1
|
Andrews SL, Ghaderi-Najafabadi M, Gong P, Shamkhi N, Carleton L, Schofield C, Kessler T, Samani NJ, Webb TR, Morris GE. SVEP1 influences monocyte to macrophage differentiation via integrin α4β1/α9β1 and Rho/Rac signalling. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119479. [PMID: 37100352 DOI: 10.1016/j.bbamcr.2023.119479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/09/2023] [Accepted: 04/15/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND The large extracellular matrix protein SVEP1 mediates cell adhesion via integrin α9β1. Recent studies have identified an association between a missense variant in SVEP1 and increased risk of coronary artery disease (CAD) in humans and in mice Svep1 deficiency alters the development of atherosclerotic plaques. However how SVEP1 functionally contributes to CAD pathogenesis is not fully understood. Monocyte recruitment and differentiation to macrophages is a key step in the development of atherosclerosis. Here, we investigated the requirement for SVEP1 in this process. METHODS SVEP1 expression was measured during monocyte-macrophage differentiation in primary monocytes and THP-1 human monocytic cells. SVEP1 knockout THP-1 cell lines and the dual integrin α4β1/α9β1 inhibitor, BOP, were utilised to investigate the effect of these proteins in THP-1 cell adhesion, migration and cell spreading assays. Subsequent activation of downstream integrin signalling intermediaries was quantified by western blotting. RESULTS SVEP1 gene expression increases in monocyte to macrophage differentiation in human primary monocytes and THP-1 cells. Using two SVEP1 knockout THP-1 cells we observed reduction in monocyte adhesion, migration, and cell spreading compared to control cells. Similar results were found with integrin α4β1/α9β1 inhibition. We demonstrate reduced activity of Rho and Rac1 in SVEP1 knockout THP-1 cells. CONCLUSIONS SVEP1 regulates monocyte recruitment and differentiation phenotypes through an integrin α4β1/α9β1 dependent mechanism. GENERAL SIGNIFICANCE These results describe a novel role for SVEP1 in monocyte behaviour relevant to CAD pathophysiology.
Collapse
Affiliation(s)
- S L Andrews
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - M Ghaderi-Najafabadi
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - P Gong
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - N Shamkhi
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - L Carleton
- Horizon Discovery Ltd., 8100 Cambridge Research Park, Cambridge CB25 9TL, United Kingdom.
| | - C Schofield
- Horizon Discovery Ltd., 8100 Cambridge Research Park, Cambridge CB25 9TL, United Kingdom.
| | - T Kessler
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Munich, Germany.; German Centre of Cardiovascular Research (DZHK e. V.), Partner Site Munich Heart Alliance, Munich, Germany.
| | - N J Samani
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - T R Webb
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| | - G E Morris
- Department of Cardiovascular Sciences, University of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester LE3 9QP, United Kingdom.
| |
Collapse
|
2
|
Boudko SP, Konopka EH, Kim W, Taga Y, Mizuno K, Springer TA, Hudson BG, Moy TI, Lin FY. A recombinant technique for mapping functional sites of heterotrimeric collagen helices: Collagen IV CB3 fragment as a prototype for integrin binding. J Biol Chem 2023; 299:104901. [PMID: 37302550 PMCID: PMC10404678 DOI: 10.1016/j.jbc.2023.104901] [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: 05/08/2023] [Revised: 06/01/2023] [Accepted: 06/05/2023] [Indexed: 06/13/2023] Open
Abstract
Collagen superfamily of proteins is a major component of the extracellular matrix. Defects in collagens underlie the cause of nearly 40 human genetic diseases in millions of people worldwide. Pathogenesis typically involves genetic alterations of the triple helix, a hallmark structural feature that bestows exceptional mechanical resistance to tensile forces and a capacity to bind a plethora of macromolecules. Yet, there is a paramount knowledge gap in understanding the functionality of distinct sites along the triple helix. Here, we present a recombinant technique to produce triple helical fragments for functional studies. The experimental strategy utilizes the unique capacity of the NC2 heterotrimerization domain of collagen IX to drive three α-chain selection and registering the triple helix stagger. For proof of principle, we produced and characterized long triple helical fragments of collagen IV that were expressed in a mammalian system. The heterotrimeric fragments encompassed the CB3 trimeric peptide of collagen IV, which harbors the binding motifs for α1β1 and α2β1 integrins. Fragments were characterized and shown to have a stable triple helix, post-translational modifications, and high affinity and specific binding of integrins. The NC2 technique is a universal tool for the high-yield production of heterotrimeric fragments of collagens. Fragments are suitable for mapping functional sites, determining coding sequences of binding sites, elucidating pathogenicity and pathogenic mechanisms of genetic mutations, and production of fragments for protein replacement therapy.
Collapse
Affiliation(s)
- Sergei P Boudko
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA.
| | | | - Woojin Kim
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA
| | - Yuki Taga
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, Japan
| | - Timothy A Springer
- Department of Biological Chemistry and Molecular Pharmacology, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Billy G Hudson
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Center for Matrix Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA; Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Terence I Moy
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA
| | - Fu-Yang Lin
- Morphic Therapeutic, Inc, Waltham, Massachusetts, USA.
| |
Collapse
|
3
|
Morris GE, Denniff MJ, Karamanavi E, Andrews SA, Kostogrys RB, Bountziouka V, Ghaderi‐Najafabadi M, Shamkhi N, McConnell G, Kaiser MA, Carleton L, Schofield C, Kessler T, Rainbow RD, Samani NJ, Webb TR. The integrin ligand SVEP1 regulates GPCR-mediated vasoconstriction via integrins α9β1 and α4β1. Br J Pharmacol 2022; 179:4958-4973. [PMID: 35802072 PMCID: PMC9805129 DOI: 10.1111/bph.15921] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 06/10/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Vascular tone is regulated by the relative contractile state of vascular smooth muscle cells (VSMCs). Several integrins directly modulate VSMC contraction by regulating calcium influx through L-type voltage-gated Ca2+ channels (VGCCs). Genetic variants in ITGA9, which encodes the α9 subunit of integrin α9β1, and SVEP1, a ligand for integrin α9β1, associate with elevated blood pressure; however, neither SVEP1 nor integrin α9β1 has reported roles in vasoregulation. We determined whether SVEP1 and integrin α9β1 can regulate VSMC contraction. EXPERIMENTAL APPROACH SVEP1 and integrin binding were confirmed by immunoprecipitation and cell binding assays. Human induced pluripotent stem cell-derived VSMCs were used in in vitro [Ca2+ ]i studies, and aortas from a Svep1+/- knockout mouse model were used in wire myography to measure vessel contraction. KEY RESULTS We confirmed the ligation of SVEP1 to integrin α9β1 and additionally found SVEP1 to directly bind to integrin α4β1. Inhibition of SVEP1, integrin α4β1 or α9β1 significantly enhanced [Ca2+ ]i levels in isolated VSMCs to Gαq/11 -vasoconstrictors. This response was confirmed in whole vessels where a greater contraction to U46619 was seen in vessels from Svep1+/- mice compared to littermate controls or when integrin α4β1 or α9β1 was inhibited. Inhibition studies suggested that this effect was mediated via VGCCs, PKC and Rho A/Rho kinase dependent mechanisms. CONCLUSIONS AND IMPLICATIONS Our studies reveal a novel role for SVEP1 and the integrins α4β1 and α9β1 in reducing VSMC contractility. This could provide an explanation for the genetic associations with blood pressure risk at the SVEP1 and ITGA9 loci.
