1
|
Discovery of antibodies and cognate surface targets for ovarian cancer by surface profiling. Proc Natl Acad Sci U S A 2023; 120:e2206751120. [PMID: 36574667 PMCID: PMC9910589 DOI: 10.1073/pnas.2206751120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Although antibodies targeting specific tumor-expressed antigens are the standard of care for some cancers, the identification of cancer-specific targets amenable to antibody binding has remained a bottleneck in development of new therapeutics. To overcome this challenge, we developed a high-throughput platform that allows for the unbiased, simultaneous discovery of antibodies and targets based on phenotypic binding profiles. Applying this platform to ovarian cancer, we identified a wide diversity of cancer targets including receptor tyrosine kinases, adhesion and migration proteins, proteases and proteins regulating angiogenesis in a single round of screening using genomics, flow cytometry, and mass spectrometry. In particular, we identified BCAM as a promising candidate for targeted therapy in high-grade serous ovarian cancers. More generally, this approach provides a rapid and flexible framework to identify cancer targets and antibodies.
Collapse
|
2
|
Escamilla-Guerrero G, García-Rosales JC. [Genotyping and its applications, a look to the future]. REVISTA MEDICA DEL INSTITUTO MEXICANO DEL SEGURO SOCIAL 2023; 61:S37-S45. [PMID: 36378105 PMCID: PMC10396029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 08/19/2022] [Indexed: 06/16/2023]
Abstract
The detection of the most significant erythrocyte antigens present in each one of the individuals is fundamental when carrying out a transfusion or a transplant. Detection to date is performed by conventional serological methods through the antigen-antibody reaction. But several drawbacks may arise depending on the pathology under study, limiting the availability of blood components. Molecular methods such as genotyping is a tool that complements sensitivity and specificity and has come to revolutionize immunohematology in the blood bank, allowing not only the detection of erythrocyte antigens but also platelet antigens. These methodologies are applicable in patients and in large-scale donors, starting from the allelic variants present in each of the genes that code for the antigens of clinical interest, using microarray systems or systems based on particles labeled with specific probes or their variants that allow an analysis from the immunohematological point of view.
Collapse
Affiliation(s)
- Guillermo Escamilla-Guerrero
- Limogen, Laboratorio de Innovación Molecular y Genética, Laboratorio de Biología Molecular e Inmunohematología. Tlalnepantla, Estado de México, MéxicoLimogenMéxico
| | - Juan Carlos García-Rosales
- Limogen, Laboratorio de Innovación Molecular y Genética, Laboratorio de Biología Molecular e Inmunohematología. Tlalnepantla, Estado de México, MéxicoLimogenMéxico
| |
Collapse
|
3
|
Kikkawa Y, Matsunuma M, Kan R, Yamada Y, Hamada K, Nomizu M, Negishi Y, Nagamori S, Toda T, Tanaka M, Kanagawa M. Laminin α5_CD239_Spectrin is a candidate association that compensates the linkage between the basement membrane and cytoskeleton in skeletal muscle fibers. Matrix Biol Plus 2022; 15:100118. [PMID: 35990309 PMCID: PMC9382564 DOI: 10.1016/j.mbplus.2022.100118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/28/2022] [Accepted: 08/02/2022] [Indexed: 11/06/2022] Open
Abstract
Laminin α5_CD239_spectrin complex is a candidate linkage in sarcolemma. The linkage molecules are expressed in embryonic and regenerative muscle fibers. CD239 expression is upregulated by steroid therapy for muscular dystrophy. The compensatory linkage may be a therapeutic target for muscular dystrophy.
The linkage between the basement membrane (BM) and cytoskeleton is crucial for muscle fiber stability and signal transduction. Mutations in the linkage molecules can cause various types of muscular dystrophies. The different severities and times of onset suggest that compensatory linkages occur at the sarcolemma. Cluster of differentiation 239 (CD239) binds to the α5 subunit of laminin-511 extracellularly and is connected to spectrin intracellularly, resulting in a linkage between the BM and cytoskeleton. In this study, we explored the linkage of laminin α5_CD239_spectrin in skeletal muscles. Although laminin α5, CD239, and spectrin were present in embryonic skeletal muscles, they disappeared in adult skeletal muscle tissues, except for the soleus and diaphragm. Laminin α5_CD239_spectrin was localized in the skeletal muscle tissues of Duchenne muscular dystrophy and congenital muscular dystrophy mouse models. The experimental regeneration of skeletal muscle increased the CD239-mediated linkage, indicating that it responds to regeneration, but not to genetic influence. Furthermore, in silico analysis showed that laminin α5_CD239_spectrin was upregulated by steroid therapy for muscular dystrophy. Therefore, CD239-mediated linkage may serve as a therapeutic target to prevent the progression of muscular dystrophy.
Collapse
|
4
|
Ekman S, Flower R, Barnard RT, Gould A, Bui XT. Computational modeling - an approach to the development of blood grouping reagents. Expert Rev Hematol 2021; 14:329-334. [PMID: 33759674 DOI: 10.1080/17474086.2021.1908119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Blood group antigens are defined by an immune response that generates antibodies against a red blood cell molecule. Antibodies against these antigens can be associated with hemolytic transfusion reactions. However, difficulties can arise when developing antibodies against antigens through the use of peptide sequences alone. Three-dimensional representations (models) of the molecular structure of antigen-bearing proteins can provide valuable insights into tertiary structures and their consequent antigenicity. This can be achieved through predictive computational modeling to produce both structural and molecular dynamics models of blood group proteins.Areas covered: Authors discuss the use of molecular dynamic simulations on existing structures, as well as the use of computational modeling techniques in the development of protein models lacking preexisting data. Finally, the authors discuss specific examples of the use of computationally derived models of the MNS blood group system and its use in attempts to produce antibodies against MNS proteins.Expert opinion: Although in silico techniques have limitations, computer-based predictive models can inform the direction of research into blood group proteins. It is to be expected that as computer-based techniques grow more powerful these contributions will be even more significant.
