction of signaling pathways involved in cell growth, proliferation, and survival. The mechanisms involved in the regulation of the pleiotropic functions of TIMP-1 are still poorly understood. Thus, this review aimed to present literature data that show its ability to form a membrane complex with CD63 and β1-integrin, and point to N-glycosylation as a potential regulatory mechanism of the functions exerted by TIMP-1. This article reviewed the characteristics and functions performed individually by TIMP1, CD63, and β1-integrin, the roles of the TIMP-1/CD63/β1-integrin complex, both in a physiological context and in cancer, and the regulatory mechanisms involved in its assembly.
Collapse
Affiliation(s)
| | - Miriam Galvonas Jasiulionis
- Department of Pharmacology, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), Rua Pedro de Toledo 669, 5 Floor, São Paulo 04039-032, Brazil;
| |
Collapse
|
19
|
Abstract
Phospholipase C γ1 (PLCγ1) is a member of the PLC family that functions as signal transducer by hydrolyzing membrane lipid to generate second messengers. The unique protein structure of PLCγ1 confers a critical role as a direct effector of VEGFR2 and signaling mediated by other receptor tyrosine kinases. The distinct vascular phenotypes in PLCγ1-deficient animal models and the gain-of-function mutations of PLCγ1 found in human endothelial cancers point to a major physiological role of PLCγ1 in the endothelial system. In this review, we discuss aspects of physiological and molecular function centering around PLCγ1 in the context of endothelial cells and provide a perspective for future investigation.
Collapse
Affiliation(s)
- Dongying Chen
- Yale Cardiovascular Research Center, Departments of Internal Medicine and Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Michael Simons
- Yale Cardiovascular Research Center, Departments of Internal Medicine and Cell Biology, Yale University School of Medicine, New Haven, CT 06511, USA.
| |
Collapse
|
20
|
Steenkiste EM, Berndt JD, Pilling C, Simpkins C, Cooper JA. A Cas-BCAR3 co-regulatory circuit controls lamellipodia dynamics. eLife 2021; 10:67078. [PMID: 34169835 PMCID: PMC8266394 DOI: 10.7554/elife.67078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/21/2021] [Indexed: 11/13/2022] Open
Abstract
Integrin adhesion complexes regulate cytoskeletal dynamics during cell migration. Adhesion activates phosphorylation of integrin-associated signaling proteins, including Cas (p130Cas, BCAR1), by Src-family kinases. Cas regulates leading-edge protrusion and migration in cooperation with its binding partner, BCAR3. However, it has been unclear how Cas and BCAR3 cooperate. Here, using normal epithelial cells, we find that BCAR3 localization to integrin adhesions requires Cas. In return, Cas phosphorylation, as well as lamellipodia dynamics and cell migration, requires BCAR3. These functions require the BCAR3 SH2 domain and a specific phosphorylation site, Tyr 117, that is also required for BCAR3 downregulation by the ubiquitin-proteasome system. These findings place BCAR3 in a co-regulatory positive-feedback circuit with Cas, with BCAR3 requiring Cas for localization and Cas requiring BCAR3 for activation and downstream signaling. The use of a single phosphorylation site in BCAR3 for activation and degradation ensures reliable negative feedback by the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Elizabeth M Steenkiste
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States.,Molecular and Cellular Biology Program, University of Washington, Seattle, United States
| | - Jason D Berndt
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Carissa Pilling
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States.,Molecular and Cellular Biology Program, University of Washington, Seattle, United States
| | - Christopher Simpkins
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Jonathan A Cooper
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States.,Molecular and Cellular Biology Program, University of Washington, Seattle, United States
| |
Collapse
|
21
|
Sarker FA, Prior VG, Bax S, O'Neill GM. Forcing a growth factor response - tissue-stiffness modulation of integrin signaling and crosstalk with growth factor receptors. J Cell Sci 2020; 133:133/23/jcs242461. [PMID: 33310867 DOI: 10.1242/jcs.242461] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Research throughout the 90s established that integrin crosstalk with growth factor receptors stimulates robust growth factor signaling. These insights were derived chiefly from comparing adherent versus suspension cell cultures. Considering the new understanding that mechanosensory inputs tune adhesion signaling, it is now timely to revisit this crosstalk in different mechanical environments. Here, we present a brief historical perspective on integrin signaling against the backdrop of the mechanically diverse extracellular microenvironment, then review the evidence supporting the mechanical regulation of integrin crosstalk with growth factor signaling. We discuss early studies revealing distinct signaling consequences for integrin occupancy (binding to matrix) and aggregation (binding to immobile ligand). We consider how the mechanical environments encountered in vivo intersect with this diverse signaling, focusing on receptor endocytosis. We discuss the implications of mechanically tuned integrin signaling for growth factor signaling, using the epidermal growth factor receptor (EGFR) as an illustrative example. We discuss how the use of rigid tissue culture plastic for cancer drug screening may select agents that lack efficacy in the soft in vivo tissue environment. Tuning of integrin signaling via external mechanical forces in vivo and subsequent effects on growth factor signaling thus has implications for normal cellular physiology and anti-cancer therapies.
Collapse
Affiliation(s)
- Farhana A Sarker
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead NSW, Westmead 2145, Australia.,Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney 2006, Australia
| | - Victoria G Prior
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead NSW, Westmead 2145, Australia.,Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney 2006, Australia
| | - Samuel Bax
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead NSW, Westmead 2145, Australia
| | - Geraldine M O'Neill
- Children's Cancer Research Unit, Kids Research Institute at the Children's Hospital at Westmead NSW, Westmead 2145, Australia .,Children's Hospital Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney 2006, Australia.,School of Medical Science, Faculty of Medicine and Health, University of Sydney, Sydney 2006, Australia
| |
Collapse
|
22
|
Frank JW, Steinhauser CB, Wang X, Burghardt RC, Bazer FW, Johnson GA. Loss of ITGB3 in ovine conceptuses decreases conceptus expression of NOS3 and SPP1: implications for the developing placental vasculature†. Biol Reprod 2020; 104:657-668. [PMID: 33232974 DOI: 10.1093/biolre/ioaa212] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/17/2020] [Accepted: 11/20/2020] [Indexed: 12/18/2022] Open
Abstract
During the peri-implantation period of pregnancy in sheep, there is an initial period of loose apposition of the elongating conceptuses (embryos and associated placental membranes) to the endometrial luminal epithelium (LE) that is followed by adhesion of the conceptus trophectoderm to the endometrial LE for implantation. Integrins and maternal extracellular matrix (ECM) molecules are major contributors to stable adhesion at implantation, and the β3 integrin subunit (ITGB3) is implicated in the adhesion cascade for implantation in several species including the sheep. We blocked mRNA translation for trophectoderm-expressed ITGB3 by infusing morpholino antisense oligonucleotides into the uterine lumen of pregnant ewes on Day 9 to assess effects on conceptus elongation, and on Day 16 to assess effects on early placental development in sheep. Results indicate that sheep conceptuses elongate and implant to the uterine wall in the absence of ITGB3 expression by the conceptuses; however, loss of ITGB3 in conceptuses decreased the growth of embryos to Day 24 of gestation, and decreased expression of secreted phosphoprotein 1 (SPP1) and nitric oxide synthase 3 (NOS3). Abundant SPP1 was localized around the blood vessels in the placental allantoic membrane in normal sheep pregnancies. We hypothesize that NOS3 and SPP1 positively influence the development of the vasculature within the allantois, and that decreased expression of NOS3 and SPP1, in response to knockdown of ITGB3 in conceptuses, alters development of the vasculature in the allantois required to transport nutrients from the endometrium to support growth and development of the embryo.
