101
|
Bonus M, Häussinger D, Gohlke H. Liver cell hydration and integrin signaling. Biol Chem 2021; 402:1033-1045. [PMID: 33915604 DOI: 10.1515/hsz-2021-0193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 04/12/2021] [Indexed: 12/21/2022]
Abstract
Liver cell hydration (cell volume) is dynamic and can change within minutes under the influence of hormones, nutrients, and oxidative stress. Such volume changes were identified as a novel and important modulator of cell function. It provides an early example for the interaction between a physical parameter (cell volume) on the one hand and metabolism, transport, and gene expression on the other. Such events involve mechanotransduction (osmosensing) which triggers signaling cascades towards liver function (osmosignaling). This article reviews our own work on this topic with emphasis on the role of β1 integrins as (osmo-)mechanosensors in the liver, but also on their role in bile acid signaling.
Collapse
Affiliation(s)
- Michele Bonus
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstr. 1, D-40225 Düsseldorf, Germany
| | - Dieter Häussinger
- Clinic for Gastroenterology, Hepatology, and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstr. 5, D-40225 Düsseldorf, Germany
| | - Holger Gohlke
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstr. 1, D-40225 Düsseldorf, Germany
- John von Neumann Institute for Computing (NIC), Jülich Supercomputing Centre (JSC), Wilhelm-Johnen-Str., D-52428 Jülich, Germany
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), and Institute of Bio- and Geosciences (IBG-4: Bioinformatics), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Str., D-52428 Jülich, Germany
| |
Collapse
|
102
|
Zheng S, Liu Q, He J, Wang X, Ye K, Wang X, Yan C, Liu P, Ding J. Critical adhesion areas of cells on micro-nanopatterns. NANO RESEARCH 2021; 15:1623-1635. [PMID: 34405038 PMCID: PMC8359768 DOI: 10.1007/s12274-021-3711-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/19/2021] [Accepted: 06/24/2021] [Indexed: 06/13/2023]
Abstract
UNLABELLED Cell adhesion to extracellular matrices (ECM) is critical to physiological and pathological processes as well as biomedical and biotechnological applications. It has been known that a cell can adhere on an adhesive microisland only over a critical size. But no publication has concerned critical adhesion areas of cells on microislands with nanoarray decoration. Herein, we fabricated a series of micro-nanopatterns with different microisland sizes and arginine-glycine-aspartate (RGD) nanospacings on a nonfouling poly(ethylene glycol) background. Besides reproducing that nanospacing of RGD, a ligand of its receptor integrin (a membrane protein), significantly influences specific cell adhesion on bioactive nanoarrays, we confirmed that the concept of critical adhesion area originally suggested in studies of cells on micropatterns was justified also on the micro-nanopatterns, yet the latter exhibited more characteristic behaviors of cell adhesion. We found increased critical adhesion areas of human mesenchymal stem cells (hMSCs) on nanoarrayed microislands with increased RGD nanospacings. However, the numbers of nanodots with respect to the critical adhesion areas were not a constant. A unified interpretation was then put forward after combining nonspecific background adhesion and specific cell adhesion. We further carried out the asymptotic analysis of a series of micro-nanopatterned surfaces to obtain the effective RGD nanospacing on unpatterned free surfaces with densely grafted RGD, which could be estimated nonzero but has never been revealed previously without the assistance of the micro-nanopatterning techniques and the corresponding analysis. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary materials and methods (details of fabrication of micro-nanopatterns), and supplementary results (selective adhesion or localization of hMSCs on nanoarrayed microislands with non-fouling background, calculation of critical number of integrin-ligand binding N*, etc.) are available in the online version of this article at 10.1007/s12274-021-3711-6.
Collapse
Affiliation(s)
- Shuang Zheng
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 China
| | - Qiong Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 China
- Navy Characteristic Medical Center, the Second Military Medical University, Shanghai, 200433 China
| | - Junhao He
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 China
| | - Xinlei Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 China
| | - Kai Ye
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 China
| | - Xuan Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 China
| | - Ce Yan
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 China
| | - Peng Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 China
- College of Bioengineering, Chongqing University, Chongqing, 400044 China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University, Shanghai, 200438 China
| |
Collapse
|
103
|
Manganese systemic distribution is modulated in vivo during tumor progression and affects tumor cell migration and invasion in vitro. Sci Rep 2021; 11:15833. [PMID: 34349175 PMCID: PMC8338990 DOI: 10.1038/s41598-021-95190-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/22/2021] [Indexed: 11/23/2022] Open
Abstract
Metastatic disease remains the leading cause of death in cancer and understanding the mechanisms involved in tumor progression continues to be challenging. This work investigates the role of manganese in tumor progression in an in vivo model of tumor growth. Our data revealed that manganese accumulates within primary tumors and secondary organs as manganese-rich niches. Consequences of such phenomenon were investigated, and we verified that short-term changes in manganese alter cell surface molecules syndecan-1 and β1-integrin, enhance collective cell migration and invasive behavior. Long-term increased levels of manganese do not affect cell growth and viability but enhance cell migration. We also observed that manganese is secreted from tumor cells in extracellular vesicles, rather than in soluble form. Finally, we describe exogenous glycosaminoglycans that counteract manganese effects on tumor cell behavior. In conclusion, our analyses describe manganese as a central element in tumor progression by accumulating in Mn-rich niches in vivo, as well as in vitro, affecting migration and extracellular vesicle secretion in vitro. Manganese accumulation in specific regions of the organism may not be a common ground for all cancers, nevertheless, it represents a new aspect of tumor progression that deserves special attention.
Collapse
|
104
|
Zhu YS, Tang K, Lv J. Peptide-drug conjugate-based novel molecular drug delivery system in cancer. Trends Pharmacol Sci 2021; 42:857-869. [PMID: 34334251 DOI: 10.1016/j.tips.2021.07.001] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/25/2021] [Accepted: 07/06/2021] [Indexed: 01/18/2023]
Abstract
Drug delivery systems are generally believed to comprise drugs and excipients. A peptide-drug conjugate is a single molecule that can simultaneously play multiple roles in a drug delivery system, such as in vivo drug distribution, targeted release, and bioactivity functions. This molecule can be regarded as an integrated drug delivery system, so it is called a molecular drug delivery system. In the context of cancer therapy, a peptide-drug conjugate comprises a tumor-targeting peptide, a payload, and a linker. Tumor-targeting peptides specifically identify membrane receptors on tumor cells, improve drug-targeted therapeutic effects, and reduce toxic and side effects. Payloads with bioactive functions connect to tumor-targeting peptides through linkers. In this review, we explored ongoing clinical work on peptide-drug conjugates targeting various receptors. We discuss the binding mechanisms of tumor-targeting peptides and related receptors, as well as the limiting factors for peptide-drug conjugate-based molecular drug delivery systems.
Collapse
Affiliation(s)
- Yi-Shen Zhu
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, Jiangsu Province, China.
| | - Kexing Tang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, Jiangsu Province, China
| | - Jiayi Lv
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, Jiangsu Province, China
| |
Collapse
|
105
|
Comparison of Linear vs. Cyclic RGD Pentapeptide Interactions with Integrin α vβ 3 by Molecular Dynamics Simulations. BIOLOGY 2021; 10:biology10070688. [PMID: 34356543 PMCID: PMC8301328 DOI: 10.3390/biology10070688] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/13/2021] [Accepted: 07/16/2021] [Indexed: 12/18/2022]
Abstract
Simple Summary The integrin αvβ3-RGD motif interaction plays a key role in the progression of malignant tumor. Although two typical cyclic and linear RGD short peptides have been widely used in tumor diagnosis and therapy, little is known about the internal dynamic mechanism for different configurations of RGD peptides with different affinities interacting with the integrin αvβ3. Our results showed that the cyclic RGD peptide had a more stable configuration in binding to integrins αvβ3, which depended on the higher binding energy and higher static electrical energy, especially in the interaction between AspRGD-MIDAS. The steered molecular dynamics simulation showed a stronger interaction for the cyclic RGD-integrin αvβ3 system than the linear one, with a larger dissociation force (average peak force) and more time to dissociate. Our findings provide insights into the dynamics of integrin αvβ3 interactions with linear and cyclic RGD ligands and offer some new therapeutic approaches for the design and development of novel antitumor drugs. Abstract Integrin αvβ3 interacting with the short Arg-Gly-Asp (RGD) motif plays a critical role in the progression of several types of tumors. However, the effects of the RGD structure (cyclic or linear) with integrin αvβ3 at the atomic level remain poorly understood. Here, we performed association and dissociation dynamic simulations for integrin αvβ3 in complex with a linear or cyclic pentapeptide by steered molecular dynamics simulations. Compared with cyclic RGD, the linear RGD peptide triggers instability of the configurational changes, mainly resting with the RGD domain due to its flexibility. The main interaction energy between Mg2+ and cyclic RGD is much stronger than that of the linear RGD system by the well shield to lessen attacks by free water molecules. The force-dependent dissociation results show that it is easier for linear RGD peptides to leave the active site and much quicker than the cyclic RGD ligand, whereas it is harder to enter the appropriate active binding site in linear RGD. The Ser123-AspRGD bond may play a critical role in the allosteric pathway. Our findings provide insights into the dynamics of αvβ3 interactions with linear and cyclic RGD ligands and contribute to the application of RGD-based strategies in preclinical therapy.
Collapse
|
106
|
Arimori T, Miyazaki N, Mihara E, Takizawa M, Taniguchi Y, Cabañas C, Sekiguchi K, Takagi J. Structural mechanism of laminin recognition by integrin. Nat Commun 2021; 12:4012. [PMID: 34188035 PMCID: PMC8241838 DOI: 10.1038/s41467-021-24184-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 06/04/2021] [Indexed: 02/06/2023] Open
Abstract
Recognition of laminin by integrin receptors is central to the epithelial cell adhesion to basement membrane, but the structural background of this molecular interaction remained elusive. Here, we report the structures of the prototypic laminin receptor α6β1 integrin alone and in complex with three-chain laminin-511 fragment determined via crystallography and cryo-electron microscopy, respectively. The laminin-integrin interface is made up of several binding sites located on all five subunits, with the laminin γ1 chain C-terminal portion providing focal interaction using two carboxylate anchor points to bridge metal-ion dependent adhesion site of integrin β1 subunit and Asn189 of integrin α6 subunit. Laminin α5 chain also contributes to the affinity and specificity by making electrostatic interactions with large surface on the β-propeller domain of α6, part of which comprises an alternatively spliced X1 region. The propeller sheet corresponding to this region shows unusually high mobility, suggesting its unique role in ligand capture.
Collapse
Affiliation(s)
- Takao Arimori
- grid.136593.b0000 0004 0373 3971Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| | - Naoyuki Miyazaki
- grid.136593.b0000 0004 0373 3971Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka Japan ,grid.20515.330000 0001 2369 4728Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Ibaraki, Japan
| | - Emiko Mihara
- grid.136593.b0000 0004 0373 3971Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| | - Mamoru Takizawa
- grid.136593.b0000 0004 0373 3971Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| | - Yukimasa Taniguchi
- grid.136593.b0000 0004 0373 3971Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| | - Carlos Cabañas
- grid.465524.4Cell-cell Communication & Inflammation Unit, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain ,grid.4795.f0000 0001 2157 7667Department of Immunology, Ophthalmology and Otorhinolaryngology (IOO), Faculty of Medicine, Universidad Complutense de Madrid, Madrid, Spain ,grid.144756.50000 0001 1945 5329Instituto de Investigación Sanitaria Hospital 12 Octubre (i+12), Madrid, Spain
| | - Kiyotoshi Sekiguchi
- grid.136593.b0000 0004 0373 3971Division of Matrixome Research and Application, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| | - Junichi Takagi
- grid.136593.b0000 0004 0373 3971Laboratory for Protein Synthesis and Expression, Institute for Protein Research, Osaka University, Suita, Osaka Japan
| |
Collapse
|
107
|
Sun T, Wei L, Tian H, Zhan W, Ma H, Nie D, Wang S, Chen X, Tang G. Novel PET/CT tracers for targeted imaging of membrane receptors to evaluate cardiomyocyte apoptosis and tissue repair process in a rat model of myocardial infarction. Apoptosis 2021; 26:460-473. [PMID: 34185202 DOI: 10.1007/s10495-021-01681-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2021] [Indexed: 12/12/2022]
Abstract
The purpose of this study was to employ novel tracers PET imaging approach to define the time course and intensity of myocardial repair after apoptosis and to correlate the imaging signal to immunohistochemical staining in myocardial infarction (MI). We designed novel αVβ3-targeted and radio-functionalized tracers for detection of apoptosis in H9C2 cells and myocardial tissue. MI rats were imaged with [18F]FDG, [18F]ANP-Cin or [18F]ANP-RGD2 using a small-animal PET/CT device. Rats were sacrificed, and tissue samples from viable and injured myocardial areas were sectioned for TUNEL assay and histology. The uncorrected radiochemical yield of [18F]ANP-Cin and [18F]ANP-RGD2 were 41.3 ± 5.4% and 21.17 ± 4.7%, respectively. Two tracers meet many criteria for cardiac imaging, including high stability, high binding, no toxicity, fast renal clearance and excellent biodistribution in rat models. The uptake of [18F]ANP-Cin was significantly higher on the 1st and 3rd day than the 7th or 28th day after MI induction, a timeframe associated with increased cardiomyocyte apoptosis. Higher uptake of [18F]ANP-Cin was observed in MI rats than in N-acetylcysteine (NAC)-treated rats on the 3rd days. In contrast with [18F]ANP-Cin, no hot-spots was observed with [18F]ANP-RGD2 on the 1st day and more hot-spots was observed from the 3rd day to the 7th day, then less on the 28th days in the high apoptotic site. There was no uptake of [18F]FDG in or around the apoptotic region. On the 7th day the uptake of [18F]ANP-RGD2 was higher in NAC-treated rats than MI rats. [18F]ANP-Cin and [18F]ANP-RGD2 are superior to [18F]FDG for PET/CT imaging for evaluation of cardiomyocyte apoptosis and tissue repair processes in the MI rats.
