1
|
Toyofuku T, Ishikawa T, Kumanogoh A. Deletion of the plexin-D1 ectodomain leads to anoikis by suppressing integrin inside-out signaling. Mol Biol Cell 2025; 36:ar71. [PMID: 40266804 DOI: 10.1091/mbc.e25-02-0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025] Open
Abstract
Plexin-D1, mainly expressed in endothelial and cancer cells, regulates diverse effects, suppresses endothelial cell growth, and induces cancer cell migration and proliferation. Here, we demonstrated that plexin-D1 was cleaved by proteinase on cancer cells. To examine the role of cleaved plexin-D1 in cells, Madin-Darby canine kidney (MDCK) cells overexpressing truncated plexin-D1 were cultured in Matrigel. MDCK cells expressing plexin-D1 lacking the ectodomain (plexin-D1 ΔEC) underwent apoptosis. An adhesion assay for extracellular matrix (ECM) molecules showed that plexin-D1 ΔEC-expressing MDCK cells lost their affinity for the ECM. These results suggest that plexin-D1 ΔEC blocks integrin inside-out signaling, leading to detachment from the ECM and apoptosis, so-called anoikis. By contrast, MDCK cells expressing full-length plexin-D1 or plexin-D1 lacking the cytoplasmic domain (plexin-D1 ΔIC) developed multicellular branching tubular structures in Matrigel. This morphological change was blocked in plexin-D1-expressing MDCK cells by the hepatocyte growth factor receptor (Met) loss of function or by Met inhibitors. These results suggest that plexin-D1 associates with Met through the plexin-D1 extracellular domain, and this activates Met cytoplasmic kinase activity. We therefore conclude that plexin-D1 contains distinct domains that determine the fate of cancer cells.
Collapse
Affiliation(s)
- Toshihiko Toyofuku
- Department of Immunology and Molecular Medicine, Graduate School of Medicine, The Center of Medical Innovation and Translational Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takako Ishikawa
- Department of Immunology and Molecular Medicine, Graduate School of Medicine, The Center of Medical Innovation and Translational Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
2
|
Yao Y. Laminin Receptors in the CNS and Vasculature. Stroke 2025. [PMID: 40421534 DOI: 10.1161/strokeaha.125.051560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025]
Abstract
Laminin exerts a variety of important functions via binding to its receptors, including integrins and dystroglycan. With the advance in gene-targeting technology, many integrin/dystroglycan knockout/mutant mice were generated in the past 3 decades. These mutants enable loss-of-function studies and have substantially enriched our knowledge of integrin/dystroglycan functions. In this review, we summarize the functions of laminin receptors during embryonic development and in the CNS and vasculature. First, the biochemical properties of integrins and dystroglycan are briefly introduced. Next, we discuss loss-of-function studies on laminin receptors, including integrin-α3, integrin-α6, integrin-α7, integrin-β1, integrin-β4, and dystroglycan, focusing on embryonic development, the CNS, and vasculature. The phenotypes of compound knockout mice are described and compared with that of single mutants. Last, important questions and challenges in the field as well as potential future directions are discussed. Our goal is to provide a synthetic review on loss-of-function studies of laminin receptors in the CNS and vasculature, which could serve as a reference for future research, encourage the formation of new hypotheses, and stimulate new research in this field.
Collapse
Affiliation(s)
- Yao Yao
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa
| |
Collapse
|
3
|
Gao T, Maskalenko NA, Kabir S, Campbell KS, Wu J. Molecular basis of β2 integrin activation by talin unveils subunit-specific mechanisms of integrin signaling. Cell Rep 2025; 44:115607. [PMID: 40310722 DOI: 10.1016/j.celrep.2025.115607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/07/2025] [Accepted: 04/02/2025] [Indexed: 05/03/2025] Open
Abstract
Integrins consist of 24 species, each with unique tissue expression profiles and distinct biological functions. The β subunit of integrin interacts with the FERM-folded head domain of talin through an NPxY/F motif, triggering integrin activation. Although this motif is conserved across most integrin-β subunits, the precise molecular mechanism governing talin's selective recognition of different integrin-β subunits remains unclear. We identify two distinct configurations of the talin head when interacting with β2 and β3 integrins, providing critical insights into subunit-specific recognition of integrins. Structural studies reveal that mutations at the subdomain interface of the talin head can shift its β2-bound configuration to a β3-bound configuration. This shift enhances β2-integrin affinity, leading to increased lymphocyte function-associated antigen-1 (LFA-1)-mediated natural killer cell activity. Together, our data elucidate the structural basis of talin's role in mediating integrin activation in a subunit-specific manner and advance our understanding of how talin may regulate diverse functions of various integrin species.
Collapse
Affiliation(s)
- Tong Gao
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Nicholas A Maskalenko
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Salvin Kabir
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA; Department of Biology, College of Science & Technology, Temple University, Philadelphia, PA 19122, USA
| | - Kerry S Campbell
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Jinhua Wu
- Cancer Signaling and Microenvironment Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA.
| |
Collapse
|
4
|
Han H, Feng P, Yuan G. Molecular typing of gliomas on the basis of integrin family genes and a functional study of ITGA7. Sci Rep 2025; 15:12306. [PMID: 40210748 PMCID: PMC11985493 DOI: 10.1038/s41598-025-97342-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025] Open
Abstract
Gliomas are highly malignant tumors of the central nervous system, and their complex molecular heterogeneity poses major therapeutic challenges. Integrins are important members of the class of cell adhesion molecules (CAMs), consisting of α-subunits and β-subunits that form 24 different heterodimers. To elucidate the complex role of integrins in glioma pathogenesis, we analyzed integrin family genes. We used a scoring system based on gene set enrichment analysis (GSEA) to identify prognostic biomarkers and nonnegative matrix factorization (NMF) to establish a new integrin-based molecular classification of gliomas. Subsequent analyses of the clinical relevance of the molecular subtypes and the underlying mechanisms demonstrated a strong correlation between integrin-based molecular subtypes and glioma malignancy. We further characterized the different clinical features and tumor microenvironments (TMEs) associated with these subtypes. We identified subtype-specific driver genes using the limma R package and weighted gene coexpression network analysis (WGCNA). We subsequently identified key integrin-mediated genes that significantly contribute to poor prognosis through a combined approach of machine learning (ML) and protein‒protein interaction (PPI) network analysis. Finally, we performed in vitro cellular experiments on the integrin family gene ITGA7 and demonstrated that ITGA7 can serve as a biomarker for gliomas. Our findings provide important insights into the multifaceted roles of integrins in glioma biology, provide an opportunity for the discovery of novel targeted therapies on the basis of the subtype-specific vulnerability of integrins, and provide a basis for the study of the role of ITGA7 in gliomas.
Collapse
Affiliation(s)
- Hongxi Han
- Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, People's Republic of China
- Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Peng Feng
- Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, People's Republic of China
- Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China
| | - Guoqiang Yuan
- Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, People's Republic of China.
- Lanzhou University, Lanzhou, 730000, Gansu, People's Republic of China.
- Department of Neurosurgery and Laboratory of Neurosurgery, Lanzhou University Second Hospital, No. 82, Cuiyingmen, Chengguan District, Lanzhou City, 730000, Gansu Province, People's Republic of China.
| |
Collapse
|
5
|
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
|
6
|
Elbakry M, Khatami N, Akoume MY, Julien C, Bouhanik S, Franco A, Caraus I, Elremaly W, Moreau A. Loss of Tyrosine Phosphatase Mu Promotes Scoliosis Progression Through Osteopontin-α5β1 Integrin Signaling and PIPK1γ90 Activity. Int J Mol Sci 2025; 26:1042. [PMID: 39940812 PMCID: PMC11816665 DOI: 10.3390/ijms26031042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/28/2024] [Accepted: 12/28/2024] [Indexed: 02/16/2025] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is characterized by a curvature of the spine affecting approximately 4% of the pediatric population, and the mechanisms driving its progression remain poorly understood. Whole-exome sequencing of a French-Canadian AIS cohort with severe scoliosis identified rare variants in the PTPRM gene, which encodes Protein Tyrosine Phosphatase μ (PTPµ). However, these rare variants alone did not account for the pronounced reduction in PTPµ at both mRNA and protein levels in severe AIS cases. This led us to investigate epigenetic regulators and the identification of five microRNAs (miR-103a-3p, miR-107, miR-148a-3p, miR-148b-3p, and miR-152-3p) that target PTPRM mRNA. These microRNAs were significantly elevated in plasma from severe AIS patients, and miR-148b-3p was also upregulated in AIS osteoblasts. Phenotypic analysis of bipedal Ptrprm knockout (PTPµ -/-) mice showed increased prevalence and severity of scoliosis, while quadrupedal PTPµ -/- mice did not develop scoliosis, underscoring PTPµ's role as a disease-modifying factor. Mechanistically, PTPµ deficiency was found to disrupt Gi-coupled receptor signaling in osteoblasts by enhancing the interaction between osteopontin (OPN) and α5β1 integrin, along with increased tyrosine phosphorylation of phosphatidylinositol-4-phosphate 5-kinase type I (PIPKIγ90). These findings provide novel insights into the molecular mechanisms underlying spinal deformity progression in AIS, linking PTPµ depletion to aberrant OPN-α5β1 integrin signaling and highlighting potential therapeutic targets to stop, mitigate, or prevent scoliosis.
Collapse
Affiliation(s)
- Mohamed Elbakry
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Azrieli Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
- Department of Chemistry, Biochemistry Section, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Nasrin Khatami
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Azrieli Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Marie-Yvonne Akoume
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Azrieli Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
- Department of Cellular, Molecular Biology and Genetics, Faculty of Medicine, Université des Sciences de la Santé (USS) de Libreville, Libreville BP 18231, Gabon
| | - Cédric Julien
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Azrieli Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Saadallah Bouhanik
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Azrieli Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Anita Franco
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Azrieli Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Iurie Caraus
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Azrieli Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Wesam Elremaly
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Azrieli Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
| | - Alain Moreau
- Viscogliosi Laboratory in Molecular Genetics of Musculoskeletal Diseases, Azrieli Research Center, CHU Sainte-Justine, Montréal, QC H3T 1C5, Canada
- Department of Cellular, Molecular Biology and Genetics, Faculty of Medicine, Université des Sciences de la Santé (USS) de Libreville, Libreville BP 18231, Gabon
- Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montréal, QC H3T 1J4, Canada
| |
Collapse
|
7
|
Adhikari B, Barakoti P, Pantcheva MB, Krebs MD. 3D printed gelatin methacryloyl hydrogels for perfusion culture of human trabecular meshwork cells and glaucoma studies. Biotechnol Bioeng 2025; 122:69-79. [PMID: 39291858 DOI: 10.1002/bit.28849] [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: 12/06/2023] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
Glaucoma, a progressive eye disease leading to irreversible blindness, currently affects over 70 million people globally. Elevated intraocular pressure (IOP) is implicated in its development. IOP is carefully regulated by the trabecular meshwork (TM). However, studying TM behavior has been limited to traditional tissue culture studies or costly ex vivo cultures of animal and donor eyes. Developing novel functional TM models could enhance cell/tissue behavior understanding and aid therapeutic development for glaucoma. In this study, we 3D printed a simplified and reproducible model of the human TM (hTM) and studied hTM cell behavior under static and dynamic cultures. Gelatin Methacryloyl bioinks proved suitable for printing with viable and proliferative hTM cells expressing crucial marker genes in response to glucocorticoid induction. This, to our knowledge, is the first functional 3D printed hTM model and aims to facilitate TM research. Moreover, this easily reproducible model could also be applicable in the study of numerous other cell types throughout the body.
Collapse
Affiliation(s)
- Bikram Adhikari
- Colorado School of Mines, Quantitative Biosciences and Engineering, Golden, Colorado, USA
| | - Prasanga Barakoti
- Colorado School of Mines, Quantitative Biosciences and Engineering, Golden, Colorado, USA
| | - Mina B Pantcheva
- Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Melissa D Krebs
- Colorado School of Mines, Quantitative Biosciences and Engineering, Golden, Colorado, USA
- Colorado School of Mines, Chemical and Biological Engineering, Golden, Colorado, USA
| |
Collapse
|
8
|
Li S, Chen A, Gui J, Zhou H, Zhu L, Mi Y. TLN1: an oncogene associated with tumorigenesis and progression. Discov Oncol 2024; 15:716. [PMID: 39589610 PMCID: PMC11599537 DOI: 10.1007/s12672-024-01593-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
Talin-1 (TLN1), encoded by the TLN1 gene, is a focal adhesion-related protein capable of binding various proteins in the cytoskeleton. It is also expressed at high levels in many cancers wherein it influences cellular adhesion and the activation of integrins. TLN1 is also capable of promoting tumor cell invasivity, proliferation, and metastatic progression, in addition to being a relevant biomarker and therapeutic target in certain cancers. The present review offers a comprehensive overview of current knowledge regarding TLN1 with respect to its structural properties, functions, and role in tumor development.