Collapse
Affiliation(s)
- James W Frank
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Chelsie B Steinhauser
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Xiaoqiu Wang
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Robert C Burghardt
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Fuller W Bazer
- Department of Animal Science, Texas A&M University, College Station, TX, USA
| | - Greg A Johnson
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| |
Collapse
|
23
|
Revach OY, Grosheva I, Geiger B. Biomechanical regulation of focal adhesion and invadopodia formation. J Cell Sci 2020; 133:133/20/jcs244848. [DOI: 10.1242/jcs.244848] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
ABSTRACT
Integrin adhesions are a structurally and functionally diverse family of transmembrane, multi-protein complexes that link the intracellular cytoskeleton to the extracellular matrix (ECM). The different members of this family, including focal adhesions (FAs), focal complexes, fibrillar adhesions, podosomes and invadopodia, contain many shared scaffolding and signaling ‘adhesome’ components, as well as distinct molecules that perform specific functions, unique to each adhesion form. In this Hypothesis, we address the pivotal roles of mechanical forces, generated by local actin polymerization or actomyosin-based contractility, in the formation, maturation and functionality of two members of the integrin adhesions family, namely FAs and invadopodia, which display distinct structures and functional properties. FAs are robust and stable ECM contacts, associated with contractile stress fibers, while invadopodia are invasive adhesions that degrade the underlying matrix and penetrate into it. We discuss here the mechanisms, whereby these two types of adhesion utilize a similar molecular machinery to drive very different – often opposing cellular activities, and hypothesize that early stages of FAs and invadopodia assembly use similar biomechanical principles, whereas maturation of the two structures, and their ‘adhesive’ and ‘invasive’ functionalities require distinct sources of biomechanical reinforcement.
Collapse
Affiliation(s)
- Or-Yam Revach
- Departments of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Inna Grosheva
- Departments of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
- Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Benjamin Geiger
- Departments of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
- Immunology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
24
|
Temporal changes guided by mesenchymal stem cells on a 3D microgel platform enhance angiogenesis in vivo at a low-cell dose. Proc Natl Acad Sci U S A 2020; 117:19033-19044. [PMID: 32709748 PMCID: PMC7430977 DOI: 10.1073/pnas.2008245117] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Therapeutic factors secreted by mesenchymal stem cells (MSCs) promote angiogenesis in vivo. However, delivery of MSCs in the absence of a cytoprotective environment offers limited efficacy due to low cell retention, poor graft survival, and the nonmaintenance of a physiologically relevant dose of growth factors at the injury site. The delivery of stem cells on an extracellular matrix (ECM)-based platform alters cell behavior, including migration, proliferation, and paracrine activity, which are essential for angiogenesis. We demonstrate the biophysical and biochemical effects of preconditioning human MSCs (hMSCs) for 96 h on a three-dimensional (3D) ECM-based microgel platform. By altering the macromolecular concentration surrounding cells in the microgels, the proangiogenic phenotype of hMSCs can be tuned in a controlled manner through cell-driven changes in extracellular stiffness and "outside-in" integrin signaling. The softest microgels were tested at a low cell dose (5 × 104 cells) in a preclinical hindlimb ischemia model showing accelerated formation of new blood vessels with a reduced inflammatory response impeding progression of tissue damage. Molecular analysis revealed that several key mediators of angiogenesis were up-regulated in the low-cell-dose microgel group, providing a mechanistic insight of pathways modulated in vivo. Our research adds to current knowledge in cell-encapsulation strategies by highlighting the importance of preconditioning or priming the capacity of biomaterials through cell-material interactions. Obtaining therapeutic efficacy at a low cell dose in the microgel platform is a promising clinical route that would aid faster tissue repair and reperfusion in "no-option" patients suffering from peripheral arterial diseases, such as critical limb ischemia (CLI).
Collapse
|
25
|
Novel fibrin-fibronectin matrix accelerates mice skin wound healing. Bioact Mater 2020; 5:949-962. [PMID: 32671290 PMCID: PMC7334397 DOI: 10.1016/j.bioactmat.2020.06.015] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 01/13/2023] Open
Abstract
Plasma fibrinogen (F1) and fibronectin (pFN) polymerize to form a fibrin clot that is both a hemostatic and provisional matrix for wound healing. About 90% of plasma F1 has a homodimeric pair of γ chains (γγF1), and 10% has a heterodimeric pair of γ and more acidic γ' chains (γγ'F1). We have synthesized a novel fibrin matrix exclusively from a 1:1 (molar ratio) complex of γγ'F1 and pFN in the presence of highly active thrombin and recombinant Factor XIII (rFXIIIa). In this matrix, the fibrin nanofibers were decorated with pFN nanoclusters (termed γγ'F1:pFN fibrin). In contrast, fibrin made from 1:1 mixture of γγF1 and pFN formed a sporadic distribution of "pFN droplets" (termed γγF1+pFN fibrin). The γγ'F1:pFN fibrin enhanced the adhesion of primary human umbilical vein endothelium cells (HUVECs) relative to the γγF1+FN fibrin. Three dimensional (3D) culturing showed that the γγ'F1:pFN complex fibrin matrix enhanced the proliferation of both HUVECs and primary human fibroblasts. HUVECs in the 3D γγ'F1:pFN fibrin exhibited a starkly enhanced vascular morphogenesis while an apoptotic growth profile was observed in the γγF1+pFN fibrin. Relative to γγF1+pFN fibrin, mouse dermal wounds that were sealed by γγ'F1:pFN fibrin exhibited accelerated and enhanced healing. This study suggests that a 3D pFN presentation on a fibrin matrix promotes wound healing.
Collapse
|
26
|
Guo X, Eitnier RA, Beard RS, Meegan JE, Yang X, Aponte AM, Wang F, Nelson PR, Wu MH. Focal adhesion kinase and Src mediate microvascular hyperpermeability caused by fibrinogen- γC- terminal fragments. PLoS One 2020; 15:e0231739. [PMID: 32352989 PMCID: PMC7192500 DOI: 10.1371/journal.pone.0231739] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 03/30/2020] [Indexed: 12/31/2022] Open
Abstract
Objectives We previously reported microvascular leakage resulting from fibrinogen-γ chain C-terminal products (γC) occurred via a RhoA-dependent mechanism. The objective of this study was to further elucidate the signaling mechanism by which γC induces endothelial hyperpermeability. Since it is known that γC binds and activates endothelial αvβ3, a transmembrane integrin receptor involved in intracellular signaling mediated by the tyrosine kinases FAK and Src, we hypothesized that γC alters endothelial barrier function by activating the FAK-Src pathway leading to junction dissociation and RhoA driven cytoskeletal stress-fiber formation. Methods and results Using intravital microscopy of rat mesenteric microvessels, we show increased extravasation of plasma protein (albumin) resulting from γC administration. In addition, capillary fluid filtration coefficient (Kfc) indicated γC-induced elevated lung vascular permeability. Furthermore, γC decreased transendothelial barrier resistance in a time-dependent and dose-related fashion in cultured rat lung microvascular endothelial cells (RLMVECs), accompanied by increased FAK/Src phosphorylation detection by western blot. Experiments with pharmacological inhibition or gene silencing of FAK showed significantly reduced γC-induced albumin and fluid leakage across microvessels, stress-fiber formation, VE-cadherin tyrosine phosphorylation, and improved γC-induced endothelial barrier dysfunction, indicating the involvement of FAK in γC mediated hyperpermeability. Comparable results were found when Src was targeted in a similar manner, however inhibition of FAK prevented Src activation, suggesting that FAK is upstream of Src in γC-mediated hyperpermeability. In addition, γC-induced cytoskeletal stress-fiber formation was attenuated during inhibition or silencing of these tyrosine kinases, concomitantly with RhoA inhibition. Conclusion The FAK-Src pathway contributes to γC-induced microvascular barrier dysfunction, junction protein phosphorylation and disorganization in a manner that involves RhoA and stress-fiber formation.
Collapse
Affiliation(s)
- Xiaohua Guo
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
| | - Rebecca A. Eitnier
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
| | - Richard S. Beard
- Department of Biomolecular Research, Boise State University, Boise, ID, United States of America
| | - Jamie E. Meegan
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
| | - Xiaoyuan Yang
- Department of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
| | - Alexandra M. Aponte
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
| | - Fang Wang
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
| | - Peter R. Nelson
- Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Mack H. Wu
- Department of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL, United States of America
- * E-mail:
| |
Collapse
|
27
|
Baek J, Cho Y, Park HJ, Choi G, Lee JS, Lee M, Yu SJ, Cho SW, Lee E, Im SG. A Surface-Tailoring Method for Rapid Non-Thermosensitive Cell-Sheet Engineering via Functional Polymer Coatings. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1907225. [PMID: 32157771 DOI: 10.1002/adma.201907225] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/30/2019] [Accepted: 02/18/2020] [Indexed: 06/10/2023]
Abstract
Cell sheet engineering, a technique utilizing a monolayer cell sheet, has recently emerged as a promising technology for scaffold-free tissue engineering. In contrast to conventional tissue-engineering approaches, the cell sheet technology allows cell harvest as a continuous cell sheet with intact extracellular matrix proteins and cell-cell junction, which facilitates cell transplantation without any other artificial biomaterials. A facile, non-thermoresponsive method is demonstrated for a rapid but highly reliable platform for cell-sheet engineering. The developed method exploits the precise modulation of cell-substrate interactions by controlling the surface energy of the substrate via a series of functional polymer coatings to enable prompt cell sheet harvesting within 100 s. The engineered surface can trigger an intrinsic cellular response upon the depletion of divalent cations, leading to spontaneous cell sheet detachment under physiological conditions (pH 7.4 and 37 °C) in a non-thermoresponsive manner. Additionally, the therapeutic potential of the cell sheet is successfully demonstrated by the transplantation of multilayered cell sheets into mouse models of diabetic wounds and ischemia. These findings highlight the ability of the developed surface for non-thermoresponsive cell sheet engineering to serve as a robust platform for regenerative medicine and provide significant breakthroughs in cell sheet technology.