Collapse
Affiliation(s)
- Gavin E. Morris
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Matthew J. Denniff
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Elisavet Karamanavi
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Sarah A. Andrews
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Renata B. Kostogrys
- Department of Human Nutrition, Faculty of Food TechnologyUniversity of Agriculture in KrakowKrakowPoland
| | - Vasiliki Bountziouka
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Maryam Ghaderi‐Najafabadi
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Noor Shamkhi
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - George McConnell
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Michael A. Kaiser
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | | | | | - Thorsten Kessler
- Department of Cardiology, German Heart Centre MunichTechnical University of MunichMunichGermany,German Centre of Cardiovascular Research (DZHK e. V.), Partner Site Munich Heart AllianceMunichGermany
| | - Richard D. Rainbow
- Department of Cardiovascular and Metabolic Medicine & Liverpool Centre for Cardiovascular ScienceUniversity of LiverpoolLiverpoolUK
| | - Nilesh J. Samani
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| | - Thomas R. Webb
- Department of Cardiovascular SciencesUniversity of Leicester and National Institute for Health Research Leicester Biomedical Research Centre, Glenfield HospitalLeicesterUK
| |
Collapse
|
4
|
Hight-Warburton W, Felix R, Burton A, Maple H, Chegkazi MS, Steiner RA, McGrath JA, Parsons M. α4/α9 Integrins Coordinate Epithelial Cell Migration Through Local Suppression of MAP Kinase Signaling Pathways. Front Cell Dev Biol 2021; 9:750771. [PMID: 34900996 PMCID: PMC8655878 DOI: 10.3389/fcell.2021.750771] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 10/31/2021] [Indexed: 11/18/2022] Open
Abstract
Adhesion of basal keratinocytes to the underlying extracellular matrix (ECM) plays a key role in the control of skin homeostasis and response to injury. Integrin receptors indirectly link the ECM to the cell cytoskeleton through large protein complexes called focal adhesions (FA). FA also function as intracellular biochemical signaling platforms to enable cells to respond to changing extracellular cues. The α4β1 and α9β1 integrins are both expressed in basal keratinocytes, share some common ECM ligands, and have been shown to promote wound healing in vitro and in vivo. However, their roles in maintaining epidermal homeostasis and relative contributions to pathological processes in the skin remain unclear. We found that α4β1 and α9β1 occupied distinct regions in monolayers of a basal keratinocyte cell line (NEB-1). During collective cell migration (CCM), α4 and α9 integrins co-localized along the leading edge. Pharmacological inhibition of α4β1 and α9β1 integrins increased keratinocyte proliferation and induced a dramatic change in cytoskeletal remodeling and FA rearrangement, detrimentally affecting CCM. Further analysis revealed that α4β1/α9β1 integrins suppress extracellular signal-regulated kinase (ERK1/2) activity to control migration through the regulation of downstream kinases including Mitogen and Stress Activated Kinase 1 (MSK1). This work demonstrates the roles of α4β1 and α9β1 in regulating migration in response to damage cues.
Collapse
Affiliation(s)
- Willow Hight-Warburton
- Parsons Group, Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | | | | | | | - Magda S Chegkazi
- Steiner Group, Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Roberto A Steiner
- Steiner Group, Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom.,Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - John A McGrath
- St Johns Institute of Dermatology, King's College London, London, United Kingdom
| | - Maddy Parsons
- Parsons Group, Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| |
Collapse
|
5
|
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: 1.2] [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
|
6
|
Baiula M, Spampinato S, Gentilucci L, Tolomelli A. Novel Ligands Targeting α 4β 1 Integrin: Therapeutic Applications and Perspectives. Front Chem 2019; 7:489. [PMID: 31338363 PMCID: PMC6629825 DOI: 10.3389/fchem.2019.00489] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 06/25/2019] [Indexed: 12/13/2022] Open
Abstract
Among the other members of the adhesion molecules' family, α4β1 integrin, a heterodimeric receptor, plays a crucial role in inflammatory diseases, cancer development, metastasis and stem cell mobilization or retention. In many cases, its function in pathogenesis is not yet completely understood and investigations on ligand binding and related stabilization of active/inactive conformations still represent an important goal. For this reason, starting from the highlight of α4β1 functions in human pathologies, we report an overview of synthetic α4β1 integrin ligands under development as potential therapeutic agents. The small molecule library that we have selected represents a collection of lead compounds. These molecules are the object of future refinement in academic and industrial research, in order to achieve a fine tuning of α4β1 integrin regulation for the development of novel agents against pathologies still eluding an effective solution.
Collapse
Affiliation(s)
- Monica Baiula
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Santi Spampinato
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Luca Gentilucci
- Department of Chemistry “G. Ciamician,” University of Bologna, Bologna, Italy
| | | |
Collapse
|
7
|
Domingues MJ, Cao H, Heazlewood SY, Cao B, Nilsson SK. Niche Extracellular Matrix Components and Their Influence on HSC. J Cell Biochem 2017; 118:1984-1993. [PMID: 28112429 DOI: 10.1002/jcb.25905] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 12/16/2022]
Abstract
Maintenance of hematopoietic stem cells (HSC) takes place in a highly specialized microenvironment within the bone marrow. Technological improvements, especially in the field of in vivo imaging, have helped unravel the complexity of the niche microenvironment and have completely changed the classical concept from what was previously believed to be a static supportive platform, to a dynamic microenvironment tightly regulating HSC homeostasis through the complex interplay between diverse cell types, secreted factors, extracellular matrix molecules, and the expression of different transmembrane receptors. To add to the complexity, non-protein based metabolites have also been recognized as a component of the bone marrow niche. The objective of this review is to discuss the current understanding on how the different extracellular matrix components of the niche regulate HSC fate, both during embryonic development and in adulthood. Special attention will be provided to the description of non-protein metabolites, such as lipids and metal ions, which contribute to the regulation of HSC behavior. J. Cell. Biochem. 118: 1984-1993, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Mélanie J Domingues
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Huimin Cao
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Shen Y Heazlewood
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Benjamin Cao
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| | - Susan K Nilsson
- Manufacturing, Commonwealth Scientific and Industrial Research Organization, Bag 10, Clayton South, VIC 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
8
|
Lu Z, Mathew S, Chen J, Hadziselimovic A, Palamuttam R, Hudson BG, Fässler R, Pozzi A, Sanders CR, Zent R. Implications of the differing roles of the β1 and β3 transmembrane and cytoplasmic domains for integrin function. eLife 2016; 5. [PMID: 27929375 PMCID: PMC5207772 DOI: 10.7554/elife.18633] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Accepted: 12/07/2016] [Indexed: 12/26/2022] Open
Abstract
Integrins are transmembrane receptors composed of α and β subunits. Although most integrins contain β1, canonical activation mechanisms are based on studies of the platelet integrin, αIIbβ3. Its inactive conformation is characterized by the association of the αIIb transmembrane and cytosolic domain (TM/CT) with a tilted β3 TM/CT that leads to activation when disrupted. We show significant structural differences between β1 and β3 TM/CT in bicelles. Moreover, the 'snorkeling' lysine at the TM/CT interface of β subunits, previously proposed to regulate αIIbβ3 activation by ion pairing with nearby lipids, plays opposite roles in β1 and β3 integrin function and in neither case is responsible for TM tilt. A range of affinities from almost no interaction to the relatively high avidity that characterizes αIIbβ3 is seen between various α subunits and β1 TM/CTs. The αIIbβ3-based canonical model for the roles of the TM/CT in integrin activation and function clearly does not extend to all mammalian integrins.