Collapse
Affiliation(s)
- Serena Ekman
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia.,Clinical Services and Research, Australian Red Cross Lifeblood (Formerly Australian Red Cross Blood Service), Research and Development, Kelvin Grove, Australia
| | - Robert Flower
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia.,Clinical Services and Research, Australian Red Cross Lifeblood (Formerly Australian Red Cross Blood Service), Research and Development, Kelvin Grove, Australia
| | - Ross T Barnard
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia.,School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, Australia
| | - Alison Gould
- Australian Red Cross Lifeblood (Formerly Australian Red Cross Blood Service), Research and Development, Alexandria, Australia
| | - Xuan T Bui
- ARC Training Centre for Biopharmaceutical Innovation, Australian Institute of Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia.,Clinical Services and Research, Australian Red Cross Lifeblood (Formerly Australian Red Cross Blood Service), Research and Development, Kelvin Grove, Australia
| |
Collapse
|
5
|
Guadall A, Cochet S, Renaud O, Colin Y, Le Van Kim C, de Brevern AG, El Nemer W. Dimerization and phosphorylation of Lutheran/basal cell adhesion molecule are critical for its function in cell migration on laminin. J Biol Chem 2019; 294:14911-14921. [PMID: 31413112 DOI: 10.1074/jbc.ra119.007521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 07/02/2019] [Indexed: 12/11/2022] Open
Abstract
Tumor cell migration depends on the interactions of adhesion proteins with the extracellular matrix. Lutheran/basal cell adhesion molecule (Lu/BCAM) promotes tumor cell migration by binding to laminin α5 chain, a subunit of laminins 511 and 521. Lu/BCAM is a type I transmembrane protein with a cytoplasmic domain of 59 (Lu) or 19 (Lu(v13)) amino acids. Here, using an array of techniques, including site-directed mutagenesis, immunoblotting, FRET, and proximity-ligation assays, we show that both Lu and Lu(v13) form homodimers at the cell surface of epithelial cancer cells. We mapped two small-XXX-small motifs in the transmembrane domain as potential sites for monomers docking and identified three cysteines in the cytoplasmic domain as being critical for covalently stabilizing dimers. We further found that Lu dimerization and phosphorylation of its cytoplasmic domain were concomitantly needed to promote cell migration. We conclude that Lu is the critical isoform supporting tumor cell migration on laminin 521 and that the Lu:Lu(v13) ratio at the cell surface may control the balance between cellular firm adhesion and migration.
Collapse
Affiliation(s)
- Anna Guadall
- Université de Paris, UMR_S1134, BIGR, Inserm, F-75015 Paris, France.,Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, 75015 Paris, France
| | - Sylvie Cochet
- Université de Paris, UMR_S1134, BIGR, Inserm, F-75015 Paris, France.,Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, 75015 Paris, France
| | - Olivier Renaud
- Institut Curie, Paris Sciences et Lettres Research University, 75005 Paris, France.,U934, Institut National de la Santé et de la Recherche Médicale, 75005 Paris, France.,UMR3215, Centre National de la Recherche Scientifique, 75005 Paris, France.,Cell and Tissue Imaging Facility (PICT-IBiSA), Institut Curie, 75005 Paris, France
| | - Yves Colin
- Université de Paris, UMR_S1134, BIGR, Inserm, F-75015 Paris, France.,Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, 75015 Paris, France
| | - Caroline Le Van Kim
- Université de Paris, UMR_S1134, BIGR, Inserm, F-75015 Paris, France.,Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, 75015 Paris, France
| | - Alexandre G de Brevern
- Université de Paris, UMR_S1134, BIGR, Inserm, F-75015 Paris, France.,Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, 75015 Paris, France
| | - Wassim El Nemer
- Université de Paris, UMR_S1134, BIGR, Inserm, F-75015 Paris, France .,Institut National de la Transfusion Sanguine, F-75015 Paris, France.,Laboratoire d'Excellence GR-Ex, 75015 Paris, France
| |
Collapse
|
6
|
Novitzky-Basso I, Spring F, Anstee D, Tripathi D, Chen F. Erythrocytes from patients with myeloproliferative neoplasms and splanchnic venous thrombosis show greater expression of Lu/BCAM. Int J Lab Hematol 2018; 40:473-477. [PMID: 29756283 DOI: 10.1111/ijlh.12838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2017] [Accepted: 03/09/2018] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Lutheran/BCAM protein (Lu) on the surface of erythrocytes is key for their adhesion to the endothelium, and erythrocytes from individuals with JAK2V617F-mutated myeloproliferative neoplasms (MPN) have increased endothelial adhesion. Splanchnic vein thrombosis (SVT) is a devastating thrombotic complication of MPN, and frequently, the only diagnostic feature is the JAK2V617F mutation. We sought to examine whether erythrocytes from patients with JAK2V617F mutated SVT (MPN-SVT) exhibited increased Lu expression, thereby supporting a mechanistic contribution to the development of thrombosis. METHODS We report the validation of a novel flow cytometry assay for Lu expression on erythrocytes. We examined the expression of Lu on erythrocytes from a cohort of MPN patients with and without SVT, and healthy controls. Samples were obtained from 20 normal individuals, 22 with MPN (both JAK2V617F-mutated and wild-type) and 8 with JAK2V617F-mutated MPN-SVT. RESULTS Lu expression by erythrocytes from patients with MPN and MPN-SVT is significantly increased compared to erythrocytes from healthy individuals (P < .05), but there was no significant difference between patients with MPN-SVT and MPN. CONCLUSIONS Patients with MPN have increased expression of the red cell Lu/BCAM adhesion molecule. Further work is required to determine the role of the increased Lu/BCAM adhesion to the endothelium in the development of thrombosis in MPN of all genotypes.
