51
|
Zhang J, Ten Dijke P, Wuhrer M, Zhang T. Role of glycosylation in TGF-β signaling and epithelial-to-mesenchymal transition in cancer. Protein Cell 2021; 12:89-106. [PMID: 32583064 PMCID: PMC7862465 DOI: 10.1007/s13238-020-00741-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/29/2020] [Indexed: 12/14/2022] Open
Abstract
Glycosylation is a common posttranslational modification on membrane-associated and secreted proteins that is of pivotal importance for regulating cell functions. Aberrant glycosylation can lead to uncontrolled cell proliferation, cell-matrix interactions, migration and differentiation, and has been shown to be involved in cancer and other diseases. The epithelial-to-mesenchymal transition is a key step in the metastatic process by which cancer cells gain the ability to invade tissues and extravasate into the bloodstream. This cellular transformation process, which is associated by morphological change, loss of epithelial traits and gain of mesenchymal markers, is triggered by the secreted cytokine transforming growth factor-β (TGF-β). TGF-β bioactivity is carefully regulated, and its effects on cells are mediated by its receptors on the cell surface. In this review, we first provide a brief overview of major types of glycans, namely, N-glycans, O-glycans, glycosphingolipids and glycosaminoglycans that are involved in cancer progression. Thereafter, we summarize studies on how the glycosylation of TGF-β signaling components regulates TGF-β secretion, bioavailability and TGF-β receptor function. Then, we review glycosylation changes associated with TGF-β-induced epithelial-to-mesenchymal transition in cancer. Identifying and understanding the mechanisms by which glycosylation affects TGF-β signaling and downstream biological responses will facilitate the identification of glycans as biomarkers and enable novel therapeutic approaches.
Collapse
Affiliation(s)
- Jing Zhang
- Oncode Institute and Cell Chemical Biology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands
| | - Peter Ten Dijke
- Oncode Institute and Cell Chemical Biology, Leiden University Medical Center, 2300 RC, Leiden, The Netherlands.
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| | - Tao Zhang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
52
|
Zhang J, Wang T, Saigal A, Johnson J, Morrisson J, Tabrizifard S, Hollingsworth SA, Eddins MJ, Mao W, O'Neill K, Garcia-Calvo M, Carballo-Jane E, Liu D, Ham T, Zhou Q, Dong W, Meng HW, Hicks J, Cai TQ, Akiyama T, Pinto S, Cheng AC, Greshock T, Marquis JC, Ren Z, Talukdar S, Shaheen HH, Handa M. Discovery of a new class of integrin antibodies for fibrosis. Sci Rep 2021; 11:2118. [PMID: 33483531 PMCID: PMC7822819 DOI: 10.1038/s41598-021-81253-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 01/05/2021] [Indexed: 12/14/2022] Open
Abstract
Lung fibrosis, or the scarring of the lung, is a devastating disease with huge unmet medical need. There are limited treatment options and its prognosis is worse than most types of cancer. We previously discovered that MK-0429 is an equipotent pan-inhibitor of αv integrins that reduces proteinuria and kidney fibrosis in a preclinical model. In the present study, we further demonstrated that MK-0429 significantly inhibits fibrosis progression in a bleomycin-induced lung injury model. In search of newer integrin inhibitors for fibrosis, we characterized monoclonal antibodies discovered using Adimab's yeast display platform. We identified several potent neutralizing integrin antibodies with unique human and mouse cross-reactivity. Among these, Ab-31 blocked the binding of multiple αv integrins to their ligands with IC50s comparable to those of MK-0429. Furthermore, both MK-0429 and Ab-31 suppressed integrin-mediated cell adhesion and latent TGFβ activation. In IPF patient lung fibroblasts, TGFβ treatment induced profound αSMA expression in phenotypic imaging assays and Ab-31 demonstrated potent in vitro activity at inhibiting αSMA expression, suggesting that the integrin antibody is able to modulate TGFβ action though mechanisms beyond the inhibition of latent TGFβ activation. Together, our results highlight the potential to develop newer integrin therapeutics for the treatment of fibrotic lung diseases.
Collapse
Affiliation(s)
- Ji Zhang
- Departments of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA.
| | - Tao Wang
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Ashmita Saigal
- Departments of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Josephine Johnson
- Quantitative Biosciences, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Jennifer Morrisson
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Sahba Tabrizifard
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Scott A Hollingsworth
- Computational & Structural Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Michael J Eddins
- Computational & Structural Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Wenxian Mao
- Quantitative Biosciences, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Kim O'Neill
- In Vitro Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Margarita Garcia-Calvo
- In Vitro Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Ester Carballo-Jane
- Quantitative Biosciences, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - DingGang Liu
- SALAR, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Taewon Ham
- SALAR, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Qiong Zhou
- SALAR, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Weifeng Dong
- SALAR, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Hsien-Wei Meng
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Jacqueline Hicks
- Discovery Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Tian-Quan Cai
- In Vivo Pharmacology, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Taro Akiyama
- Departments of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Shirly Pinto
- Departments of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Alan C Cheng
- Computational & Structural Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Thomas Greshock
- Discovery Chemistry, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - John C Marquis
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Zhao Ren
- Quantitative Biosciences, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Saswata Talukdar
- Departments of Cardiometabolic Diseases, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Hussam Hisham Shaheen
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA
| | - Masahisa Handa
- Discovery Biologics, MRL, Merck & Co., Inc., 2000 Galloping Hill Road, Kenilworth, NJ, 07033, USA.
| |
Collapse
|
53
|
Mészáros B, Sámano-Sánchez H, Alvarado-Valverde J, Čalyševa J, Martínez-Pérez E, Alves R, Shields DC, Kumar M, Rippmann F, Chemes LB, Gibson TJ. Short linear motif candidates in the cell entry system used by SARS-CoV-2 and their potential therapeutic implications. Sci Signal 2021; 14:eabd0334. [PMID: 33436497 PMCID: PMC7928535 DOI: 10.1126/scisignal.abd0334] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 12/10/2020] [Indexed: 12/12/2022]
Abstract
The first reported receptor for SARS-CoV-2 on host cells was the angiotensin-converting enzyme 2 (ACE2). However, the viral spike protein also has an RGD motif, suggesting that cell surface integrins may be co-receptors. We examined the sequences of ACE2 and integrins with the Eukaryotic Linear Motif (ELM) resource and identified candidate short linear motifs (SLiMs) in their short, unstructured, cytosolic tails with potential roles in endocytosis, membrane dynamics, autophagy, cytoskeleton, and cell signaling. These SLiM candidates are highly conserved in vertebrates and may interact with the μ2 subunit of the endocytosis-associated AP2 adaptor complex, as well as with various protein domains (namely, I-BAR, LC3, PDZ, PTB, and SH2) found in human signaling and regulatory proteins. Several motifs overlap in the tail sequences, suggesting that they may act as molecular switches, such as in response to tyrosine phosphorylation status. Candidate LC3-interacting region (LIR) motifs are present in the tails of integrin β3 and ACE2, suggesting that these proteins could directly recruit autophagy components. Our findings identify several molecular links and testable hypotheses that could uncover mechanisms of SARS-CoV-2 attachment, entry, and replication against which it may be possible to develop host-directed therapies that dampen viral infection and disease progression. Several of these SLiMs have now been validated to mediate the predicted peptide interactions.
Collapse
Affiliation(s)
- Bálint Mészáros
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.
| | - Hugo Sámano-Sánchez
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Jesús Alvarado-Valverde
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences
| | - Jelena Čalyševa
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences
| | - Elizabeth Martínez-Pérez
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
- Laboratorio de bioinformática estructural, Fundación Instituto Leloir, C1405BWE Buenos Aires, Argentina
| | - Renato Alves
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany
| | - Denis C Shields
- School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Manjeet Kumar
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.
| | - Friedrich Rippmann
- Computational Chemistry & Biology, Merck KGaA, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Lucía B Chemes
- Instituto de Investigaciones Biotecnológicas "Dr. Rodolfo A. Ugalde", IIB-UNSAM, IIBIO-CONICET, Universidad Nacional de San Martín, CP1650 San Martín, Buenos Aires, Argentina.
| | - Toby J Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg 69117, Germany.
| |
Collapse
|
54
|
Mezu-Ndubuisi OJ, Maheshwari A. The role of integrins in inflammation and angiogenesis. Pediatr Res 2021; 89:1619-1626. [PMID: 33027803 PMCID: PMC8249239 DOI: 10.1038/s41390-020-01177-9] [Citation(s) in RCA: 218] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/18/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023]
Abstract
Integrins are heterodimeric transmembrane cell adhesion molecules made up of alpha (α) and beta (β) subunits arranged in numerous dimeric pairings. These complexes have varying affinities to extracellular ligands. Integrins regulate cellular growth, proliferation, migration, signaling, and cytokine activation and release and thereby play important roles in cell proliferation and migration, apoptosis, tissue repair, as well as in all processes critical to inflammation, infection, and angiogenesis. This review presents current evidence from human and animal studies on integrin structure and molecular signaling, with particular emphasis on signal transduction in infants. We have included evidence from our own laboratory studies and from an extensive literature search in databases PubMed, EMBASE, Scopus, and the electronic archives of abstracts presented at the annual meetings of the Pediatric Academic Societies. To avoid bias in identification of existing studies, key words were short-listed prior to the actual search both from anecdotal experience and from PubMed's Medical Subject Heading (MeSH) thesaurus. IMPACT: Integrins are a family of ubiquitous αβ heterodimeric receptors that interact with numerous ligands in physiology and disease. Integrins play a key role in cell proliferation, tissue repair, inflammation, infection, and angiogenesis. This review summarizes current evidence from human and animal studies on integrin structure and molecular signaling and promising role in diseases of inflammation, infection, and angiogenesis in infants. This review shows that integrin receptors and ligands are novel therapeutic targets of clinical interest and hold promise as novel therapeutic targets in the management of several neonatal diseases.
Collapse
Affiliation(s)
- Olachi J. Mezu-Ndubuisi
- grid.14003.360000 0001 2167 3675Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI USA
| | - Akhil Maheshwari
- grid.21107.350000 0001 2171 9311Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| |
Collapse
|
55
|
Urquiza M, Guevara V, Diaz-Sana E, Mora F. The Role of αvβ6 Integrin Binding Molecules in the Diagnosis and Treatment of Cancer. CURR ORG CHEM 2020. [DOI: 10.2174/1385272824999200528124936] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Peptidic and non-peptidic αvβ6 integrin-binding molecules have been used in
the clinic for detection and treatment of tumors expressing αvβ6 integrin, because this protein
is expressed in malignant epithelial cells of the oral cavity, pancreas, breast, ovary,
colon and stomach carcinomas but it is not expressed in healthy adult tissue except during
wound healing and inflammation. This review focuses on the landscape of αvβ6 integrinbinding
molecules and their use in cancer treatment and detection, and discusses recent
designs for tumor detection, treatment, and immunotherapy. In the last ten years, several
reviews abamp;#945;vβ6 integrin-binding molecules and their role in cancer detection and treatment.