Collapse
Affiliation(s)
- Zhenwei Lu
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, United States
| | - Sijo Mathew
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, United States
| | - Jiang Chen
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, United States
| | - Arina Hadziselimovic
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, United States
| | - Riya Palamuttam
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, United States
| | - Billy G Hudson
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, United States.,Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, United States.,Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, United States
| | - Reinhard Fässler
- Department of Molecular Medicine, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Ambra Pozzi
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, United States.,Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, United States.,Veterans Affairs Hospital, Nashville, United States.,Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, United States
| | - Charles R Sanders
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, United States
| | - Roy Zent
- Division of Nephrology, Department of Medicine, Vanderbilt Medical Center, Nashville, United States.,Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, United States.,Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, United States.,Veterans Affairs Hospital, Nashville, United States
| |
Collapse
|
9
|
Cao B, Zhang Z, Grassinger J, Williams B, Heazlewood CK, Churches QI, James SA, Li S, Papayannopoulou T, Nilsson SK. Therapeutic targeting and rapid mobilization of endosteal HSC using a small molecule integrin antagonist. Nat Commun 2016; 7:11007. [PMID: 26975966 PMCID: PMC4796355 DOI: 10.1038/ncomms11007] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 02/10/2016] [Indexed: 12/24/2022] Open
Abstract
The inherent disadvantages of using granulocyte colony-stimulating factor (G-CSF) for hematopoietic stem cell (HSC) mobilization have driven efforts to identify alternate strategies based on single doses of small molecules. Here, we show targeting α9β1/α4β1 integrins with a single dose of a small molecule antagonist (BOP (N-(benzenesulfonyl)-L-prolyl-L-O-(1-pyrrolidinylcarbonyl)tyrosine)) rapidly mobilizes long-term multi-lineage reconstituting HSC. Synergistic engraftment augmentation is observed when BOP is co-administered with AMD3100. Impressively, HSC in equal volumes of peripheral blood (PB) mobilized with this combination effectively out-competes PB mobilized with G-CSF. The enhanced mobilization observed using BOP and AMD3100 is recapitulated in a humanized NODSCIDIL2Rγ−/− model, demonstrated by a significant increase in PB CD34+ cells. Using a related fluorescent analogue of BOP (R-BC154), we show that this class of antagonists preferentially bind human and mouse HSC and progenitors via endogenously primed/activated α9β1/α4β1 within the endosteal niche. These results support using dual α9β1/α4β1 inhibitors as effective, rapid and transient mobilization agents with promising clinical applications. Mobilizing haematopoietic stem cells to the peripheral blood has largely replaced bone marrow transplants as a strategy in the clinic. Here, Cao et al. report the use of an α9β1/α4β1 integrin antagonist to induce rapid mobilization of blood stem cells from the bone marrow in a humanized mouse model.
Collapse
Affiliation(s)
- Benjamin Cao
- Biomedical Manufacturing, CSIRO Manufacturing, Bag 10, Clayton South, Victoria 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Zhen Zhang
- Biomedical Manufacturing, CSIRO Manufacturing, Bag 10, Clayton South, Victoria 3169, Australia
| | - Jochen Grassinger
- University Hospital Regensberg, Department of Hematology and Oncology, Franz-Josef-Strauß-Allee 11, Regensburg 93053, Germany
| | - Brenda Williams
- Biomedical Manufacturing, CSIRO Manufacturing, Bag 10, Clayton South, Victoria 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Chad K Heazlewood
- Biomedical Manufacturing, CSIRO Manufacturing, Bag 10, Clayton South, Victoria 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Quentin I Churches
- Biomedical Manufacturing, CSIRO Manufacturing, Bag 10, Clayton South, Victoria 3169, Australia
| | - Simon A James
- Biomedical Manufacturing, CSIRO Manufacturing, Bag 10, Clayton South, Victoria 3169, Australia.,Australian Synchrotron, Clayton, Victoria 3168, Australia
| | - Songhui Li
- Biomedical Manufacturing, CSIRO Manufacturing, Bag 10, Clayton South, Victoria 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| | - Thalia Papayannopoulou
- Department of Medicine/Hematology, University of Washington Seattle, 1705 NE Pacific, Box 357710, Seattle, Washington 98195-7710, USA
| | - Susan K Nilsson
- Biomedical Manufacturing, CSIRO Manufacturing, Bag 10, Clayton South, Victoria 3169, Australia.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
10
|
Kanemaru K, Noguchi E, Tokunaga T, Nagai K, Hiroyama T, Nakamura Y, Tahara-Hanaoka S, Shibuya A. Tie2 Signaling Enhances Mast Cell Progenitor Adhesion to Vascular Cell Adhesion Molecule-1 (VCAM-1) through α4β1 Integrin. PLoS One 2015; 10:e0144436. [PMID: 26659448 PMCID: PMC4687632 DOI: 10.1371/journal.pone.0144436] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 11/18/2015] [Indexed: 12/21/2022] Open
Abstract
Mast cell (MC) activation contributes considerably to immune responses, such as host protection and allergy. Cell surface immunoreceptors expressed on MCs play an important role in MC activation. Although various immunoreceptors on MCs have been identified, the regulatory mechanism of MC activation is not fully understood. To understand the regulatory mechanisms of MC activation, we used gene expression analyses of human and mouse MCs to identify a novel immunoreceptor expressed on MCs. We found that Tek, which encodes Tie2, was preferentially expressed in the MCs of both humans and mice. However, Tie2 was not detected on the cell surface of the mouse MCs of the peritoneal cavity, ear skin, or colon lamina propria. In contrast, it was expressed on mouse bone marrow–derived MCs and bone marrow MC progenitors (BM-MCps). Stimulation of Tie2 by its ligand angiopoietin-1 induced tyrosine phosphorylation of Tie2 in MEDMC-BRC6, a mouse embryonic stem cell-derived mast cell line, and enhanced MEDMC-BRC6 and mouse BM-MCp adhesion to vascular cell adhesion molecule-1 (VCAM-1) through α4β1 integrin. These results suggest that Tie2 signaling induces α4β1 integrin activation on BM-MCps for adhesion to VCAM-1.