Collapse
Affiliation(s)
- I Novitzky-Basso
- Scottish Blood and Marrow Transplant Unit, Queen Elizabeth University Hospital, Glasgow, UK
| | - F Spring
- NHS Blood and Transplant, Filton, Bristol, UK
| | - D Anstee
- NHS Blood and Transplant, Filton, Bristol, UK
| | - D Tripathi
- Liver Unit, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, UK
- Centre for Rare Disease, University of Birmingham, Birmingham, UK
| | - F Chen
- Centre for Clinical Haematology, Queen Elizabeth Hospital Birmingham NHS Trust, Birmingham, UK
- Centre for Rare Disease, University of Birmingham, Birmingham, UK
- NHS Blood and Transplant, Birmingham, UK
| |
Collapse
|
7
|
Rohn F, Kordes C, Castoldi M, Götze S, Poschmann G, Stühler K, Herebian D, Benk AS, Geiger F, Zhang T, Spatz JP, Häussinger D. Laminin-521 promotes quiescence in isolated stellate cells from rat liver. Biomaterials 2018; 180:36-51. [PMID: 30014965 DOI: 10.1016/j.biomaterials.2018.07.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 07/06/2018] [Indexed: 12/15/2022]
Abstract
The laminin α5 protein chain is an element of basement membranes and important to maintain stem cells. Hepatic stellate cells (HSC) are liver-resident mesenchymal stem cells, which reside in a quiescent state on a basement membrane-like structure in the space of Dissé. In the present study, laminin α5 chain was detected in the space of Dissé of normal rat liver. Since HSC are critical for liver regeneration and can contribute to fibrosis in chronic liver diseases, the effect of laminins on HSC maintenance was investigated. Therefore, isolated rat HSC were seeded on uncoated polystyrene (PS) or PS coated with either laminin-521 (PS/LN-521) or laminin-211 (PS/LN-211). PS/LN-521 improved HSC adhesion and better preserved their retinoid stores as well as quiescence- and stem cell-associated phenotype, whereas HSC on PS/LN-211 or PS developed into myofibroblasts-like cells. To improve the homogeneity as well as the presentation of laminin molecules on the culture surface to HSC, laminin-functionalized, gold-nanostructured glass surfaces were generated. This approach further enhanced the expression of quiescence-associated genes in HSC. In conclusion, the results indicate that LN-521 supports the quiescent state of HSC and laminin α5 can be regarded as an important element of their niche in the space of Dissé.
Collapse
Affiliation(s)
- Friederike Rohn
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Claus Kordes
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Mirco Castoldi
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Silke Götze
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Gereon Poschmann
- Molecular Proteomics Laboratory, Biologisch-Medizinisches Forschungszentrum, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory, Biologisch-Medizinisches Forschungszentrum, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; Institute of Molecular Medicine, University Hospital Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany
| | - Diran Herebian
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty, Heinrich Heine University, 40225 Düsseldorf, Germany
| | - Amelie S Benk
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, 69120 Heidelberg, Germany; Department of Biophysical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
| | - Fania Geiger
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, 69120 Heidelberg, Germany; Department of Biophysical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
| | - Tingyu Zhang
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, 69120 Heidelberg, Germany; Department of Biophysical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
| | - Joachim P Spatz
- Max-Planck-Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, 69120 Heidelberg, Germany; Department of Biophysical Chemistry, University of Heidelberg, Im Neuenheimer Feld 253, 69120 Heidelberg, Germany
| | - Dieter Häussinger
- Clinic of Gastroenterology, Hepatology and Infectious Diseases, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany.
| |
Collapse
|
8
|
Kikkawa Y, Enomoto-Okawa Y, Fujiyama A, Fukuhara T, Harashima N, Sugawara Y, Negishi Y, Katagiri F, Hozumi K, Nomizu M, Ito Y. Internalization of CD239 highly expressed in breast cancer cells: a potential antigen for antibody-drug conjugates. Sci Rep 2018; 8:6612. [PMID: 29700410 PMCID: PMC5919910 DOI: 10.1038/s41598-018-24961-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 04/12/2018] [Indexed: 01/25/2023] Open
Abstract
Antibody-drug conjugates (ADCs) are attractive in cancer therapy because they can directly bind to cancer cells and provide anticancer activity. To kill cancer cells with ADCs, the target antigens are required not only to be highly and/or selectively expressed on cancer cells but also internalized by the cells. CD239, also known as the Lutheran blood group glycoprotein (Lu) or basal cell adhesion molecule (B-CAM), is a specific receptor for laminin α5, a major component of basement membranes. Here, we show that CD239 is strongly expressed in a subset of breast cancer cells and internalized into the cells. We also produced a human single-chain variable fragment (scFv) specific to CD239 fused with human IgG1 Fc, called C7-Fc. The binding affinity of the C7-Fc antibody is similar to that of mouse monoclonal antibodies. Although the C7-Fc antibody alone does not influence cellular functions, when conjugated with a fragment of diphtheria toxin lacking the receptor-binding domain (fDT), it can selectively kill breast cancer cells. Interestingly, fDT-bound C7-Fc shows anticancer activity in CD239-highly positive SKBR3 cells, but not in weakly positive cells. Our results show that CD239 is a promising antigen for ADC-based breast cancer therapy.
Collapse
Affiliation(s)
- Yamato Kikkawa
- Department of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392, Japan.
| | - Yurie Enomoto-Okawa
- Graduate School of Science and Engineering, Kagoshima University, Kagoshima, 890-0065, Japan
| | - Aiko Fujiyama
- Graduate School of Science and Engineering, Kagoshima University, Kagoshima, 890-0065, Japan
| | - Takeshi Fukuhara
- Laboratory of Oncology, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392, Japan.,Department of Neurology, Graduate School of Medicine, Juntendo University, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Nozomi Harashima
- Department of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392, Japan
| | - Yumika Sugawara
- Department of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392, Japan
| | - Yoichi Negishi
- Department of Drug Delivery and Molecular Biopharmaceutics, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392, Japan
| | - Fumihiko Katagiri
- Department of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392, Japan
| | - Kentaro Hozumi
- Department of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392, Japan
| | - Motoyoshi Nomizu
- Department of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Tokyo, 192-0392, Japan
| | - Yuji Ito
- Graduate School of Science and Engineering, Kagoshima University, Kagoshima, 890-0065, Japan
| |
Collapse
|
9
|
Glycophorin-C sialylation regulates Lu/BCAM adhesive capacity during erythrocyte aging. Blood Adv 2018; 2:14-24. [PMID: 29344581 DOI: 10.1182/bloodadvances.2017013094] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/30/2017] [Indexed: 11/20/2022] Open
Abstract
Lutheran/basal cell adhesion molecule (Lu/BCAM) is a transmembrane adhesion molecule expressed by erythrocytes and endothelial cells that can interact with the extracellular matrix protein laminin-α5. In sickle cell disease, Lu/BCAM is thought to contribute to adhesion of sickle erythrocytes to the vascular wall, especially during vaso-occlusive crises. On healthy erythrocytes however, its function is unclear. Here we report that Lu/BCAM is activated during erythrocyte aging. We show that Lu/BCAM-mediated binding to laminin-α5 is restricted by interacting, in cis, with glycophorin-C-derived sialic acid residues. Following loss of sialic acid during erythrocyte aging, Lu/BCAM is released from glycophorin-C and allowed to interact with sialic acid residues on laminin-α5. Decreased glycophorin-C sialylation, as observed in individuals lacking exon 3 of glycophorin-C, the so-called Gerbich phenotype, was found to correlate with increased Lu/BCAM-dependent binding to laminin-α5. In addition, we identified the sialic acid-binding site within the third immunoglobulin-like domain within Lu/BCAM that accounts for the interaction with glycophorin-C and laminin-α5. Last, we present evidence that neuraminidase-expressing pathogens, such as Streptococcus pneumoniae, can similarly induce Lu/BCAM-mediated binding to laminin-α5, by cleaving terminal sialic acid residues from the erythrocyte membrane. These results shed new light on the mechanisms contributing to increased adhesiveness of erythrocytes at the end of their lifespan, possibly facilitating their clearance. Furthermore, this work may contribute to understanding the pathology induced by neuraminidase-positive bacteria, because they are especially harmful to patients suffering from sickle cell disease and are associated with the occurrence of vaso-occlusive crises.