Firstly, this review describes the role of the αvβ6 integrin in normal tissues, how the expression
of this protein is correlated with cancer severity and its role in cancer development. Taking into account
the potential of αvβ6 integrin-binding molecules in detection and treatment of specific tumors, special
attention is given to several high-affinity αvβ6 integrin-binding peptides used for tumor imaging; particularly,
the αvβ6-binding peptide NAVPNLRGDLQVLAQKVART [A20FMDV2], derived from the foot and mouth
disease virus. This peptide labeled with either 18F, 111In or with 68Ga has been used for PET imaging of αvβ6
integrin-positive tumors. Moreover, αvβ6 integrin-binding peptides have been used for photoacoustic and fluorescence
imaging and could potentially be used in clinical application in cancer diagnosis and intraoperative
imaging of αvβ6-integrin positive tumors. Additionally, non-peptidic αvβ6-binding molecules have been designed
and used in the clinic for the detection and treatment of αvβ6-expressing tumors. Anti-αvβ6 integrin antibodies
are another useful tool for selective identification and treatment of αvβ6 (+) tumors. The utility of
these αvβ6 integrin-binding molecules as a tool for tumor detection and treatment is discussed, considering
specificity, sensitivity and serum stability. Another use of the αvβ6 integrin-binding peptides is to modify the
Ad5 cell tropism for inducing oncolytic activity of αvβ6-integrin positive tumor cells by expressing
A20FMDV2 peptide within the fiber knob protein (Ad5NULL-A20). The newly designed oncolytic
Ad5NULL-A20 virotherapy is promising for local and systemic targeting of αvβ6-overexpressing cancers. Finally,
new evidence has emerged, indicating that chimeric antigen receptor (CAR) containing the αvβ6 integrin-
binding peptide on top of CD28+CD3 endodomain displays a potent therapeutic activity in a diverse
repertoire of solid tumor models.
Collapse
Affiliation(s)
- Mauricio Urquiza
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| | - Valentina Guevara
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| | - Erika Diaz-Sana
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| | - Felipe Mora
- Grupo de Investigacion en Hormonas (GIH), Department of Chemistry, National University of Columbia, Cra 30 # 45-03, Bogota, zip code 111321, Colombia
| |
Collapse
|
56
|
Henderson NC, Rieder F, Wynn TA. Fibrosis: from mechanisms to medicines. Nature 2020; 587:555-566. [PMID: 33239795 DOI: 10.1038/s41586-020-2938-9] [Citation(s) in RCA: 1058] [Impact Index Per Article: 211.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022]
Abstract
Fibrosis can affect any organ and is responsible for up to 45% of all deaths in the industrialized world. It has long been thought to be relentlessly progressive and irreversible, but both preclinical models and clinical trials in various organ systems have shown that fibrosis is a highly dynamic process. This has clear implications for therapeutic interventions that are designed to capitalize on this inherent plasticity. However, despite substantial progress in our understanding of the pathobiology of fibrosis, a translational gap remains between the identification of putative antifibrotic targets and conversion of this knowledge into effective treatments in humans. Here we discuss the transformative experimental strategies that are being leveraged to dissect the key cellular and molecular mechanisms that regulate fibrosis, and the translational approaches that are enabling the emergence of precision medicine-based therapies for patients with fibrosis.
Collapse
Affiliation(s)
- Neil C Henderson
- University of Edinburgh Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, Edinburgh, UK.,MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Florian Rieder
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, USA.,Department of Gastroenterology, Hepatology and Nutrition, Digestive Diseases and Surgery Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Thomas A Wynn
- Inflammation & Immunology Research Unit, Pfizer Worldwide Research, Development & Medical, Cambridge, MA, USA.
| |
Collapse
|
57
|
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: 6] [Impact Index Per Article: 1.2] [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
|
58
|
Bugatti K, Bruno A, Arosio D, Sartori A, Curti C, Augustijn L, Zanardi F, Battistini L. Shifting Towards α
V
β
6
Integrin Ligands Using Novel Aminoproline‐Based Cyclic Peptidomimetics. Chemistry 2020; 26:13468-13475. [DOI: 10.1002/chem.202002554] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Indexed: 01/04/2023]
Affiliation(s)
- Kelly Bugatti
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27A 43124 Parma Italy
| | - Agostino Bruno
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27A 43124 Parma Italy
| | - Daniela Arosio
- Istituto di Scienze e Tecnologie Chimiche (SCITEC) “Giulio Natta” CNR, Consiglio Nazionale delle Ricerche Via C. Golgi 19 20133 Milano Italy
| | - Andrea Sartori
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27A 43124 Parma Italy
| | - Claudio Curti
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27A 43124 Parma Italy
| | - Lisa Augustijn
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS) Division of Medicinal Chemistry Vrije Universiteit Amsterdam De Boelelaan 1108, 1081 HZ Amsterdam Noord-Holland The Netherlands
| | - Franca Zanardi
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27A 43124 Parma Italy
| | - Lucia Battistini
- Dipartimento di Scienze degli Alimenti e del Farmaco Università di Parma Parco Area delle Scienze 27A 43124 Parma Italy
| |
Collapse
|
59
|
Duffy C, Sorolla A, Wang E, Golden E, Woodward E, Davern K, Ho D, Johnstone E, Pfleger K, Redfern A, Iyer KS, Baer B, Blancafort P. Honeybee venom and melittin suppress growth factor receptor activation in HER2-enriched and triple-negative breast cancer. NPJ Precis Oncol 2020; 4:24. [PMID: 32923684 PMCID: PMC7463160 DOI: 10.1038/s41698-020-00129-0] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Despite decades of study, the molecular mechanisms and selectivity of the biomolecular components of honeybee (Apis mellifera) venom as anticancer agents remain largely unknown. Here, we demonstrate that honeybee venom and its major component melittin potently induce cell death, particularly in the aggressive triple-negative and HER2-enriched breast cancer subtypes. Honeybee venom and melittin suppress the activation of EGFR and HER2 by interfering with the phosphorylation of these receptors in the plasma membrane of breast carcinoma cells. Mutational studies reveal that a positively charged C-terminal melittin sequence mediates plasma membrane interaction and anticancer activity. Engineering of an RGD motif further enhances targeting of melittin to malignant cells with minimal toxicity to normal cells. Lastly, administration of melittin enhances the effect of docetaxel in suppressing breast tumor growth in an allograft model. Our work unveils a molecular mechanism underpinning the anticancer selectivity of melittin, and outlines treatment strategies to target aggressive breast cancers.
Collapse
Affiliation(s)
- Ciara Duffy
- School of Human Sciences, The University of Western Australia, Perth, WA 6009 Australia.,Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Plant Energy Biology, The University of Western Australia, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia
| | - Anabel Sorolla
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia
| | - Edina Wang
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia
| | - Emily Golden
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia
| | - Eleanor Woodward
- Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia
| | - Kathleen Davern
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia.,Monoclonal Antibody (MAb) Facility, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia
| | - Diwei Ho
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009 Australia
| | - Elizabeth Johnstone
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia.,Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, Australia
| | - Kevin Pfleger
- Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia.,Molecular Endocrinology and Pharmacology, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Australian Research Council Centre for Personalised Therapeutics Technologies, Perth, Australia.,Dimerix Limited; Nedlands, Perth, WA 6009 Australia
| | - Andrew Redfern
- School of Medicine, The University of Western Australia, Perth, WA 6009 Australia
| | - K Swaminathan Iyer
- Monoclonal Antibody (MAb) Facility, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia
| | - Boris Baer
- Centre for Integrative Bee Research (CIBER), Department of Entomology; University of California Riverside, Riverside, CA 92521 USA
| | - Pilar Blancafort
- School of Human Sciences, The University of Western Australia, Perth, WA 6009 Australia.,Cancer Epigenetics Group, Harry Perkins Institute of Medical Research, Perth, WA 6009 Australia.,Centre for Medical Research, The University of Western Australia, Perth, WA 6009 Australia.,The Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| |
Collapse
|
60
|
Mao D, Lü S, Zhang X, Long M. Mechanically Regulated Outside-In Activation of an I-Domain-Containing Integrin. Biophys J 2020; 119:966-977. [PMID: 32814058 DOI: 10.1016/j.bpj.2020.07.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 07/18/2020] [Accepted: 07/27/2020] [Indexed: 10/23/2022] Open
Abstract
Integrins are heterodimeric transmembrane proteins that mediate cellular adhesion and bidirectional mechanotransductions through their conformational allostery. The allosteric pathway of an I-domain-containing integrin remains unclear because of its complexity and lack of effective experiments. For a typical I-domain-containing integrin αXβ2, molecular dynamics simulations were employed here to investigate the conformational dynamics in the first two steps of outside-in activation, the bindings of both the external and internal ligands. Results showed that the internal ligand binding is a prerequisite to the allosteric transmission from the α- to β-subunits and the exertion of external force to integrin-ligand complex. The opening state of αI domain with downward movement and lower half unfolding of α7-helix ensures the stable intersubunit conformational transmission through external ligand binding first and internal ligand binding later. Reverse binding order induces a, to our knowledge, novel but unstable swingout of β-subunit Hybrid domain with the retained close states of both αI and βI domains. Prebinding of external ligand greatly facilitates the following internal ligand binding and vice versa. These simulations furthered the understanding in the outside-in activation of I-domain-containing integrins from the viewpoint of internal allosteric pathways.
Collapse
Affiliation(s)
- Debin Mao
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Shouqin Lü
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.
| | - Xiao Zhang
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China
| | - Mian Long
- Center of Biomechanics and Bioengineering, Key Laboratory of Microgravity (National Microgravity Laboratory), Beijing Key Laboratory of Engineered Construction and Mechanobiology, and CAS Center for Excellence in Complex System Mechanics, Institute of Mechanics, Chinese Academy of Sciences, Beijing, China; School of Engineering Science, University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
61
|
Sorolla A, Sorolla MA, Wang E, Ceña V. Peptides, proteins and nanotechnology: a promising synergy for breast cancer targeting and treatment. Expert Opin Drug Deliv 2020; 17:1597-1613. [PMID: 32835538 DOI: 10.1080/17425247.2020.1814733] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION The use of nanoparticles for breast cancer targeting and treatment has become a reality. They are safe and possess interesting peculiarities such as the unspecific accumulation into the tumor site and the possibility to activate controlled drug release as compared to free drugs. However, there are still many areas of improvement which can certainly be addressed with the use of peptide-based elements. AREAS COVERED The article reviews different preclinical strategies employing peptides and proteins in combination with nanoparticles for breast cancer targeting and treatment as well as peptide and protein-targeted encapsulated drugs, and it lists the current clinical status of therapies using peptides and proteins for breast cancer. EXPERT OPINION The conjugation of protein and peptides can improve tumor homing of nanoparticles, increase cellular penetration and attack specific drivers and vulnerabilities of the breast cancer cell to promote tumor cytotoxicity while reducing secondary effects in healthy tissues. Examples are the use of antibodies, arginylglycylaspartic acid (RGD) peptides, membrane disruptive peptides, interference peptides, and peptide vaccines. Although their implementation in the clinic has been relatively slow up to now, we anticipate great progress in the field which will translate into more efficacious and selective nanotherapies for breast cancer.
Collapse
Affiliation(s)
- Anabel Sorolla
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia , Crawley, Australia
| | - Maria Alba Sorolla
- Biomedical Research Institute (IRB Lleida), Research Group of Cancer Biomarkers , Lleida, Spain
| | - Edina Wang
- Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, The University of Western Australia , Crawley, Australia
| | - Valentín Ceña
- Unidad Asociada Neurodeath, Universidad De Castilla-La Mancha , Albacete, Spain.,Centro De Investigación En Red Sobre Enfermedades Neurodegenerativas (CIBERNED), ISCIII , Madrid, Spain
| |
Collapse
|
62
|
Lippa RA, Barrett J, Pal S, Rowedder JE, Murphy JA, Barrett TN. Discovery of the first potent and selective α vβ 5 integrin inhibitor based on an amide-containing core. Eur J Med Chem 2020; 208:112719. [PMID: 32865176 DOI: 10.1016/j.ejmech.2020.112719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/31/2020] [Accepted: 08/02/2020] [Indexed: 01/25/2023]
Abstract
Integrins αvβ5 and αvβ3 are closely related, proangiogenic members of the wider RGD-binding integrin family. Due to their high sequence homology, the development of αvβ5-selective compounds has remained elusive to synthetic and medicinal chemists. Herein, we describe a survey of SAR around a series of amide-containing 3-aryl-succinamic acid-based RGD mimetics. This resulted in the discovery of α,α,α-trifluorotolyl 12 which exhibits 800 × selectivity for αvβ5versus αvβ3 with a pyrrolidine amide linker that confers selectivity for αvβ5 by positioning a key aryl ring in the SDL of αvβ5 with good complementarity; binding in this mode is disfavoured in αvβ3 due to clashes with key residues in the β3-subunit. Compound 12 exhibits selective inhibition by a cell adhesion assay, high passive permeability and solubility which enables potential use of this inhibitor as an αvβ5-selective in vitro tool compound.
