1
|
Ansa‐Addo EA, Pathak P, McCrossan MV, Volpato Rossi I, Abdullahi M, Stratton D, Lange S, Ramirez MI, Inal JM. Monocyte-derived extracellular vesicles, stimulated by Trypanosoma cruzi, enhance cellular invasion in vitro via activated TGF-β1. J Extracell Vesicles 2024; 13:e70014. [PMID: 39611395 PMCID: PMC11605483 DOI: 10.1002/jev2.70014] [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: 05/25/2023] [Revised: 09/06/2024] [Accepted: 10/24/2024] [Indexed: 11/30/2024] Open
Abstract
During cell invasion, large Extracellular Vesicle (lEV) release from host cells was dose-dependently triggered by Trypanosoma cruzi metacyclic trypomastigotes (Mtr). This lEV release was inhibited when IP3-mediated Ca2+ exit from the ER and further Ca2+ entry from plasma membrane channels was blocked, but whilst any store-independent Ca2+ entry (SICE) could continue unabated. That lEV release was equally inhibited if all entry from external sources was blocked by chelation of external Ca2+ points to the major contributor to Mtr-triggered host cell lEV release being IP3/store-mediated Ca2+ release, SICE playing a minor role. Host cell lEVs were released through Mtr interaction with host cell lipid raft domains, integrins, and mechanosensitive ion channels, whereupon [Ca2+]cyt increased (50 to 750 nM) within 15 s. lEV release and cell entry of T. cruzi, which increased up to 30 and 60 mpi, respectively, as well as raised actin depolymerization at 60 mpi, were all reduced by TRPC inhibitor, GsMTx-4. Vesicle release and infection was also reduced with RGD peptide, methyl-β-cyclodextrin, knockdown of calpain and with the calpain inhibitor, calpeptin. Restoration of lEV levels, whether with lEVs from infected or uninfected epithelial cells, did not restore invasion, but supplementation with lEVs from infected monocytes, did. We provide evidence of THP-1 monocyte-derived lEV interaction with Mtr (lipid mixing by R18-dequenching; flow cytometry showing transfer to Mtr of R18 from R18-lEVs and of LAP(TGF-β1). Active, mature TGF-β1 (at 175 pg/×105 in THP-1 lEVs) was detected in concentrated lEV-/cell-free supernatant by western blotting, only after THP-1 lEVs had interacted with Mtr. The TGF-β1 receptor (TβRI) inhibitor, SB-431542, reduced the enhanced cellular invasion due to monocyte-lEVs.
Collapse
Affiliation(s)
- Ephraim A. Ansa‐Addo
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- Pelotonia Institute for Immuno‐Oncology, Department of Internal MedicineThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Paras Pathak
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- Medical Research Council HarwellHarwell Science and Innovation Campus, Genotyping CoreOxfordshireUK
| | | | - Izadora Volpato Rossi
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- School of Life and Medical Sciences, Biosciences Research GroupUniversity of HertfordshireHatfieldUK
- Carlos Chagas InstituteFundacao Oswaldo Cruz, (FIOCRUZ‐PR)CuritibaBrazil
- Postgraduate Program in Cellular and Molecular BiologyFederal University of ParanáCuritibaBrazil
| | - Mahamed Abdullahi
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- National Mycobacterium Reference Service‐South (NMRS‐South) ColindaleLondonUK
| | - Dan Stratton
- School of Life, Health & Chemical SciencesThe Open UniversityMilton KeynesUK
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life SciencesUniversity of WestminsterLondonUK
- University College London, Institute of Women's HealthLondonUK
| | - Marcel I. Ramirez
- Carlos Chagas InstituteFundacao Oswaldo Cruz, (FIOCRUZ‐PR)CuritibaBrazil
| | - Jameel M. Inal
- School of Human Sciences, Cell Communication in Disease PathologyLondon Metropolitan UniversityLondonUK
- School of Life and Medical Sciences, Biosciences Research GroupUniversity of HertfordshireHatfieldUK
| |
Collapse
|
2
|