Collapse
|
10
|
Reppin F, Cochet S, El Nemer W, Fritz G, Schmidt G. High Affinity Binding of Escherichia coli Cytotoxic Necrotizing Factor 1 (CNF1) to Lu/BCAM Adhesion Glycoprotein. Toxins (Basel) 2017; 10:toxins10010003. [PMID: 29267242 PMCID: PMC5793090 DOI: 10.3390/toxins10010003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 01/07/2023] Open
Abstract
The protein toxin Cytotoxic Necrotizing Factor 1 (CNF1) is a major virulence factor of pathogenic Escherichia coli strains. It belongs to a family of single chain AB-toxins, which enter mammalian cells by receptor-mediated endocytosis. Recently, we identified the Lutheran (Lu) adhesion glycoprotein/basal cell adhesion molecule (BCAM) as a cellular receptor for CNF1. Here, we identified the Ig-like domain 2 of Lu/BCAM as main interaction site of the toxin by direct protein-protein interaction and competition studies. Using surface plasmon resonance, we showed a high affinity CNF-Lu/BCAM interaction with a KD of 2.8 nM. Furthermore, we performed small-angle X-ray scattering to define the molecular envelope of the Lu/BCAM-CNF1 complex, suggesting a 6:1 ratio of Lu/BCAM to CNF1 in the receptor-toxin complex. This study leads to a deeper understanding of the interaction between CNF1 and Lu/BCAM, and presents novel opportunities for the development of future anti-toxin strategies.
Collapse
Affiliation(s)
- Franziska Reppin
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Albert-Street 25, 79104 Freiburg, Germany.
- Biological Faculty, Albert-Ludwigs-University of Freiburg, Albert-Street 25, 79104 Freiburg, Germany.
| | - Sylvie Cochet
- Universite Sorbonne Paris Cite, Universite Paris Diderot, Inserm, INTS, Unite Biologie Integree du Globule Rouge, Laboratoire d'Excellence GR-Ex, 75013 Paris, France.
| | - Wassim El Nemer
- Universite Sorbonne Paris Cite, Universite Paris Diderot, Inserm, INTS, Unite Biologie Integree du Globule Rouge, Laboratoire d'Excellence GR-Ex, 75013 Paris, France.
| | - Günter Fritz
- Department of Neuropathology, Albert-Ludwigs-University of Freiburg, Breisacher Strasse 64, 79106 Freiburg, Germany.
| | - Gudula Schmidt
- Institute for Experimental and Clinical Pharmacology and Toxicology, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Albert-Street 25, 79104 Freiburg, Germany.
| |
Collapse
|
11
|
An Anti-Human Lutheran Glycoprotein Phage Antibody Inhibits Cell Migration on Laminin-511: Epitope Mapping of the Antibody. PLoS One 2017; 12:e0167860. [PMID: 28060819 PMCID: PMC5218393 DOI: 10.1371/journal.pone.0167860] [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: 09/03/2016] [Accepted: 11/21/2016] [Indexed: 11/24/2022] Open
Abstract
The Lutheran glycoprotein (Lu), also known as basal cell adhesion molecule (B-CAM), is an Ig superfamily (IgSF) transmembrane receptor for laminin α5. Although Lu is not present in normal hepatocytes, its expression is significantly increased in hepatocellular carcinoma (HCC). In this study, we isolated thirteen phage antibodies to Lu from a phage library of peripheral blood from HCC patients, suggesting that these patients produced autoantibodies against endogenous Lu. To characterize the phage antibodies, we determined the Lu domains they recognize. The extracellular domain of Lu contains five IgSF domains, D1-D2-D3-D4-D5. The epitope of one phage antibody (A7) was localized to the D5 domain. The other phage antibodies recognized the D2 domain, which is also recognized by a function blocking mouse monoclonal antibody. One of the antibodies to D2 (C7) inhibited the binding of Lu to ligand, and it also prevented tumor cell migration on laminin-511 (LM-511). However, the C7 scFv purified from the periplasm fraction of bacteria did not exhibit the inhibitory effects, indicating that the scFv form could not sterically inhibit the binding of Lu to LM-511. We also identified the amino acid residues that form the epitope recognized by the C7 phage antibody. Mutagenesis studies showed that Arg247 is necessary for forming the epitope. The C7 phage antibody and its epitope may be useful for developing drugs to prevent HCC progression and/or metastasis.
Collapse
|
12
|
The macrophage contribution to stress erythropoiesis: when less is enough. Blood 2016; 128:1756-65. [PMID: 27543439 DOI: 10.1182/blood-2016-05-714527] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 08/12/2016] [Indexed: 12/15/2022] Open
Abstract
Although the importance of native bone marrow and spleen macrophages in enhancing baseline and stress erythropoiesis has been emphasized over several decades, their kinetic and phenotypic changes during a variety of stress responses have been unclear. Furthermore, whether monocyte-derived recruited macrophages can functionally substitute for inadequate or functionally impaired native macrophages has been controversial and seem to be not only tissue- but also stress-type dependent. To provide further insight into these issues, we made detailed observations at baseline and post-erythroid stress (E-stress) in 2 mouse models with genetically depressed macrophage numbers and compared them to their controls. We documented that, irrespective of the stress-induced (hemolytic or post-erythropoietin [Epo]) treatment, only native CD11b(lo) splenic macrophages expand dramatically post-stress in normal mice without significant changes in the monocyte-derived CD11b(hi) subset. The latter remained a minority and did not change post-stress in 2 genetic models lacking either Spi-C or VCAM-1 with impaired native macrophage proliferative expansion. Although CD11b(lo) macrophages in these mice were one-fifth of normal at their peak response, surprisingly, their erythroid response was not compromised and was similar to controls. Thus, despite the prior emphasis on numerical macrophage reliance to provide functional rescue from E-stress, our data highlight the importance of previously described non-macrophage-dependent pathways activated under certain stress conditions to compensate for low macrophage numbers.
