1
|
Ding X, Liang Y, Zhou S, Wu Y, Sałata P, Mikolajczk-Martinez A, Khosrawipour V, Zhang Z. Targeting tumor extracellular matrix with nanoparticles to circumvent therapeutic resistance. J Control Release 2025:113786. [PMID: 40306575 DOI: 10.1016/j.jconrel.2025.113786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
Each stage of tumor development is intrinsically linked to the tumor microenvironment (TME), wherein the extracellular matrix (ECM) serves as a vital and abundant component in tumor tissues. The ECM is a non-cellular, three-dimensional macromolecular network scaffold that provides structural support to cells, stores bioactive molecules, and mediates signaling pathways through specific binding to cell surface receptors. Moreover, the ECM in tumor tissues plays a crucial role in impeding drug diffusion and resisting apoptosis induced by conventional anti-cancer therapies that primarily target cancer cells. Therefore, directing attentions towards the tumor ECM can facilitate the identification of novel targets and the development of new therapies. This review aims to summarize the composition, structure, remodeling, and function of tumor ECM, its association with drug resistance, and current targeting strategies, with a specific emphasis on nanoparticles (NPs).
Collapse
Affiliation(s)
- Xinyue Ding
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yiyu Liang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Siyuan Zhou
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Yao Wu
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China
| | - Patricia Sałata
- Wrocław University of Environmental and Life Sciences, Wrocław, Poland
| | | | | | - Zhiwen Zhang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, China.
| |
Collapse
|
2
|
Miao MZ, Lee JS, Yamada KM, Loeser RF. Integrin signalling in joint development, homeostasis and osteoarthritis. Nat Rev Rheumatol 2024; 20:492-509. [PMID: 39014254 PMCID: PMC11886400 DOI: 10.1038/s41584-024-01130-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2024] [Indexed: 07/18/2024]
Abstract
Integrins are key regulators of cell-matrix interactions during joint development and joint tissue homeostasis, as well as in the development of osteoarthritis (OA). The signalling cascades initiated by the interactions of integrins with a complex network of extracellular matrix (ECM) components and intracellular adaptor proteins orchestrate cellular responses necessary for maintaining joint tissue integrity. Dysregulated integrin signalling, triggered by matrix degradation products such as matrikines, disrupts this delicate balance, tipping the scales towards an environment conducive to OA pathogenesis. The interplay between integrin signalling and growth factor pathways further underscores the multifaceted nature of OA. Moreover, emerging insights into the role of endocytic trafficking in regulating integrin signalling add a new layer of complexity to the understanding of OA development. To harness the therapeutic potential of targeting integrins for mitigation of OA, comprehensive understanding of their molecular mechanisms across joint tissues is imperative. Ultimately, deciphering the complexities of integrin signalling will advance the ability to treat OA and alleviate its global burden.
Collapse
Affiliation(s)
- Michael Z Miao
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Division of Rheumatology, Allergy, and Immunology and the Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Janice S Lee
- Craniofacial Anomalies and Regeneration Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
- Office of the Clinical Director, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth M Yamada
- Cell Biology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA.
| | - Richard F Loeser
- Division of Rheumatology, Allergy, and Immunology and the Thurston Arthritis Research Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
3
|
Zhang QD, Duan QY, Tu J, Wu FG. Thrombin and Thrombin-Incorporated Biomaterials for Disease Treatments. Adv Healthc Mater 2024; 13:e2302209. [PMID: 37897228 DOI: 10.1002/adhm.202302209] [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: 07/12/2023] [Revised: 10/20/2023] [Indexed: 10/29/2023]
Abstract
Thrombin, a coagulation-inducing protease, has long been used in the hemostatic field. During the past decades, many other therapeutic uses of thrombin have been developed. For instance, burn treatment, pseudoaneurysm therapy, wound management, and tumor vascular infarction (or tumor vasculature blockade therapy) can all utilize the unique and powerful function of thrombin. Based on their therapeutic effects, many thrombin-associated products have been certificated by the Food and Drug Administration, including bovine thrombin, human thrombin, recombinant thrombin, fibrin glue, etc. Besides, several thrombin-based drugs are currently undergoing clinical trials. In this article, the therapeutic uses of thrombin (from the initial hemostasis to the latest cancer therapy), the commercially available drugs associated with thrombin, and the pros and cons of thrombin-based therapeutics (e.g., adverse immune responses related to bovine thrombin, thromboinflammation, and vasculogenic "rebounds") are summarized. Further, the current challenges and possible future research directions of thrombin-incorporated biomaterials and therapies are discussed. It is hoped that this review may provide a valuable reference for researchers in this field and help them to design safer and more effective thrombin-based drugs for fighting against various intractable diseases.
Collapse
Affiliation(s)
- Qiong-Dan Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, Jiangsu, 211189, P. R. China
| | - Qiu-Yi Duan
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, Jiangsu, 211189, P. R. China
| | - Jing Tu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, Jiangsu, 211189, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, 2 Southeast University Road, Nanjing, Jiangsu, 211189, P. R. China
| |
Collapse
|
4
|
Fu Q, Gao H, Liu K, Su J, Zhang J, Guo X, Yang F. Identification of circRNA-miRNA-mRNA Network Regulated by Hsp90 in Human Melanoma A375 Cells. Comb Chem High Throughput Screen 2024; 27:307-316. [PMID: 37303182 DOI: 10.2174/1386207326666230609145247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/15/2023] [Accepted: 05/25/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Melanoma is the deadliest form of skin cancer. Heat shock protein 90 (Hsp90) is highly expressed in human melanoma. Hsp90 inhibitors can suppress the growth of human melanoma A375 cells; however, the underlying mechanism remains unclear. METHODS A375 cells were treated with SNX-2112, an Hsp90 inhibitor, for 48 h, and wholetranscriptome sequencing was performed. RESULTS A total of 2,528 differentially expressed genes were identified, including 895 upregulated and 1,633 downregulated genes. Pathway enrichment analyses of differentially expressed mRNAs identified the extracellular matrix (ECM)-receptor interaction pathway as the most significantly enriched pathway. The ECM receptor family mainly comprises integrins (ITGs) and collagens (COLs), wherein ITGs function as the major cell receptors for COLs. 19 upregulated miRNAs were found to interact with 6 downregulated ITG genes and 8 upregulated miRNAs were found to interact with 3 downregulated COL genes. 9 differentially expressed circRNAs in SNX-2112- treated A375 cells were identified as targets of the ITG- and COL-related miRNAs. Based on the differentially expressed circRNAs, miRNAs, and mRNAs, ITGs- and COL-based circRNAmiRNA- mRNA regulatory networks were mapped, revealing a novel regulatory mechanism of Hsp90-regulated melanoma. CONCLUSION Targeting the ITG-COL network is a promising approach to the treatment of melanoma.
Collapse
Affiliation(s)
- Qiang Fu
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Hengyuan Gao
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Kaisheng Liu
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Juan Su
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jianglin Zhang
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Candidate Branch of the National Clinical Research Center for Skin Diseases, Shenzhen, 518020, Guangdong, China
| | - Xiaojing Guo
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Fang Yang
- Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Candidate Branch of the National Clinical Research Center for Skin Diseases, Shenzhen, 518020, Guangdong, China
| |
Collapse
|
5
|
Zhu S, Jaffiol R, Crunteanu A, Vézy C, Chan ST, Yuan W, Ho HP, Zeng S. Label-free biosensing with singular-phase-enhanced lateral position shift based on atomically thin plasmonic nanomaterials. LIGHT, SCIENCE & APPLICATIONS 2024; 13:2. [PMID: 38161210 PMCID: PMC10757996 DOI: 10.1038/s41377-023-01345-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 11/18/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024]
Abstract
Rapid plasmonic biosensing has attracted wide attention in early disease diagnosis and molecular biology research. However, it was still challenging for conventional angle-interrogating plasmonic sensors to obtain higher sensitivity without secondary amplifying labels such as plasmonic nanoparticles. To address this issue, we developed a plasmonic biosensor based on the enhanced lateral position shift by phase singularity. Such singularity presents as a sudden phase retardation at the dark point of reflection from resonating plasmonic substrate, leading to a giant position shift on reflected beam. Herein, for the first time, the atomically thin layer of Ge2Sb2Te5 (GST) on silver nanofilm was demonstrated as a novel phase-response-enhancing plasmonic material. The GST layer was not only precisely engineered to singularize phase change but also served as a protective layer for active silver nanofilm. This new configuration has achieved a record-breaking largest position shift of 439.3 μm measured in calibration experiments with an ultra-high sensitivity of 1.72 × 108 nm RIU-1 (refractive index unit). The detection limit was determined to be 6.97 × 10-7 RIU with a 0.12 μm position resolution. Besides, a large figure of merit (FOM) of 4.54 × 1011 μm (RIU∙°)-1 was evaluated for such position shift interrogation, enabling the labelfree detection of trace amounts of biomolecules. In targeted biosensing experiments, the optimized sensor has successfully detected small cytokine biomarkers (TNF-α and IL-6) with the lowest concentration of 1 × 10-16 M. These two molecules are the key proinflammatory cancer markers in clinical diagnosis, which cannot be directly screened by current clinical techniques. To further validate the selectivity of our sensing systems, we also measured the affinity of integrin binding to arginylglycylaspartic acid (RGD) peptide (a key protein interaction in cell adhesion) with different Mn2+ ion concentrations, ranging from 1 nM to 1 mM.
Collapse
Affiliation(s)
- Shaodi Zhu
- Light, Nanomaterials & Nanotechnologies (L2n), CNRS-EMR 7004, University of Technology of Troyes, 10000, Troyes, France
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Rodolphe Jaffiol
- Light, Nanomaterials & Nanotechnologies (L2n), CNRS-EMR 7004, University of Technology of Troyes, 10000, Troyes, France
| | - Aurelian Crunteanu
- XLIM Research Institute, UMR 7252 CNRS/University of Limoges, 123, Avenue Albert Thomas, Limoges, France
| | - Cyrille Vézy
- Light, Nanomaterials & Nanotechnologies (L2n), CNRS-EMR 7004, University of Technology of Troyes, 10000, Troyes, France
| | - Sik-To Chan
- Light, Nanomaterials & Nanotechnologies (L2n), CNRS-EMR 7004, University of Technology of Troyes, 10000, Troyes, France
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Wu Yuan
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Ho-Pui Ho
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China.
| | - Shuwen Zeng
- Light, Nanomaterials & Nanotechnologies (L2n), CNRS-EMR 7004, University of Technology of Troyes, 10000, Troyes, France.
| |
Collapse
|
6
|
Herrera-Quiterio GA, Encarnación-Guevara S. The transmembrane proteins (TMEM) and their role in cell proliferation, migration, invasion, and epithelial-mesenchymal transition in cancer. Front Oncol 2023; 13:1244740. [PMID: 37936608 PMCID: PMC10627164 DOI: 10.3389/fonc.2023.1244740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/11/2023] [Indexed: 11/09/2023] Open
Abstract
Transmembrane proteins (TMEM) are located in the different biological membranes of the cell and have at least one passage through these cellular compartments. TMEM proteins carry out a wide variety of functions necessary to maintain cell homeostasis TMEM165 participates in glycosylation protein, TMEM88 in the development of cardiomyocytes, TMEM45A in epidermal keratinization, and TMEM74 regulating autophagy. However, for many TMEM proteins, their physiological function remains unknown. The role of these proteins is being recently investigated in cancer since transcriptomic and proteomic studies have revealed that exits differential expression of TMEM proteins in different neoplasms concerning cancer-free tissues. Among the cellular processes in which TMEM proteins have been involved in cancer are the promotion or suppression of cell proliferation, epithelial-mesenchymal transition, invasion, migration, intravasation/extravasation, metastasis, modulation of the immune response, and response to antineoplastic drugs. Inclusive data suggests that the participation of TMEM proteins in these cellular events could be carried out through involvement in different cell signaling pathways. However, the exact mechanisms not clear. This review shows a description of the involvement of TMEM proteins that promote or decrease cell proliferation, migration, and invasion in cancer cells, describes those TMEM proteins for which both a tumor suppressor and a tumor promoter role have been identified, depending on the type of cancer in which the protein is expressed. As well as some TMEM proteins involved in chemoresistance. A better characterization of these proteins is required to improve the understanding of the tumors in which their expression and function are altered; in addition to improving the understanding of the role of these proteins in cancer will show those TMEM proteins be potential candidates as biomarkers of response to chemotherapy or prognostic biomarkers or as potential therapeutic targets in cancer.