Li X, Li S, Cheng J, Zhang Y, Zhan A. Deciphering protein-mediated underwater adhesion in an invasive biofouling ascidian: Discovery, validation, and functional mechanism of an interfacial protein. Acta Biomater 2024; 181:146-160. [PMID: 38679406 DOI: 10.1016/j.actbio.2024.04.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 04/11/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Discovering macromolecules and understanding the associated mechanisms involved in underwater adhesion are essential for both studying the fundamental ecology of benthos in aquatic ecosystems and developing biomimetic adhesive materials in industries. Here, we employed quantitative proteomics to assess protein expression variations during the development of the distinct adhesive structure - stolon in the model fouling ascidian, Ciona robusta. We found 16 adhesive protein candidates with increased expression in the stolon, with ascidian adhesive protein 1 (AAP1) being particularly rich in adhesion-related signal peptides, amino acids, and functional domains. Western blot and immunolocalization analyses confirmed the prominent AAP1 signals in the mantle, tunic, stolon, and adhesive footprints, indicating the interfacial role of this protein. Surface coating and atomic force microscopy experiments verified AAP1's adhesion to diverse materials, likely through the specific electrostatic and hydrophobic amino acid interactions with various substrates. In addition, molecular docking calculations indicated the AAP1's potential for cross-linking via hydrogen bonds and salt bridges among Von Willebrand factor type A domains, enhancing its adhesion capability. Altogether, the newly discovered interfacial protein responsible for permanent underwater adhesion, along with the elucidated adhesion mechanisms, are expected to contribute to the development of biomimetic adhesive materials and anti-fouling strategies. STATEMENT OF SIGNIFICANCE: Discovering macromolecules and studying their associated mechanisms involved in underwater adhesion are essential for understanding the fundamental ecology of benthos in aquatic ecosystems and developing innovative bionic adhesive materials in various industries. Using multidisciplinary analytical methods, we identified an interfacial protein - Ascidian Adhesive Protein 1 (AAP1) from the model marine fouling ascidian, Ciona robusta. The interfacial functions of AAP1 are achieved by electrostatic and hydrophobic interactions, and the Von Willebrand factor type A domain-based cross-linking likely enhances AAP1's interfacial adhesion. The identification and validation of the interfacial functions of AAP1, combined with the elucidation of adhesion mechanisms, present a promising target for the development of biomimetic adhesive materials and the formulation of effective anti-fouling strategies.
Collapse
Affiliation(s)
- Xi Li
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Shiguo Li
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Shijingshan District, Beijing 100049, China.
| | - Jiawei Cheng
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Ying Zhang
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Aibin Zhan
- Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, 18 Shuangqing Road, Haidian District, Beijing 100085, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Shijingshan District, Beijing 100049, China.
| |
Collapse
|
3
|
Jorfi S, Ansa-Addo EA, Mariniello K, Warde P, Bin Senian AA, Stratton D, Bax BE, Levene M, Lange S, Inal JM. A Coxsackievirus B1-mediated nonlytic Extracellular Vesicle-to-cell mechanism of virus transmission and its possible control through modulation of EV release. J Gen Virol 2023; 104. [PMID: 37665326 DOI: 10.1099/jgv.0.001884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Abstract
Like most non-enveloped viruses, CVB1 mainly uses cell lysis to spread. Details of a nonlytic virus transmission remain unclear. Extracellular Vesicles (EVs) transfer biomolecules between cells. We show that CVB1 entry into HeLa cells results in apoptosis and release of CVB1-induced 'medium-sized' EVs (CVB1i-mEVs). These mEVs (100-300 nm) harbour CVB1 as shown by immunoblotting with anti-CVB1-antibody; viral capsids were detected by transmission electron microscopy and RT-PCR revealed CVB1 RNA. The percentage of mEVs released from CVB1-infected HeLa cells harbouring virus was estimated from TEM at 34 %. Inhibition of CVB1i-mEV production, with calpeptin or siRNA knockdown of CAPNS1 in HeLa cells limited spread of CVB1 suggesting these vesicles disseminate CVB1 virions to new host cells by a nonlytic EV-to-cell mechanism. This was confirmed by detecting CVB1 virions inside HeLa cells after co-culture with CVB1i-mEVs; EV release may also prevent apoptosis of infected cells whilst spreading apoptosis to secondary sites of infection.