Collapse
|
13
|
Bartolini A, Cardaci S, Lamba S, Oddo D, Marchiò C, Cassoni P, Amoreo CA, Corti G, Testori A, Bussolino F, Pasqualini R, Arap W, Corà D, Di Nicolantonio F, Marchiò S. BCAM and LAMA5 Mediate the Recognition between Tumor Cells and the Endothelium in the Metastatic Spreading of KRAS-Mutant Colorectal Cancer. Clin Cancer Res 2016; 22:4923-4933. [PMID: 27143691 DOI: 10.1158/1078-0432.ccr-15-2664] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 03/31/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE KRAS mutations confer adverse prognosis to colorectal cancer, and no targeted therapies have shown efficacy in this patient subset. Paracrine, nongenetic events induced by KRAS-mutant tumor cells are expected to result in specific deregulation and/or relocation of tumor microenvironment (TME) proteins, which in principle can be exploited as alternative therapeutic targets. EXPERIMENTAL DESIGN A multimodal strategy combining ex vivo/in vitro phage display screens with deep-sequencing and bioinformatics was applied to uncover TME-specific targets in KRAS-mutant hepatic metastasis from colorectal cancer. Expression and localization of BCAM and LAMA5 were validated by immunohistochemistry in preclinical models of human hepatic metastasis and in a panel of human specimens (n = 71). The antimetastatic efficacy of two BCAM-mimic peptides was evaluated in mouse models. The role of BCAM in the interaction of KRAS-mutant colorectal cancer cells with TME cells was investigated by adhesion assays. RESULTS BCAM and LAMA5 were identified as molecular targets within both tumor cells and TME of KRAS-mutant hepatic metastasis from colorectal cancer, where they were specifically overexpressed. Two BCAM-mimic peptides inhibited KRAS-mutant hepatic metastasis in preclinical models. Genetic suppression and biochemical inhibition of either BCAM or LAMA5 impaired adhesion of KRAS-mutant colorectal cancer cells specifically to endothelial cells, whereas adhesion to pericytes and hepatocytes was unaffected. CONCLUSIONS These data show that the BCAM/LAMA5 system plays a functional role in the metastatic spreading of KRAS-mutant colorectal cancer by mediating tumor-TME interactions and as such represents a valuable therapeutic candidate for this large, currently untreatable patient group. Clin Cancer Res; 22(19); 4923-33. ©2016 AACR.
Collapse
Affiliation(s)
- Alice Bartolini
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (Turin), Italy
| | - Sabrina Cardaci
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (Turin), Italy
| | - Simona Lamba
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (Turin), Italy
| | - Daniele Oddo
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (Turin), Italy. Department of Oncology, University of Turin, Candiolo (Turin), Italy
| | - Caterina Marchiò
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Paola Cassoni
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | - Giorgio Corti
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (Turin), Italy
| | | | - Federico Bussolino
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (Turin), Italy. Department of Oncology, University of Turin, Candiolo (Turin), Italy
| | - Renata Pasqualini
- University of New Mexico Comprehensive Cancer Center. Albuquerque, New Mexico. Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Wadih Arap
- University of New Mexico Comprehensive Cancer Center. Albuquerque, New Mexico. Division of Hematology/Oncology, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico
| | - Davide Corà
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (Turin), Italy. Department of Oncology, University of Turin, Candiolo (Turin), Italy
| | - Federica Di Nicolantonio
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (Turin), Italy. Department of Oncology, University of Turin, Candiolo (Turin), Italy.
| | - Serena Marchiò
- Candiolo Cancer Institute-FPO, IRCCS, Candiolo (Turin), Italy. Department of Oncology, University of Turin, Candiolo (Turin), Italy. University of New Mexico Comprehensive Cancer Center. Albuquerque, New Mexico. Division of Molecular Medicine, Department of Internal Medicine, University of New Mexico School of Medicine, Albuquerque, New Mexico.
| |
Collapse
|
14
|
Kikkawa Y, Miwa T, Tanimizu N, Kadoya Y, Ogawa T, Katagiri F, Hozumi K, Nomizu M, Mizuguchi T, Hirata K, Mitaka T. Soluble Lutheran/basal cell adhesion molecule is detectable in plasma of hepatocellular carcinoma patients and modulates cellular interaction with laminin-511 in vitro. Exp Cell Res 2014; 328:197-206. [PMID: 25051049 DOI: 10.1016/j.yexcr.2014.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 07/05/2014] [Accepted: 07/11/2014] [Indexed: 11/26/2022]
Abstract
Lutheran (Lu), an immunoglobulin superfamily transmembrane receptor, is also known as basal cell adhesion molecule (B-CAM). Lu/B-CAM is a specific receptor for laminin α5, a subunit of laminin-511 (LM-511) that is a major component of basement membranes in various tissues. Our previous study showed that Lu/B-CAM was cleaved by MT1-MMP and released from cell surfaces. In this study we examined the soluble Lu/B-CAM in culture media and in plasma of mice bearing HuH-7 hepatocellular carcinoma (HCC) cells and patients with HCC. Two HCC cell lines, HepG2 and HuH-7, released Lu/B-CAM into the culture media. Although Lu/B-CAM was cleaved by MT1-MMP in HuH-7 cells, HepG2 cells released Lu/B-CAM in a MMP-independent manner. The concentration of Lu/B-CAM released into mouse plasma correlated with tumor size. Moreover the soluble Lu/B-CAM in plasma of HCC patients was significantly decreased after resection of the tumor. Immunohistochemical studies showed that although the expression of Lu/B-CAM was observed in most HCCs, MT1-MMP was not always expressed in tumor tissues, suggesting that a part of Lu/B-CAM in plasma of HCC patients was also released in a MMP-independent manner. In vitro studies showed that the soluble Lu/B-CAM released from HCC cells bound to LM-511. Moreover the soluble Lu/B-CAM influenced cell migration on LM-511. These results suggest that soluble Lu/B-CAM serves as not only a novel marker for HCC but also a modulator in tumor progression.