Collapse
Affiliation(s)
- Rhys A Lippa
- Department of Pure & Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, Scotland, UK
| | - John Barrett
- GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Sandeep Pal
- GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - James E Rowedder
- GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - John A Murphy
- Department of Pure & Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, Scotland, UK
| | - Tim N Barrett
- GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK.
| |
Collapse
|
63
|
Weil P, van den Bruck R, Ziegenhals T, Juranek S, Goedde D, Orth V, Wirth S, Jenke AC, Postberg J. β6 integrinosis: a new lethal autosomal recessive ITGB6 disorder leading to impaired conformational transitions of the α Vβ6 integrin receptor. Gut 2020; 69:1359-1361. [PMID: 31201286 PMCID: PMC7306976 DOI: 10.1136/gutjnl-2019-319015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 05/26/2019] [Accepted: 05/30/2019] [Indexed: 01/01/2023]
Affiliation(s)
- Patrick Weil
- Clinical Molecular Genetics and Epigenetics, Centre for Biomedical Education and Research (ZBAF), HELIOS University Hospital Wuppertal, Witten/Herdecke University, Wuppertal, Germany
| | - Rhea van den Bruck
- Department of Paediatrics, HELIOS University Hospital Wuppertal, Wuppertal, Germany
| | - Thomas Ziegenhals
- Chair of Biochemistry, Theodor-Boveri-Institute at the Biocenter, University of Würzburg, Wurzburg, Germany
| | - Stefan Juranek
- Chair of Biochemistry, Theodor-Boveri-Institute at the Biocenter, University of Würzburg, Wurzburg, Germany
| | - Daniel Goedde
- Department of Pathology, HELIOS University Hospital Wuppertal, Wuppertal, Germany
| | - Valerie Orth
- Department of Surgery II, HELIOS University Hospital Wuppertal, Wuppertal, Germany
| | - Stefan Wirth
- Department of Paediatrics, HELIOS University Hospital Wuppertal, Wuppertal, Germany
| | - Andreas C Jenke
- Clinical Molecular Genetics and Epigenetics, Centre for Biomedical Education and Research (ZBAF), HELIOS University Hospital Wuppertal, Witten/Herdecke University, Wuppertal, Germany,Department of Neonatology and General Pediatrics, Children’s Hospital Kassel, Kassel, Germany
| | - Jan Postberg
- Clinical Molecular Genetics and Epigenetics, Centre for Biomedical Education and Research (ZBAF), HELIOS University Hospital Wuppertal, Witten/Herdecke University, Wuppertal, Germany
| |
Collapse
|
64
|
Robinson H, Oatley SA, Rowedder JE, Slade P, Macdonald SJF, Argent SP, Hirst JD, McInally T, Moody CJ. Late-Stage Functionalization by Chan-Lam Amination: Rapid Access to Potent and Selective Integrin Inhibitors. Chemistry 2020; 26:7678-7684. [PMID: 32129907 DOI: 10.1002/chem.202001059] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Indexed: 11/07/2022]
Abstract
A late-stage functionalization of the aromatic ring in amino acid derivatives is described. The key step is a copper-catalysed diversification of a boronate ester by amination (Chan-Lam reaction) that can be carried out on a complex β-aryl-β-amino acid scaffold. This not only considerably extends the substrate scope of amination partners, but also delivers an array of potent and selective integrin inhibitors as potential treatment agents of idiopathic pulmonary fibrosis (IPF). This versatile chemical strategy, which is amenable to high-throughput-array protocols, allows the installation of pharmaceutically valuable heteroaromatic fragments at a late stage by direct coupling to NH heterocycles, leading to compounds with drug-like attributes. It thus constitutes a useful addition to the medicinal chemist's repertoire.
Collapse
Affiliation(s)
- Henry Robinson
- School of Chemistry, GSK Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham, NG7 2TU, UK
| | - Steven A Oatley
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - James E Rowedder
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Pawel Slade
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Simon J F Macdonald
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, UK
| | - Stephen P Argent
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Jonathan D Hirst
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| | - Thomas McInally
- School of Chemistry, GSK Carbon Neutral Laboratories for Sustainable Chemistry, University of Nottingham, Jubilee Campus, Triumph Road, Nottingham, NG7 2TU, UK
| | - Christopher J Moody
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, UK
| |
Collapse
|
65
|
McCarty JH. αvβ8 integrin adhesion and signaling pathways in development, physiology and disease. J Cell Sci 2020; 133:133/12/jcs239434. [PMID: 32540905 DOI: 10.1242/jcs.239434] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Cells must interpret a complex milieu of extracellular cues to modulate intracellular signaling events linked to proliferation, differentiation, migration and other cellular processes. Integrins are heterodimeric transmembrane proteins that link the extracellular matrix (ECM) to the cytoskeleton and control intracellular signaling events. A great deal is known about the structural and functional properties for most integrins; however, the adhesion and signaling pathways controlled by αvβ8 integrin, which was discovered nearly 30 years ago, have only recently been characterized. αvβ8 integrin is a receptor for ECM-bound forms of latent transforming growth factor β (TGFβ) proteins and promotes the activation of TGFβ signaling pathways. Studies of the brain, lung and immune system reveal that the αvβ8 integrin-TGFβ axis mediates cell-cell contact and communication within complex multicellular structures. Perturbing components of this axis results in aberrant cell-cell adhesion and signaling leading to the initiation of various pathologies, including neurodegeneration, fibrosis and cancer. As discussed in this Review, understanding the functions for αvβ8 integrin, its ECM ligands and intracellular effector proteins is not only an important topic in cell biology, but may lead to new therapeutic strategies to treat human pathologies related to integrin dysfunction.
Collapse
Affiliation(s)
- Joseph H McCarty
- Department of Neurosurgery, Brain Tumor Center, M.D. Anderson Cancer Center, 6767 Bertner Avenue, Unit 1004, Houston, TX 77030, USA
| |
Collapse
|
66
|
Nešić D, Zhang Y, Spasic A, Li J, Provasi D, Filizola M, Walz T, Coller BS. Cryo-Electron Microscopy Structure of the αIIbβ3-Abciximab Complex. Arterioscler Thromb Vasc Biol 2020; 40:624-637. [PMID: 31969014 PMCID: PMC7047619 DOI: 10.1161/atvbaha.119.313671] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/30/2019] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The αIIbβ3 antagonist antiplatelet drug abciximab is the chimeric antigen-binding fragment comprising the variable regions of murine monoclonal antibody 7E3 and the constant domains of human IgG1 and light chain κ. Previous mutagenesis studies suggested that abciximab binds to the β3 C177-C184 specificity-determining loop (SDL) and Trp129 on the adjacent β1-α1 helix. These studies could not, however, assess whether 7E3 or abciximab prevents fibrinogen binding by steric interference, disruption of either the αIIbβ3-binding pocket for fibrinogen or the β3 SDL (which is not part of the binding pocket but affects fibrinogen binding), or some combination of these effects. To address this gap, we used cryo-electron microscopy to determine the structure of the αIIbβ3-abciximab complex at 2.8 Å resolution. Approach and Results: The interacting surface of abciximab is comprised of residues from all 3 complementarity-determining regions of both the light and heavy chains, with high representation of aromatic residues. Binding is primarily to the β3 SDL and neighboring residues, the β1-α1 helix, and β3 residues Ser211, Val212 and Met335. Unexpectedly, the structure also indicated several interactions with αIIb. As judged by the cryo-electron microscopy model, molecular-dynamics simulations, and mutagenesis, the binding of abciximab does not appear to rely on the interaction with the αIIb residues and does not result in disruption of the fibrinogen-binding pocket; it does, however, compress and reduce the flexibility of the SDL. CONCLUSIONS We deduce that abciximab prevents ligand binding by steric interference, with a potential contribution via displacement of the SDL and limitation of the flexibility of the SDL residues.
Collapse
Affiliation(s)
- Dragana Nešić
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, Rockefeller University, New York, NY
| | - Yixiao Zhang
- Laboratory of Molecular Electron Microscopy, Rockefeller University, New York, NY
| | - Aleksandar Spasic
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jihong Li
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, Rockefeller University, New York, NY
| | - Davide Provasi
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Marta Filizola
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Thomas Walz
- Laboratory of Molecular Electron Microscopy, Rockefeller University, New York, NY
| | - Barry S. Coller
- Allen and Frances Adler Laboratory of Blood and Vascular Biology, Rockefeller University, New York, NY
| |
Collapse
|
67
|
Campbell MG, Cormier A, Ito S, Seed RI, Bondesson AJ, Lou J, Marks JD, Baron JL, Cheng Y, Nishimura SL. Cryo-EM Reveals Integrin-Mediated TGF-β Activation without Release from Latent TGF-β. Cell 2020; 180:490-501.e16. [PMID: 31955848 PMCID: PMC7238552 DOI: 10.1016/j.cell.2019.12.030] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 10/15/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023]
Abstract
Integrin αvβ8 binds with exquisite specificity to latent transforming growth factor-β (L-TGF-β). This binding is essential for activating L-TGF-β presented by a variety of cell types. Inhibiting αvβ8-mediated TGF-β activation blocks immunosuppressive regulatory T cell differentiation, which is a potential therapeutic strategy in cancer. Using cryo-electron microscopy, structure-guided mutagenesis, and cell-based assays, we reveal the binding interactions between the entire αvβ8 ectodomain and its intact natural ligand, L-TGF-β, as well as two different inhibitory antibody fragments to understand the structural underpinnings of αvβ8 binding specificity and TGF-β activation. Our studies reveal a mechanism of TGF-β activation where mature TGF-β signals within the confines of L-TGF-β and the release and diffusion of TGF-β are not required. The structural details of this mechanism provide a rational basis for therapeutic strategies to inhibit αvβ8-mediated L-TGF-β activation.
Collapse
Affiliation(s)
- Melody G Campbell
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
| | - Anthony Cormier
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Saburo Ito
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Robert I Seed
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew J Bondesson
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Jianlong Lou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - James D Marks
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Jody L Baron
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA.
| | - Stephen L Nishimura
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
68
|
Zheng Y, Leftheris K. Insights into Protein–Ligand Interactions in Integrin Complexes: Advances in Structure Determinations. J Med Chem 2020; 63:5675-5696. [DOI: 10.1021/acs.jmedchem.9b01869] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Yajun Zheng
- Pliant Therapeutics, South San Francisco, California 94080, United States
| | - Katerina Leftheris
- Pliant Therapeutics, South San Francisco, California 94080, United States
| |
Collapse
|
69
|
Nardelli F, Ghitti M, Quilici G, Gori A, Luo Q, Berardi A, Sacchi A, Monieri M, Bergamaschi G, Bermel W, Chen F, Corti A, Curnis F, Musco G. A stapled chromogranin A-derived peptide is a potent dual ligand for integrins αvβ6 and αvβ8. Chem Commun (Camb) 2020; 55:14777-14780. [PMID: 31755501 DOI: 10.1039/c9cc08518a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Combining 2D STD-NMR, computation, biochemical assays and click-chemistry, we have identified a chromogranin-A derived compound (5) that has high affinity and bi-selectivity for αvβ6 and αvβ8 integrins and is stable in microsomal preparations. 5 is suitable for nanoparticle functionalization and delivery to cancer cells, holding promise for diagnostic and/or therapeutic applications.