Collapse
Affiliation(s)
- Ting Sun
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Lijiang Wei
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.,Nanfang PET Center and Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hua Tian
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200032, China.
| | - Wanlin Zhan
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Hui Ma
- Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Dahong Nie
- Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Shilin Wang
- Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xin Chen
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ganghua Tang
- Nanfang PET Center and Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China. .,Department of Radiotherapy and Medical Imaging, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
108
|
Ozsvar J, Wang R, Tarakanova A, Buehler MJ, Weiss AS. Fuzzy binding model of molecular interactions between tropoelastin and integrin alphaVbeta3. Biophys J 2021; 120:3138-3151. [PMID: 34197806 DOI: 10.1016/j.bpj.2021.04.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 03/30/2021] [Accepted: 04/12/2021] [Indexed: 12/30/2022] Open
Abstract
Tropoelastin is the highly flexible monomer subunit of elastin, required for the resilience of the extracellular matrix in elastic tissues. To elicit biological signaling, multiple sites on tropoelastin bind to cell surface integrins in a poorly understood multifactorial process. We constructed a full atomistic molecular model of the interactions between tropoelastin and integrin αvβ3 using ensemble-based computational methodologies. Conformational changes of integrin αvβ3 associated with outside-in signaling were more frequently facilitated in an ensemble in which tropoelastin bound the integrin's α1 helix rather than the upstream canonical binding site. Our findings support a model of fuzzy binding, whereby many tropoelastin conformations and defined sites cooperatively interact with multiple αvβ3 regions. This model explains prior experimental binding to distinct tropoelastin regions, domains 17 and 36, and points to the cooperative participation of domain 20. Our study highlights the utility of ensemble-based approaches in helping to understand the interactive mechanisms of functionally significant flexible proteins.
Collapse
Affiliation(s)
- Jazmin Ozsvar
- Charles Perkins Centre, The University of Sydney, Sydney, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Richard Wang
- Charles Perkins Centre, The University of Sydney, Sydney, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Anna Tarakanova
- Department of Biomedical Engineering, University of Connecticut, Storrs, Connecticut; Department of Mechanical Engineering, University of Connecticut, Storrs, Connecticut
| | - Markus J Buehler
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Anthony S Weiss
- Charles Perkins Centre, The University of Sydney, Sydney, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia; Sydney Nano Institute, The University of Sydney, Sydney, Australia.
| |
Collapse
|
109
|
Nader D, Fletcher N, Curley GF, Kerrigan SW. SARS-CoV-2 uses major endothelial integrin αvβ3 to cause vascular dysregulation in-vitro during COVID-19. PLoS One 2021; 16:e0253347. [PMID: 34161337 PMCID: PMC8221465 DOI: 10.1371/journal.pone.0253347] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 06/02/2021] [Indexed: 12/14/2022] Open
Abstract
The unprecedented global COVID-19 pandemic has prompted a desperate international effort to accelerate the development of anti-viral candidates. For unknown reasons, COVID-19 infections are associated with adverse cardiovascular complications, implicating that vascular endothelial cells are essential in viral propagation. The etiological pathogen, SARS-CoV-2, has a higher reproductive number and infection rate than its predecessors, indicating it possesses novel characteristics that infers enhanced transmissibility. A unique K403R spike protein substitution encodes an Arg-Gly-Asp (RGD) motif, introducing a potential role for RGD-binding host integrins. Integrin αVβ3 is widely expressed across the host, particularly in the endothelium, which acts as the final barrier before microbial entry into the bloodstream. This mutagenesis creates an additional binding site, which may be sufficient to increase SARS-CoV-2 pathogenicity. Here, we investigate how SARS-CoV-2 passes from the epithelium to endothelium, the effects of αVβ3 antagonist, Cilengitide, on viral adhesion, vasculature permeability and leakage, and also report on a simulated interaction between the viral and host protein in-silico.
Collapse
Affiliation(s)
- Danielle Nader
- Cardiovascular Infection Research Group, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Nicola Fletcher
- School of Veterinary Medicine, Veterinary Science Centre, University College Dublin, Belfield, Dublin 4, Ireland
| | - Gerard F. Curley
- Department of Anaesthesia and Critical Care Medicine, RCSI University of Medicine and Health Sciences, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin, Ireland
| | - Steven W. Kerrigan
- Cardiovascular Infection Research Group, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- * E-mail:
| |
Collapse
|
110
|
Liu J, Wu L, Liu Z, Seery S, Li J, Gao Z, Yu J, Meng X. 18F-RGD PET/CT and Systemic Inflammatory Biomarkers Predict Outcomes of Patients With Advanced NSCLC Receiving Combined Antiangiogenic Treatment. Front Oncol 2021; 11:671912. [PMID: 34150635 PMCID: PMC8212050 DOI: 10.3389/fonc.2021.671912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/12/2021] [Indexed: 12/09/2022] Open
Abstract
Background The aim of this study was to evaluate 18F-AlF-NOTA-PRGD2 positron emission tomography/computed tomography (18F-RGD PET/CT) and serum inflammation biomarkers for predicting outcomes of patients receiving combined antiangiogenic treatment for advanced non-small cell lung cancer (NSCLC). Methods Patients with advanced NSCLC underwent 18F-RGD PET/CT examination and provided blood samples before treatments commenced. PET/CT parameters included maximum standard uptake value (SUVmax) and mean standard uptake value (SUVmean), peak standard uptake value (SUVpeak) and metabolic tumor volume (MTV) for all contoured lesions. Biomarkers for inflammation included pretreatment neutrophil-to-lymphocyte ratio (PreNLR), pretreatment platelet-to-lymphocyte ratio (PrePLR), and pretreatment lymphocyte-to-monocyte ratio (PreLMR). Receiver operating characteristic (ROC) curve analysis was used to describe response prediction accuracy. Logistic regression and Cox’s regression analysis was implemented to identify independent factors for short-term responses and progression-free survival (PFS). Results This study included 23 patients. According to ROC curve analysis, there were significant correlations between the SUVmax, SUVmean, and 18F-RGD PET/CT MTV and short-term responses (p<0.05). SUVmax was identified using logistic regression analysis as a significant predictor of treatment sensitivity (p=0.008). Cox’s multivariate regression analysis suggested that high SUVpeak (p=0.021) and high PreLMR (p=0.03) were independent PFS predictors. Combining SUVpeak and PreLMR may also increase the prognostic value for PFS, enabling us to identify a subgroup of patients with intermediate PFS. Conclusion 18F-RGD uptake on PET/CT and serum inflammation biomarker pretreatment may predict outcomes for combined antiangiogenic treatments for advanced NSCLC patients. Higher 18F-RGD uptake and higher PreLMR also appear to predict improved short-term responses and PFS. Combining biomarkers may therefore provide a basis for risk stratification, although further research is required.
Collapse
Affiliation(s)
- Jie Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Leilei Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Department of Medical Oncology, Cancer Center, Qilu Hospital of Shandong University, Jinan, China
| | - Zhiguo Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Samuel Seery
- Department of Humanities and Social Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Faculty of Health and Medicine, Division of Health Research, Lancaster University, Lancaster, United Kingdom
| | - Jianing Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhenhua Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xue Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
111
|
Farina B, Andrea C, Del Gatto A, Comegna D, Di Gaetano S, Capasso D, Paladino A, Acconcia C, Teresa Gentile M, Saviano M, Fattorusso R, Zaccaro L, Russo L. A novel approach for studying receptor-ligand interactions on living cells surface by using NUS/T1ρ-NMR methodologies combined with computational techniques: The RGDechi15D-α vβ 5 integrin complex. Comput Struct Biotechnol J 2021; 19:3303-3318. [PMID: 34188779 PMCID: PMC8207173 DOI: 10.1016/j.csbj.2021.05.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 05/22/2021] [Accepted: 05/27/2021] [Indexed: 11/30/2022] Open
Abstract
Structural investigations of receptor-ligand interactions on living cells surface by high-resolution Nuclear Magnetic Resonance (NMR) are problematic due to their short lifetime, which often prevents the acquisition of experiments longer than few hours. To overcome these limitations, we developed an on-cell NMR-based approach for exploring the molecular determinants driving the receptor-ligand recognition mechanism under native conditions. Our method relies on the combination of high-resolution structural and dynamics NMR data with Molecular Dynamics simulations and Molecular Docking studies. The key point of our strategy is the use of Non Uniform Sampling (NUS) and T1ρ-NMR techniques to collect atomic-resolution structural and dynamics information on the receptor-ligand interactions with living cells, that can be used as conformational constraints in computational studies. In fact, the application of these two NMR methodologies allows to record spectra with high S/N ratio and resolution within the lifetime of cells. In particular, 2D NUS [1H–1H] trNOESY spectra are used to explore the ligand conformational changes induced by receptor binding; whereas T1ρ-based experiments are applied to characterize the ligand binding epitope by defining two parameters: T1ρ Attenuation factor and T1ρ Binding Effect. This approach has been tested to characterize the molecular determinants regulating the recognition mechanism of αvβ5-integrin by a selective cyclic binder peptide named RGDechi15D. Our data demonstrate that the developed strategy represents an alternative in-cell NMR tool for studying, at atomic resolution, receptor-ligand recognition mechanism on living cells surface. Additionally, our application may be extremely useful for screening of the interaction profiling of drugs with their therapeutic targets in their native cellular environment.
Collapse
Affiliation(s)
- Biancamaria Farina
- Institute of Biostructures and Bioimaging-CNR, Via Mezzocannone 16, 80134 Naples, Italy.,Advanced Accelerator Applications, a Novartis Company, via Vivaldi 43, 81100 Caserta, Italy
| | - Corvino Andrea
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania - Luigi Vanvitelli, via Vivaldi 43, 81100 Caserta, Italy
| | - Annarita Del Gatto
- Institute of Biostructures and Bioimaging-CNR, Via Mezzocannone 16, 80134 Naples, Italy.,Interdepartmental Center of Bioactive Peptide, University of Naples Federico II, Via Mezzocannone 16, 80134 Naples, Italy
| | - Daniela Comegna
- Institute of Biostructures and Bioimaging-CNR, Via Mezzocannone 16, 80134 Naples, Italy
| | - Sonia Di Gaetano
- Institute of Biostructures and Bioimaging-CNR, Via Mezzocannone 16, 80134 Naples, Italy.,Interdepartmental Center of Bioactive Peptide, University of Naples Federico II, Via Mezzocannone 16, 80134 Naples, Italy
| | - Domenica Capasso
- Interdepartmental Center of Bioactive Peptide, University of Naples Federico II, Via Mezzocannone 16, 80134 Naples, Italy.,Center for Life Sciences and Technologies (CESTEV) University of Naples Federico II, Via Tommaso De Amicis 95, 80145 Naples, Italy
| | - Antonella Paladino
- Department of Science and Technology, University of Sannio, via Francesco de Sanctis, Benevento 82100, Italy
| | - Clementina Acconcia
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania - Luigi Vanvitelli, via Vivaldi 43, 81100 Caserta, Italy
| | - Maria Teresa Gentile
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania - Luigi Vanvitelli, via Vivaldi 43, 81100 Caserta, Italy
| | - Michele Saviano
- Institute of Crystallography-CNR, Via Amendola 122/O, 70126 Bari, Italy
| | - Roberto Fattorusso
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania - Luigi Vanvitelli, via Vivaldi 43, 81100 Caserta, Italy.,Interdepartmental Center of Bioactive Peptide, University of Naples Federico II, Via Mezzocannone 16, 80134 Naples, Italy
| | - Laura Zaccaro
- Institute of Biostructures and Bioimaging-CNR, Via Mezzocannone 16, 80134 Naples, Italy.,Interdepartmental Center of Bioactive Peptide, University of Naples Federico II, Via Mezzocannone 16, 80134 Naples, Italy
| | - Luigi Russo
- Department of Environmental, Biological and Pharmaceutical Science and Technology, University of Campania - Luigi Vanvitelli, via Vivaldi 43, 81100 Caserta, Italy
| |
Collapse
|
112
|
Schumacher S, Vazquez Nunez R, Biertümpfel C, Mizuno N. Bottom-up reconstitution of focal adhesion complexes. FEBS J 2021; 289:3360-3373. [PMID: 33999507 PMCID: PMC9290908 DOI: 10.1111/febs.16023] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/13/2021] [Accepted: 05/14/2021] [Indexed: 12/28/2022]
Abstract
Focal adhesions (FA) are large macromolecular assemblies relevant for various cellular and pathological events such as migration, polarization, and metastatic cancer formation. At FA sites at the migrating periphery of a cell, hundreds of players gather and form a network to respond to extra cellular stimuli transmitted by the integrin receptor, the most upstream component within a cell, initiating the FA signaling pathway. Numerous cellular experiments have been performed to understand the FA architecture and functions; however, their intricate network formation hampers unraveling the precise molecular actions of individual players. Here, in vitro bottom‐up reconstitution presents an advantageous approach for elucidating the FA machinery and the hierarchical crosstalk of involved cellular players.