Collapse
Affiliation(s)
- Jieung Baek
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Younghak Cho
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Hyun-Ji Park
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Goro Choi
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jong Seung Lee
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Minseok Lee
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Seung Jung Yu
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, 03772, Republic of Korea
- Yonsei-IBS Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Eunjung Lee
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering and KI for NanoCentury, Korea Advanced Institute of Science and Technology, 291 Daehak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| |
Collapse
|
28
|
Otasevic V, Stancic A, Korac A, Jankovic A, Korac B. Reactive oxygen, nitrogen, and sulfur species in human male fertility. A crossroad of cellular signaling and pathology. Biofactors 2020; 46:206-219. [PMID: 31185138 DOI: 10.1002/biof.1535] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 05/17/2019] [Indexed: 12/23/2022]
Abstract
Infertility is a significant global health problem that currently affects one of six couples in reproductive age. The quality of male reproductive cells dramatically decreased over the last years and almost every aspect of modern life additionally worsen sperm functional parameters that consequently markedly increase male infertility. This clearly points out the importance of finding a new approach to treat male infertility. Redox signaling mediated by reactive oxygen, nitrogen and sulfur species (ROS, RNS, and RSS respectively), has appeared important for sperm reproductive function. Present review summarizes the current knowledge of ROS, RNS, and RSS in male reproductive biology and identifies potential targets for development of novel pharmacological and therapeutic approaches for male infertility by targeted therapeutic modulation of redox signaling.
Collapse
Affiliation(s)
- Vesna Otasevic
- University of Belgrade, Institute for Biological Research "Sinisa Stankovic", Belgrade, Serbia
| | - Ana Stancic
- University of Belgrade, Institute for Biological Research "Sinisa Stankovic", Belgrade, Serbia
| | - Aleksandra Korac
- University of Belgrade, Faculty of Biology, Center for Electron Microscopy, Belgrade, Serbia
| | - Aleksandra Jankovic
- University of Belgrade, Institute for Biological Research "Sinisa Stankovic", Belgrade, Serbia
| | - Bato Korac
- University of Belgrade, Institute for Biological Research "Sinisa Stankovic", Belgrade, Serbia
| |
Collapse
|
29
|
Rosalem GS, Las Casas EB, Lima TP, González-Torres LA. A mechanobiological model to study upstream cell migration guided by tensotaxis. Biomech Model Mechanobiol 2020; 19:1537-1549. [PMID: 32006123 DOI: 10.1007/s10237-020-01289-5] [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] [Received: 07/23/2019] [Accepted: 01/11/2020] [Indexed: 01/06/2023]
Abstract
Cell migration is a process of crucial importance for the human body. It is responsible for important processes such as wound healing and tumor metastasis. Migration may occur in response to stimuli of chemical, physical and mechanical nature occurring in the cellular microenvironment. The interstitial flow (IF) can generate mechanical stimuli in cells that influence the cell behavior and interactions of the cells with the extracellular matrix (ECM). One of the phenomena is upstream migration, which is observed in some tumors. In this work, we present a new approach to study the adherent cell migration in a porous medium using a mechanobiological model, attempting to understand if upstream migration can be generated exclusively by mechanical factors. The influence of IF on the behavior of cells and the extracellular matrix was considered. The model is based on a system of coupled nonlinear differential equations solved by the finite element method. Several simulations were performed to study the upstream cell migration and evaluate the effects of pressure, permeability, ECM stiffness and cellular concentration variations on the cell velocity. The results indicated that upstream migration can occur in the presence of mechanical stimuli generated by IF and that the tested parameters have a direct influence on the cellular velocity, especially the pressure and the permeability.
Collapse
Affiliation(s)
- Gabriel Santos Rosalem
- Department of Mechanical Engineering, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Thiago Parente Lima
- Institute of Science and Technology, Federal University of Jequitinhonha and Mucuri Valleys, Diamantina, Brazil
| | | |
Collapse
|
30
|
Jakubec M, Bariås E, Kryuchkov F, Hjørnevik LV, Halskau Ø. Fast and Quantitative Phospholipidomic Analysis of SH-SY5Y Neuroblastoma Cell Cultures Using Liquid Chromatography-Tandem Mass Spectrometry and 31P Nuclear Magnetic Resonance. ACS OMEGA 2019; 4:21596-21603. [PMID: 31867556 PMCID: PMC6921604 DOI: 10.1021/acsomega.9b03463] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 11/08/2019] [Indexed: 05/04/2023]
Abstract
Global lipid analysis still lags behind proteomics with respect to the availability of databases, experimental protocols, and specialized software. Determining the lipidome of cellular model systems in common use is of particular importance, especially when research questions involve lipids directly. In Parkinson's disease research, there is a growing awareness for the role of the biological membrane, where individual lipids may contribute to provoking α-synuclein oligomerisation and fibrillation. We present an analysis of the whole cell and plasma membrane lipid isolates of a neuroblastoma cell line, SH-SY5Y, a commonly used model system for research on this and other neurodegenerative diseases. We have used two complementary lipidomics methods. The relative quantities of PC, PE, SMs, CL, PI, PG, and PS were determined by 31P NMR. Fatty acid chain composition and their relative abundances within each phospholipid group were evaluated by liquid chromatography-tandem mass spectrometry. For this part of the analysis, we have developed and made available a set of Matlab scripts, LipMat. Our approach allowed us to observe several deviations of lipid abundances when compared to published reports regarding phospholipid analysis of cell cultures or brain matter. The most striking was the high abundance of PC (54.7 ± 1.9%) and low abundance of PE (17.8 ± 4.8%) and SMs (2.7 ± 1.2%). In addition, the observed abundance of PS was smaller than expected (4.7 ± 2.7%), similar to the observed abundance of PG (4.5 ± 1.8%). The observed fatty acid chain distribution was similar to the whole brain content with some notable differences: a higher abundance of 16:1 PC FA (17.4 ± 3.4% in PC whole cell content), lower abundance of 22:6 PE FA (15.9 ± 2.2% in plasma membrane fraction), and a complete lack of 22:6 PS FA.