Collapse
Affiliation(s)
- Francesca Nardelli
- IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.
| | - Michela Ghitti
- IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.
| | - Giacomo Quilici
- IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.
| | - Alessandro Gori
- Istituto di Chimica del Riconoscimento Molecolare, C.N.R., Via Mario Bianco 9, 20131 Milan, Italy
| | - Qingqiong Luo
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Andrea Berardi
- IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.
| | - Angelina Sacchi
- IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.
| | - Matteo Monieri
- IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.
| | - Greta Bergamaschi
- Istituto di Chimica del Riconoscimento Molecolare, C.N.R., Via Mario Bianco 9, 20131 Milan, Italy
| | - Wolfgang Bermel
- Bruker BioSpin GmbH, Silberstreifen 4, Rheinstetten, 76287, Germany
| | - Fuxiang Chen
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Angelo Corti
- IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy. and Vita Salute San Raffaele University, Via Olgettina 58, 20132 Milan, Italy
| | - Flavio Curnis
- IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.
| | - Giovanna Musco
- IRCCS San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milan, Italy.
| |
Collapse
|
70
|
Ajmal M, Yunus U, Graham RM, Leblanc RM. Design, Synthesis, and Targeted Delivery of Fluorescent 1,2,4-Triazole-Peptide Conjugates to Pediatric Brain Tumor Cells. ACS OMEGA 2019; 4:22280-22291. [PMID: 31909311 PMCID: PMC6941177 DOI: 10.1021/acsomega.9b01903] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/18/2019] [Indexed: 06/10/2023]
Abstract
Most of the chemotherapeutics and drug-delivery models pose serious health problems and several undesirable side effects due to nonspecificity, lack of proper targeting system, and their large sizes. The rational design and synthesis of target-specific chemotherapeutics are highly important. This research work is focused on the rational design, synthesis, and anticancer studies of fluorescent 1,2,4-triazole-peptide conjugates for the development of target-specific anticancer drugs. Three novel 1,2,4-triazole derivatives: 4-(4-fluorobenzylidenamino)-3-hydrazino-5-mercapto-1,2,4-triazole (4FBAHMT, 2a), 4-(3,4,5-trimethoxybenzylidenamino)-3-hydrazino-5-mercapto-1,2,4-triazole (TMOBAHMT, 2b), and 4-(4-benzyloxy-2-methyloxbenzylidenamino)-3-hydrazino-5-mercapto-1,2,4-triazole (4BO2MOBAHMT, 2c) were synthesized after screening through molecular docking procedures. The docking studies were performed between ligand molecules and αvβ6 integrin protein. Fluorescent carbon nanoparticles (CNPs, 3) were conjugated with 1,2,4-triazole derivatives (2a-c) and l-carnosine (LC) dipeptide to get their corresponding conjugates (4a-c). The title double conjugates were characterized by spectroscopic (UV/vis spectroscopy, fluorescence spectroscopy, and FTIR spectroscopy) and microscopic (scanning electron microscopy, transmission electron microscopy, and atomic force microscopy) techniques. In vitro efficacy of fluorescent 1,2,4-triazole-peptide conjugates was investigated against two pediatric brain tumor cell lines (CHLA-200 & SJGBM2) and human embryonic kidney cell line (HEK293 as a control) by employing cell proliferation assay/MTS assay and fluorescence microscopy. 1,2,4-Triazole derivatives and their conjugates showed potent and selective anticancer activity against CHLA-200 and SJGBM2 cell lines. Cell proliferation assay and fluorescence microscopy results revealed that conjugates were more highly selective and cytotoxic than control drug temozolomide (TM) against both cell lines. CNPs are highly biocompatible and the quantum-sized conjugates were nontoxic for normal embryonic kidney cell line (HEK 293). The experimental results of MTS bioactivity assay and fluorescence microscopy were in close agreement with the theoretical results of molecular docking studies.
Collapse
Affiliation(s)
- Muhammad Ajmal
- Department
of Chemistry, Allama Iqbal Open University, Islamabad 44000, Pakistan
| | - Uzma Yunus
- Department
of Chemistry, Allama Iqbal Open University, Islamabad 44000, Pakistan
| | - Regina M. Graham
- Department
of Neurological Surgery, Miller School of
Medicine, University of Miami, Miami, Florida 33136, United States
| | - Roger M. Leblanc
- Department
of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida 33146, United States
| |
Collapse
|
71
|
Wang J, Su Y, Iacob RE, Engen JR, Springer TA. General structural features that regulate integrin affinity revealed by atypical αVβ8. Nat Commun 2019; 10:5481. [PMID: 31792290 PMCID: PMC6889490 DOI: 10.1038/s41467-019-13248-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/22/2019] [Indexed: 01/16/2023] Open
Abstract
Integrin αVβ8, which like αVβ6 functions to activate TGF-βs, is atypical. Its β8 subunit binds to a distinctive cytoskeleton adaptor and does not exhibit large changes in conformation upon binding to ligand. Here, crystal structures, hydrogen-deuterium exchange dynamics, and affinity measurements on mutants are used to compare αVβ8 and αVβ6. Lack of a binding site for one of three βI domain divalent cations and a unique β6-α7 loop conformation in β8 facilitate movements of the α1 and α1’ helices at the ligand binding pocket toward the high affinity state, without coupling to β6-α7 loop reshaping and α7-helix pistoning that drive large changes in βI domain-hybrid domain orientation seen in other integrins. Reciprocal swaps between β6 and β8 βI domains increase affinity of αVβ6 and decrease affinity of αVβ8 and define features that regulate affinity of the βI domain and its coupling to the hybrid domain. The activation mechanism of integrin αVβ8 differs from other integrins. Combining X-ray crystallography, hydrogen deuterium exchange mass spectrometry and mutation, the authors reveal structural features responsible for these differences and provide insights into how typical integrins are regulated.
Collapse
Affiliation(s)
- Jianchuan Wang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Yang Su
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA
| | - Roxana E Iacob
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - John R Engen
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA. .,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
72
|
Anderson NA, Campos S, Butler S, Copley RCB, Duncan I, Harrison S, Le J, Maghames R, Pastor-Garcia A, Pritchard JM, Rowedder JE, Smith CE, Thomas J, Vitulli G, Macdonald SJF. Discovery of an Orally Bioavailable Pan αv Integrin Inhibitor for Idiopathic Pulmonary Fibrosis. J Med Chem 2019; 62:8796-8808. [PMID: 31497959 DOI: 10.1021/acs.jmedchem.9b00962] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The heterodimeric transmembrane αv integrin receptors have recently emerged as potential targets for the treatment of idiopathic pulmonary fibrosis. Herein, we describe how subtle modifications of the central aromatic ring of a series of phenylbutyrate-based antagonists of the vitronectin receptors αvβ3 and αvβ5 significantly change the biological activities against αvβ6 and αvβ8. This resulted in the discovery of a pan αv antagonist (compound 39, 4-40 nM for the integrin receptors named above) possessing excellent oral pharmacokinetic properties in rats (with a clearance of 7.6 mL/(min kg) and a bioavailability of 97%).
Collapse
Affiliation(s)
- Niall A Anderson
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Sebastien Campos
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Sharon Butler
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Royston C B Copley
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Ian Duncan
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Stephen Harrison
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Joelle Le
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Rosemary Maghames
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Aleix Pastor-Garcia
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - John M Pritchard
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - James E Rowedder
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Claire E Smith
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Jack Thomas
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Giovanni Vitulli
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| | - Simon J F Macdonald
- GlaxoSmithKline Medicines Research Centre , Gunnels Wood Road , Stevenage SG1 2NY , U.K
| |
Collapse
|
73
|
Integrin-Mediated TGFβ Activation Modulates the Tumour Microenvironment. Cancers (Basel) 2019; 11:cancers11091221. [PMID: 31438626 PMCID: PMC6769837 DOI: 10.3390/cancers11091221] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/12/2019] [Accepted: 08/15/2019] [Indexed: 12/26/2022] Open
Abstract
TGFβ (transforming growth factor-beta) is a pleotropic cytokine with contrasting effects in cancer. In normal tissue and early tumours, TGFβ acts as a tumour suppressor, limiting proliferation and inducing apoptosis. However, these effects are eventually abrogated by the loss or inactivation of downstream signalling within the TGFβ pathway, and in established tumours, TGFβ then acts as a tumour promotor through multiple mechanisms including inducing epithelial-to-mesenchymal transition (EMT), promoting formation of cancer-associated fibroblasts (CAFs) and increasing angiogenesis. TGFβ is secrereted as a large latent complex and is embedded in the extracellular matrix or held on the surface of cells and must be activated before mediating its multiple functions. Thus, whilst TGFβ is abundant in the tumour microenvironment (TME), its functionality is regulated by local activation. The αv-integrins are major activators of latent-TGFβ. The potential benefits of manipulating the immune TME have been highlighted by the clinical success of immune-checkpoint inhibitors in a number of solid tumour types. TGFβ is a potent suppressor of T-cell-mediated immune surveillance and a key cause of resistance to checkpoint inhibitors. Therefore, as certain integrins locally activate TGFβ, they are likely to have a role in the immunosuppressive TME, although this remains to be confirmed. In this review, we discussed the role of TGFβ in cancer, the role of integrins in activating TGFβ in the TME, and the potential benefits of targeting integrins to augment immunotherapies.
Collapse
|
74
|
Barrett TN, Taylor JA, Barker D, Procopiou PA, Thompson JDF, Barrett J, Le J, Lynn SM, Pogany P, Pratley C, Pritchard JM, Roper JA, Rowedder JE, Slack RJ, Vitulli G, Macdonald SJF, Kerr WJ. Profile of a Highly Selective Quaternized Pyrrolidine Betaine αvβ6 Integrin Inhibitor—(3S)-3-(3-(3,5-Dimethyl-1H-pyrazol-1-yl)phenyl)-4-((1S and 1R,3R)-1-methyl-3-(2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl)pyrrolidin-1-ium-1-yl)butanoate Synthesized by Stereoselective Methylation. J Med Chem 2019; 62:7543-7556. [DOI: 10.1021/acs.jmedchem.9b00819] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Tim N. Barrett
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Jonathan A. Taylor
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Daniel Barker
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Panayiotis A. Procopiou
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - James D. F. Thompson
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
- Department of Pure & Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, U.K
| | - John Barrett
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Joelle Le
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Sean M. Lynn
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Peter Pogany
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Cassie Pratley
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - John M. Pritchard
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - James A. Roper
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - James E. Rowedder
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Robert J. Slack
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Giovanni Vitulli
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Simon J. F. Macdonald
- Medicinal Science & Technology, GlaxoSmithKline Medicines Research Centre, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - William J. Kerr
- Department of Pure & Applied Chemistry, University of Strathclyde, 295 Cathedral Street, Glasgow, G1 1XL, U.K
| |
Collapse
|
75
|
Buß M, Tegtmeyer N, Schnieder J, Dong X, Li J, Springer TA, Backert S, Niemann HH. Specific high affinity interaction of Helicobacter pylori CagL with integrin α V β 6 promotes type IV secretion of CagA into human cells. FEBS J 2019; 286:3980-3997. [PMID: 31197920 DOI: 10.1111/febs.14962] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 04/12/2019] [Accepted: 06/10/2019] [Indexed: 12/21/2022]
Abstract
CagL is an essential pilus surface component of the virulence-associated type IV secretion system (T4SS) employed by Helicobacter pylori to translocate the oncogenic effector protein CagA into human gastric epithelial cells. CagL contains an RGD motif and integrin α5 β1 is widely accepted as its host cell receptor. Here, we show that CagL binds integrin αV β6 with substantially higher affinity and that this interaction is functionally important. Cell surface expression of αV β6 on various cell lines correlated perfectly with cell adhesion to immobilized CagL and with binding of soluble CagL to cells. We found no such correlation for α5 β1 . The purified αV β6 ectodomain bound CagL with high affinity. This interaction was highly specific, as the affinity of CagL for other RGD-binding integrins was two to three orders of magnitude weaker. Mutation of either conserved leucine in the CagL RGDLXXL motif, a motif that generally confers specificity for integrin αV β6 and αV β8 , lowered the affinity of CagL for αV β6 . Stable expression of αV β6 in αV β6 -negative but α5 β1 -expressing human cells promoted two hallmarks of the functional H. pylori T4SS, namely translocation of CagA into host cells and induction of interleukin-8 secretion by host cells. These findings suggest that integrin αV β6 , although not essential for T4SS function, represents an important host cell receptor for CagL.