Collapse
Affiliation(s)
- Stephanie Schumacher
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Roberto Vazquez Nunez
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christian Biertümpfel
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Naoko Mizuno
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.,National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
113
|
Goyal R, Jerath G, Akhil R, Chandrasekharan A, Puppala ER, Ponneganti S, Sarma A, Naidu VGM, Santhoshkumar TR, Ramakrishnan V. Geometry encoded functional programming of tumor homing peptides for targeted drug delivery. J Control Release 2021; 333:16-27. [PMID: 33722612 DOI: 10.1016/j.jconrel.2021.03.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/24/2021] [Accepted: 03/07/2021] [Indexed: 01/10/2023]
Abstract
Poly-peptide molecules have shown promising applications in drug delivery and tumor targeting. A series of tumor homing peptides were designed by exhaustively sampling low energy geometrical basins of amino acids at specific sites of a peptide molecule to induce a conformational lock. This peptide library was pruned to a limited set of eight molecules, employing electrostatic interactions, docking, and molecular dynamics simulations. These designed and optimized peptides were synthesized and tested on various cell lines, including breast cancer (MDA-MB-231), cervical cancer (HeLa), osteosarcoma (U2-OS), and non-cancerous mammary epithelial cells (MCF-10A) using confocal microscopy and flow cytometry. Peptides show differential uptake in cancerous MDA-MB-231, HeLa, U2-OS, and non-cancerous MCF-10A cells. Confocal imaging verified their ability to penetrate even in 3D tumorospheres of MDA-MB-231 cells. Further, experiments of mitochondrial membrane potential depolarization and Caspase-3 activation confirmed that their cytotoxic effects are by apoptosis. Homing ability of the designed peptides in in vivo system and fluorescence imaging with clinical samples of human origin have further confirmed that the in vitro studies are qualitatively identical and quantitatively comparable in their ability to selectively recognize tumor cells. Overall, we present a roadmap for the functional programming of peptide-based homing and penetrating molecules that can perform selective tumor targeting.
Collapse
Affiliation(s)
- Ruchika Goyal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Gaurav Jerath
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - R Akhil
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Aneesh Chandrasekharan
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Eswara Rao Puppala
- National Institute of Pharmaceutical Education and Research Guwahati, Guwahati 781101, Assam, India
| | - Srikanth Ponneganti
- National Institute of Pharmaceutical Education and Research Guwahati, Guwahati 781101, Assam, India
| | - Anupam Sarma
- Dr. Bhubaneswar Borooah Cancer Institute, Tata Memorial Centre (Mumbai), Guwahati 781016, Assam, India
| | - V G M Naidu
- National Institute of Pharmaceutical Education and Research Guwahati, Guwahati 781101, Assam, India
| | - T R Santhoshkumar
- Cancer Research Program-1, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.
| | - Vibin Ramakrishnan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
114
|
Schumacher S, Dedden D, Nunez RV, Matoba K, Takagi J, Biertümpfel C, Mizuno N. Structural insights into integrin α 5β 1 opening by fibronectin ligand. SCIENCE ADVANCES 2021; 7:7/19/eabe9716. [PMID: 33962943 PMCID: PMC8104898 DOI: 10.1126/sciadv.abe9716] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 03/19/2021] [Indexed: 05/08/2023]
Abstract
Integrin α5β1 is a major fibronectin receptor critical for cell migration. Upon complex formation, fibronectin and α5β1 undergo conformational changes. While this is key for cell-tissue connections, its mechanism is unknown. Here, we report cryo-electron microscopy structures of native human α5β1 with fibronectin to 3.1-angstrom resolution, and in its resting state to 4.6-angstrom resolution. The α5β1-fibronectin complex revealed simultaneous interactions at the arginine-glycine-aspartate loop, the synergy site, and a newly identified binding site proximal to adjacent to metal ion-dependent adhesion site, inducing the translocation of helix α1 to secure integrin opening. Resting α5β1 adopts an incompletely bent conformation, challenging the model of integrin sharp bending inhibiting ligand binding. Our biochemical and structural analyses showed that affinity of α5β1 for fibronectin is increased with manganese ions (Mn2+) while adopting the half-bent conformation, indicating that ligand-binding affinity does not depend on conformation, and α5β1 opening is induced by ligand-binding.
Collapse
Affiliation(s)
- Stephanie Schumacher
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, D-82152 Martinsried, Germany
| | - Dirk Dedden
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, D-82152 Martinsried, Germany
| | - Roberto Vazquez Nunez
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Dr., Bethesda, MD 20892, USA
| | - Kyoko Matoba
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Junichi Takagi
- Institute for Protein Research, Osaka University, 3-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Christian Biertümpfel
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Dr., Bethesda, MD 20892, USA.
| | - Naoko Mizuno
- Department of Structural Cell Biology, Max Planck Institute of Biochemistry, Am Klopferspitz 18, D-82152 Martinsried, Germany.
- Laboratory of Structural Cell Biology, National Heart, Lung, and Blood Institute, National Institutes of Health, 50 South Dr., Bethesda, MD 20892, USA
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, 50 South Dr., Bethesda, MD 20892, USA
| |
Collapse
|
115
|
Tomas NM, Mortensen SA, Wilmanns M, Huber TB. Across scales: novel insights into kidney health and disease by structural biology. Kidney Int 2021; 100:281-288. [PMID: 33940110 DOI: 10.1016/j.kint.2021.03.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/16/2021] [Accepted: 03/25/2021] [Indexed: 11/25/2022]
Abstract
Over the past decades, structural biology methods such as X-ray crystallography and cryo-electron microscopy have been increasingly used to study protein functions, molecular interactions, physiological processes, and disease mechanisms. This review outlines a selection of structural biology methods, highlights recent examples of how structural analyses have contributed to a more profound understanding of the machinery of life, and gives a perspective on how these methods can be applied to investigate functions of kidney molecules and pathogenic mechanisms of renal diseases.
Collapse
Affiliation(s)
- Nicola M Tomas
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simon A Mortensen
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany
| | - Matthias Wilmanns
- European Molecular Biology Laboratory, Hamburg Unit, Hamburg, Germany; University Hamburg Clinical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
116
|
Candelotti E, De Luca R, Megna R, Maiolo M, De Vito P, Gionfra F, Percario ZA, Borgatti M, Gambari R, Davis PJ, Lin HY, Polticelli F, Persichini T, Colasanti M, Affabris E, Pedersen JZ, Incerpi S. Inhibition by Thyroid Hormones of Cell Migration Activated by IGF-1 and MCP-1 in THP-1 Monocytes: Focus on Signal Transduction Events Proximal to Integrin αvβ3. Front Cell Dev Biol 2021; 9:651492. [PMID: 33898447 PMCID: PMC8060509 DOI: 10.3389/fcell.2021.651492] [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: 01/10/2021] [Accepted: 03/04/2021] [Indexed: 02/04/2023] Open
Abstract
Interaction between thyroid hormones and the immune system is reported in the literature. Thyroid hormones, thyroxine, T4, but also T3, act non-genomically through mechanisms that involve a plasma membrane receptor αvβ3 integrin, a co-receptor for insulin-like growth factor-1 (IGF-1). Previous data from our laboratory show a crosstalk between thyroid hormones and IGF-1 because thyroid hormones inhibit the IGF-1-stimulated glucose uptake and cell proliferation in L-6 myoblasts, and the effects are mediated by integrin αvβ3. IGF-1 also behaves as a chemokine, being an important factor for tissue regeneration after damage. In the present study, using THP-1 human leukemic monocytes, expressing αvβ3 integrin in their cell membrane, we focused on the crosstalk between thyroid hormones and either IGF-1 or monocyte chemoattractant protein-1 (MCP-1), studying cell migration and proliferation stimulated by the two chemokines, and the role of αvβ3 integrin, using inhibitors of αvβ3 integrin and downstream pathways. Our results show that IGF-1 is a potent chemoattractant in THP-1 monocytes, stimulating cell migration, and thyroid hormone inhibits the effect through αvβ3 integrin. Thyroid hormone also inhibits IGF-1-stimulated cell proliferation through αvβ3 integrin, an example of a crosstalk between genomic and non-genomic effects. We also studied the effects of thyroid hormone on cell migration and proliferation induced by MCP-1, together with the pathways involved, by a pharmacological approach and docking simulation. Our findings show a different downstream signaling for IGF-1 and MCP-1 in THP-1 monocytes mediated by the plasma membrane receptor of thyroid hormones, integrin αvβ3.
Collapse
Affiliation(s)
| | - Roberto De Luca
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Roberto Megna
- Department of Science, Roma Tre University, Rome, Italy
| | | | - Paolo De Vito
- Department of Biology, Tor Vergata University, Rome, Italy
| | - Fabio Gionfra
- Department of Science, Roma Tre University, Rome, Italy
| | | | - Monica Borgatti
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Paul J Davis
- Department of Medicine, Albany Medical College, Albany, NY, United States.,Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, United States
| | - Hung-Yun Lin
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY, United States.,Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan.,Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
117
|
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers (Basel) 2021; 13:1711. [PMID: 33916607 PMCID: PMC8038522 DOI: 10.3390/cancers13071711] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Integrins have been extensively investigated as therapeutic targets over the last decades, which has been inspired by their multiple functions in cancer progression, metastasis, and angiogenesis as well as a continuously expanding number of other diseases, e.g., sepsis, fibrosis, and viral infections, possibly also Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Although integrin-targeted (cancer) therapy trials did not meet the high expectations yet, integrins are still valid and promising targets due to their elevated expression and surface accessibility on diseased cells. Thus, for the future successful clinical translation of integrin-targeted compounds, revisited and innovative treatment strategies have to be explored based on accumulated knowledge of integrin biology. For this, refined approaches are demanded aiming at alternative and improved preclinical models, optimized selectivity and pharmacological properties of integrin ligands, as well as more sophisticated treatment protocols considering dose fine-tuning of compounds. Moreover, integrin ligands exert high accuracy in disease monitoring as diagnostic molecular imaging tools, enabling patient selection for individualized integrin-targeted therapy. The present review comprehensively analyzes the state-of-the-art knowledge on the roles of RGD-binding integrin subtypes in cancer and non-cancerous diseases and outlines the latest achievements in the design and development of synthetic ligands and their application in biomedical, translational, and molecular imaging approaches. Indeed, substantial progress has already been made, including advanced ligand designs, numerous elaborated pre-clinical and first-in-human studies, while the discovery of novel applications for integrin ligands remains to be explored.
Collapse
Affiliation(s)
- Beatrice S. Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, University Hospital Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
118
|
Ayo A, Laakkonen P. Peptide-Based Strategies for Targeted Tumor Treatment and Imaging. Pharmaceutics 2021; 13:pharmaceutics13040481. [PMID: 33918106 PMCID: PMC8065807 DOI: 10.3390/pharmaceutics13040481] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 02/03/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide. The development of cancer-specific diagnostic agents and anticancer toxins would improve patient survival. The current and standard types of medical care for cancer patients, including surgery, radiotherapy, and chemotherapy, are not able to treat all cancers. A new treatment strategy utilizing tumor targeting peptides to selectively deliver drugs or applicable active agents to solid tumors is becoming a promising approach. In this review, we discuss the different tumor-homing peptides discovered through combinatorial library screening, as well as native active peptides. The different structure–function relationship data that have been used to improve the peptide’s activity and conjugation strategies are highlighted.
Collapse
Affiliation(s)
- Abiodun Ayo
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
| | - Pirjo Laakkonen
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland;
- Laboratory Animal Center, HiLIFE—Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-50-4489100
| |
Collapse
|
119
|
Sun H, Zhi K, Hu L, Fan Z. The Activation and Regulation of β2 Integrins in Phagocytes and Phagocytosis. Front Immunol 2021; 12:633639. [PMID: 33868253 PMCID: PMC8044391 DOI: 10.3389/fimmu.2021.633639] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/11/2021] [Indexed: 01/10/2023] Open
Abstract
Phagocytes, which include neutrophils, monocytes, macrophages, and dendritic cells, protect the body by removing foreign particles, bacteria, and dead or dying cells. Phagocytic integrins are greatly involved in the recognition of and adhesion to specific antigens on cells and pathogens during phagocytosis as well as the recruitment of immune cells. β2 integrins, including αLβ2, αMβ2, αXβ2, and αDβ2, are the major integrins presented on the phagocyte surface. The activation of β2 integrins is essential to the recruitment and phagocytic function of these phagocytes and is critical for the regulation of inflammation and immune defense. However, aberrant activation of β2 integrins aggravates auto-immune diseases, such as psoriasis, arthritis, and multiple sclerosis, and facilitates tumor metastasis, making them double-edged swords as candidates for therapeutic intervention. Therefore, precise regulation of phagocyte activities by targeting β2 integrins should promote their host defense functions with minimal side effects on other cells. Here, we reviewed advances in the regulatory mechanisms underlying β2 integrin inside-out signaling, as well as the roles of β2 integrin activation in phagocyte functions.