Collapse
Affiliation(s)
- Kazumasa Kanemaru
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Emiko Noguchi
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Takahiro Tokunaga
- Department of Medical Genetics, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Otorhinolaryngology Head and Neck Surgery, University of Fukui, Fukui, Japan
| | - Kei Nagai
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takashi Hiroyama
- Cell Engineering Division, RIKEN BioResource Center, Kounodai, Tsukuba, Ibaraki, Japan
| | - Yukio Nakamura
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Cell Engineering Division, RIKEN BioResource Center, Kounodai, Tsukuba, Ibaraki, Japan
| | - Satoko Tahara-Hanaoka
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center of Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
| | - Akira Shibuya
- Department of Immunology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Life Science Center of Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
- * E-mail:
| |
Collapse
|
11
|
Yang Y, Enis D, Zheng H, Chia S, Yang J, Chen M, Dhillon V, Papayannapoulou T, Kahn ML. Cell Adhesion Mediated by VCAM-ITGα9 Interactions Enables Lymphatic Development. Arterioscler Thromb Vasc Biol 2015; 35:1179-89. [PMID: 25745057 DOI: 10.1161/atvbaha.114.304997] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 02/22/2015] [Indexed: 01/16/2023]
Abstract
OBJECTIVE Adhesive ligand-receptor interactions play key roles in blood vessel angiogenesis but remain poorly characterized during lymphatic vessel growth. In this study, we use genetic approaches in both fish and mice to address the roles of cell surface integrin ligand vascular cell adhesion molecule (VCAM) and its 2 receptors, integrins α9 and α4, during lymphatic vascular development. APPROACH AND RESULTS Conditional deletion of the Vcam gene was used to test VCAM function in lymphatic growth in midgestation mice. Morpholino knockdown and cRNA rescue of the 2 zebrafish vcam alleles, as well as integrins α9 and 4, were used to test the role of these ligands and receptors during lymphatic growth in the developing fish. We show that VCAM is essential for lymphatic development in the zebrafish embryo and that integrin α9 (Itgα9) rather than Itgα4 is the required VCAM receptor in the developing fish. VCAM is expressed along lines of lymphatic migration in the mouse intestine, but its loss only retards lymphatic growth. CONCLUSIONS These studies reveal an unexpected role for cell-cell adhesion mediated by Itgα9-VCAM interactions during lymphatic development in the fish but not in the mouse. We propose that the relative importance of cellular adhesive ligands is magnified under conditions of rapid tissue growth when the cell number increases faster than cell matrix, such as in the early zebrafish embryo.
Collapse
Affiliation(s)
- Yiqing Yang
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - David Enis
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Hui Zheng
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Stephanie Chia
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Jisheng Yang
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Mei Chen
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Veerpal Dhillon
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Thalia Papayannapoulou
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.)
| | - Mark L Kahn
- From the Department of Medicine and Cardiovascular Institute (Y.Y., D.E., H.Z., S.C., J.Y., M.C., V.D., M.L.K.) and Department of Dermatology (D.E.), University of Pennsylvania, Philadelphia; and Department of Medicine, University of Washington, Seattle (T.P.).
| |
Collapse
|
12
|
Osteopontin binding to the alpha 4 integrin requires highest affinity integrin conformation, but is independent of post-translational modifications of osteopontin. Matrix Biol 2014; 41:19-25. [PMID: 25446551 DOI: 10.1016/j.matbio.2014.11.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/20/2014] [Accepted: 11/22/2014] [Indexed: 12/17/2022]
Abstract
Osteopontin (OPN) is a ligand for the α4ß1 integrin, but the physiological importance of this binding is not well understood. Here, we have assessed the effect of post-translational modifications on OPN binding to the α4 integrin on cultured human leukocyte cell lines and compared OPN interaction with α4 integrin to that of VCAM and fibronectin. Jurkat cells, whose α4 integrins are inherently activated, adhered to different preparations of OPN in the presence of Mn(2+): the EC50 of adhesion was not affected by phosphorylation or glycosylation status. Thrombin cleavage of OPN at the C-terminus of the α4 integrin-binding site also did not affect binding affinity. THP-1 cells express a low-affinity conformation of the integrin and adhered to OPN only in the presence of Mn(2+) plus PMA or an activating antibody. This was in contrast to VCAM and fibronectin: THP-1 cells adhered to these ligands without integrin activation. Studies with ligand-induced binding site antibodies demonstrated that the SVVYGLR peptide of OPN bound to the α4 integrin with a similar affinity as the LDV peptide of fibronectin, suggesting that a high off-rate is responsible for the reduced binding of OPN to the low-affinity forms of this integrin. Together, the results suggest OPN has very low affinity for the α4 integrin on human leukocytes under physiological conditions.
Collapse
|
13
|
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: 0.9] [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
|
14
|
Abstract
Sphingosine 1-phosphate (S1P) plays a role in lymphocyte egress from lymphoid organs. However, it remains unclear how S1P production and secretion are regulated. We show that under inflammatory conditions, α9 integrin, which is closely associated with activated β1 integrin, and its ligand, tenascin-C, colocalize on medullary and cortical sinuses of draining lymph nodes (dLNs), which is a gate for lymphocyte exit, and that inhibition of lymphocyte egress is evident by blockade of α9 integrin-mediated signaling at dLNs. Based on in vitro analysis using lymphatic endothelial cells obtained from mice embryos, we suggested the possibility that stimulation of lymphatic endothelial cells by tenascin-C enhances S1P secretion in an α9 integrin-dependent manner without affecting S1P synthesis and/or degradation. Blockade of α9 integrin-mediated signaling reduced lymphocyte egress from dLNs in several models, including experimental autoimmune encephalomyelitis, where it improved clinical scores and pathology. Therefore, manipulating α9 integrin function may offer a therapeutic strategy for treating various inflammatory disorders.
Collapse
|
15
|
Vanderslice P, Biediger RJ, Woodside DG, Brown WS, Khounlo S, Warier ND, Gundlach CW, Caivano AR, Bornmann WG, Maxwell DS, McIntyre BW, Willerson JT, Dixon RAF. Small molecule agonist of very late antigen-4 (VLA-4) integrin induces progenitor cell adhesion. J Biol Chem 2013; 288:19414-28. [PMID: 23703610 DOI: 10.1074/jbc.m113.479634] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Activation of the integrin family of cell adhesion receptors on progenitor cells may be a viable approach to enhance the effects of stem cell-based therapies by improving cell retention and engraftment. Here, we describe the synthesis and characterization of the first small molecule agonist identified for the integrin α4β1 (also known as very late antigen-4 or VLA-4). The agonist, THI0019, was generated via two structural modifications to a previously identified α4β1 antagonist. THI0019 greatly enhanced the adhesion of cultured cell lines and primary progenitor cells to α4β1 ligands VCAM-1 and CS1 under both static and flow conditions. Furthermore, THI0019 facilitated the rolling and spreading of cells on VCAM-1 and the migration of cells toward SDF-1α. Molecular modeling predicted that the compound binds at the α/β subunit interface overlapping the ligand-binding site thus indicating that the compound must be displaced upon ligand binding. In support of this model, an analog of THI0019 modified to contain a photoreactive group was used to demonstrate that when cross-linked to the integrin, the compound behaves as an antagonist instead of an agonist. In addition, THI0019 showed cross-reactivity with the related integrin α4β7 as well as α5β1 and αLβ2. When cross-linked to αLβ2, the photoreactive analog of THI0019 remained an agonist, consistent with it binding at the α/β subunit interface and not at the ligand-binding site in the inserted ("I") domain of the αL subunit. Co-administering progenitor cells with a compound such as THI0019 may provide a mechanism for enhancing stem cell therapy.
