1
|
Paulus J, Sewald N. Small molecule- and peptide-drug conjugates addressing integrins: A story of targeted cancer treatment. J Pept Sci 2024; 30:e3561. [PMID: 38382900 DOI: 10.1002/psc.3561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 02/23/2024]
Abstract
Targeted cancer treatment should avoid side effects and damage to healthy cells commonly encountered during traditional chemotherapy. By combining small molecule or peptidic ligands as homing devices with cytotoxic drugs connected by a cleavable or non-cleavable linker in peptide-drug conjugates (PDCs) or small molecule-drug conjugates (SMDCs), cancer cells and tumours can be selectively targeted. The development of highly affine, selective peptides and small molecules in recent years has allowed PDCs and SMDCs to increasingly compete with antibody-drug conjugates (ADCs). Integrins represent an excellent target for conjugates because they are overexpressed by most cancer cells and because of the broad knowledge about native binding partners as well as the multitude of small-molecule and peptidic ligands that have been developed over the last 30 years. In particular, integrin αVβ3 has been addressed using a variety of different PDCs and SMDCs over the last two decades, following various strategies. This review summarises and describes integrin-addressing PDCs and SMDCs while highlighting points of great interest.
Collapse
Affiliation(s)
- Jannik Paulus
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Bielefeld, Germany
| | - Norbert Sewald
- Organic and Bioorganic Chemistry, Faculty of Chemistry, Bielefeld University, Bielefeld, Germany
| |
Collapse
|
2
|
Sabat M, Carney DW, Hernandez-Torres G, Gibson TS, Balakrishna D, Zou H, Xu R, Chen CH, de Jong R, Dougan DR, Qin L, Bigi-Botterill SV, Chambers A, Miura J, Johnson LK, Ermolieff J, Johns D, Selimkhanov J, Kwok L, DeMent K, Proffitt C, Vu P, Lindsey EA, Ivetac T, Jennings A, Wang H, Manam P, Santos C, Fullenwider C, Manohar R, Flick AC. Design and Discovery of a Potent and Selective Inhibitor of Integrin αvβ1. J Med Chem 2024; 67:10306-10320. [PMID: 38872300 DOI: 10.1021/acs.jmedchem.4c00743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Selective inhibition of the RGD (Arg-Gly-Asp) integrin αvβ1 has been recently identified as an attractive therapeutic approach for the treatment of liver fibrosis given its function, target expression, and safety profile. Our identification of a non-RGD small molecule lead followed by focused, systematic changes to the core structure utilizing a crystal structure, in silico modeling, and a tractable synthetic approach resulted in the identification of a potent small molecule exhibiting a remarkable affinity for αvβ1 relative to several other integrin isoforms measured. Azabenzimidazolone 25 demonstrated antifibrotic efficacy in an in vivo rat liver fibrosis model and represents a tool compound capable of further exploring the biological consequences of selective αvβ1 inhibition.
Collapse
Affiliation(s)
- Mark Sabat
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Daniel W Carney
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Gloria Hernandez-Torres
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Tony S Gibson
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Deepika Balakrishna
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Hua Zou
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Rui Xu
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Chien-Hung Chen
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Ron de Jong
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Douglas R Dougan
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Ling Qin
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Simone V Bigi-Botterill
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Alison Chambers
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Joanne Miura
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Lucas K Johnson
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Jacques Ermolieff
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Deidre Johns
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Jangir Selimkhanov
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Lily Kwok
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Kevin DeMent
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Chris Proffitt
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Phong Vu
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Erick A Lindsey
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Tony Ivetac
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Andy Jennings
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Haixia Wang
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Padma Manam
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Cipriano Santos
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Cody Fullenwider
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Rohan Manohar
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| | - Andrew C Flick
- Gastroenterology Drug Discovery Unit, Takeda Development Center Americas, Inc., 9625 Towne Centre Dr., San Diego, California 92121 United States
| |
Collapse
|
3
|
Huycke TR, Häkkinen TJ, Miyazaki H, Srivastava V, Barruet E, McGinnis CS, Kalantari A, Cornwall-Scoones J, Vaka D, Zhu Q, Jo H, Oria R, Weaver VM, DeGrado WF, Thomson M, Garikipati K, Boffelli D, Klein OD, Gartner ZJ. Patterning and folding of intestinal villi by active mesenchymal dewetting. Cell 2024; 187:3072-3089.e20. [PMID: 38781967 PMCID: PMC11166531 DOI: 10.1016/j.cell.2024.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 12/30/2023] [Accepted: 04/26/2024] [Indexed: 05/25/2024]
Abstract
Tissue folds are structural motifs critical to organ function. In the intestine, bending of a flat epithelium into a periodic pattern of folds gives rise to villi, finger-like protrusions that enable nutrient absorption. However, the molecular and mechanical processes driving villus morphogenesis remain unclear. Here, we identify an active mechanical mechanism that simultaneously patterns and folds the intestinal epithelium to initiate villus formation. At the cellular level, we find that PDGFRA+ subepithelial mesenchymal cells generate myosin II-dependent forces sufficient to produce patterned curvature in neighboring tissue interfaces. This symmetry-breaking process requires altered cell and extracellular matrix interactions that are enabled by matrix metalloproteinase-mediated tissue fluidization. Computational models, together with in vitro and in vivo experiments, revealed that these cellular features manifest at the tissue level as differences in interfacial tensions that promote mesenchymal aggregation and interface bending through a process analogous to the active dewetting of a thin liquid film.
Collapse
Affiliation(s)
- Tyler R Huycke
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Teemu J Häkkinen
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Hikaru Miyazaki
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Emilie Barruet
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Christopher S McGinnis
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Ali Kalantari
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Jake Cornwall-Scoones
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Dedeepya Vaka
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Qin Zhu
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Roger Oria
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Valerie M Weaver
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Comprehensive Cancer Center, Helen Diller Family Cancer Research Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Bioengineering and Therapeutic Sciences, Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Krishna Garikipati
- Departments of Mechanical Engineering, and Mathematics, University of Michigan, Ann Arbor, MI, USA
| | - Dario Boffelli
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA
| | - Ophir D Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California, San Francisco, San Francisco, CA, USA; Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA, USA.
| | - Zev J Gartner
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
4
|
Huycke TR, Miyazaki H, Häkkinen TJ, Srivastava V, Barruet E, McGinnis CS, Kalantari A, Cornwall-Scoones J, Vaka D, Zhu Q, Jo H, DeGrado WF, Thomson M, Garikipati K, Boffelli D, Klein OD, Gartner ZJ. Patterning and folding of intestinal villi by active mesenchymal dewetting. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.25.546328. [PMID: 37425793 PMCID: PMC10326967 DOI: 10.1101/2023.06.25.546328] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Tissue folding generates structural motifs critical to organ function. In the intestine, bending of a flat epithelium into a periodic pattern of folds gives rise to villi, the numerous finger-like protrusions that are essential for nutrient absorption. However, the molecular and mechanical mechanisms driving the initiation and morphogenesis of villi remain a matter of debate. Here, we identify an active mechanical mechanism that simultaneously patterns and folds intestinal villi. We find that PDGFRA+ subepithelial mesenchymal cells generate myosin II-dependent forces sufficient to produce patterned curvature in neighboring tissue interfaces. At the cell-level, this occurs through a process dependent upon matrix metalloproteinase-mediated tissue fluidization and altered cell-ECM adhesion. By combining computational models with in vivo experiments, we reveal these cellular features manifest at the tissue-level as differences in interfacial tensions that promote mesenchymal aggregation and interface bending through a process analogous to the active de-wetting of a thin liquid film.
Collapse
Affiliation(s)
- Tyler R. Huycke
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Equal contribution
| | - Hikaru Miyazaki
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Equal contribution
| | - Teemu J. Häkkinen
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Equal contribution
| | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Emilie Barruet
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Christopher S. McGinnis
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Ali Kalantari
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Jake Cornwall-Scoones
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Dedeepya Vaka
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Qin Zhu
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - William F. DeGrado
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Krishna Garikipati
- Departments of Mechanical Engineering, and Mathematics, University of Michigan, Ann Arbor, USA
| | - Dario Boffelli
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Ophir D. Klein
- Program in Craniofacial Biology and Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
5
|
Borodina TN, Smirnov VI, Serykh VY, Rozentsveig IB. Structural and theoretical study of π-stacking interactions in new complexes based on CuCl 2 and 3-sulfonamide-substituted imidazo[2,1-b]thiazoles. J Mol Model 2023; 29:136. [PMID: 37045992 DOI: 10.1007/s00894-023-05549-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/03/2023] [Indexed: 04/14/2023]
Abstract
CONTEXT At present, sulfonamides and their metal complexes have received a new impetus for development. Of particular interest is the study of molecular and crystal structures, which takes into account weak non-valent interactions. Despite the low energy of such interactions, in many cases, they act collectively, and the sum of their actions can play a significant role. As a result, the spectrum of medical and biological activity of new metal complexes is expanded. In this regard, the synthesis and study of the molecular and crystal structure of sulfonamides and their metal complexes is of undoubted relevance. In this work, we studied non-valent intra- and intermolecular interactions in ligands of sulfonamide-substituted imidazo[2,1-b]thiazoles and their previously unknown complexes with CuCl2. The performed analysis of the data obtained by X-ray diffraction analysis made it possible to establish the intramolecular π-stacking interaction in imidazothiazole ligands, which is retained in their complexes with CuCl2. Within the framework of QTAIM topological analysis of electron density and DORI analysis, stereoelectronic and topological structures were studied. In the complexes, tetral, chalcogen, and pnycogen new interligand non-valent interactions were established. The energies of all established types of non-valent interactions have been calculated, and their comparative evaluation has been made. METHODS X-ray data of new arylsulfonylamino-substituted derivatives of imidazo[2,1-b]thiazoles and their metal complexes with CuCl2 have been studied. To determine the theoretical prerequisites for the occurrence of π-stacking in the molecules under study, the QTAIM method was used in the framework of the DFT/B3LYP/6-311 + G(d) calculation using the GAUSSIAN 09 program. In addition, the DORI electron density region overlap indicator and the Multiwfn program were used to analyze non-valent interactions.