Collapse
Affiliation(s)
- Maren Buß
- Structural Biochemistry, Department of Chemistry, Bielefeld University, Germany
| | - Nicole Tegtmeyer
- Division of Microbiology, Department of Biology, Friedrich-Alexander-University, Erlangen-Nuremberg, Germany
| | - Jennifer Schnieder
- Structural Biochemistry, Department of Chemistry, Bielefeld University, Germany
| | - Xianchi Dong
- Children's Hospital Boston, MA, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Jing Li
- Children's Hospital Boston, MA, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Timothy A Springer
- Children's Hospital Boston, MA, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Steffen Backert
- Division of Microbiology, Department of Biology, Friedrich-Alexander-University, Erlangen-Nuremberg, Germany
| | - Hartmut H Niemann
- Structural Biochemistry, Department of Chemistry, Bielefeld University, Germany
| |
Collapse
|
76
|
Sorolla A, Wang E, Clemons TD, Evans CW, Plani-Lam JH, Golden E, Dessauvagie B, Redfern AD, Swaminathan-Iyer K, Blancafort P. Triple-hit therapeutic approach for triple negative breast cancers using docetaxel nanoparticles, EN1-iPeps and RGD peptides. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 20:102003. [PMID: 31055077 DOI: 10.1016/j.nano.2019.04.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
Triple negative breast cancers (TNBC) are aggressive malignancies for which chemotherapy is the only treatment option. Many TNBC acquire chemotherapy resistance, notably docetaxel, which has been associated with the overexpression of transcription factors (TFs), such as ENGRAILED1 (EN1). Here, we have developed a tumor delivery system for docetaxel-PGMA-PAA-nanoparticles and interference peptides designed to specifically inhibit EN1 (EN1-iPeps). To promote tumor specific targeting, we functionalized these nanoparticles with EN1-iPeps engineered with RGD sequences. We found that these peptides reduce cell viability and induce apoptosis in TNBC cells with negligible effects on normal cells (EN1-). Moreover, EN1-RGD-iPeps-mediated nanoparticle internalization into breast cancer cells was via integrins and intravenous injection of this nanoformulation increased tumor accumulation. Furthermore, docetaxel nanoparticles functionalized with EN1-RGD-iPeps significantly reduced TNBC growth both in vitro and in vivo without showing toxicity. Our results suggest that this targeted nanoformulation represents a new and safe therapeutic approach for chemoresistant TNBCs.
Collapse
Affiliation(s)
- Anabel Sorolla
- Cancer Epigenetics, Harry Perkins Institute of Medical Research Nedlands, WA 6009, Australia; School of Human Sciences, The University of Western Australia Crawley, WA 6009, Australia.
| | - Edina Wang
- Cancer Epigenetics, Harry Perkins Institute of Medical Research Nedlands, WA 6009, Australia; School of Human Sciences, The University of Western Australia Crawley, WA 6009, Australia
| | - Tristan D Clemons
- School of Molecular Science, The University of Western Australia, Crawley, WA 6009, Australia
| | - Cameron W Evans
- School of Molecular Science, The University of Western Australia, Crawley, WA 6009, Australia
| | - Janice Hc Plani-Lam
- Cancer Epigenetics, Harry Perkins Institute of Medical Research Nedlands, WA 6009, Australia; School of Human Sciences, The University of Western Australia Crawley, WA 6009, Australia
| | - Emily Golden
- Cancer Epigenetics, Harry Perkins Institute of Medical Research Nedlands, WA 6009, Australia; School of Human Sciences, The University of Western Australia Crawley, WA 6009, Australia
| | - Ben Dessauvagie
- Division of Pathology and Laboratory Medicine, Medical School, The University of Western Australia, Crawley, WA 6009, Australia
| | - Andrew D Redfern
- School of Medicine, The University of Western Australia, Crawley, WA 6009, Australia
| | - K Swaminathan-Iyer
- School of Molecular Science, The University of Western Australia, Crawley, WA 6009, Australia
| | - Pilar Blancafort
- Cancer Epigenetics, Harry Perkins Institute of Medical Research Nedlands, WA 6009, Australia; School of Human Sciences, The University of Western Australia Crawley, WA 6009, Australia.
| |
Collapse
|
77
|
Reichart F, Maltsev OV, Kapp TG, Räder AFB, Weinmüller M, Marelli UK, Notni J, Wurzer A, Beck R, Wester HJ, Steiger K, Di Maro S, Di Leva FS, Marinelli L, Nieberler M, Reuning U, Schwaiger M, Kessler H. Selective Targeting of Integrin αvβ8 by a Highly Active Cyclic Peptide. J Med Chem 2019; 62:2024-2037. [DOI: 10.1021/acs.jmedchem.8b01588] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Florian Reichart
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Oleg V. Maltsev
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Tobias G. Kapp
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Andreas F. B. Räder
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Michael Weinmüller
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Udaya Kiran Marelli
- Central NMR Facility and Division of Organic Chemistry, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India
| | - Johannes Notni
- Lehrstuhl für Pharmazeutische Radiochemie, Technische Universität München, Walther-Meißner Straße 3, 85748 Garching, Germany
| | - Alexander Wurzer
- Lehrstuhl für Pharmazeutische Radiochemie, Technische Universität München, Walther-Meißner Straße 3, 85748 Garching, Germany
| | - Roswitha Beck
- Lehrstuhl für Pharmazeutische Radiochemie, Technische Universität München, Walther-Meißner Straße 3, 85748 Garching, Germany
| | - Hans-Jürgen Wester
- Lehrstuhl für Pharmazeutische Radiochemie, Technische Universität München, Walther-Meißner Straße 3, 85748 Garching, Germany
| | - Katja Steiger
- Department of Pathology, Technische Universität München, Trogerstraße 18, 81675 München, Germany
| | - Salvatore Di Maro
- DiSTABiF, Università degli Studi della Campania “Luigi Vanvitelli”, Via Vivaldi 43, 81100 Caserta, Italy
| | - Francesco Saverio Di Leva
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Luciana Marinelli
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Markus Nieberler
- Department of Oral and Maxillofacial Surgery, University Hospital Rechts der Isar, Technische Universität München, Ismaninger Straße 22, 81679 München, Germany
| | | | | | - Horst Kessler
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| |
Collapse
|
78
|
Identification, Characterization, and Optimization of Integrin α vβ₆-Targeting Peptides from a One-Bead One-Compound (OBOC) Library: Towards the Development of Positron Emission Tomography (PET) Imaging Agents. Molecules 2019; 24:molecules24020309. [PMID: 30654483 PMCID: PMC6359284 DOI: 10.3390/molecules24020309] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/13/2022] Open
Abstract
The current translation of peptides identified through the one-bead one-compound (OBOC) technology into positron emission tomography (PET) imaging agents is a slow process, with a major delay between ligand identification and subsequent lead optimization. This work aims to streamline the development process of 18F-peptide based PET imaging agents to target the integrin αvβ6. By directly identify αvβ6–targeting peptides from a 9-mer 4-fluorobenzoyl peptide library using the on-bead two-color (OBTC) cell-screening assay, a total of 185 peptide beads were identified and 5 beads sequenced for further evaluation. The lead peptide 1 (VGDLTYLKK(FB), IC50 = 0.45 ± 0.06 μM, 25% stable in serum at 1 h) was further modified at the N-, C-, and bi-termini. C-terminal PEGylation increased the metabolic stability (>95% stable), but decreased binding affinity (IC50 = 3.7 ± 1 μM) was noted. C-terminal extension (1i, VGDLTYLKK(FB)KVART) significantly increased binding affinity for integrin αvβ6 (IC50 = 0.021 ± 0.002 μM), binding selectivity for αvβ6-expressing cells (3.1 ± 0.8:1), and the serum stability (>99% stable). Our results demonstrate the challenges in optimizing OBOC-derived peptides, indicate both termini of 1 are sensitive to modifications, and show that further modification of 1 is necessary to demonstrate utility as an 18F-peptide imaging agent.
Collapse
|
79
|
Procopiou PA, Anderson NA, Barrett J, Barrett TN, Crawford MHJ, Fallon BJ, Hancock AP, Le J, Lemma S, Marshall RP, Morrell J, Pritchard JM, Rowedder JE, Saklatvala P, Slack RJ, Sollis SL, Suckling CJ, Thorp LR, Vitulli G, Macdonald SJF. Discovery of ( S)-3-(3-(3,5-Dimethyl-1 H-pyrazol-1-yl)phenyl)-4-(( R)-3-(2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl)pyrrolidin-1-yl)butanoic Acid, a Nonpeptidic α vβ 6 Integrin Inhibitor for the Inhaled Treatment of Idiopathic Pulmonary Fibrosis. J Med Chem 2018; 61:8417-8443. [PMID: 30215258 DOI: 10.1021/acs.jmedchem.8b00959] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
A series of 3-aryl(pyrrolidin-1-yl)butanoic acids were synthesized using a diastereoselective route, via a rhodium catalyzed asymmetric 1,4-addition of arylboronic acids in the presence of ( R)-BINAP to a crotonate ester to provide the ( S) absolute configuration for the major product. A variety of aryl substituents including morpholine, pyrazole, triazole, imidazole, and cyclic ether were screened in cell adhesion assays for affinity against αvβ1, αvβ3, αvβ5, αvβ6, and αvβ8 integrins. Numerous analogs with high affinity and selectivity for the αvβ6 integrin were identified. The analog ( S)-3-(3-(3,5-dimethyl-1 H-pyrazol-1-yl)phenyl)-4-(( R)-3-(2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl)pyrrolidin-1-yl)butanoic acid hydrochloride salt was found to have very high affinity for αvβ6 integrin in a radioligand binding assay (p Ki = 11), a long dissociation half-life (7 h), very high solubility in saline at pH 7 (>71 mg/mL), and pharmacokinetic properties commensurate with inhaled dosing by nebulization. It was selected for further clinical investigation as a potential therapeutic agent for the treatment of idiopathic pulmonary fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Colin J Suckling
- Department of Pure & Applied Chemistry , University of Strathclyde , 295 Cathedral Street , Glasgow G1 1XL , Scotland, U.K
| | | | | | | |
Collapse
|
80
|
Autonomous conformational regulation of β 3 integrin and the conformation-dependent property of HPA-1a alloantibodies. Proc Natl Acad Sci U S A 2018; 115:E9105-E9114. [PMID: 30209215 DOI: 10.1073/pnas.1806205115] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Integrin α/β heterodimer adopts a compact bent conformation in the resting state, and upon activation undergoes a large-scale conformational rearrangement. During the inside-out activation, signals impinging on the cytoplasmic tail of β subunit induce the α/β separation at the transmembrane and cytoplasmic domains, leading to the extended conformation of the ectodomain with the separated leg and the opening headpiece that is required for the high-affinity ligand binding. It remains enigmatic which integrin subunit drives the bent-to-extended conformational rearrangement in the inside-out activation. The β3 integrins, including αIIbβ3 and αVβ3, are the prototypes for understanding integrin structural regulation. The Leu33Pro polymorphism located at the β3 PSI domain defines the human platelet-specific alloantigen (HPA) 1a/b, which provokes the alloimmune response leading to clinically important bleeding disorders. Some, but not all, anti-HPA-1a alloantibodies can distinguish the αIIbβ3 from αVβ3 and affect their functions with unknown mechanisms. Here we designed a single-chain β3 subunit that mimics a separation of α/β heterodimer on inside-out activation. Our crystallographic and functional studies show that the single-chain β3 integrin folds into a bent conformation in solution but spontaneously extends on the cell surface. This demonstrates that the β3 subunit autonomously drives the membrane-dependent conformational rearrangement during integrin activation. Using the single-chain β3 integrin, we identified the conformation-dependent property of anti-HPA-1a alloantibodies, which enables them to differently recognize the β3 in the bent state vs. the extended state and in the complex with αIIb vs. αV This study provides deeper understandings of integrin conformational activation on the cell surface.