Collapse
Affiliation(s)
- Sixin Li
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Anjie Chen
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Jiandong Gui
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Hangsheng Zhou
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China
- Wuxi School of Medicine, Jiangnan University, 1800 Lihudadao, Wuxi, 214122, Jiangsu, China
| | - Lijie Zhu
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China.
| | - Yuanyuan Mi
- Department of Urology, Affiliated Hospital of Jiangnan University, 1000 Hefeng Road, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
9
|
Bogdanović B, Fagret D, Ghezzi C, Montemagno C. Integrin Targeting and Beyond: Enhancing Cancer Treatment with Dual-Targeting RGD (Arginine-Glycine-Aspartate) Strategies. Pharmaceuticals (Basel) 2024; 17:1556. [PMID: 39598465 PMCID: PMC11597078 DOI: 10.3390/ph17111556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Integrins, an important superfamily of cell adhesion receptors, play an essential role in cancer progression, metastasis, and angiogenesis, establishing them as prime targets for both diagnostic and therapeutic applications. Despite their significant potential, integrin-targeted therapies have faced substantial challenges in clinical trials, including variable efficacy and unmet high expectations. Nevertheless, the consistent expression of integrins on tumor and stromal cells underscores their ongoing relevance and potential. Traditional RGD-based imaging and therapeutic agents have faced limitations, such as inconsistent target expression and rapid systemic clearance, which have reduced their effectiveness. To overcome these challenges, recent research has focused on advancing RGD-based strategies and exploring innovative solutions. This review offers a thorough analysis of the latest developments in the RGD-integrin field, with a particular focus on addressing previous limitations. It delves into new dual-targeting approaches and cutting-edge RGD-based agents designed to improve both tumor diagnosis and therapeutic outcomes. By examining these advancements, this review illuminates new pathways for enhancing the specificity and efficacy of integrin-targeted therapies, paving the way for more effective cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Bojana Bogdanović
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | - Daniel Fagret
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | - Catherine Ghezzi
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | | |
Collapse
|
10
|
Lopuhaä BV, Guzel C, van der Lee A, van den Bosch TPP, van Kemenade FJ, Huisman MV, Kruip MJHA, Luider TM, von der Thüsen JH. Increase in venous thromboembolism in SARS-CoV-2 infected lung tissue: proteome analysis of lung parenchyma, isolated endothelium, and thrombi. Histopathology 2024; 84:967-982. [PMID: 38253958 DOI: 10.1111/his.15143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024]
Abstract
AIMS COVID-19 pneumonia is characterized by an increased rate of deep venous thrombosis and pulmonary embolism. To better understand the pathophysiology behind thrombosis in COVID-19, we performed proteomics analysis on SARS-CoV-2 infected lung tissue. METHODS Liquid chromatography mass spectrometry was performed on SARS-CoV-2 infected postmortem lung tissue samples. Five protein profiling analyses were performed: whole slide lung parenchyma analysis, followed by analysis of isolated thrombi and endothelium, both stratified by disease (COVID-19 versus influenza) and thrombus morphology (embolism versus in situ). Influenza autopsy cases with pulmonary thrombi were used as controls. RESULTS Compared to influenza controls, both analyses of COVID-19 whole-tissue and isolated endothelium showed upregulation of proteins and pathways related to liver metabolism including urea cycle activation, with arginase being among the top upregulated proteins in COVID-19 lung tissue. Analysis of isolated COVID-19 thrombi showed significant downregulation of pathways related to platelet activation compared to influenza thrombi. Analysis of isolated thrombi based on histomorphology shows that in situ thrombi have significant upregulation of coronavirus pathogenesis proteins. CONCLUSIONS The decrease in platelet activation pathways in severe COVID-19 thrombi suggests a relative increase in venous thromboembolism, as thrombi from venous origin tend to contain fewer platelets than arterial thrombi. Based on histomorphology, in situ thrombi show upregulation of various proteins related to SARS-CoV-2 pathogenesis compared to thromboemboli, which may indicate increased in situ pulmonary thrombosis in COVID-19. Therefore, this study supports the increase of venous thromboembolism without undercutting the involvement of in situ thrombosis in severe COVID-19.
Collapse
Affiliation(s)
- Boaz V Lopuhaä
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Coşkun Guzel
- Laboratory of Neuro-Oncology, Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | | | | | | | - Menno V Huisman
- Department of Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, the Netherlands
| | - Marieke J H A Kruip
- Department of Haematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Theo M Luider
- Laboratory of Neuro-Oncology, Clinical and Cancer Proteomics, Department of Neurology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Jan H von der Thüsen
- Department of Pathology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| |
Collapse
|
11
|
Lazzarino M, Zanetti M, Chen SN, Gao S, Peña B, Lam CK, Wu JC, Taylor MRG, Mestroni L, Sbaizero O. Defective Biomechanics and Pharmacological Rescue of Human Cardiomyocytes with Filamin C Truncations. Int J Mol Sci 2024; 25:2942. [PMID: 38474188 PMCID: PMC10932268 DOI: 10.3390/ijms25052942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Actin-binding filamin C (FLNC) is expressed in cardiomyocytes, where it localizes to Z-discs, sarcolemma, and intercalated discs. Although FLNC truncation variants (FLNCtv) are an established cause of arrhythmias and heart failure, changes in biomechanical properties of cardiomyocytes are mostly unknown. Thus, we investigated the mechanical properties of human-induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) carrying FLNCtv. CRISPR/Cas9 genome-edited homozygous FLNCKO-/- hiPSC-CMs and heterozygous knock-out FLNCKO+/- hiPSC-CMs were analyzed and compared to wild-type FLNC (FLNCWT) hiPSC-CMs. Atomic force microscopy (AFM) was used to perform micro-indentation to evaluate passive and dynamic mechanical properties. A qualitative analysis of the beating traces showed gene dosage-dependent-manner "irregular" peak profiles in FLNCKO+/- and FLNCKO-/- hiPSC-CMs. Two Young's moduli were calculated: E1, reflecting the compression of the plasma membrane and actin cortex, and E2, including the whole cell with a cytoskeleton and nucleus. Both E1 and E2 showed decreased stiffness in mutant FLNCKO+/- and FLNCKO-/- iPSC-CMs compared to that in FLNCWT. The cell adhesion force and work of adhesion were assessed using the retraction curve of the SCFS. Mutant FLNC iPSC-CMs showed gene dosage-dependent decreases in the work of adhesion and adhesion forces from the heterozygous FLNCKO+/- to the FLNCKO-/- model compared to FLNCWT, suggesting damaged cytoskeleton and membrane structures. Finally, we investigated the effect of crenolanib on the mechanical properties of hiPSC-CMs. Crenolanib is an inhibitor of the Platelet-Derived Growth Factor Receptor α (PDGFRA) pathway which is upregulated in FLNCtv hiPSC-CMs. Crenolanib was able to partially rescue the stiffness of FLNCKO-/- hiPSC-CMs compared to control, supporting its potential therapeutic role.
Collapse
Affiliation(s)
- Marco Lazzarino
- CNR-IOM, Area Science Park, 34149 Trieste, Italy; (M.L.); (M.Z.)
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Michele Zanetti
- CNR-IOM, Area Science Park, 34149 Trieste, Italy; (M.L.); (M.Z.)
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Suet Nee Chen
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Shanshan Gao
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Brisa Peña
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
- Bioengineering Department, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Chi Keung Lam
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA; (C.K.L.); (J.C.W.)
| | - Joseph C. Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA; (C.K.L.); (J.C.W.)
| | - Matthew R. G. Taylor
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Luisa Mestroni
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
| | - Orfeo Sbaizero
- Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (S.N.C.); (S.G.); (B.P.); (M.R.G.T.); (L.M.)
- Engineering and Architecture Department, University of Trieste, 34127 Trieste, Italy
| |
Collapse
|
12
|
Kwon HW, Shin JH, Rhee MH, Park CE, Lee DH. Anti-thrombotic effects of ginsenoside Rk3 by regulating cAMP and PI3K/MAPK pathway on human platelets. J Ginseng Res 2023; 47:706-713. [PMID: 38107398 PMCID: PMC10721468 DOI: 10.1016/j.jgr.2023.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 12/19/2023] Open
Abstract
Background and objective The ability to inhibit aggregation has been demonstrated with synthetically derived ginsenoside compounds G-Rp (1, 3, and 4) and ginsenosides naturally found in Panax ginseng 20(S)-Rg3, Rg6, F4, and Ro. Among these compounds, Rk3 (G-Rk3) from Panax ginseng needs to be further explored in order to reveal the mechanisms of action during inhibition. Methodology Our study focused to investigate the action of G-Rk3 on agonist-stimulated human platelet aggregation, inhibition of platelet signaling molecules such as fibrinogen binding with integrin αIIbβ3 using flow cytometry, intracellular calcium mobilization, dense granule secretion, and thromboxane B2 secretion. In addition, we checked the regulation of phosphorylation on PI3K/MAPK pathway, and thrombin-induced clot retraction was also observed in platelets rich plasma. Key Results G-Rk3 significantly increased amounts of cyclic adenosine monophosphate (cAMP) and led to significant phosphorylation of cAMP-dependent kinase substrates vasodilator-stimulated phosphoprotein (VASP) and inositol 1,4,5-trisphosphate receptor (IP3R). In the presence of G-Rk3, dense tubular system Ca2+ was inhibited, and platelet activity was lowered by inactivating the integrin αIIb/β3 and reducing the binding of fibrinogen. Furthermore, the effect of G-Rk3 extended to the inhibition of MAPK and PI3K/Akt phosphorylation resulting in the reduced secretion of intracellular granules and reduced production of TXA2. Lastly, G-Rk3 inhibited platelet aggregation and thrombus formation via fibrin clot. Conclusions and implications These results suggest that when dealing with cardiovascular diseases brought upon by faulty aggregation among platelets or through the formation of a thrombus, the G-Rk3 compound can play a role as an effective prophylactic or therapeutic agent.
Collapse
Affiliation(s)
- Hyuk-Woo Kwon
- Department of Biomedical Laboratory Science, Far East University, Eumseong, Republic of Korea
| | - Jung-Hae Shin
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Man Hee Rhee
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Chang-Eun Park
- Department of Biomedical Laboratory Science, Namseoul University, Cheonan, Republic of Korea
- Molecular Diagnostics Research Institute, Namseoul University, Cheonan, Republic of Korea
| | - Dong-Ha Lee
- Department of Biomedical Laboratory Science, Namseoul University, Cheonan, Republic of Korea
- Molecular Diagnostics Research Institute, Namseoul University, Cheonan, Republic of Korea
| |
Collapse
|
13
|
Honasoge KS, Karagöz Z, Goult BT, Wolfenson H, LaPointe VLS, Carlier A. Force-dependent focal adhesion assembly and disassembly: A computational study. PLoS Comput Biol 2023; 19:e1011500. [PMID: 37801464 PMCID: PMC10584152 DOI: 10.1371/journal.pcbi.1011500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 10/18/2023] [Accepted: 09/07/2023] [Indexed: 10/08/2023] Open
Abstract
Cells interact with the extracellular matrix (ECM) via cell-ECM adhesions. These physical interactions are transduced into biochemical signals inside the cell which influence cell behaviour. Although cell-ECM interactions have been studied extensively, it is not completely understood how immature (nascent) adhesions develop into mature (focal) adhesions and how mechanical forces influence this process. Given the small size, dynamic nature and short lifetimes of nascent adhesions, studying them using conventional microscopic and experimental techniques is challenging. Computational modelling provides a valuable resource for simulating and exploring various "what if?" scenarios in silico and identifying key molecular components and mechanisms for further investigation. Here, we present a simplified mechano-chemical model based on ordinary differential equations with three major proteins involved in adhesions: integrins, talin and vinculin. Additionally, we incorporate a hypothetical signal molecule that influences adhesion (dis)assembly rates. We find that assembly and disassembly rates need to vary dynamically to limit maturation of nascent adhesions. The model predicts biphasic variation of actin retrograde velocity and maturation fraction with substrate stiffness, with maturation fractions between 18-35%, optimal stiffness of ∼1 pN/nm, and a mechanosensitive range of 1-100 pN/nm, all corresponding to key experimental findings. Sensitivity analyses show robustness of outcomes to small changes in parameter values, allowing model tuning to reflect specific cell types and signaling cascades. The model proposes that signal-dependent disassembly rate variations play an underappreciated role in maturation fraction regulation, which should be investigated further. We also provide predictions on the changes in traction force generation under increased/decreased vinculin concentrations, complementing previous vinculin overexpression/knockout experiments in different cell types. In summary, this work proposes a model framework to robustly simulate the mechanochemical processes underlying adhesion maturation and maintenance, thereby enhancing our fundamental knowledge of cell-ECM interactions.