Collapse
Affiliation(s)
- T N Borodina
- A.E. Favorsky Irkutsk Institute of Chemistry Siberian Branch of the Russian Academy of Sciences, 1 Favorsky Street, 664033, Irkutsk, Russian Federation.
| | - V I Smirnov
- A.E. Favorsky Irkutsk Institute of Chemistry Siberian Branch of the Russian Academy of Sciences, 1 Favorsky Street, 664033, Irkutsk, Russian Federation
| | - V Yu Serykh
- A.E. Favorsky Irkutsk Institute of Chemistry Siberian Branch of the Russian Academy of Sciences, 1 Favorsky Street, 664033, Irkutsk, Russian Federation
| | - I B Rozentsveig
- A.E. Favorsky Irkutsk Institute of Chemistry Siberian Branch of the Russian Academy of Sciences, 1 Favorsky Street, 664033, Irkutsk, Russian Federation
| |
Collapse
|
6
|
Hunter EJ, Hamaia SW, Kim PSK, Malcor JDM, Farndale RW. The effects of inhibition and siRNA knockdown of collagen-binding integrins on human umbilical vein endothelial cell migration and tube formation. Sci Rep 2022; 12:21601. [PMID: 36517525 PMCID: PMC9751114 DOI: 10.1038/s41598-022-25937-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Blood vessels in the body are lined with endothelial cells which have vital roles in numerous physiological and pathological processes. Collagens are major constituents of the extracellular matrix, and many adherent cells express several collagen-binding adhesion receptors. Here, we study the endothelium-collagen interactions mediated by the collagen-binding integrins, α1β1, α2β1, α10β1 and α11β1 expressed in human umbilical vein endothelial cells (HUVECs). Using qPCR, we found expression of the α10 transcript of the chondrocyte integrin, α10β1, along with the more abundant α2, and low-level expression of α1. The α11 transcript was not detected. Inhibition or siRNA knockdown of the α2-subunit resulted in impaired HUVEC adhesion, spreading and migration on collagen-coated surfaces, whereas inhibition or siRNA knockdown of α1 had no effect on these processes. In tube formation assays, inhibition of either α1 or α2 subunits impaired the network complexity, whereas siRNA knockdown of these integrins had no such effect. Knockdown of α10 had no effect on cell spreading, migration or tube formation in these conditions. Overall, our results indicate that the collagen-binding integrins, α1β1 and α2β1 play a central role in endothelial cell motility and self-organisation.
Collapse
Affiliation(s)
- Emma J Hunter
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, UK
- Stem Cell and Brain Research Institute, Université Lyon 1, INSERM U1208, 18 Avenue Doyen Lépine, 69500, Bron, France
| | - Samir W Hamaia
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, UK
| | - Peter S-K Kim
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, UK
| | - Jean-Daniel M Malcor
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, UK
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, UMS3444 BioSciences Gerland-Lyon Sud, UMR5305, CNRS/Université Lyon 1, Lyon, France
| | - Richard W Farndale
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge, CB2 1QW, UK.
- CambCol Laboratories Ltd, 18 Oak Lane, Littleport, Ely, CB6 1QZ, UK.
| |
Collapse
|
7
|
Clift CL, Saunders J, Drake RR, Angel PM. Perspectives on pediatric congenital aortic valve stenosis: Extracellular matrix proteins, post translational modifications, and proteomic strategies. Front Cardiovasc Med 2022; 9:1024049. [PMID: 36439995 PMCID: PMC9685993 DOI: 10.3389/fcvm.2022.1024049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022] Open
Abstract
In heart valve biology, organization of the extracellular matrix structure is directly correlated to valve function. This is especially true in cases of pediatric congenital aortic valve stenosis (pCAVS), in which extracellular matrix (ECM) dysregulation is a hallmark of the disease, eventually leading to left ventricular hypertrophy and heart failure. Therapeutic strategies are limited, especially in pediatric cases in which mechanical and tissue engineered valve replacements may not be a suitable option. By identifying mechanisms of translational and post-translational dysregulation of ECM in CAVS, potential drug targets can be identified, and better bioengineered solutions can be developed. In this review, we summarize current knowledge regarding ECM proteins and their post translational modifications (PTMs) during aortic valve development and disease and contributing factors to ECM dysregulation in CAVS. Additionally, we aim to draw parallels between other fibrotic disease and contributions to ECM post-translational modifications. Finally, we explore the current treatment options in pediatrics and identify how the field of proteomics has advanced in recent years, highlighting novel characterization methods of ECM and PTMs that may be used to identify potential therapeutic strategies relevant to pCAVS.
Collapse
Affiliation(s)
- Cassandra L. Clift
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
- Division of Cardiovascular Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
| | - Janet Saunders
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Richard R. Drake
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
| | - Peggi M. Angel
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, United States
- *Correspondence: Peggi M. Angel,
| |
Collapse
|
8
|
Kashif Khan R, Meanwell NA, Hager HH. Pseudoprolines as stereoelectronically tunable proline isosteres. Bioorg Med Chem Lett 2022; 75:128983. [PMID: 36096342 DOI: 10.1016/j.bmcl.2022.128983] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 11/18/2022]
Abstract
The cyclic structure of proline (Pro) confers unique conformational properties on this natural amino acid that influences polypeptide structure and function. Pseudoprolines are a family of Pro isosteres that incorporate a heteroatom, most prominently oxygen or sulfur but also silicon and selenium, to replace the Cβ or Cγ carbon atom of the pyrrolidine ring. These readily synthetically accessible structural motifs can facilitate facile molecular editing in a fashion that allows modulation of the amide bond topology of dipeptide elements and influence over ring pucker. While the properties of pseudoprolines have been exploited most prominently in the design of oligopeptide analogues, they have potential application in the design and optimization of small molecules. In this Digest, we summarize the physicochemical properties of pseudoprolines and illustrate their potential in drug discovery by surveying examples of applications in the design of bioactive molecules.
Collapse
Affiliation(s)
- R Kashif Khan
- Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, 100 Binney Street, Cambridge, MA 02142, USA.
| | - Nicholas A Meanwell
- Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, NJ 08543-4000, USA.
| | - Harry H Hager
- Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, 200 Cambridgepark Drive, Cambridge, MA 02140, USA.
| |
Collapse
|
9
|
Zheng S, Ye Z, Ji Y, Li N, Zhang L. Development of the Antithrombotic Peptide LEKNSTY Targeting the Collagen Surface: II. Improvement of Plasma Stability. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2022; 38:7114-7120. [PMID: 35623058 DOI: 10.1021/acs.langmuir.2c00587] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The development of antithrombotic peptides targeting collagen was proven effective, and an effective antithrombotic peptide LEKNSTY was obtained in part I. However, the plasma stability of LEKNSTY was found to be not good enough. In this part, the LEKNSTY was further optimized for improvement in plasma stability by substitution using d-amino acid residues. Two novel antithrombotic peptides LekNStY and lEKnsTy were designed, where lowercase letters represent d-amino acid residues. Improvements in plasma stability of both LekNStY and lEKnsTy were experimentally confirmed. Moreover, good binding of these antithrombotic peptides on the collagen surface was confirmed by molecular dynamics simulation and experimental validation. For example, a Kd of only 0.75 ± 0.10 μM was observed for lEKnsTy. Moreover, LekNStY and lEKnsTy were found to inhibit platelet adhesion on the collagen surface more effectively than LEKNSTY, and the IC50 of lEKnsTy was only 2/5 of that of LEKNSTY. These results confirmed the successful design of LekNStY and lEKnsTy that had good plasma stability and could effectively inhibit arterial thrombosis, which would be helpful for the research into interfaces involved in thrombus formation and the development of antithrombotic nanomedicine.
Collapse
Affiliation(s)
- Si Zheng
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Zhao Ye
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Yufan Ji
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Nanxing Li
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Lin Zhang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| |
Collapse
|
10
|
Singh S, Winkelstein BA. Inhibiting the β1integrin subunit increases the strain threshold for neuronal dysfunction under tensile loading in collagen gels mimicking innervated ligaments. Biomech Model Mechanobiol 2022; 21:885-898. [DOI: 10.1007/s10237-022-01565-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 02/13/2022] [Indexed: 11/28/2022]
|
11
|
Hunter EJ, Hamaia SW, Gullberg D, Malcor JD, Farndale RW. Selectivity of the collagen-binding integrin inhibitors, TC-I-15 and obtustatin. Toxicol Appl Pharmacol 2021; 428:115669. [PMID: 34363821 PMCID: PMC8444087 DOI: 10.1016/j.taap.2021.115669] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/29/2021] [Accepted: 08/02/2021] [Indexed: 11/22/2022]
Abstract
Integrins are a family of 24 adhesion receptors which are both widely-expressed and important in many pathophysiological cellular processes, from embryonic development to cancer metastasis. Hence, integrin inhibitors are valuable research tools which may have promising therapeutic uses. Here, we focus on the four collagen-binding integrins α1β1, α2β1, α10β1 and α11β1. TC-I-15 is a small molecule inhibitor of α2β1 that inhibits platelet adhesion to collagen and thrombus deposition, and obtustatin is an α1β1-specific disintegrin that inhibits angiogenesis. Both inhibitors were applied in cellular adhesion studies, using synthetic collagen peptide coatings with selective affinity for the different collagen-binding integrins and testing the adhesion of C2C12 cells transfected with each. Obtustatin was found to be specific for α1β1, as described, whereas TC-I-15 is shown to be non-specific, since it inhibits both α1β1 and α11β1 as well as α2β1. TC-I-15 was 100-fold more potent against α2β1 binding to a lower-affinity collagen peptide, suggestive of a competitive mechanism. These results caution against the use of integrin inhibitors in a therapeutic or research setting without testing for cross-reactivity.
Collapse
Affiliation(s)
- Emma J Hunter
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge CB2 1QW, UK
| | - Samir W Hamaia
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge CB2 1QW, UK
| | - Donald Gullberg
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, N-5009 Bergen, Norway
| | - Jean-Daniel Malcor
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge CB2 1QW, UK
| | - Richard W Farndale
- Department of Biochemistry, University of Cambridge, Downing Site, Cambridge CB2 1QW, UK.
| |
Collapse
|
12
|
Liu S, Ngo U, Tang XZ, Ren X, Qiu W, Huang X, DeGrado W, Allen CD, Jo H, Sheppard D, Sundaram AB. Integrin α2β1 regulates collagen I tethering to modulate hyperresponsiveness in reactive airway disease models. J Clin Invest 2021; 131:138140. [PMID: 33956668 DOI: 10.1172/jci138140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/05/2021] [Indexed: 12/26/2022] Open
Abstract
Severe asthma remains challenging to manage and has limited treatment options. We have previously shown that targeting smooth muscle integrin α5β1 interaction with fibronectin can mitigate the effects of airway hyperresponsiveness by impairing force transmission. In this study, we show that another member of the integrin superfamily, integrin α2β1, is present in airway smooth muscle and capable of regulating force transmission via cellular tethering to the matrix protein collagen I and, to a lesser degree, laminin-111. The addition of an inhibitor of integrin α2β1 impaired IL-13-enhanced contraction in mouse tracheal rings and human bronchial rings and abrogated the exaggerated bronchoconstriction induced by allergen sensitization and challenge. We confirmed that this effect was not due to alterations in classic intracellular myosin light chain phosphorylation regulating muscle shortening. Although IL-13 did not affect surface expression of α2β1, it did increase α2β1-mediated adhesion and the level of expression of an activation-specific epitope on the β1 subunit. We developed a method to simultaneously quantify airway narrowing and muscle shortening using 2-photon microscopy and demonstrated that inhibition of α2β1 mitigated IL-13-enhanced airway narrowing without altering muscle shortening by impairing the tethering of muscle to the surrounding matrix. Our data identified cell matrix tethering as an attractive therapeutic target to mitigate the severity of airway contraction in asthma.