Collapse
Affiliation(s)
| | - Sergio Encarnación-Guevara
- Laboratorio de Proteómica, Centro de Ciencias Genómicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos, Mexico
| |
Collapse
|
7
|
Campbell S, Mendoza MC, Rammohan A, McKenzie ME, Bidone TC. Computational model of integrin adhesion elongation under an actin fiber. PLoS Comput Biol 2023; 19:e1011237. [PMID: 37410718 PMCID: PMC10325090 DOI: 10.1371/journal.pcbi.1011237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 06/02/2023] [Indexed: 07/08/2023] Open
Abstract
Cells create physical connections with the extracellular environment through adhesions. Nascent adhesions form at the leading edge of migrating cells and either undergo cycles of disassembly and reassembly, or elongate and stabilize at the end of actin fibers. How adhesions assemble has been addressed in several studies, but the exact role of actin fibers in the elongation and stabilization of nascent adhesions remains largely elusive. To address this question, here we extended our computational model of adhesion assembly by incorporating an actin fiber that locally promotes integrin activation. The model revealed that an actin fiber promotes adhesion stabilization and elongation. Actomyosin contractility from the fiber also promotes adhesion stabilization and elongation, by strengthening integrin-ligand interactions, but only up to a force threshold. Above this force threshold, most integrin-ligand bonds fail, and the adhesion disassembles. In the absence of contraction, actin fibers still support adhesions stabilization. Collectively, our results provide a picture in which myosin activity is dispensable for adhesion stabilization and elongation under an actin fiber, offering a framework for interpreting several previous experimental observations.
Collapse
Affiliation(s)
- Samuel Campbell
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
| | - Michelle C. Mendoza
- Department of Oncological Sciences, University of Utah, Salt Lake City, Utah, United States of America
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah, United States of America
| | - Aravind Rammohan
- Corning Life Sciences, Tewksburry, Massachusetts, United States of America
| | - Matthew E. McKenzie
- Corning Research and Development Corporation, Corning, New York, United States of America
| | - Tamara C. Bidone
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, United States of America
- Scientific Computing and Imaging Institute, University of Utah, Salt Lake City, Utah, United States of America
| |
Collapse
|
8
|
Jing H, Wu X, Xiang M, Wang C, Novakovic VA, Shi J. Microparticle Phosphatidylserine Mediates Coagulation: Involvement in Tumor Progression and Metastasis. Cancers (Basel) 2023; 15:cancers15071957. [PMID: 37046617 PMCID: PMC10093313 DOI: 10.3390/cancers15071957] [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: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Tumor progression and cancer metastasis has been linked to the release of microparticles (MPs), which are shed upon cell activation or apoptosis and display parental cell antigens, phospholipids such as phosphatidylserine (PS), and nucleic acids on their external surfaces. In this review, we highlight the biogenesis of MPs as well as the pathophysiological processes of PS externalization and its involvement in coagulation activation. We review the available evidence, suggesting that coagulation factors (mainly tissue factor, thrombin, and fibrin) assist in multiple steps of tumor dissemination, including epithelial-mesenchymal transition, extracellular matrix remodeling, immune escape, and tumor angiogenesis to support the formation of the pre-metastatic niche. Platelets are not just bystander cells in circulation but are functional players in primary tumor growth and metastasis. Tumor-induced platelet aggregation protects circulating tumor cells (CTCs) from the blood flow shear forces and immune cell attack while also promoting the binding of CTCs to endothelial cells and extravasation, which activates tumor invasion and sustains metastasis. Finally, in terms of therapy, lactadherin can inhibit coagulation by competing effectively with coagulation factors for PS binding sites and may similarly delay tumor progression. Furthermore, we also investigate the therapeutic potential of coagulation factor inhibitors within the context of cancer treatment. The development of multiple therapies targeting platelet activation and platelet-tumor cell interactions may not only reduce the lethal consequences of thrombosis but also impede tumor growth and spread.
Collapse
Affiliation(s)
- Haijiao Jing
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Xiaoming Wu
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Mengqi Xiang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Chengyue Wang
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
| | - Valerie A Novakovic
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02132, USA
| | - Jialan Shi
- Department of Hematology, The First Hospital, Harbin Medical University, Harbin 150001, China
- Department of Research, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02132, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02132, USA
| |
Collapse
|
9
|
Trzaskowska M, Vivcharenko V, Przekora A. The Impact of Hydroxyapatite Sintering Temperature on Its Microstructural, Mechanical, and Biological Properties. Int J Mol Sci 2023; 24:ijms24065083. [PMID: 36982158 PMCID: PMC10049015 DOI: 10.3390/ijms24065083] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/22/2023] [Accepted: 03/05/2023] [Indexed: 03/09/2023] Open
Abstract
Hydroxyapatite (HA), the principal mineral of bone tissue, can be fabricated as an artificial calcium phosphate (CaP) ceramic and potentially used as bioceramic material for bone defect treatment. Nevertheless, the production method (including the applied sintering temperature) of synthetic hydroxyapatite directly affects its basic properties, such as its microstructure, mechanical parameters, bioabsorbability, and osteoconductivity, and in turn influences its biomedical potential as an implantable biomaterial. The wide application of HA in regenerative medicine makes it necessary to explain the validity of the selection of the sintering temperature. The main emphasis of this article is on the description and summarization of the key features of HA depending on the applied sintering temperature during the synthesis process. The review is mainly focused on the dependence between the HA sintering temperature and its microstructural features, mechanical properties, biodegradability/bioabsorbability, bioactivity, and biocompatibility.
Collapse
|
10
|
Russo V, Fabiani D. Put out the fire: The pleiotropic anti-inflammatory action of non-vitamin K oral anticoagulants. Pharmacol Res 2022; 182:106335. [PMID: 35781059 DOI: 10.1016/j.phrs.2022.106335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/15/2022] [Accepted: 06/28/2022] [Indexed: 11/28/2022]
Abstract
Non-vitamin K antagonist oral anticoagulants (NOACs) should be the preferred anticoagulant strategy for preventing ischemic stroke in patients with atrial fibrillation (AF) at increased thromboembolic risk and for treating deep venous thromboembolism (DVT) in the general population. Beyond their inhibiting action on the activated factor X (FXa) or thrombin (FIIa), NOACs showed some pleiotropic anti-inflammatory effects. The present review aimed to describe the role of FXa and FIIa in the inflammation pathway and the potential anti-inflammatory effects of NOACs.
Collapse
Affiliation(s)
- Vincenzo Russo
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania "Luigi Vanvitelli" - Monaldi Hospital, Naples, Italy.
| | - Dario Fabiani
- Cardiology Unit, Department of Medical Translational Sciences, University of Campania "Luigi Vanvitelli" - Monaldi Hospital, Naples, Italy
| |
Collapse
|
11
|
Mishchenko EL, Mishchenko AM, Ivanisenko VA. Mechanosensitive molecular interactions in atherogenic regions of the arteries: development of atherosclerosis. Vavilovskii Zhurnal Genet Selektsii 2021; 25:552-561. [PMID: 34595377 PMCID: PMC8453358 DOI: 10.18699/vj21.062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/26/2021] [Accepted: 04/08/2021] [Indexed: 11/30/2022] Open
Abstract
A terrible disease of the cardiovascular system, atherosclerosis, develops in the areas of bends and
branches of arteries, where the direction and modulus of the blood flow velocity vector change, and consequently
so does the mechanical effect on endothelial cells in contact with the blood flow. The review focuses on topical
research studies on the development of atherosclerosis – mechanobiochemical events that transform the proatherogenic
mechanical stimulus of blood flow – low and low/oscillatory arterial wall shear stress in the chains of biochemical
reactions in endothelial cells, leading to the expression of specific proteins that cause the progression
of the pathological process. The stages of atherogenesis, systemic risk factors for atherogenesis and its important
hemodynamic factor, low and low/oscillatory wall shear stress exerted by blood flow on the endothelial cells lining
the arterial walls, have been described. The interactions of cell adhesion molecules responsible for the development
of atherosclerosis under low and low/oscillating shear stress conditions have been demonstrated. The activation
of the regulator of the expression of cell adhesion molecules, the transcription factor NF-κB, and the factors
regulating its activation under these conditions have been described. Mechanosensitive signaling pathways leading
to the expression of NF-κB in endothelial cells have been described. Studies of the mechanobiochemical signaling
pathways and interactions involved in the progression of atherosclerosis provide valuable information for the
development of approaches that delay or block the development of this disease.
Key words: atherogenesis; shear stress; transcription factor NF-κB; RelA expression; mechanosensitive receptors;
cell adhesion molecules; signaling pathways; mechanotransduction.
Collapse
Affiliation(s)
- E L Mishchenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | | - V A Ivanisenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
12
|
Wang P, Yao L, Luo M, Zhou W, Jin X, Xu Z, Yan S, Li Y, Xu C, Cheng R, Huang Y, Lin X, Ma K, Cao H, Liu H, Xue G, Han F, Nie H, Jiang Q. Single-cell transcriptome and TCR profiling reveal activated and expanded T cell populations in Parkinson's disease. Cell Discov 2021; 7:52. [PMID: 34282123 PMCID: PMC8289849 DOI: 10.1038/s41421-021-00280-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023] Open
Abstract
Given the chronic inflammatory nature of Parkinson's disease (PD), T cell immunity may be important for disease onset. Here, we performed single-cell transcriptome and TCR sequencing, and conducted integrative analyses to decode composition, function and lineage relationship of T cells in the blood and cerebrospinal fluid of PD. Combined expression and TCR-based lineage tracking, we discovered a large population of CD8+ T cells showing continuous progression from central memory to terminal effector T cells in PD patients. Additionally, we identified a group of cytotoxic CD4+ T cells (CD4 CTLs) remarkably expanded in PD patients, which derived from Th1 cells by TCR-based fate decision. Finally, we screened putative TCR-antigen pairs that existed in both blood and cerebrospinal fluid of PD patients to provide potential evidence for peripheral T cells to participate in neuronal degeneration. Our study provides valuable insights and rich resources for understanding the adaptive immune response in PD.