Collapse
Affiliation(s)
- Kailas Shankar Honasoge
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Zeynep Karagöz
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Benjamin T. Goult
- School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
| | - Vanessa L. S. LaPointe
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| | - Aurélie Carlier
- Department of Cell Biology–Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
14
|
Chen Y, Chen Y, Jian B, Feng Q, Liu L. Identification and Expression of Integrins during Testicular Fusion in Spodoptera litura. Genes (Basel) 2023; 14:1452. [PMID: 37510356 PMCID: PMC10379305 DOI: 10.3390/genes14071452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Integrin members are cell adhesion receptors that bind to extracellular matrix (ECM) proteins to regulate cell-cell adhesion and cell-ECM adhesion. This process is essential for tissue development and organogenesis. The fusion of two testes is a physiological phenomenon that is required for sperm production and effective reproduction in many Lepidoptera. However, the molecular mechanism of testicular fusion is unclear. In Spodoptera litura, two separated testes fuse into a single testis during the larva-to-pupa transformation. We identified five α and five β integrin subunits that were closely associated with testicular fusion. Integrin α1 and α2 belong to the position-specific 1 (PS1) and PS2 groups, respectively. Integrin α3, αPS1/αPS2, and αPS3 were clustered into the PS3 group. Integrin β1 belonged to the insect β group, and β2, β3, and β5 were clustered in the βν group. Among these integrins, α1, α2, α3, αPS1/PS2, αPS3, β1, and β4 subunits were highly expressed when the testes fused. However, their expression levels were much lower before and after the fusion of the testis. The qRT-PCR and immunohistochemistry analyses indicated that integrin β1 mRNA and the protein were highly expressed in the peritoneal sheath of the testis, particularly when the testes fused. These results indicate that integrins might participate in S. litura testicular fusion.
Collapse
Affiliation(s)
- Yaqing Chen
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Yu Chen
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Baozhu Jian
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Qili Feng
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| | - Lin Liu
- Guangdong Provincial Key Laboratory of Insect Developmental Biology and Applied Technology, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China
| |
Collapse
|
15
|
Hong L, Li W, Li Y, Yin S. Nanoparticle-based drug delivery systems targeting cancer cell surfaces. RSC Adv 2023; 13:21365-21382. [PMID: 37465582 PMCID: PMC10350659 DOI: 10.1039/d3ra02969g] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/11/2023] [Indexed: 07/20/2023] Open
Abstract
Traditional cancer chemotherapy easily produces serious toxic and side effects due to the lack of specific selection of tumor cells, which restricts its curative effect. Targeted delivery can increase the concentration of drugs in the target site and reduce their toxic and side effects on normal tissues and cells. Biocompatible and surface-modifiable nanocarriers are novel drug delivery systems, which are used to specifically target tumor sites in a controllable way. One of the effective ways to design effective targeting nanocarriers is to decorate with functional ligands, which can bind to specific receptors overexpressed on the surfaces of cancer cells. Various functional ligands, including transferrin, folic acid, polypeptide and hyaluronic acid, have been widely explored to develop tumor-selective drug delivery systems. This review focuses on the research progress of various receptors overexpressed on the surfaces of cancer cells and different nano-delivery systems of anticancer drugs targeted on the surfaces of cancer cells. We believe that through continuous research and development, actively targeted cancer nano-drugs will make a breakthrough and become an indispensable platform for accurate cancer treatment.
Collapse
Affiliation(s)
- Liquan Hong
- Deqing Hospital of Hangzhou Normal University, The Third People's Hospital of Deqing Deqing 313200 China
| | - Wen Li
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology Zhejiang Province Hangzhou 311121 China
| | - Yang Li
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology Zhejiang Province Hangzhou 311121 China
| | - Shouchun Yin
- Deqing Hospital of Hangzhou Normal University, The Third People's Hospital of Deqing Deqing 313200 China
- College of Material, Chemistry and Chemical Engineering, Hangzhou Normal University, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Key Laboratory of Organosilicon Material Technology Zhejiang Province Hangzhou 311121 China
| |
Collapse
|
16
|
Medrano-Bosch M, Simón-Codina B, Jiménez W, Edelman ER, Melgar-Lesmes P. Monocyte-endothelial cell interactions in vascular and tissue remodeling. Front Immunol 2023; 14:1196033. [PMID: 37483594 PMCID: PMC10360188 DOI: 10.3389/fimmu.2023.1196033] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
Monocytes are circulating leukocytes of innate immunity derived from the bone marrow that interact with endothelial cells under physiological or pathophysiological conditions to orchestrate inflammation, angiogenesis, or tissue remodeling. Monocytes are attracted by chemokines and specific receptors to precise areas in vessels or tissues and transdifferentiate into macrophages with tissue damage or infection. Adherent monocytes and infiltrated monocyte-derived macrophages locally release a myriad of cytokines, vasoactive agents, matrix metalloproteinases, and growth factors to induce vascular and tissue remodeling or for propagation of inflammatory responses. Infiltrated macrophages cooperate with tissue-resident macrophages during all the phases of tissue injury, repair, and regeneration. Substances released by infiltrated and resident macrophages serve not only to coordinate vessel and tissue growth but cellular interactions as well by attracting more circulating monocytes (e.g. MCP-1) and stimulating nearby endothelial cells (e.g. TNF-α) to expose monocyte adhesion molecules. Prolonged tissue accumulation and activation of infiltrated monocytes may result in alterations in extracellular matrix turnover, tissue functions, and vascular leakage. In this review, we highlight the link between interactions of infiltrating monocytes and endothelial cells to regulate vascular and tissue remodeling with a special focus on how these interactions contribute to pathophysiological conditions such as cardiovascular and chronic liver diseases.
Collapse
Affiliation(s)
- Mireia Medrano-Bosch
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Blanca Simón-Codina
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Wladimiro Jiménez
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
| | - Elazer R. Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Pedro Melgar-Lesmes
- Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
- Biochemistry and Molecular Genetics Service, Hospital Clínic Universitari, Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
17
|
Gumina DL, Su EJ. Mechanistic insights into the development of severe fetal growth restriction. Clin Sci (Lond) 2023; 137:679-695. [PMID: 37186255 PMCID: PMC10241202 DOI: 10.1042/cs20220284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/28/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023]
Abstract
Fetal growth restriction (FGR), which most commonly results from suboptimal placental function, substantially increases risks for adverse perinatal and long-term outcomes. The only "treatment" that exists is delivery, which averts stillbirth but does not improve outcomes in survivors. Furthermore, the potential long-term consequences of FGR to the fetus, including cardiometabolic disorders, predispose these individuals to developing FGR in their future pregnancies. This creates a multi-generational cascade of adverse effects stemming from a single dysfunctional placenta, and understanding the mechanisms underlying placental-mediated FGR is critically important if we are to improve outcomes and overall health. The mechanisms behind FGR remain unknown. However, placental insufficiency derived from maldevelopment of the placental vascular systems is the most common etiology. To highlight important mechanistic interactions within the placenta, we focus on placental vascular development in the setting of FGR. We delve into fetoplacental angiogenesis, a robust and ongoing process in normal pregnancies that is impaired in severe FGR. We review cellular models of FGR, with special attention to fetoplacental angiogenesis, and we highlight novel integrin-extracellular matrix interactions that regulate placental angiogenesis in severe FGR. In total, this review focuses on key developmental processes, with specific focus on the human placenta, an underexplored area of research.
Collapse
Affiliation(s)
- Diane L Gumina
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, CO, U.S.A
| | - Emily J Su
- Department of Obstetrics and Gynecology, University of Colorado School of Medicine, CO, U.S.A
| |
Collapse
|
18
|
Ødegaard KS, Westhrin M, Afif AB, Ma Q, Mela P, Standal T, Elverum CW, Torgersen J. The effects of surface treatments on electron beam melted Ti-6Al-4V disks on osteogenesis of human mesenchymal stromal cells. BIOMATERIALS ADVANCES 2023; 147:213327. [PMID: 36841111 DOI: 10.1016/j.bioadv.2023.213327] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/18/2023] [Accepted: 01/31/2023] [Indexed: 02/12/2023]
Abstract
Additive manufactured (AM) Titanium-6Aluminum-4Vanadium (Ti64) scaffolds display unique mechanical and biological properties for implant devices. The elastic modulus can be tailored by adjusting the porosity, further facilitating bone ingrowth. Although Ti64 implants are biocompatible, the effects of AM surfaces without porous structures, and how the topography and surface chemistry of the respective surfaces affect the osteogenesis of bone marrow-derived mesenchymal stromal cells (BMSCs) has not yet been revealed. In this paper, we cultured BMSCs on solid electron beam melted Ti64 disks subjected to three surface treatments: chemical etching (HF), atomic-layer deposition of TiO2 (TiO2), and polished (POL), or left untreated (AB). The biocompatibility and osteogenic properties of these surfaces were investigated, and the results were compared to cells cultured in regular tissue-culture polystyrene culturing wells (TCPS). The surfaces were hydrophobic, except for the polished surface which was hydrophilic. All surface treatments are biocompatible and allow for osteogenic differentiation, as revealed by viability assays and gene expression analysis. Scanning electron microscopy shows that cells adhere differently depending on the surface properties, with more filopodia on the rougher surfaces, AB and TiO2 disks, and more lamellipodia on the smoother surfaces, HF and POL disks. All groups stimulated with beta glycerophosphate, ascorbic acid, and dexamethasone, have elevated expression of genes related to matrix formation, where the cells cultured on the disks treated with TiO2, HF and POL have the overall highest expression. The AB group appears to be less favorable in regards to matrix formation. Considering the matrix mineralization, the rougher surfaces, AB and TiO2, are able to induce matrix mineralization, with an elevated gene expression of vitamin D receptors and calcium deposition of unstimulated cells. Finally, imaging at day 21 revealed an even amount of cells and matrix, covering most of the partially melted particles. Our results suggests that surface topography is more important to osteogenesis than the wettability of the surface. Overall, the present study contributes to the understanding of using surface modifications to AM Ti64 implant materials and reveals how they affect bone growth.
Collapse
Affiliation(s)
- Kristin S Ødegaard
- Department of Mechanical and Industrial Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Marita Westhrin
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Abdulla Bin Afif
- Department of Mechanical and Industrial Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Qianli Ma
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Petra Mela
- Chair of Medical Materials and Implants, Department of Mechanical Engineering, TUM School of Engineering and Design, Munich Institute for Biomedical Engineering, Technical University of Munich, Boltzmannstrasse 15, 85748 Garching bei München, Germany
| | - Therese Standal
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Christer W Elverum
- Department of Mechanical and Industrial Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Jan Torgersen
- Chair of Materials Science, Department of Materials Engineering, TUM School of Engineering and Design, Technical University of Munich, Boltzmannstrasse 15, 85748 Garching bei München, Germany.
| |
Collapse
|
19
|
Ihog proteins contribute to integrin-mediated focal adhesions. SCIENCE CHINA. LIFE SCIENCES 2023; 66:366-375. [PMID: 36103028 DOI: 10.1007/s11427-022-2154-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/27/2022] [Indexed: 10/14/2022]
Abstract
Integrin expression forms focal adhesions, but how this process is physiologically regulated is unclear. Ihog proteins are evolutionarily conserved, playing roles in Hedgehog signaling and serving as trans-homophilic adhesion molecules to mediate cell-cell interactions. Whether these proteins are also engaged in other cell adhesion processes remains unknown. Here, we report that Drosophila Ihog proteins function in the integrin-mediated adhesions. Removal of Ihog proteins causes blister and spheroidal muscle in wings and embryos, respectively. We demonstrate that Ihog proteins interact with integrin via the extracellular portion and that their removal perturbs integrin distribution. Finally, we show that Boc, a mammalian Ihog protein, rescues the embryonic defects caused by removing its Drosophila homologs. We thus propose that Ihog proteins contribute to integrin-mediated focal adhesions.
Collapse
|
20
|
Wang K, Wen D, Xu X, Zhao R, Jiang F, Yuan S, Zhang Y, Gao Y, Li Q. Extracellular matrix stiffness-The central cue for skin fibrosis. Front Mol Biosci 2023; 10:1132353. [PMID: 36968277 PMCID: PMC10031116 DOI: 10.3389/fmolb.2023.1132353] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/20/2023] [Indexed: 03/29/2023] Open
Abstract
Skin fibrosis is a physiopathological process featuring the excessive deposition of extracellular matrix (ECM), which is the main architecture that provides structural support and constitutes the microenvironment for various cellular behaviors. Recently, increasing interest has been drawn to the relationship between the mechanical properties of the ECM and the initiation and modulation of skin fibrosis, with the engagement of a complex network of signaling pathways, the activation of mechanosensitive proteins, and changes in immunoregulation and metabolism. Simultaneous with the progression of skin fibrosis, the stiffness of ECM increases, which in turn perturbs mechanical and humoral homeostasis to drive cell fate toward an outcome that maintains and enhances the fibrosis process, thus forming a pro-fibrotic "positive feedback loop". In this review, we highlighted the central role of the ECM and its dynamic changes at both the molecular and cellular levels in skin fibrosis. We paid special attention to signaling pathways regulated by mechanical cues in ECM remodeling. We also systematically summarized antifibrotic interventions targeting the ECM, hopefully enlightening new strategies for fibrotic diseases.