Collapse
Affiliation(s)
- Hao Sun
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Kangkang Zhi
- Department of Vascular Surgery, Changzheng Hospital, Shanghai, China
| | - Liang Hu
- Department of Cardiology, Cardiovascular Institute of Zhengzhou University, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, United States
| |
Collapse
|
120
|
Jeong J, Kim J. Cyclic RGD Pentapeptide Cilengitide Enhances Efficacy of Gefitinib on TGF-β1-Induced Epithelial-to-Mesenchymal Transition and Invasion in Human Non-Small Cell Lung Cancer Cells. Front Pharmacol 2021; 12:639095. [PMID: 33967774 PMCID: PMC8104086 DOI: 10.3389/fphar.2021.639095] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
During non-small cell lung cancer (NSCLC) progression, transforming growth factor (TGF)-β mediated epithelial-to-mesenchymal transition (EMT) is an important process leading to high mortality and poor prognosis. The EMT is a fundamental process for morphogenesis characterized by the transformation of cancer cells into invasive forms that can be transferred to other organs during human lung cancer progression. Gefitinib, an epidermal growth factor receptor (EGFR) inhibitor, has shown anti-proliferative effects in EGFR-mutated NSCLC cells and an inhibitory effect on migration and invasion of NSCLC cells to other organs. In this study, we evaluated the combinatorial treatment effect of cilengitide, a cyclic RGD pentapeptide, on TGF-β1-induced EMT phenotype and invasion. Gefitinib suppressed the expression of TGF-β1-induced mesenchymal markers by inhibiting Smad and non-Smad signaling pathways. Cilengitide enhanced the inhibitory effect of gefitinib on TGF-β1-induced expression of mesenchymal markers, phosphorylation of Smad2/3, and invasion of NSCLC A549 cells. We suggested that the use of cilengitide can improve the efficacy of anti-cancer drugs in combination drug-based chemotherapy. These results provide an improved therapeutic strategy for treating and preventing EMT-related disorders, such as NSCLC, lung fibrosis, cancer metastasis, and relapse.
Collapse
Affiliation(s)
- Jisu Jeong
- Department of Medical Laboratory Science, School of Health Science, Dankook University, Cheonan, Korea
| | - Jiyeon Kim
- Department of Medical Laboratory Science, School of Health Science, Dankook University, Cheonan, Korea
| |
Collapse
|
121
|
Sacco G, Stammwitz S, Belvisi L, Pignataro L, Dal Corso A, Gennari C. Functionalized 2‐Hydroxybenzaldehyde‐PEG Modules as Portable Tags for the Engagement of Protein Lysine ϵ‐Amino Groups. European J Org Chem 2021. [DOI: 10.1002/ejoc.202100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Giovanni Sacco
- Dipartimento di Chimica Università degli Studi di Milano Via C. Golgi, 19 20133 Milan Italy
| | - Simon Stammwitz
- Dipartimento di Chimica Università degli Studi di Milano Via C. Golgi, 19 20133 Milan Italy
| | - Laura Belvisi
- Dipartimento di Chimica Università degli Studi di Milano Via C. Golgi, 19 20133 Milan Italy
| | - Luca Pignataro
- Dipartimento di Chimica Università degli Studi di Milano Via C. Golgi, 19 20133 Milan Italy
| | - Alberto Dal Corso
- Dipartimento di Chimica Università degli Studi di Milano Via C. Golgi, 19 20133 Milan Italy
| | - Cesare Gennari
- Dipartimento di Chimica Università degli Studi di Milano Via C. Golgi, 19 20133 Milan Italy
| |
Collapse
|
122
|
Caulier B, Stofleth G, Hannani D, Guidetti M, Josserand V, Laurin D, Chroboczek J, Mossuz P, Plantaz D. Evaluation of the human type 3 adenoviral dodecahedron as a vector to target acute myeloid leukemia. Mol Ther Methods Clin Dev 2021; 20:181-190. [PMID: 33473357 PMCID: PMC7797482 DOI: 10.1016/j.omtm.2020.11.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023]
Abstract
Intensive systemic chemotherapy is the gold standard of acute myeloid leukemia (AML) treatment and is associated with considerable off-target toxicities. Safer and targeted delivery systems are thus urgently needed. In this study, we evaluated a virus-like particle derived from the human type 3 adenovirus, called the adenoviral dodecahedron (Dd) to target AML cells. The vectorization of leukemic cells was proved very effective at nanomolar concentrations in a time- and dose-dependent manner, without vector toxicity. The internalization involved clathrin-mediated energy-dependent endocytosis and strongly correlated with the expression of αVβ3 integrin. The treatment of healthy donor peripheral blood mononuclear cells showed a preferential targeting of monocytes compared to lymphocytes and granulocytes. Similarly, monocytes but also AML blasts were the best-vectorized populations in patients while acute lymphoid leukemia blasts were less efficiently targeted. Importantly, AML leukemic stem cells (LSCs) could be addressed. Finally, Dd reached peripheral monocytes and bone marrow hematopoietic stem and progenitor cells following intravenous injection in mice, without excessive spreading in other organs. These findings reveal Dd as a promising myeloid vector especially for therapeutic purposes in AML blasts, LSCs, and progenitor cells.
Collapse
Affiliation(s)
- Benjamin Caulier
- University Grenoble Alpes, CNRS, CHU Grenoble Alpes, Grenoble INP, TIMC-IMAG, 38000 Grenoble, France
- Institute of Biology and Pathology, Laboratory of Cellular Hematology, University Grenoble Alpes Hospital, Grenoble, France
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, Grenoble, France
| | - Gaëlle Stofleth
- Institute of Biology and Pathology, Laboratory of Cellular Hematology, University Grenoble Alpes Hospital, Grenoble, France
- Department of Pediatric Onco-Immuno-Hematology, University Grenoble Alpes Hospital, Grenoble, France
| | - Dalil Hannani
- University Grenoble Alpes, CNRS, CHU Grenoble Alpes, Grenoble INP, TIMC-IMAG, 38000 Grenoble, France
| | - Mélanie Guidetti
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, Grenoble, France
| | - Véronique Josserand
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, Grenoble, France
| | - David Laurin
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, Grenoble, France
- Etablissement Français du Sang Auvergne Rhône-Alpes, Grenoble, France
| | - Jadwiga Chroboczek
- University Grenoble Alpes, CNRS, CHU Grenoble Alpes, Grenoble INP, TIMC-IMAG, 38000 Grenoble, France
| | - Pascal Mossuz
- Institute of Biology and Pathology, Laboratory of Cellular Hematology, University Grenoble Alpes Hospital, Grenoble, France
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR 5309, University Grenoble Alpes, Grenoble, France
| | - Dominique Plantaz
- Department of Pediatric Onco-Immuno-Hematology, University Grenoble Alpes Hospital, Grenoble, France
| |
Collapse
|
123
|
Min S, Ko MJ, Jung HJ, Kim W, Han SB, Kim Y, Bae G, Lee S, Thangam R, Choi H, Li N, Shin JE, Jeon YS, Park HS, Kim YJ, Sukumar UK, Song JJ, Park SK, Yu SH, Kang YC, Lee KB, Wei Q, Kim DH, Han SM, Paulmurugan R, Kim YK, Kang H. Remote Control of Time-Regulated Stretching of Ligand-Presenting Nanocoils In Situ Regulates the Cyclic Adhesion and Differentiation of Stem Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2008353. [PMID: 33527502 DOI: 10.1002/adma.202008353] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Indexed: 06/12/2023]
Abstract
Native extracellular matrix (ECM) can exhibit cyclic nanoscale stretching and shrinking of ligands to regulate complex cell-material interactions. Designing materials that allow cyclic control of changes in intrinsic ligand-presenting nanostructures in situ can emulate ECM dynamicity to regulate cellular adhesion. Unprecedented remote control of rapid, cyclic, and mechanical stretching ("ON") and shrinking ("OFF") of cell-adhesive RGD ligand-presenting magnetic nanocoils on a material surface in five repeated cycles are reported, thereby independently increasing and decreasing ligand pitch in nanocoils, respectively, without modulating ligand-presenting surface area per nanocoil. It is demonstrated that cyclic switching "ON" (ligand nanostretching) facilitates time-regulated integrin ligation, focal adhesion, spreading, YAP/TAZ mechanosensing, and differentiation of viable stem cells, both in vitro and in vivo. Fluorescence resonance energy transfer (FRET) imaging reveals magnetic switching "ON" (stretching) and "OFF" (shrinking) of the nanocoils inside animals. Versatile tuning of physical dimensions and elements of nanocoils by regulating electrodeposition conditions is also demonstrated. The study sheds novel insight into designing materials with connected ligand nanostructures that exhibit nanocoil-specific nano-spaced declustering, which is ineffective in nanowires, to facilitate cell adhesion. This unprecedented, independent, remote, and cytocompatible control of ligand nanopitch is promising for regulating the mechanosensing-mediated differentiation of stem cells in vivo.
Collapse
Affiliation(s)
- Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Min Jun Ko
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hee Joon Jung
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
- International Institute for Nanotechnology, Evanston, IL, USA
- NUANCE Center, Northwestern University, Evanston, IL, USA
| | - Wonsik Kim
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Seong-Beom Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Yuri Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Gunhyu Bae
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sungkyu Lee
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyojun Choi
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Na Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Jeong Eun Shin
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yoo Sang Jeon
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyeon Su Park
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yu Jin Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Uday Kumar Sukumar
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Seung-Keun Park
- Department of Chemical Engineering, Kongju National University, Cheonan, 31080, Republic of Korea
| | - Seung-Ho Yu
- Department of Chemical and Biological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yun Chan Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, China
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Seung Min Han
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Young Keun Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
124
|
Damjanovic J, Miao J, Huang H, Lin YS. Elucidating Solution Structures of Cyclic Peptides Using Molecular Dynamics Simulations. Chem Rev 2021; 121:2292-2324. [PMID: 33426882 DOI: 10.1021/acs.chemrev.0c01087] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Protein-protein interactions are vital to biological processes, but the shape and size of their interfaces make them hard to target using small molecules. Cyclic peptides have shown promise as protein-protein interaction modulators, as they can bind protein surfaces with high affinity and specificity. Dozens of cyclic peptides are already FDA approved, and many more are in various stages of development as immunosuppressants, antibiotics, antivirals, or anticancer drugs. However, most cyclic peptide drugs so far have been natural products or derivatives thereof, with de novo design having proven challenging. A key obstacle is structural characterization: cyclic peptides frequently adopt multiple conformations in solution, which are difficult to resolve using techniques like NMR spectroscopy. The lack of solution structural information prevents a thorough understanding of cyclic peptides' sequence-structure-function relationship. Here we review recent development and application of molecular dynamics simulations with enhanced sampling to studying the solution structures of cyclic peptides. We describe novel computational methods capable of sampling cyclic peptides' conformational space and provide examples of computational studies that relate peptides' sequence and structure to biological activity. We demonstrate that molecular dynamics simulations have grown from an explanatory technique to a full-fledged tool for systematic studies at the forefront of cyclic peptide therapeutic design.
Collapse
Affiliation(s)
- Jovan Damjanovic
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Jiayuan Miao
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - He Huang
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| | - Yu-Shan Lin
- Department of Chemistry, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
125
|
Song G, Luo BH. Effects of the association of the α v β 8 lower legs on integrin ligand binding. J Cell Biochem 2021; 122:801-813. [PMID: 33619784 DOI: 10.1002/jcb.29912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/18/2022]
Abstract
Many integrins transmit signals through global conformational changes. However, it is unclear whether integrin αv β8 adopts a similar mechanism during integrin activation and signaling on the cell surface. Here, we showed that disulfide-bonded mutants, which prevented integrin αv β8 lower leg dissociation, bound ligands with similar level as the wild-type protein, suggesting that αv β8 ligand binding did not require lower leg disassociation. We further showed that the N-glycosylation mutant at the interface between the β I and hybrid domains did not affect ligand binding, suggesting that the αv β8 open headpiece was not present on the cell surface. We proposed that αv β8 integrin may adopt only one state, that is, the extended conformation with a closed headpiece. Our results showed that two lower legs retained heterodimeric interfaces, and this association might be important for stabilizing integrin in the extended conformation. Therefore, αv β8 may not transmit bidirectional signals across the plasma membrane but instead may serve as an anchoring site with high affinity and high accessibility for extracellular ligands.