Collapse
Affiliation(s)
- Pingping Wang
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Lifen Yao
- grid.412596.d0000 0004 1797 9737Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang China
| | - Meng Luo
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Wenyang Zhou
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Xiyun Jin
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Zhaochun Xu
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Shi Yan
- grid.412596.d0000 0004 1797 9737Department of Neurology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang China
| | - Yiqun Li
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Chang Xu
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Rui Cheng
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Yan Huang
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Xiaoyu Lin
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Kexin Ma
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Huimin Cao
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Hongxin Liu
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Guangfu Xue
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Fang Han
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Huan Nie
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China
| | - Qinghua Jiang
- grid.19373.3f0000 0001 0193 3564School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Big Data (Harbin Institute of Technology), Ministry of Education, Harbin, Heilongjiang China
| |
Collapse
|
13
|
Ide K, Takahashi S, Sakai K, Taga Y, Ueno T, Dickens D, Jenkins R, Falciani F, Sasaki T, Ooi K, Kawashiri S, Mizuno K, Hattori S, Sakai T. The dipeptide prolyl-hydroxyproline promotes cellular homeostasis and lamellipodia-driven motility via active β1-integrin in adult tendon cells. J Biol Chem 2021; 297:100819. [PMID: 34029590 PMCID: PMC8239475 DOI: 10.1016/j.jbc.2021.100819] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 05/08/2021] [Accepted: 05/20/2021] [Indexed: 12/14/2022] Open
Abstract
Collagen-derived hydroxyproline (Hyp)-containing peptides have a variety of biological effects on cells. These bioactive collagen peptides are locally generated by the degradation of endogenous collagen in response to injury. However, no comprehensive study has yet explored the functional links between Hyp-containing peptides and cellular behavior. Here, we show that the dipeptide prolyl-4-hydroxyproline (Pro-Hyp) exhibits pronounced effects on mouse tendon cells. Pro-Hyp promotes differentiation/maturation of tendon cells with modulation of lineage-specific factors and induces significant chemotactic activity in vitro. In addition, Pro-Hyp has profound effects on cell proliferation, with significantly upregulated extracellular signal-regulated kinase phosphorylation and extracellular matrix production and increased type I collagen network organization. Using proteomics, we have predicted molecular transport, cellular assembly and organization, and cellular movement as potential linked-network pathways that could be altered in response to Pro-Hyp. Mechanistically, cells treated with Pro-Hyp demonstrate increased directional persistence and significantly increased directed motility and migration velocity. They are accompanied by elongated lamellipodial protrusions with increased levels of active β1-integrin-containing focal contacts, as well as reorganization of thicker peripheral F-actin fibrils. Pro-Hyp-mediated chemotactic activity is significantly reduced (p < 0.001) in cells treated with the mitogen-activated protein kinase kinase 1/2 inhibitor PD98059 or the α5β1-integrin antagonist ATN-161. Furthermore, ATN-161 significantly inhibits uptake of Pro-Hyp into adult tenocytes. Thus, our findings document the molecular basis of the functional benefits of the Pro-Hyp dipeptide in cellular behavior. These dynamic properties of collagen-derived Pro-Hyp dipeptide could lead the way to its application in translational medicine.
Collapse
Affiliation(s)
- Kentaro Ide
- Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Sanai Takahashi
- Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Keiko Sakai
- Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Yuki Taga
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Tomonori Ueno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - David Dickens
- Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Rosalind Jenkins
- Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Francesco Falciani
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Takako Sasaki
- Department of Biochemistry, Faculty of Medicine, Oita University, Oita, Japan
| | - Kazuhiro Ooi
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa, Japan
| | - Shuichi Kawashiri
- Department of Oral and Maxillofacial Surgery, Kanazawa University Graduate School of Medical Science, Kanazawa, Ishikawa, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Takao Sakai
- Department of Pharmacology and Therapeutics, MRC Centre for Drug Safety Science, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
14
|
Reina-Torres E, De Ieso ML, Pasquale LR, Madekurozwa M, van Batenburg-Sherwood J, Overby DR, Stamer WD. The vital role for nitric oxide in intraocular pressure homeostasis. Prog Retin Eye Res 2020; 83:100922. [PMID: 33253900 DOI: 10.1016/j.preteyeres.2020.100922] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/13/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Catalyzed by endothelial nitric oxide (NO) synthase (eNOS) activity, NO is a gaseous signaling molecule maintaining endothelial and cardiovascular homeostasis. Principally, NO regulates the contractility of vascular smooth muscle cells and permeability of endothelial cells in response to either biochemical or biomechanical cues. In the conventional outflow pathway of the eye, the smooth muscle-like trabecular meshwork (TM) cells and Schlemm's canal (SC) endothelium control aqueous humor outflow resistance, and therefore intraocular pressure (IOP). The mechanisms by which outflow resistance is regulated are complicated, but NO appears to be a key player as enhancement or inhibition of NO signaling dramatically affects outflow function; and polymorphisms in NOS3, the gene that encodes eNOS modifies the relation between various environmental exposures and glaucoma. Based upon a comprehensive review of past foundational studies, we present a model whereby NO controls a feedback signaling loop in the conventional outflow pathway that is sensitive to changes in IOP and its oscillations. Thus, upon IOP elevation, the outflow pathway tissues distend, and the SC lumen narrows resulting in increased SC endothelial shear stress and stretch. In response, SC cells upregulate the production of NO, relaxing neighboring TM cells and increasing permeability of SC's inner wall. These IOP-dependent changes in the outflow pathway tissues reduce the resistance to aqueous humor drainage and lower IOP, which, in turn, diminishes the biomechanical signaling on SC. Similar to cardiovascular pathogenesis, dysregulation of the eNOS/NO system leads to dysfunctional outflow regulation and ocular hypertension, eventually resulting in primary open-angle glaucoma.
Collapse
Affiliation(s)
| | | | - Louis R Pasquale
- Eye and Vision Research Institute of New York Eye and Ear Infirmary at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, UK.
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, NC, USA.
| |
Collapse
|
15
|
Cha B, Kim J, Bello A, Lee G, Kim D, Kim BJ, Arai Y, Choi B, Park H, Lee S. Efficient Isolation and Enrichment of Mesenchymal Stem Cells from Human Embryonic Stem Cells by Utilizing the Interaction between Integrin α5 β1 and Fibronectin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001365. [PMID: 32995130 PMCID: PMC7507081 DOI: 10.1002/advs.202001365] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/02/2020] [Indexed: 05/09/2023]
Abstract
Human pluripotent stem cells (hPSCs) are a potent source of clinically relevant mesenchymal stem cells (MSCs) that confer functional and structural benefits in cell therapy and tissue regeneration. Obtaining sufficient numbers of MSCs in a short period of time and enhancing the differentiation potential of MSCs can be offered the potential to improve the regenerative activity of MSCs therapy. In addition, the underlying processes in the isolation and derivation of MSCs from hPSCs are still poorly understood and controlled. To overcome these clinical needs, an efficient and simplified technique on the isolation of MSCs from spontaneously differentiated human embryonic stem cells (hESCs) via integrin α5β1 (fibronectin (FN) receptor)-to-FN interactions (hESC-FN-MSCs) is successfully developed. It is demonstrated that hESC-FN-MSCs exhibit a typical MSC surface phenotype, cellular morphology, with the whole transcriptome similar to conventional adult MSCs; but show higher proliferative capacity, more efficient trilineage differentiation, enhanced cytokine secretion, and attenuated cellular senescence. In addition, the therapeutic potential and regenerative capacity of the isolated hESC-FN-MSCs are confirmed by in vitro and in vivo multilineage differentiation. This novel method will be useful in the generation of abundant amounts of clinically relevant MSCs for stem cell therapeutics and regenerative medicine.
Collapse
Affiliation(s)
- Byung‐Hyun Cha
- Division of Cardio‐Thoracic SurgeryDepartment of SurgeryCollege of MedicineUniversity of ArizonaTucsonAZ85724USA
| | - Jin‐Su Kim
- CellenGene R&D CenterOpen Innovation BuildingSeoul02455Republic of Korea
- Department of Biomedical ScienceCHA UniversityCHA BiocomplexSeongnam‐siGyeonggi‐do13488Republic of Korea
| | - Alvin Bello
- Department of Integrative EngineeringChung‐Ang UniversitySeoul06974Republic of Korea
| | - Geun‐Hui Lee
- Department of Biomedical ScienceCHA UniversityCHA BiocomplexSeongnam‐siGyeonggi‐do13488Republic of Korea
| | - Do‐Hyun Kim
- Department of Medical BiotechnologyDongguk University32 Dongguk‐ro, Ilsandong‐guGoyangGyeonggi10326Republic of Korea
| | - Byoung Ju Kim
- Department of Medical BiotechnologyDongguk University32 Dongguk‐ro, Ilsandong‐guGoyangGyeonggi10326Republic of Korea
| | - Yoshie Arai
- Department of Medical BiotechnologyDongguk University32 Dongguk‐ro, Ilsandong‐guGoyangGyeonggi10326Republic of Korea
| | - Bogyu Choi
- Department of Biomedical ScienceCHA UniversityCHA BiocomplexSeongnam‐siGyeonggi‐do13488Republic of Korea
| | - Hansoo Park
- Department of Integrative EngineeringChung‐Ang UniversitySeoul06974Republic of Korea
| | - Soo‐Hong Lee
- Department of Medical BiotechnologyDongguk University32 Dongguk‐ro, Ilsandong‐guGoyangGyeonggi10326Republic of Korea
| |
Collapse
|
16
|
Yang Y, Sun X, Xu J, Cui C, Safari Yazd H, Pan X, Zhu Y, Chen X, Li X, Li J, Tan W. Circular Bispecific Aptamer-Mediated Artificial Intercellular Recognition for Targeted T Cell Immunotherapy. ACS NANO 2020; 14:9562-9571. [PMID: 32584540 DOI: 10.1021/acsnano.9b09884] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Adoptive T cell immunotherapy, such as chimeric antigen receptor (CAR) T cell therapy, has proven to be highly efficient in the treatment of hematologic malignancies. However, it is challenged by complicated ex vivo engineering, systemic side effects, and low expression of tumor-specific antigen, especially in solid tumors. In this paper, we present a "recognition-then-activation" strategy, which first assists naïve T cells to recognize and adhere to cancer cells and then activates the accumulated T cell in situ to specifically kill cancer cells. In this way, we could unleash the antitumor power of the T cell without complicated and time-consuming cell engineering. To this end, circular bispecific aptamers (cb-aptamers), a class of chemically cyclized aptamers with improved stability and molecular recognition ability which can simultaneously bind to two different types of cells, were first constructed to form artificial intercellular recognition between naïve T cells and tumor cells. After T cell accumulation in the tumor mediated by cb-aptamers, T cells in the tumor site were subsequently activated in situvia commercial CD3/CD28 T cell activator beads to induce tumor-specific killing. Furthermore, by simply choosing different anticancer aptamers, the application of this "recognition-then-activation" strategy can be expanded for targeted treatment of various types of cancer. This may represent a simple T cell immunotherapy that is useful for the treatment of multiple cancers.
Collapse
Affiliation(s)
- Yu Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Xiaoqi Sun
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jun Xu
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Cheng Cui
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Hoda Safari Yazd
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Xiaoshu Pan
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Yujie Zhu
- Institute of Functional Nano & Soft Materials Laboratory (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China
| | - Xigao Chen
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Xiaowei Li
- Department of Chemistry, Department of Physiology and Functional Genomics, Center for Research at Bio/Nano Interface, Health Cancer Center, UF Genetics Institute and McKnight Brain Institute, University of Florida, Gainesville, Florida 32611-7200, United States
| | - Jin Li
- Molecular Science and Biomedicine Laboratory(MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), and Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, and College of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
- Molecular Science and Biomedicine Laboratory(MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha 410082, China
| |
Collapse
|
17
|
Pokhrel S, Triplett KD, Daly SM, Joyner JA, Sharma G, Hathaway HJ, Prossnitz ER, Hall PR. Complement Receptor 3 Contributes to the Sexual Dimorphism in Neutrophil Killing of Staphylococcus aureus. THE JOURNAL OF IMMUNOLOGY 2020; 205:1593-1600. [PMID: 32769122 DOI: 10.4049/jimmunol.2000545] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 07/17/2020] [Indexed: 12/24/2022]
Abstract
We previously reported sex differences in innate susceptibility to Staphylococcus aureus skin infection and that bone marrow neutrophils (BMN) from female mice have an enhanced ability to kill S. aureus ex vivo compared with those of male mice. However, the mechanism(s) driving this sex bias in neutrophil killing have not been reported. Given the role of opsonins such as complement, as well as their receptors, in S. aureus recognition and clearance, we investigated their contribution to the enhanced bactericidal capacity of female BMN. We found that levels of C3 in the serum and CR3 (CD11b/CD18) on the surface of BMN were higher in female compared with male mice. Consistent with increased CR3 expression following TNF-α priming, production of reactive oxygen species (ROS), an important bactericidal effector, was also increased in female versus male BMN in response to serum-opsonized S. aureus Furthermore, blocking CD11b reduced both ROS levels and S. aureus killing by murine BMN from both sexes. However, at the same concentration of CD11b blocking Ab, S. aureus killing by female BMN was greatly reduced compared with those from male mice, suggesting CR3-dependent differences in bacterial killing between sexes. Overall, this work highlights the contributions of CR3, C3, and ROS to innate sex bias in the neutrophil response to S. aureus Given that neutrophils are crucial for S. aureus clearance, understanding the mechanism(s) driving the innate sex bias in neutrophil bactericidal capacity could identify novel host factors important for host defense against S. aureus.