Collapse
Affiliation(s)
- Kang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dongsheng Wen
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuewen Xu
- Department of Burn and Plastic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rui Zhao
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Feipeng Jiang
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Shengqin Yuan
- School of Public Administration, Sichuan University, Chengdu, Sichuan, China
| | - Yifan Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Ya Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Yifan Zhang, ; Ya Gao, ; Qingfeng Li,
| |
Collapse
|
21
|
Al-Maslamani NA, Oldershaw R, Tew S, Curran J, D’Hooghe P, Yamamoto K, Horn HF. Chondrocyte De-Differentiation: Biophysical Cues to Nuclear Alterations. Cells 2022; 11:cells11244011. [PMID: 36552775 PMCID: PMC9777101 DOI: 10.3390/cells11244011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 12/14/2022] Open
Abstract
Autologous chondrocyte implantation (ACI) is a cell therapy to repair cartilage defects. In ACI a biopsy is taken from a non-load bearing area of the knee and expanded in-vitro. The expansion process provides the benefit of generating a large number of cells required for implantation; however, during the expansion these cells de-differentiate and lose their chondrocyte phenotype. In this review we focus on examining the de-differentiation phenotype from a mechanobiology and biophysical perspective, highlighting some of the nuclear mechanics and chromatin changes in chondrocytes seen during the expansion process and how this relates to the gene expression profile. We propose that manipulating chondrocyte nuclear architecture and chromatin organization will highlight mechanisms that will help to preserve the chondrocyte phenotype.
Collapse
Affiliation(s)
- Noor A. Al-Maslamani
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Correspondence:
| | - Rachel Oldershaw
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Simon Tew
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Jude Curran
- Department of Mechanical, Materials and Aerospace Engineering, School of Engineering, University of Liverpool, Liverpool L69 3GH, UK
| | - Pieter D’Hooghe
- Department of Orthopaedic Surgery, Aspetar Orthopaedic and Sports Medicine Hospital, Doha P.O. Box 29222, Qatar
| | - Kazuhiro Yamamoto
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Henning F. Horn
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| |
Collapse
|
22
|
Pedrosa L, Hoyos J, Reyes L, Llull L, Santana D, de Riva N, Mellado R, Sala X, Rodríguez-Hernández A, Enseñat J, Amaro S, Torné R. MicroRNA cerebrospinal fluid profile during the early brain injury period as a biomarker in subarachnoid hemorrhage patients. Front Cell Neurosci 2022; 16:1016814. [PMID: 36505512 PMCID: PMC9732100 DOI: 10.3389/fncel.2022.1016814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
Introduction Delayed cerebral ischemia (DCI) is a dreadful complication present in up to 30% of patients with spontaneous subarachnoid hemorrhage (SAH). Indeed, DCI is one of the main causes of long-term disability in SAH, yet its prediction and prevention are troublesome in poor-grade SAH cases. In this prospective study, we explored the potential role of micro ribonucleic acid (microRNA, abbreviated miRNAs)-small non-coding RNAs involved in clue gene regulation at the post-transcriptional level-as biomarkers of neurological outcomes in SAH patients. Methods We analyzed the expression of several miRNAs present in the cerebrospinal fluid (CSF) of SAH patients during the early stage of the disease (third-day post-hemorrhage). NanoString Technologies were used for the characterization of the CSF samples. Results We found an overexpression of miRNAs in the acute stage of 57 SAH in comparison with 10 non-SAH controls. Moreover, a differential expression of specific miRNAs was detected according to the severity of clinical onset, but also regarding the development of DCI and the midterm functional outcomes. Conclusion These observations reinforce the potential utility of miRNAs as prognostic and diagnostic biomarkers in SAH patients. In addition, the identification of specific miRNAs related to SAH evolution might provide insights into their regulatory functions of pathophysiological pathways, such as the TGF-β inflammatory pathway and blood-brain barrier disruption.
Collapse
Affiliation(s)
- Leire Pedrosa
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Jhon Hoyos
- Department of Neurosurgery, Institute of Neuroscience, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Luis Reyes
- Department of Neurosurgery, Institute of Neuroscience, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Laura Llull
- Comprehensive Stroke Center, Institute of Neuroscience, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Daniel Santana
- Comprehensive Stroke Center, Institute of Neuroscience, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Nicolás de Riva
- Neuroanesthesia Division, Department of Anesthesiology, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Ricard Mellado
- Department of Anesthesiology and Critical Care, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Xavier Sala
- Neuroanesthesia Division, Department of Anesthesiology, Hospital Clinic of Barcelona, Barcelona, Spain
| | | | - Joaquim Enseñat
- Department of Neurosurgery, Institute of Neuroscience, Hospital Clinic of Barcelona, Barcelona, Spain
| | - Sergio Amaro
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain,Comprehensive Stroke Center, Institute of Neuroscience, Hospital Clinic of Barcelona, Barcelona, Spain,Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain,*Correspondence: Sergio Amaro,
| | - Ramon Torné
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain,Department of Neurosurgery, Institute of Neuroscience, Hospital Clinic of Barcelona, Barcelona, Spain,Department of Medicine, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain,Ramon Torné,
| |
Collapse
|
23
|
Ji Y, Zhang Z, Hou W, Wu M, Wu H, Hu N, Ni M, Tang C, Wu F, Xu H. Enhanced antitumor effect of icariin nanoparticles coated with iRGD functionalized erythrocyte membrane. Eur J Pharmacol 2022; 931:175225. [PMID: 36002038 DOI: 10.1016/j.ejphar.2022.175225] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 01/10/2023]
Abstract
Lung cancer is the most common cause of incidence and mortality among tumor diseases. Icariin (ICA), a potential Chinese medicine monomer, has been reported to show outstanding antitumor effects. However, the hydrophobic nature and less tumor penetration limit its potential as a topical healing agent. There are few studies report the efficacy of ICA on lung cancer, moreover, there is no biomimetic targeted delivery system in the application of ICA. Herein, we firstly develop a novel ICA bionic targeted nano-preparation, camouflaged by the tumor penetrating peptide iRGD (cRGDKGPDC), functionalized red blood cell membrane (RBCM), has the increased solubility, utilized biocompatibility, and aggravated tumor penetration of ICA. In this study, we constructed the iRGD functionalized RBCM mimetic targeted ICA-loaded nanoparticles (iRINPs) and explored the anti-tumor effect of iRINPs against lung cancer with biochemical and behavioral analysis. The results suggested that iRINPs showed improved biocompatibility and stability, and reduced phagocytic uptakes by macrophages. Besides, the modification of iRGD significantly improved the targeting ability of iRINPs. In vitro and in vivo the treatment effects and safety assays showed that iRINPs attained better therapeutic effects than ICA by inhibiting A549 cell migration, proliferation and invasion, as well as reducing side effects of ICA. Overall, we expected that the new bionic nanocarriers would be a promising nano-platform for ICA in the precise therapy of lung cancer.
Collapse
Affiliation(s)
- You Ji
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211116, China
| | - Ziting Zhang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211116, China
| | - Wenjun Hou
- Department of Dermatology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, Nanjing, 210093, China
| | - Min Wu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211116, China
| | - Haisi Wu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211116, China
| | - Nan Hu
- Department of Oncology, The Affiliated Hospital of Kangda College of Nanjing Medical University & the First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Mengnan Ni
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211116, China
| | - Chunming Tang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211116, China.
| | - Fenglei Wu
- Department of Oncology, The Affiliated Hospital of Kangda College of Nanjing Medical University & the First People's Hospital of Lianyungang, Lianyungang, 222000, China.
| | - Huae Xu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211116, China.
| |
Collapse
|
24
|
Kausar S, Abbas MN, Gul I, Liu Y, Tang BP, Maqsood I, Liu QN, Dai LS. Integrins in the Immunity of Insects: A Review. Front Immunol 2022; 13:906294. [PMID: 35757717 PMCID: PMC9218073 DOI: 10.3389/fimmu.2022.906294] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/02/2022] [Indexed: 12/30/2022] Open
Abstract
Integrins are a large group of cell-surface proteins that are classified as transmembrane proteins. Integrins are classified into different types based on sequence variations, leading to structural and functional diversity. They are broadly distributed in animals and have a wide range of biological functions such as cell-to-cell communication, intracellular cytoskeleton organization, cellular signaling, immune responses, etc. Integrins are among the most abundant cell surface proteins in insects, exhibiting their indispensability in insect physiology. Because of their critical biological involvement in physiological processes, they appear to be a novel target for designing effective pest control strategies. In the current literature review, we first discuss the discovery and expression responses of integrins against various types of pathogens. Secondly, we examine the specific biological roles of integrins in controlling microbial pathogens, such as phagocytosis, encapsulation, nodulation, immune signaling, and so on. Finally, we describe the possible uses of integrins to control agricultural insect pests.
Collapse
Affiliation(s)
- Saima Kausar
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Isma Gul
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, China
| | - Yu Liu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bo-Ping Tang
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng, China
| | - Iram Maqsood
- Department of Zoology, Shaheed Benazir Bhutto Woman University, Peshawar, Pakistan
| | - Qiu-Ning Liu
- Jiangsu Key Laboratory for Bioresources of Saline Soils, Jiangsu Synthetic Innovation Center for Coastal Bio-agriculture, Jiangsu Provincial Key Laboratory of Coastal Wetland Bioresources and Environmental Protection, School of Wetlands, Yancheng Teachers University, Yancheng, China.,Key Laboratory of Insect Developmental and Evolutionary Biology, Chinese Academy of Sciences (CAS) Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Li-Shang Dai
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
25
|
Kamal NAMA, Abdulmalek E, Fakurazi S, Cordova KE, Abdul Rahman MB. Dissolution and Biological Assessment of Cancer-Targeting Nano-ZIF-8 in Zebrafish Embryos. ACS Biomater Sci Eng 2022; 8:2445-2454. [PMID: 35583465 DOI: 10.1021/acsbiomaterials.2c00186] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cancer-targeting nanotherapeutics offer promising opportunities for selective delivery of cytotoxic chemotherapeutics to cancer cells. However, the understanding of dissolution behavior and safety profiles of such nanotherapeutics is scarce. In this study, we report the dissolution profile of a cancer-targeting nanotherapeutic, gemcitabine (GEM) encapsulated within RGD-functionalized zeolitic imidazolate framework-8 (GEM⊂RGD@nZIF-8), in dissolution media having pH = 6.0 and 7.4. GEM⊂RGD@nZIF-8 was not only responsive in acidic media (pH = 6.0) but also able to sustain the dissolution rate (57.6%) after 48 h compared to non-targeting nanotherapeutic GEM⊂nZIF-8 (76%). This was reflected by the f2 value of 36.1, which indicated a difference in the dissolution behaviors of GEM⊂RGD@nZIF-8 and GEM⊂nZIF-8 in acidic media compared to those in neutral media (pH = 7.4). A dissolution kinetic study showed that the GEM release mechanism from GEM⊂RGD@nZIF-8 followed the Higuchi model. In comparison to a non-targeting nanotherapeutic, the cancer-targeting nanotherapeutic exhibited an enhanced permeability rate in healthy zebrafish embryos but did not induce lethality to 50% of the embryos (LC50 > 250 μg mL-1) with significantly improved survivability (75%) after 96 h of incubation. Monitoring malformation showed minimal adverse effects with only 8.3% of edema at 62.5 μg mL-1. This study indicates that cancer-targeting GEM⊂RGD@nZIF, with its pH-responsive behavior for sustaining chemotherapeutic dissolution in a physiologically relevant environment and its non-toxicity toward the healthy embryos within the tested concentrations, has considerable potential for use in cancer treatment.
Collapse
Affiliation(s)
- Nurul Akmarina Mohd Abdul Kamal
- Integrated Chemical BioPhysics Research, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia.,UPM-MAKNA Cancer Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.,Foundry of Reticular Materials for Sustainability (FORMS), Institute of Advanced Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.,Faculty of Chemical and Process Engineering Technology, Universiti Malaysia Pahang (UMP), Pekan 26600, Pahang, Malaysia
| | - Emilia Abdulmalek
- Integrated Chemical BioPhysics Research, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia.,Foundry of Reticular Materials for Sustainability (FORMS), Institute of Advanced Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Sharida Fakurazi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Kyle E Cordova
- Foundry of Reticular Materials for Sustainability (FORMS), Institute of Advanced Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.,Materials Discovery Research Unit, Advanced Research Centre, Royal Scientific Society, Amman 11941, Jordan
| | - Mohd Basyaruddin Abdul Rahman
- Integrated Chemical BioPhysics Research, Faculty of Science, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia.,UPM-MAKNA Cancer Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.,Foundry of Reticular Materials for Sustainability (FORMS), Institute of Advanced Technology, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
26
|
Wang Y, Han B, Liu K, Wang X. Effects of DDR1 on migration and adhesion of periodontal ligament cells and the underlying mechanism. J Periodontal Res 2022; 57:568-577. [PMID: 35297053 DOI: 10.1111/jre.12986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 02/13/2022] [Accepted: 03/09/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND OBJECTIVE As one of the widely expressed cell surface receptors binding to collagen, the most abundant component of the extracellular matrix (ECM), knowledge of the expression, functions, and mechanisms underlying the role of discoidin domain receptor 1 (DDR1) in human periodontal ligament cells (hPDLCs) is incomplete. This study determined the expression of DDR1 in hPDLCs and the effect of DDR1 upon migration and adhesion to hPDLCs, as well as the related regulatory mechanisms. MATERIALS AND METHODS The expression of DDR1 and the DDR1 isoforms in hPDLCs from six donors were tested. The migratory ability (horizontal and vertical) and adhesive capacity of hPDLCs with or without specific knockdown of DDR1 were evaluated. After treatment with MEK-ERK1/2 inhibitors (PD98059 and U0126) with or without RNAi, the migratory and adhesive capacity of hPDLCs were re-tested. Western blotting was performed to verify p-MEK1/2 and p-ERK1/2, the key factors of the MEK-ERK1/2 signaling pathways. RESULTS DDR1 was detected in hPDLCs in the mRNA and protein level; DDR1b was the dominant isoform. Knockdown of DDR1 almost halved the migratory capacity and significantly downregulated the adhesive capacity of hPDLCs. The use of MEK-ERK1/2 inhibitors caused declined migratory and adhesive capacity of hPDLCs as well. After DDR1 was knocked down, the expression of p-MEK and p-ERK protein declined significantly while total MEK and ERK showed no obvious change, which means the ratio of p-MEK/MEK and p-ERK/ERK was markedly reduced. CONCLUSIONS DDR1 plays an important role in the migration and adhesion of hPDLCs and might be regulated via the MEK-ERK1/2 signaling pathway.