Collapse
Affiliation(s)
- Guannan Song
- Department of Life Science, University of Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bing-Hao Luo
- Department of Life Science, University of Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|
126
|
Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, Duca L, Durbeej M, Afratis NA, Troeberg L, Franchi M, Masola V, Onisto M. A guide to the composition and functions of the extracellular matrix. FEBS J 2021; 288:6850-6912. [PMID: 33605520 DOI: 10.1111/febs.15776] [Citation(s) in RCA: 362] [Impact Index Per Article: 120.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Extracellular matrix (ECM) is a dynamic 3-dimensional network of macromolecules that provides structural support for the cells and tissues. Accumulated knowledge clearly demonstrated over the last decade that ECM plays key regulatory roles since it orchestrates cell signaling, functions, properties and morphology. Extracellularly secreted as well as cell-bound factors are among the major members of the ECM family. Proteins/glycoproteins, such as collagens, elastin, laminins and tenascins, proteoglycans and glycosaminoglycans, hyaluronan, and their cell receptors such as CD44 and integrins, responsible for cell adhesion, comprise a well-organized functional network with significant roles in health and disease. On the other hand, enzymes such as matrix metalloproteinases and specific glycosidases including heparanase and hyaluronidases contribute to matrix remodeling and affect human health. Several cell processes and functions, among them cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immunity regulation are affected by certain matrix components. Structural alterations have been also well associated with disease progression. This guide on the composition and functions of the ECM gives a broad overview of the matrisome, the major ECM macromolecules, and their interaction networks within the ECM and with the cell surface, summarizes their main structural features and their roles in tissue organization and cell functions, and emphasizes the importance of specific ECM constituents in disease development and progression as well as the advances in molecular targeting of ECM to design new therapeutic strategies.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece.,Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems- Functional Molecular Systems, Eggenstein-Leopoldshafen, Germany
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246, ICBMS, Université Lyon 1, CNRS, Villeurbanne Cedex, France
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany.,Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2: Matrix Aging and Vascular Remodelling, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, Sweden
| | - Nikolaos A Afratis
- Department Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich, UK
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | | | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
127
|
Yu K, Yu Y, Yao Y, Wu Z, Fu S, Cheng RH, Chen YW, Chen HY, Zhou J, Hwang DW, Ding S. A Polypeptide-Based, Membrane-Penetrating, Target-Specific Contrast Agent for Magnetic Resonance Molecular Imaging. ACS APPLIED BIO MATERIALS 2021; 4:1597-1604. [DOI: 10.1021/acsabm.0c01442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Kaichao Yu
- Department of Chemistry and Centre for Nanoscience and Nanotechnology, National Sun Yat-sen University, 70 Lien-Hai Road, Kaohsiung 80424, Taiwan
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yamian Yu
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yao Yao
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhen Wu
- Department of Chemistry and Centre for Nanoscience and Nanotechnology, National Sun Yat-sen University, 70 Lien-Hai Road, Kaohsiung 80424, Taiwan
| | - Shitao Fu
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Ren-Hao Cheng
- Department of Chemistry and Centre for Nanoscience and Nanotechnology, National Sun Yat-sen University, 70 Lien-Hai Road, Kaohsiung 80424, Taiwan
| | - Yu-Wen Chen
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd. Nankang, Taipei 115, Taiwan
| | - Hsuan-Ying Chen
- Department of Medicinal and Applied Chemistry, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung 80708, Taiwan
| | - Jinlan Zhou
- School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dennis W. Hwang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd. Nankang, Taipei 115, Taiwan
| | - Shangwu Ding
- Department of Chemistry and Centre for Nanoscience and Nanotechnology, National Sun Yat-sen University, 70 Lien-Hai Road, Kaohsiung 80424, Taiwan
| |
Collapse
|
128
|
Vandendriessche S, Cambier S, Proost P, Marques PE. Complement Receptors and Their Role in Leukocyte Recruitment and Phagocytosis. Front Cell Dev Biol 2021; 9:624025. [PMID: 33644062 PMCID: PMC7905230 DOI: 10.3389/fcell.2021.624025] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/15/2021] [Indexed: 12/21/2022] Open
Abstract
The complement system is deeply embedded in our physiology and immunity. Complement activation generates a multitude of molecules that converge simultaneously on the opsonization of a target for phagocytosis and activation of the immune system via soluble anaphylatoxins. This response is used to control microorganisms and to remove dead cells, but also plays a major role in stimulating the adaptive immune response and the regeneration of injured tissues. Many of these effects inherently depend on complement receptors expressed on leukocytes and parenchymal cells, which, by recognizing complement-derived molecules, promote leukocyte recruitment, phagocytosis of microorganisms and clearance of immune complexes. Here, the plethora of information on the role of complement receptors will be reviewed, including an analysis of how this functionally and structurally diverse group of molecules acts jointly to exert the full extent of complement regulation of homeostasis.
Collapse
Affiliation(s)
- Sofie Vandendriessche
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Seppe Cambier
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| | - Pedro E Marques
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit Leuven (KU Leuven), Leuven, Belgium
| |
Collapse
|
129
|
Torikai M, Higuchi H, Yamamoto N, Ishikawa D, Fujita H, Taguchi K, Sakai F, Soejima K, Nakashima T. A novel monoclonal antibody cross-reactive with both human and mouse α9 integrin useful for therapy against rheumatoid arthritis. J Biochem 2021; 168:231-241. [PMID: 32271918 DOI: 10.1093/jb/mvaa040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/28/2020] [Indexed: 11/14/2022] Open
Abstract
This study introduces a novel monoclonal anti-α9 integrin antibody (MA9-413) with human variable regions, isolated by phage display technology. MA9-413 specifically binds to both human and mouse α9 integrin by recognizing a conserved loop region designated as L1 (amino acids 104-122 of human α9 integrin). MA9-413 inhibits human and mouse α9 integrin-dependent cell adhesion to ligands and suppresses synovial inflammation and osteoclast activation in a mouse model of arthritis. This is the first monoclonal anti-α9 integrin antibody that can react with and functionally inhibit both human and mouse α9 integrin. MA9-413 allows data acquisition both in animal and human pharmacological studies without resorting to surrogate antibodies. Since MA9-413 showed certain therapeutic effects in the mouse arthritis model, it can be considered as a useful therapy against rheumatoid arthritis and other α9 integrin-associated diseases.
Collapse
Affiliation(s)
- Masaharu Torikai
- Research & Development Division, KM Biologics Co., Ltd, 1314-1 Kyokushi-Kawabe, Kikuchi, Kumamoto 869-1298, Japan
| | - Hirofumi Higuchi
- Research & Development Division, KM Biologics Co., Ltd, 1314-1 Kyokushi-Kawabe, Kikuchi, Kumamoto 869-1298, Japan
| | | | - Daisuke Ishikawa
- Research & Development Division, KM Biologics Co., Ltd, 1314-1 Kyokushi-Kawabe, Kikuchi, Kumamoto 869-1298, Japan
| | - Hirotada Fujita
- Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Katsunari Taguchi
- Astellas Pharma Inc., 21 Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Fumihiko Sakai
- EVEC Inc., 6 Odori Nishi, Chuo-ku, Sapporo 060-0042, Japan
| | - Kenji Soejima
- Research & Development Division, KM Biologics Co., Ltd, 1314-1 Kyokushi-Kawabe, Kikuchi, Kumamoto 869-1298, Japan
| | - Toshihiro Nakashima
- The Chemo-Sero-Therapeutic Research Institute (Kaketsuken), 4-7 Hanabatacho, Chuo-ku, Kumamoto 860-0806, Japan
| |
Collapse
|
130
|
Deguchi S, Kato A, Wu P, Hakamada M, Mabuchi M. Heterogeneous role of integrins in fibroblast response to small cyclic mechanical stimulus generated by a nanoporous gold actuator. Acta Biomater 2021; 121:418-430. [PMID: 33326880 DOI: 10.1016/j.actbio.2020.12.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023]
Abstract
It is important to understand the effects of mechanical stimulation on cell behaviors for homeostasis. Many studies have been performed on cell responses to mechanical stimuli, but the mechanosensing mechanism is still under debate. In the present study, experiments employing molecular dynamics (MD) simulations concerning the effects of cyclic mechanical stimulus on cell proliferation were performed based on the hypothesis that mechanosensing depends on integrin types. We used a nanoporous gold (NPG) actuator to prevent transfer of a mechanical stimulus via molecules other than integrins. Surprisingly, a small cyclic strain of only 0.5% enhanced the proliferation of fibroblasts. α5β1 and αvβ3 integrins showed high sensitivity to the mechanical stimulus, whereas α1β1 and α2β1 integrins exhibited low mechanosensitivity. The MD simulations showed that different conformational changes of the integrin headpiece induced by binding to the ECM led to a difference in mechanosensitivity between αI and αI-less integrin types. Thus, the present study provides evidence to support the hypothesis and suggests the mechanism for the heterogeneous roles of integrins in mechanosensing.
Collapse
Affiliation(s)
- Soichiro Deguchi
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan.
| | - Atsushi Kato
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| | - Peizheng Wu
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| | - Masataka Hakamada
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| | - Mamoru Mabuchi
- Graduate School of Energy Science, Kyoto University, Yoshidahonmachi, Sakyo, Kyoto 606-8501, Japan
| |
Collapse
|
131
|
Cirillo M, Giacomini D. Molecular Delivery of Cytotoxic Agents via Integrin Activation. Cancers (Basel) 2021; 13:299. [PMID: 33467465 PMCID: PMC7830197 DOI: 10.3390/cancers13020299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/16/2022] Open
Abstract
Integrins are cell adhesion receptors overexpressed in tumor cells. A direct inhibition of integrins was investigated, but the best inhibitors performed poorly in clinical trials. A gained attention towards these receptors arouse because they could be target for a selective transport of cytotoxic agents. Several active-targeting systems have been developed to use integrins as a selective cell entrance for some antitumor agents. The aim of this review paper is to report on the most recent results on covalent conjugates between integrin ligands and antitumor drugs. Cytotoxic drugs thus conjugated through specific linker to integrin ligands, mainly RGD peptides, demonstrated that the covalent conjugates were more selective against tumor cells and hopefully with fewer side effects than the free drugs.
Collapse
Affiliation(s)
| | - Daria Giacomini
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum University of Bologna, Via Selmi 2, 40126 Bologna, Italy;
| |
Collapse
|
132
|
De Melo N, Murrell L, Islam MT, Titman JJ, Macri-Pellizzeri L, Ahmed I, Sottile V. Tailoring Pyro-and Orthophosphate Species to Enhance Stem Cell Adhesion to Phosphate Glasses. Int J Mol Sci 2021; 22:ijms22020837. [PMID: 33467686 PMCID: PMC7829838 DOI: 10.3390/ijms22020837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/29/2022] Open
Abstract
Phosphate-based glasses (PBGs) offer significant therapeutic potential due to their bioactivity, controllable compositions, and degradation rates. Several PBGs have already demonstrated their ability to support direct cell growth and in vivo cytocompatibility for bone repair applications. This study investigated development of PBG formulations with pyro- and orthophosphate species within the glass system (40 − x)P2O5·(16 + x)CaO·20Na2O·24MgO (x = 0, 5, 10 mol%) and their effect on stem cell adhesion properties. Substitution of phosphate for calcium revealed a gradual transition within the glass structure from Q2 to Q0 phosphate species. Human mesenchymal stem cells were cultured directly onto discs made from three PBG compositions. Analysis of cells seeded onto the discs revealed that PBG with higher concentration of pyro- and orthophosphate content (61% Q1 and 39% Q0) supported a 4.3-fold increase in adhered cells compared to glasses with metaphosphate connectivity (49% Q2 and 51% Q1). This study highlights that tuning the composition of PBGs to possess pyro- and orthophosphate species only, enables the possibility to control cell adhesion performance. PBGs with superior cell adhesion profiles represent ideal candidates for biomedical applications, where cell recruitment and support for tissue ingrowth are of critical importance for orthopaedic interventions.
Collapse
Affiliation(s)
- Nigel De Melo
- School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK; (N.D.M.); (L.M.-P.)
| | - Lauren Murrell
- Advanced Materials Research Group, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, UK; (L.M.); (M.T.I.)
| | - Md Towhidul Islam
- Advanced Materials Research Group, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, UK; (L.M.); (M.T.I.)
- Department of Applied Chemistry and Chemical Engineering, Faculty of Engineering, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Jeremy J. Titman
- School of Chemistry, University of Nottingham, Nottingham NG7 2RD, UK;
| | - Laura Macri-Pellizzeri
- School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK; (N.D.M.); (L.M.-P.)
| | - Ifty Ahmed
- Advanced Materials Research Group, Faculty of Engineering, University of Nottingham, Nottingham NG7 2RD, UK; (L.M.); (M.T.I.)
- Correspondence: (I.A.); (V.S.)
| | - Virginie Sottile
- School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK; (N.D.M.); (L.M.-P.)
- Department of Molecular Medicine, The University of Pavia, 27100 Pavia, Italy
- Correspondence: (I.A.); (V.S.)
| |
Collapse
|
133
|
Feng W, Nguyen H, Shen D, Deng H, Jiang Z, Podolnikova N, Ugarova T, Wang X. Structural Characterization of the Interaction between the α MI-Domain of the Integrin Mac-1 (α Mβ 2) and the Cytokine Pleiotrophin. Biochemistry 2021; 60:182-193. [PMID: 33427449 DOI: 10.1021/acs.biochem.0c00700] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Integrin Mac-1 (αMβ2) is an adhesion receptor vital to many functions of myeloid leukocytes. It is also the most promiscuous member of the integrin family capable of recognizing a broad range of ligands. In particular, its ligand-binding αMI-domain is known to bind cationic proteins/peptides depleted in acidic residues. This contradicts the canonical ligand-binding mechanism of αI-domains, which requires an acidic amino acid in the ligand to coordinate the divalent cation within the metal ion-dependent adhesion site (MIDAS) of αI-domains. The lack of acidic amino acids in the αMI-domain-binding sequences suggests the existence of an as-yet uncharacterized interaction mechanism. In the present study, we analyzed interactions of the αMI-domain with a representative Mac-1 ligand, the cationic cytokine pleiotrophin (PTN). Through NMR chemical shift perturbation analysis, cross saturation, NOESY, and mutagenesis studies, we found the interaction between the αMI-domain and PTN is divalent cation-independent and mediated mostly by hydrophobic contacts between the N-terminal domain of PTN and residues in the α5-β5 loop of αMI-domain. The observation that increased ionic strength weakens the interaction between the proteins indicates electrostatic forces may also play a significant role in the binding. On the basis of the results from these experiments, we formulated a model of the interaction between the αMI-domain and PTN.
Collapse
|
134
|
Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res 2021; 89:1619-1626. [PMID: 33027803 PMCID: PMC8249239 DOI: 10.1038/s41390-020-01177-9] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/18/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Integrins are heterodimeric transmembrane cell adhesion molecules made up of alpha (α) and beta (β) subunits arranged in numerous dimeric pairings. These complexes have varying affinities to extracellular ligands. Integrins regulate cellular growth, proliferation, migration, signaling, and cytokine activation and release and thereby play important roles in cell proliferation and migration, apoptosis, tissue repair, as well as in all processes critical to inflammation, infection, and angiogenesis. This review presents current evidence from human and animal studies on integrin structure and molecular signaling, with particular emphasis on signal transduction in infants. We have included evidence from our own laboratory studies and from an extensive literature search in databases PubMed, EMBASE, Scopus, and the electronic archives of abstracts presented at the annual meetings of the Pediatric Academic Societies. To avoid bias in identification of existing studies, key words were short-listed prior to the actual search both from anecdotal experience and from PubMed's Medical Subject Heading (MeSH) thesaurus. IMPACT: Integrins are a family of ubiquitous αβ heterodimeric receptors that interact with numerous ligands in physiology and disease. Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis. This review summarizes current evidence from human and animal studies on integrin structure and molecular signaling and promising role in diseases of inflammation, infection, and angiogenesis in infants. This review shows that integrin receptors and ligands are novel therapeutic targets of clinical interest and hold promise as novel therapeutic targets in the management of several neonatal diseases.