Collapse
Affiliation(s)
- Srijana Pokhrel
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131
| | - Kathleen D Triplett
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131
| | - Seth M Daly
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131
| | - Jason A Joyner
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131
| | - Geetanjali Sharma
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131; and
| | - Helen J Hathaway
- Department of Cell Biology and Physiology, University of New Mexico School of Medicine, Albuquerque, NM 87131
| | - Eric R Prossnitz
- Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, NM 87131; and
| | - Pamela R Hall
- Department of Pharmaceutical Sciences, University of New Mexico College of Pharmacy, Albuquerque, NM 87131;
| |
Collapse
|
18
|
Amyloid Peptide β1-42 Induces Integrin αIIb β3 Activation, Platelet Adhesion, and Thrombus Formation in a NADPH Oxidase-Dependent Manner. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1050476. [PMID: 31007831 PMCID: PMC6441506 DOI: 10.1155/2019/1050476] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 11/05/2018] [Accepted: 12/13/2018] [Indexed: 01/02/2023]
Abstract
The progression of Alzheimer's dementia is associated with neurovasculature impairment, which includes inflammation, microthromboses, and reduced cerebral blood flow. Here, we investigate the effects of β amyloid peptides on the function of platelets, the cells driving haemostasis. Amyloid peptide β1-42 (Aβ1-42), Aβ1-40, and Aβ25-35 were tested in static adhesion experiments, and it was found that platelets preferentially adhere to Aβ1-42 compared to other Aβ peptides. In addition, significant platelet spreading was observed over Aβ1-42, while Aβ1-40, Aβ25-35, and the scAβ1-42 control did not seem to induce any platelet spreading, which suggested that only Aβ1-42 activates platelet signalling in our experimental conditions. Aβ1-42 also induced significant platelet adhesion and thrombus formation in whole blood under venous flow condition, while other Aβ peptides did not. The molecular mechanism of Aβ1-42 was investigated by flow cytometry, which revealed that this peptide induces a significant activation of integrin αIIbβ3, but does not induce platelet degranulation (as measured by P-selectin membrane translocation). Finally, Aβ1-42 treatment of human platelets led to detectable levels of protein kinase C (PKC) activation and tyrosine phosphorylation, which are hallmarks of platelet signalling. Interestingly, the NADPH oxidase (NOX) inhibitor VAS2870 completely abolished Aβ1-42-dependent platelet adhesion in static conditions, thrombus formation in physiological flow conditions, integrin αIIbβ3 activation, and tyrosine- and PKC-dependent platelet signalling. In summary, this study highlights the importance of NOXs in the activation of platelets in response to amyloid peptide β1-42. The molecular mechanisms described in this manuscript may play an important role in the neurovascular impairment observed in Alzheimer's patients.
Collapse
|
19
|
Mahtta D, Bavry AA. αIIbβ3 (GPIIb-IIIa) Antagonists. Platelets 2019. [DOI: 10.1016/b978-0-12-813456-6.00052-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
20
|
De Marco R, Greco A, Calonghi N, Dattoli SD, Baiula M, Spampinato S, Picchetti P, De Cola L, Anselmi M, Cipriani F, Gentilucci L. Selective detection of α4β1 integrin (VLA-4)-expressing cells using peptide-functionalized nanostructured materials mimicking endothelial surfaces adjacent to inflammatory sites. Biopolymers 2017; 110. [PMID: 29178262 DOI: 10.1002/bip.23081] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/06/2017] [Accepted: 10/18/2017] [Indexed: 01/01/2023]
Abstract
Persistent accumulation of immune cells mediated by α4β1 integrin (VLA-4) is a hallmark of the inflammatory diseases and of chronic inflammation observed in the affected tissues of autoimmune diseases. Aiming at exploring new methods for monitoring the course of the inflammatory processes, we designed the first peptide-functionalized nanostructured devices capable to mimic the high-density multivalency binding between the α4β1 integrin-expressing cells and the ligands overexpressed on the endothelial surfaces, in the proximity of the sites of inflammation. Specifically, we describe the first examples of monolayers constituted by dye-loaded zeolite L crystals, coated with α4β1 integrin peptide ligands, and we analyze the adhesion of model Jurkat cells in comparison to non-α4β1 integrin-expressing cells. In particular, the peptidomimetic diphenylurea-Leu-Asp-Val-diamine allows significant and selective detection of α4β1 integrin-expressing Jurkat cells, after very rapid incubation time, supporting the possible implementation in a diagnostic device capable to detect the desired cells from biological fluids, obtainable from patients in a noninvasive way.
Collapse
Affiliation(s)
- Rossella De Marco
- Department of Chemistry "G. Ciamician", University of Bologna, via Selmi 2, Bologna, 40126, Italy
| | - Arianna Greco
- Department of Chemistry "G. Ciamician", University of Bologna, via Selmi 2, Bologna, 40126, Italy
| | - Natalia Calonghi
- Department of Pharmacy and Biotechnology, University of Bologna, via Irnerio 48, Bologna, 40126, Italy
| | - Samantha D Dattoli
- Department of Pharmacy and Biotechnology, University of Bologna, via Irnerio 48, Bologna, 40126, Italy
| | - Monica Baiula
- Department of Pharmacy and Biotechnology, University of Bologna, via Irnerio 48, Bologna, 40126, Italy
| | - Santi Spampinato
- Department of Pharmacy and Biotechnology, University of Bologna, via Irnerio 48, Bologna, 40126, Italy
| | - Pierre Picchetti
- Institut de science et d'ingénierie supramoléculaires (ISIS), Université de Strasbourg and CNR UMR 7006, 8 Allée Gaspard Monge, Strasbourg, 67000, France
| | - Luisa De Cola
- Institut de science et d'ingénierie supramoléculaires (ISIS), Université de Strasbourg and CNR UMR 7006, 8 Allée Gaspard Monge, Strasbourg, 67000, France
- Institut fűr Nanotechnologie (INT), Karlsruhe Institute of Technology (KIT) - Campus Nord, Hermann-von-Helmholtz-Platz 1, Eggenstein-Leopoldshafen, 76344, Germany
| | - Michele Anselmi
- Department of Chemistry "G. Ciamician", University of Bologna, via Selmi 2, Bologna, 40126, Italy
| | - Francesca Cipriani
- Department of Medical and Surgical Sciences, University of Bologna, Via Massarenti 11, Bologna, 40138, Italy
| | - Luca Gentilucci
- Department of Chemistry "G. Ciamician", University of Bologna, via Selmi 2, Bologna, 40126, Italy
| |
Collapse
|
21
|
Abdullahi M, Olotu FA, Soliman ME. Dynamics of allosteric modulation of lymphocyte function associated antigen-1 closure-open switch: unveiling the structural mechanisms associated with outside-in signaling activation. Biotechnol Lett 2017; 39:1843-1851. [PMID: 28918491 DOI: 10.1007/s10529-017-2432-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 08/31/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVES To provide insight into the dynamics of the shape-shifting mechanistic events associated with the opening (activation) of Lymphocyte Function Associated Antigen-1 upon allosteric modulation by an activator, ICAM Binding Enhancer-667 (IBE-667), using molecular dynamics simulation. RESULTS Various parameters were used to appropriately describe and understand the sequence of events that characterized its activation across the simulation period such as residual distances, TriCα angles; as well as the dihedral angle. Our findings revealed a significant residual fluctuation and stability difference between both systems. Also, there was a synergistic coordination of the active MIDAS site by the downward pull of the α7 helix upon ligand binding, which appeared to be directly proportional to each other. CONCLUSION Allosteric binding of IBE-667, activated LFA-1 integrin as evidenced by residual motion at the MIDAS region which appears to be synergistically coordinated by the downward pull of the α7 helix.
Collapse
Affiliation(s)
- Maryam Abdullahi
- Molecular Modeling and Drug Design Research, Group School of Health Sciences, University of KwaZulu- Natal, Westville Campus, Durban, 4001, South Africa
| | - Fisayo A Olotu
- Molecular Modeling and Drug Design Research, Group School of Health Sciences, University of KwaZulu- Natal, Westville Campus, Durban, 4001, South Africa
| | - Mahmoud E Soliman
- Molecular Modeling and Drug Design Research, Group School of Health Sciences, University of KwaZulu- Natal, Westville Campus, Durban, 4001, South Africa. .,College of Pharmacy and Pharmaceutical Sciences, Florida Agricultural and Mechanical University (FAMU), Tallahassee, FL, 32307, USA. .,Department of Pharmaceutical Organic Chemistry Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
| |
Collapse
|
22
|
Hoshiba T, Yoshihiro A, Tanaka M. Evaluation of initial cell adhesion on poly (2-methoxyethyl acrylate) (PMEA) analogous polymers. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2017; 28:986-999. [DOI: 10.1080/09205063.2017.1312738] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Takashi Hoshiba
- Frontier Center for Organic Materials, Yamagata University, Yonezawa, Japan
- Innovative Flex Course for Frontier Organic Materials Systems, Yamagata University, Yonezawa, Japan
- International Center for Materials Nanoarchitectonics, National Institute for Materials Science, Tsukuba, Japan
| | - Ayano Yoshihiro
- Faculty of Engineering, Yamagata University, Yonezawa, Japan
| | - Masaru Tanaka
- Frontier Center for Organic Materials, Yamagata University, Yonezawa, Japan
- Institute for Materials Chemistry and Engineering, Kyushu University, Fukuoka, Japan
| |
Collapse
|
23
|
Ebrahimi S, Rahmani F, Behnam-Rassouli R, Hoseinkhani F, Parizadeh MR, Keramati MR, Khazaie M, Avan A, Hassanian SM. Proinflammatory signaling functions of thrombin in cancer. J Cell Physiol 2017; 232:2323-2329. [PMID: 28004386 DOI: 10.1002/jcp.25753] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 12/19/2022]
Abstract
Thrombin-induced activation of protease-activated receptors (PARs) represents a link between inflammation and cancer. Proinflammatory signaling functions of thrombin are associated with several inflammatory diseases including neurodegenerative, cardiovascular, and of special interest in this review cancer. Thrombin-induced inflammatory responses up-regulates expression of cytokines, adhesion molecules, angiogenic factors, and matrix-degrading proteases that facilitate tumor cells proliferation, angiogenesis, invasion, and metastasis. This review summarizes the current knowledge about the mechanisms of thrombin-mediated proinflammatory responses in cancer pathology for a better understanding and hence a better management of this disease.
Collapse
Affiliation(s)
- Safieh Ebrahimi
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farzad Rahmani
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Fatemeh Hoseinkhani
- Department of Medical Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Parizadeh
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Keramati
- Cancer Molecular Pathology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaie
- Department of Medical Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Molecular Medicine Group, Department of Modern Sciences and Technologies, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
24
|
Integrin signaling in atherosclerosis. Cell Mol Life Sci 2017; 74:2263-2282. [PMID: 28246700 DOI: 10.1007/s00018-017-2490-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/24/2017] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
Abstract
Atherosclerosis, a chronic lipid-driven inflammatory disease affecting large arteries, represents the primary cause of cardiovascular disease in the world. The local remodeling of the vessel intima during atherosclerosis involves the modulation of vascular cell phenotype, alteration of cell migration and proliferation, and propagation of local extracellular matrix remodeling. All of these responses represent targets of the integrin family of cell adhesion receptors. As such, alterations in integrin signaling affect multiple aspects of atherosclerosis, from the earliest induction of inflammation to the development of advanced fibrotic plaques. Integrin signaling has been shown to regulate endothelial phenotype, facilitate leukocyte homing, affect leukocyte function, and drive smooth muscle fibroproliferative remodeling. In addition, integrin signaling in platelets contributes to the thrombotic complications that typically drive the clinical manifestation of cardiovascular disease. In this review, we examine the current literature on integrin regulation of atherosclerotic plaque development and the suitability of integrins as potential therapeutic targets to limit cardiovascular disease and its complications.