Collapse
Affiliation(s)
- Sean Liu
- Lung Biology Center, Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine
| | - Uyen Ngo
- Lung Biology Center, Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine
| | - Xin-Zi Tang
- Cardiovascular Research Institute.,Sandler Asthma Basic Research Center.,Biomedical Sciences Graduate Program
| | - Xin Ren
- Lung Biology Center, Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine
| | - Wenli Qiu
- Lung Biology Center, Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine
| | - Xiaozhu Huang
- Lung Biology Center, Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine
| | - William DeGrado
- Cardiovascular Research Institute.,Department of Pharmaceutical Chemistry, and
| | - Christopher Dc Allen
- Cardiovascular Research Institute.,Sandler Asthma Basic Research Center.,Department of Anatomy, UCSF, San Francisco, California, USA
| | - Hyunil Jo
- Cardiovascular Research Institute.,Department of Pharmaceutical Chemistry, and
| | - Dean Sheppard
- Lung Biology Center, Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine.,Cardiovascular Research Institute
| | - Aparna B Sundaram
- Lung Biology Center, Division of Pulmonary, Critical Care, Allergy and Sleep, Department of Medicine
| |
Collapse
|
13
|
Sun N, Ye Z, Hao T, Zheng S, Sun Y, Zhang Y, Zhang L. Inhibition of Arterial Thrombus Formation by Blocking Exposed Collagen Surface Using LWWNSYY-Poly(l-Glutamic Acid) Nanoconjugate. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:6792-6799. [PMID: 34047558 DOI: 10.1021/acs.langmuir.1c00894] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Exposed collagen surface on diseased blood vessel wall is a trigger of platelet adhesion and subsequent thrombus formation, which is associated with many serious diseases such as myocardial infarction and stroke. Various antithrombotic agents have been developed, but are usually targeted on blood components such as platelet, which suffered from the risk of bleeding due to interference with hemostasis. In contrast, blocking the exposed collagen surface would prevent thrombus formation without the risk of bleeding. In the present study, an antithrombotic nanoconjugate (LWWNSYY-poly glutamic acid, L7-PGA) targeting collagen surface was designed by immobilizing heptapeptide LWWNSYY, a biomimetic inhibitor designed in our previous work, on poly(l-glutamic acid). Successful binding of L7-PGA on the collagen surface was confirmed by a negative ΔG of -5.99 ± 0.26 kcal/mol. L7-PGA was found to effectively inhibit platelet adhesion on the collagen surface, with a reduced IC50 of only 1/5 of that of free LWWNSYY. The inhibition of thrombus formation by L7-PGA was also validated in vivo by a reduction of 31.2% in the weight of thrombus. These results highlight L7-PGA as an effective inhibitor of arterial thrombus formation via blocking exposed collagen surface, which would be helpful for the development of novel antithrombotic nanomedicine.
Collapse
Affiliation(s)
- Na Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Zhao Ye
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Tanyi Hao
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Si Zheng
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Yan Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| | - Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, People's Republic of China
| | - Lin Zhang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, People's Republic of China
| |
Collapse
|
14
|
Gallorini M, Carradori S. Understanding collagen interactions and their targeted regulation by novel drugs. Expert Opin Drug Discov 2021; 16:1239-1260. [PMID: 34034595 DOI: 10.1080/17460441.2021.1933426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Introduction: Among protein and fibers in the extracellular matrix (ECM), collagen is the most copious and widely employed in cosmetic, food, pharmaceutical, and biomedical industries due to its extensive biocompatible and versatile properties. In the last years, the knowledge about functions of collagens increased and expanded dramatically. Once considered only crucial for the ECM scaffolding and mechanotransduction, additional functional roles have now been ascribed to the collagen superfamily which are defined by other recently discovered domains, supramolecular assembly and receptors.Areas covered: Given the importance of each step in the collagen biosynthesis, folding and signaling, medicinal chemists have explored small molecules, peptides, and monoclonal antibodies to modulate enzymes, receptors and interactions with the physiological ligands of collagen. These compounds were also explored toward diseases and pathological conditions. The authors discuss this providing their expert perspectives on the subject area.Expert opinion: Understanding collagen protein properties and its interactome is beneficial for therapeutic drug design. Nevertheless, compounds targeting collagen-based interactome suffered from the presence of different isoforms for each target and the lack of specific 3D crystal structures able to guide properly drug design.
Collapse
Affiliation(s)
- Marialucia Gallorini
- Department of Pharmacy, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Simone Carradori
- Department of Pharmacy, "G. D'Annunzio" University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
15
|
Okada T, Suzuki H. The Role of Tenascin-C in Tissue Injury and Repair After Stroke. Front Immunol 2021; 11:607587. [PMID: 33552066 PMCID: PMC7859104 DOI: 10.3389/fimmu.2020.607587] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/04/2020] [Indexed: 12/16/2022] Open
Abstract
Stroke is still one of the most common causes for mortality and morbidity worldwide. Following acute stroke onset, biochemical and cellular changes induce further brain injury such as neuroinflammation, cell death, and blood-brain barrier disruption. Matricellular proteins are non-structural proteins induced by many stimuli and tissue damage including stroke induction, while its levels are generally low in a normal physiological condition in adult tissues. Currently, a matricellular protein tenascin-C (TNC) is considered to be an important inducer to promote neuroinflammatory cascades and the resultant pathology in stroke. TNC is upregulated in cerebral arteries and brain tissues including astrocytes, neurons, and brain capillary endothelial cells following subarachnoid hemorrhage (SAH). TNC may be involved in blood-brain barrier disruption, neuronal apoptosis, and cerebral vasospasm via the activation of mitogen-activated protein kinases and nuclear factor-kappa B following SAH. In addition, post-SAH TNC levels in cerebrospinal fluid predicted the development of delayed cerebral ischemia and angiographic vasospasm in clinical settings. On the other hand, TNC is reported to promote fibrosis and exert repair effects for an experimental aneurysm via macrophages-induced migration and proliferation of smooth muscle cells. The authors review TNC-induced inflammatory signal cascades and the relationships with other matricellular proteins in stroke-related pathology.
Collapse
Affiliation(s)
- Takeshi Okada
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan.,Department of Neurosurgery, Kuwana City Medical Center, Kuwana, Japan
| | - Hidenori Suzuki
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
16
|
Fluksman A, Steinberg E, Orehov N, Shai E, Lahiani A, Katzhendler J, Marcinkiewicz C, Lazarovici P, Benny O. Integrin α 2β 1-Targeted Self-Assembled Nanocarriers for Tumor Bioimaging. ACS APPLIED BIO MATERIALS 2020; 3:6059-6070. [DOI: 10.1021/acsabm.0c00662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Arnon Fluksman
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91121, Israel
| | - Eliana Steinberg
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91121, Israel
| | - Natalie Orehov
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91121, Israel
| | - Ela Shai
- Department of Hematology, Coagulation Unit, Hadassah−Hebrew University Medical Center, Jerusalem 91121, Israel
| | - Adi Lahiani
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91121, Israel
| | - Jehoshua Katzhendler
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91121, Israel
| | - Cezary Marcinkiewicz
- Department of Biology, Temple University College of Science and Technology, Philadelphia, Pennsylvania 19122, United States
| | - Philip Lazarovici
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91121, Israel
| | - Ofra Benny
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91121, Israel
| |
Collapse
|
17
|
Li B, Wang X, Wang R, Rutz B, Ciotkowska A, Gratzke C, Herlemann A, Spek A, Tamalunas A, Waidelich R, Stief CG, Hennenberg M. Inhibition of neurogenic and thromboxane A 2 -induced human prostate smooth muscle contraction by the integrin α2β1 inhibitor BTT-3033 and the integrin-linked kinase inhibitor Cpd22. Prostate 2020; 80:831-849. [PMID: 32449814 DOI: 10.1002/pros.23998] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 01/23/2023]
Abstract
INTRODUCTION Prostate smooth muscle contraction is critical for etiology and treatment of lower urinary tract symptoms in benign prostatic hyperplasia (BPH). Integrins connect the cytoskeleton to membranes and cells to extracellular matrix, what is essential for force generation in smooth muscle contraction. Integrins are composed of different subunits and may cooperate with integrin-linked kinase (ILK). Here, we examined effects of inhibitors for different integrin heterodimers and ILK on contraction of human prostate tissues. METHODS Prostate tissues were obtained from radical prostatectomy. Integrins and ILK were detected by Western blot, real-time polymerase chain reaction (RT-PCR), and double fluorescence staining. Smooth muscle contractions of prostate strips were studied in an organ bath. Contractions were compared after application of solvent (controls), the ILK inhibitor Cpd22 (N-methyl-3-(1-(4-(piperazin-1-yl)phenyl)-5-(4'-(trifluoromethyl)-[1,1'-biphenyl]-4-yl)-1H-pyrazol-3-yl)propanamide), the integrin α2β1 inhibitor BTT-3033 (1-(4-fluorophenyl)-N-methyl-N-[4[[(phenylamino)carbonyl]amino]phenyl]-1H-pyrazole-4-sulfonamide), or the integrin α4β1/α9β1 inhibitor BOP (N-(benzenesulfonyl)- l-prolyl- l-O-(1-pyrrolidinylcarbonyl)tyrosine sodium salt). RESULTS Western blot analyses of prostate tissues using antibodies raised against integrins α2b, α4, α9, β1, and ILK revealed bands matching the expected sizes of corresponding antigens. Expression of integrins and ILK was confirmed by RT-PCR. Individual variations of expression levels occurred independently from divergent degree of BPH, reflected by different contents of prostate-specific antigen. Double fluorescence staining of prostate sections using antibodies raised against integrins α2 and β1, or against ILK resulted in immunoreactivity colocalizing with calponin, suggesting localization in prostate smooth muscle cells. Electric field stimulation (EFS) induced frequency-dependent contractions, which were inhibited by Cpd22 (3 µM) and BTT-3033 (1 µM) (inhibition around 37% by Cpd22 and 46% by BTT-3033 at 32 Hz). The thromboxane A2 analog U46619-induced concentration-dependent contractions, which were inhibited by Cpd22 and BTT-3033 (around 67% by Cpd22 and 39% by BTT-3033 at 30 µM U46619). Endothelin-1 induced concentration-dependent contractions, which were not affected by Cpd22 or BTT-3033. Noradrenaline and the α1 -adrenergic agonists methoxamine and phenylephrine-induced concentration-dependent contractions, which were not or very slightly inhibited by Cpd22 and BTT-3033. BOP did not change EFS- or agonist-induced contraction. CONCLUSIONS Integrin α2β1 and ILK inhibitors inhibit neurogenic and thromboxane A2 -induced prostate smooth muscle contraction in human BPH. A role for these targets for prostate smooth muscle contraction may appear possible.