Collapse
Affiliation(s)
- Yamato Kikkawa
- Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan.
| | - Takahiro Miwa
- Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Naoki Tanimizu
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| | - Yuichi Kadoya
- Department of Anatomy, Kitasato University School of Allied Health Sciences, Minami-ku, Sagamihara, Japan
| | - Takaho Ogawa
- Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Fumihiko Katagiri
- Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Kentaro Hozumi
- Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Motoyoshi Nomizu
- Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Toru Mizuguchi
- Department of 1st Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Koichi Hirata
- Department of 1st Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Toshihiro Mitaka
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo, Japan
| |
Collapse
|
15
|
Sessa L, Gatti E, Zeni F, Antonelli A, Catucci A, Koch M, Pompilio G, Fritz G, Raucci A, Bianchi ME. The receptor for advanced glycation end-products (RAGE) is only present in mammals, and belongs to a family of cell adhesion molecules (CAMs). PLoS One 2014; 9:e86903. [PMID: 24475194 PMCID: PMC3903589 DOI: 10.1371/journal.pone.0086903] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 12/17/2013] [Indexed: 12/20/2022] Open
Abstract
The human receptor for advanced glycation endproducts (RAGE) is a multiligand cell surface protein belonging to the immunoglobulin superfamily, and is involved in inflammatory and immune responses. Most importantly, RAGE is considered a receptor for HMGB1 and several S100 proteins, which are Damage-Associated Molecular Pattern molecules (DAMPs) released during tissue damage. In this study we show that the Ager gene coding for RAGE first appeared in mammals, and is closely related to other genes coding for cell adhesion molecules (CAMs) such as ALCAM, BCAM and MCAM that appeared earlier during metazoan evolution. RAGE is expressed at very low levels in most cells, but when expressed at high levels, it mediates cell adhesion to extracellular matrix components and to other cells through homophilic interactions. Our results suggest that RAGE evolved from a family of CAMs, and might still act as an adhesion molecule, in particular in the lung where it is highly expressed or under pathological conditions characterized by an increase of its protein levels.
Collapse
Affiliation(s)
- Luca Sessa
- Chromatin Dynamics Unit, San Raffaele University and Research Institute, Milano, Italy
| | - Elena Gatti
- Chromatin Dynamics Unit, San Raffaele University and Research Institute, Milano, Italy
| | - Filippo Zeni
- Laboratory of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, Milano, Italy
| | - Antonella Antonelli
- Chromatin Dynamics Unit, San Raffaele University and Research Institute, Milano, Italy
| | - Alessandro Catucci
- Chromatin Dynamics Unit, San Raffaele University and Research Institute, Milano, Italy
| | - Michael Koch
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Giulio Pompilio
- Laboratory of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, Milano, Italy
| | - Günter Fritz
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Angela Raucci
- Laboratory of Vascular Biology and Regenerative Medicine, Centro Cardiologico Monzino, Milano, Italy
- * E-mail: (MEB); (AR)
| | - Marco E. Bianchi
- Chromatin Dynamics Unit, San Raffaele University and Research Institute, Milano, Italy
- * E-mail: (MEB); (AR)
| |
Collapse
|
16
|
Kikkawa Y, Ogawa T, Sudo R, Yamada Y, Katagiri F, Hozumi K, Nomizu M, Miner JH. The lutheran/basal cell adhesion molecule promotes tumor cell migration by modulating integrin-mediated cell attachment to laminin-511 protein. J Biol Chem 2013; 288:30990-1001. [PMID: 24036115 DOI: 10.1074/jbc.m113.486456] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cell-matrix interactions are critical for tumor cell migration. Lutheran (Lu), also known as basal cell adhesion molecule (B-CAM), competes with integrins for binding to laminin α5, a subunit of LM-511, a major component of basement membranes. Here we show that the preferential binding of Lu/B-CAM to laminin α5 promotes tumor cell migration. The attachment of Lu/B-CAM transfectants to LM-511 was slightly weaker than that of control cells, and this was because Lu/B-CAM disturbed integrin binding to laminin α5. Lu/B-CAM induced a spindle cell shape with pseudopods and promoted cell migration on LM-511. In addition, blocking with an anti-Lu/B-CAM antibody led to a flat cell shape and inhibited migration on LM-511, similar to the effects of an activating integrin β1 antibody. We conclude that tumor cell migration on LM-511 requires that Lu/B-CAM competitively modulates cell attachment through integrins. We suggest that this competitive interaction is involved in a balance between static and migratory cell behaviors.
Collapse
Affiliation(s)
- Yamato Kikkawa
- From the Laboratory of Clinical Biochemistry, Tokyo University of Pharmacy and Life Sciences, Hachioji, 192-0392, Japan
| | | | | | | | | | | | | | | |
Collapse
|
17
|
An antibody to the lutheran glycoprotein (Lu) recognizing the LU4 blood type variant inhibits cell adhesion to laminin α5. PLoS One 2011; 6:e23329. [PMID: 21858073 PMCID: PMC3155534 DOI: 10.1371/journal.pone.0023329] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 07/13/2011] [Indexed: 11/23/2022] Open
Abstract
Background The Lutheran blood group glycoprotein (Lu), an Ig superfamily (IgSF) transmembrane receptor, is also known as basal cell adhesion molecule (B-CAM). Lu/B-CAM is a specific receptor for laminin α5, a major component of basement membranes in various tissues. Previous reports have shown that Lu/B-CAM binding to laminin α5 contributes to sickle cell vaso-occlusion. However, as there are no useful tools such as function-blocking antibodies or drugs, it is unclear how epithelial and sickled red blood cells adhere to laminin α5 via Lu/B-CAM. Methodology/Principal Findings In this study, we discovered a function-blocking antibody that inhibits Lu binding to laminin α5 using a unique binding assay on tissue sections. To characterize the function-blocking antibody, we identified the site on Lu/B-CAM recognized by this antibody. The extracellular domain of Lu/B-CAM contains five IgSF domains, D1-D2-D3-D4-D5. The antibody epitope was localized to D2, but not to the D3 domain containing the major part of the laminin α5 binding site. Furthermore, mutagenesis studies showed that Arg175, the LU4 blood group antigenic site, was crucial for forming the epitope and the antibody bound sufficiently close to sterically hinder the interaction with α5. Cell adhesion assay using the antibody also showed that Lu/B-CAM serves as a secondary receptor for the adhesion of carcinoma cells to laminin α5. Conclusion/Significance This function-blocking antibody against Lu/B-CAM should be useful for not only investigating cell adhesion to laminin α5 but also for developing drugs to inhibit sickle cell vaso-occlusion.