Collapse
|
25
|
Kanazawa H, Imoto K, Okada M, Yamawaki H. Canstatin inhibits hypoxia-induced apoptosis through activation of integrin/focal adhesion kinase/Akt signaling pathway in H9c2 cardiomyoblasts. PLoS One 2017; 12:e0173051. [PMID: 28235037 PMCID: PMC5325616 DOI: 10.1371/journal.pone.0173051] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 02/14/2017] [Indexed: 12/19/2022] Open
Abstract
A hypoxic stress which causes apoptosis of cardiomyocytes is the main problem in the ischemic heart disease. Canstatin, a non-collagenous fragment of type IV collagen α2 chain, is an endogenous anti-angiogenic factor. We have previously reported that canstatin has a cytoprotective effect on cardiomyoblasts. In the present study, we examined the effects of canstatin on hypoxia-induced apoptosis in H9c2 cardiomyoblasts. Cell counting assay was performed to determine a cell viability. Western blotting was performed to detect expression of cleaved casepase-3 and phosphorylation of focal adhesion kinase (FAK) and Akt. Immunocytochemical staining was performed to observe a distribution of αv integrin. Hypoxia (1% O2, 48 h) significantly decreased cell viability and increased cleaved caspase-3 expression. Canstatin (10–250 ng/ml) significantly inhibited these changes in a concentration-dependent manner. Cilengitide (1 μM), an αvβ3 and αvβ5 integrin inhibitor, significantly prevented the protective effects of canstatin on cell viability. Canstatin significantly increased phosphorylation of FAK and Akt under hypoxic condition, which were inhibited by cilengitide. LY294002, an inhibitor of phosphatidylinositol-3 kinase/Akt pathway, suppressed the canstatin-induced Akt phosphorylation and reversed the protective effects of canstatin. It was observed that hypoxia caused a localization of αv integrin to focal adhesion. In summary, we for the first time clarified that canstatin inhibits hypoxia-induced apoptosis via FAK and Akt pathways through activating integrins in H9c2 cardiomyoblasts.
Collapse
Affiliation(s)
- Hiroki Kanazawa
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Keisuke Imoto
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| | - Muneyoshi Okada
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
- * E-mail:
| | - Hideyuki Yamawaki
- Laboratory of Veterinary Pharmacology, School of Veterinary Medicine, Kitasato University, Towada, Aomori, Japan
| |
Collapse
|
26
|
Russell-Puleri S, Dela Paz NG, Adams D, Chattopadhyay M, Cancel L, Ebong E, Orr AW, Frangos JA, Tarbell JM. Fluid shear stress induces upregulation of COX-2 and PGI 2 release in endothelial cells via a pathway involving PECAM-1, PI3K, FAK, and p38. Am J Physiol Heart Circ Physiol 2016; 312:H485-H500. [PMID: 28011582 PMCID: PMC5402016 DOI: 10.1152/ajpheart.00035.2016] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 10/31/2016] [Accepted: 11/21/2016] [Indexed: 12/16/2022]
Abstract
Vascular endothelial cells play an important role in the regulation of vascular function in response to mechanical stimuli in both healthy and diseased states. Prostaglandin I2 (PGI2) is an important antiatherogenic prostanoid and vasodilator produced in endothelial cells through the action of the cyclooxygenase (COX) isoenzymes COX-1 and COX-2. However, the mechanisms involved in sustained, shear-induced production of COX-2 and PGI2 have not been elucidated but are determined in the present study. We used cultured endothelial cells exposed to steady fluid shear stress (FSS) of 10 dyn/cm2 for 5 h to examine shear stress-induced induction of COX-2/PGI2 Our results demonstrate the relationship between the mechanosensor platelet endothelial cell adhesion molecule-1 (PECAM-1) and the intracellular mechanoresponsive molecules phosphatidylinositol 3-kinase (PI3K), focal adhesion kinase (FAK), and mitogen-activated protein kinase p38 in the FSS induction of COX-2 expression and PGI2 release. Knockdown of PECAM-1 (small interference RNA) expression inhibited FSS-induced activation of α5β1-integrin, upregulation of COX-2, and release of PGI2 in both bovine aortic endothelial cells (BAECs) and human umbilical vein endothelial cells (HUVECs). Furthermore, inhibition of the PI3K pathway (LY294002) substantially inhibited FSS activation of α5β1-integrin, upregulation of COX-2 gene and protein expression, and release of PGI2 in BAECs. Inhibition of integrin-associated FAK (PF573228) and MAPK p38 (SB203580) also inhibited the shear-induced upregulation of COX-2. Finally, a PECAM-1-/- mouse model was characterized by reduced COX-2 immunostaining in the aorta and reduced plasma PGI2 levels compared with wild-type mice, as well as complete inhibition of acute flow-induced PGI2 release compared with wild-type animals.NEW & NOTEWORTHY In this study we determined the major mechanotransduction pathway by which blood flow-driven shear stress activates cyclooxygenase-2 (COX-2) and prostaglandin I2 (PGI2) release in endothelial cells. Our work has demonstrated for the first time that COX-2/PGI2 mechanotransduction is mediated by the mechanosensor platelet endothelial cell adhesion molecule-1 (PECAM-1).
Collapse
Affiliation(s)
| | | | - Diana Adams
- La Jolla Bioengineering Institute, La Jolla, California
| | | | - Limary Cancel
- Department of Biomedical Engineering, City College of New York, New York, New York
| | - Eno Ebong
- Department of Chemical Engineering, Northeastern University, Boston, Massachusetts; and
| | - A Wayne Orr
- Department of Pathology, Louisiana State University, Shreveport, Louisiana
| | | | - John M Tarbell
- Department of Biomedical Engineering, City College of New York, New York, New York;
| |
Collapse
|
27
|
Cytohesin 1 regulates homing and engraftment of human hematopoietic stem and progenitor cells. Blood 2016; 129:950-958. [PMID: 27899358 DOI: 10.1182/blood-2016-06-720649] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 11/17/2016] [Indexed: 12/25/2022] Open
Abstract
Adhesion is a key component of hematopoietic stem cell regulation mediating homing and retention to the niche in the bone marrow. Here, using an RNA interference screen, we identify cytohesin 1 (CYTH1) as a critical mediator of adhesive properties in primary human cord blood-derived hematopoietic stem and progenitor cells (HSPCs). Knockdown of CYTH1 disrupted adhesion of HSPCs to primary human mesenchymal stroma cells. Attachment to fibronectin and ICAM1, 2 integrin ligands, was severely impaired, and CYTH1-deficient cells showed a reduced integrin β1 activation response, suggesting that CYTH1 mediates integrin-dependent functions. Transplantation of CYTH1-knockdown cells to immunodeficient mice resulted in significantly lower long-term engraftment levels, associated with a reduced capacity of the transplanted cells to home to the bone marrow. Intravital microscopy showed that CYTH1 deficiency profoundly affects HSPC mobility and localization within the marrow space and thereby impairs proper lodgment into the niche. Thus, CYTH1 is a novel major regulator of adhesion and engraftment in human HSPCs through mechanisms that, at least in part, involve the activation of integrins.
Collapse
|
28
|
Fuhrmann A, Engler AJ. The cytoskeleton regulates cell attachment strength. Biophys J 2016; 109:57-65. [PMID: 26153702 DOI: 10.1016/j.bpj.2015.06.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 05/19/2015] [Accepted: 06/03/2015] [Indexed: 11/25/2022] Open
Abstract
Quantitative information about adhesion strength is a fundamental part of our understanding of cell-extracellular matrix (ECM) interactions. Adhesion assays should measure integrin-ECM bond strength, but reports now suggest that cell components remain behind after exposure to acute force for radial shear assays in the presence of divalent cations that increase integrin-ECM affinity. Here, we show that focal adhesion proteins FAK, paxillin, and vinculin but not the cytoskeletal protein actin remain behind after shear-induced detachment of HT1080 fibrosarcoma cells. Cytoskeletal stabilization increased attachment strength by eightfold, whereas cross-linking integrins to the substrate only caused a 1.5-fold increase. Reducing temperature-only during shear application-also increased attachment strength eightfold, with detachment again occurring between focal adhesion proteins and actin. Detachment at the focal adhesion-cytoskeleton interface was also observed in mouse and human fibroblasts and was ligand-independent, highlighting the ubiquity of this mode of detachment in the presence of divalent cations. These data show that the cytoskeleton and its dynamic coupling to focal adhesions are critically important for cell adhesion in niche with divalent cations.
Collapse
Affiliation(s)
- Alexander Fuhrmann
- Department of Bioengineering, University of California, San Diego, La Jolla, California
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, California; Sanford Consortium for Regenerative Medicine, La Jolla, California.
| |
Collapse
|
29
|
Long K, Moss L, Laursen L, Boulter L, Ffrench-Constant C. Integrin signalling regulates the expansion of neuroepithelial progenitors and neurogenesis via Wnt7a and Decorin. Nat Commun 2016; 7:10354. [PMID: 26838601 PMCID: PMC4742793 DOI: 10.1038/ncomms10354] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2015] [Accepted: 12/03/2015] [Indexed: 01/14/2023] Open
Abstract
Development of the cerebral cortex requires regulation of proliferation and differentiation of neural stem cells and a diverse range of progenitors. Recent work suggests a role for extracellular matrix (ECM) and the major family of ECM receptors, the integrins. Here we show that enhancing integrin beta-1 signalling, by expressing a constitutively active integrin beta-1 (CA*β1) in the embryonic chick mesencephalon, enhances neurogenesis and increases the number of mitotic cells dividing away from the ventricular surface, analogous to sub-apical progenitors in mouse. Only non-integrin-expressing neighbouring cells (lacking CA*β1) contributed to the increased neurogenesis. Transcriptome analysis reveals upregulation of Wnt7a within the CA*β1 cells and upregulation of the ECM protein Decorin in the neighbouring non-expressing cells. Experiments using inhibitors in explant models and genetic knock-downs in vivo reveal an integrin-Wnt7a-Decorin pathway that promotes proliferation and differentiation of neuroepithelial cells, and identify Decorin as a novel neurogenic factor in the central nervous system. The extracellular matrix is suggested to play a role in neurogenesis, but it is unclear what role integrin signalling may play in the developing neuroepithelium. Here, in chick, Long et al. show that expression of constitutively active integrin beta-1 enhances neurogenesis via a novel Wnt7 and decorin pathway.