Collapse
Affiliation(s)
- Samireh Jorfi
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
| | - Ephraim Abrokwa Ansa-Addo
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
- Present address: Pelotonia Institute for Immuno-Oncology, The James, Ohio State University, Columbus, OH 43210, USA
| | - Katia Mariniello
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
- Present address: William Harvey Research Institute, Queen Mary, University of London, London, UK
| | - Purva Warde
- Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
| | - Ahmad Asyraf Bin Senian
- Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
- Present address: Clinical Research Centre, Sarawak General Hospital, Kuching, Malaysia
| | - Dan Stratton
- School of Life, Health & Chemical Sciences, The Open University, Milton Keynes MK7 6AE, UK
| | - Bridget E Bax
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Michelle Levene
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London SW17 0RE, UK
| | - Sigrun Lange
- Tissue Architecture and Regeneration Research Group, School of Life Sciences, University of Westminster, 116, New Cavendish St., London, UK
- University College London School of Pharmacy, Brunswick Sq., London, UK
| | - Jameel Malhador Inal
- Cell Communication in Disease Pathology, School of Human Sciences, London Metropolitan University, London N7 8DB, UK
- Biosciences Research Group, School of Life and Medical Sciences, University of Hertfordshire, Hatfield AL10 9EU, UK
| |
Collapse
|
4
|
Microvesicles released constitutively from prostate cancer cells differ biochemically and functionally to stimulated microvesicles released through sublytic C5b-9. Biochem Biophys Res Commun 2015; 460:589-95. [PMID: 25817790 DOI: 10.1016/j.bbrc.2015.03.074] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/12/2015] [Indexed: 01/12/2023]
Abstract
We have classified microvesicles into two subtypes: larger MVs released upon stimulation of prostate cancer cells, sMVs, and smaller cMVs, released constitutively. cMVs are released as part of cell metabolism and sMVs, released at 10-fold higher levels, produced upon activation, including sublytic C5b-9. From electron microscopy, nanosight tracking analysis, dynamic light scattering and flow cytometry, cMVs (194-210 nm in diameter) are smaller than sMVs (333-385 nm). Furthermore, using a Quartz Crystal Microbalance measuring changes in resonant frequency (Δf) that equate to mass deposited on a sensor, an sMV and a cMV are estimated at 0.267 and 0.241 pg, respectively. sMVs carry more calcium and protein, express higher levels of lipid rafts, GPI-anchored CD55 and phosphatidylserine including deposited C5b-9 compared to cMVs. This may allude to biological differences such as increased bound C4BP on sMVs inhibiting complement more effectively.
Collapse
|
5
|
Stratton D, Moore C, Zheng L, Lange S, Inal J. Prostate cancer cells stimulated by calcium-mediated activation of protein kinase C undergo a refractory period before re-releasing calcium-bearing microvesicles. Biochem Biophys Res Commun 2015; 460:511-7. [PMID: 25797625 DOI: 10.1016/j.bbrc.2015.03.061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/11/2015] [Indexed: 10/23/2022]
Abstract
MVs are released in response to several stress agents, in an attempt to prevent continued cellular damage. After an initial stimulus of prostate cancer cells with sublytic C5b-9 and activation of MV release through PKC, cells take at least 20 min to fully recover their ability to microvesiculate. This release of MVs through activation of sublytic C5b-9 was inhibited by the PKC inhibitor bisindoylmaleimide I but not the Rho kinase inhibitor, Y27632. After stimulus there is a rise of 79 nMs(-1) over 11 s, reaching a peak [Ca(2+)]i of 920 nM. The concentration of cytosolic calcium then falls steadily at 2.4 nMs(-1) over 109 s reaching baseline levels (50-100 nM) within 10-15 min. In PC3 cells the rate of release of MVs from stimulated cells also reaches a minimum within 10-15 min. Using fura-2 AM-loaded cells, upon stimulation, cells were found to release MVs with a concentration of intravesicular calcium estimated at ∼ 430 nM.