Collapse
Affiliation(s)
- Yuhan Wang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Bing Han
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Kaining Liu
- Department of Periodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| | - Xiaoyan Wang
- Department of Cariology and Endodontology, Peking University School and Hospital of Stomatology & National Center of Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Laboratory for Digital and Material Technology of Stomatology & Beijing Key Laboratory of Digital Stomatology & Research Center of Engineering and Technology for Computerized Dentistry Ministry of Health & NMPA Key Laboratory for Dental Materials, Beijing, China
| |
Collapse
|
27
|
Bergonzini C, Kroese K, Zweemer AJM, Danen EHJ. Targeting Integrins for Cancer Therapy - Disappointments and Opportunities. Front Cell Dev Biol 2022; 10:863850. [PMID: 35356286 PMCID: PMC8959606 DOI: 10.3389/fcell.2022.863850] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 02/16/2022] [Indexed: 12/29/2022] Open
Abstract
Integrins mediate adhesive interactions between cells and their environment, including neighboring cells and extracellular matrix (ECM). These heterodimeric transmembrane receptors bind extracellular ligands with their globular head domains and connect to the cytoskeleton through multi-protein interactions at their cytoplasmic tails. Integrin containing cell–matrix adhesions are dynamic force-responsive protein complexes that allow bidirectional mechanical coupling of cells with their environment. This allows cells to sense and modulate tissue mechanics and regulates intracellular signaling impacting on cell faith, survival, proliferation, and differentiation programs. Dysregulation of these functions has been extensively reported in cancer and associated with tumor growth, invasion, angiogenesis, metastasis, and therapy resistance. This central role in multiple hallmarks of cancer and their localization on the cell surface makes integrins attractive targets for cancer therapy. However, despite a wealth of highly encouraging preclinical data, targeting integrin adhesion complexes in clinical trials has thus far failed to meet expectations. Contributing factors to therapeutic failure are 1) variable integrin expression, 2) redundancy in integrin function, 3) distinct roles of integrins at various disease stages, and 4) sequestering of therapeutics by integrin-containing tumor-derived extracellular vesicles. Despite disappointing clinical results, new promising approaches are being investigated that highlight the potential of integrins as targets or prognostic biomarkers. Improvement of therapeutic delivery at the tumor site via integrin binding ligands is emerging as another successful approach that may enhance both efficacy and safety of conventional therapeutics. In this review we provide an overview of recent encouraging preclinical findings, we discuss the apparent disagreement between preclinical and clinical results, and we consider new opportunities to exploit the potential of integrin adhesion complexes as targets for cancer therapy.
Collapse
|
28
|
Lamrani L, Adam F, Soukaseum C, Denis CV, Raslova H, Rosa J, Bryckaert M. New insights into regulation of αIIbβ3 integrin signaling by filamin A. Res Pract Thromb Haemost 2022; 6:e12672. [PMID: 35316942 PMCID: PMC8924993 DOI: 10.1002/rth2.12672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/29/2021] [Accepted: 01/05/2022] [Indexed: 12/14/2022] Open
Abstract
Background Filamin (FLN) regulates many cell functions through its scaffolding activity cross-linking cytoskeleton and integrins. FLN was shown to inhibit integrin activity, but the exact mechanism remains unclear. Objectives The aim of this study was to evaluate the role of filamin A (FLNa) subdomains on the regulation of integrin αIIbβ3 signaling. Methods Three FLNa deletion mutants were overexpressed in the erythro-megakaryocytic leukemic cell line HEL: Del1, which lacks the N-terminal CH1-CH2 domains mediating the FLNa-actin interaction; Del2, lacking the Ig-like repeat 21, which mediates the FLNa-β3 interaction; and Del3, lacking the C-terminal Ig repeat 24, responsible for FLNa dimerization and interaction with the small Rho guanosine triphosphatase involved in actin cytoskeleton reorganisation. Fibrinogen binding to HEL cells in suspension and talin-β3 proximity in cells adherent to immobilized fibrinogen were assessed before and after αIIbβ3 activation by the protein kinase C agonist phorbol 12-myristate 13-acetate. Results Our results show that FLNa-actin and FLNa-β3 interactions negatively regulate αIIbβ3 activation. Moreover, FLNa-actin interaction represses Rac activation, contributing to the negative regulation of αIIbβ3 activation. In contrast, the FLNa dimerization domain, which maintains Rho inactive, was found to negatively regulate αIIbβ3 outside-in signaling. Conclusion We conclude that FLNa negatively controls αIIbβ3 activation by regulating actin polymerization and restraining activation of Rac, as well as outside-in signaling by repressing Rho.
Collapse
Affiliation(s)
- Lamia Lamrani
- HIThUMR_S1176Institut National de la Santé et de la Recherche MédicaleUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Frédéric Adam
- HIThUMR_S1176Institut National de la Santé et de la Recherche MédicaleUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Christelle Soukaseum
- HIThUMR_S1176Institut National de la Santé et de la Recherche MédicaleUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Cécile V. Denis
- HIThUMR_S1176Institut National de la Santé et de la Recherche MédicaleUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Hana Raslova
- UMR_S1170Institut National de la Santé et de la Recherche MédicaleUniversité Paris‐SudUniversité Paris‐SaclayGustave Roussy Cancer CampusEquipe Labellisée Ligue Nationale Contre le CancerVillejuifFrance
| | - Jean‐Philippe Rosa
- HIThUMR_S1176Institut National de la Santé et de la Recherche MédicaleUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| | - Marijke Bryckaert
- HIThUMR_S1176Institut National de la Santé et de la Recherche MédicaleUniversité Paris‐SaclayLe Kremlin‐BicêtreFrance
| |
Collapse
|
29
|
Yoon SS, Kwon HW, Shin JH, Rhee MH, Park CE, Lee DH. Anti-Thrombotic Effects of Artesunate through Regulation of cAMP and PI3K/MAPK Pathway on Human Platelets. Int J Mol Sci 2022; 23:1586. [PMID: 35163507 PMCID: PMC8836205 DOI: 10.3390/ijms23031586] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/29/2022] Open
Abstract
Normal activation of platelets and their aggregation are crucial for proper hemostasis. It appears that excessive or abnormal aggregation of platelets may bring about cardiovascular diseases such as stroke, atherosclerosis, and thrombosis. For this reason, finding a substance that can regulate platelet aggregation or suppress aggregation will aid in the prevention and treatment of cardiovascular diseases. Artesunate is a compound extracted from the plant roots of Artemisia or Scopolia, and its effects have shown to be promising in areas of anticancer and Alzheimer's disease. However, the role and mechanisms by which artesunate affects the aggregation of platelets and the formation of a thrombus are currently not understood. This study examines the ways artesunate affects the aggregation of platelets and the formation of a thrombus on platelets induced by U46619. As a result, cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) production were increased significantly by artesunate relative to the doses, as well as phosphorylated vasodilator-stimulated phosphoprotein (VASP) and inositol 1,4,5-trisphosphate receptor (IP3R), substrates to cAMP-dependent kinase and cGMP-dependent kinase, in a significant manner. The Ca2+, normally mobilized from the dense tubular system, was inhibited due to IP3R phosphorylation from artesunate, and phosphorylated VASP aided in inhibiting platelet activity via αIIb/β3 platelet membrane inactivation and inhibiting fibrinogen binding. In addition, MAPK and PI3K/Akt phosphorylation was inhibited via artesunate in a significant manner, causing the production of TXA2 and intracellular granular secretion (serotonin and ATP release) to be reduced. Therefore, we suggest that artesunate has value as a substance that inhibits platelet aggregation and thrombus formation through an antiplatelet mechanism.
Collapse
Affiliation(s)
- Shin-Sook Yoon
- Department of Biomedical Laboratory Science, Namseoul University, Cheonan 31020, Korea; (S.-S.Y.); (C.-E.P.)
| | - Hyuk-Woo Kwon
- Department of Biomedical Laboratory Science, Far East University, Eumseong 27601, Korea;
| | - Jung-Hae Shin
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea; (J.-H.S.); (M.H.R.)
| | - Man Hee Rhee
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea; (J.-H.S.); (M.H.R.)
| | - Chang-Eun Park
- Department of Biomedical Laboratory Science, Namseoul University, Cheonan 31020, Korea; (S.-S.Y.); (C.-E.P.)
- Molecular Diagnostics Research Institute, Namseoul University, Cheonan 31020, Korea
| | - Dong-Ha Lee
- Department of Biomedical Laboratory Science, Namseoul University, Cheonan 31020, Korea; (S.-S.Y.); (C.-E.P.)
- Molecular Diagnostics Research Institute, Namseoul University, Cheonan 31020, Korea
| |
Collapse
|
30
|
Irfan M, Kwon TH, Kwon HW, Rhee MH. Pharmacological actions of Dieckol on modulation of platelet functions and thrombus formation via integrin α IIbβ 3 and cAMP signaling. Pharmacol Res 2022; 177:106088. [PMID: 35038555 DOI: 10.1016/j.phrs.2022.106088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/29/2021] [Accepted: 01/12/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND PURPOSE Dieckol is a phlorotannin that can be found in seaweeds, particularly in Eisenia bicyclis (brown algae) and is known to have anti-oxidant, anti-inflammatory, and anti-microbial properties. It also possesses anti-thrombotic and pro-fibrinolytic activities; however, the mechanistic aspects of anti-platelet and anti-thrombotic activity are yet to be explored. STUDY DESIGN and Methodology: We investigated the pharmacological effects of dieckol on the modulation of platelet functions using human, rat, and mice models. Inhibitory effects of dieckol on platelet aggregation were assessed using platelet-rich plasma and washed platelets, followed by measurement of dense granule secretions, fibrinogen binding to integrin αIIbβ3, fibronectin adhesion assay, platelet spreading on immobilized fibrinogen, and clot retraction. Cyclic nucleotide signaling events were evaluated, such as cyclic-AMP production followed by vasodilator-stimulated phosphoprotein (VASP) stimulation. The in vivo anti-thrombotic potential was evaluated in mice using an acute pulmonary thromboembolism model and tail bleeding assay. RESULTS Dieckol markedly inhibited platelet aggregation and granule secretion; furthermore, it down-regulated integrin αIIbβ3-mediated inside-out and outside-in signaling events, including platelet adhesion, spreading, and clot retraction, whereas it upregulated the cAMP-PKA-VASP pathway. Dieckol-treated mice significantly survived the thrombosis than vehicle treated mice, without affecting hemostasis. Histological examinations of lungs revealed minimum occluded vasculature in dieckol-treated mice. CONCLUSION Dieckol possesses strong anti-platelet and anti-thrombotic properties and is a potential therapeutic drug candidate to treat and prevent platelet-related cardiovascular disorders.