Collapse
Affiliation(s)
- Martin Jakubec
- Faculty of Mathematics
and Natural Sciences, Department of Biological Sciences, University of Bergen, PB 7803, Bergen NO 5020, Norway
| | - Espen Bariås
- Faculty of Mathematics
and Natural Sciences, Department of Biological Sciences, University of Bergen, PB 7803, Bergen NO 5020, Norway
| | - Fedor Kryuchkov
- Faculty of Veterinary and Biosciences, Norwegian University of Life Sciences, Ullevålsveien 68, Oslo, Akershus NO 0033, Norway
| | - Linda Veka Hjørnevik
- Faculty of Mathematics
and Natural Sciences, Department of Biological Sciences, University of Bergen, PB 7803, Bergen NO 5020, Norway
| | - Øyvind Halskau
- Faculty of Mathematics
and Natural Sciences, Department of Biological Sciences, University of Bergen, PB 7803, Bergen NO 5020, Norway
- E-mail:
| |
Collapse
|
31
|
Deville SS, Cordes N. The Extracellular, Cellular, and Nuclear Stiffness, a Trinity in the Cancer Resistome-A Review. Front Oncol 2019; 9:1376. [PMID: 31867279 PMCID: PMC6908495 DOI: 10.3389/fonc.2019.01376] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Alterations in mechano-physiological properties of a tissue instigate cancer burdens in parallel to common genetic and epigenetic alterations. The chronological and mechanistic interrelation between the various extra- and intracellular aspects remains largely elusive. Mechano-physiologically, integrins and other cell adhesion molecules present the main mediators for transferring and distributing forces between cells and the extracellular matrix (ECM). These cues are channeled via focal adhesion proteins, termed the focal adhesomes, to cytoskeleton and nucleus and vice versa thereby affecting the pathophysiology of multicellular cancer tissues. In combination with simultaneous activation of diverse downstream signaling pathways, the phenotypes of cancer cells are created and driven characterized by deregulated transcriptional and biochemical cues that elicit the hallmarks of cancer. It, however, remains unclear how elastostatic modifications, i.e., stiffness, in the extracellular, intracellular, and nuclear compartment contribute and control the resistance of cancer cells to therapy. In this review, we discuss how stiffness of unique tumor components dictates therapy response and what is known about the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Sara Sofia Deville
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | - Nils Cordes
- OncoRay – National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Helmholtz-Zentrum Dresden - Rossendorf, Technische Universität Dresden, Dresden, Germany
- Department of Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Helmholtz-Zentrum Dresden - Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
- German Cancer Consortium (DKTK), Dresden, Germany
- Germany German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
32
|
Turner KR, Adams C, Staelens S, Deckmyn H, San Antonio J. Crucial Role for Endothelial Cell α2β1 Integrin Receptor Clustering in Collagen-Induced Angiogenesis. Anat Rec (Hoboken) 2019; 303:1604-1618. [PMID: 31581346 DOI: 10.1002/ar.24277] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 04/11/2019] [Accepted: 04/26/2019] [Indexed: 11/07/2022]
Abstract
Angiogenesis is a crucial mechanism of vascular growth and regeneration that requires biosynthesis and cross-linking of collagens in vivo and is induced by collagen in vitro. Here, we use an in vitro model in which apical Type I collagen gels rapidly induce angiogenesis in endothelial monolayers. We extend previous studies demonstrating the importance of the endothelial α2β1 integrin, a key collagen receptor, in angiogenesis by investigating the roles of receptor clustering and conformational activation. Immunocytochemical localization of α2β1 integrins in endothelial monolayers showed a concentration of integrins along cell-cell borders. After inducing angiogenesis with collagen, the receptors redistributed to apical cell surfaces, aligning with collagen fibers, which were also redistributed during angiogenesis. Levels of conformationally activated α2β1 integrins were unchanged during angiogenesis and undetected on endothelial cells binding collagen in suspension. We mimicked the polyvalency of collagen fibrils using antibody-coated polystyrene beads to cluster endothelial cell surface α2β1 integrins, which induced rapid angiogenesis in the absence of collagen gels. Clustering of αvβ3 integrins and PECAM-1 but not of α1 integrins also induced angiogenesis. Soluble antibodies alone had no effect. Thus, the angiogenic property of collagen may reside in its ability to ligate and cluster cell surface receptors such as α2β1 integrins. Furthermore, synthetic substrates that promote the clustering of select endothelial cell surface receptors mimic the angiogenic properties of Type I collagen and may have applications in promoting vascularization of engineered tissues. Anat Rec, 2019. © 2019 American Association for Anatomy.
Collapse
Affiliation(s)
- Kevin R Turner
- Cardeza Foundation for Hematologic Research, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania.,Department of Pathology, Oregon Health and Science University, Portland, Oregon
| | - Christopher Adams
- Department of Anatomy, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania
| | - Stephanie Staelens
- Agrosavfe NV, Ghent, Zwijnaarde, Belgium.,Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Hans Deckmyn
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - James San Antonio
- Cardeza Foundation for Hematologic Research, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
33
|
Mosier JA, Rahman-Zaman A, Zanotelli MR, VanderBurgh JA, Bordeleau F, Hoffman BD, Reinhart-King CA. Extent of Cell Confinement in Microtracks Affects Speed and Results in Differential Matrix Strains. Biophys J 2019; 117:1692-1701. [PMID: 31623884 DOI: 10.1016/j.bpj.2019.09.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 08/15/2019] [Accepted: 09/18/2019] [Indexed: 12/27/2022] Open
Abstract
During metastasis, cancer cells navigate through a spatially heterogeneous extracellular matrix (ECM). Physical properties of ECM, including the degree of confinement, influence cell migration behavior. Here, utilizing in vitro three-dimensional collagen microtracks, we demonstrate that cell-ECM interactions, specifically the degree of spatial confinement, regulate migratory behavior. We found that cells migrate faster when they are fully confined, contacting all four walls (top, bottom, and two sides) of a collagen microtrack, compared with cells that are partially confined, contacting less than four walls. When fully confined, cells exhibit fewer but larger vinculin-containing adhesions and create greater strains in the surrounding matrix directed toward the cell body. In contrast, partially confined cells develop a more elongated morphology with smaller but significantly more vinculin-containing adhesions and displace the surrounding matrix less than fully confined cells. The resulting effect of increasing cell contractility via Rho activation is dependent on the number of walls with which the cell is in contact. Although matrix strains increase in both fully and partially confined cells, cells that are partially confined increase speed, whereas those in full confinement decrease speed. Together, these results suggest that the degree of cell-ECM contact during confined migration is a key determinant of speed, morphology, and cell-generated substrate strains during motility, and these factors may work in tandem to facilitate metastatic cell migration.
Collapse
Affiliation(s)
- Jenna A Mosier
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
| | - Aniqua Rahman-Zaman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Matthew R Zanotelli
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jacob A VanderBurgh
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Francois Bordeleau
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Dép. Biologie Moléculaire, Biochimie Médicale et Pathologie, Université Laval, Québec, Canada
| | - Brenton D Hoffman
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Cynthia A Reinhart-King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee; Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.
| |
Collapse
|
34
|
Jang YH, Jin X, Shankar P, Lee JH, Jo K, Lim KI. Molecular-Level Interactions between Engineered Materials and Cells. Int J Mol Sci 2019; 20:E4142. [PMID: 31450647 PMCID: PMC6747072 DOI: 10.3390/ijms20174142] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/19/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022] Open
Abstract
Various recent experimental observations indicate that growing cells on engineered materials can alter their physiology, function, and fate. This finding suggests that better molecular-level understanding of the interactions between cells and materials may guide the design and construction of sophisticated artificial substrates, potentially enabling control of cells for use in various biomedical applications. In this review, we introduce recent research results that shed light on molecular events and mechanisms involved in the interactions between cells and materials. We discuss the development of materials with distinct physical, chemical, and biological features, cellular sensing of the engineered materials, transfer of the sensing information to the cell nucleus, subsequent changes in physical and chemical states of genomic DNA, and finally the resulting cellular behavior changes. Ongoing efforts to advance materials engineering and the cell-material interface will eventually expand the cell-based applications in therapies and tissue regenerations.
Collapse
Affiliation(s)
- Yoon-Ha Jang
- Department of Chemical and Biological Engineering, Sookmyung Women's University, Seoul 04310, Korea
| | - Xuelin Jin
- Department of Chemistry and Integrated Biotechnology, Sogang University, Seoul 04107, Korea
| | - Prabakaran Shankar
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea
| | - Jung Heon Lee
- School of Advanced Materials Science and Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Korea.
| | - Kyubong Jo
- Department of Chemistry and Integrated Biotechnology, Sogang University, Seoul 04107, Korea.
| | - Kwang-Il Lim
- Department of Chemical and Biological Engineering, Sookmyung Women's University, Seoul 04310, Korea.
| |
Collapse
|
35
|
Moura CEB, Queiroz Neto MF, Braz JKFS, de Medeiros Aires M, Silva Farias NB, Barboza CAG, Cavalcanti Júnior GB, Rocha HAO, Alves Junior C. Effect of plasma-nitrided titanium surfaces on the differentiation of pre-osteoblastic cells. Artif Organs 2019; 43:764-772. [PMID: 30779451 DOI: 10.1111/aor.13438] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 01/07/2019] [Accepted: 02/16/2019] [Indexed: 12/15/2022]
Abstract
A titanium surface nitrided by plasma contains nitrogen ions that guarantee resistance to corrosion and biocompatibility. Despite this, no descriptions concerning the influence of the expression of cell adhesion proteins and their influence on osteogenic cell differentiation are available. Thus, the present study aimed to assess the response of murine pre-osteoblastic cells (MC3T3-E1) cultured on nitrided titanium surfaces. Pre-osteoblastic cells were grown on polished titanium discs, used as controls, and on previously characterized plasma-nitrided titanium discs. Cells from both groups were submitted to the MTT cell viability test. The expressions of α5, α2, and β1 integrin were assessed by flow cytometry and immunofluorescence, while osteocalcin expression was assessed by flow cytometry. The nitrided surface presented higher α2 and β1 integrin expressions, as well as osteocalcin expression, when compared to the polished surface, with no alterations in cell viability. These findings seem to suggest that the plasma nitriding treatment produces a titanium surface with the potential for effective in vitro osseointegration.