Collapse
Affiliation(s)
- Dan Stratton
- Cellular and Molecular Immunology Research Centre, School of Human Sciences, London Metropolitan University, London, UK
| | - Colin Moore
- Cellular and Molecular Immunology Research Centre, School of Human Sciences, London Metropolitan University, London, UK
| | - Lei Zheng
- Dept. of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Sigrun Lange
- University College London School of Pharmacy, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Jameel Inal
- Cellular and Molecular Immunology Research Centre, School of Human Sciences, London Metropolitan University, London, UK.
| |
Collapse
|
6
|
Moussavi-Harami SF, Annis DS, Ma W, Berry SM, Coughlin EE, Strotman LN, Maurer LM, Westphall MS, Coon JJ, Mosher DF, Beebe DJ. Characterization of molecules binding to the 70K N-terminal region of fibronectin by IFAST purification coupled with mass spectrometry. J Proteome Res 2013; 12:3393-404. [PMID: 23750785 DOI: 10.1021/pr400225p] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Fibronectin (Fn) is a large glycoprotein present in plasma and extracellular matrix and is important for many processes. Within Fn the 70 kDa N-terminal region (70k-Fn) is involved in cell-mediated Fn assembly, a process that contributes to embryogenesis, development, and platelet thrombus formation. In addition, major human pathogens including Staphlycoccus aureus and Streptococcus pyogenes bind the 70k-Fn region by a novel form of protein-protein interaction called β-zipper formation, facilitating bacterial spread and colonization. Knowledge of blood plasma and platelet proteins that interact with 70k-Fn by β-zipper formation is incomplete. In the current study, we aimed to characterize these proteins through affinity purification. For this affinity purification, we used a novel purification technique termed immiscible filtration assisted by surface tension (IFAST). The foundation of this technology is immiscible phase filtration, using a magnet to draw paramagnetic particle (PMP)-bound analyte through an immiscible barrier (oil or organic solvent) that separates an aqueous sample from an aqueous eluting buffer. The immiscible barrier functions to remove unbound proteins via exclusion rather than dilutive washing used in traditional isolation methods. We identified 31 interactors from plasma, of which only seven were previously known to interact with Fn. Furthermore, five proteins were identified to interact with 70k-Fn from platelet lysate, of which one was previously known. These results demonstrate that IFAST offers advantages for proteomic studies of interacting molecules in that the technique requires small sample volumes, can be done with high enough throughput to sample multiple interaction conditions, and is amenable to exploratory mass spectrometric and confirmatory immuno-blotting read-outs.
Collapse
|
7
|
Abstract
SUMMARY A trispanning orphan receptor (TOR) has been described in Schistosoma haematobium and S. mansoni. Here we report the complete molecular organization of the S. mansoni TOR gene, also known as SmCRIT (complement C2 receptor inhibitor trispanning). The SmTOR gene consists of 4 exons and 3 introns as shown by cloning the single exons from S. mansoni genomic DNA and the corresponding cDNA from the larval stage (cercaria) and the adult worm. The SmTOR ORF consists of 1260 bp and is longer than previously reported, with a fourth trans-membrane domain (proposed new name: Tetraspanning Orphan Receptor) and with, however, an unchanged C2-binding domain on the extracellular domain 1 (ed1). This domain differs in S. japonicum. A protein at the approximate expected molecular weight (55 kDa) was detected in adult worm extracts with polyclonal and monoclonal antibodies, and was found to be expressed on the tegumental surface of cercariae.