Collapse
Affiliation(s)
- Olachi J. Mezu-Ndubuisi
- grid.14003.360000 0001 2167 3675Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Akhil Maheshwari
- grid.21107.350000 0001 2171 9311Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
135
|
Mukai H, Watanabe Y. Review: PET imaging with macro- and middle-sized molecular probes. Nucl Med Biol 2021; 92:156-170. [PMID: 32660789 DOI: 10.1016/j.nucmedbio.2020.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022]
Abstract
Recent progress in radiolabeling of macro- and middle-sized molecular probes has been extending possibilities to use PET molecular imaging for dynamic application to drug development and therapeutic evaluation. Theranostics concept also accelerated the use of macro- and middle-sized molecular probes for sharpening the contrast of proper target recognition even the cellular types/subtypes and proper selection of the patients who should be treated by the same molecules recognition. Here, brief summary of the present status of immuno-PET, and then further development of advanced technologies related to immuno-PET, peptidic PET probes, and nucleic acids PET probes are described.
Collapse
Affiliation(s)
- Hidefumi Mukai
- Laboratory for Molecular Delivery and Imaging Technology, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, 6-7-3 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan.
| |
Collapse
|
136
|
HIV-1 Tat Protein Enters Dysfunctional Endothelial Cells via Integrins and Renders Them Permissive to Virus Replication. Int J Mol Sci 2020; 22:ijms22010317. [PMID: 33396807 PMCID: PMC7796023 DOI: 10.3390/ijms22010317] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/21/2020] [Accepted: 12/26/2020] [Indexed: 12/21/2022] Open
Abstract
Previous work has shown that the Tat protein of Human Immunodeficiency Virus (HIV)-1 is released by acutely infected cells in a biologically active form and enters dendritic cells upon the binding of its arginine-glycine-aspartic acid (RGD) domain to the α5β1, αvβ3, and αvβ5 integrins. The up-regulation/activation of these integrins occurs in endothelial cells exposed to inflammatory cytokines that are increased in HIV-infected individuals, leading to endothelial cell dysfunction. Here, we show that inflammatory cytokine-activated endothelial cells selectively bind and rapidly take up nano-micromolar concentrations of Tat, as determined by flow cytometry. Protein oxidation and low temperatures reduce Tat entry, suggesting a conformation- and energy-dependent process. Consistently, Tat entry is competed out by RGD-Tat peptides or integrin natural ligands, and it is blocked by anti-α5β1, -αvβ3, and -αvβ5 antibodies. Moreover, modelling–docking calculations identify a low-energy Tat-αvβ3 integrin complex in which Tat makes contacts with both the αv and β3 chains. It is noteworthy that internalized Tat induces HIV replication in inflammatory cytokine-treated, but not untreated, endothelial cells. Thus, endothelial cell dysfunction driven by inflammatory cytokines renders the vascular system a target of Tat, which makes endothelial cells permissive to HIV replication, adding a further layer of complexity to functionally cure and/or eradicate HIV infection.
Collapse
|
137
|
Panzeri S, Arosio D, Gazzola S, Belvisi L, Civera M, Potenza D, Vasile F, Kemker I, Ertl T, Sewald N, Reiser O, Piarulli U. Cyclic RGD and isoDGR Integrin Ligands Containing cis-2-amino-1-cyclopentanecarboxylic ( cis-β-ACPC) Scaffolds. Molecules 2020; 25:molecules25245966. [PMID: 33339382 PMCID: PMC7766232 DOI: 10.3390/molecules25245966] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
Integrin ligands containing the tripeptide sequences Arg-Gly-Asp (RGD) and iso-Asp-Gly- Arg (isoDGR) were actively investigated as inhibitors of tumor angiogenesis and directing unit in tumor-targeting drug conjugates. Reported herein is the synthesis, of two RGD and one isoDGR cyclic peptidomimetics containing (1S,2R) and (1R,2S) cis-2-amino-1-cyclopentanecarboxylic acid (cis-β-ACPC), using a mixed solid phase/solution phase synthetic protocol. The three ligands were examined in vitro in competitive binding assays to the purified αvβ3 and α5β1 receptors using biotinylated vitronectin (αvβ3) and fibronectin (α5β1) as natural displaced ligands. The IC50 values of the ligands ranged from nanomolar (the two RGD ligands) to micromolar (the isoDGR ligand) with a pronounced selectivity for αvβ3 over α5β1. In vitro cell adhesion assays were also performed using the human skin melanoma cell line WM115 (rich in integrin αvβ3). The two RGD ligands showed IC50 values in the same micromolar range as the reference compound (cyclo[RGDfV]), while for the isoDGR derivative an IC50 value could not be measured for the cell adhesion assay. A conformational analysis of the free RGD and isoDGR ligands by NMR (VT-NMR and NOESY experiments) and computational studies (MC/EM and MD), followed by docking simulations performed in the αVβ3 integrin active site, provided a rationale for the behavior of these ligands toward the receptor.
Collapse
Affiliation(s)
- Silvia Panzeri
- Dipartimento di Scienza e Alta Tecnologia, Università degli Studi dell’Insubria, Via Valleggio 11, 22100 Como, Italy; (S.P.); (S.G.)
- Institute of Organic Chemistry, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany; (T.E.); (O.R.)
| | - Daniela Arosio
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Scienze e Tecnologie Chimiche (SCITEC), Giulio Natta, Via C. Golgi 19, 20133 Milan, Italy;
| | - Silvia Gazzola
- Dipartimento di Scienza e Alta Tecnologia, Università degli Studi dell’Insubria, Via Valleggio 11, 22100 Como, Italy; (S.P.); (S.G.)
| | - Laura Belvisi
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy; (L.B.); (M.C.); (D.P.); (F.V.)
| | - Monica Civera
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy; (L.B.); (M.C.); (D.P.); (F.V.)
| | - Donatella Potenza
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy; (L.B.); (M.C.); (D.P.); (F.V.)
| | - Francesca Vasile
- Dipartimento di Chimica, Università degli Studi di Milano, Via C. Golgi 19, 20133 Milan, Italy; (L.B.); (M.C.); (D.P.); (F.V.)
| | - Isabell Kemker
- Department of Chemistry, Organic and Bioorganic Chemistry, University of Bielefeld, Universitätsstraße 25, 33615 Bielefeld, Germany; (I.K.); (N.S.)
| | - Thomas Ertl
- Institute of Organic Chemistry, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany; (T.E.); (O.R.)
| | - Norbert Sewald
- Department of Chemistry, Organic and Bioorganic Chemistry, University of Bielefeld, Universitätsstraße 25, 33615 Bielefeld, Germany; (I.K.); (N.S.)
| | - Oliver Reiser
- Institute of Organic Chemistry, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany; (T.E.); (O.R.)
| | - Umberto Piarulli
- Dipartimento di Scienza e Alta Tecnologia, Università degli Studi dell’Insubria, Via Valleggio 11, 22100 Como, Italy; (S.P.); (S.G.)
- Correspondence:
| |
Collapse
|
138
|
Ma J, Huang C. Composition and Mechanism of Three-Dimensional Hydrogel System in Regulating Stem Cell Fate. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:498-518. [PMID: 32272868 DOI: 10.1089/ten.teb.2020.0021] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Three-dimensional (3D) hydrogel systems integrating different types of stem cells and scaffolding biomaterials have an important application in tissue engineering. The biomimetic hydrogels that pattern cell suspensions within 3D configurations of biomaterial networks allow for the transport of bioactive factors and mimic the stem cell niche in vivo, thereby supporting the proliferation and differentiation of stem cells. The composition of a 3D hydrogel system determines the physical and chemical characteristics that regulate stem cell function through a biological mechanism. Here, we discuss the natural and synthetic hydrogel compositions that have been employed in 3D scaffolding, focusing on their characteristics, fabrication, biocompatibility, and regulatory effects on stem cell proliferation and differentiation. We also discuss the regulatory mechanisms of cell-matrix interaction and cell-cell interaction in stem cell activities in various types of 3D hydrogel systems. Understanding hydrogel compositions and their cellular mechanisms can yield insights into how scaffolding biomaterials and stem cells interact and can lead to the development of novel hydrogel systems of stem cells in tissue engineering and stem cell-based regenerative medicine. Impact statement Three-dimensional hydrogel system of stem cell mimicking the stemcell niche holds significant promise in tissue engineering and regenerative medicine. Exactly how hydrogel composition regulates stem cell fate is not well understood. This review focuses on the composition of hydrogel, and how the hydrogel composition and its properties regulate the stem cell adhesion, growth, and differentiation. We propose that cell-matrix interaction and cell-cell interaction are important regulatory mechanisms in stem cell activities. Our review provides key insights into how the hydrogel composition regulates the stem cell fate, untangling the engineering of three-dimensional hydrogel systems for stem cells.
Collapse
Affiliation(s)
- Jianrui Ma
- Center for Neurobiology, Shantou University Medical College, Shantou, China
| | - Chengyang Huang
- Center for Neurobiology, Shantou University Medical College, Shantou, China
- Department of Biological Chemistry, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, California, USA
| |
Collapse
|
139
|
Sani S, Messe M, Fuchs Q, Pierrevelcin M, Laquerriere P, Entz-Werle N, Reita D, Etienne-Selloum N, Bruban V, Choulier L, Martin S, Dontenwill M. Biological Relevance of RGD-Integrin Subtype-Specific Ligands in Cancer. Chembiochem 2020; 22:1151-1160. [PMID: 33140906 DOI: 10.1002/cbic.202000626] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/02/2020] [Indexed: 12/13/2022]
Abstract
Integrins are heterodimeric transmembrane proteins able to connect cells with the micro-environment. They represent a family of receptors involved in almost all the hallmarks of cancer. Integrins recognizing the Arg-Gly-Asp (RGD) peptide in their natural extracellular matrix ligands have been particularly investigated as tumoral therapeutic targets. In the last 30 years, intense research has been dedicated to designing specific RGD-like ligands able to discriminate selectively the different RGD-recognizing integrins. Chemists' efforts have led to the proposition of modified peptide or peptidomimetic libraries to be used for tumor targeting and/or tumor imaging. Here we review, from the biological point of view, the rationale underlying the need to clearly delineate each RGD-integrin subtype by selective tools. We describe the complex roles of RGD-integrins (mainly the most studied αvβ3 and α5β1 integrins) in tumors, the steps towards selective ligands and the current usefulness of such ligands. Although the impact of integrins in cancer is well acknowledged, the biological characteristics of each integrin subtype in a specific tumor are far from being completely resolved. Selective ligands might help us to reconsider integrins as therapeutic targets in specific clinical settings.