Collapse
Affiliation(s)
- Peter Vanderslice
- Department of Molecular Cardiology, Texas Heart Institute at St. Luke's Episcopal Hospital, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kohri K, Yasui T, Okada A, Hirose M, Hamamoto S, Fujii Y, Niimi K, Taguchi K. Biomolecular mechanism of urinary stone formation involving osteopontin. ACTA ACUST UNITED AC 2012; 40:623-37. [PMID: 23124115 DOI: 10.1007/s00240-012-0514-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 09/13/2012] [Indexed: 12/15/2022]
Abstract
Urinary stones consist of two phases-an inorganic (mineral) phase and an organic (matrix) phase. Studies on the organic components of kidney stones have been undertaken later than those on the inorganic components. After osteopontin was identified as one of the matrix components, the biomolecular mechanism of urinary stone formation became clearer. It also triggered the development of new preventive treatments. Osteopontin expression is sporadically observed in normal distal tubular cells and is markedly increased in stone-forming kidneys. Calcium oxalate crystals adhering to renal tubular cells are incorporated into cells by the involvement of osteopontin. Stimulation of crystal-cell adhesion impairs the opening of mitochondrial permeability transition pores (mPTP) in tubular cells and produces oxidative stress, apoptosis, and osteopontin expression. Macrophages phagocytose and digest a small amount of crystals, but many crystals aggregate into a mass containing osteopontin and epithelial cell debris and are excreted into the renal tubular lumen, becoming nuclei of urinary stones. This biomolecular mechanism is similar to atherosclerotic calcification. Based on these findings, new preventive treatments have been developed. Dietary control such as low-cholesterol intake and the ingestion of antioxidative foods and vegetables have successfully reduced the 5-year recurrence rate. Osteopontin antibodies and cyclosporine A, which blocks the opening of mPTP, have markedly inhibited the expression of osteopontin and urinary stone formation in animal models.
Collapse
Affiliation(s)
- Kenjiro Kohri
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Wallace M, Allentoff AJ, Bonacorsi S. Synthesis of acyl[35S]sulfonamides: Coupling of high specific activity [35S]methane sulfonamide with acids and acid chlorides. J Labelled Comp Radiopharm 2012. [DOI: 10.1002/jlcr.2974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Michael Wallace
- Radiochemistry; Bristol-Myers Squibb Company; P.O. Box 4000; Princeton; NJ; 08543; USA
| | - Alban J. Allentoff
- Radiochemistry; Bristol-Myers Squibb Company; P.O. Box 4000; Princeton; NJ; 08543; USA
| | - Samuel Bonacorsi
- Radiochemistry; Bristol-Myers Squibb Company; P.O. Box 4000; Princeton; NJ; 08543; USA
| |
Collapse
|
18
|
Application of LC/MS to determine specific activity variation in a series of sulfonamide tracers from the [35S]methane sulfonate reagent. J Labelled Comp Radiopharm 2012. [DOI: 10.1002/jlcr.2976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
19
|
Hamamoto S, Yasui T, Okada A, Hirose M, Matsui Y, Kon S, Sakai F, Kojima Y, Hayashi Y, Tozawa K, Uede T, Kohri K. Crucial role of the cryptic epitope SLAYGLR within osteopontin in renal crystal formation of mice. J Bone Miner Res 2011; 26:2967-77. [PMID: 21898593 DOI: 10.1002/jbmr.495] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Osteopontin plays a crucial role in the formation of renal calcium crystals, which are primarily induced by renal tubular cell injury, especially mitochondrial damage. We have previously shown that the impaired Arg-Gly-Asp (RGD) sequence of osteopontin inhibits renal crystal formation by using OPN-transgenic mice and OPN-knockout (OPN-KO) mice. Here, we investigated the effects of an antimurine osteopontin antibody (35B6-Ab) that specifically reacts with the (162) SLAYGLR(168) sequence, which is exposed by thrombin cleavage and is located adjacent to the RGD sequence, on renal crystal formation. Renal crystals induced by daily administration of glyoxylate over 9 days (from days 1 to 9) in a murine model were sporadically detected in the renal tubular cells at the corticomedullary junction, where thrombin-cleaved osteopontin expression was also coincidentally detected. On days 0, 3, 6, and 9, 35B6-Ab administration inhibited renal crystal formation and induced significant morphological changes in a dose-dependent manner (250, 500, and 1000 µg per mouse). Scanning electron microscopy showed that the crystals in 35B6-Ab-treated mice were aberrantly formed and their density was low; in contrast, the crystals in untreated mice that were not administered 35B6-Ab had a radial pattern of growth (rosette petal-like crystals), and their density was high. Microstructure analysis of renal tubular cells by transmission electron microscopy revealed that untreated mice showed collapsed mitochondria in the flattened cytoplasm of renal tubular cells, unlike the corresponding structures in 35B6-Ab-treated mice, in which renal tubular cell injury was inhibited. In vitro, 35B6-Ab was found to inhibit the attachment of (14) C-labeled crystals to renal tubular culture cells and reduce morphological damage to these cells. We conclude that thrombin-cleaved osteopontin plays an important role in formation of renal calcium crystals and that 35B6-Ab contributes to the remarkable inhibition of early-stage renal crystal formation by preventing renal tubular cell injury and crystal-cell attachment.
Collapse
Affiliation(s)
- Shuzo Hamamoto
- Department of Nephro-urology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Lomakina EB, Waugh RE. Adhesion between human neutrophils and immobilized endothelial ligand vascular cell adhesion molecule 1: divalent ion effects. Biophys J 2010; 96:276-84. [PMID: 19134480 DOI: 10.1016/j.bpj.2008.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Accepted: 10/09/2008] [Indexed: 10/21/2022] Open
Abstract
Integrin-mediated adhesion of circulating neutrophils to endothelium during inflammation involves multiple adhesion molecules on both neutrophils and endothelium. Most studies of neutrophil adhesion have focused on adhesion to ICAM-1 (mediated by beta(2) integrins), but interaction with the endothelial ligand vascular cell adhesion molecule 1 (VCAM-1) may also play a role in neutrophil adhesion to activated endothelium. In this study we demonstrate significant adhesion between neutrophils and VCAM-1 mediated by beta(1) integrins, principally via alpha(4)beta(1) (VLA-4). We characterize the dynamics of adhesion in terms of rate constants for a two-step bond formation process, the first involving juxtaposition of active molecules with substrate and the second involving bond formation. The results indicate that the first step is rate limiting for VLA-4-VCAM-1 interactions. Changing divalent cation composition affects these coefficients, implicating molecular conformational changes as a key step in the process.
Collapse
Affiliation(s)
- Elena B Lomakina
- Department of Pharmacology and Physiology, University of Rochester, Medical Center, Rochester, New York, USA
| | | |
Collapse
|
21
|
Abstract
Two thirds of patients with multiple sclerosis have the relapsing-remitting form, which often progresses to more debilitating disease. Striking clinical recovery, termed remission, often follows these periodic neurological defects, termed relapses. Recent work has revealed the role of three key molecules in relapse and remission: alpha4beta1 integrin (also known as VLA4) is an adhesion molecule that mediates T cell migration from the blood into the brain; osteopontin binds to alpha4beta1 integrin, stimulating the production of pro-inflammatory cytokines and inhibiting apoptosis; and alphaB crystallin inhibits inflammation in the brain. This Review discusses how this molecular trio interacts to initiate relapses (in the case of osteopontin and alpha4beta1 integrin) and then to terminate them as remissions in multiple sclerosis (in the case of alphaB crystallin).