Collapse
Affiliation(s)
- Bingsheng Li
- Department of Urology, LMU Munich, University Hospital, Munich, Germany
| | - Xiaolong Wang
- Department of Urology, LMU Munich, University Hospital, Munich, Germany
| | - Ruixiao Wang
- Department of Urology, LMU Munich, University Hospital, Munich, Germany
| | - Beata Rutz
- Department of Urology, LMU Munich, University Hospital, Munich, Germany
| | - Anna Ciotkowska
- Department of Urology, LMU Munich, University Hospital, Munich, Germany
| | | | - Annika Herlemann
- Department of Urology, LMU Munich, University Hospital, Munich, Germany
| | - Annabel Spek
- Department of Urology, LMU Munich, University Hospital, Munich, Germany
| | | | | | - Christian G Stief
- Department of Urology, LMU Munich, University Hospital, Munich, Germany
| | - Martin Hennenberg
- Department of Urology, LMU Munich, University Hospital, Munich, Germany
| |
Collapse
|
18
|
Xiong G, Chen J, Zhang G, Wang S, Kawasaki K, Zhu J, Zhang Y, Nagata K, Li Z, Zhou BP, Xu R. Hsp47 promotes cancer metastasis by enhancing collagen-dependent cancer cell-platelet interaction. Proc Natl Acad Sci U S A 2020; 117:3748-3758. [PMID: 32015106 PMCID: PMC7035603 DOI: 10.1073/pnas.1911951117] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Increased expression of extracellular matrix (ECM) proteins in circulating tumor cells (CTCs) suggests potential function of cancer cell-produced ECM in initiation of cancer cell colonization. Here, we showed that collagen and heat shock protein 47 (Hsp47), a chaperone facilitating collagen secretion and deposition, were highly expressed during the epithelial-mesenchymal transition (EMT) and in CTCs. Hsp47 expression induced mesenchymal phenotypes in mammary epithelial cells (MECs), enhanced platelet recruitment, and promoted lung retention and colonization of cancer cells. Platelet depletion in vivo abolished Hsp47-induced cancer cell retention in the lung, suggesting that Hsp47 promotes cancer cell colonization by enhancing cancer cell-platelet interaction. Using rescue experiments and functional blocking antibodies, we identified type I collagen as the key mediator of Hsp47-induced cancer cell-platelet interaction. We also found that Hsp47-dependent collagen deposition and platelet recruitment facilitated cancer cell clustering and extravasation in vitro. By analyzing DNA/RNA sequencing data generated from human breast cancer tissues, we showed that gene amplification and increased expression of Hsp47 were associated with cancer metastasis. These results suggest that targeting the Hsp47/collagen axis is a promising strategy to block cancer cell-platelet interaction and cancer colonization in secondary organs.
Collapse
Affiliation(s)
- Gaofeng Xiong
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536
| | - Jie Chen
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536
| | - Guoying Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536
| | - Shike Wang
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536
| | - Kunito Kawasaki
- Department of Molecular and Cellular Biology, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Jieqing Zhu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536
| | - Yan Zhang
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536
| | - Kazuhiro Nagata
- Department of Molecular and Cellular Biology, Kyoto Sangyo University, Kyoto 603-8555, Japan
| | - Zhenyu Li
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Kentucky, Lexington, KY 40536
| | - Binhua P Zhou
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, KY 40536
| | - Ren Xu
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536;
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
19
|
Lodochnikova OA, Akhmetshina EA, Mingaleva ER, Startseva VA, Litvinov IA, Plemenkov VV, Nikitina LE. Stable and reproducible supramolecular motif in the crystal structure of sulfonamides of the benzothiazine series fused to an epoxybornane moiety. Russ Chem Bull 2020. [DOI: 10.1007/s11172-020-2762-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
20
|
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: 4.0] [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
|
21
|
Structure-guided design of pure orthosteric inhibitors of αIIbβ3 that prevent thrombosis but preserve hemostasis. Nat Commun 2020; 11:398. [PMID: 31964886 PMCID: PMC6972956 DOI: 10.1038/s41467-019-13928-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023] Open
Abstract
A prevailing dogma is that inhibition of vascular thrombosis by antagonizing platelet integrin αIIbβ3 cannot be achieved without compromising hemostasis, thus causing serious bleeding and increased morbidity and mortality. It is speculated that these adverse outcomes result from drug-induced activating conformational changes in αIIbβ3 but direct proof is lacking. Here, we report the structure-guided design of peptide Hr10 and a modified form of the partial agonist drug tirofiban that act as "pure" antagonists of αIIbβ3, i.e., they no longer induce the conformational changes in αIIbβ3. Both agents inhibit human platelet aggregation but preserve clot retraction. Hr10 and modified tirofiban are as effective as partial agonist drugs in inhibiting vascular thrombosis in humanized mice, but neither causes serious bleeding, establishing a causal link between partial agonism and impaired hemostasis. Pure orthosteric inhibitors of αIIbβ3 may thus provide safer alternatives for human therapy, and valuable tools to probe structure-activity relationships in integrins.
Collapse
|
22
|
Kong J, Tian H, Zhang F, Zhang Z, Li J, Liu X, Li X, Liu J, Li X, Jin D, Yang X, Sun B, Guo T, Luo Y, Lu Y, Lin B, Liu T. Extracellular vesicles of carcinoma-associated fibroblasts creates a pre-metastatic niche in the lung through activating fibroblasts. Mol Cancer 2019; 18:175. [PMID: 31796058 PMCID: PMC6892147 DOI: 10.1186/s12943-019-1101-4] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 11/11/2019] [Indexed: 02/07/2023] Open
Abstract
Objectives Carcinoma-associated fibroblasts (CAFs) have been known to promote cancer progression by modifying the primary tumor microenvironment. We aimed to elucidate the intercellular communication between CAFs and secondary organs in salivary adenoid cystic carcinoma (SACC) metastasis. Methods Pre-metastatic and metastatic animal models of SACC were established using extracellular vesicles (EVs) from CAFs and SACC cells. Lung fibroblasts (LFs) were treated with EVs and their transcriptomic alterations were identified by RNA sequencing. ITRAQ were performed to analyze EV cargos. TC I-15 was used to inhibit EV uptake by LFs and SACC lung metastasis in vivo. Results Here, we show that CAF EVs induced lung pre-metastatic niche formation in mice and consequently increased SACC lung metastasis. The pre-metastatic niche induced by CAF EVs was different from that induced by SACC EVs. CAF EVs presented a great ability for matrix remodeling and periostin is a potential biomarker characterizing the CAF EV-induced pre-metastatic niche. We found that lung fibroblast activation promoted by CAF EVs was a critical event at the pre-metastatic niche. Integrin α2β1 mediated CAF EV uptake by lung fibroblasts, and its blockage by TC I-15 prevented lung pre-metastatic niche formation and subsequent metastasis. Plasma EV integrin β1 was considerably upregulated in the mice bearing xenografts with high risk of lung metastasis. Conclusions We demonstrated that CAF EVs participated in the pre-metastatic niche formation in the lung. Plasma EV integrin β1 might be a promising biomarker to predict SACC metastasis at an early stage. An integrated strategy targeting both tumor and stromal cells is necessary to prevent SACC metastasis.
Collapse
Affiliation(s)
- Jing Kong
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Hongzhu Tian
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Fuyin Zhang
- Department of Oral Surgery, the Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zebing Zhang
- Department of Oral Pathology, College of Stomatology, Jilin University, Changchun, China
| | - Jiao Li
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Xue Liu
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Xiancheng Li
- Department of Urology, the Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jing Liu
- Sino-UK Regenerative Medicine Center, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiaojie Li
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Dong Jin
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China
| | - Xuesong Yang
- Department of Biochemistry and Molecular Biology, Liaoning Provincial Core Lab of Glycobiology and Glycoengineering, Dalian Medical University, Dalian, China
| | - Bo Sun
- Department of Oral Surgery, the Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Tao Guo
- Department of Thoracic Surgery, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yong Luo
- Faculty of Chemical, Environmental and Biological Science and Technology, Dalian Technology University, Dalian, China
| | - Yao Lu
- Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Bingcheng Lin
- Faculty of Chemical, Environmental and Biological Science and Technology, Dalian Technology University, Dalian, China.,Department of Biotechnology, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Tingjiao Liu
- Department of Oral Pathology, College of Stomatology, Dalian Medical University, West Section No. 9, South Road of Lvshun, Dalian, 116044, China.
| |
Collapse
|
23
|
Tanaka S, Senda N, Iida A, Sehara-Fujisawa A, Ishii T, Sato F, Toi M, Itou J. In silico analysis-based identification of the target residue of integrin α6 for metastasis inhibition of basal-like breast cancer. Genes Cells 2019; 24:596-607. [PMID: 31295752 DOI: 10.1111/gtc.12714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 01/24/2023]
Abstract
Metastasis causes death in breast cancer patients. To inhibit breast cancer metastasis, we focused on integrin α6, a membrane protein that contributes to cell migration and metastasis. According to in silico analysis, we identified Asp-358 as an integrin α6-specific vertebrate-conserved residue and consequently as a potential therapeutic target. Because Asp-358 is located on the surface of the β propeller domain that interacts with other molecules for integrin α6 function, we hypothesized that a peptide with the sequence around Asp-358 competitively inhibits integrin α6 complex formation. We treated basal-like breast cancer cells with the peptide and observed reductions in cell migration and metastasis. The result of the immunoprecipitation assay showed that the peptide inhibited integrin α6 complex formation. Our immunofluorescence for phosphorylated paxillin, a marker of integrin-regulated focal adhesion, showed that the peptide reduced the number of focal adhesions. These results indicate that the peptide inhibits integrin α6 function. This study identified the functional residue of integrin α6 and designed the inhibitory peptide. For breast cancer patients, metastasis inhibition therapy may be developed in the future based on this study.
Collapse
Affiliation(s)
- Sunao Tanaka
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Noriko Senda
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsuo Iida
- Department of Regeneration Science and Engineering, Institute of Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Atsuko Sehara-Fujisawa
- Department of Regeneration Science and Engineering, Institute of Frontier Medical Sciences, Kyoto University, Kyoto, Japan
| | - Tomoko Ishii
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumiaki Sato
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Breast Surgery, Kansai Electric Power Hospital and Kansai Electric Power Medical Research Institute, Osaka, Japan
| | - Masakazu Toi
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junji Itou
- Department of Breast Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
24
|
Kotani N, Yamaguchi A, Ohnishi T, Kuwahara R, Nakano T, Nakano Y, Ida Y, Murakoshi T, Honke K. Proximity proteomics identifies cancer cell membrane cis-molecular complex as a potential cancer target. Cancer Sci 2019; 110:2607-2619. [PMID: 31228215 PMCID: PMC6676139 DOI: 10.1111/cas.14108] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 06/10/2019] [Accepted: 06/15/2019] [Indexed: 12/22/2022] Open
Abstract
Cancer‐specific antigens expressed in the cell membrane have been used as targets for several molecular targeted strategies in the last 20 years with remarkable success. To develop more effective cancer treatments, novel targets and strategies for targeted therapies are needed. Here, we examined the cancer cell membrane‐resident “cis‐bimolecular complex” as a possible cancer target (cis‐bimolecular cancer target: BiCAT) using proximity proteomics, a technique that has attracted attention in the last 10 years. BiCAT were detected using a previously developed method termed the enzyme‐mediated activation of radical source (EMARS), to label the components proximal to a given cell membrane molecule. EMARS analysis identified some BiCAT, such as close homolog of L1 (CHL1), fibroblast growth factor 3 (FGFR3) and α2 integrin, which are commonly expressed in mouse primary lung cancer cells and human lung squamous cell carcinoma cells. Analysis of cancer specimens from 55 lung cancer patients revealed that CHL1 and α2 integrin were highly co–expressed in almost all cancer tissues compared with normal lung tissues. As an example of BiCAT application, in vitro simulation of effective drug combinations used for multiple drug treatment strategies was performed using reagents targeted to BiCAT molecules. The combination treatment based on BiCAT information moderately suppressed cancer cell proliferation compared with single administration, suggesting that the information about BiCAT in cancer cells is useful for the appropriate selection of the combination among molecular targeted reagents. Thus, BiCAT has the potential to contribute to several molecular targeted strategies in future.