Collapse
Affiliation(s)
- Saidu Sani
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
- Cancer and Diabetic Research Group, Department of Biochemistry and Molecular Biology, Faculty of Science, Federal University Ndufu-Alike Ikwo, P.M.B, 1010, Abakaliki, Ebonyi State, Nigeria
| | - Mélissa Messe
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
- Institut Pluridisciplinaire Hubert Curien (IPHC), UMR CNRS 7178, Université de Strasbourg, 67000, Strasbourg, France
| | - Quentin Fuchs
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| | - Marina Pierrevelcin
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| | - Patrice Laquerriere
- Institut Pluridisciplinaire Hubert Curien (IPHC), UMR CNRS 7178, Université de Strasbourg, 67000, Strasbourg, France
| | - Natacha Entz-Werle
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
- Pediatric Onco-Hematology Department, Pediatrics, University Hospital of Strasbourg, 1 avenue Molière, 67098, Strasbourg, France
| | - Damien Reita
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
- Department of Oncobiology, Laboratory of Biochemistry and Molecular Biology, University Hospital of Strasbourg, France
| | - Nelly Etienne-Selloum
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
- Institut du Cancer Strasbourg Europe (ICANS), Service de Pharmacie, 17 rue Albert Calmette, 67200 Strasbourg, France
| | - Véronique Bruban
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| | - Laurence Choulier
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| | - Sophie Martin
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| | - Monique Dontenwill
- Laboratoire de Bioimagerie et Pathologies (LBP), UMR CNRS 7021, Institut Thématique Interdisciplinaire InnoVec, Université de Strasbourg, Faculté de Pharmacie, 74 route du, Rhin, CS 60024, 67401, Illkirch Cedex, France
| |
Collapse
|
140
|
Liu J, Zhang Y, Chen T, Chen H, He H, Jin T, Wang J, Ke Y. Environmentally Self-Adaptative Nanocarriers Suppress Glioma Proliferation and Stemness via Codelivery of shCD163 and Doxorubicin. ACS APPLIED MATERIALS & INTERFACES 2020; 12:52354-52369. [PMID: 33196179 DOI: 10.1021/acsami.0c14288] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Gliomas-devastating intracranial tumors with a dismal outcome-are in dire need of innovative treatment. Although nanodrugs have been utilized as a target therapy for certain types of solid tumors, their therapeutic effects in gliomas are limited due to the complications of the systemic circulation, blood-brain barrier (BBB), and specific glioma environment. Thus, we aimed to establish a nanoliposome adaptable to different environments by codelivery of shCD163 and doxorubicin (DOX) to treat gliomas. In this study, we first synthesized pH-sensitive DSPE-cRGD-Hz-PEG2000 to form an environmentally self-adaptative nanoliposome (cRGD-DDD Lip) via a thin film method. We used in vitro BBB models, in vitro cell uptake experiments, and in vivo biodistribution assays to confirm the long circulation time and low cell uptake of the cRGD-DDD Lip as a result of the poly(ethylene glycol) (PEG) shell of cRGD-DDD Lip in the neutral pH systemic circulation. Moreover, the cRGD-DDD Lip bypassed the BBB and attached to the intracranial glioma following the removal of the PEG shell and the exposure of cRGD to the weakly acidic tumor microenvironment. We further assembled the shCD163/DOX@cRGD-DDD Lip through cRGD-DDD Lip loading of shCD163 and DOX. In vitro, cell proliferation and self-renewal of glioma cells were inhibited by the shCD163/DOX@cRGD-DDD Lip due to the toxicity of DOX and the suppression of shCD163 via the CD163 pathway. In vivo, the shCD163/DOX@cRGD-DDD Lip disturbed the progression of in situ gliomas by inhibiting the growth and stemness of glioma cells and prevented the recurrence of gliomas after resection. In conclusion, the cRGD-DDD Lip may be a promising nanodrug-loading platform to cope with different environments and the shCD163/DOX@cRGD-DDD Lip may potentially be a novel nanodrug for glioma therapy.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/pharmacology
- Antibiotics, Antineoplastic/therapeutic use
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Antigens, Differentiation, Myelomonocytic/metabolism
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/metabolism
- Brain Neoplasms/drug therapy
- Brain Neoplasms/mortality
- Brain Neoplasms/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Doxorubicin/chemistry
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Glioma/drug therapy
- Glioma/mortality
- Glioma/pathology
- Humans
- Liposomes/chemistry
- Mice
- Mice, Nude
- Nanoparticles/chemistry
- Nanoparticles/metabolism
- Oligopeptides/chemistry
- Polyethylene Glycols/chemistry
- RNA Interference
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/metabolism
- Receptors, Cell Surface/antagonists & inhibitors
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Survival Rate
- Tissue Distribution
Collapse
Affiliation(s)
- Jie Liu
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yuxuan Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Taoliang Chen
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Huajian Chen
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Haoqi He
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Tao Jin
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Jihui Wang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yiquan Ke
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| |
Collapse
|
141
|
Zhang M, Sun Q, Liu Y, Chu Z, Yu L, Hou Y, Kang H, Wei Q, Zhao W, Spatz JP, Zhao C, Cavalcanti-Adam EA. Controllable ligand spacing stimulates cellular mechanotransduction and promotes stem cell osteogenic differentiation on soft hydrogels. Biomaterials 2020; 268:120543. [PMID: 33260094 DOI: 10.1016/j.biomaterials.2020.120543] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 01/14/2023]
Abstract
Hydrogels with tunable mechanical properties have provided a tremendous opportunity to regulate stem cell differentiation. Hydrogels with osteoid (about 30-40 kPa) or higher stiffness are usually required to induce the osteogenic differentiation of mesenchymal stem cells (MSCs). It is yet difficult to achieve the same differentiation on very soft hydrogels, because of low environmental mechanical stimuli and restricted cellular mechanotransduction. Here, we modulate cellular spatial sensing of integrin-adhesive ligands via quasi-hexagonally arranged nanopatterns to promote cell mechanosensing on hydrogels having low stiffness (about 3 kPa). The increased interligand spacing has been shown to regulate actomyosin force loading to recruit extra integrins on soft hydrogels. It therefore activates mechanotransduction and promotes the osteogenic differentiation of MSCs on soft hydrogels to the level comparable with the one observed on osteoid stiffness. Our work opens up new possibilities for the design of biomaterials and tissue scaffolds for regenerative therapeutics.
Collapse
Affiliation(s)
- Man Zhang
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, 610065, Chengdu, China; Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany; Department of Biophysical Chemistry, Heidelberg University, INF 253, 69120, Heidelberg, Germany
| | - Qian Sun
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, 610065, Chengdu, China
| | - Yiling Liu
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, 610065, Chengdu, China
| | - Zhiqin Chu
- Department of Electrical and Electronic Engineering, Joint Appointment with School of Biomedical Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Leixiao Yu
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195, Berlin, Germany
| | - Yong Hou
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustr. 3, 14195, Berlin, Germany
| | - Heemin Kang
- Department of Biomicrosystem Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, 610065, Chengdu, China; College of Biomedical Engineering, Sichuan University, 610064, Chengdu, China.
| | - Weifeng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, 610065, Chengdu, China.
| | - Joachim P Spatz
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany; Department of Biophysical Chemistry, Heidelberg University, INF 253, 69120, Heidelberg, Germany
| | - Changsheng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, 610065, Chengdu, China; College of Biomedical Engineering, Sichuan University, 610064, Chengdu, China
| | - Elisabetta A Cavalcanti-Adam
- Department of Cellular Biophysics, Max Planck Institute for Medical Research, Jahnstraße 29, 69120, Heidelberg, Germany; Department of Biophysical Chemistry, Heidelberg University, INF 253, 69120, Heidelberg, Germany
| |
Collapse
|
142
|
Guest EE, Oatley SA, Macdonald SJF, Hirst JD. Molecular Simulation of αvβ6 Integrin Inhibitors. J Chem Inf Model 2020; 60:5487-5498. [PMID: 32421320 DOI: 10.1021/acs.jcim.0c00254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The urgent need for new treatments for the chronic lung disease idiopathic pulmonary fibrosis (IPF) motivates research into antagonists of the RGD binding integrin αvβ6, a protein linked to the initiation and progression of the disease. Molecular dynamics (MD) simulations of αvβ6 in complex with its natural ligand, pro-TGF-β1, show the persistence over time of a bidentate Arg-Asp ligand-receptor interaction and a metal chelate interaction between an aspartate on the ligand and an Mg2+ ion in the active site. This is typical of RGD binding ligands. Additional binding site interactions, which are not observed in the static crystal structure, are also identified. We investigate an RGD mimetic, which serves as a framework for a series of potential αvβ6 antagonists. The scaffold includes a derivative of the widely utilized 1,8-naphthyridine moiety, for which we present force field parameters, to enable MD and relative free energy perturbation (FEP) simulations. The MD simulations highlight the importance of hydrogen bonding and cation-π interactions. The FEP calculations predict relative binding affinities, within 1.5 kcal mol-1, on average, of experiments.
Collapse
Affiliation(s)
- Ellen E Guest
- School of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Steven A Oatley
- School of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | | | - Jonathan D Hirst
- School of Chemistry, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| |
Collapse
|
143
|
Maynard SA, Winter CW, Cunnane EM, Stevens MM. Advancing Cell-Instructive Biomaterials Through Increased Understanding of Cell Receptor Spacing and Material Surface Functionalization. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2020; 7:553-547. [PMID: 34805482 PMCID: PMC8594271 DOI: 10.1007/s40883-020-00180-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Abstract Regenerative medicine is aimed at restoring normal tissue function and can benefit from the application of tissue engineering and nano-therapeutics. In order for regenerative therapies to be effective, the spatiotemporal integration of tissue-engineered scaffolds by the native tissue, and the binding/release of therapeutic payloads by nano-materials, must be tightly controlled at the nanoscale in order to direct cell fate. However, due to a lack of insight regarding cell–material interactions at the nanoscale and subsequent downstream signaling, the clinical translation of regenerative therapies is limited due to poor material integration, rapid clearance, and complications such as graft-versus-host disease. This review paper is intended to outline our current understanding of cell–material interactions with the aim of highlighting potential areas for knowledge advancement or application in the field of regenerative medicine. This is achieved by reviewing the nanoscale organization of key cell surface receptors, the current techniques used to control the presentation of cell-interactive molecules on material surfaces, and the most advanced techniques for characterizing the interactions that occur between cell surface receptors and materials intended for use in regenerative medicine. Lay Summary The combination of biology, chemistry, materials science, and imaging technology affords exciting opportunities to better diagnose and treat a wide range of diseases. Recent advances in imaging technologies have enabled better understanding of the specific interactions that occur between human cells and their immediate surroundings in both health and disease. This biological understanding can be used to design smart therapies and tissue replacements that better mimic native tissue. Here, we discuss the advances in molecular biology and technologies that can be employed to functionalize materials and characterize their interaction with biological entities to facilitate the design of more sophisticated medical therapies.
Collapse
Affiliation(s)
- Stephanie A. Maynard
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Charles W. Winter
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Eoghan M. Cunnane
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Molly M. Stevens
- Department of Materials, Department of Bioengineering and Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| |
Collapse
|
144
|
Kollmetz T, Monteiro A I, Gerrard JA, Malmström J. Polystyrene- block-poly(ethylene oxide) Thin Films Fabricated from a Solvent Mixture for the Co-Assembly of Polymers and Proteins. ACS OMEGA 2020; 5:26365-26373. [PMID: 33110964 PMCID: PMC7581074 DOI: 10.1021/acsomega.0c02392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 09/28/2020] [Indexed: 06/11/2023]
Abstract
The co-assembly of peptides and proteins in poly(styrene-block-ethylene oxide) (PS-b-PEO) thin films has proven to be a promising method to fabricate polymer-biomolecule functional materials. Contrary to the covalent immobilization of biomolecules on surfaces, co-assembly presents the opportunity to arrange cargo within thin films, which can be released upon exposure to an aqueous environment. The use of a mixed solvent system ensures the solubilization of hydrophobic polymer as well as the solubilization and protection of the biomolecule cargo. However, to produce largely defect-free films of PS-b-PEO from a solvent mixture containing water is challenging due to the narrow range of solvent miscibility and polymer/protein solubility. This work explores the limits of using a benzene/methanol/water solvent mixture for the production of thin PS-b-PEO films and provides a template for the fabrication optimization of block copolymer thin films in different complex solvent systems. The film quality is analyzed using optical microscopy and atomic force microscopy and correlated to the solvent composition. By adjusting the solvent composition to 80/18.8/1.2 vol % benzene/methanol/water, it was possible to reliably fabricate thin films with less than 1% macroscopic defect surface coverage. Using the optimized solvent composition, we also demonstrate the fabrication of ordered PS-b-PEO films containing lysozyme. Furthermore, we show the release of lysozyme into aqueous media, which highlights the potential use of such films for drug delivery applications.
Collapse
Affiliation(s)
- Tarek Kollmetz
- Department
of Chemical and Materials Engineering, The
University of Auckland, Auckland 1010, New Zealand
- The
MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Isabela Monteiro A
- Department
of Chemical and Materials Engineering, The
University of Auckland, Auckland 1010, New Zealand
- The
MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| | - Juliet A. Gerrard
- The
MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
- School
of Biological Sciences, The University of
Auckland, Auckland 1010, New Zealand
- School
of Chemical Sciences, The University of
Auckland, Auckland 1010, New Zealand
| | - Jenny Malmström
- Department
of Chemical and Materials Engineering, The
University of Auckland, Auckland 1010, New Zealand
- The
MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington 6140, New Zealand
| |
Collapse
|
145
|
Min S, Jeon YS, Choi H, Khatua C, Li N, Bae G, Jung HJ, Kim Y, Hong H, Shin J, Ko MJ, Ko HS, Kim T, Moon JH, Song JJ, Dravid VP, Kim YK, Kang H. Large and Externally Positioned Ligand-Coated Nanopatches Facilitate the Adhesion-Dependent Regenerative Polarization of Host Macrophages. NANO LETTERS 2020; 20:7272-7280. [PMID: 32910662 DOI: 10.1021/acs.nanolett.0c02655] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Macrophages can associate with extracellular matrix (ECM) demonstrating nanosequenced cell-adhesive RGD ligand. In this study, we devised barcoded materials composed of RGD-coated gold and RGD-absent iron nanopatches to show various frequencies and position of RGD-coated nanopatches with similar areas of iron and RGD-gold nanopatches that maintain macroscale and nanoscale RGD density invariant. Iron patches were used for substrate coupling. Both large (low frequency) and externally positioned RGD-coated nanopatches stimulated robust attachment in macrophages, compared with small (high frequency) and internally positioned RGD-coated nanopatches, respectively, which mediate their regenerative/anti-inflammatory M2 polarization. The nanobarcodes exhibited stability in vivo. We shed light into designing ligand-engineered nanostructures in an external position to facilitate host cell attachment, thereby eliciting regenerative host responses.