Collapse
Affiliation(s)
- Muhammad Irfan
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea; Department of Oral Biology, College of Dentistry, University of Illinois at Chicago, Chicago 60612, IL, USA
| | - Tae-Hyung Kwon
- Chuncheon Bio Industry Foundation, Chuncheon 24232, Republic of Korea
| | - Hyuk-Woo Kwon
- Department of Biomedical Laboratory Science, Far East University, Eumseong 27601, Republic of Korea
| | - Man Hee Rhee
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
31
|
Sharifi-Rad J, Quispe C, Zam W, Kumar M, Cardoso SM, Pereira OR, Ademiluyi AO, Adeleke O, Moreira AC, Živković J, Noriega F, Ayatollahi SA, Kobarfard F, Faizi M, Martorell M, Cruz-Martins N, Butnariu M, Bagiu IC, Bagiu RV, Alshehri MM, Cho WC. Phenolic Bioactives as Antiplatelet Aggregation Factors: The Pivotal Ingredients in Maintaining Cardiovascular Health. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:2195902. [PMID: 34447485 PMCID: PMC8384526 DOI: 10.1155/2021/2195902] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 07/28/2021] [Indexed: 01/22/2023]
Abstract
Cardiovascular diseases (CVD) are one of the main causes of mortality in the world. The development of these diseases has a specific factor-alteration in blood platelet activation. It has been shown that phenolic compounds have antiplatelet aggregation abilities and a positive impact in the management of CVD, exerting prominent antioxidant, anti-inflammatory, antitumor, cardioprotective, antihyperglycemic, and antimicrobial effects. Thus, this review is intended to address the antiplatelet activity of phenolic compounds with special emphasis in preventing CVD, along with the mechanisms of action through which they are able to prevent and treat CVD. In vitro and in vivo studies have shown beneficial effects of phenolic compound-rich plant extracts and isolated compounds against CVD, despite that the scientific literature available on the antiplatelet aggregation ability of phenolic compounds in vivo is scarce. Thus, despite the current advances, further studies are needed to confirm the cardioprotective potential of phenolic compounds towards their use alone or in combination with conventional drugs for effective therapeutic interventions.
Collapse
Affiliation(s)
- Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Cristina Quispe
- Facultad de Ciencias de la Salud, Universidad Arturo Prat, Avda. Arturo Prat 2120, Iquique 1110939, Chile
| | - Wissam Zam
- Department of Analytical and Food Chemistry, Faculty of Pharmacy, Al-Andalus University for Medical Sciences, Tartous, Syria
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR-Central Institute for Research on Cotton Technology, Mumbai 400019, India
| | - Susana M. Cardoso
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Olivia R. Pereira
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Adedayo O. Ademiluyi
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| | - Oluwakemi Adeleke
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
- Department of Science Laboratory Technology, Ekiti State University, Ado-Ekiti, Nigeria
| | | | - Jelena Živković
- Institute for Medicinal Plants Research “Dr. Josif Pančić”, Tadeuša Košćuška 1, 11000 Belgrade, Serbia
| | - Felipe Noriega
- Department of Plant Production, Faculty of Agronomy, Universidad de Concepción, Chillan 4070386, Chile
| | - Seyed Abdulmajid Ayatollahi
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
- Department of Pharmacognosy and Biotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farzad Kobarfard
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehrdad Faizi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Miquel Martorell
- Department of Nutrition and Dietetics, Faculty of Pharmacy, and Centre for Healthy Living, University of Concepción, 4070386 Concepción, Chile
| | - Natália Cruz-Martins
- Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute for Research and Innovation in Health (i3S), University of Porto, 4200-135 Porto, Portugal
- Institute of Research and Advanced Training in Health Sciences and Technologies (CESPU), Rua Central de Gandra, 1317, 4585-116 Gandra PRD, Portugal
| | - Monica Butnariu
- Banat's University of Agricultural Sciences and Veterinary Medicine “King Michael I of Romania” from Timisoara, Timisoara, Romania
| | - Iulia Cristina Bagiu
- Victor Babes University of Medicine and Pharmacy of Timisoara, Department of Microbiology, Timisoara, Romania
- Multidisciplinary Research Center on Antimicrobial Resistance, Timisoara, Romania
| | - Radu Vasile Bagiu
- Victor Babes University of Medicine and Pharmacy of Timisoara, Department of Microbiology, Timisoara, Romania
- Preventive Medicine Study Center, Timisoara, Romania
| | - Mohammed M. Alshehri
- Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong
| |
Collapse
|
32
|
Benwell CJ, Taylor JAGE, Robinson SD. Endothelial neuropilin-2 influences angiogenesis by regulating actin pattern development and α5-integrin-p-FAK complex recruitment to assembling adhesion sites. FASEB J 2021; 35:e21679. [PMID: 34314542 DOI: 10.1096/fj.202100286r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 01/02/2023]
Abstract
The ability to form a variety of cell-matrix connections is crucial for angiogenesis to take place. Without stable anchorage to the extracellular matrix (ECM), endothelial cells (ECs) are unable to sense, integrate and disseminate growth factor stimulated responses that drive growth of a vascular bed. Neuropilin-2 (NRP2) is a widely expressed membrane-bound multifunctional non-tyrosine kinase receptor, which has previously been implicated in influencing cell adhesion and migration by interacting with α5-integrin and regulating adhesion turnover. α5-integrin, and its ECM ligand fibronectin (FN) are both known to be upregulated during the formation of neo-vasculature. Despite being descriptively annotated as a candidate biomarker for aggressive cancer phenotypes, the EC-specific roles for NRP2 during developmental and pathological angiogenesis remain unexplored. The data reported here support a model whereby NRP2 actively promotes EC adhesion and migration by regulating dynamic cytoskeletal remodeling and by stimulating Rab11-dependent recycling of α5-integrin-p-FAK complexes to newly assembling adhesion sites. Furthermore, temporal depletion of EC-NRP2 in vivo impairs primary tumor growth by disrupting vessel formation. We also demonstrate that EC-NRP2 is required for normal postnatal retinal vascular development, specifically by regulating cell-matrix adhesion. Upon loss of endothelial NRP2, vascular outgrowth from the optic nerve during superficial plexus formation is disrupted, likely due to reduced FAK phosphorylation within sprouting tip cells.
Collapse
Affiliation(s)
- Christopher J Benwell
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - James A G E Taylor
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Stephen D Robinson
- Gut Microbes and Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.,School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich, UK
| |
Collapse
|
33
|
Nabai L, Pourghadiri A, Ghahary A. Hypertrophic Scarring: Current Knowledge of Predisposing Factors, Cellular and Molecular Mechanisms. J Burn Care Res 2021; 41:48-56. [PMID: 31999336 DOI: 10.1093/jbcr/irz158] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hypertrophic scarring (HSc) is an age-old problem that still affects millions of people physically, psychologically, and economically. Despite advances in surgical techniques and wound care, prevention and treatment of HSc remains a challenge. Elucidation of factors involved in the development of this common fibroproliferative disorder is crucial for further progress in preventive and/or therapeutic measures. Our knowledge about pathophysiology of HSc at the cellular and molecular level has grown considerably in recent decades. In this article, current knowledge of predisposing factors and the cellular and molecular mechanisms of HSc has been reviewed.
Collapse
Affiliation(s)
- Layla Nabai
- BC Professional Firefighters' Burn & Wound Healing Research Laboratory, Department of Surgery, Division of Plastic Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Amir Pourghadiri
- BC Professional Firefighters' Burn & Wound Healing Research Laboratory, Department of Surgery, Division of Plastic Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aziz Ghahary
- BC Professional Firefighters' Burn & Wound Healing Research Laboratory, Department of Surgery, Division of Plastic Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
34
|
Irfan M, Lee YY, Lee KJ, Kim SD, Rhee MH. Comparative antiplatelet and antithrombotic effects of red ginseng and fermented red ginseng extracts. J Ginseng Res 2021; 46:387-395. [PMID: 35600768 PMCID: PMC9120646 DOI: 10.1016/j.jgr.2021.05.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/10/2021] [Accepted: 05/30/2021] [Indexed: 12/13/2022] Open
Affiliation(s)
- Muhammad Irfan
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Oral Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Yuan Yee Lee
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ki-Ja Lee
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sung Dae Kim
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Man Hee Rhee
- Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
- Corresponding author. Department of Veterinary Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
35
|
Zhu GX, Zuo JL, Xu L, Li SQ. Ginsenosides in vascular remodeling: Cellular and molecular mechanisms of their therapeutic action. Pharmacol Res 2021; 169:105647. [PMID: 33964471 DOI: 10.1016/j.phrs.2021.105647] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/09/2021] [Accepted: 04/26/2021] [Indexed: 12/14/2022]
Abstract
Evidence is mounting that abnormal vascular remodeling (VR) is a vital pathological event that precedes many cardiovascular diseases (CVD). This provides us with a new research perspective that VR can be a pivotal target for CVD treatment and prevention. However, the current drugs for treating CVD do not fundamentally reverse VR and repair vascular function. The reason may be that a complicated regulatory network is formed between the various signaling pathways involved in VR. Recently, ginsenoside, the main active substance of ginseng, has become increasingly the focus of many researchers for its multiple targets, multiple pathways, and few side effects. Several data have revealed that ginsenosides can improve VR caused by vasodilation dysfunction, abnormal vascular structure and blood pressure. This review is intended to discuss the therapeutic effects and mechanisms of ginsenosides in some diseases involved in VR. Besides, we herein also give a new and contradictory insight into intracellular and molecular signaling of ginsenosides in all kinds of vascular cells. Most importantly, we also discuss the feasibility of ginsenosides Rb1/Rg1/Rg3 in drug development by combining the pharmacodynamics and pharmacokinetics of ginsenosides, and provide a pharmacological basis for the development of ginsenosides in clinical applications.
Collapse
Affiliation(s)
- Guang-Xuan Zhu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China.
| | - Jian-Li Zuo
- College of Pharmacy, Chongqing Medical University, Chongqing 410016, China
| | - Lin Xu
- College of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563000, China
| | - Shu-Qing Li
- The Second Xiangya Hospital of Central South University Shenzhen Hospital, Shenzhen, Guangdong 518067, China
| |
Collapse
|
36
|
Alday-Parejo B, Ghimire K, Coquoz O, Albisetti GW, Tamò L, Zaric J, Stalin J, Rüegg C. MAGI1 localizes to mature focal adhesion and modulates endothelial cell adhesion, migration and angiogenesis. Cell Adh Migr 2021; 15:126-139. [PMID: 33823745 PMCID: PMC8115569 DOI: 10.1080/19336918.2021.1911472] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
MAGI1 is an intracellular adaptor protein that stabilizes cell junctions and regulates epithelial and endothelial integrity. Here, we report that that in endothelial cells MAGI1 colocalizes with paxillin, β3-integrin, talin 1, tensin 3 and α-4-actinin at mature focal adhesions and actin stress fibers, and regulates their dynamics. Downregulation of MAGI1 reduces focal adhesion formation and maturation, cell spreading, actin stress fiber formation and RhoA/Rac1 activation. MAGI1 silencing increases phosphorylation of paxillin at Y118, an indicator of focal adhesion turnover. MAGI1 promotes integrin-dependent endothelial cells adhesion to ECM, reduces invasion and tubulogenesisin vitro and suppresses angiogenesis in vivo. Our results identify MAGI1 as anovel component of focal adhesions, and regulator of focal adhesion dynamics, cell adhesion, invasion and angiogenesis.
Collapse
Affiliation(s)
- Begoña Alday-Parejo
- Laboratory of Experimental and Translational Oncology, Pathology, Department of Oncology, Microbiology and Immunology, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Kedar Ghimire
- Laboratory of Experimental and Translational Oncology, Pathology, Department of Oncology, Microbiology and Immunology, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland.,Westmead Institute for Medical Research, University of Sydney, Sydney, Australia
| | - Oriana Coquoz
- Laboratory of Experimental and Translational Oncology, Pathology, Department of Oncology, Microbiology and Immunology, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Gioele W Albisetti
- Laboratory of Experimental and Translational Oncology, Pathology, Department of Oncology, Microbiology and Immunology, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland.,Institute of Pharmacology and Toxicology, Section of Neuropharmacology, University of Zürich, Zürich, Switzerland
| | - Luca Tamò
- Laboratory of Experimental and Translational Oncology, Pathology, Department of Oncology, Microbiology and Immunology, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland.,Clinical Trials Unit, University of Bern, Bern, Switzerland
| | - Jelena Zaric
- Laboratory of Experimental and Translational Oncology, Pathology, Department of Oncology, Microbiology and Immunology, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland.,Swiss Institute for Experimental Cancer Research, Ecole Polytechnique Fédérale De Lausanne, Lausanne, Switzerland
| | - Jimmy Stalin
- Laboratory of Experimental and Translational Oncology, Pathology, Department of Oncology, Microbiology and Immunology, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Curzio Rüegg
- Laboratory of Experimental and Translational Oncology, Pathology, Department of Oncology, Microbiology and Immunology, Faculty of Sciences and Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
37
|
Okubo K, Brenner MD, Cullere X, Saggu G, Patchen ML, Bose N, Mihori S, Yuan Z, Lowell CA, Zhu C, Mayadas TN. Inhibitory affinity modulation of FcγRIIA ligand binding by glycosphingolipids by inside-out signaling. Cell Rep 2021; 35:109142. [PMID: 34010642 PMCID: PMC8218468 DOI: 10.1016/j.celrep.2021.109142] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 01/19/2021] [Accepted: 04/26/2021] [Indexed: 12/22/2022] Open
Abstract
The interaction of the human FcγRIIA with immune complexes (ICs) promotes neutrophil activation and thus must be tightly controlled to avoid damage to healthy tissue. Here, we demonstrate that a fungal-derived soluble β-1,3/1,6-glucan binds to the glycosphingolipid long-chain lactosylceramide (LacCer) to reduce FcγRIIA-mediated recruitment to immobilized ICs under flow, a process requiring high-affinity FcγRIIA-immunoglobulin G (IgG) interactions. The inhibition requires Lyn phosphorylation of SHP-1 phosphatase and the FcγRIIA immunotyrosine-activating motif. β-glucan reduces the effective 2D affinity of FcγRIIA for IgG via Lyn and SHP-1 and, in vivo, inhibits FcγRIIA-mediated neutrophil recruitment to intravascular IgG deposited in the kidney glomeruli in a glycosphingolipid- and Lyn-dependent manner. In contrast, β-glucan did not affect FcγR functions that bypass FcγR affinity for IgG. In summary, we have identified a pathway for modulating the 2D affinity of FcγRIIA for ligand that relies on LacCer-Lyn-SHP-1-mediated inhibitory signaling triggered by β-glucan, a previously described activator of innate immunity. Okubo et al. demonstrate that β-glucan binding to the glycosphingolipid lactosylceramide engages a Lyn kinase to SHP-1 phosphatase pathway that reduces FcγRIIA binding propensity for IgG, which suggests FcγRIIA affinity regulation by “inside-out” signaling. The β-glucan-lactosylceramide-Lyn axis prevents FcγRIIA-dependent neutrophil recruitment in vitro and to intravascular IgG deposits following glomerulonephritis.