Collapse
Affiliation(s)
- Lawrence Steinman
- Department of Neurology and Neurological Sciences, Interdepartmental Program in Immunology, Beckman Center for Molecular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA.
| |
Collapse
|
22
|
Treonze KM, Alves K, Fischer P, Hagmann WK, Hora D, Kulick A, Vakerich K, Smith ND, Lingham RB, Maniar S, Reger TS, Zunic J, Munoz B, Prasit P, Nicholson D, Si Q, Judd K, Nicolich S, Kellerhouse P, Thompson D, Mumford RA. Characterization of alpha(4)beta(1) (CD49d/CD29) on equine leukocytes: potential utility of a potent alpha(4)beta(1) (CD49d/CD29) receptor antagonist in the treatment of equine heaves (recurrent airway obstruction). Vet Immunol Immunopathol 2009; 130:79-87. [PMID: 19250687 DOI: 10.1016/j.vetimm.2009.01.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2008] [Revised: 01/15/2009] [Accepted: 01/26/2009] [Indexed: 11/28/2022]
Abstract
The purpose of this study was to characterize the alpha(4)beta(1) receptor (CD49d/CD29, very late antigen-4, VLA-4) on circulating equine leukocytes and to evaluate the intrinsic potency of an alpha(4)beta(1) receptor antagonist (Compound B) in the horse. Ultimately, these studies would allow us to determine the suitability of treating recurrent airway obstruction (RAO; heaves) affected horses by blocking the cellular recruitment of lymphocytes and neutrophils into the lung. The data demonstrates the alpha(4)beta(1) integrin is present on horse lymphocytes and neutrophils (fluorescence-assisted cell sorter, FACS) and can bind low molecular weight alpha(4)beta(1) antagonists (Compounds A and B) with high affinity. K(D) values for the binding of Compound A to non-activated alpha(4)beta(1) on isolated horse PBMCs (peripheral blood mononuclear cells) and activated neutrophils were 17 pM and 27 pM, respectively. Compound B was identified as a suitable antagonist for performing a series of in vivo experiments. Compound B was found to possess excellent potency in horse whole blood, possessing IC(50) and IC(90) values of 39 pM and 172 pM, respectively. This represents a 3.9-fold molar excess of drug over the alpha(4)beta(1) concentration in blood. Following oral administration of Compound B (5 mg/kg) to beagle dogs and rhesus monkeys, rapid and sustained alpha(4)beta(1) receptor occupancy (>80%) was achieved and maintained for a period of 24 h. When Compound B was administered intravenously to the horse, by either a slow or rapid infusion at a dose of 0.3 mg/kg, receptor blockade of >80% was observed out to 24 h with a concomitant leukocytosis. We believe that Compound B possesses suitable intrinsic and pharmacological properties to be evaluated clinically in horses affected by RAO.
Collapse
Affiliation(s)
- Kelly M Treonze
- Department of Immunology, Merck Research Laboratories, PO Box 2000, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Staniszewska I, Zaveri S, Del Valle L, Oliva I, Rothman VL, Croul SE, Roberts DD, Mosher DF, Tuszynski GP, Marcinkiewicz C. Interaction of alpha9beta1 integrin with thrombospondin-1 promotes angiogenesis. Circ Res 2007; 100:1308-16. [PMID: 17413041 DOI: 10.1161/01.res.0000266662.98355.66] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Thrombospondin-1 is a multifunctional protein interacting with several cell surface receptors including integrins. We found that it is a ligand for alpha9beta1 integrin, and has an integrin binding site within its N-terminal domain (NoC1). Interaction of thrombospondin-1 and its recombinant NoC1 domain with alpha9beta1 integrin was confirmed in ELISA and cell adhesion assays. Binding of NoC1 to cells expressing alpha9beta1 integrin activated signaling proteins such as Erk1/2 and paxillin. Blocking of this integrin by monoclonal antibody and the met-leu-asp-disintegrin inhibited dermal human microvascular endothelial cell proliferation and NoC1-induced migration of these cells. Immunohistochemical studies revealed that alpha9beta1 is expressed on microvascular endothelium in several organs including skin, lung, heart and brain. NoC1 induced neovascularization in an experimental quail chorioallantoic membrane system and Matrigel plug formation assay in mice. This proangiogenic activity of NoC1 in vivo was inhibited by alpha9beta1 inhibitors. In summary, our results revealed that alpha9beta1 integrin expressed on microvascular endothelial cells interacts with thrombospondin-1, and this interaction is involved in modulation of angiogenesis.
Collapse
Affiliation(s)
- Izabela Staniszewska
- Department of Neuroscience, Center for Neurovirology, Temple University, School of Medicine, Philadelphia, PA 19122, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Wallace MA, Raab C, Dean D, Melillo D. Synthesis of aryl [35S]sulfones: Friedel–Crafts sulfonylation of aryl ethers with high specific activity [35S]methanesulfonyl chloride. J Labelled Comp Radiopharm 2007. [DOI: 10.1002/jlcr.1300] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
25
|
Priestley GV, Scott LM, Ulyanova T, Papayannopoulou T. Lack of alpha4 integrin expression in stem cells restricts competitive function and self-renewal activity. Blood 2005; 107:2959-67. [PMID: 16357327 PMCID: PMC1895392 DOI: 10.1182/blood-2005-07-2670] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Alpha4 integrin or VLA4 (CD49d/CD29) is a multitask molecule with wide expression within and outside the hematopoietic system. Because targeted ablation of alpha4 integrin leads to embryonic lethality, to study its effects on adult hematopoiesis, we used animals with conditional excision of alpha4 integrin (alpha4Delta/Delta) in hematopoietic cells. In such animals, we previously documented weakened bone marrow retention of progenitor cells during homeostasis and impaired homing and short-term engraftment after transplantation. In the present study we show that long-term repopulating cells lacking alpha4 integrins display a competitive disadvantage in hematopoietic reconstitution compared to normal competitors. Although initial dominance of alpha4+ competitors is due to their better homing and proliferative expansion early after transplantation, a progressive decline in contribution of alpha4Delta/Delta hematopoiesis is compatible with neither normal homing nor normal function of alpha4Delta/Delta hematopoietic stem cells (HSCs) in post-homing hematopoiesis. In the absence of alpha4+ competitor cells, alpha4Delta/Delta HSCs can establish long-term hematopoiesis in primary recipients, however, some resurgence of host hematopoiesis is evident, and it becomes dominant in secondary transplants, so that no survivors with exclusively alpha4Delta/Delta cells are seen in tertiary transplants. Collectively, our data provide compelling evidence that under regenerative stress alpha4 integrin assumes a greater importance than for maintenance of steady-state hematopoiesis.