Collapse
Affiliation(s)
- Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Yoo Sang Jeon
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
- Research Institute of Engineering and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Hyojun Choi
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Chandra Khatua
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Na Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Gunhyu Bae
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Hee Joon Jung
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- NUANCE Center, Northwestern University, Evanston, Illinois 60208, United States
| | - Yuri Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Jeongeun Shin
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Min Jun Ko
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Han Seok Ko
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Taesoon Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Jun Hwan Moon
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul 08308, Republic of Korea
| | - Vinayak P Dravid
- Department of Materials Science and Engineering, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- NUANCE Center, Northwestern University, Evanston, Illinois 60208, United States
| | - Young Keun Kim
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul 02841, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
146
|
Li J, Liu H, Li N, Wang J, Song L. TDCPP mimics thyroid hormones associated with the activation of integrin α vβ 3 and ERK1/2. CHEMOSPHERE 2020; 256:127066. [PMID: 32434091 DOI: 10.1016/j.chemosphere.2020.127066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 04/24/2020] [Accepted: 05/11/2020] [Indexed: 06/11/2023]
Abstract
Tri(1,3-dichloropropyl) phosphate (TDCPP) potentially damages the thyroid system in humans and animals. However, knowledge of its toxic effects and underlying mechanisms is limited. The present study was conducted to determine the thyroid hormone-disrupting effects of TDCPP and its major metabolite, bis(1,3-dichloro-2-propyl) phosphate (BDCPP) in rat pituitary cell lines (GH3). TDCPP and BDCPP, that mimic the thyroid hormone (TH), promoted GH3 cell proliferation and modulated the progression of the cell cycle at 20 and 200 μmol/L, respectively. Similar to T3, TDCPP and BDCPP also significantly upregulated c-fos and downregulated Tshβ gene expression. Although the binding affinity of these chemicals for thyroid receptor β (TRβ) was not measured, significant competition between these chemicals to bind to the membrane thyroid hormone receptor (integrin αvβ3) was found, suggesting that TDCPP and BDCPP were strongly bound to integrin αvβ3. Results from a molecular docking analysis provided further evidence of strong binding affinities of TDCPP and BDCPP for integrin αvβ3, and the ligand binding site of Arg-Gly-Asp (RGD) was identified. Real-time PCR also supported the supposition that, after binding to integrin αvβ3, TDCPP and BDCPP may induce the activation of the extracellular signal-regulated protein kinase (ERK1/2) signal transduction pathway. Taken together, our data suggest that TDCPP and BDCPP have the ability to mimic THs and that the underlying mechanism might be associated with their interactions with integrin αvβ3 and the activation of the ERK1/2 pathway, providing new insight into the mechanism of TDCPP- and BDCPP-induced cytotoxicity.
Collapse
Affiliation(s)
- Jian Li
- Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, College of Water Sciences, Beijing Normal University, Beijing, 100875, China.
| | - Hedan Liu
- Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, College of Water Sciences, Beijing Normal University, Beijing, 100875, China
| | - Na Li
- Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, 100085, China
| | - Jinsheng Wang
- Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, College of Water Sciences, Beijing Normal University, Beijing, 100875, China
| | - Liuting Song
- Engineering Research Center of Groundwater Pollution Control and Remediation, Ministry of Education, College of Water Sciences, Beijing Normal University, Beijing, 100875, China
| |
Collapse
|
147
|
Mierke CT. Mechanical Cues Affect Migration and Invasion of Cells From Three Different Directions. Front Cell Dev Biol 2020; 8:583226. [PMID: 33043017 PMCID: PMC7527720 DOI: 10.3389/fcell.2020.583226] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Cell migration and invasion is a key driving factor for providing essential cellular functions under physiological conditions or the malignant progression of tumors following downward the metastatic cascade. Although there has been plentiful of molecules identified to support the migration and invasion of cells, the mechanical aspects have not yet been explored in a combined and systematic manner. In addition, the cellular environment has been classically and frequently assumed to be homogeneous for reasons of simplicity. However, motility assays have led to various models for migration covering only some aspects and supporting factors that in some cases also include mechanical factors. Instead of specific models, in this review, a more or less holistic model for cell motility in 3D is envisioned covering all these different aspects with a special emphasis on the mechanical cues from a biophysical perspective. After introducing the mechanical aspects of cell migration and invasion and presenting the heterogeneity of extracellular matrices, the three distinct directions of cell motility focusing on the mechanical aspects are presented. These three different directions are as follows: firstly, the commonly used invasion tests using structural and structure-based mechanical environmental signals; secondly, the mechano-invasion assay, in which cells are studied by mechanical forces to migrate and invade; and thirdly, cell mechanics, including cytoskeletal and nuclear mechanics, to influence cell migration and invasion. Since the interaction between the cell and the microenvironment is bi-directional in these assays, these should be accounted in migration and invasion approaches focusing on the mechanical aspects. Beyond this, there is also the interaction between the cytoskeleton of the cell and its other compartments, such as the cell nucleus. In specific, a three-element approach is presented for addressing the effect of mechanics on cell migration and invasion by including the effect of the mechano-phenotype of the cytoskeleton, nucleus and the cell's microenvironment into the analysis. In precise terms, the combination of these three research approaches including experimental techniques seems to be promising for revealing bi-directional impacts of mechanical alterations of the cellular microenvironment on cells and internal mechanical fluctuations or changes of cells on the surroundings. Finally, different approaches are discussed and thereby a model for the broad impact of mechanics on cell migration and invasion is evolved.
Collapse
Affiliation(s)
- Claudia Tanja Mierke
- Faculty of Physics and Earth Science, Peter Debye Institute of Soft Matter Physics, Biological Physics Division, University of Leipzig, Leipzig, Germany
| |
Collapse
|
148
|
John AE, Graves RH, Pun KT, Vitulli G, Forty EJ, Mercer PF, Morrell JL, Barrett JW, Rogers RF, Hafeji M, Bibby LI, Gower E, Morrison VS, Man Y, Roper JA, Luckett JC, Borthwick LA, Barksby BS, Burgoyne RA, Barnes R, Le J, Flint DJ, Pyne S, Habgood A, Organ LA, Joseph C, Edwards-Pritchard RC, Maher TM, Fisher AJ, Gudmann NS, Leeming DJ, Chambers RC, Lukey PT, Marshall RP, Macdonald SJF, Jenkins RG, Slack RJ. Translational pharmacology of an inhaled small molecule αvβ6 integrin inhibitor for idiopathic pulmonary fibrosis. Nat Commun 2020; 11:4659. [PMID: 32938936 PMCID: PMC7494911 DOI: 10.1038/s41467-020-18397-6] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 08/17/2020] [Indexed: 12/16/2022] Open
Abstract
The αvβ6 integrin plays a key role in the activation of transforming growth factor-β (TGFβ), a pro-fibrotic mediator that is pivotal to the development of idiopathic pulmonary fibrosis (IPF). We identified a selective small molecule αvβ6 RGD-mimetic, GSK3008348, and profiled it in a range of disease relevant pre-clinical systems. To understand the relationship between target engagement and inhibition of fibrosis, we measured pharmacodynamic and disease-related end points. Here, we report, GSK3008348 binds to αvβ6 with high affinity in human IPF lung and reduces downstream pro-fibrotic TGFβ signaling to normal levels. In human lung epithelial cells, GSK3008348 induces rapid internalization and lysosomal degradation of the αvβ6 integrin. In the murine bleomycin-induced lung fibrosis model, GSK3008348 engages αvβ6, induces prolonged inhibition of TGFβ signaling and reduces lung collagen deposition and serum C3M, a marker of IPF disease progression. These studies highlight the potential of inhaled GSK3008348 as an anti-fibrotic therapy. The αvβ6 integrin is key in activating the pro-fibrotic cytokine TGFβ in idiopathic pulmonary fibrosis. Here, the authors show an inhaled small molecule αvβ6 inhibitor GSK3008348 induces prolonged inhibition of TGFβ signaling pathways in human and murine models of lung fibrosis via αvβ6 degradation.
Collapse
Affiliation(s)
- Alison E John
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Rebecca H Graves
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - K Tao Pun
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Giovanni Vitulli
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Ellen J Forty
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Paul F Mercer
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Josie L Morrell
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - John W Barrett
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Rebecca F Rogers
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Maryam Hafeji
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Lloyd I Bibby
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Elaine Gower
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Valerie S Morrison
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Yim Man
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - James A Roper
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Jeni C Luckett
- Radiological Sciences, University of Nottingham, Nottingham, UK
| | - Lee A Borthwick
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Ben S Barksby
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Rachel A Burgoyne
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Rory Barnes
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Joelle Le
- Drug Design and Selection - Molecular Design, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - David J Flint
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Anthony Habgood
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Louise A Organ
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Chitra Joseph
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | | | - Toby M Maher
- NIHR Respiratory Clinical Research Facility, Royal Brompton Hospital, London, UK.,Fibrosis Research Group, National Heart and Lung Institute, Imperial College, London, UK
| | - Andrew J Fisher
- Fibrosis Research Group, Newcastle University Biosciences Institute and Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK.,Institute of Transplantation, Freeman Hospital, Newcastle Upon Tyne Hospitals NHS, Foundation Trust, Newcastle upon Tyne, UK
| | - Natasja Stæhr Gudmann
- Nordic Bioscience A/S, Biomarkers and Research, Herlev Hovedgade 205-207, Herlev, Denmark
| | - Diana J Leeming
- Nordic Bioscience A/S, Biomarkers and Research, Herlev Hovedgade 205-207, Herlev, Denmark
| | - Rachel C Chambers
- Centre for Inflammation and Tissue Repair, University College London, London, UK
| | - Pauline T Lukey
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Richard P Marshall
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Simon J F Macdonald
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - R Gisli Jenkins
- Respiratory Medicine NIHR Biomedical Research Centre, University of Nottingham, Nottingham, UK.
| | - Robert J Slack
- Fibrosis DPU, Respiratory TAU, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| |
Collapse
|
149
|
Ghabraie E, Kemker I, Tonali N, Ismail M, Dodero VI, Sewald N. Phenothiazine-Biaryl-Containing Fluorescent RGD Peptides. Chemistry 2020; 26:12036-12042. [PMID: 32297686 PMCID: PMC7540173 DOI: 10.1002/chem.202001312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Indexed: 12/22/2022]
Abstract
Cyclic RGD peptides are well-known ligands of integrins. The integrins αV β3 and α5 β1 are involved in angiogenesis, and integrin αV β3 is abundantly present on cancer cells, thus representing a therapeutic target. Hence, synthetic and biophysical studies continuously are being directed towards the understanding of ligand-integrin interaction. In this context, the development of versatile synthetic strategies to obtain fluorescent building blocks that can add molecular diversity and modular spectral characteristics while not compromising binding affinity or selectivity is a relevant task. An on-resin intramolecular Suzuki-Miyaura cross-coupling (SMC) between l- or d-7-bromotryptophan (7BrTrp) and a phenothiazine (Ptz) boronic acid affords fluorescent cyclic RGD pseudopeptides, c(RGD(W/w)Ptz). Ring closure by SMC establishes a phenothiazine-indole moiety with axial chirality. An array of eight novel compounds has been synthesized, among them one fluorescent compound with good affinity to integrin αV β3 . The fluorescence properties of the analogues can be efficiently tuned depending on the substituents in Ptz moiety even for fluorescence emission in the visible (red) spectral range.
Collapse
Affiliation(s)
- Elmira Ghabraie
- Department of ChemistryOrganic and Bioorganic ChemistryBielefeld UniversityPO Box 10013133501BielefeldGermany
| | - Isabell Kemker
- Department of ChemistryOrganic and Bioorganic ChemistryBielefeld UniversityPO Box 10013133501BielefeldGermany
| | - Nicolo Tonali
- Department of ChemistryOrganic and Bioorganic ChemistryBielefeld UniversityPO Box 10013133501BielefeldGermany
| | - Mohamed Ismail
- Department of ChemistryOrganic and Bioorganic ChemistryBielefeld UniversityPO Box 10013133501BielefeldGermany
| | - Veronica I. Dodero
- Department of ChemistryOrganic and Bioorganic ChemistryBielefeld UniversityPO Box 10013133501BielefeldGermany
| | - Norbert Sewald
- Department of ChemistryOrganic and Bioorganic ChemistryBielefeld UniversityPO Box 10013133501BielefeldGermany
| |
Collapse
|
150
|
Redondo-Gómez C, Padilla-Lopategui S, Azevedo HS, Mata A. Host-Guest-Mediated Epitope Presentation on Self-Assembled Peptide Amphiphile Hydrogels. ACS Biomater Sci Eng 2020; 6:4870-4880. [PMID: 33455284 DOI: 10.1021/acsbiomaterials.0c00549] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A key feature in biomaterial design is the incorporation of bioactive signals into artificial constructs to stimulate tissue regeneration. Most currently used hydrogel cell culture systems depend on the covalent attachment of extracellular matrix (ECM)-derived peptides to either macromolecular units or smaller self-assembling building blocks, thereby restricting biosignal presentation and adaptability. However, new ways to rationally incorporate adhesion epitopes through noncovalent interactions would offer opportunities to better recreate the dynamic and reversible nature of the native ECM. Here, we report on a noncovalent epitope presentation approach mediated by host-guest interactions. Using peptide amphiphile hydrogels, we demonstrate that the adamantane/β-cyclodextrin pair can be used to anchor RGDS cell adhesion signals onto self-assembled hydrogels via host-guest interactions. We evaluate hydrogel morphological and rheological properties as well as fibroblast attachment, organization, and spreading when cultured atop these scaffolds. This host-guest-mediated epitope display might lead to new self-assembling hydrogels for improved cell culture applications in fields such as tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Carlos Redondo-Gómez
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, U.K.,Institute of Bioengineering, Queen Mary University of London, London E1 4NS, U.K
| | - Soraya Padilla-Lopategui
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, U.K.,Institute of Bioengineering, Queen Mary University of London, London E1 4NS, U.K
| | - Helena S Azevedo
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, U.K.,Institute of Bioengineering, Queen Mary University of London, London E1 4NS, U.K
| | - Alvaro Mata
- School of Engineering & Materials Science, Queen Mary University of London, London E1 4NS, U.K.,Institute of Bioengineering, Queen Mary University of London, London E1 4NS, U.K.,School of Pharmacy, University of Nottingham, University Park, Nottingham NG7 2RD, U.K.,Department of Chemical and Environmental Engineering, University of Nottingham, University Park, Nottingham NG7 2RD, U.K.,Biodiscovery Institute, University of Nottingham, University Park, Nottingham NG7 2RD, U.K
| |
Collapse
|