Collapse
Affiliation(s)
- Norihiro Kotani
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | - Arisa Yamaguchi
- Department of Biochemistry, Kochi University Medical School, Kochi, Japan
| | - Tomoko Ohnishi
- Department of Biochemistry, Kochi University Medical School, Kochi, Japan
| | - Ryusuke Kuwahara
- Quantum Wave Microscopy Unit, Okinawa Institute of Science and Technology Graduate University (OIST), Okinawa, Japan
| | - Takanari Nakano
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | - Yuka Nakano
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | - Yui Ida
- Department of Biochemistry, Saitama Medical University, Saitama, Japan
| | | | - Koichi Honke
- Department of Biochemistry, Kochi University Medical School, Kochi, Japan
| |
Collapse
|
25
|
Lazarovici P, Marcinkiewicz C, Lelkes PI. From Snake Venom's Disintegrins and C-Type Lectins to Anti-Platelet Drugs. Toxins (Basel) 2019; 11:toxins11050303. [PMID: 31137917 PMCID: PMC6563238 DOI: 10.3390/toxins11050303] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 05/16/2019] [Accepted: 05/24/2019] [Indexed: 12/13/2022] Open
Abstract
Snake venoms are attractive natural sources for drug discovery and development, with a number of substances either in clinical use or in research and development. These drugs were developed based on RGD-containing snake venom disintegrins, which efficiently antagonize fibrinogen activation of αIIbβ3 integrin (glycoprotein GP IIb/IIIa). Typical examples of anti-platelet drugs found in clinics are Integrilin (Eptifibatide), a heptapeptide derived from Barbourin, a protein found in the venom of the American Southeastern pygmy rattlesnake and Aggrastat (Tirofiban), a small molecule based on the structure of Echistatin, and a protein found in the venom of the saw-scaled viper. Using a similar drug discovery approach, linear and cyclic peptides containing the sequence K(R)TS derived from VP12, a C-type lectin protein found in the venom of Israeli viper venom, were used as a template to synthesize Vipegitide, a novel peptidomimetic antagonist of α2β1 integrin, with anti-platelet activity. This review focus on drug discovery of these anti-platelet agents, their indications for clinical use in acute coronary syndromes and percutaneous coronary intervention based on several clinical trials, as well as their adverse effects.
Collapse
Affiliation(s)
- Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel.
| | - Cezary Marcinkiewicz
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA.
| | - Peter I Lelkes
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA 19122, USA.
| |
Collapse
|
26
|
Li Q, Deng Q, Chao HP, Liu X, Lu Y, Lin K, Liu B, Tang GW, Zhang D, Tracz A, Jeter C, Rycaj K, Calhoun-Davis T, Huang J, Rubin MA, Beltran H, Shen J, Chatta G, Puzanov I, Mohler JL, Wang J, Zhao R, Kirk J, Chen X, Tang DG. Linking prostate cancer cell AR heterogeneity to distinct castration and enzalutamide responses. Nat Commun 2018; 9:3600. [PMID: 30190514 PMCID: PMC6127155 DOI: 10.1038/s41467-018-06067-7] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/02/2018] [Indexed: 12/31/2022] Open
Abstract
Expression of androgen receptor (AR) in prostate cancer (PCa) is heterogeneous but the functional significance of AR heterogeneity remains unclear. Screening ~200 castration-resistant PCa (CRPC) cores and whole-mount sections (from 89 patients) reveals 3 AR expression patterns: nuclear (nuc-AR), mixed nuclear/cytoplasmic (nuc/cyto-AR), and low/no expression (AR-/lo). Xenograft modeling demonstrates that AR+ CRPC is enzalutamide-sensitive but AR-/lo CRPC is resistant. Genome editing-derived AR+ and AR-knockout LNCaP cell clones exhibit distinct biological and tumorigenic properties and contrasting responses to enzalutamide. RNA-Seq and biochemical analyses, coupled with experimental combinatorial therapy, identify BCL-2 as a critical therapeutic target and provide proof-of-concept therapeutic regimens for both AR+/hi and AR-/lo CRPC. Our study links AR expression heterogeneity to distinct castration/enzalutamide responses and has important implications in understanding the cellular basis of prostate tumor responses to AR-targeting therapies and in facilitating development of novel therapeutics to target AR-/lo PCa cells/clones.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Benzamides
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Male
- Mice, Inbred NOD
- Mice, Knockout
- Molecular Targeted Therapy
- Nitriles
- Phenylthiohydantoin/analogs & derivatives
- Phenylthiohydantoin/pharmacology
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/pathology
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Receptors, Androgen/genetics
- Receptors, Androgen/metabolism
- Signal Transduction
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Qiuhui Li
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology,, Wuhan University, 430079, Wuhan, China
| | - Qu Deng
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
- Program in Molecular Carcinogenesis, University of Texas Graduate School for Biomedical Sciences (GSBS), Houston, TX, 77030, USA
| | - Hsueh-Ping Chao
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
- Program in Molecular Carcinogenesis, University of Texas Graduate School for Biomedical Sciences (GSBS), Houston, TX, 77030, USA
| | - Xin Liu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Bigang Liu
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Gregory W Tang
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Dingxiao Zhang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Amanda Tracz
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Collene Jeter
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Kiera Rycaj
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Tammy Calhoun-Davis
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Jiaoti Huang
- Department of Pathology, Duke University of School of Medicine, Durham, NC, 27710, USA
| | - Mark A Rubin
- Caryl and Israel Englander Institute for Precision Medicine, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center at Weill Cornell Medicine, New York, NY, 10021, USA
| | - Himisha Beltran
- Caryl and Israel Englander Institute for Precision Medicine, New York-Presbyterian Hospital, Weill Cornell Medical College, New York, NY, 10065, USA
- Sandra and Edward Meyer Cancer Center at Weill Cornell Medicine, New York, NY, 10021, USA
| | - Jianjun Shen
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA
| | - Gurkamal Chatta
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - James L Mohler
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Ruizhe Zhao
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jason Kirk
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Xin Chen
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA.
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China.
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Epigenetics and Molecular Carcinogenesis, University of Texas M.D. Anderson Cancer Center, Science Park, Smithville, TX, 78957, USA.
- Cancer Stem Cell Institute, Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, 200120, Shanghai, China.
| |
Collapse
|
27
|
Zhang S, Zhao E, Winkelstein BA. A Nociceptive Role for Integrin Signaling in Pain After Mechanical Injury to the Spinal Facet Capsular Ligament. Ann Biomed Eng 2017; 45:2813-2825. [PMID: 28924864 PMCID: PMC5693676 DOI: 10.1007/s10439-017-1917-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 09/01/2017] [Indexed: 12/17/2022]
Abstract
Integrins modulate chemically-induced nociception in a variety of inflammatory and neuropathic pain models. Yet, the role of integrins in mechanically-induced pain remains undefined, despite its well-known involvement in cell adhesion and mechanotransduction. Excessive spinal facet capsular ligament stretch is a common injury that induces morphological and functional changes in its innervating afferent neurons and can lead to pain. However, the local mechanisms underlying the translation from tissue deformation to pain signaling are unclear, impeding effective treatment. Therefore, the involvement of the integrin subunit β1 in pain signaling from facet injury was investigated in complementary in vivo and in vitro studies. An anatomical study in the rat identified expression of the integrin subunit β1 in dorsal root ganglion (DRG) neurons innervating the facet, with greater expression in peptidergic than non-peptidergic DRG neurons. Painful facet capsule stretch in the rat upregulated the integrin subunit β1 in small- and medium-diameter DRG neurons at day 7. Inhibiting the α2β1 integrin in a DRG-collagen culture prior to its stretch injury prevented strain-induced increases in axonal substance P (SP) in a dose-dependent manner. Together, these findings suggest that integrin subunit β1-dependent pathways may contribute to SP-mediated pain from mechanical injury of the facet capsular ligament.
Collapse
Affiliation(s)
- Sijia Zhang
- Department of Bioengineering, University of Pennsylvania, 240 Skirkanich Hall, 210 S. 33rd St, Philadelphia, PA, 19104-6321, USA
| | - Ethan Zhao
- Department of Bioengineering, University of Pennsylvania, 240 Skirkanich Hall, 210 S. 33rd St, Philadelphia, PA, 19104-6321, USA
| | - Beth A Winkelstein
- Department of Bioengineering, University of Pennsylvania, 240 Skirkanich Hall, 210 S. 33rd St, Philadelphia, PA, 19104-6321, USA.
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
28
|
Reed NI, Tang YZ, McIntosh J, Wu Y, Molnar KS, Civitavecchia A, Sheppard D, DeGrado WF, Jo H. Exploring N-Arylsulfonyl-l-proline Scaffold as a Platform for Potent and Selective αvβ1 Integrin Inhibitors. ACS Med Chem Lett 2016; 7:902-907. [PMID: 27774126 DOI: 10.1021/acsmedchemlett.6b00196] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/30/2016] [Indexed: 02/08/2023] Open
Abstract
One small molecule inhibitor of αvβ1 integrin, c8, shows antifibrotic effects in multiple in vivo mouse models. Here we synthesized c8 analogues and systematically investigate their structure-activity relationships (SAR) in αvβ1 integrin inhibition. N-Phenylsulfonyl-l-homoproline analogues of c8 maintained excellent potency against αvβ1 integrin while retaining good selectivity over other RGD integrins. In addition, 2-aminopyridine or cyclic guanidine analogues were shown to be equally potent to c8. A rigid phenyl linker increased the potency compared to c8, but the selectivity over other RGD integrins diminished. These results can provide further insights on design of αvβ1 integrin inhibitors as antifibrotics.