Collapse
|
81
|
Di Leva FS, Tomassi S, Di Maro S, Reichart F, Notni J, Dangi A, Marelli UK, Brancaccio D, Merlino F, Wester HJ, Novellino E, Kessler H, Marinelli L. Von einer Helix zu einem kleinen Ring: Metadynamik-inspirierte, selektive Liganden für αvβ6-Integrin. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201803250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Francesco Saverio Di Leva
- Dipartimento di Farmacia; Università degli Studi di Napoli Federico II; Via D. Montesano 49 80131 Naples Italien
| | - Stefano Tomassi
- DiSTABiF; Università degli Studi della Campania Luigi Vanvitelli; Via Vivaldi 43 81100 Caserta Italien
| | - Salvatore Di Maro
- DiSTABiF; Università degli Studi della Campania Luigi Vanvitelli; Via Vivaldi 43 81100 Caserta Italien
| | - Florian Reichart
- Institute for Advanced Study and Center of Integrated Protein Science; Department Chemie; Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Johannes Notni
- Lehrstuhl für Pharmazeutische Radiochemie; Technische Universität München; Walther-Meißner Straße 3 85748 Garching Deutschland
| | - Abha Dangi
- Central NMR Facility and Division of Organic Chemistry; CSIR-National Chemical Laboratory; Dr. Homi Bhabha Road 411008 Pune Indien
| | - Udaya Kiran Marelli
- Institute for Advanced Study and Center of Integrated Protein Science; Department Chemie; Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
- Central NMR Facility and Division of Organic Chemistry; CSIR-National Chemical Laboratory; Dr. Homi Bhabha Road 411008 Pune Indien
| | - Diego Brancaccio
- Dipartimento di Farmacia; Università degli Studi di Napoli Federico II; Via D. Montesano 49 80131 Naples Italien
| | - Francesco Merlino
- Dipartimento di Farmacia; Università degli Studi di Napoli Federico II; Via D. Montesano 49 80131 Naples Italien
| | - Hans-Jürgen Wester
- Lehrstuhl für Pharmazeutische Radiochemie; Technische Universität München; Walther-Meißner Straße 3 85748 Garching Deutschland
| | - Ettore Novellino
- Dipartimento di Farmacia; Università degli Studi di Napoli Federico II; Via D. Montesano 49 80131 Naples Italien
| | - Horst Kessler
- Institute for Advanced Study and Center of Integrated Protein Science; Department Chemie; Technische Universität München; Lichtenbergstraße 4 85748 Garching Deutschland
| | - Luciana Marinelli
- Dipartimento di Farmacia; Università degli Studi di Napoli Federico II; Via D. Montesano 49 80131 Naples Italien
| |
Collapse
|
82
|
Di Leva FS, Tomassi S, Di Maro S, Reichart F, Notni J, Dangi A, Marelli UK, Brancaccio D, Merlino F, Wester HJ, Novellino E, Kessler H, Marinelli L. From a Helix to a Small Cycle: Metadynamics-Inspired αvβ6 Integrin Selective Ligands. Angew Chem Int Ed Engl 2018; 57:14645-14649. [DOI: 10.1002/anie.201803250] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Francesco Saverio Di Leva
- Dipartimento di Farmacia; Università degli Studi di Napoli Federico II; Via D. Montesano 49 80131 Naples Italy
| | - Stefano Tomassi
- DiSTABiF; Università degli Studi della Campania Luigi Vanvitelli; Via Vivaldi 43 81100 Caserta Italy
| | - Salvatore Di Maro
- DiSTABiF; Università degli Studi della Campania Luigi Vanvitelli; Via Vivaldi 43 81100 Caserta Italy
| | - Florian Reichart
- Institute for Advanced Study and Center of Integrated Protein Science; Department Chemie; Technische Universität München; Lichtenbergstraße 4 85748 Garching Germany
| | - Johannes Notni
- Lehrstuhl für Pharmazeutische Radiochemie; Technische Universität München; Walther-Meißner Straße 3 85748 Garching Germany
| | - Abha Dangi
- Central NMR Facility and Division of Organic Chemistry; CSIR-National Chemical Laboratory; Dr. Homi Bhabha Road 411008 Pune India
| | - Udaya Kiran Marelli
- Institute for Advanced Study and Center of Integrated Protein Science; Department Chemie; Technische Universität München; Lichtenbergstraße 4 85748 Garching Germany
- Central NMR Facility and Division of Organic Chemistry; CSIR-National Chemical Laboratory; Dr. Homi Bhabha Road 411008 Pune India
| | - Diego Brancaccio
- Dipartimento di Farmacia; Università degli Studi di Napoli Federico II; Via D. Montesano 49 80131 Naples Italy
| | - Francesco Merlino
- Dipartimento di Farmacia; Università degli Studi di Napoli Federico II; Via D. Montesano 49 80131 Naples Italy
| | - Hans-Jürgen Wester
- Lehrstuhl für Pharmazeutische Radiochemie; Technische Universität München; Walther-Meißner Straße 3 85748 Garching Germany
| | - Ettore Novellino
- Dipartimento di Farmacia; Università degli Studi di Napoli Federico II; Via D. Montesano 49 80131 Naples Italy
| | - Horst Kessler
- Institute for Advanced Study and Center of Integrated Protein Science; Department Chemie; Technische Universität München; Lichtenbergstraße 4 85748 Garching Germany
| | - Luciana Marinelli
- Dipartimento di Farmacia; Università degli Studi di Napoli Federico II; Via D. Montesano 49 80131 Naples Italy
| |
Collapse
|
83
|
Caja L, Dituri F, Mancarella S, Caballero-Diaz D, Moustakas A, Giannelli G, Fabregat I. TGF-β and the Tissue Microenvironment: Relevance in Fibrosis and Cancer. Int J Mol Sci 2018. [PMID: 29701666 DOI: 10.3390/ijms19051294.pmid:29701666;pmcid:pmc5983604] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Transforming growth factor-β (TGF-β) is a cytokine essential for the induction of the fibrotic response and for the activation of the cancer stroma. Strong evidence suggests that a strong cross-talk exists among TGF-β and the tissue extracellular matrix components. TGF-β is stored in the matrix as part of a large latent complex bound to the latent TGF-β binding protein (LTBP) and matrix binding of latent TGF-β complexes, which is required for an adequate TGF-β function. Once TGF-β is activated, it regulates extracellular matrix remodelling and promotes a fibroblast to myofibroblast transition, which is essential in fibrotic processes. This cytokine also acts on other cell types present in the fibrotic and tumour microenvironment, such as epithelial, endothelial cells or macrophages and it contributes to the cancer-associated fibroblast (CAF) phenotype. Furthermore, TGF-β exerts anti-tumour activity by inhibiting the host tumour immunosurveillance. Aim of this review is to update how TGF-β and the tissue microenvironment cooperate to promote the pleiotropic actions that regulate cell responses of different cell types, essential for the development of fibrosis and tumour progression. We discuss recent evidences suggesting the use of TGF-β chemical inhibitors as a new line of defence against fibrotic disorders or cancer.
Collapse
Affiliation(s)
- Laia Caja
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Francesco Dituri
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Serena Mancarella
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Daniel Caballero-Diaz
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Aristidis Moustakas
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, L'Hospitalet, 08907 Barcelona, Spain.
| |
Collapse
|
84
|
Caja L, Dituri F, Mancarella S, Caballero-Diaz D, Moustakas A, Giannelli G, Fabregat I. TGF-β and the Tissue Microenvironment: Relevance in Fibrosis and Cancer. Int J Mol Sci 2018; 19:ijms19051294. [PMID: 29701666 PMCID: PMC5983604 DOI: 10.3390/ijms19051294] [Citation(s) in RCA: 249] [Impact Index Per Article: 35.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/17/2018] [Accepted: 04/24/2018] [Indexed: 12/14/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a cytokine essential for the induction of the fibrotic response and for the activation of the cancer stroma. Strong evidence suggests that a strong cross-talk exists among TGF-β and the tissue extracellular matrix components. TGF-β is stored in the matrix as part of a large latent complex bound to the latent TGF-β binding protein (LTBP) and matrix binding of latent TGF-β complexes, which is required for an adequate TGF-β function. Once TGF-β is activated, it regulates extracellular matrix remodelling and promotes a fibroblast to myofibroblast transition, which is essential in fibrotic processes. This cytokine also acts on other cell types present in the fibrotic and tumour microenvironment, such as epithelial, endothelial cells or macrophages and it contributes to the cancer-associated fibroblast (CAF) phenotype. Furthermore, TGF-β exerts anti-tumour activity by inhibiting the host tumour immunosurveillance. Aim of this review is to update how TGF-β and the tissue microenvironment cooperate to promote the pleiotropic actions that regulate cell responses of different cell types, essential for the development of fibrosis and tumour progression. We discuss recent evidences suggesting the use of TGF-β chemical inhibitors as a new line of defence against fibrotic disorders or cancer.
Collapse
Affiliation(s)
- Laia Caja
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Francesco Dituri
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Serena Mancarella
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Daniel Caballero-Diaz
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
| | - Aristidis Moustakas
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Box 582, 75123 Uppsala, Sweden.
| | - Gianluigi Giannelli
- National Institute of Gastroenterology, "S. de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy.
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Gran Via de l'Hospitalet, 199, 08908 Barcelona, Spain.
- Oncology Program, CIBEREHD, National Biomedical Research Institute on Liver and Gastrointestinal Diseases, Instituto de Salud Carlos III, 28029 Madrid, Spain.
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona, L'Hospitalet, 08907 Barcelona, Spain.
| |
Collapse
|
85
|
Alpha-v-containing integrins are host receptors for the Plasmodium falciparum sporozoite surface protein, TRAP. Proc Natl Acad Sci U S A 2018; 115:4477-4482. [PMID: 29632205 PMCID: PMC5924908 DOI: 10.1073/pnas.1719660115] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Malaria is caused by a parasite that is deposited in the skin through the bite of an infected mosquito. From the skin, parasites navigate through host tissues where they must locate and invade liver cells. We know that a parasite surface protein called TRAP is important for this process, making it a leading vaccine candidate. TRAP is thought to work by specifically binding a defined host cell surface protein, but its identity has remained a long-standing mystery. Our research has identified an integrin—a class of host cell surface proteins—as a TRAP receptor. This finding provides an important piece of the puzzle relating to TRAP function and may help improve the development of an effective malaria vaccine. Malaria-causing Plasmodium sporozoites are deposited in the dermis by the bite of an infected mosquito and move by gliding motility to the liver where they invade and develop within host hepatocytes. Although extracellular interactions between Plasmodium sporozoite ligands and host receptors provide important guidance cues for productive infection and are good vaccine targets, these interactions remain largely uncharacterized. Thrombospondin-related anonymous protein (TRAP) is a parasite cell surface ligand that is essential for both gliding motility and invasion because it couples the extracellular binding of host receptors to the parasite cytoplasmic actinomyosin motor; however, the molecular nature of the host TRAP receptors is poorly defined. Here, we use a systematic extracellular protein interaction screening approach to identify the integrin αvβ3 as a directly interacting host receptor for Plasmodium falciparum TRAP. Biochemical characterization of the interaction suggests a two-site binding model, requiring contributions from both the von Willebrand factor A domain and the RGD motif of TRAP for integrin binding. We show that TRAP binding to cells is promoted in the presence of integrin-activating proadhesive Mn2+ ions, and that cells genetically targeted so that they lack cell surface expression of the integrin αv-subunit are no longer able to bind TRAP. P. falciparum sporozoites moved with greater speed in the dermis of Itgb3-deficient mice, suggesting that the interaction has a role in sporozoite migration. The identification of the integrin αvβ3 as the host receptor for TRAP provides an important demonstration of a sporozoite surface ligand that directly interacts with host receptors.