Collapse
Affiliation(s)
- Carlos Eduardo B Moura
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | - Moacir F Queiroz Neto
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | - Janine Karla F S Braz
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | | | - Naisandra B Silva Farias
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | - Carlos Augusto G Barboza
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | | | - Hugo Alexandre O Rocha
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| | - Clodomiro Alves Junior
- Department of Animal Sciences, Federal Rural University of Semiarid Region (UFERSA), Mossoró, Brazil
| |
Collapse
|
36
|
Jiang T, Zhao J, Yu S, Mao Z, Gao C, Zhu Y, Mao C, Zheng L. Untangling the response of bone tumor cells and bone forming cells to matrix stiffness and adhesion ligand density by means of hydrogels. Biomaterials 2019; 188:130-143. [PMID: 30343256 PMCID: PMC6279509 DOI: 10.1016/j.biomaterials.2018.10.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/14/2018] [Accepted: 10/14/2018] [Indexed: 12/22/2022]
Abstract
How cancer cells and their anchorage-dependent normal counterparts respond to the adhesion ligand density and stiffness of the same extracellular matrix (ECM) is still not very clear. Here we investigated the effects of ECM adhesion ligand density and stiffness on bone tumor cells (osteosarcoma cells) and bone forming cells (osteoblasts) by using poly (ethylene glycol) diacrylate (PEGDA) and methacrylated gelatin (GelMA) hydrogels. By independently changing the PEGDA and GelMA content in the hydrogels, we achieved crosslinked hydrogel matrix with independently tunable stiffness (1.6, 6 and 25 kPa for 5%, 10%, 15% PEDGA, respectively) and adhesion ligand density (low, medium and high for 0.05%, 0.2%, 0.5% GelMA respectively). By using a series of biochemical and cell biological characterizations as well as in vivo studies, we confirmed that osteosarcoma and osteoblastic cells responded differently to the stiffness and adhesion ligand density within 3D ECM. When cultured within the 3D PEGDA/GelMA hydrogel matrix, osteosarcoma cells are highly dependent on the matrix stiffness via regulating the integrin-mediated focal adhesion (FA) pathway, whereas osteoblasts are highly sensitive to the matrix adhesion ligand density through regulating the integrin-mediated adherens junction (AJ) pathway. However, when seeded on the 2D surface of the hydrogels, osteosarcoma cells behaved differently and became sensitive to the matrix adhesion ligand density because they were "forced" to attach to the substrate, similar to anchorage-dependent osteoblasts. This study might provide new insights into rational design of scaffolds for generating in vitro tumor models to test anticancer therapeutics and for regenerating tissue to repair defects.
Collapse
Affiliation(s)
- Tongmeng Jiang
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Jinmin Zhao
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China; Department of Orthopaedics Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China; Guangxi Key Laboratory of Regenerative Medicine, International Joint Laboratory on Regeneration of Bone and Soft Tissue, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Shan Yu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 38# Zheda Road, Hangzhou, 310027, China
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 38# Zheda Road, Hangzhou, 310027, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 38# Zheda Road, Hangzhou, 310027, China
| | - Ye Zhu
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Institute for Biomedical Engineering, Sience and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019-5300, USA
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Sciences Research Center, Institute for Biomedical Engineering, Sience and Technology, University of Oklahoma, 101 Stephenson Parkway, Norman, OK 73019-5300, USA; School of Materials Science and Engineering, Zhejiang University, Hangzhou, 310027, China.
| | - Li Zheng
- Guangxi Engineering Center in Biomedical Materials for Tissue and Organ Regeneration & Guangxi Collaborative Innovation Center for Biomedicine, Life Sciences Institute, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
37
|
Afewerki T, Ahmed S, Warren D. Emerging regulators of vascular smooth muscle cell migration. J Muscle Res Cell Motil 2019; 40:185-196. [PMID: 31254136 PMCID: PMC6726670 DOI: 10.1007/s10974-019-09531-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 06/21/2019] [Indexed: 12/30/2022]
Abstract
Vascular smooth muscle cells (VSMCs) are the predominant cell type in the blood vessel wall and normally adopt a quiescent, contractile phenotype. VSMC migration is tightly controlled, however, disease associated changes in the soluble and insoluble environment promote VSMC migration. Classically, studies investigating VSMC migration have described the influence of soluble factors. Emerging data has highlighted the importance of insoluble factors, including extracellular matrix stiffness and porosity. In this review, we will recap on the important signalling pathways that regulate VSMC migration and reflect on the potential importance of emerging regulators of VSMC function.
Collapse
Affiliation(s)
- TecLino Afewerki
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ UK
| | - Sultan Ahmed
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ UK
| | - Derek Warren
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ UK
| |
Collapse
|
38
|
Protein-protein interactions reveal key canonical pathways, upstream regulators, interactome domains, and novel targets in ALS. Sci Rep 2018; 8:14732. [PMID: 30283000 PMCID: PMC6170493 DOI: 10.1038/s41598-018-32902-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/18/2018] [Indexed: 02/08/2023] Open
Abstract
Developing effective treatment strategies for neurodegenerative diseases require an understanding of the underlying cellular pathways that lead to neuronal vulnerability and progressive degeneration. To date, numerous mutations in 147 distinct genes are identified to be "associated" with, "modifier" or "causative" of amyotrophic lateral sclerosis (ALS). Protein products of these genes and their interactions helped determine the protein landscape of ALS, and revealed upstream modulators, key canonical pathways, interactome domains and novel therapeutic targets. Our analysis originates from known human mutations and circles back to human, revealing increased PPARG and PPARGC1A expression in the Betz cells of sALS patients and patients with TDP43 pathology, and emphasizes the importance of lipid homeostasis. Downregulation of YWHAZ, a 14-3-3 protein, and cytoplasmic accumulation of ZFYVE27 especially in diseased Betz cells of ALS patients reinforce the idea that perturbed protein communications, interactome defects, and altered converging pathways will reveal novel therapeutic targets in ALS.
Collapse
|
39
|
Okada T, Ogura T. Nanoscale imaging of the adhesion core including integrin β1 on intact living cells using scanning electron-assisted dielectric-impedance microscopy. PLoS One 2018; 13:e0204133. [PMID: 30235285 PMCID: PMC6147470 DOI: 10.1371/journal.pone.0204133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 09/03/2018] [Indexed: 12/13/2022] Open
Abstract
The integrins are a superfamily of transmembrane proteins composed of α and β subunit dimers involved in cell-cell and cell-extracellular matrix interactions. The largest integrin subgroup is integrin β1, which contributes to several malignant phenotypes. Recently, we have developed a novel imaging technology named scanning electron-assisted dielectric-impedance microscopy (SE-ADM), which visualizes untreated living mammalian cells in aqueous conditions with high contrast. Using the SE-ADM system, we observed 60-nm gold colloids with antibodies directly binding to the focal adhesion core containing integrin β1 on mammalian cancer cells without staining and fixation. The adhesion core contains three or four high-density regions of integrin β1 and connects to the actin filament. An adhesion core with high-density integrin β1 is suggested to contain 10-20 integrin dimers. Our SE-ADM system can also visualize various other membrane proteins in living cells in medium without staining and fixation.