Collapse
|
18
|
Novel role for the Lu/BCAM-spectrin interaction in actin cytoskeleton reorganization. Biochem J 2011; 436:699-708. [PMID: 21434869 DOI: 10.1042/bj20101717] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Lu/BCAM (Lutheran/basal cell-adhesion molecule) is a laminin 511/521 receptor expressed in erythroid and endothelial cells, and in epithelial tissues. The RK573-574 (Arg573-Lys574) motif of the Lu/BCAM cytoplasmic domain interacts with αI-spectrin, the main component of the membrane skeleton in red blood cells. In the present paper we report that Lu/BCAM binds to the non-erythroid αII-spectrin via the RK573-574 motif. Alanine substitution of this motif abolished the Lu/BCAM-spectrin interaction, enhanced the half-life of Lu/BCAM at the MDCK (Madin-Darby canine kidney) cell surface, and increased Lu/BCAM-mediated cell adhesion and spreading on laminin 511/521. We have shown that the Lu/BCAM-spectrin interaction mediated actin reorganization during cell adhesion and spreading on laminin 511/521. This interaction was involved in a laminin 511/521-to-actin signalling pathway leading to stress fibre formation. This skeletal rearrangement was associated with an activation of the small GTP-binding protein RhoA, which depended on the integrity of the Lu/BCAM laminin 511/521-binding site. It also required a Lu/BCAM-αII-spectrin interaction, since its disruption decreased stress fibre formation and RhoA activation. We conclude that the Lu/BCAM-spectrin interaction is required for stress fibre formation during cell spreading on laminin 511/521, and that spectrin acts as a signal relay between laminin 511/521 and actin that is involved in actin dynamics.
Collapse
|
19
|
Abstract
Antigens of 23 of the 30 human blood group systems are defined by the amino acid sequence of red cell membrane proteins. The antigens of DI, RH, RHAG, MNS, GE and CO systems are carried on blood group-active proteins (Band 3, D and CE polypeptides, RhAG, Glycophorins A and B, Glycophorins C and D and Aquaporin 1, respectively) which are expressed at high levels (>200,000 copies/red cell). These major proteins contribute to essential red cell functions either directly as membrane transporters and by providing linkage to the underlying red cell skeleton or by facilitating the membrane assembly of the protein complexes involved in these processes. The proteins expressing antigens of the remaining 17 blood group systems are much less abundant (<20,000 copies/red cell) and their functional importance for the circulating red cell is largely unknown. Human gene knock-outs (null phenotypes) have been described for many of these minor blood group-active proteins, but only absence of Kx glycoprotein has been clearly linked with pathology directly related to the function of circulating red cells. Recent evidence suggesting the normal quality control system for glycoprotein synthesis is altered during the latter stages of red cell production raises the possibility that many of these low abundance blood group-active proteins are vestigial. In sickle cell disease and polycythaemia vera, elevated Lutheran glycoprotein expression may contribute to pathology. Dyserythropoiesis with reduced antigen expression can result from mutations in the erythroid transcription factors GATA-1 and EKLF.
Collapse
Affiliation(s)
- D J Anstee
- Bristol Institute for Transfusion Sciences, NHS Blood and Transplant, Bristol, UK.
| |
Collapse
|
20
|
|
21
|
El Nemer W, Colin Y, Le Van Kim C. Role of Lu/BCAM glycoproteins in red cell diseases. Transfus Clin Biol 2010; 17:143-7. [DOI: 10.1016/j.tracli.2010.06.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Accepted: 06/08/2010] [Indexed: 11/24/2022]
|
22
|
Hatherley D, Graham SC, Harlos K, Stuart DI, Barclay AN. Structure of signal-regulatory protein alpha: a link to antigen receptor evolution. J Biol Chem 2009; 284:26613-9. [PMID: 19628875 PMCID: PMC2785349 DOI: 10.1074/jbc.m109.017566] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2009] [Revised: 06/18/2009] [Indexed: 11/24/2022] Open
Abstract
Signal-regulatory protein alpha (SIRPalpha) is a myeloid membrane receptor that interacts with the membrane protein CD47, a marker of self. We have solved the structure of the complete extracellular portion of SIRPalpha, comprising three immunoglobulin superfamily domains, by x-ray crystallography to 2.5 A resolution. These data, together with previous data on the N-terminal domain and its ligand CD47 (possessing a single immunoglobulin superfamily domain), show that the CD47-SIRPalpha interaction will span a distance of around 14 nm between interacting cells, comparable with that of an immunological synapse. The N-terminal (V-set) domain mediates binding to CD47, and the two others are found to be constant (C1-set) domains. C1-set domains are restricted to proteins involved in vertebrate antigen recognition: T cell antigen receptors, immunoglobulins, major histocompatibility complex antigens, tapasin, and beta2-microglobulin. The domains of SIRPalpha (domains 2 and 3) are structurally more similar to C1-set domains than any cell surface protein not involved in antigen recognition. This strengthens the suggestion from sequence analysis that SIRP is evolutionarily closely related to antigen recognition proteins.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Differentiation/chemistry
- Antigens, Differentiation/genetics
- Antigens, Differentiation/metabolism
- Binding Sites/genetics
- CD47 Antigen/chemistry
- CD47 Antigen/metabolism
- CHO Cells
- Cluster Analysis
- Cricetinae
- Cricetulus
- Crystallography, X-Ray
- Evolution, Molecular
- Humans
- Models, Molecular
- Molecular Sequence Data
- Protein Binding
- Protein Conformation
- Protein Structure, Secondary
- Protein Structure, Tertiary
- Receptors, Immunologic/chemistry
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Sequence Homology, Amino Acid
Collapse
Affiliation(s)
- Deborah Hatherley
- From the Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom and
| | - Stephen C. Graham
- the Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - Karl Harlos
- the Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - David I. Stuart
- the Division of Structural Biology and Oxford Protein Production Facility, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, United Kingdom
| | - A. Neil Barclay
- From the Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom and
| |
Collapse
|
23
|
Abstract
Abstract
Over the past 20 years the molecular bases of almost all the major blood group antigens have been determined. This research has enabled development of DNA-based methods for determining blood group genotype. The most notable application of these DNA-based methods has been for determining fetal blood group in pregnancies when the fetus is at risk for hemolytic disease of the fetus and newborn. The replacement of all conventional serologic methods for pretransfusion testing by molecular methods is not straightforward. For the majority of transfusion recipients matching beyond ABO and D type is unnecessary, and the minority of untransfused patients at risk of alloimmunization who would benefit from more extensively blood group–matched blood cannot be identified reliably. Even if a method to identify persons most likely to make alloantibodies were available, this would not of itself guarantee the provision of extensively phenotype-matched blood for these patients because this is determined by the size and racial composition of blood donations available for transfusion. However, routine use of DNA-based extended phenotyping to provide optimally matched donations for patients with preexisting antibodies or patients with a known predisposition to alloimmunization, such as those with sickle cell disease, is widely used.