Collapse
Affiliation(s)
- Gregory V Priestley
- University of Washington, Department of Medicine/Division of Hematology, Box 357710, Seattle, WA 98195-7710.
| | | | | | | |
Collapse
|
26
|
Pepinsky RB, Lee WC, Cornebise M, Gill A, Wortham K, Chen LL, Leone DR, Giza K, Dolinski BM, Perper S, Nickerson-Nutter C, Lepage D, Chakraborty A, Whalley ET, Petter RC, Adams SP, Lobb RR, Scott DM. Design, synthesis, and analysis of a polyethelene glycol-modified (PEGylated) small molecule inhibitor of integrin {alpha}4{beta}1 with improved pharmaceutical properties. J Pharmacol Exp Ther 2005; 312:742-50. [PMID: 15485895 DOI: 10.1124/jpet.104.075648] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Integrin alpha4beta1 plays an important role in inflammatory processes by regulating the migration of leukocytes into inflamed tissues. Previously, we identified BIO5192 [2(S)-{[1-(3,5-dichloro-benzenesulfonyl)-pyrrolidine-2(S)-carbonyl]-amino}-4-[4-methyl-2(S)-(methyl-{2-[4-(3-o-tolyl-ureido)-phenyl]-acetyl}-amino)-pentanoylamino]-butyric acid], a highly selective and potent (K(D) of 9 pM) small molecule inhibitor of alpha4beta1. Although BIO5192 is efficacious in various animal models of inflammatory disease, high doses and daily treatment of the compound are needed to achieve a therapeutic effect because of its relatively short serum half-life. To address this issue, polyethylene glycol modification (PEGylation) was used as an approach to improve systemic exposure. BIO5192 was PEGylated by a targeted approach in which derivatizable amino groups were incorporated into the molecule. Two sites were identified that could be modified, and from these, five PEGylated compounds were synthesized and characterized. One compound, 2a-PEG (K(D) of 19 pM), was selected for in vivo studies. The pharmacokinetic and pharmacodynamic properties of 2a-PEG were dramatically improved relative to the unmodified compound. The PEGylated compound was efficacious in a rat model of experimental autoimmune encephalomyelitis at a 30-fold lower molar dose than the parent compound and required only a once-a-week dosing regimen compared with a daily treatment for BIO5192. Compound 2a-PEG was highly selective for alpha4beta1. These studies demonstrate the feasibility of PEGylation of alpha4beta1-targeted small molecules with retention of activity in vitro and in vivo. 2a-PEG, and related compounds, will be valuable reagents for assessing alpha4beta1 biology and may provide a new therapeutic approach to treatment of human inflammatory diseases.
Collapse
Affiliation(s)
- R B Pepinsky
- Biogen Idec, Inc., 14 Cambridge Center, Cambridge, MA 02142, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wallace MA, Raab CE, Dean DC, Melillo DG. Synthesis of [35S]aryl sulfonyl chlorides from [35S]elemental sulfur. J Labelled Comp Radiopharm 2005. [DOI: 10.1002/jlcr.920] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
28
|
Ajroud K, Sugimori T, Goldmann WH, Fathallah DM, Xiong JP, Arnaout MA. Binding Affinity of Metal Ions to the CD11b A-domain Is Regulated by Integrin Activation and Ligands. J Biol Chem 2004; 279:25483-8. [PMID: 15070893 DOI: 10.1074/jbc.m402901200] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The divalent cations Mg(2+) and Ca(2+) regulate the interaction of integrins with their cognate ligands, with Mg(2+) uniformly facilitating and Ca(2+) generally inhibiting such interactions in vitro. Because both cations are present in mm concentrations in vivo, the physiologic relevance of the in vitro observations is unclear. We measured the affinity of both cations to the inactive and active states of the ligand- and cation-binding A-domain (CD11bA) from integrin CD11b/CD18 in the absence and presence of the single-chain 107 antibody (scFv107), an activation-insensitive ligand-mimetic antibody. Using titration calorimetry, we found that Mg(2+) and Ca(2+) display equivalent (mm) affinities to inactive CD11bA. Activation induced a approximately 10-fold increase in the binding affinity of Mg(2+) to CD11bA with no change in that of Ca(2+) (106 microm +/- 16 and 2.1 mm +/- 0.19, respectively, n = 4). This increase is largely driven by favorable enthalpy. scFv107 induced a 50-80-fold increase in the binding affinity of Ca(2+) (but not Mg(2+) or Mn(2+)) to either form of CD11bA. Thus the affinity of metal ions to integrins is itself regulated by the activation state of these receptors and by certain ligands. These findings, which we expect will be applicable in vivo, elucidate a new level of regulation of the integrin-metal-ligand ternary complex and help explain some of the discrepant effects of Ca(2+) on integrin-ligand interactions.
Collapse
Affiliation(s)
- Kaouther Ajroud
- Leukocyte Biology and Inflammation Program, Renal Unit, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | |
Collapse
|
29
|
Higashibata Y, Sakuma T, Kawahata H, Fujihara S, Moriyama K, Okada A, Yasui T, Kohri K, Kitamura Y, Nomura S. Identification of promoter regions involved in cell- and developmental stage-specific osteopontin expression in bone, kidney, placenta, and mammary gland: an analysis of transgenic mice. J Bone Miner Res 2004; 19:78-88. [PMID: 14753740 DOI: 10.1359/jbmr.2004.19.1.78] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
UNLABELLED Cell-specific expression of GFP under the control of different lengths of the osteopontin promoter in transgenic mice identified the positive and negative regulatory regions for respective cell types. The results provide new insights for physiological and pathological expression of the osteopontin gene. INTRODUCTION Osteopontin (OPN) is a major non-collagenous bone matrix protein that is involved in normal and pathological calcification and is expressed in a tissue-specific manner. To investigate how such tissue-specific OPN gene expression is regulated in vivo, transgenic mice expressing the green fluorescent protein (GFP) reporter gene controlled by different lengths of the OPN promoter were generated. MATERIALS AND METHODS Cell- and developmental stage-specific osteopontin expression in transgenic mice was examined by Northern blotting, immunoblotting, fluorescence examination, and in situ hybridization and compared with those of OPN. RESULTS AND CONCLUSIONS The line bearing the -5505 to +14 region of the OPN promoter was shown by Northern blotting and immunoblotting to express GFP in the same cells that express endogenous OPN (osteoblasts, hypertrophic chondrocytes, renal and mammary gland epithelial cells, and granulated metrial gland [GMG] placental cells) at the same stage in development. Thus, the 5.5-kb -5505 to +14 promoter region is sufficient for proper tissue-specific OPN expression. The lines carrying shorter segments of the OPN promoter showed different expression patterns. These patterns revealed a putative cis-acting element in the -5269 to -5263 region that restricts OPN expression to hypertrophic chondrocytes and a mammary gland-specific expressing element and a GMG cell-specific enhancing element in the -5505 to -3156 region. Furthermore, the -3155 to -1576 region seems to contain positive renal epithelial cell- and GMG cell-specific expression motif(s) as well as a negative regulatory element that prevents OPN expression in fibroblasts. Moreover, the -1576 to -910 region seems to contain a positive osteoblast-specific-expressing element. Thus, the 5.5-kb OPN promoter contains multiple cis-acting elements encoding positive and negative cell-specific regulatory systems.