Collapse
Affiliation(s)
- Tomoko Okada
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Higashi, Tsukuba, Ibaraki, Japan
| | - Toshihiko Ogura
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), Higashi, Tsukuba, Ibaraki, Japan
- * E-mail:
| |
Collapse
|
40
|
Rittweger J, Albracht K, Flück M, Ruoss S, Brocca L, Longa E, Moriggi M, Seynnes O, Di Giulio I, Tenori L, Vignoli A, Capri M, Gelfi C, Luchinat C, Francheschi C, Bottinelli R, Cerretelli P, Narici M. Sarcolab pilot study into skeletal muscle's adaptation to long-term spaceflight. NPJ Microgravity 2018; 4:18. [PMID: 30246141 PMCID: PMC6141586 DOI: 10.1038/s41526-018-0052-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 07/04/2018] [Accepted: 07/10/2018] [Indexed: 12/31/2022] Open
Abstract
Spaceflight causes muscle wasting. The Sarcolab pilot study investigated two astronauts with regards to plantar flexor muscle size, architecture, and function, and to the underlying molecular adaptations in order to further the understanding of muscular responses to spaceflight and exercise countermeasures. Two crew members (A and B) spent 6 months in space. Crew member A trained less vigorously than B. Postflight, A showed substantial decrements in plantar flexor volume, muscle architecture, in strength and in fiber contractility, which was strongly mitigated in B. The difference between these crew members closely reflected FAK-Y397 abundance, a molecular marker of muscle's loading history. Moreover, crew member A showed downregulation of contractile proteins and enzymes of anaerobic metabolism, as well as of systemic markers of energy and protein metabolism. However, both crew members exhibited decrements in muscular aerobic metabolism and phosphate high energy transfer. We conclude that countermeasures can be effective, particularly when resistive forces are of sufficient magnitude. However, to fully prevent space-related muscular deterioration, intersubject variability must be understood, and intensive exercise countermeasures programs seem mandatory. Finally, proteomic and metabolomic analyses suggest that exercise benefits in space may go beyond mere maintenance of muscle mass, but rather extend to the level of organismic metabolism.
Collapse
Affiliation(s)
- Jörn Rittweger
- 1Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany.,2Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Kirsten Albracht
- 3Faculty of Medical Engineering and Technomathematics, FH Aachen University of Applied Science Aachen, Aachen, Germany.,4Institute of Biomechanics and Orthopaedics, German Sport University, Cologne, Germany
| | - Martin Flück
- 5Department of Orthopaedics, University of Zürich, Zürich, Switzerland
| | - Severin Ruoss
- 5Department of Orthopaedics, University of Zürich, Zürich, Switzerland
| | - Lorenza Brocca
- 6Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Emanuela Longa
- 6Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | | | - Olivier Seynnes
- 8Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Irene Di Giulio
- 9Centre for Human and Applied Physiological Sciences, King's College London, London, UK
| | - Leonardo Tenori
- 10Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessia Vignoli
- CERM Centro di Ricerca di Risonanze Magnetiche, Florence, Italy
| | - Miriam Capri
- 12Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Cecilia Gelfi
- 13Department of Biomedical Sciences for Health, University of Milan, Milano, Italy
| | | | - Claudio Francheschi
- 12Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Roberto Bottinelli
- 6Department of Molecular Medicine, University of Pavia, Pavia, Italy.,14Fondazione Salvatore Maugeri (IRCSS), Scientific Institute of Pavia, Pavia, Italy
| | | | - Marco Narici
- 15Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
41
|
Barr SP, Hill EW, Bayat A. Novel Proteomic Assay of Breast Implants Reveals Proteins With Significant Binding Differences: Implications for Surface Coating and Biocompatibility. Aesthet Surg J 2018; 38:962-969. [PMID: 29590306 DOI: 10.1093/asj/sjy018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Silicone elastomer, a ubiquitous biomaterial and main constituent of breast implants, has been used for breast augmentation and reconstruction for over 50 years. Breast implants have direct local and purported systemic effects on normal tissue homeostasis dictated by the chemical and physical presence of the implant. OBJECTIVES Protein adsorption has been demonstrated to be a key driver of local reactions to silicone. We sought to develop an assay and identify the proteins that coat implants during breast implantation. METHODS Wound fluid was salvaged from women who had undergone breast reduction and incubated in contact with the surface of 13 commercially available implant surfaces. An in situ digestion technique was optimized to elute bound proteins. Samples were analyzed on an Orbitrap elite analyser, proteins identified in Mascot Demon and analyzed in Progenesis. RESULTS A total of 822 proteins were identified, bound to the surfaces of the implants. Extracellular proteins were the most abundant ontology, followed by intracellular proteins. Fibrinogen, a proinflammatory protein and Albumin, an anti-inflammatory protein had significant (P < 0.0001) binding differences between the surfaces studied. Complement C3, C5, and factor H were also shown to have significantly different binding affinities for the implants included in the study (P < 0.05). CONCLUSIONS We have developed a novel assay of breast implant protein binding and demonstrated significant binding affinities for relevant proteins derived from breast tissue wound fluid. LEVEL OF EVIDENCE 5
Collapse
Affiliation(s)
| | - Ernie W Hill
- University of Manchester, Manchester, United Kingdom
| | | |
Collapse
|
42
|
Brilha S, Chong DLW, Khawaja AA, Ong CWM, Guppy NJ, Porter JC, Friedland JS. Integrin α2β1 Expression Regulates Matrix Metalloproteinase-1-Dependent Bronchial Epithelial Repair in Pulmonary Tuberculosis. Front Immunol 2018; 9:1348. [PMID: 29988449 PMCID: PMC6024194 DOI: 10.3389/fimmu.2018.01348] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/31/2018] [Indexed: 11/22/2022] Open
Abstract
Pulmonary tuberculosis (TB) is caused by inhalation of Mycobacterium tuberculosis, which damages the bronchial epithelial barrier to establish local infection. Matrix metalloproteinase-1 plays a crucial role in the immunopathology of TB, causing breakdown of type I collagen and cavitation, but this collagenase is also potentially involved in bronchial epithelial repair. We hypothesized that the extracellular matrix (ECM) modulates M. tuberculosis-driven matrix metalloproteinase-1 expression by human bronchial epithelial cells (HBECs), regulating respiratory epithelial cell migration and repair. Medium from monocytes stimulated with M. tuberculosis induced collagenase activity in bronchial epithelial cells, which was reduced by ~87% when cells were cultured on a type I collagen matrix. Matrix metalloproteinase-1 had a focal localization, which is consistent with cell migration, and overall secretion decreased by 32% on type I collagen. There were no associated changes in the specific tissue inhibitors of metalloproteinases. Decreased matrix metalloproteinase-1 secretion was due to ligand-binding to the α2β1 integrin and was dependent on the actin cytoskeleton. In lung biopsies, samples from patients with pulmonary TB, integrin α2β1 is highly expressed on the bronchial epithelium. Areas of lung with disrupted collagen matrix showed an increase in matrix metalloproteinases-1 expression compared with areas where collagen was comparable to control lung. Type I collagen matrix increased respiratory epithelial cell migration in a wound-healing assay, and this too was matrix metalloproteinase-dependent, since it was blocked by the matrix metalloproteinase inhibitor GM6001. In summary, we report a novel mechanism by which α2β1-mediated signals from the ECM modulate matrix metalloproteinase-1 secretion by HBECs, regulating their migration and epithelial repair in TB.
Collapse
Affiliation(s)
- Sara Brilha
- Infectious Diseases and Immunity, Imperial College London, London, United Kingdom.,Centre for Inflammation and Tissue Repair, University College London, London, United Kingdom
| | - Deborah L W Chong
- Centre for Inflammation and Tissue Repair, University College London, London, United Kingdom
| | - Akif A Khawaja
- Centre for Inflammation and Tissue Repair, University College London, London, United Kingdom
| | - Catherine W M Ong
- Infectious Diseases and Immunity, Imperial College London, London, United Kingdom.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Naomi J Guppy
- UCL Advanced Diagnostics, University College London, London, United Kingdom
| | - Joanna C Porter
- Centre for Inflammation and Tissue Repair, University College London, London, United Kingdom
| | - Jon S Friedland
- Infectious Diseases and Immunity, Imperial College London, London, United Kingdom
| |
Collapse
|
43
|
|
44
|
Cook JH, Ueno N, Lodoen MB. Toxoplasma gondii disrupts β1 integrin signaling and focal adhesion formation during monocyte hypermotility. J Biol Chem 2018; 293:3374-3385. [PMID: 29295815 PMCID: PMC5836128 DOI: 10.1074/jbc.m117.793281] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 12/14/2017] [Indexed: 01/13/2023] Open
Abstract
The motility of blood monocytes is orchestrated by the activity of cell-surface integrins, which translate extracellular signals into cytoskeletal changes to mediate adhesion and migration. Toxoplasma gondii is an intracellular parasite that infects migratory cells and enhances their motility, but the mechanisms underlying T. gondii-induced hypermotility are incompletely understood. We investigated the molecular basis for the hypermotility of primary human peripheral blood monocytes and THP-1 cells infected with T. gondii Compared with uninfected monocytes, T. gondii infection of monocytes reduced cell spreading and the number of activated β1 integrin clusters in contact with fibronectin during settling, an effect not observed in monocytes treated with lipopolysaccharide (LPS) or Escherichia coli Furthermore, T. gondii infection disrupted the phosphorylation of focal adhesion kinase (FAK) at tyrosine 397 (Tyr-397) and Tyr-925 and of the related protein proline-rich tyrosine kinase (Pyk2) at Tyr-402. The localization of paxillin, FAK, and vinculin to focal adhesions and the colocalization of these proteins with activated β1 integrins were also impaired in T. gondii-infected monocytes. Using time-lapse confocal microscopy of THP-1 cells expressing enhanced GFP (eGFP)-FAK during settling on fibronectin, we found that T. gondii-induced monocyte hypermotility was characterized by a reduced number of enhanced GFP-FAK-containing clusters over time compared with uninfected cells. This study demonstrates an integrin conformation-independent regulation of the β1 integrin adhesion pathway, providing further insight into the molecular mechanism of T. gondii-induced monocyte hypermotility.
