1
|
Zhu H, Ma Q, Li R, Fu M, Lu P. Blood group-related antigen Le y on human platelets and its involvement in platelet aggregation via a possible interaction with CD61. Transfusion 2020; 60:2130-2138. [PMID: 32757229 DOI: 10.1111/trf.15981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND The Ley antigen is a carbohydrate chain belonging to the ABH-Lewis blood group family. Ley has been reported to be present on red blood cells (RBCs) and granulocytes, but its distribution and function in platelets remain unknown. There are a variety of glycoproteins on platelets, which may carry the Ley antigen. This study aims to investigate the expression pattern and the function of Ley in human platelets. STUDY DESIGN AND METHODS Flow cytometry, Western blot, and immunofluorescence assays were performed to determine Ley expression on human platelets. ADP (1.25-10 μM) and thrombin (0.05-1 IU/mL) were used to activate platelets in the presence or absence of prostaglandin E1 (PGE1) and the Ley expression was evaluated again by flow cytometry. Blockade was performed with an anti-Ley monoclonal antibody to verify the role of this epitope in platelet function. Finally, coimmunoprecipitation was performed to identify glycoproteins associated with Ley . RESULTS Ley was expressed on human platelets independent of ABO blood type. Ley expression was decreased in a dose-dependent manner after activation with either ADP or thrombin, and this effect could be partially reversed by PGE1. Anti- Ley mAb treatment increased alpha-granule release and neutralized the inhibitory effect of the anti-CD61 antibody on platelet aggregation. In addition, Ley was proven to interact and colocalize with CD61. CONCLUSIONS These results demonstrate nondifferential expression of Ley in platelets of different ABO blood types and suggest the involvement of Ley in platelet function, possibly via interaction with CD61.
Collapse
Affiliation(s)
- Huijun Zhu
- Immunohematology Platelet Lab, Shanghai Blood Center, Shanghai, China
| | - Qinqin Ma
- Immunohematology Platelet Lab, Shanghai Blood Center, Shanghai, China
| | - Ruishu Li
- Immunohematology Platelet Lab, Shanghai Blood Center, Shanghai, China
| | - Min Fu
- Immunohematology Platelet Lab, Shanghai Blood Center, Shanghai, China
| | - Ping Lu
- Immunohematology Platelet Lab, Shanghai Blood Center, Shanghai, China
| |
Collapse
|
2
|
Li C, Ouyang N, Wang X, Liang A, Mo Y, Li S, Qiu J, Fang G, Fu Y, Song B, Chen Z, Ding Y. Association between the ABO blood group and primary knee osteoarthritis: A case-control study. J Orthop Translat 2020; 21:129-135. [PMID: 32309138 PMCID: PMC7152790 DOI: 10.1016/j.jot.2019.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/03/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Recent studies have suggested association between the ABO blood group and inflammation, which was a crucial pathological process of primary knee osteoarthritis. The aim of this study was to investigate the association between the ABO blood group and primary knee osteoarthritis and the severity of primary knee osteoarthritis evaluated by the Kellgren/Lawrence score, as well as the histopathologic association in a subgroup of patients. METHODS We performed a retrospective review of patients with primary knee osteoarthritis that served as the case group and a random sampling of healthy blood donors that served as the control group. The severity of knee osteoarthritis at the first outpatient visit was evaluated by the Kellgren/Lawrence scoring system. Further study was performed to investigate the expression of blood group antigens in synovial tissue of the knee in both cases and controls. RESULTS A total of 1126 cases and 30299 controls were involved. The proportion of AB blood group was higher in the case group than in the control group (9.7% vs. 7.8%), and logistic regression revealed that the AB blood group was a risk factor of primary knee osteoarthritis (P = 0.025 and 0.048 for univariate and multivariate analysis, respectively), independent of age (P = 0.973) and sex (P = 0.520). Patients of the blood group AB had a higher Kellgren/Lawrence score (P = 0.017). The immunohistochemical study indicated association between LeY antigen and primary knee osteoarthritis (P = 0.029). CONCLUSIONS This study suggested that the blood group AB was associated with primary knee osteoarthritis, as well as its radiological severity. Further study indicated that LeY antigen, which was related to the blood group, was associated with primary knee osteoarthritis. TRANSLATIONAL POTENTIAL OF THIS ARTICLE This study revealed that blood group AB and LeY antigen was associated with primary knee osteoarthritis, which shed new light on the nature of osteoarthritis, and the development of novel therapy for osteoarthritis.