Collapse
MESH Headings
- Animals
- Blotting, Northern
- Blotting, Southern
- Blotting, Western
- Bone and Bones/chemistry
- Bone and Bones/cytology
- Bone and Bones/metabolism
- Cartilage/chemistry
- Cartilage/cytology
- Cartilage/metabolism
- Crosses, Genetic
- Female
- Gene Dosage
- Gene Expression
- Gene Expression Regulation, Developmental
- Green Fluorescent Proteins
- Immunohistochemistry
- In Situ Hybridization
- Kidney/chemistry
- Kidney/cytology
- Kidney/metabolism
- Leg Bones/chemistry
- Leg Bones/cytology
- Leg Bones/metabolism
- Luminescent Proteins/analysis
- Luminescent Proteins/genetics
- Mammary Glands, Animal/chemistry
- Mammary Glands, Animal/cytology
- Mammary Glands, Animal/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Transgenic
- Models, Genetic
- Osteopontin
- Placenta/chemistry
- Placenta/cytology
- Placenta/metabolism
- Pregnancy
- Promoter Regions, Genetic/genetics
- Recombinant Fusion Proteins/analysis
- Recombinant Fusion Proteins/genetics
- Regulatory Sequences, Nucleic Acid/genetics
- Sialoglycoproteins/genetics
- Skin/chemistry
- Skin/metabolism
- Spine/chemistry
- Spine/metabolism
Collapse
Affiliation(s)
- Yuji Higashibata
- Department of Pathology, Graduate School of Osaka University Medical School, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Egger LA, Cao J, McCallum C, Kidambi U, Van Riper G, McCauley E, Mumford RA, Lanza TJ, Lin LS, de Laszlo SE, Young DN, Yang G, Dean DC, Raab CE, Wallace MA, Jones AN, Hagmann WK, Schmidt JA, Pepinsky RB, Scott DM, Lee WC, Cornebise MA, Detmers PA. A small molecule alpha4beta1/alpha4beta7 antagonist differentiates between the low-affinity states of alpha4beta1 and alpha4beta7: characterization of divalent cation dependence. J Pharmacol Exp Ther 2003; 306:903-13. [PMID: 12766251 DOI: 10.1124/jpet.102.047704] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An alpha4beta1/alpha4beta7 dual antagonist, 35S-compound 1, was used as a model ligand to study the effect of divalent cations on the activation state and ligand binding properties of alpha4 integrins. In the presence of 1 mM each Ca2+/Mg2+, 35S-compound 1 bound to several cell lines expressing both alpha4beta1 and alpha4beta7, but 2S-[(1-benzenesulfonyl-pyrrolidine-2S-carbonyl)-amino]-4-[4-methyl-2S-(methyl-[2-[4-(3-o-tolyl-ureido)-phenyl]-acetyl]-amino) pentanoylamino]-butyric acid (BIO7662), a specific alpha4beta1 antagonist, completely inhibited 35S-compound 1 binding, suggesting that alpha4beta1 was responsible for the observed binding. 35S-Compound 1 bound RPMI-8866 cells expressing predominantly alpha4beta7 with a KD of 1.9 nM in the presence of 1 mM Mn2+, and binding was inhibited only 29% by BIO7662, suggesting that the probe is a potent antagonist of activated alpha4beta7. With Ca2+/Mg2+, 35S-compound 1 bound Jurkat cells expressing primarily alpha4beta1 with a KD of 18 nM. In contrast, the binding of 35S-compound 1 to Mn2+-activated Jurkat cells occurred slowly, reaching equilibrium by 60 min, and failed to dissociate within another 60 min. The ability of four alpha4beta1/alpha4beta7 antagonists to block binding of activated alpha4beta1 or alpha4beta7 to vascular cell adhesion molecule-1 or mucosal addressin cell adhesion molecule-1, respectively, or to 35S-compound 1 was measured, and a similar rank order of potency was observed for native ligand and probe. Inhibition of 35S-compound 1 binding to alpha4beta1 in Ca2+/Mg2+ was used to identify nonselective antagonists among these four. These studies demonstrate that alpha4beta1 and alpha4beta7 have distinct binding properties for the same ligand, and binding parameters are dependent on the state of integrin activation in response to different divalent cations.
Collapse
Affiliation(s)
- Linda A Egger
- Pharmacology, Merck & Co, Inc, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
Integrins are cell adhesion receptors that couple extracellular divalent cation-dependent recognition events with intracellular mechanical and biochemical responses and vice versa, thus affecting every function of nucleated cells. The structural basis of this bidirectional signaling and its dependency on cations has been the focus of intensive study over the past three decades. Significant progress made recently in elucidating the three-dimensional structure of the extracellular and cytoplasmic segments of integrins is giving valuable new insights into the tertiary and quaternary changes that underlie activation, ligand recognition and signaling by these receptors.
Collapse
Affiliation(s)
- J-P Xiong
- Renal Unit, Leukocyte Biology & Inflammation Program, Structural Biology Program, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | | | | | | |
Collapse
|
32
|
Doherty GA, Yang GX, Borges E, Tong S, McCauley ED, Treonz KM, Van Riper G, Pacholok S, Si Q, Koo GC, Shah K, Mumford RA, Hagmann WK. N-isonicotinoyl-(L)-4-aminophenylalanine derivatives as tight binding VLA-4 antagonists. Bioorg Med Chem Lett 2003; 13:1891-5. [PMID: 12749892 DOI: 10.1016/s0960-894x(03)00308-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
A series of isonicotinoyl-(L)-aminophenylalanine derivatives was prepared and evaluated as VLA-4 antagonists. These compounds exhibit subnanomolar binding affinity to VLA-4 and significant off-rates. The interplay between off-rate, protein binding and pharmacokinetics is discussed.
Collapse
Affiliation(s)
- George A Doherty
- Department of Medicinal Chemistry, Merck Research Laboratories, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Leone DR, Giza K, Gill A, Dolinski BM, Yang W, Perper S, Scott DM, Lee WC, Cornebise M, Wortham K, Nickerson-Nutter C, Chen LL, LePage D, Spell JC, Whalley ET, Petter RC, Adams SP, Lobb RR, Pepinsky RB. An assessment of the mechanistic differences between two integrin alpha 4 beta 1 inhibitors, the monoclonal antibody TA-2 and the small molecule BIO5192, in rat experimental autoimmune encephalomyelitis. J Pharmacol Exp Ther 2003; 305:1150-62. [PMID: 12626659 DOI: 10.1124/jpet.102.047332] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Integrin alpha 4 beta 1 plays an important role in inflammatory processes by regulating the migration of lymphocytes into inflamed tissues. Here we evaluated the biochemical, pharmacological, and pharmacodynamic properties and efficacy in experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis, of two types of alpha 4 beta 1 inhibitors, the anti-rat alpha 4 monoclonal antibody TA-2 and the small molecule inhibitor BIO5192 [2(S)-[[1-(3,5-dichloro-benzenesulfonyl)-pyrrolidine-2(S)-carbonyl]-amino]-4-[4-methyl-2(S)-(methyl-[2-[4-(3-o-tolyl-ureido)-phenyl]-acetyl]-amino)-pentanoylamino]-butyric acid]. TA-2 has been extensively studied in rats and provides a benchmark for assessing function. BIO5192 is a highly selective and potent (KD of <10 pM) inhibitor of alpha 4 beta 1. Dosing regimens were identified for both inhibitors, which provided full receptor occupancy during the duration of the study. Both inhibitors induced leukocytosis, an effect that was used as a pharmacodynamic marker of activity, and both were efficacious in the EAE model. Treatment with TA-2 caused a decrease in alpha 4 integrin expression on the cell surface, which resulted from internalization of alpha 4 integrin/TA-2 complexes. In contrast, BIO5192 did not modulate cell surface alpha 4 beta 1. Our results with BIO5192 indicate that alpha 4 beta 7 does not play a role in this model and that blockade of alpha 4 beta 1/ligand interactions without down-modulation is sufficient for efficacy in rat EAE. BIO5192 is highly selective and binds with high affinity to alpha 4 beta 1 from four of four species tested. These studies demonstrate that BIO5192, a novel, potent, and selective inhibitor of alpha 4 beta 1 integrin, will be a valuable reagent for assessing alpha 4 beta 1 biology and may provide a new therapeutic for treatment of human inflammatory diseases.
Collapse
Affiliation(s)
- D R Leone
- Biogen, Inc., 12 Cambridge Center, Cambridge, MA 02142, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|