Collapse
|
8
|
Cestari IDS, Evans-Osses I, Freitas JC, Inal JM, Ramirez MI. Complement C2 receptor inhibitor trispanning confers an increased ability to resist complement-mediated lysis in Trypanosoma cruzi. J Infect Dis 2008; 198:1276-83. [PMID: 18781865 DOI: 10.1086/592167] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The ability to resist complement differs between the Y and Colombiana Trypanosoma cruzi strains. We found that the Y strain of T. cruzi was more able to resist the classical and lectin pathways of complement activation than the Colombiana strain. The complement C2 receptor inhibitor trispanning gene (CRIT) is highly conserved in both strains. At the protein level, CRIT is expressed only in stationary-phase epimastigotes of the Y but not the Colombiana strain and is expressed in infectious metacyclic trypomastigotes of both strains. Y strain epimastigotes with an overexpressed CRIT gene (pTEX-CRIT) had higher survival in normal human serum (NHS). Overexpression of the Y strain CRIT gene in Colombiana epimastigote forms increased the parasite's resistance to lysis mediated by the classical and lectin pathways but not to lysis mediated by alternative pathways. CRIT involvement on the parasite surface was confirmed by showing that the lytic activity of NHS against epimastigotes could be restored by adding excess C2.
Collapse
Affiliation(s)
- Igor Dos S Cestari
- Departamento de Bioquímica e Biologia Molecular, Instituto Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | | | | | | | | |
Collapse
|
9
|
Abstract
Inter-alpha-trypsin inhibitor family proteins are mainly detected in plasma and urine and comprise the common light chain bikunin and at least 6 closely related heavy chains. The bikunin moiety exhibits protease inhibitory activity and has been studied extensively; however, the heavy chains have been largely overlooked. Recent studies clearly indicate that the heavy chain moieties have important biological functions either in association with or independent of bikunin. Because the heavy chains comprise the main part of the protein structure of this family, it is important to understand their functions. This review summarizes the domain structural features of heavy chains, the heavy chain-interacting molecules identified thus far, and the association of heavy chains with diseases to encourage the discovery of novel heavy chains-interacting molecules and to gain a deeper insight into their functions.
Collapse
Affiliation(s)
- Lisheng Zhuo
- Institute for Molecular Science of Medicine, Aichi Medical University, Nagakute, Aichi, Japan
| | | |
Collapse
|
10
|
Garantziotis S, Hollingsworth JW, Ghanayem RB, Timberlake S, Zhuo L, Kimata K, Schwartz DA. Inter-alpha-trypsin inhibitor attenuates complement activation and complement-induced lung injury. THE JOURNAL OF IMMUNOLOGY 2007; 179:4187-92. [PMID: 17785858 DOI: 10.4049/jimmunol.179.6.4187] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Complement activation is a central component of inflammation and sepsis and can lead to significant tissue injury. Complement factors are serum proteins that work through a cascade of proteolytic reactions to amplify proinflammatory signals. Inter-alpha-trypsin inhibitor (IaI) is an abundant serum protease inhibitor that contains potential complement-binding domains, and has been shown to improve survival in animal sepsis models. We hypothesized that IaI can bind complement and inhibit complement activation, thus ameliorating complement-dependent inflammation. We evaluated this hypothesis with in vitro complement activation assays and in vivo in a murine model of complement-dependent lung injury. We found that IaI inhibited complement activation through the classical and alternative pathways, inhibited complement-dependent phagocytosis in vitro, and reduced complement-dependent lung injury in vivo. This novel function of IaI provides a mechanistic explanation for its observed salutary effects in sepsis and opens new possibilities for its use as a treatment agent in inflammatory diseases.
Collapse
|
11
|
Oda-Ishii I, Di Gregorio A. Lineage-independent mosaic expression and regulation of theCiona multidomgene in the ancestral notochord. Dev Dyn 2007; 236:1806-19. [PMID: 17576134 DOI: 10.1002/dvdy.21213] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The transcription factor Ciona Brachyury (Ci-Bra) plays an essential role in notochord development in the ascidian Ciona intestinalis. We characterized a putative Ci-Bra target gene, which we named Ci-multidom, and analyzed in detail its expression pattern in normal embryos and in embryos where Ci-Bra was misexpressed. Ci-multidom encodes a novel protein, which contains eight CCP domains and a partial VWFA domain. We show that an EGFP-multidom fusion protein localizes preferentially to the endoplasmic reticulum (ER), and is excluded from the nucleus. In situ hybridization experiments demonstrate that Ci-multidom is expressed in the notochord and in the anterior neural boundary (ANB). We found that the expression in the ANB is fully recapitulated by an enhancer element located upstream of Ci-multidom. By means of misexpression experiments, we provide evidence that Ci-Bra controls transcription of Ci-multidom in the notochord; however, while Ci-Bra is homogeneously expressed throughout this structure, Ci-multidom is transcribed at detectable levels only in a random subset of notochord cells. The number of notochord cells expressing Ci-multidom varies among different embryos and is independent of developmental stage, lineage, and position along the anterior-posterior axis. These results suggest that despite its morphological simplicity and invariant cell-lineage, the ancestral notochord is a mosaic of cells in which the gene cascade downstream of Brachyury is differentially modulated.