Collapse
|
24
|
Rich RL, Myszka DG. Survey of the year 2007 commercial optical biosensor literature. J Mol Recognit 2008; 21:355-400. [DOI: 10.1002/jmr.928] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
25
|
Abstract
The Lutheran (Lu) and Lu(v13) blood group glycoproteins function as receptors for extracellular matrix laminins. Lu and Lu(v13) are linked to the erythrocyte cytoskeleton through a direct interaction with spectrin. However, neither the molecular basis of the interaction nor its functional consequences have previously been delineated. In the present study, we defined the binding motifs of Lu and Lu(v13) on spectrin and identified a functional role for this interaction. We found that the cytoplasmic domains of both Lu and Lu(v13) bound to repeat 4 of the alpha spectrin chain. The interaction of full-length spectrin dimer to Lu and Lu(v13) was inhibited by repeat 4 of alpha-spectrin. Further, resealing of this repeat peptide into erythrocytes led to weakened Lu-cytoskeleton interaction as demonstrated by increased detergent extractability of Lu. Importantly, disruption of the Lu-spectrin linkage was accompanied by enhanced cell adhesion to laminin. We conclude that the interaction of the Lu cytoplasmic tail with the cytoskeleton regulates its adhesive receptor function.
Collapse
|
26
|
Abstract
Erythroblastic islands, the specialized niches in which erythroid precursors proliferate, differentiate, and enucleate, were first described 50 years ago by analysis of transmission electron micrographs of bone marrow. These hematopoietic subcompartments are composed of erythroblasts surrounding a central macrophage. A hiatus of several decades followed, during which the importance of erythroblastic islands remained unrecognized as erythroid progenitors were shown to possess an autonomous differentiation program with a capacity to complete terminal differentiation in vitro in the presence of erythropoietin but without macrophages. However, as the extent of proliferation, differentiation, and enucleation efficiency documented in vivo could not be recapitulated in vitro, a resurgence of interest in erythroid niches has emerged. We now have an increased molecular understanding of processes operating within erythroid niches, including cell-cell and cell-extracellular matrix adhesion, positive and negative regulatory feedback, and central macrophage function. These features of erythroblast islands represent important contributors to normal erythroid development, as well as altered erythropoiesis found in such diverse diseases as anemia of inflammation and chronic disease, myelodysplasia, thalassemia, and malarial anemia. Coupling of historical, current, and future insights will be essential to understand the tightly regulated production of red cells both in steady state and stress erythropoiesis.
Collapse
|
27
|
Cartron JP, Elion J. Erythroid adhesion molecules in sickle cell disease: effect of hydroxyurea. Transfus Clin Biol 2008; 15:39-50. [PMID: 18515167 DOI: 10.1016/j.tracli.2008.05.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Accepted: 05/05/2008] [Indexed: 01/02/2023]
Abstract
In sickle cell disease, the complex scenario of vaso-occlusive crisis (VOC) typical of this disease is clearly multifactorial and not fully understood. Cell-cell and cell-cell matrix interactions mediated by adhesive molecules present on blood cells and endothelial cells (ECs) are thought to play an important role. Early studies have shown that sickle red blood cells (RBCs) are abnormally adherent to ECs and some of the molecules involved in these interactions have been identified, such as the alpha4beta1 integrin and CD36, exclusively present on stress reticulocytes, and CD47 on mature RBCs. More recently, attention focused on Lu/BCAM, the unique RBC receptor for laminin, and on ICAM-4, a red cell-specific adhesion receptor, which is a ligand for a large repertoire of integrins (alphaLbeta2, alphaMbeta2, alphaxbeta2, alphaVbeta3). The counter-receptors on ECs and the role of plasma proteins forming bridges between blood cells and ECs have been clarified in part. It has also been shown that reticulocytes from SCD patients express higher levels of alpha4beta1 integrin and CD36, and that under hydroxyurea (HU) therapy, both cell adhesion to ECs or extracellular matrix proteins and the levels of these adhesion molecules are reduced. These findings are consistent with the view that enhanced adhesion of blood cells to ECs is largely determined by the membrane expression level of adhesion molecules and could be a crucial factor for triggering or aggravating vaso-occlusion. In SCD patients, membrane expression of Lu/BCAM (and perhaps ICAM-4) is enhanced on RBCs whose adherence to laminin or ECs is also increased. Interestingly, Lu/BCAM- and ICAM-4-mediated adhesion are enhanced by the stress mediator epinephrine through a PKA-dependent pathway initiated by a rise in intracellular cAMP and leading to receptor activation by phosphorylation according to the same signaling pathway. More recently, studies based on quantitative expression analysis of adhesion molecules on RBCs and during erythroid differentiation in patients undergoing HU therapy, surprisingly revealed that Lu/BCAM level was enhanced, although alpha4beta1, CD36 and ICAM-4 (to a lower extent) levels were indeed reduced. CD47 and CD147 expression were also enhanced in HU-treated patients. Based on these findings we suggest that the signalization cascade leading to receptor activation rather than the expression level only of adhesion molecules may be the critical factor regulating cell adhesion, although both mechanisms are not mutually exclusive.
Collapse
Affiliation(s)
- J-P Cartron
- Inserm U665, 6, rue Alexandre-Cabanel, 75015 Paris, France.
| | | |
Collapse
|
28
|
Burton NM, Brady RL. Molecular structure of the extracellular region of Lutheran blood group glycoprotein and location of the laminin binding site. Blood Cells Mol Dis 2008; 40:446-8. [PMID: 18302987 DOI: 10.1016/j.bcmd.2008.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Accepted: 01/04/2008] [Indexed: 10/22/2022]
|