Collapse
Affiliation(s)
- Nilgun Isik Reed
- Department of Medicine, University of California−San Francisco, San Francisco, California 94153, United States
| | - You-Zhi Tang
- Department of Pharmaceutical Chemistry, University of California−San Francisco, San Francisco, California 94158, United States
- College of Veterinary Medicine, South China Agricultural University, Guangdong 510642, China
| | - Joel McIntosh
- Department of Pharmaceutical Chemistry, University of California−San Francisco, San Francisco, California 94158, United States
| | - Yibing Wu
- Department of Pharmaceutical Chemistry, University of California−San Francisco, San Francisco, California 94158, United States
| | - Kathleen S. Molnar
- Department of Pharmaceutical Chemistry, University of California−San Francisco, San Francisco, California 94158, United States
| | - Annafelicia Civitavecchia
- Department of Pharmaceutical Chemistry, University of California−San Francisco, San Francisco, California 94158, United States
| | - Dean Sheppard
- Department of Medicine, University of California−San Francisco, San Francisco, California 94153, United States
| | - William F. DeGrado
- Department of Pharmaceutical Chemistry, University of California−San Francisco, San Francisco, California 94158, United States
- Cardiovascular
Research Institute, University of California−San Francisco, San Francisco, California 94158, United States
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California−San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
29
|
Xu XR, Carrim N, Neves MAD, McKeown T, Stratton TW, Coelho RMP, Lei X, Chen P, Xu J, Dai X, Li BX, Ni H. Platelets and platelet adhesion molecules: novel mechanisms of thrombosis and anti-thrombotic therapies. Thromb J 2016; 14:29. [PMID: 27766055 PMCID: PMC5056500 DOI: 10.1186/s12959-016-0100-6] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Platelets are central mediators of thrombosis and hemostasis. At the site of vascular injury, platelet accumulation (i.e. adhesion and aggregation) constitutes the first wave of hemostasis. Blood coagulation, initiated by the coagulation cascades, is the second wave of thrombin generation and enhance phosphatidylserine exposure, can markedly potentiate cell-based thrombin generation and enhance blood coagulation. Recently, deposition of plasma fibronectin and other proteins onto the injured vessel wall has been identified as a new "protein wave of hemostasis" that occurs prior to platelet accumulation (i.e. the classical first wave of hemostasis). These three waves of hemostasis, in the event of atherosclerotic plaque rupture, may turn pathogenic, and cause uncontrolled vessel occlusion and thrombotic disorders (e.g. heart attack and stroke). Current anti-platelet therapies have significantly reduced cardiovascular mortality, however, on-treatment thrombotic events, thrombocytopenia, and bleeding complications are still major concerns that continue to motivate innovation and drive therapeutic advances. Emerging evidence has brought platelet adhesion molecules back into the spotlight as targets for the development of novel anti-thrombotic agents. These potential antiplatelet targets mainly include the platelet receptors glycoprotein (GP) Ib-IX-V complex, β3 integrins (αIIb subunit and PSI domain of β3 subunit) and GPVI. Numerous efforts have been made aiming to balance the efficacy of inhibiting thrombosis without compromising hemostasis. This mini-review will update the mechanisms of thrombosis and the current state of antiplatelet therapies, and will focus on platelet adhesion molecules and the novel anti-thrombotic therapies that target them.
Collapse
Affiliation(s)
- Xiaohong Ruby Xu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong People’s Republic of China
| | - Naadiya Carrim
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada
- Canadian Blood Services, Toronto, ON Canada
| | - Miguel Antonio Dias Neves
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada
| | - Thomas McKeown
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada
| | - Tyler W. Stratton
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada
| | - Rodrigo Matos Pinto Coelho
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada
| | - Xi Lei
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada
| | - Pingguo Chen
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada
- Canadian Blood Services, Toronto, ON Canada
| | - Jianhua Xu
- CCOA Therapeutics Inc, Toronto, ON Canada
| | - Xiangrong Dai
- Lee’s Pharmaceutical holdings limited, Shatin Hong Kong, China
- Zhaoke Pharmaceutical co. limited, Hefei, Anhui China
| | - Benjamin Xiaoyi Li
- Lee’s Pharmaceutical holdings limited, Shatin Hong Kong, China
- Zhaoke Pharmaceutical co. limited, Hefei, Anhui China
- Hong Kong University of Science and technology, Hong Kong, China
| | - Heyu Ni
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Toronto, ON Canada
- Canadian Blood Services, Toronto, ON Canada
- CCOA Therapeutics Inc, Toronto, ON Canada
- Department of Medicine and Department of Physiology, University of Toronto, Toronto, ON Canada
| |
Collapse
|
30
|
A Nitric Oxide-Releasing Self-Assembled Peptide Amphiphile Nanomatrix for Improving the Biocompatibility of Microporous Hollow Fibers. ASAIO J 2016; 61:589-95. [PMID: 26102178 DOI: 10.1097/mat.0000000000000257] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Oxygenators are critical components of extracorporeal circuits used frequently in cardiopulmonary bypass and intensive care, but platelet activation and induction of a complex inflammatory response are usually observed with their use. To improve the biocompatibility of oxygenators, we developed a nitric oxide (NO)-releasing, self-assembled peptide amphiphile nanomatrix. The nanomatrix formed a homogenous coating over the microporous hollow fibers as demonstrated by scanning electron microscopy. We quantitated platelet adhesion to the artificial fibers by measuring absorbance/area of platelets (Abs/A; nm/m2) using acid phosphatase assay. There was a 17-fold decrease in platelet adhesion to the nanomatrix (Abs/A = 0.125) compared with collagen controls (Abs/A = 2.07; p < 0.05) and a 22-fold decrease compared with uncoated fibers (Abs/A = 2.75; p < 0.05). Importantly, the nanomatrix coating did not impede oxygen transfer in water through coated fiber modules (p > 0.05) in a benchtop test circuit at different flow rates as estimated by change in partial pressure of oxygen in relation to water velocity through fibers. These findings demonstrate the feasibility of coating microporous hollow fibers with a NO-releasing self-assembled amphiphile nanomatrix that may improve the biocompatibility of the hollow fibers without affecting their gas exchange capacity.
Collapse
|
31
|
Development of antithrombotic nanoconjugate blocking integrin α2β1-collagen interactions. Sci Rep 2016; 6:26292. [PMID: 27195826 PMCID: PMC4872532 DOI: 10.1038/srep26292] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/28/2016] [Indexed: 01/07/2023] Open
Abstract
An antithrombotic nanoconjugate was designed in which a designed biomimetic peptide LWWNSYY was immobilized to the surface of poly(glycidyl methacrylate) nanoparticles (PGMA NPs). Our previous work has demonstrated LWWNSYY to be an effective inhibitor of integrin α2β1-collagen interaction and subsequent thrombus formation, however its practical application suffered from the formation of clusters in physiological environment caused by its high hydrophobicity. In our present study, the obtained LWWNSYY-PGMA nanoparticles (L-PGMA NPs) conjugate, with an improved dispersibility of LWWNSYY by PGMA NPs, have shown binding to collagen receptors with a Kd of 3.45 ± 1.06 μM. L-PGMA NPs have also proven capable of inhibiting platelet adhesion in vitro with a reduced IC50 of 1.83 ± 0.29 μg/mL. High inhibition efficiency of L-PGMA NPs in thrombus formation was further confirmed in vivo with a 50% reduction of thrombus weight. Therefore, L-PGMA NPs were developed as a high-efficiency antithrombotic nanomedicine targeted for collagen exposed on diseased blood vessel wall.
Collapse
|
32
|
Conroy KP, Kitto LJ, Henderson NC. αv integrins: key regulators of tissue fibrosis. Cell Tissue Res 2016; 365:511-9. [PMID: 27139180 PMCID: PMC5010580 DOI: 10.1007/s00441-016-2407-9] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 04/08/2016] [Indexed: 01/09/2023]
Abstract
Chronic tissue injury with fibrosis results in the disruption of tissue architecture, organ dysfunction and eventual organ failure. Therefore, the development of effective anti-fibrotic therapies is urgently required. During fibrogenesis, complex interplay occurs between cellular and extracellular matrix components of the wound healing response. Integrins, a family of transmembrane cell adhesion molecules, play a key role in mediating intercellular and cell-matrix interactions. Thus, integrins provide a major node of communication between the extracellular matrix, inflammatory cells, fibroblasts and parenchymal cells and, as such, are intimately involved in the initiation, maintenance and resolution of tissue fibrosis. Modulation of members of the αv integrin family has exhibited profound effects on fibrosis in multiple organs and disease states. In this review, we discuss the current knowledge of the mechanisms of αv-integrin-mediated regulation of fibrogenesis and show that the therapeutic targeting of specific αv integrins represents a promising avenue to treat patients with a broad range of fibrotic diseases.
Collapse
Affiliation(s)
- Kylie P Conroy
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Laura J Kitto
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Neil C Henderson
- MRC Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
33
|
Reed NI, Jo H, Chen C, Tsujino K, Arnold TD, DeGrado WF, Sheppard D. The αvβ1 integrin plays a critical in vivo role in tissue fibrosis. Sci Transl Med 2016; 7:288ra79. [PMID: 25995225 DOI: 10.1126/scitranslmed.aaa5094] [Citation(s) in RCA: 223] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Integrins are transmembrane heterodimeric receptors that contribute to diverse biological functions and play critical roles in many human diseases. Studies using integrin subunit knockout mice and inhibitory antibodies have identified important roles for nearly every integrin heterodimer and led to the development of a number of potentially useful therapeutics. One notable exception is the αvβ1 integrin. αv and β1 subunits are individually present in numerous dimer pairs, making it challenging to infer specific roles for αvβ1 by genetic inactivation of individual subunits, and αvβ1 complex-specific blocking antibodies do not yet exist. We therefore developed a potent and highly specific small-molecule inhibitor of αvβ1 to probe the function of this understudied integrin. We found that αvβ1, which is highly expressed on activated fibroblasts, directly binds to the latency-associated peptide of transforming growth factor-β1 (TGFβ1) and mediates TGFβ1 activation. Therapeutic delivery of this αvβ1 inhibitor attenuated bleomycin-induced pulmonary fibrosis and carbon tetrachloride-induced liver fibrosis, suggesting that drugs based on this lead compound could be broadly useful for treatment of diseases characterized by excessive tissue fibrosis.
Collapse
Affiliation(s)
- Nilgun I Reed
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Hyunil Jo
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Chun Chen
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kazuyuki Tsujino
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Thomas D Arnold
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - William F DeGrado
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Dean Sheppard
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
34
|
Arosio D, Casagrande C. Advancement in integrin facilitated drug delivery. Adv Drug Deliv Rev 2016; 97:111-43. [PMID: 26686830 DOI: 10.1016/j.addr.2015.12.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/27/2015] [Accepted: 12/03/2015] [Indexed: 02/06/2023]
Abstract
The research of integrin-targeted anticancer agents has recorded important advancements in ingenious design of delivery systems, based either on the prodrug approach, or on nanoparticle carriers, but for now, none of these has reached a clinical stage of development. Past work in this area has been extensively reviewed by us and others. Thus, the purpose and scope of the present review is to survey the advancement reported in the last 3years, with focus on innovative delivery systems that appear to afford openings for future developments. These systems exploit the labelling with conventional and novel integrin ligands for targeting the interface of cancer cells and of endothelial cells involved in cancer angiogenesis, with the proteins of the extracellular matrix, in the circulation, in tissues, and in tumour stroma, as the site of progression and metastatic evolution of the disease. Furthermore, these systems implement the expertise in the development of nanomedicines to the purpose of achieving preferential biodistribution and uptake in cancer tissues, internalisation in cancer cells, and release of the transported drugs at intracellular sites. The assessment of the value of controlling these factors, and their combination, for future developments requires support of biological testing in appropriate mechanistic models, but also imperatively demand confirmation in therapeutically relevant in vivo models for biodistribution, efficacy, and lack of off-target effects. Thus, among many studies, we have tried to point out the results supported by relevant in vivo studies, and we have emphasised in specific sections those addressing the medical needs of drug delivery to brain tumours, as well as the delivery of oligonucleotides modulating gene-dependent pathological mechanism. The latter could constitute the basis of a promising third branch in the therapeutic armamentarium against cancer, in addition to antibody-based agents and to cytotoxic agents.