Collapse
Affiliation(s)
- K Long
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - L Moss
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - L Laursen
- Department of Molecular biology and Genetics, Aarhus University, Gustav Wieds Vej 10 C, 8000 Aarhus C, Denmark
| | - L Boulter
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, Crewe Road, Edinburgh EH4 2XU, UK
| | - C Ffrench-Constant
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, UK
| |
Collapse
|
30
|
Integrative Utilization of Microenvironments, Biomaterials and Computational Techniques for Advanced Tissue Engineering. J Biotechnol 2015; 212:71-89. [DOI: 10.1016/j.jbiotec.2015.08.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 08/02/2015] [Accepted: 08/11/2015] [Indexed: 01/13/2023]
|
31
|
Lemaire M, Negro Silva LF, Lemarié CA, Bolt AM, Flores Molina M, Krohn RM, Smits JE, Lehoux S, Mann KK. Arsenic Exposure Increases Monocyte Adhesion to the Vascular Endothelium, a Pro-Atherogenic Mechanism. PLoS One 2015; 10:e0136592. [PMID: 26332580 PMCID: PMC4557830 DOI: 10.1371/journal.pone.0136592] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/05/2015] [Indexed: 01/24/2023] Open
Abstract
Epidemiological studies have shown that arsenic exposure increases atherosclerosis, but the mechanisms underlying this relationship are unknown. Monocytes, macrophages and platelets play an important role in the initiation of atherosclerosis. Circulating monocytes and macrophages bind to the activated vascular endothelium and migrate into the sub-endothelium, where they become lipid-laden foam cells. This process can be facilitated by platelets, which favour monocyte recruitment to the lesion. Thus, we assessed the effects of low-to-moderate arsenic exposure on monocyte adhesion to endothelial cells, platelet activation and platelet-monocyte interactions. We observed that arsenic induces human monocyte adhesion to endothelial cells in vitro. These findings were confirmed ex vivo using a murine organ culture system at concentrations as low as 10 ppb. We found that both cell types need to be exposed to arsenic to maximize monocyte adhesion to the endothelium. This adhesion process is specific to monocyte/endothelium interactions. Hence, no effect of arsenic on platelet activation or platelet/leukocyte interaction was observed. We found that arsenic increases adhesion of mononuclear cells via increased CD29 binding to VCAM-1, an adhesion molecule found on activated endothelial cells. Similar results were observed in vivo, where arsenic-exposed mice exhibit increased VCAM-1 expression on endothelial cells and increased CD29 on circulating monocytes. Interestingly, expression of adhesion molecules and increased binding can be inhibited by antioxidants in vitro and in vivo. Together, these data suggest that arsenic might enhance atherosclerosis by increasing monocyte adhesion to endothelial cells, a process that is inhibited by antioxidants.
Collapse
Affiliation(s)
- Maryse Lemaire
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
| | - Luis Fernando Negro Silva
- Division of Experimental Medicine, Department of Medicine, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
| | - Catherine A. Lemarié
- Division of Experimental Medicine, Department of Medicine, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
- Department of Medicine, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
| | - Alicia M. Bolt
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
| | - Manuel Flores Molina
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
| | - Regina M. Krohn
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Judit E. Smits
- Department of Ecosystem and Public Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Stéphanie Lehoux
- Department of Medicine, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
| | - Koren K. Mann
- Department of Oncology, Lady Davis Institute for Medical Research, McGill University, Montréal, Québec, Canada
- * E-mail:
| |
Collapse
|
32
|
Affiliation(s)
- Yukio Takeshita
- Department of Neurology and Clinical Neuroscience; Yamaguchi University Graduate School of Medicine; Ube Yamaguchi Japan
| | | |
Collapse
|
33
|
Qu T, Zhang SM, Yu LL, Zhang S, Yuan DZ, Xu Q, Zhang JH, He YP, Yue LM. Relocalisation and activation of integrins induced rapidly by oestrogen via G-protein-coupled receptor 30 in mouse blastocysts. Reprod Fertil Dev 2015; 28:RD14227. [PMID: 25950704 DOI: 10.1071/rd14227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 03/21/2015] [Indexed: 12/20/2022] Open
Abstract
Integrins are the dominant and final adhesion molecules in the attachment process between the blastocysts and endometrium. It is necessary for oestrogen to rapidly activate mouse blastocysts so that they attach to the endometrial epithelium. Our previous study suggested that oestrogen can rapidly induce an increase in intracellular calcium in mouse blastocysts via G-protein-coupled receptor 30 (GPR30). Thus, we deduced that integrins may be involved in GPR30 mediation of the fast effect of oestrogen on mouse blastocysts in implantation. To prove our hypothesis, we used immunofluorescence staining and in vitro coculture of mouse blastocysts and endometrial epithelial cell line (EECs), Ishikawa cells, in the present study. We found that αv and β1 integrin clustered in mouse blastocysts, and that β3 integrin was relocalised to the apical membrane of blastocyst cells when embryos were treated with 1 μM 17β-estradiol (E2), 1 μM E2 conjugated to bovine serum albumin (E2-BSA) and 1 μM G-1, a specific GPR30 agonist, for 30 min respectively, whereas pretreatment with 1 μM G15, a specific GPR30 antagonist, and 5 μM 1,2-Bis(2-aminophenoxy)ethane-N,N,N'',N''-tetraacetic acid tetrakis (acetoxymethyl ester)(BAPTA/AM), a cellular Ca2+ chelator, blocked the localisation of integrins induced by oestrogen via GPR30 in mouse blastocyst cells. E2, E2-BSA and G-1 increased the fibronectin (FN)-binding activity of integrins in blastocysts, whereas G15 and BAPTA/AM blocked the activation of integrins induced by oestrogen via GPR30 in mouse blastocysts. Inhibition of integrins by Arg-Gly-Asp peptide in blastocysts resulted in their failure to adhere to EECs in vitro, even if oestrogen or G-1 was provided. Together, the results indicate the fast effect of oestrogen via the GPR30 membrane receptor further induces relocalisation and activation of integrins in mouse blastocysts, which play important roles in the adhesion of blastocysts to EECs.
Collapse
|
34
|
Roll, adhere, spread and contract: structural mechanics of platelet function. Eur J Cell Biol 2015; 94:129-38. [PMID: 25655000 DOI: 10.1016/j.ejcb.2015.01.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 12/26/2014] [Accepted: 01/07/2015] [Indexed: 12/31/2022] Open
Abstract
Platelets are involved in life-sustaining processes such as hemostasis, wound healing, atherothrombosis and angiogenesis. Mechanical trauma to blood vessels causes platelet activation resulting in their adherence and clot formation at the damaged site, culminating in clot retraction and tissue repair. Two of the major players underlying this process are the cytoskeleton, i.e., actin and microtubules, and the membrane integrin receptors. Rare congenital bleeding disorders such as Glanzmann thrombasthenia and Bernard-Soulier syndrome are associated with genetic alterations of platelet surface receptors, also affecting the platelet cytoskeletal structure. In this review, we summarize the current knowledge about platelet structure and adhesion, and delve into the mechanical aspects of platelet function. Platelets lack a nucleus, and can thus provide a minimal model of a biological cell. New biophysical tools may help to scrutinize platelets anew and to extend the existing knowledge on cell biology.
Collapse
|
35
|
MUC1 extracellular domain confers resistance of epithelial cancer cells to anoikis. Cell Death Dis 2014; 5:e1438. [PMID: 25275599 PMCID: PMC4649521 DOI: 10.1038/cddis.2014.421] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/29/2014] [Accepted: 09/01/2014] [Indexed: 01/10/2023]
Abstract
Anoikis, a special apoptotic process occurring in response to loss of cell adhesion to the extracellular matrix, is a fundamental surveillance process for maintaining tissue homeostasis. Resistance to anoikis characterises cancer cells and is a pre-requisite for metastasis. This study shows that overexpression of the transmembrane mucin protein MUC1 prevents initiation of anoikis in epithelial cancer cells in response to loss of adhesion. We show that this effect is largely attributed to the elongated and heavily glycosylated extracellular domain of MUC1 that protrudes high above the cell membrane and hence prevents activation of the cell surface anoikis-initiating molecules such as integrins and death receptors by providing them a mechanically ‘homing' microenvironment. As overexpression of MUC1 is a common feature of epithelial cancers and as resistance to anoikis is a hallmark of both oncogenic epithelial–mesenchymal transition and metastasis, MUC1-mediated cell resistance to anoikis may represent one of the fundamental regulatory mechanisms in tumourigenesis and metastasis.
Collapse
|
36
|
Herrera-García AM, Domínguez-Luis MJ, Arce-Franco M, Armas-González E, Álvarez de La Rosa D, Machado JD, Pec MK, Feria M, Barreiro O, Sánchez-Madrid F, Díaz-González F. Prevention of neutrophil extravasation by α2-adrenoceptor-mediated endothelial stabilization. THE JOURNAL OF IMMUNOLOGY 2014; 193:3023-35. [PMID: 25114107 DOI: 10.4049/jimmunol.1400255] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adrenergic receptors are expressed on the surface of inflammation-mediating cells, but their potential role in the regulation of the inflammatory response is still poorly understood. The objectives of this work were to study the effects of α2-adrenergic agonists on the inflammatory response in vivo and to determine their mechanism of action. In two mouse models of inflammation, zymosan air pouch and thioglycolate-induced peritonitis models, the i.m. treatment with xylazine or UK14304, two α2-adrenergic agonists, reduced neutrophil migration by 60%. The α2-adrenergic antagonist RX821002 abrogated this effect. In flow cytometry experiments, the basal surface expression of L-selectin and CD11b was modified neither in murine nor in human neutrophils upon α2-agonist treatment. Similar experiments in HUVEC showed that UK14304 prevented the activation-dependent upregulation of ICAM-1. In contrast, UK14304 augmented electrical resistance and reduced macromolecular transport through a confluent HUVEC monolayer. In flow chamber experiments, under postcapillary venule-like flow conditions, the pretreatment of HUVECs, but not neutrophils, with α2-agonists decreased transendothelial migration, without affecting neutrophil rolling. Interestingly, α2-agonists prevented the TNF-α-mediated decrease in expression of the adherens junctional molecules, VE-cadherin, β-catenin, and plakoglobin, and reduced the ICAM-1-mediated phosphorylation of VE-cadherin by immunofluorescence and confocal analysis and Western blot analysis, respectively. These findings indicate that α2-adrenoceptors trigger signals that protect the integrity of endothelial adherens junctions during the inflammatory response, thus pointing at the vascular endothelium as a therapeutic target for the management of inflammatory processes in humans.
Collapse
Affiliation(s)
- Ada María Herrera-García
- Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - María Jesús Domínguez-Luis
- Centro para la Investigación Biomédica de las Islas Canarias, Instituto de Tecnologías Biomedicas, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - María Arce-Franco
- Servicio de Reumatología, Hospital Universitario de Canarias, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Estefanía Armas-González
- Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Diego Álvarez de La Rosa
- Departamento de Fisiología, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - José David Machado
- Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Martina K Pec
- Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Manuel Feria
- Departamento de Farmacología, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain
| | - Olga Barreiro
- Departamento de Biología Vascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares, Consejo Superior de Investigaciones Cientificas, 28029 Madrid, Spain; and
| | - Francisco Sánchez-Madrid
- Departamento de Biología Vascular e Inflamación, Centro Nacional de Investigaciones Cardiovasculares, Consejo Superior de Investigaciones Cientificas, 28029 Madrid, Spain; and Servicio de Inmunología, Hospital Universitario de La Princesa, 28006 Madrid, Spain
| | - Federico Díaz-González
- Departamento de Medicina, Facultad de Medicina, Universidad de La Laguna, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain; Servicio de Reumatología, Hospital Universitario de Canarias, La Cuesta, 38320 San Cristóbal de La Laguna, Santa Cruz de Tenerife, Spain;
| |
Collapse
|
37
|
ZHOU TIAN, ZHENG YIMING, QIU JUHUI, HU JIANJUN, SUN DAMING, TANG CHAOJUN, WANG GUIXUE. ENDOTHELIAL MECHANOTRANSDUCTION MECHANISMS FOR VASCULAR PHYSIOLOGY AND ATHEROSCLEROSIS. J MECH MED BIOL 2014. [DOI: 10.1142/s0219519414300063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Vascular physiology and disease progression, such as atherosclerosis, are mediated by hemodynamic force generated from blood flow. The hemodynamic force exerts on vascular endothelial cells (ECs), which could perceive the mechanical signals and transmit them into cell interior by multiple potential shear sensors, collectively known as mechanotransduction. However, we do not understand completely how these shear-sensitive components orchestrate physiological and atherosclerotic responses to shear stress. In this review, we provide an overview of biomechanical mechanisms underlying vascular physiology and atherosclerotic progression. Additionally, we summarize current evidences to illustrate that atherosclerotic lesions preferentially develop in arterial regions experiencing disturbance in blood flow, during which endothelial dysfunction is the initial event of atherosclerosis, inflammation plays dominant roles in atherosclerotic progression, and angiogenesis emerges as compensatory explanation for atherosclerotic plaque rupture. Especially in the presence of systemic risk factors (e.g., hyperlipidaemia, hypertension and hyperglycemia), the synergy between these systemic risk factors with hemodynamic factors aggravates atherosclerosis by co-stimulating some of these biomechanical events. Given the hemodynamic environment of vasculature, understanding how the rapid shear-mediated signaling, particularly in combination with systemic risk factors, contribute to atherosclerotic progression through endothelial dysfunction, inflammation and angiogenesis helps to elucidate the role for atherogenic shear stress in specifically localizing atherosclerotic lesions in arterial regions with disturbed flow.