Collapse
Affiliation(s)
- Izumi Oda-Ishii
- Department of Cell and Developmental Biology, Weill Medical College of Cornell University, New York, New York 10021, USA
| | | |
Collapse
|
12
|
Hui KM, Magnadóttir B, Schifferli JA, Inal JM. CRIT peptide interacts with factor B and interferes with alternative pathway activation. Biochem Biophys Res Commun 2006; 344:308-14. [PMID: 16600177 DOI: 10.1016/j.bbrc.2006.03.101] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Accepted: 03/17/2006] [Indexed: 01/11/2023]
Abstract
Complement C2 receptor inhibitor trispanning (CRIT) inhibits the classical pathway (CP) C3 convertase formation by competing with C4b for the binding of C2. The C-terminal 11-amino-acid of the first CRIT-extracellular domain (CRIT-H17) has a strong homology with a sequence in the C4beta chain, which is responsible for the binding of C2. Since the CP and alternative pathway (AP) C3 convertases have many functional and structural similarities, we further investigated the effects of CRIT-H17 on the AP. The factor D-mediated cleavage of factor B (FB) was blocked by CRIT-H17. By ELISA and immunoblot, CRIT-H17 was shown to bind FB. CRIT-H17 had no decay activity on the C3bBb complex as compared to decay-accelerating factor. Binding of CRIT-H17 to FB did not interfere with the assembly of C3bB complex. In a haemolytic assay using C2-deficient serum, CRIT-H17 interfered with AP complement activation.
Collapse
Affiliation(s)
- Kwok-Min Hui
- University Hospital Basel, Immunonephrology, Department of Research, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | | | | | | |
Collapse
|
13
|
Inal J, Miot S, Schifferli JA. The complement inhibitor, CRIT, undergoes clathrin-dependent endocytosis. Exp Cell Res 2005; 310:54-65. [PMID: 16112669 DOI: 10.1016/j.yexcr.2005.07.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2005] [Revised: 07/07/2005] [Accepted: 07/07/2005] [Indexed: 01/22/2023]
Abstract
Complement C2 receptor inhibitor trispanning (CRIT) is a receptor for the second component of complement and is found in various tissues and hemopoietic cells. On binding to CRIT, C2 cannot be activated to potentially form a variant-C3 convertase as it is rendered non-cleavable by C1s. CRIT thus limits the amount of C3 convertase formed on the cell surface. In this study we have shown, using flow cytometry and immunofluorescence microscopy, that human CRIT undergoes endocytosis from the plasma membrane. The endocytosis, possibly ligand mediated, occurs via clathrin-coated pits as it can be inhibited by prior incubation of cells in hypertonic medium or with chlorpromazine, at 37 degrees C. However, inhibition of endocytosis was not possible after treatment with nystatin, or filipin, inhibitors of caveolae/raft-dependent endocytosis. In the presence of C2 alone, CRIT associates with the adapter protein, beta-arrestin-2, and whether in association with C2 or not, then appears in the perinuclear region, but does not appear to be translocated into the nucleus. Apart from the C3aR and C5aR that internalize the anaphylatoxic peptides, this is the first report of the internalization via the clathrin pathway of a receptor for a complement serum protein.
Collapse
Affiliation(s)
- Jameel Inal
- University Hospital Basel, Immunonephrology, Department of Research, 4031 Basel, Switzerland.
| | | | | |
Collapse
|