Collapse
Affiliation(s)
- Daniela Arosio
- Istituto di Scienze e Tecnologie Molecolari (ISTM), CNR, Via C. Golgi 19, I-20133 Milan, Italy.
| | - Cesare Casagrande
- Università degli Studi di Milano, Dipartimento di Chimica, Via C. Golgi 19, I-20133 Milan, Italy.
| |
Collapse
|
35
|
Jiang L, Sun Z, Chen X, Li J, Xu Y, Zu Y, Hu J, Han D, Yang C. Cells Sensing Mechanical Cues: Stiffness Influences the Lifetime of Cell-Extracellular Matrix Interactions by Affecting the Loading Rate. ACS NANO 2016; 10:207-17. [PMID: 26701367 DOI: 10.1021/acsnano.5b03157] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
The question of how cells sense substrate mechanical cues has gained increasing attention among biologists. By introducing contour-based data analysis to single-cell force spectroscopy, we identified a loading-rate threshold for the integrin α2β1-DGEA bond beyond which a dramatic increase in bond lifetime was observed. On the basis of mechanical cues (elasticity or topography), the effective spring constant of substrates k is mapped to the loading rate r under actomyosin pulling speed v, which, in turn, affects the lifetime of the integrin-ligand bond. Additionally, downregulating v with a low-dose blebbistatin treatment promotes the neuronal lineage specification of mesenchymal stem cells on osteogenic stiff substrates. Thus, sensing of the loading rate is central to how cells sense mechanical cues that affect cell-extracellular matrix interactions and stem cell differentiation.
Collapse
Affiliation(s)
- Li Jiang
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| | - Zhenglong Sun
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
- Suzhou Institute of Biomedical Engineering and Technology, China Academy of Science , Suzhou 215163, People's Republic of China
| | - Xiaofei Chen
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
- Beijing Institute of Aerospace Systems Engineering , Beijing 100076, People's Republic of China
| | - Jing Li
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| | - Yue Xu
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| | - Yan Zu
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| | - Jiliang Hu
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| | - Dong Han
- National Center for Nanoscience and Technology , Beijing 100190, People's Republic of China
| | - Chun Yang
- Institute of Biomechanics and Medical Engineering, School of Aerospace, Tsinghua University , Beijing 100084, People's Republic of China
| |
Collapse
|
36
|
Nissinen L, Rappu P, Ollikka P, Nieminen J, Marjamäki A, Heino J. Platelet response to a small molecule inhibitor of α2β1 integrin is associated with ITGA2 C807T dimorphism. Platelets 2015; 27:378-80. [PMID: 26556301 DOI: 10.3109/09537104.2015.1095877] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
High expression of the collagen receptor, α2β1 integrin, on platelets of ITGA2 807T-allele carriers has been identified as a risk factor for thromboembolic conditions, and α2β1 inhibitors are considered to be potential therapeutic agents. In 59 genotyped individuals, we measured α2 expression levels on platelets and analyzed platelet adhesion to collagen under flow conditions. A sulfonamide-type small-molecule inhibitor of α2β1 integrin decreased average platelet adhesion in individuals with the C/T807T genotype but not in those harboring C807C. Thus, genotype can be used to select a human subpopulation that has the highest probability of showing a positive response to α2β1 inhibitors.
Collapse
Affiliation(s)
- Liisa Nissinen
- a Department of Biochemistry , University of Turku , Turku , Finland.,b BioTie Therapies Corp ., Turku , Finland
| | - Pekka Rappu
- a Department of Biochemistry , University of Turku , Turku , Finland
| | | | | | - Anne Marjamäki
- a Department of Biochemistry , University of Turku , Turku , Finland.,b BioTie Therapies Corp ., Turku , Finland
| | - Jyrki Heino
- a Department of Biochemistry , University of Turku , Turku , Finland
| |
Collapse
|
37
|
Zeng Y, Zhang L, Hu Z, Yang Q, Ma M, Liu B, Xia J, Xu H, Liu Y, Du X. 807C/T polymorphism of platelet glycoprotein Ia gene is associated with cerebral hemorrhage in a Chinese population. Int J Neurosci 2015; 126:729-33. [DOI: 10.3109/00207454.2015.1067891] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
38
|
Nissinen L, Ojala M, Langen B, Dost R, Pihlavisto M, Käpylä J, Marjamäki A, Heino J. Sulfonamide inhibitors of α2β1 integrin reveal the essential role of collagen receptors in in vivo models of inflammation. Pharmacol Res Perspect 2015; 3:e00146. [PMID: 26171226 PMCID: PMC4492762 DOI: 10.1002/prp2.146] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 03/17/2015] [Accepted: 03/25/2015] [Indexed: 01/30/2023] Open
Abstract
Small molecule inhibitors of α2β1 integrin, a major cellular collagen receptor, have been reported to inhibit platelet function, kidney injury, and angiogenesis. Since α2β1 integrin is abundantly expressed on various inflammation-associated cells, we tested whether recently developed α2β1 blocking sulfonamides have anti-inflammatory properties. Integrin α2β1 inhibitors were shown to reduce the signs of inflammation in arachidonic acid-induced ear edema, PAF stimulated air pouch, ovalbumin-induced skin hypersensitivity, adjuvant arthritis, and collagen-induced arthritis. Thus, these sulfonamides are potential drugs for acute and allergic inflammation, hypersensitivity, and arthritis. One sulfonamide with potent anti-inflammatory activity has previously been reported to be selective for activated integrins, but not to inhibit platelet function. Thus, the experiments also revealed fundamental differences in the action of nonactivated and activated α2β1 integrins in inflammation when compared to thrombosis.
Collapse
Affiliation(s)
- Liisa Nissinen
- Department of Biochemistry, University of Turku 20014, Turku, Finland ; Biotie Therapies Corp Turku, Finland
| | | | | | - Rita Dost
- BioTie Therapies GmbH Radebeul, Germany
| | | | - Jarmo Käpylä
- Department of Biochemistry, University of Turku 20014, Turku, Finland
| | - Anne Marjamäki
- Department of Biochemistry, University of Turku 20014, Turku, Finland ; Biotie Therapies Corp Turku, Finland
| | - Jyrki Heino
- Department of Biochemistry, University of Turku 20014, Turku, Finland
| |
Collapse
|
39
|
Momic T, Katzhendler J, Shai E, Noy E, Senderowitz H, Eble JA, Marcinkiewicz C, Varon D, Lazarovici P. Vipegitide: a folded peptidomimetic partial antagonist of α2β1 integrin with antiplatelet aggregation activity. Drug Des Devel Ther 2015; 9:291-304. [PMID: 25609915 PMCID: PMC4294129 DOI: 10.2147/dddt.s72844] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Linear peptides containing the sequence WKTSRTSHY were used as lead compounds to synthesize a novel peptidomimetic antagonist of α2β1 integrin, with platelet aggregation-inhibiting activity, named Vipegitide. Vipegitide is a 13-amino acid, folded peptidomimetic molecule, containing two α-aminoisobutyric acid residues at positions 6 and 8 and not stable in human serum. Substitution of glycine and tryptophan residues at positions 1 and 2, respectively, with a unit of two polyethylene glycol (PEG) molecules yielded peptidomimetic Vipegitide-PEG2, stable in human serum for over 3 hours. Vipegitide and Vipegitide-PEG2 showed high potency (7×10(-10) M and 1.5×10(-10) M, respectively) and intermediate efficacy (40% and 35%, respectively) as well as selectivity toward α2 integrin in inhibition of adhesion of α1/α2 integrin overexpressing cells toward respective collagens. Interaction of both peptidomimetics with extracellular active domain of α2 integrin was confirmed in cell-free binding assay with recombinant α2 A-domain. Integrin α2β1 receptor is found on the platelet membrane and triggers collagen-induced platelet aggregation. Vipegitide and Vipegitide-PEG2 inhibited α2β1 integrin-mediated adhesion of human and murine platelets under the flow condition, by 50%. They efficiently blocked adenosine diphosphate- and collagen I-induced platelet aggregation in platelet rich plasma and whole human blood. Higher potency of Vipegitide than Vipegitide-PEG2 is consistent with results of computer modeling of the molecules in water. These peptidomimetic molecules were acutely tolerated in mice upon intravenous bolus injection of 50 mg/kg. These results underline the potency of Vipegitide and Vipegitide-PEG2 molecules as platelet aggregation-inhibiting drug lead compounds in antithrombotic therapy.
Collapse
Affiliation(s)
- Tatjana Momic
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jehoshua Katzhendler
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ela Shai
- Department of Hematology, Coagulation Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Efrat Noy
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, Israel
| | | | - Johannes A Eble
- Institute for Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Cezary Marcinkiewicz
- Department of Bioengineering, College of Engineering, Temple University, Philadelphia, PA, USA
| | - David Varon
- Department of Hematology, Coagulation Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
40
|
Borza CM, Chen X, Zent R, Pozzi A. Cell Receptor-Basement Membrane Interactions in Health and Disease: A Kidney-Centric View. CURRENT TOPICS IN MEMBRANES 2015; 76:231-53. [PMID: 26610916 PMCID: PMC4913201 DOI: 10.1016/bs.ctm.2015.07.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell-extracellular matrix (ECM) interactions are essential for tissue development, homeostasis, and response to injury. Basement membranes (BMs) are specialized ECMs that separate epithelial or endothelial cells from stromal components and interact with cells via cellular receptors, including integrins and discoidin domain receptors. Disruption of cell-BM interactions due to either injury or genetic defects in either the ECM components or cellular receptors often lead to irreversible tissue injury and loss of organ function. Animal models that lack specific BM components or receptors either globally or in selective tissues have been used to help with our understanding of the molecular mechanisms whereby cell-BM interactions regulate organ function in physiological and pathological conditions. We review recently published works on animal models that explore how cell-BM interactions regulate kidney homeostasis in both health and disease.
Collapse
Affiliation(s)
- Corina M. Borza
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Xiwu Chen
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
| | - Roy Zent
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Medicine, Veterans Administration Hospital, Nashville, TN, 37232
| | - Ambra Pozzi
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, 37232
- Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN, 37232
- Department of Medicine, Veterans Administration Hospital, Nashville, TN, 37232
| |
Collapse
|
41
|
Cao B, Hutt OE, Zhang Z, Li S, Heazlewood SY, Williams B, Smith JA, Haylock DN, Savage GP, Nilsson SK. Design, synthesis and binding properties of a fluorescent α₉β₁/α₄β₁ integrin antagonist and its application as an in vivo probe for bone marrow haemopoietic stem cells. Org Biomol Chem 2014; 12:965-78. [PMID: 24363056 DOI: 10.1039/c3ob42332h] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The α9β1 and α4β1 integrin subtypes are expressed on bone marrow haemopoietic stem cells and have important roles in stem cell regulation and trafficking. Although the roles of α4β1 integrin have been thoroughly investigated with respect to HSC function, the role of α9β1 integrin remains poorly characterised. Small molecule fluorescent probes are useful tools for monitoring biological processes in vivo, to determine cell-associated protein localisation and activation, and to elucidate the mechanism of small molecule mediated protein interactions. Herein, we report the design, synthesis and integrin-dependent cell binding properties of a new fluorescent α9β1 integrin antagonist (R-BC154), which was based on a series of N-phenylsulfonyl proline dipeptides and assembled using the Cu(I)-catalyzed azide alkyne cycloaddition (CuAAC) reaction. Using transfected human glioblastoma LN18 cells, we show that R-BC154 exhibits high nanomolar binding affinities to α9β1 integrin with potent cross-reactivity against α4β1 integrin under physiological mimicking conditions. On-rate and off-rate measurements revealed distinct differences in the binding kinetics between α9β1 and α4β1 integrins, which showed faster binding to α4β1 integrin relative to α9β1, but more prolonged binding to the latter. Finally, we show that R-BC154 was capable of binding rare populations of bone marrow haemopoietic stem and progenitor cells when administered to mice. Thus, R-BC154 represents a useful multi-purpose fluorescent integrin probe that can be used for (1) screening small molecule inhibitors of α9β1 and α4β1 integrins; (2) investigating the biochemical properties of α9β1 and α4β1 integrin binding and (3) investigating integrin expression and activation on defined cell phenotypes in vivo.