Collapse
Affiliation(s)
- TIAN ZHOU
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - YIMING ZHENG
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - JUHUI QIU
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - JIANJUN HU
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - DAMING SUN
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - CHAOJUN TANG
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| | - GUIXUE WANG
- Key Laboratory of Biorheological Science and Technology (Chongqing University), Ministry of Education, Chongqing Engineering Laboratory in Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, P. R. China
| |
Collapse
|
38
|
Fuhrmann A, Li J, Chien S, Engler AJ. Cation type specific cell remodeling regulates attachment strength. PLoS One 2014; 9:e102424. [PMID: 25014042 PMCID: PMC4094514 DOI: 10.1371/journal.pone.0102424] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 06/18/2014] [Indexed: 11/19/2022] Open
Abstract
Single-molecule experiments indicate that integrin affinity is cation-type-dependent, but in spread cells integrins are engaged in complex focal adhesions (FAs), which can also regulate affinity. To better understand cation-type-dependent adhesion in fully spread cells, we investigated attachment strength by application of external shear. While cell attachment strength is indeed modulated by cations, the regulation of integrin-mediated adhesion is also exceedingly complex, cell specific, and niche dependent. In the presence of magnesium only, fibroblasts and fibrosarcoma cells remodel their cytoskeleton to align in the direction of applied shear in an α5-integrin/fibronectin-dependent manner, which allows them to withstand higher shear. In the presence of calcium or on collagen in modest shear, fibroblasts undergo piecewise detachment but fibrosarcoma cells exhibit increased attachment strength. These data augment the current understanding of force-mediated detachment by suggesting a dynamic interplay in situ between cell adhesion and integrins depending on local niche cation conditions.
Collapse
Affiliation(s)
- Alexander Fuhrmann
- Department of Bioengineering, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Julie Li
- Department of Bioengineering, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Shu Chien
- Department of Bioengineering, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
- Department of Medicine, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
- Sanford Consortium for Regenerative Medicine, La Jolla, California, United States of America
| | - Adam J. Engler
- Department of Bioengineering, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, United States of America
- Sanford Consortium for Regenerative Medicine, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
39
|
Fu R, Shen Q, Xu P, Luo JJ, Tang Y. Phagocytosis of microglia in the central nervous system diseases. Mol Neurobiol 2014; 49:1422-34. [PMID: 24395130 PMCID: PMC4012154 DOI: 10.1007/s12035-013-8620-6] [Citation(s) in RCA: 476] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/15/2013] [Indexed: 12/20/2022]
Abstract
Microglia, the resident macrophages of the central nervous system, rapidly activate in nearly all kinds of neurological diseases. These activated microglia become highly motile, secreting inflammatory cytokines, migrating to the lesion area, and phagocytosing cell debris or damaged neurons. During the past decades, the secretory property and chemotaxis of microglia have been well-studied, while relatively less attention has been paid to microglial phagocytosis. So far there is no obvious concordance with whether it is beneficial or detrimental in tissue repair. This review focuses on phagocytic phenotype of microglia in neurological diseases such as Alzheimer's disease, multiple sclerosis, Parkinson's disease, traumatic brain injury, ischemic and other brain diseases. Microglial morphological characteristics, involved receptors and signaling pathways, distribution variation along with time and space changes, and environmental factors that affecting phagocytic function in each disease are reviewed. Moreover, a comparison of contributions between macrophages from peripheral circulation and the resident microglia to these pathogenic processes will also be discussed.
Collapse
Affiliation(s)
- Ruying Fu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
| | - Qingyu Shen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
- Department of Neurology, Zengcheng People’s Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Pengfei Xu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
| | - Jin Jun Luo
- Department of Neurology, School of Medicine, Temple University, Philadelphia, PA USA
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Number 107, Yan Jiang Xi Road, Guangzhou, 510120 Guangdong Province China
- Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
40
|
Shih CH, Chiang TB, Wang WJ. A critical role for the regulation of Syk from agglutination to aggregation in human platelets. Biochem Biophys Res Commun 2013; 443:580-5. [PMID: 24326074 DOI: 10.1016/j.bbrc.2013.12.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 12/02/2013] [Indexed: 10/25/2022]
Abstract
Agglucetin, a tetrameric glycoprotein (GP) Ibα agonist from Formosan Agkistrodon acutus venom, has been characterized as an agglutination inducer in human washed platelets (WPs). In platelet-rich plasma (PRP), agglucetin dramatically elicits a biphasic response of agglutination and subsequent aggregation. For clarifying the intracellular signaling events from agglutination to aggregation in human platelets, we examined the essential signaling molecules involved through the detection of protein tyrosine phosphorylation (PTP). In WPs, an anti-GPIbα monoclonal antibody (mAb) AP1, but not a Src kinase inhibitor PP1, completely inhibited agglucetin-induced agglutination. However, PP1 but not AP1 had a potent suppression on platelet aggregation by a GPVI activator convulxin. The PTP analyses showed agglucetin alone can cause a weak pattern involving sequential phosphorylation of Lyn/Fyn, Syk, SLP-76 and phospholipase Cγ2 (PLCγ2). Furthermore, a Syk-selective kinase inhibitor, piceatannol, significantly suppressed the aggregating response in agglucetin-activated PRP. Analyzed by flow cytometry, the binding capacity of fluorophore-conjugated PAC-1, a mAb recognizing activated integrin αIIbβ3, was shown to increase in agglucetin-stimulated platelets. Again, piceatannol but not PP1 had a concentration-dependent suppression on agglucetin-induced αIIbβ3 exposure. Moreover, the formation of signalosome, including Syk, SLP-76, VAV, adhesion and degranulation promoting adapter protein (ADAP) and PLCγ2, are required for platelet aggregation in agglucetin/fibrinogen-activated platelets. In addition, GPIbα-ligation via agglucetin can substantially promote the interactions between αIIbβ3 and fibrinogen. Therefore, the signal pathway of Lyn/Fyn/Syk/SLP-76/ADAP/VAV/PLCγ2/PKC is sufficient to trigger platelet aggregation in agglucetin/fibrinogen-pretreated platelets. Importantly, Syk may function as a major regulator for the response from GPIbα-initiated agglutination to integrin αIIbβ3-dependent aggregation in human platelets.
Collapse
Affiliation(s)
- Chun-Ho Shih
- Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan, Taiwan
| | - Tin-Bin Chiang
- Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan, Taiwan
| | - Wen-Jeng Wang
- Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan, Taiwan.
| |
Collapse
|
41
|
Junctional adhesion molecule-A suppresses platelet integrin αIIbβ3 signaling by recruiting Csk to the integrin-c-Src complex. Blood 2013; 123:1393-402. [PMID: 24300854 DOI: 10.1182/blood-2013-04-496232] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Fibrinogen binding to activated integrin induces outside-in signaling that results in stable platelet aggregates and clot retraction. How integrin αIIbβ3 is discouraged from spontaneous activation is not known. We have recently shown that junctional adhesion molecule-A (JAM-A) renders protection from thrombosis by suppressing integrin outside-in signaling. In this study, we show that JAM-A associates with integrin αIIbβ3 in resting platelets and dissociates upon platelet activation by agonists. We also show that integrin-associated JAM-A is tyrosine phosphorylated and is rapidly dephosphorylated upon platelet activation. C-terminal Src kinase (Csk) binds to tyrosine phosphorylated JAM-A through its Src homology 2 domain. Thus, JAM-A recruits Csk to the integrin-c-Src complex in resting platelets. Csk, in turn, keeps integrin-associated c-Src in an inactive state by phosphorylating Y(529) in its regulatory domain. Absence of JAM-A results in impaired c-SrcY(529) phosphorylation and augmentation of outside-in signaling-dependent c-Src activation. Our results strongly suggest that tyrosine-phosphorylated JAM-A is a Csk-binding protein and functions as an endogenous inhibitor of integrin signaling. JAM-A recruits Csk to the integrin-c-Src complex, where Csk negatively regulates c-Src activation, thereby suppressing the initiation of outside-in signaling. Upon agonist stimulation, JAM-A is dephosphorylated on the tyrosine, allowing the dissociation of Csk from the integrin complex, and thus facilitating outside-in signaling.
Collapse
|
42
|
Zhong R, Tang X, Zhan W, Xing J, Sheng X. Expression kinetics of β-integrin in Chinese shrimp (Fenneropenaeus chinensis) hemocytes following infection with white spot syndrome virus. FISH & SHELLFISH IMMUNOLOGY 2013; 35:539-545. [PMID: 23711468 DOI: 10.1016/j.fsi.2013.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Revised: 04/15/2013] [Accepted: 05/06/2013] [Indexed: 06/02/2023]
Abstract
Our previous study has demonstrated that an integrin β subunit of Chinese shrimp (Fenneropenaeus chinensis) (FcβInt) involved in WSSV infection. In order to further elucidate the potential role of the FcβInt in the WSSV infection, expression response of FcβInt to WSSV infection in shrimp hemocytes was investigated after intra-muscular injection with the virus. Following time-course hemocytes sampling, the expression variation of FcβInt in hemocytes was examined by flow cytometric immunofluorescence assay (FCIFA) and enzyme linked immunosorbent assay (ELISA) using the monoclonal antibody (Mab) 2C5 against FcβInt, which was successfully produced with recombinant partial FcβInt and exhibited binding to a 120 kDa hemocyte protein. Meanwhile, the dynamic state of FcβInt mRNA level and WSSV copies in hemocytes were determined by quantitative real-time PCR. The result of FCIFA showed that FcβInt was mainly expressed on the semi-granular and granular cells, which was down-regulated at 6 h post infection (p.i.), and significantly increased to the peak level at 12 h p.i., then decreased to the control level by 24 h. However, FcβInt on the hyaline cells was lowly expressed and didn't show active response to the viral infection. The variation of FcβInt concentrations in total hemocytes determined by ELISA was roughly in accordance with the changing tendency of FcβInt expressed on the semi-granular and granular cells. FcβInt mRNA level in total hemocytes was significantly up-regulated to the peak level at 12 h p.i. Moreover, the number of WSSV copies in hemocytes began to exhibit a significant increase at 24 h p.i. The present study demonstrated that WSSV infection would induce a differential regulation of FcβInt expression in different type hemocytes, which provided valuable evidences for the close correlation between FcβInt and WSSV infection.