Collapse
Affiliation(s)
- Joshua H Cook
- From the Department of Molecular Biology and Biochemistry and the Institute for Immunology, University of California, Irvine, California, 92697
| | - Norikiyo Ueno
- From the Department of Molecular Biology and Biochemistry and the Institute for Immunology, University of California, Irvine, California, 92697
| | - Melissa B Lodoen
- From the Department of Molecular Biology and Biochemistry and the Institute for Immunology, University of California, Irvine, California, 92697
| |
Collapse
|
45
|
Barton M, Filardo EJ, Lolait SJ, Thomas P, Maggiolini M, Prossnitz ER. Twenty years of the G protein-coupled estrogen receptor GPER: Historical and personal perspectives. J Steroid Biochem Mol Biol 2018; 176:4-15. [PMID: 28347854 PMCID: PMC5716468 DOI: 10.1016/j.jsbmb.2017.03.021] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/24/2022]
Abstract
Estrogens play a critical role in many aspects of physiology, particularly female reproductive function, but also in pathophysiology, and are associated with protection from numerous diseases in premenopausal women. Steroids and the effects of estrogen have been known for ∼90 years, with the first evidence for a receptor for estrogen presented ∼50 years ago. The original ancestral steroid receptor, extending back into evolution more than 500 million years, was likely an estrogen receptor, whereas G protein-coupled receptors (GPCRs) trace their origins back into history more than one billion years. The classical estrogen receptors (ERα and ERβ) are ligand-activated transcription factors that confer estrogen sensitivity upon many genes. It was soon apparent that these, or novel receptors may also be responsible for the "rapid"/"non-genomic" membrane-associated effects of estrogen. The identification of an orphan GPCR (GPR30, published in 1996) opened a new field of research with the description in 2000 that GPR30 expression is required for rapid estrogen signaling. In 2005-2006, the field was greatly stimulated by two studies that described the binding of estrogen to GPR30-expressing cell membranes, followed by the identification of a GPR30-selective agonist (that lacked binding and activity towards ERα and ERβ). Renamed GPER (G protein-coupled estrogen receptor) by IUPHAR in 2007, the total number of articles in PubMed related to this receptor recently surpassed 1000. In this article, the authors present personal perspectives on how they became involved in the discovery and/or advancement of GPER research. These areas include non-genomic effects on vascular tone, receptor cloning, molecular and cellular biology, signal transduction mechanisms and pharmacology of GPER, highlighting the roles of GPER and GPER-selective compounds in diseases such as obesity, diabetes, and cancer and the obligatory role of GPER in propagating cardiovascular aging, arterial hypertension and heart failure through the stimulation of Nox expression.
Collapse
Affiliation(s)
- Matthias Barton
- Molecular Internal Medicine, University of Zürich, 8057 Zürich, Switzerland.
| | - Edward J Filardo
- Rhode Island Hospital, Brown University, Providence, RI 02903, USA
| | - Stephen J Lolait
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, School of Clinical Sciences, University of Bristol, Bristol, UK
| | - Peter Thomas
- Marine Science Institute, University of Texas at Austin, Port Aransas, TX 78373, USA
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico Health Sciences Center and University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
46
|
Hu S, Chen TH, Zhao Y, Wang Z, Lam RHW. Protein-Substrate Adhesion in Microcontact Printing Regulates Cell Behavior. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:1750-1759. [PMID: 29304548 DOI: 10.1021/acs.langmuir.7b02935] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Microcontact printing (μCP) is widely used to create patterns of biomolecules essential for studies of cell mechanics, migration, and tissue engineering. However, different types of μCPs may create micropatterns with varied protein-substrate adhesion, which may change cell behaviors and pose uncertainty in result interpretation. Here, we characterize two μCP methods for coating extracellular matrix (ECM) proteins (stamp-off and covalent bond) and demonstrate for the first time the important role of protein-substrate adhesion in determining cell behavior. We found that, as compared to cells with weaker traction force (e.g., endothelial cells), cells with strong traction force (e.g., vascular smooth muscle cells) may delaminate the ECM patterns, which reduced cell viability as a result. Importantly, such ECM delamination was observed on patterns by stamp-off but not on the patterns by covalent bonds. Further comparisons of the displacement of the ECM patterns between the normal VSMCs and the force-reduced VSMCs suggested that the cell traction force plays an essential role in this ECM delamination. Together, our results indicated that μCPs with insufficient adhesion may lead to ECM delamination and cause cell death, providing new insight for micropatterning in cell-biomaterial interaction on biointerfaces.
Collapse
Affiliation(s)
- Shuhuan Hu
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong , Hong Kong
| | - Ting-Hsuan Chen
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong , Hong Kong
- City University of Hong Kong, Shenzhen Research Institute , Shenzhen, China
| | - Yanhua Zhao
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong , Hong Kong
| | - Zuankai Wang
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong , Hong Kong
- City University of Hong Kong, Shenzhen Research Institute , Shenzhen, China
| | - Raymond H W Lam
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong , Hong Kong
- City University of Hong Kong, Shenzhen Research Institute , Shenzhen, China
| |
Collapse
|
47
|
Jia T, Choi J, Ciccione J, Henry M, Mehdi A, Martinez J, Eymin B, Subra G, Coll JL. Heteromultivalent targeting of integrin α vβ 3 and neuropilin 1 promotes cell survival via the activation of the IGF-1/insulin receptors. Biomaterials 2017; 155:64-79. [PMID: 29169039 DOI: 10.1016/j.biomaterials.2017.10.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/19/2017] [Accepted: 10/25/2017] [Indexed: 12/29/2022]
Abstract
Angiogenesis strongly depends on the activation of integrins, especially integrin αvβ3, and of neuropilin-1 (NRP-1), a co-receptor of VEGFR2. Dual-targeted molecules that simultaneously block both of them are expected have increased anti-angiogenic and antitumor activity. Toward this goal, we generated bifunctional 40 nm-sized silica nanoparticles (NPs) coated with controlled amounts of cRGD and ATWLPPR peptides and studied their affinity, selectivity and biological activity in HUVECs. Sub-nanomolar concentrations of NPs grafted either with ATWLPPR alone or in combination with cRGD exhibit potent and specific antagonist activity against VEGFR2/AKT signaling. However, a 1 nM concentration of the cRGD/ATWLPPR-heteromultivalent particles (RGD/ATW-NPs) also blocks the phosphorylation of VEGFR2 while co-inducing an unexpected long-lasting activation of AKT via IGF-1R/IR-AKT/GSK3β/eNOS signaling that stimulates cell survival and abrogates the intrinsic toxicity of silica-NPs to serum-starved HUVECs. We also showed that their repeated intravenous administration was associated with the proliferation of human U87MG tumor cells engrafted in nude mice and a dilatation of the tumor blood vessels. We present biochemical evidence for the complex cross-talk generated by the binding of the heteromultivalent NPs with αvβ3-integrin and with NRP1. In particular, we show for the first time that such heteromultivalent NPs can trans-activate IGF-1/insulin receptors and exert dose-dependent pro-survival activity. This study demonstrates the difficulties in designing targeted silica-based NPs for antiangiogenic therapies and the possible risks posed by undesirable side effects.