Collapse
|
86
|
Abstract
Activation of TGF-β1 initiates a program of temporary collagen accumulation important to wound repair in many organs. However, the outcome of temporary extracellular matrix strengthening all too frequently morphs into progressive fibrosis, contributing to morbidity and mortality worldwide. To avoid this maladaptive outcome, TGF-β1 signaling is regulated at numerous levels and intimately connected to feedback signals that limit accumulation. Here, we examine the current understanding of the core functions of TGF-β1 in promoting collagen accumulation, parallel pathways that promote physiological repair, and pathological triggers that tip the balance toward progressive fibrosis. Implicit in better understanding of these processes is the identification of therapeutic opportunities that will need to be further advanced to limit or reverse organ fibrosis.
Collapse
Affiliation(s)
- Kevin K Kim
- Department of Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan 48109
| | - Dean Sheppard
- Department of Medicine, Cardiovascular Research Institute, and Lung Biology Center, University of California, San Francisco, San Francisco, California 94143
| | - Harold A Chapman
- Department of Medicine, Cardiovascular Research Institute, and Lung Biology Center, University of California, San Francisco, San Francisco, California 94143
| |
Collapse
|
87
|
Kapp TG, Di Leva FS, Notni J, Räder AFB, Fottner M, Reichart F, Reich D, Wurzer A, Steiger K, Novellino E, Marelli UK, Wester HJ, Marinelli L, Kessler H. N-Methylation of isoDGR Peptides: Discovery of a Selective α5β1-Integrin Ligand as a Potent Tumor Imaging Agent. J Med Chem 2018; 61:2490-2499. [DOI: 10.1021/acs.jmedchem.7b01752] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Tobias G. Kapp
- Institute for Advanced Study and Center of Integrated Protein Science, Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Francesco Saverio Di Leva
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Johannes Notni
- Lehrstuhl für Pharmazeutische Radiochemie, Technische Universität München, Walther-Meißner Straße 3, 85748 Garching, Germany
| | - Andreas F. B. Räder
- Institute for Advanced Study and Center of Integrated Protein Science, Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Maximilian Fottner
- Institute for Advanced Study and Center of Integrated Protein Science, Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Florian Reichart
- Institute for Advanced Study and Center of Integrated Protein Science, Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Dominik Reich
- Lehrstuhl für Pharmazeutische Radiochemie, Technische Universität München, Walther-Meißner Straße 3, 85748 Garching, Germany
| | - Alexander Wurzer
- Lehrstuhl für Pharmazeutische Radiochemie, Technische Universität München, Walther-Meißner Straße 3, 85748 Garching, Germany
| | - Katja Steiger
- Department of Pathology, Technische Universität München, Trogerstraße 18, 81675 München, Germany
| | - Ettore Novellino
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Udaya Kiran Marelli
- Central NMR Facility and Division of Organic Chemistry, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, 411008 Pune, India
| | - Hans-Jürgen Wester
- Lehrstuhl für Pharmazeutische Radiochemie, Technische Universität München, Walther-Meißner Straße 3, 85748 Garching, Germany
| | - Luciana Marinelli
- Dipartimento di Farmacia, Università degli Studi di Napoli “Federico II”, Via D. Montesano 49, 80131 Naples, Italy
| | - Horst Kessler
- Institute for Advanced Study and Center of Integrated Protein Science, Department Chemie, Technische Universität München, Lichtenbergstraße 4, 85748 Garching, Germany
| |
Collapse
|
88
|
Hatley RJD, Macdonald SJF, Slack RJ, Le J, Ludbrook SB, Lukey PT. An αv-RGD Integrin Inhibitor Toolbox: Drug Discovery Insight, Challenges and Opportunities. Angew Chem Int Ed Engl 2018; 57:3298-3321. [DOI: 10.1002/anie.201707948] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Richard J. D. Hatley
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Simon J. F. Macdonald
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Robert J. Slack
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Joelle Le
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Steven B. Ludbrook
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| | - Pauline T. Lukey
- Fibrosis DPU; Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY UK
| |
Collapse
|
89
|
Hatley RJD, Macdonald SJF, Slack RJ, Le J, Ludbrook SB, Lukey PT. Ein Instrumentarium von αv-RGD-Integrin-Inhibitoren: Wirkstoffsuche, Herausforderungen und Möglichkeiten. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201707948] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Richard J. D. Hatley
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Simon J. F. Macdonald
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Robert J. Slack
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Joelle Le
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Steven B. Ludbrook
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| | - Pauline T. Lukey
- Fibrosis and Lung Injury DPU, Respiratory Therapeutic Area; GlaxoSmithKline Medicines Research Centre; Gunnels Wood Road Stevenage SG1 2NY Großbritannien
| |
Collapse
|
90
|
Vishwakarma P, Parmar N, Chandrakar P, Sharma T, Kathuria M, Agnihotri PK, Siddiqi MI, Mitra K, Kar S. Ammonium trichloro [1,2-ethanediolato-O,O']-tellurate cures experimental visceral leishmaniasis by redox modulation of Leishmania donovani trypanothione reductase and inhibiting host integrin linked PI3K/Akt pathway. Cell Mol Life Sci 2018; 75:563-588. [PMID: 28900667 PMCID: PMC11105478 DOI: 10.1007/s00018-017-2653-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/11/2017] [Accepted: 09/05/2017] [Indexed: 10/18/2022]
Abstract
In an endeavor to search for affordable and safer therapeutics against debilitating visceral leishmaniasis, we examined antileishmanial potential of ammonium trichloro [1,2-ethanediolato-O,O']-tellurate (AS101); a tellurium based non toxic immunomodulator. AS101 showed significant in vitro efficacy against both Leishmania donovani promastigotes and amastigotes at sub-micromolar concentrations. AS101 could also completely eliminate organ parasite load from L. donovani infected Balb/c mice along with significant efficacy against infected hamsters (˃93% inhibition). Analyzing mechanistic details revealed that the double edged AS101 could directly induce apoptosis in promastigotes along with indirectly activating host by reversing T-cell anergy to protective Th1 mode, increased ROS generation and anti-leishmanial IgG production. AS101 could inhibit IL-10/STAT3 pathway in L. donovani infected macrophages via blocking α4β7 integrin dependent PI3K/Akt signaling and activate host MAPKs and NF-κB for Th1 response. In silico docking and biochemical assays revealed AS101's affinity to form thiol bond with cysteine residues of trypanothione reductase in Leishmania promastigotes leading to its inactivation and inducing ROS-mediated apoptosis of the parasite via increased Ca2+ level, loss of ATP and mitochondrial membrane potential along with metacaspase activation. Our findings provide the first evidence for the mechanism of action of AS101 with excellent safety profile and suggest its promising therapeutic potential against experimental visceral leishmaniasis.
Collapse
Affiliation(s)
- Preeti Vishwakarma
- Division of Parasitology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India
| | - Naveen Parmar
- Division of Parasitology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India
| | - Pragya Chandrakar
- Division of Parasitology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India
| | - Tanuj Sharma
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Manoj Kathuria
- Electron Microscopy Unit, Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Pramod K Agnihotri
- Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mohammad Imran Siddiqi
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India
- Molecular and Structural Biology Division, CSIR-Central Drug Research Institute, Lucknow, India
| | - Kalyan Mitra
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India
- Electron Microscopy Unit, Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Susanta Kar
- Division of Parasitology, CSIR-Central Drug Research Institute, Jankipuram Extension, Sitapur Road, Lucknow, Uttar Pradesh, 226031, India.
- Academy of Scientific and Innovative Research, Anusandhan Bhawan, New Delhi, India.
| |
Collapse
|
91
|
Oglic D, Oatley SA, Macdonald SJF, Mcinally T, Garnett R, Hirst JD, Gärtner T. Active Search for Computer-aided Drug Design. Mol Inform 2018; 37. [PMID: 29388736 DOI: 10.1002/minf.201700130] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/03/2018] [Indexed: 01/08/2023]
Abstract
We consider lead discovery as active search in a space of labelled graphs. In particular, we extend our recent data-driven adaptive Markov chain approach, and evaluate it on a focused drug design problem, where we search for an antagonist of an αv integrin, the target protein that belongs to a group of Arg-Gly-Asp integrin receptors. This group of integrin receptors is thought to play a key role in idiopathic pulmonary fibrosis, a chronic lung disease of significant pharmaceutical interest. As an in silico proxy of the binding affinity, we use a molecular docking score to an experimentally determined αvβ6 protein structure. The search is driven by a probabilistic surrogate of the activity of all molecules from that space. As the process evolves and the algorithm observes the activity scores of the previously designed molecules, the hypothesis of the activity is refined. The algorithm is guaranteed to converge in probability to the best hypothesis from an a priori specified hypothesis space. In our empirical evaluations, the approach achieves a large structural variety of designed molecular structures for which the docking score is better than the desired threshold. Some novel molecules, suggested to be active by the surrogate model, provoke a significant interest from the perspective of medicinal chemistry and warrant prioritization for synthesis. Moreover, the approach discovered 19 out of the 24 active compounds which are known to be active from previous biological assays.
Collapse
Affiliation(s)
- Dino Oglic
- School of Computer Science, University of Nottingham, Jubilee Campus, Wollaton Road, Nottingham, NG8 1BB, United Kingdom.,Institut für Informatik III, Universität Bonn, Römerstraße 164, 53117, Bonn, Germany
| | - Steven A Oatley
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, United Kingdom
| | - Simon J F Macdonald
- GlaxoSmithKline, Medicines Research Centre, Gunnels Wood Road, Stevenage, Hertfordshire, SG1 2NY, United Kingdom
| | - Thomas Mcinally
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, United Kingdom
| | - Roman Garnett
- Department of Computer Science and Engineering Washington University in St. Louis, One Brookings Drive CB 1045, St. Louis, MO, 63130, USA
| | - Jonathan D Hirst
- School of Chemistry, University of Nottingham, University Park, Nottingham, NG7 2RD, United Kingdom
| | - Thomas Gärtner
- School of Computer Science, University of Nottingham, Jubilee Campus, Wollaton Road, Nottingham, NG8 1BB, United Kingdom
| |
Collapse
|
92
|
High integrin α Vβ 6 affinity reached by hybrid domain deletion slows ligand-binding on-rate. Proc Natl Acad Sci U S A 2018; 115:E1429-E1436. [PMID: 29378937 DOI: 10.1073/pnas.1718662115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The role of the hybrid domain in integrin affinity regulation is unknown, as is whether the kinetics of ligand binding is modulated by integrin affinity state. Here, we compare cell surface and soluble integrin αVβ6 truncation mutants for ligand-binding affinity, kinetics, and thermodynamics. Removal of the integrin transmembrane/cytoplasmic domains or lower legs has little effect on αVβ6 affinity, in contrast to β1 integrins. In integrin opening, rearrangement at the interface between the βI and hybrid domains is linked to remodeling at the ligand-binding site at the opposite end of the βI domain, which greatly increases in affinity in the open conformation. The larger size of the βI-hybrid interface in the closed state suggests that the hybrid domain stabilizes closing. In agreement, deletion of the hybrid domain raised affinity by 50-fold. Surface plasmon resonance and isothermal titration calorimetry gave similar results and the latter revealed tradeoffs between enthalpy and entropy not apparent from affinity. At extremely high affinity reached in Mn2+ with hybrid domain truncation, αVβ6 on-rate for both pro-TGF-β1 and fibronectin declined. The results suggest that the open conformation of αVβ6 has lower on-rate than the closed conformation, correlate with constriction of the ligand-binding pocket in open αVβ6 structures, and suggest that the extended-closed conformation is kinetically selected for ligand binding. Subsequent transition to the extended-open conformation is stabilized by its much higher affinity for ligand and would also be stabilized by force exerted across ligand-bound integrins by the actin cytoskeleton.