Collapse
Affiliation(s)
- Koshu Okubo
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Michael D Brenner
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Xavier Cullere
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Gurpanna Saggu
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | | | - Nandita Bose
- Biothera Pharmaceuticals, Inc., Eagan, Minnesota, MN 55121, USA
| | - Saki Mihori
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA
| | - Zhou Yuan
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Clifford A Lowell
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143, USA
| | - Cheng Zhu
- Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Tanya N Mayadas
- Department of Pathology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Karagöz Z, Geuens T, LaPointe VLS, van Griensven M, Carlier A. Win, Lose, or Tie: Mathematical Modeling of Ligand Competition at the Cell-Extracellular Matrix Interface. Front Bioeng Biotechnol 2021; 9:657244. [PMID: 33996781 PMCID: PMC8117103 DOI: 10.3389/fbioe.2021.657244] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/07/2021] [Indexed: 12/19/2022] Open
Abstract
Integrin transmembrane proteins conduct mechanotransduction at the cell–extracellular matrix (ECM) interface. This process is central to cellular homeostasis and therefore is particularly important when designing instructive biomaterials and organoid culture systems. Previous studies suggest that fine-tuning the ECM composition and mechanical properties can improve organoid development. Toward the bigger goal of fully functional organoid development, we hypothesize that resolving the dynamics of ECM–integrin interactions will be highly instructive. To this end, we developed a mathematical model that enabled us to simulate three main interactions, namely integrin activation, ligand binding, and integrin clustering. Different from previously published computational models, we account for the binding of more than one type of ligand to the integrin. This competition between ligands defines the fate of the system. We have demonstrated that an increase in the initial concentration of ligands does not ensure an increase in the steady state concentration of ligand-bound integrins. The ligand with higher binding rate occupies more integrins at the steady state than does the competing ligand. With cell type specific, quantitative input on integrin-ligand binding rates, this model can be used to develop instructive cell culture systems.
Collapse
Affiliation(s)
- Zeynep Karagöz
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Thomas Geuens
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Vanessa L S LaPointe
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Martijn van Griensven
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Aurélie Carlier
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
39
|
Antiplatelet and Antithrombotic Effects of Epimedium koreanum Nakai. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:7071987. [PMID: 33953788 PMCID: PMC8068545 DOI: 10.1155/2021/7071987] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 02/26/2021] [Accepted: 04/03/2021] [Indexed: 12/02/2022]
Abstract
Background and Objective. Epimedium koreanum Nakai is a medicinal plant known for its health beneficial effects on impotence, arrhythmia, oxidation, aging, osteoporosis, and cardiovascular diseases. However, there is no report available that shows its effects on platelet functions. Here, we elucidated antiplatelet and antithrombotic effects of ethyl acetate fraction of E. koreanum. Methodology. We analyzed the antiplatelet properties using standard in vitro and in vivo techniques, such as light transmission aggregometry, scanning electron microscopy, intracellular calcium mobilization measurement, dense granule secretion, and flow cytometry to assess integrin αIIbβ3 activation, clot retraction, and Western blot, on washed platelets. The antithrombotic effects of E. koreanum were assessed by arteriovenous- (AV-) shunt model in rats, and its effects on hemostasis were analyzed by tail bleeding assay in mice. Key Results. E. koreanum inhibited platelet aggregation in agonist-stimulated human and rat washed platelets, and it also reduced calcium mobilization, ATP secretion, and TXB2 formation. Fibrinogen binding, fibronectin adhesion, and clot retraction by attenuated integrin αIIbβ3-mediated inside-out and outside-in signaling were also decreased. Reduced phosphorylation of extracellular signal-regulated kinases (ERK), Akt, PLCγ2, and Src was observed. Moreover, the fraction inhibited thrombosis. HPLC results revealed that the fraction predominantly contained icariin. Conclusion and Implications. E. koreanum inhibited platelet aggregation and thrombus formation by attenuating calcium mobilization, ATP secretion, TXB2 formation, and integrin αIIbβ3 activation. Therefore, it may be considered as a potential candidate to treat and prevent platelet-related cardiovascular disorders.
Collapse
|
40
|
Goggins BJ, Minahan K, Sherwin S, Soh WS, Pryor J, Bruce J, Liu G, Mathe A, Knight D, Horvat JC, Walker MM, Keely S. Pharmacological HIF-1 stabilization promotes intestinal epithelial healing through regulation of α-integrin expression and function. Am J Physiol Gastrointest Liver Physiol 2021; 320:G420-G438. [PMID: 33470153 DOI: 10.1152/ajpgi.00192.2020] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 01/31/2023]
Abstract
Intestinal epithelia are critical for maintaining gastrointestinal homeostasis. Epithelial barrier injury, causing inflammation and vascular damage, results in inflammatory hypoxia, and thus, healing occurs in an oxygen-restricted environment. The transcription factor hypoxia-inducible factor (HIF)-1 regulates genes important for cell survival and repair, including the cell adhesion protein β1-integrin. Integrins function as αβ-dimers, and α-integrin-matrix binding is critical for cell migration. We hypothesized that HIF-1 stabilization accelerates epithelial migration through integrin-dependent pathways. We aimed to examine functional and posttranslational activity of α-integrins during HIF-1-mediated intestinal epithelial healing. Wound healing was assessed in T84 monolayers over 24 h with/without prolyl-hydroxylase inhibitor (PHDi) (GB-004), which stabilizes HIF-1. Gene and protein expression were measured by RT-PCR and immunoblot, and α-integrin localization was assessed by immunofluorescence. α-integrin function was assessed by antibody-mediated blockade, and integrin α6 regulation was determined by HIF-1α chromatin immunoprecipitation. Models of mucosal wounding and 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced colitis were used to examine integrin expression and localization in vivo. PHDi treatment accelerated wound closure and migration within 12 h, associated with increased integrin α2 and α6 protein, but not α3. Functional blockade of integrins α2 and α6 inhibited PHDi-mediated accelerated wound closure. HIF-1 bound directly to the integrin α6 promoter. PHDi treatment accelerated mucosal healing, which was associated with increased α6 immunohistochemical staining in wound-associated epithelium and wound-adjacent tissue. PHDi treatment increased α6 protein levels in colonocytes of TNBS mice and induced α6 staining in regenerating crypts and reepithelialized inflammatory lesions. Together, these data demonstrate a role for HIF-1 in regulating both integrin α2 and α6 responses during intestinal epithelial healing.NEW & NOTEWORTHY HIF-1 plays an important role in epithelial restitution, selectively inducing integrins α6 and α2 to promote migration and proliferation, respectively. HIF-stabilizing prolyl-hydroxylase inhibitors accelerate intestinal mucosal healing by inducing epithelial integrin expression.
Collapse
Affiliation(s)
- Bridie J Goggins
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Kyra Minahan
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Simonne Sherwin
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Wai S Soh
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Jennifer Pryor
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Jessica Bruce
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Gang Liu
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Andrea Mathe
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Darryl Knight
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Jay C Horvat
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Marjorie M Walker
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| | - Simon Keely
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
- Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Gastrointestinal Research Group, University of Newcastle, New South Wales, Australia
- Priority Research Centre for Digestive Health and Neurogastroenterology, Hunter Medical Research Institute and University of Newcastle, Newcastle, New South Wales, Australia
| |
Collapse
|
41
|
Young KA, Biggins L, Sharpe HJ. Protein tyrosine phosphatases in cell adhesion. Biochem J 2021; 478:1061-1083. [PMID: 33710332 PMCID: PMC7959691 DOI: 10.1042/bcj20200511] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 02/07/2023]
Abstract
Adhesive structures between cells and with the surrounding matrix are essential for the development of multicellular organisms. In addition to providing mechanical integrity, they are key signalling centres providing feedback on the extracellular environment to the cell interior, and vice versa. During development, mitosis and repair, cell adhesions must undergo extensive remodelling. Post-translational modifications of proteins within these complexes serve as switches for activity. Tyrosine phosphorylation is an important modification in cell adhesion that is dynamically regulated by the protein tyrosine phosphatases (PTPs) and protein tyrosine kinases. Several PTPs are implicated in the assembly and maintenance of cell adhesions, however, their signalling functions remain poorly defined. The PTPs can act by directly dephosphorylating adhesive complex components or function as scaffolds. In this review, we will focus on human PTPs and discuss their individual roles in major adhesion complexes, as well as Hippo signalling. We have collated PTP interactome and cell adhesome datasets, which reveal extensive connections between PTPs and cell adhesions that are relatively unexplored. Finally, we reflect on the dysregulation of PTPs and cell adhesions in disease.
Collapse
Affiliation(s)
- Katherine A. Young
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Laura Biggins
- Bioinformatics, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Hayley J. Sharpe
- Signalling Programme, Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, U.K
| |
Collapse
|
42
|
Ghosh N, Garg I, Srivastava S, Kumar B. Influence of integrins on thrombus formation: a road leading to the unravelling of DVT. Mol Cell Biochem 2021; 476:1489-1504. [PMID: 33398665 DOI: 10.1007/s11010-020-03961-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/23/2020] [Indexed: 01/18/2023]
Abstract
Integrins are a group of transmembrane glycoprotein receptors that are responsible for platelet activation through bidirectional signalling. These receptors have left their footprints in various cellular events and have intrigued many groups of scientists that have led to some significant discoveries. A lot of the recent understanding of haemostasis has been possible due to the integrins filling the gaps in between several cellular mechanism. Apart from this, other important functions carried out by integrins are growth and maturation of cardiomyocytes, mechano-transduction, and interaction with actin cytoskeleton. The signalling cascade for integrin activation involves certain intracellular interacting proteins, which initiates the step-by-step activation procedure through 'inside-out' signalling. The signalling cascade gets activated through 'outside-in' signalling with the involvement of agonists such as ADP, Fibronectin, Vitronectin, and so on. This is a crucial step for the downstream processes of platelet spreading, followed by aggregation, clot progression and finally thrombus formation. Researchers throughout the world have shown direct relation of integrins with CVD and cardiac remodelling. The present review aims to summarize the information available so far on the involvement of integrins in thrombosis and its relationship to DVT. This information could be a bedrock of hidden answers to several questions on pathogenesis of deep vein thrombosis.
Collapse
Affiliation(s)
- Nilanjana Ghosh
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Iti Garg
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India.
| | - Swati Srivastava
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| | - Bhuvnesh Kumar
- Defence Institute of Physiology and Allied Sciences (DIPAS), Defence Research and Development Organization (DRDO), Lucknow Road, Timarpur, Delhi, 110054, India
| |
Collapse
|
43
|
Yin M, Li C, Jiang J, Le J, Luo B, Yang F, Fang Y, Yang M, Deng Z, Ni W, Shao J. Cell adhesion molecule-mediated therapeutic strategies in atherosclerosis: From a biological basis and molecular mechanism to drug delivery nanosystems. Biochem Pharmacol 2021; 186:114471. [PMID: 33587918 DOI: 10.1016/j.bcp.2021.114471] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/30/2021] [Accepted: 02/08/2021] [Indexed: 01/13/2023]
Abstract
Atherosclerosis (AS), characterized by pathological constriction of blood vessels due to chronic low-grade inflammation and lipid deposition, is a leading cause of human morbidity and mortality worldwide. Cell adhesion molecules (CAMs) have the ability to regulate the inflammatory response and endothelial function, as well as potentially driving plaque rupture, which all contribute to the progression of AS. Moreover, recent advances in the development of clinical agents in the cardiovascular field are based on CAMs, which show promising results in the fight against AS. Here, we review the current literature on mechanisms by which CAMs regulate atherosclerotic progression from the earliest induction of inflammation to plaques formation. In particular, we focused on therapeutic strategies based on CAMs inhibitors that prevent leukocyte from migrating to endothelium, including high-affinity antibodies and antagonists, nonspecific traditional medicinal formulas and lipid lowering drugs. The CAMs-based drug delivery nanosystem and the available data on the more reasonable and effective clinical application of CAMs inhibitors have been emphasized, raising hope for further progress in the field of AS therapy.