Collapse
Affiliation(s)
- Tao Jia
- INSERM-UGA U1209, CNRS UMR5309, Institute for Advanced Biosciences, La Tronche, France
| | - Jungyoon Choi
- INSERM-UGA U1209, CNRS UMR5309, Institute for Advanced Biosciences, La Tronche, France
| | - Jéremy Ciccione
- Institut des Biomolécules Max Mousseron (IBMM), UMR5247 CNRS, Université de Montpellier, ENSCM, Montpellier, France
| | - Maxime Henry
- INSERM-UGA U1209, CNRS UMR5309, Institute for Advanced Biosciences, La Tronche, France
| | - Ahmad Mehdi
- Institut Charles Gerhardt, UMR5253, CNRS, Université de Montpellier, ENSCM, Montpellier Cedex 05, France
| | - Jean Martinez
- Institut des Biomolécules Max Mousseron (IBMM), UMR5247 CNRS, Université de Montpellier, ENSCM, Montpellier, France
| | - Béatrice Eymin
- INSERM-UGA U1209, CNRS UMR5309, Institute for Advanced Biosciences, La Tronche, France
| | - Gilles Subra
- Institut des Biomolécules Max Mousseron (IBMM), UMR5247 CNRS, Université de Montpellier, ENSCM, Montpellier, France
| | - Jean-Luc Coll
- INSERM-UGA U1209, CNRS UMR5309, Institute for Advanced Biosciences, La Tronche, France.
| |
Collapse
|
48
|
Idoux-Gillet Y, Nassour M, Lakis E, Bonini F, Theillet C, Du Manoir S, Savagner P. Slug/Pcad pathway controls epithelial cell dynamics in mammary gland and breast carcinoma. Oncogene 2017; 37:578-588. [PMID: 28991231 DOI: 10.1038/onc.2017.355] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 08/16/2017] [Accepted: 08/23/2017] [Indexed: 12/28/2022]
Abstract
Mammary gland morphogenesis results from the coordination of proliferation, cohort migration, apoptosis and stem/progenitor cell dynamics. We showed earlier that the transcription repressor Slug is involved in these functions during mammary tubulogenesis. Slug is expressed by a subpopulation of basal epithelial cells, co-expressed with P-cadherin (Pcad). Slug-knockout mammary glands showed excessive branching, similarly to Pcad-knockout. Here, we found that Slug unexpectedly binds and activates Pcad promoter through E-boxes, inducing Pcad expression. We determined that Pcad can mediate several functions of Slug: Pcad promoted clonal mammosphere growth, basal epithelial differentiation, cell-cell dissociation and cell migration, rescuing Slug depletion. Pcad also promoted cell migration in isolated cells, in association with Src activation, focal adhesion reorganization and cell polarization. Pcad, similarly to Slug, was required for in vitro 3D tubulogenesis. Therefore, Pcad appears to be responsible for epithelial-mesenchymal transition-linked plasticity in mammary epithelial cells. In addition, we found that genes from the Slug/Pcad pathway components were co-expressed and specifically correlated in human breast carcinomas subtypes, carrying pathophysiological significance.
Collapse
Affiliation(s)
- Y Idoux-Gillet
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - M Nassour
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - E Lakis
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - F Bonini
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - C Theillet
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - S Du Manoir
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| | - P Savagner
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Inserm U1194, Université de Montpellier, Institut Régional du Cancer de Montpellier (ICM), Montpellier, France
| |
Collapse
|
49
|
Biegel HR, Quackenbush A, Highlander HC. Sensitivity analysis for stochastic and deterministic models of nascent focal adhesion dynamics. INT J BIOMATH 2017. [DOI: 10.1142/s1793524517501054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Sensitivity analysis (SA) is a critical part of modeling any biological system due to the inherent uncertainty in model output, as introduced by parameter values that have not been experimentally determined. SA therefore provides deeper understanding of the system by painting a picture of the extent to which certain model outputs vary in relationship to changes in model parameters. Here we explore two types of global SA for recently developed models of nascent focal adhesion formation, a key step in cellular movement. While many SA methods have been used for deterministic methods, we utilize methods for both stochastic and deterministic models, providing a more complete description of the parameters to which the focal adhesion model is most sensitive. Specific recommendations for further experimentation in the process of cellular motility are proposed in response to the SA.
Collapse
Affiliation(s)
- Hannah R. Biegel
- Departments of Mathematics and Biology, University of Portland, 5000 North Willamette Boulevard, Portland, Oregon 97203, USA
| | - Alex Quackenbush
- Departments of Mathematics and Biology, University of Portland, 5000 North Willamette Boulevard, Portland, Oregon 97203, USA
| | - Hannah Callender Highlander
- Department of Mathematics, University of Portland, 5000 North Willamette Boulevard, Portland, Oregon 97203, USA
| |
Collapse
|
50
|
Gao M, Wu S, Ji J, Zhang J, Liu Q, Yue Y, Liu L, Liu X, Liu W. The influence of actin depolymerization induced by Cytochalasin D and mechanical stretch on interleukin-8 expression and JNK phosphorylation levels in human retinal pigment epithelial cells. BMC Ophthalmol 2017; 17:43. [PMID: 28388885 PMCID: PMC5384187 DOI: 10.1186/s12886-017-0437-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 04/04/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND This study explores the role of actin cytoskeleton depolymerization induced by Cytochalasin D and mechanical stretch on the interleukin-8 (IL-8) expression and c-jun N-terminal kinase (JNK) phosphorylation levels in human retinal pigment epithelial (RPE) cells. METHODS A Flexcell FX-5000 Tension system was used to apply cyclic stretch to cultured human RPE cells (ARPE-19) at 0.33 Hz with 20% elongation for 0 h, 6 h or 24 h. The cells were stretched alone or pre-treated with Cytochalasin D. The redistribution of the actin cytoskeleton was evaluated using phalloidin immunofluorescence staining. The protein expression levels of IL-8 and JNK in the RPE cells were determined via Western blotting. RESULTS The cells in the control groups displayed abundant and uniform phalloidin staining. After exposure to mechanical stretch for 24 h, phalloidin staining revealed an unclear and irregular actin cytoskeleton. In all Cytochalasin D-treated cells, the shrinkage and disruption of the cytoskeletal structure was observed regardless of mechanical stress. The stimulation of the RPE cells with cyclic stretch alone did not induce a significant increase in IL-8 expression and JNK phosphorylation levels, which were similar to those of the control groups. After pre-treatment with Cytochalasin D alone, IL-8 expression and JNK phosphorylation levels were not significantly different at 6 h but were significantly increased by approximately 1.2-fold (1.18 ± 0.05; P<0.01) and 3.0-fold (3.01 ± 0.02; P<0.01) at 24 h, respectively. After the pre-incubation of the RPE cells with Cytochalasin D followed by exposure to cyclic stretch, IL-8 expression and JNK phosphorylation levels increased by approximately 1.3-fold (1.31 ± 0.02; P<0.01) and 1.3-fold (1.31 ± 0.02; P<0.01) at 6 h, respectively, and by 1.7-fold (1.69 ± 0.06; P<0.01) and 3.2-fold (3.21 ± 0.12; P<0.01) at 24 h, respectively. CONCLUSIONS This study demonstrates that disruption of actin polymerization by cytochalasin D and mechanical stretch upregulates interleukin-8 expression and JNK phosphorylation levels in human RPE cells, which indicates that the integrity of the actin cytoskeleton may play important roles in the pro-inflammatory processes in RPE cells.
Collapse
Affiliation(s)
- Meng Gao
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing, China
| | - Shen Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing, China
| | - Jing Ji
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - JingXue Zhang
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing, China
| | - Qian Liu
- Beijing Institute of Ophthalmology, Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing, China
| | - YanKun Yue
- Department of Ophthalmology, Beijing Fuxing Hospital, Capital Medical University, Beijing, China
| | - Lu Liu
- Department of Ophthalmology, Beijing Fuxing Hospital, Capital Medical University, Beijing, China
| | - XinXin Liu
- Department of Ophthalmology, Kailuan General Hospital, Tangshan, China
| | - Wu Liu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University; Beijing Ophthalmology and Visual Sciences Key Laboratory, Beijing, China.
| |
Collapse
|