Collapse
Affiliation(s)
- Benjamin Cao
- CSIRO Materials Science and Engineering, Bag 10, Clayton Sth MDC, VIC 3169, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: catalytic receptors. Br J Pharmacol 2014; 170:1676-705. [PMID: 24528241 PMCID: PMC3892291 DOI: 10.1111/bph.12449] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Catalytic receptors are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, nuclear hormone receptors, transporters and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen P H Alexander
- School of Life Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
A 3D matrix platform for the rapid generation of therapeutic anti-human carcinoma monoclonal antibodies. Proc Natl Acad Sci U S A 2014; 111:14882-7. [PMID: 25267635 DOI: 10.1073/pnas.1410996111] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Efforts to develop unbiased screens for identifying novel function-blocking monoclonal antibodies (mAbs) in human carcinomatous states have been hampered by the limited ability to design in vitro models that recapitulate tumor cell behavior in vivo. Given that only invasive carcinoma cells gain permanent access to type I collagen-rich interstitial tissues, an experimental platform was established in which human breast cancer cells were embedded in 3D aldimine cross-linked collagen matrices and used as an immunogen to generate mAb libraries. In turn, cancer-cell-reactive antibodies were screened for their ability to block carcinoma cell proliferation within collagen hydrogels that mimic the in vivo environment. As a proof of principle, a single function-blocking mAb out of 15 identified was selected for further analysis and found to be capable of halting carcinoma cell proliferation, inducing apoptosis, and exerting global changes in gene expression in vitro. The ability of this mAb to block carcinoma cell proliferation and metastatic activity was confirmed in vivo, and the target antigen was identified by mass spectroscopy as the α2 subunit of the α2β1 integrin, one of the major type I collagen-binding receptors in mammalian cells. Validating the ability of the in vitro model to predict patterns of antigen expression in the disease setting, immunohistochemical analyses of tissues from patients with breast cancer verified markedly increased expression of the α2 subunit in vivo. These results not only highlight the utility of this discovery platform for rapidly selecting and characterizing function-blocking, anticancer mAbs in an unbiased fashion, but also identify α2β1 as a potential target in human carcinomatous states.
Collapse
|
44
|
Mironova EV, Lodochnikova OA, Krivolapov DB, Veremeichik YV, Plemenkov VV, Litvinov IA. Crystal structure of cyclic sulfin- and sulfonamides of the thiazine series: Conformation, intra- and intermolecular interactions. J STRUCT CHEM+ 2014. [DOI: 10.1134/s0022476614030238] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
45
|
Abstract
While platelet activation is essential to maintain blood vessel patency and minimize loss of blood upon injury, untimely or excessive activity can lead to unwanted platelet activation and aggregation. Resultant thrombosis has the potential to block blood vessels, causing myocardial infarction or stroke. To tackle this major cause of mortality, clinical therapies that target platelet responsiveness (antiplatelet therapy) can successfully reduce cardiovascular events, especially in people at higher risk; however, all current antiplatelet therapies carry an increased probability of bleeding. This review will evaluate new and emerging targets for antithrombotics, focusing particularly on platelet glycoprotein VI, as blockade or depletion of this platelet-specific receptor conveys benefits in experimental models of thrombosis and thromboinflammation without causing major bleeding complications.
Collapse
Affiliation(s)
- Robert K Andrews
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Jane F Arthur
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| | - Elizabeth E Gardiner
- Australian Centre for Blood Diseases, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
46
|
Anil Kumar KS, Misra A, Siddiqi TI, Srivastava S, Jain M, Bhatta RS, Barthwal M, Dikshit M, Dikshit DK. Synthesis and identification of chiral aminomethylpiperidine carboxamides as inhibitor of collagen induced platelet activation. Eur J Med Chem 2014; 81:456-72. [PMID: 24859764 DOI: 10.1016/j.ejmech.2014.05.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 05/01/2014] [Accepted: 05/04/2014] [Indexed: 11/19/2022]
Abstract
A series of chiral lactam carboxamides of aminomethylpiperidine were synthesized and investigated for the collagen induced in vitro anti-platelet efficacy and collagen plus epinephrine induced in vivo pulmonary thromboembolism. The compound 31a (30 μM/kg) displayed a remarkable antithrombotic efficacy (60% protection) which was sustained for more than 24 h and points to its excellent bioavailability. The compounds 31a (IC50 = 6.6 μM) and 32a (IC50 = 37 μM), as well as their racemic mixture 28i (IC50 = 16 μM) significantly inhibited collagen-induced human platelet aggregation in vitro. Compound 34c displayed dual mechanism of action against both collagen (IC50 = 3.3 μM) and U46619 (IC50 = 2.7 μM) induced platelet aggregation. The pharmacokinetic study of 31a indicated very faster absorption, prolonged and constant systemic exposure and thereby exhibiting better therapeutic response.
Collapse
Affiliation(s)
- K S Anil Kumar
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sec-10, Janakipuram Ext., Sitapur Road, Lucknow 226 031, India
| | - Ankita Misra
- Pharmacology Division, CSIR-Central Drug Research Institute, Sec-10, Janakipuram Ext., Sitapur Road, Lucknow 226 031, India
| | - Tanveer Irshad Siddiqi
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sec-10, Janakipuram Ext., Sitapur Road, Lucknow 226 031, India
| | - Stuti Srivastava
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sec-10, Janakipuram Ext., Sitapur Road, Lucknow 226 031, India
| | - Manish Jain
- Pharmacology Division, CSIR-Central Drug Research Institute, Sec-10, Janakipuram Ext., Sitapur Road, Lucknow 226 031, India
| | - Rabi Sankar Bhatta
- Pharmacokinetics & Metabolism Division, CSIR-Central Drug Research Institute, Sec-10, Janakipuram Ext., Sitapur Road, Lucknow 226 031, India
| | - Manoj Barthwal
- Pharmacology Division, CSIR-Central Drug Research Institute, Sec-10, Janakipuram Ext., Sitapur Road, Lucknow 226 031, India
| | - Madhu Dikshit
- Pharmacology Division, CSIR-Central Drug Research Institute, Sec-10, Janakipuram Ext., Sitapur Road, Lucknow 226 031, India.
| | - Dinesh K Dikshit
- Medicinal and Process Chemistry Division, CSIR-Central Drug Research Institute, Sec-10, Janakipuram Ext., Sitapur Road, Lucknow 226 031, India.
| |
Collapse
|
47
|
Halland N, Blum H, Buning C, Kohlmann M, Lindenschmidt A. Small Macrocycles As Highly Active Integrin α2β1 Antagonists. ACS Med Chem Lett 2014; 5:193-8. [PMID: 24900800 DOI: 10.1021/ml4004556] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Accepted: 01/08/2014] [Indexed: 01/17/2023] Open
Abstract
Starting from clinical candidates Firategrast, Valategrast, and AJM-300, a series of novel macrocyclic platelet collagen receptor α2β1 antagonists were developed. The amino acid derived low molecular weight 14-18-membered macrocycles turned out to be highly active toward integrin α2β1 with IC50s in the low nanomolar range. The conformation of the macrocycles was found to be highly important for the activity, and an X-ray crystal structure was obtained to clarify this. Subsequent docking into the metal-ion-dependent adhesion site (MIDAS) of a β1 unit revealed a binding model indicating key binding features. Macrocycle 38 was selected for further in vitro and in vivo profiling.
Collapse
Affiliation(s)
- Nis Halland
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Horst Blum
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Christian Buning
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | - Markus Kohlmann
- Sanofi R&D, Industriepark Höchst Building G838, D-65926 Frankfurt am Main, Germany
| | | |
Collapse
|
48
|
Madamanchi A, Santoro SA, Zutter MM. α2β1 Integrin. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 819:41-60. [PMID: 25023166 DOI: 10.1007/978-94-017-9153-3_3] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The α2β1 integrin, also known as VLA-2, GPIa-IIa, CD49b, was first identified as an extracellular matrix receptor for collagens and/or laminins [55, 56]. It is now recognized that the α2β1 integrin serves as a receptor for many matrix and nonmatrix molecules [35, 79, 128]. Extensive analyses have clearly elucidated the α2 I domain structural motifs required for ligand binding, and also defined distinct conformations that lead to inactive, partially active or highly active ligand binding [3, 37, 66, 123, 136, 137, 140]. The mechanisms by which the α2β1 integrin plays a critical role in platelet function and homeostasis have been carefully defined via in vitro and in vivo experiments [76, 104, 117, 125]. Genetic and epidemiologic studies have confirmed human physiology and disease states mediated by this receptor in immunity, cancer, and development [6, 20, 21, 32, 43, 90]. The role of the α2β1 integrin in these multiple complex biologic processes will be discussed in the chapter.
Collapse
Affiliation(s)
- Aasakiran Madamanchi
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | |
Collapse
|
49
|
Brennan M, Cox D. The therapeutic potential of I-domain integrins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 819:157-78. [PMID: 25023174 DOI: 10.1007/978-94-017-9153-3_11] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Due to their role in processes central to cancer and autoimmune disease I-domain integrins are an attractive drug target. Both antibodies and small molecule antagonists have been discovered and tested in the clinic. Much of the effort has focused on αLβ2 antagonists. Maybe the most successful was the monoclonal antibody efalizumab, which was approved for the treatment of psoriasis but subsequently withdrawn from the market due to the occurrence of a serious adverse effect (progressive multifocal leukoencephalopathy). Other monoclonal antibodies were tested for the treatment of reperfusion injury, post-myocardial infarction, but failed to progress due to lack of efficacy. New potent small molecule inhibitors of αv integrins are promising reagents for treating fibrotic disease. Small molecule inhibitors targeting collagen-binding integrins have been discovered and future work will focus on identifying molecules selectively targeting each of the collagen receptors and identifying appropriate target diseases for future clinical studies.
Collapse
Affiliation(s)
- Marian Brennan
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | | |
Collapse
|
50
|
Affiliation(s)
- Matthew D. Linden
- The Centre for Microscopy; Characterisation and Analysis; The University of Western Australia; Nedlands WA 6009 Australia
| |
Collapse
|