Collapse
Affiliation(s)
- Rujie Zhong
- Laboratory of Pathology and Immunology of Aquatic Animals, Ocean University of China, No. 5, Yushan Road, Qingdao 266003, PR China
| | | | | | | | | |
Collapse
|
43
|
P38MAP kinase, but not phosphoinositol-3 kinase, signal downstream of glutamine-mediated fibronectin-integrin signaling after intestinal injury. Nutr J 2013; 12:88. [PMID: 24499047 PMCID: PMC3691833 DOI: 10.1186/1475-2891-12-88] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 06/17/2013] [Indexed: 12/31/2022] Open
Abstract
Background Glutamine appears to mediate protection against gut injury via multiple pathways. These include fibronectin-integrin, PI3-K/MAPK pathways, and activation of heat shock protein (HSP) response. We hypothesize there may be a relationship between these pathways mediating glutamine’s protection in intestinal epithelial-6 cells after heat stress. We assessed whether p38MAPK and PI3-K/Akt signaling are involved in glutamine’s cytoprotective mechanism and the role they play in glutamine-mediated protection in conjunction with fibronectin-integrin osmosignaling after hyperthermia. Methods Intestinal epithelial cells were treated for 15 min with glutamine, with/without the fibronectin-integrin interaction inhibitor GRGDSP, inactive control peptide GRGESP, p38MAPK inhibitor SB203580, or PI3-K/Akt inhibitor LY294002 under basal (37°C) and stressed (43°C or 44°C) conditions. Cell survival was measured via MTS assay 24 h post-heat stress (44°C × 50 min). Total p38MAPK, [T(P)180/Y(P)182]p38MAPK, total Akt, [S(P)473]Akt, HSP70, FN, and caspase-3 levels were determined via Western blot after non-lethal HS (43°C × 50 min). Additionally, HSP70 levels were assessed via Western blot and ELISA. Results We were able to show that GRGDSP and LY294002 attenuated glutamine’s protective effect. However, SB203580 increased cell survival after heat stress. LY294002 attenuated glutamine-mediated increases in fibronectin and in HSP70 expression after hyperthermia. GRGDSP increased glutamine-mediated attenuations in p38MAPK phosphorylation, but had no effect on glutamine-mediated augmentations in Akt phosphorylation. Conclusions These data suggest that glutamine is protective after heat stress by activating PI3-K/Akt signaling preventing fibronectin-integrin expression and increasing HSP70 expression. Furthermore, dephosphorylation of p38MAPK after heat stress plays an important role in glutamine-mediated cellular protection. However, p38MAPK, but not PI3-K/Akt, signals downstream of glutamine-mediated fibronectin-integrin signaling after hyperthermia.
Collapse
|
44
|
Integrin α3 is required for late postnatal stability of dendrite arbors, dendritic spines and synapses, and mouse behavior. J Neurosci 2013; 33:6742-52. [PMID: 23595732 DOI: 10.1523/jneurosci.0528-13.2013] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Most dendrite branches and a large fraction of dendritic spines in the adult rodent forebrain are stable for extended periods of time. Destabilization of these structures compromises brain function and is a major contributing factor to psychiatric and neurodegenerative diseases. Integrins are a class of transmembrane extracellular matrix receptors that function as αβ heterodimers and activate signaling cascades regulating the actin cytoskeleton. Here we identify integrin α3 as a key mediator of neuronal stability. Dendrites, dendritic spines, and synapses develop normally in mice with selective loss of integrin α3 in excitatory forebrain neurons, reaching their mature sizes and densities through postnatal day 21 (P21). However, by P42, integrin α3 mutant mice exhibit significant reductions in hippocampal dendrite arbor size and complexity, loss of dendritic spine and synapse densities, and impairments in hippocampal-dependent behavior. Furthermore, gene-dosage experiments demonstrate that integrin α3 interacts functionally with the Arg nonreceptor tyrosine kinase to activate p190RhoGAP, which inhibits RhoA GTPase and regulates hippocampal dendrite and synapse stability and mouse behavior. Together, our data support a fundamental role for integrin α3 in regulating dendrite arbor stability, synapse maintenance, and proper hippocampal function. In addition, these results provide a biochemical and structural explanation for the defects in long-term potentiation, learning, and memory reported previously in mice lacking integrin α3.
Collapse
|
45
|
Abstract
Mechanical forces influence nearly all aspects of biology. Cells are equipped with numerous mechanosensitive proteins that activate various signaling cascades in response to mechanical cues from their environment. Much interest lies in understanding how cells respond to external stresses. A number of studies have highlighted the coordination of mechanical and chemical signaling cascades downstream of integrins. In recent years, the study of mechanotransduction has expanded to other mechanosensitive adhesion receptors, such as platelet endothelial cell adhesion molecule-1 (PECAM-1). This commentary will highlight our current understanding of integrin and PECAM-1-mediated mechanotransduction and expand on the observation that a localized mechanical stress can elicit a global mechanosignaling response.
Collapse
Affiliation(s)
- Caitlin Collins
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
46
|
Takeshita Y, Ransohoff RM. Inflammatory cell trafficking across the blood-brain barrier: chemokine regulation and in vitro models. Immunol Rev 2012; 248:228-39. [PMID: 22725965 DOI: 10.1111/j.1600-065x.2012.01127.x] [Citation(s) in RCA: 224] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The blood-brain barrier (BBB) is the brain-specific capillary barrier that is critical for preventing toxic substances from entering the central nervous system (CNS). In contrast to vessels of peripheral organs, the BBB limits the exchange of inflammatory cells and mediators under physiological and pathological conditions. Clarifying these limitations and the role of chemokines in regulating the BBB would provide new insights into neuroprotective strategies in neuroinflammatory diseases. Because there is a paucity of in vitro BBB models, however, some mechanistic aspects of transmigration across the BBB still remain largely unknown. In this review, we summarize current knowledge of BBB cellular components, the multistep process of inflammatory cells crossing the BBB, functions of CNS-derived chemokines, and in vitro BBB models for transmigration, with a particular focus on new and recent findings.
Collapse
Affiliation(s)
- Yukio Takeshita
- Neuroinflammation Research Center, Department of Neuroscience, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | |
Collapse
|
47
|
Parnell E, Smith BO, Palmer TM, Terrin A, Zaccolo M, Yarwood SJ. Regulation of the inflammatory response of vascular endothelial cells by EPAC1. Br J Pharmacol 2012; 166:434-46. [PMID: 22145651 DOI: 10.1111/j.1476-5381.2011.01808.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Life-threatening diseases of the cardiovascular system, like atherosclerosis, are exacerbated by unwanted inflammation within the structures of large blood vessels. This inflammation involves increased permeability of the vascular endothelial cells (VECs) that form the lining of blood vessels, leading to exaggerated extravasation of blood components and accumulation of fluid in the extravascular space. This results in tissue dysfunction and increased secretion of chemokines that attract leukocytes and monocytes to the inflamed endothelium. Cyclic AMP is synthesized in VECs in response to endogenous Gs-coupled receptors and is known to limit cytokine action and reduce endothelial hyperpermeability induced by multiple pro-inflammatory stimuli. The mechanisms underlying this anti-inflammatory action of cyclic AMP are now being elucidated and it is becoming clear that the cyclic AMP sensor, exchange protein activated by cyclic AMP (EPAC1), appears to play a key role in suppressing unwanted inflammation. EPAC1 mediates at least three anti-inflammatory pathways in VECs by down-regulating inflammatory signalling through the induction of the suppressors of cytokine signalling 3 (SOCS-3) gene, limiting integrin-dependent vascular permeability and enhancing endothelial barrier function through the stabilization of VE-cadherin junctions. Given that manipulation of cellular cyclic AMP levels currently forms the basis of many effective pharmaceuticals and that EPAC1 is involved in multiple anti-inflammatory protective processes in VECs, does this make EPAC1 an attractive target for the development of activators capable of eliciting a coordinated programme of 'protection' against the development of endothelial dysfunction? Here we discuss whether EPAC1 represents an attractive therapeutic target for limiting endothelial dysfunction associated with cardiovascular diseases like atherosclerosis. LINKED ARTICLES This article is part of a themed section on Novel cAMP Signalling Paradigms. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.166.issue-2.
Collapse
Affiliation(s)
- Euan Parnell
- Institute of Molecular, Cell and Systems Biology, University of Glasgow, Glasgow, UK
| | | | | | | | | | | |
Collapse
|
48
|
Draskovic-Pavlovic B, Vucevic D, Bozic B, Majstorovic I, Colic M. Functional properties of granulocytes after thermal injury. Immunol Res 2012; 52:133-8. [PMID: 22388640 DOI: 10.1007/s12026-012-8280-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Thermal injury, as well as other forms of severe trauma, induces simultaneous hyper- and anti-inflammatory response. While data about decreased number and responsiveness of T lymphocytes are largely consistent, reports concerning granulocytes following trauma are contradictory. Contrary to the evidence on the increased accumulation of granulocytes in the lungs or liver, the results from our laboratory demonstrated reduced granulocyte influx in the wound that heals in conditions of thermal injury. We also demonstrated evidence that indicates impaired signal transduction in granulocytes following thermal injury, as well as their divergent response regarding the adhesiveness, oxidative burst and nitric oxide production at the wound site.
Collapse
Affiliation(s)
- Biljana Draskovic-Pavlovic
- Medical Faculty of the Military Medical Academy, University of Defense in Belgrade, Crnotravska 17, 11002 Belgrade, Serbia
| | | | | | | | | |
Collapse
|
49
|
JAM-A protects from thrombosis by suppressing integrin αIIbβ3-dependent outside-in signaling in platelets. Blood 2012; 119:3352-60. [PMID: 22271446 DOI: 10.1182/blood-2011-12-397398] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mounting evidence suggests that agonist-initiated signaling in platelets is closely regulated to avoid excessive responses to injury. A variety of physiologic agonists induce a cascade of signaling events termed as inside-out signaling that culminate in exposure of high-affinity binding sites on integrin α(IIb)β(3). Once platelet activation has occurred, integrin α(IIb)β(3) stabilizes thrombus formation by providing agonist-independent "outside-in" signals mediated in part by contractile signaling. Junctional adhesion molecule A (JAM-A), a member of the cortical thymocyte marker of the Xenopus (CTX) family, was initially identified as a receptor for a platelet stimulatory mAb. Here we show that JAM-A in resting platelets functions as an endogenous inhibitor of platelet function. Genetic ablation of Jam-A in mice enhances thrombotic function of platelets in vivo. The absence of Jam-A results in increase in platelet aggregation ex vivo. This gain of function is not because of enhanced inside-out signaling because granular secretion, Thromboxane A2 (TxA2) generation, as well as fibrinogen receptor activation, are normal in the absence of Jam-A. Interestingly, integrin outside-in signaling such as platelet spreading and clot retraction is augmented in Jam-A-deficient platelets. We conclude that JAM-A normally limits platelet accumulation by inhibiting integrin outside-in signaling thus preventing premature platelet activation.
Collapse
|
50
|
Papst S, Noisier AFM, Brimble MA, Yang Y, Chan YC, Krissansen GW. Synthesis and SAR of a Library of Cell-Permeable Biotin-R8ERY* Peptidomimetics Inhibiting α4β7 Integrin Mediated Adhesion of TK-1 Cells to MAdCAM-1-Fc. Aust J Chem 2012. [DOI: 10.1071/ch12227] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The α4β7 integrin is a well‐known target for the development of drugs against various inflammatory disease states including inflammatory bowel disease, type 1 diabetes, and multiple sclerosis. The β7 subunit contains the cell adhesion regulatory domain (CARD) motif YDRREY within its cytoplasmic domain, which is an effective peptide agent for inhibiting T-cell adhesion. The synthesis of a library of cell-permeable β7 integrin inhibitors based on the shortened biotin-R8ERY (R8 = (l-arginine)8) motif is reported, wherein the tyrosine residue has been modified. The synthesised peptidomimetics were evaluated in a cell adhesion assay and shown to inhibit Mn2+-activated adhesion of mouse TK-1 T-cells to mouse MAdCAM-1. Several analogues exhibited improved activity to that of the tyrosine-containing lead compound 1 (biotin-R8ERY). Specifically, analogues 4, 10, and 22 bearing a 4-chloro, a 4-nitro, and a 3,3-diphenyl substituent showed an increase in activity of approximately two-fold compared with that of the initial lead compound. The six most active compounds of the tested series had IC50’s between 25 and 50 μM.
Collapse
|