Collapse
Affiliation(s)
- Mengdie Yin
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Chao Li
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jiali Jiang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Jingqing Le
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Bangyue Luo
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Fang Yang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Yifan Fang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Mingyue Yang
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Zhenhua Deng
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China
| | - Wenxin Ni
- Ocean College, Minjiang University, Fuzhou 350108, China
| | - Jingwei Shao
- Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry, Fuzhou University, Fuzhou 350116, China.
| |
Collapse
|
44
|
Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res 2021; 89:1619-1626. [PMID: 33027803 PMCID: PMC8249239 DOI: 10.1038/s41390-020-01177-9] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/18/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Integrins are heterodimeric transmembrane cell adhesion molecules made up of alpha (α) and beta (β) subunits arranged in numerous dimeric pairings. These complexes have varying affinities to extracellular ligands. Integrins regulate cellular growth, proliferation, migration, signaling, and cytokine activation and release and thereby play important roles in cell proliferation and migration, apoptosis, tissue repair, as well as in all processes critical to inflammation, infection, and angiogenesis. This review presents current evidence from human and animal studies on integrin structure and molecular signaling, with particular emphasis on signal transduction in infants. We have included evidence from our own laboratory studies and from an extensive literature search in databases PubMed, EMBASE, Scopus, and the electronic archives of abstracts presented at the annual meetings of the Pediatric Academic Societies. To avoid bias in identification of existing studies, key words were short-listed prior to the actual search both from anecdotal experience and from PubMed's Medical Subject Heading (MeSH) thesaurus. IMPACT: Integrins are a family of ubiquitous αβ heterodimeric receptors that interact with numerous ligands in physiology and disease. Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis. This review summarizes current evidence from human and animal studies on integrin structure and molecular signaling and promising role in diseases of inflammation, infection, and angiogenesis in infants. This review shows that integrin receptors and ligands are novel therapeutic targets of clinical interest and hold promise as novel therapeutic targets in the management of several neonatal diseases.
Collapse
Affiliation(s)
- Olachi J. Mezu-Ndubuisi
- grid.14003.360000 0001 2167 3675Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Akhil Maheshwari
- grid.21107.350000 0001 2171 9311Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
45
|
Kim HN, Ruan Y, Ogana H, Kim YM. Cadherins, Selectins, and Integrins in CAM-DR in Leukemia. Front Oncol 2020; 10:592733. [PMID: 33425742 PMCID: PMC7793796 DOI: 10.3389/fonc.2020.592733] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/22/2020] [Indexed: 12/12/2022] Open
Abstract
The interaction between leukemia cells and the bone microenvironment is known to provide drug resistance in leukemia cells. This phenomenon, called cell adhesion-mediated drug resistance (CAM-DR), has been demonstrated in many subsets of leukemia including B- and T-acute lymphoblastic leukemia (B- and T-ALL) and acute myeloid leukemia (AML). Cell adhesion molecules (CAMs) are surface molecules that allow cell-cell or cell-extracellular matrix (ECM) adhesion. CAMs not only recognize ligands for binding but also initiate the intracellular signaling pathways that are associated with cell proliferation, survival, and drug resistance upon binding to their ligands. Cadherins, selectins, and integrins are well-known cell adhesion molecules that allow binding to neighboring cells, ECM proteins, and soluble factors. The expression of cadherin, selectin, and integrin correlates with the increased drug resistance of leukemia cells. This paper will review the role of cadherins, selectins, and integrins in CAM-DR and the results of clinical trials targeting these molecules.
Collapse
Affiliation(s)
- Hye Na Kim
- Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Cancer and Blood Disease Institute, Los Angeles, CA, United States
| | - Yongsheng Ruan
- Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Cancer and Blood Disease Institute, Los Angeles, CA, United States.,Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Heather Ogana
- Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Cancer and Blood Disease Institute, Los Angeles, CA, United States
| | - Yong-Mi Kim
- Children's Hospital Los Angeles, Keck School of Medicine of University of Southern California, Cancer and Blood Disease Institute, Los Angeles, CA, United States
| |
Collapse
|
46
|
Henning Stumpf B, Ambriović-Ristov A, Radenovic A, Smith AS. Recent Advances and Prospects in the Research of Nascent Adhesions. Front Physiol 2020; 11:574371. [PMID: 33343382 PMCID: PMC7746844 DOI: 10.3389/fphys.2020.574371] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 11/09/2020] [Indexed: 01/08/2023] Open
Abstract
Nascent adhesions are submicron transient structures promoting the early adhesion of cells to the extracellular matrix. Nascent adhesions typically consist of several tens of integrins, and serve as platforms for the recruitment and activation of proteins to build mature focal adhesions. They are also associated with early stage signaling and the mechanoresponse. Despite their crucial role in sampling the local extracellular matrix, very little is known about the mechanism of their formation. Consequently, there is a strong scientific activity focused on elucidating the physical and biochemical foundation of their development and function. Precisely the results of this effort will be summarized in this article.
Collapse
Affiliation(s)
- Bernd Henning Stumpf
- PULS Group, Institute for Theoretical Physics, Interdisciplinary Center for Nanostructured Films, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreja Ambriović-Ristov
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Aleksandra Radenovic
- Laboratory of Nanoscale Biology, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Ana-Sunčana Smith
- PULS Group, Institute for Theoretical Physics, Interdisciplinary Center for Nanostructured Films, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Group for Computational Life Sciences, Division of Physical Chemistry, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
47
|
Grimm TM, Dierdorf NI, Betz K, Paone C, Hauck CR. PPM1F controls integrin activity via a conserved phospho-switch. J Cell Biol 2020; 219:211512. [PMID: 33119040 PMCID: PMC7604772 DOI: 10.1083/jcb.202001057] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 07/20/2020] [Accepted: 09/11/2020] [Indexed: 01/04/2023] Open
Abstract
Control of integrin activity is vital during development and tissue homeostasis, while derailment of integrin function contributes to pathophysiological processes. Phosphorylation of a conserved threonine motif (T788/T789) in the integrin β cytoplasmic domain increases integrin activity. Here, we report that T788/T789 functions as a phospho-switch, which determines the association with either talin and kindlin-2, the major integrin activators, or filaminA, an integrin activity suppressor. A genetic screen identifies the phosphatase PPM1F as the critical enzyme, which selectively and directly dephosphorylates the T788/T789 motif. PPM1F-deficient cell lines show constitutive integrin phosphorylation, exaggerated talin binding, increased integrin activity, and enhanced cell adhesion. These gain-of-function phenotypes are reverted by reexpression of active PPM1F, but not a phosphatase-dead mutant. Disruption of the ppm1f gene in mice results in early embryonic death at day E10.5. Together, PPM1F controls the T788/T789 phospho-switch in the integrin β1 cytoplasmic tail and constitutes a novel target to modulate integrin activity.
Collapse
Affiliation(s)
- Tanja M. Grimm
- Lehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, Konstanz, Germany,Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany
| | - Nina I. Dierdorf
- Lehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, Konstanz, Germany,Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany
| | - Karin Betz
- Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany,Lehrstuhl Zelluläre Chemie, Fachbereich Chemie, Universität Konstanz, Konstanz, Germany
| | - Christoph Paone
- Lehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, Konstanz, Germany,Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany
| | - Christof R. Hauck
- Lehrstuhl Zellbiologie, Fachbereich Biologie, Universität Konstanz, Konstanz, Germany,Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz, Germany,Correspondence to Christof R. Hauck:
| |
Collapse
|
48
|
Park EJ, Myint PK, Ito A, Appiah MG, Darkwah S, Kawamoto E, Shimaoka M. Integrin-Ligand Interactions in Inflammation, Cancer, and Metabolic Disease: Insights Into the Multifaceted Roles of an Emerging Ligand Irisin. Front Cell Dev Biol 2020; 8:588066. [PMID: 33195249 PMCID: PMC7649757 DOI: 10.3389/fcell.2020.588066] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/05/2020] [Indexed: 01/10/2023] Open
Abstract
Integrins are transmembrane proteins that mediate cellular adhesion and migration to neighboring cells or the extracellular matrix, which is essential for cells to undertake diverse physiological and pathological pathways. For integrin activation and ligand binding, bidirectional signaling across the cell membrane is needed. Integrins aberrantly activated under pathologic conditions facilitate cellular infiltration into tissues, thereby causing inflammatory or tumorigenic progressions. Thus, integrins have emerged to the forefront as promising targets for developing therapeutics to treat autoimmune and cancer diseases. In contrast, it remains a fact that integrin-ligand interactions are beneficial for improving the health status of different tissues. Among these ligands, irisin, a myokine produced mainly by skeletal muscles in an exercise-dependent manner, has been shown to bind to integrin αVβ5, alleviating symptoms under unfavorable conditions. These findings may provide insights into some of the underlying mechanisms by which exercise improves quality of life. This review will discuss the current understanding of integrin-ligand interactions in both health and disease. Likewise, we not only explain how diverse ligands play different roles in mediating cellular functions under both conditions via their interactions with integrins, but also specifically highlight the potential roles of the emerging ligand irisin in inflammation, cancer, and metabolic disease.
Collapse
Affiliation(s)
- Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Phyoe Kyawe Myint
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Atsushi Ito
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Thoracic and Cardiovascular Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Michael G Appiah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Samuel Darkwah
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Emergency and Disaster Medicine, Mie University Graduate School of Medicine, Tsu, Japan
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
49
|
Bhatwadekar AD, Kansara V, Luo Q, Ciulla T. Anti-integrin therapy for retinovascular diseases. Expert Opin Investig Drugs 2020; 29:935-945. [PMID: 32657172 DOI: 10.1080/13543784.2020.1795639] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Integrins are a family of multi-functional cell-adhesion molecules, heterodimeric receptors that connect extracellular matrix (ECM) to actin cytoskeleton in the cell cortex, thus regulating cellular adhesion, migration, proliferation, invasion, survival, and apoptosis. Consequently, integrins play a role in inflammation, angiogenesis and fibrosis. AREAS COVERED This review examines individual anti-integrin agents in terms of their chemical nature, route of administration, and anti-integrin action. It also provides a summary of preclinical and clinical studies. Current clinical candidates include risuteganib, THR-687, and SF-0166, which have shown promise in treating diabetic macular edema (DME) and/or age-related macular degeneration (AMD) in early clinical studies. Preclinical candidates include SB-267268, AXT-107, JNJ-26076713, Cilengitide and Lebecetin, which exhibit a decrease in retinal permeability, angiogenesis and/or choroidal neovascularization (CNV). EXPERT OPINION Anti-integrin therapies show potential in treating retinal diseases. Anti-integrin agents tackle the multi-factorial nature of diabetic retinopathy (DR) and AMD and show promise as injectable and topical agents in preclinical and early clinical studies. Integrin inhibition has potential to serve as primary therapy, adjunctive therapy to anti-vascular endothelial growth factor agents, or secondary therapy in refractory cases.
Collapse
Affiliation(s)
- Ashay D Bhatwadekar
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University , Indianapolis, IN, USA
| | - Viral Kansara
- Preclinical Development Department, Clearside Biomedical, Inc , Alpharetta, GA, USA
| | - Qianyi Luo
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University , Indianapolis, IN, USA
| | - Thomas Ciulla
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University , Indianapolis, IN, USA.,Preclinical Development Department, Clearside Biomedical, Inc , Alpharetta, GA, USA.,Retina Department, Midwest Eye Institute , Indianapolis, IN, USA
| |
Collapse
|
50
|
Kang S, Lee S, Park S. iRGD Peptide as a Tumor-Penetrating Enhancer for Tumor-Targeted Drug Delivery. Polymers (Basel) 2020; 12:E1906. [PMID: 32847045 PMCID: PMC7563641 DOI: 10.3390/polym12091906] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/20/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023] Open
Abstract
The unique structure and physiology of a tumor microenvironment impede intra-tumoral penetration of chemotherapeutic agents. A novel iRGD peptide that exploits the tumor microenvironment can activate integrin-dependent binding to tumor vasculatures and neuropilin-1 (NRP-1)-dependent transport to tumor tissues. Recent studies have focused on its dual-targeting ability to achieve enhanced penetration of chemotherapeutics for the efficient eradication of cancer cells. Both the covalent conjugation and the co-administration of iRGD with chemotherapeutic agents and engineered delivery vehicles have been explored. Interestingly, the iRGD-mediated drug delivery also enhances penetration through the blood-brain barrier (BBB). Recent studies have shown its synergistic effect with BBB disruptive techniques. The efficacy of immunotherapy involving immune checkpoint blockades has also been amplified by using iRGD as a targeting moiety. In this review, we presented the recent advances in iRGD technology, focusing on cancer treatment modalities, including the current clinical trials using iRGD. The iRGD-mediated nano-carrier system could serve as a promising strategy in drug delivery to the deeper tumor regions, and be combined with various therapeutic interventions due to its novel targeting ability.
Collapse
Affiliation(s)
| | | | - Soyeun Park
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldaero, Dalseo-gu, Daegu 42601, Korea; (S.K.); (S.L.)
| |
Collapse
|