Collapse
|
93
|
Li J, Springer TA. Energy landscape differences among integrins establish the framework for understanding activation. J Cell Biol 2017; 217:397-412. [PMID: 29122968 PMCID: PMC5748972 DOI: 10.1083/jcb.201701169] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 09/15/2017] [Accepted: 10/04/2017] [Indexed: 11/22/2022] Open
Abstract
Li and Springer demonstrate differences between integrins α4β1 and α5β1 in intrinsic affinities and relative free energies of three conformational states. Integrin conformational equilibria are both subunit and cell type specific. The energy landscapes of intact receptors on the cell surface provide a framework for understanding regulation of integrin adhesiveness. Why do integrins differ in basal activity, and how does affinity for soluble ligand correlate with cellular adhesiveness? We show that basal conformational equilibrium set points for integrin α4β1 are cell type specific and differ from integrin α5β1 when the two integrins are coexpressed on the same cell. Although α4β1 is easier to activate, its high-affinity state binds vascular cell adhesion molecule and fibronectin 100- to 1,000-fold more weakly than α5β1 binds fibronectin. Furthermore, the difference in affinity between the high- and low-affinity states is more compressed in α4β1 (600- to 800-fold) than in α5β1 (4,000- to 6,000-fold). α4β1 basal conformational equilibria differ among three cell types, define affinity for soluble ligand and readiness for priming, and may reflect differences in interactions with intracellular adaptors but do not predict cellular adhesiveness for immobilized ligand. The measurements here provide a necessary framework for understanding integrin activation in intact cells, including activation of integrin adhesiveness by application of tensile force by the cytoskeleton, across ligand–integrin–adaptor complexes.
Collapse
Affiliation(s)
- Jing Li
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA .,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| |
Collapse
|
94
|
Blocking immunosuppression by human Tregs in vivo with antibodies targeting integrin αVβ8. Proc Natl Acad Sci U S A 2017; 114:E10161-E10168. [PMID: 29109269 DOI: 10.1073/pnas.1710680114] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Human regulatory T cells (Tregs) suppress other T cells by converting the latent, inactive form of TGF-β1 into active TGF-β1. In Tregs, TGF-β1 activation requires GARP, a transmembrane protein that binds and presents latent TGF-β1 on the surface of Tregs stimulated through their T cell receptor. However, GARP is not sufficient because transduction of GARP in non-Treg T cells does not induce active TGF-β1 production. RGD-binding integrins were shown to activate TGF-β1 in several non-T cell types. Here we show that αVβ8 dimers are present on stimulated human Tregs but not in other T cells, and that antibodies against αV or β8 subunits block TGF-β1 activation in vitro. We also show that αV and β8 interact with GARP/latent TGF-β1 complexes in human Tregs. Finally, a blocking antibody against β8 inhibited immunosuppression by human Tregs in a model of xenogeneic graft-vs.-host disease induced by the transfer of human T cells in immunodeficient mice. These results show that TGF-β1 activation on the surface of human Tregs implies an interaction between the integrin αVβ8 and GARP/latent TGF-β1 complexes. Immunosuppression by human Tregs can be inhibited by antibodies against GARP or against the integrin β8 subunit. Such antibodies may prove beneficial against cancer or chronic infections.
Collapse
|
95
|
Zhao B, Xu S, Dong X, Lu C, Springer TA. Prodomain-growth factor swapping in the structure of pro-TGF-β1. J Biol Chem 2017; 293:1579-1589. [PMID: 29109152 PMCID: PMC5798290 DOI: 10.1074/jbc.m117.809657] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/24/2017] [Indexed: 12/20/2022] Open
Abstract
TGF-β is synthesized as a proprotein that dimerizes in the endoplasmic reticulum. After processing in the Golgi to cleave the N-terminal prodomain from the C-terminal growth factor (GF) domain in each monomer, pro-TGF-β is secreted and stored in latent complexes. It is unclear which prodomain and GF monomer are linked before proprotein convertase cleavage and how much conformational change occurs following cleavage. We have determined a structure of pro-TGF-β1 with the proprotein convertase cleavage site mutated to mimic the structure of the TGF-β1 proprotein. Structure, mutation, and model building demonstrate that the prodomain arm domain in one monomer is linked to the GF that interacts with the arm domain in the other monomer in the dimeric structure (i.e. the prodomain arm domain and GF domain in each monomer are swapped). Swapping has important implications for the mechanism of biosynthesis in the TGF-β family and is relevant to the mechanism for preferential formation of heterodimers over homodimers for some members of the TGF-β family. Our structure, together with two previous ones, also provides insights into which regions of the prodomain–GF complex are highly structurally conserved and which are perturbed by crystal lattice contacts.
Collapse
Affiliation(s)
- Bo Zhao
- From the Children's Hospital Boston and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115 and
| | - Shutong Xu
- the Department of Medical Oncology and Department of Cancer Biology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts 02215
| | - Xianchi Dong
- From the Children's Hospital Boston and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115 and
| | - Chafen Lu
- From the Children's Hospital Boston and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115 and
| | - Timothy A Springer
- From the Children's Hospital Boston and Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02115 and
| |
Collapse
|
96
|
MacFarlane EG, Haupt J, Dietz HC, Shore EM. TGF-β Family Signaling in Connective Tissue and Skeletal Diseases. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022269. [PMID: 28246187 DOI: 10.1101/cshperspect.a022269] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The transforming growth factor β (TGF-β) family of signaling molecules, which includes TGF-βs, activins, inhibins, and numerous bone morphogenetic proteins (BMPs) and growth and differentiation factors (GDFs), has important functions in all cells and tissues, including soft connective tissues and the skeleton. Specific TGF-β family members play different roles in these tissues, and their activities are often balanced with those of other TGF-β family members and by interactions with other signaling pathways. Perturbations in TGF-β family pathways are associated with numerous human diseases with prominent involvement of the skeletal and cardiovascular systems. This review focuses on the role of this family of signaling molecules in the pathologies of connective tissues that manifest in rare genetic syndromes (e.g., syndromic presentations of thoracic aortic aneurysm), as well as in more common disorders (e.g., osteoarthritis and osteoporosis). Many of these diseases are caused by or result in pathological alterations of the complex relationship between the TGF-β family of signaling mediators and the extracellular matrix in connective tissues.
Collapse
Affiliation(s)
- Elena Gallo MacFarlane
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Julia Haupt
- Department of Orthopedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Harry C Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.,Howard Hughes Medical Institute, Bethesda, Maryland 21205
| | - Eileen M Shore
- Department of Orthopedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
97
|
Abstract
Transforming growth factor βs (TGF-βs) are closely related ligands that have pleiotropic activity on most cell types of the body. They act through common heterotetrameric TGF-β type II and type I transmembrane dual specificity kinase receptor complexes, and the outcome of signaling is context-dependent. In normal tissue, they serve a role in maintaining homeostasis. In many diseased states, particularly fibrosis and cancer, TGF-β ligands are overexpressed and the outcome of signaling is diverted toward disease progression. There has therefore been a concerted effort to develop drugs that block TGF-β signaling for therapeutic benefit. This review will cover the basics of TGF-β signaling and its biological activities relevant to oncology, present a summary of pharmacological TGF-β blockade strategies, and give an update on preclinical and clinical trials for TGF-β blockade in a variety of solid tumor types.
Collapse
Affiliation(s)
- Rosemary J Akhurst
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94158-9001
| |
Collapse
|
98
|
Abstract
Transforming growth factor βs (TGF-βs) are closely related ligands that have pleiotropic activity on most cell types of the body. They act through common heterotetrameric TGF-β type II and type I transmembrane dual specificity kinase receptor complexes, and the outcome of signaling is context-dependent. In normal tissue, they serve a role in maintaining homeostasis. In many diseased states, particularly fibrosis and cancer, TGF-β ligands are overexpressed and the outcome of signaling is diverted toward disease progression. There has therefore been a concerted effort to develop drugs that block TGF-β signaling for therapeutic benefit. This review will cover the basics of TGF-β signaling and its biological activities relevant to oncology, present a summary of pharmacological TGF-β blockade strategies, and give an update on preclinical and clinical trials for TGF-β blockade in a variety of solid tumor types.
Collapse
Affiliation(s)
- Rosemary J Akhurst
- Department of Anatomy and Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, California 94158-9001
| |
Collapse
|
99
|
Investigating the Interaction of Cyclic RGD Peptidomimetics with α Vβ₆ Integrin by Biochemical and Molecular Docking Studies. Cancers (Basel) 2017; 9:cancers9100128. [PMID: 28934103 PMCID: PMC5664067 DOI: 10.3390/cancers9100128] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 09/13/2017] [Accepted: 09/18/2017] [Indexed: 01/15/2023] Open
Abstract
The interaction of a small library of cyclic RGD (Arg-Gly-Asp) peptidomimetics with αVβ6 integrin has been investigated by means of competitive solid phase binding assays to the isolated receptor and docking calculations in the crystal structure of the αVβ6 binding site. To this aim, a rigid receptor-flexible ligand docking protocol has been set up and then applied to predict the binding mode of the cyclic RGD peptidomimetics to αVβ6 integrin. Although the RGD interaction with αVβ6 recapitulates the RGD binding mode observed in αVβ3, differences between the integrin binding pockets can strongly affect the ligand binding ability. In general, the peptidomimetics exhibited IC50 values for integrin αVβ6 (i.e., the concentration of compound required for 50% inhibition of biotinylated fibronectin binding to isolated αVβ6 integrin) in the nanomolar range (77–345 nM), about 10–100 times higher than those for the related αVβ3 receptor, with a single notable ligand displaying a low nanomolar IC50 value (2.3 nM). Insights from the properties of the binding pocket combined with the analysis of the docking poses provided a rationale for ligand recognition and selectivity.
Collapse
|
100
|
Arimori T, Kitago Y, Umitsu M, Fujii Y, Asaki R, Tamura-Kawakami K, Takagi J. Fv-clasp: An Artificially Designed Small Antibody Fragment with Improved Production Compatibility, Stability, and Crystallizability. Structure 2017; 25:1611-1622.e4. [PMID: 28919443 DOI: 10.1016/j.str.2017.08.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/22/2017] [Accepted: 08/15/2017] [Indexed: 12/13/2022]
Abstract
Antibody fragments are frequently used as a "crystallization chaperone" to aid structural analysis of complex macromolecules that are otherwise crystallization resistant, but conventional fragment formats have not been designed for this particular application. By fusing an anti-parallel coiled-coil structure derived from the SARAH domain of human Mst1 kinase to the variable region of an antibody, we succeeded in creating a novel chimeric antibody fragment of ∼37 kDa, termed "Fv-clasp," which exhibits excellent crystallization compatibility while maintaining the binding ability of the original IgG molecule. The "clasp" and the engineered disulfide bond at the bottom of the Fv suppressed the internal mobility of the fragment and shielded hydrophobic residues, likely contributing to the high heat stability and the crystallizability of the Fv-clasp. Finally, Fv-clasp antibodies showed superior "chaperoning" activity over conventional Fab fragments, and facilitated the structure determination of an ectodomain fragment of integrin α6β1.
Collapse
Affiliation(s)
- Takao Arimori
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yu Kitago
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Masataka Umitsu
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuki Fujii
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ryoko Asaki
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | | | - Junichi Takagi
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan.
| |
Collapse
|