Collapse
Key Words
- ACR, American College of Rheumatology
- ANOVA, analysis of variance
- Blood group
- CI, confidence interval
- FUT, fucosyltransferase
- HR, hazard ratio
- IHC, immunohistochemistry
- IRB, institutional review board
- K/L, Kellgren/Lawrence
- Kellgren/Lawrence
- Knee
- LeY antigen
- OA, osteoarthritis
- Osteoarthritis
- ST, synovial tissue
Collapse
Affiliation(s)
- Changchuan Li
- Department of Orthopaedic Surgery, Sun Yat-sen Memorial Hospital, China
| | - Nengtai Ouyang
- Department of Pathology, Sun Yat-sen Memorial Hospital, China
| | - Xiuju Wang
- Department of Blood Transfusion, Sun Yat-sen Memorial Hospital, China
| | - Anjing Liang
- Department of Orthopaedic Surgery, Sun Yat-sen Memorial Hospital, China
| | - Yingqian Mo
- Department of Rheumatology and Immunology, Sun Yat-sen Memorial Hospital, China
| | - Shixun Li
- Department of Orthopaedic Surgery, Sun Yat-sen Memorial Hospital, China
| | - Junxiong Qiu
- Department of Orthopaedic Surgery, Sun Yat-sen Memorial Hospital, China
| | - Guibin Fang
- Department of Orthopaedic Surgery, Sun Yat-sen Memorial Hospital, China
| | - Yuan Fu
- Department of Orthopaedic Surgery, Sun Yat-sen Memorial Hospital, China
| | - Bin Song
- Department of Orthopaedic Surgery, Sun Yat-sen Memorial Hospital, China
| | - Zhong Chen
- Department of Orthopaedic Surgery, Sun Yat-sen Memorial Hospital, China
| | - Yue Ding
- Department of Orthopaedic Surgery, Sun Yat-sen Memorial Hospital, China
| |
Collapse
|
3
|
Du Y, Lu C, Morgan RL, Stinson WA, Campbell PL, Cealey E, Fu W, Lepore NJ, Hervoso JL, Cui H, Urquhart AG, Lawton JN, Chung KC, Fox DA, Amin MA. Angiogenic and Arthritogenic Properties of the Soluble Form of CD13. THE JOURNAL OF IMMUNOLOGY 2019; 203:360-369. [PMID: 31189572 DOI: 10.4049/jimmunol.1801276] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 05/15/2019] [Indexed: 11/19/2022]
Abstract
Aminopeptidase N/CD13 is expressed by fibroblast-like synoviocytes (FLS) and monocytes (MNs) in inflamed human synovial tissue (ST). This study examined the role of soluble CD13 (sCD13) in angiogenesis, MN migration, phosphorylation of signaling molecules, and induction of arthritis. The contribution of sCD13 was examined in angiogenesis and MN migration using sCD13 and CD13-depleted rheumatoid arthritis (RA) synovial fluids (SFs). An enzymatically inactive mutant CD13 and intact wild-type (WT) CD13 were used to determine whether its enzymatic activity contributes to the arthritis-related functions. CD13-induced phosphorylation of signaling molecules was determined by Western blotting. The effect of sCD13 on cytokine secretion from RA ST and RA FLS was evaluated. sCD13 was injected into C57BL/6 mouse knees to assess its arthritogenicity. sCD13 induced angiogenesis and was a potent chemoattractant for MNs and U937 cells. Inhibitors of Erk1/2, Src, NF-κB, Jnk, and pertussis toxin, a G protein-coupled receptor inhibitor, decreased sCD13-stimulated chemotaxis. CD13-depleted RA SF induced significantly less MN migration than sham-depleted SF, and addition of mutant or WT CD13 to CD13-depleted RA SF equally restored MN migration. sCD13 and recombinant WT or mutant CD13 had similar effects on signaling molecule phosphorylation, indicating that the enzymatic activity of CD13 had no role in these functions. CD13 increased the expression of proinflammatory cytokines by RA FLS, and a CD13 neutralizing Ab inhibited cytokine secretion from RA ST organ culture. Mouse knee joints injected with CD13 exhibited increased circumference and proinflammatory mediator expression. These data support the concept that sCD13 plays a pivotal role in RA and acute inflammatory arthritis.
Collapse
Affiliation(s)
- Yuxuan Du
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109.,Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.,National Center for Clinical Laboratories/Beijing Engineering Research Center of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, Beijing 100730, China
| | - Chenyang Lu
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Rachel L Morgan
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - William A Stinson
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Phillip L Campbell
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Ellen Cealey
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Wenyi Fu
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109.,Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, China; and
| | - Nicholas J Lepore
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Jonatan L Hervoso
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Huadong Cui
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109.,Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110022, China; and
| | - Andrew G Urquhart
- Department of Orthopaedic Surgery, University of Michigan Health System, A. Alfred Taubman Health Care Center, Ann Arbor, MI 48109
| | - Jeffrey N Lawton
- Department of Orthopaedic Surgery, University of Michigan Health System, A. Alfred Taubman Health Care Center, Ann Arbor, MI 48109
| | - Kevin C Chung
- Department of Orthopaedic Surgery, University of Michigan Health System, A. Alfred Taubman Health Care Center, Ann Arbor, MI 48109
| | - David A Fox
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109;
| | - Mohammad A Amin
- Division of Rheumatology, Clinical Autoimmunity Center of Excellence, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
4
|
Erden A, Batu ED, Armagan B, Sönmez HE, Sarı A, Demir S, Bilgin E, Fırat E, Kılıc L, Bilginer Y, Karadag O, Kiraz S, Kalyoncu U. Blood group 'A' may have a possible modifier effect on familial Mediterranean fever and blood group '0' may be associated with colchicine resistance. Biomark Med 2018; 12:565-572. [PMID: 29873519 DOI: 10.2217/bmm-2017-0344] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Aim/purpose: Our aim was to investigate the association between blood groups and colchicine resistance in familial Mediterranean fever (FMF) patients. METHODS This is a single-center, cross-sectional study. Between January and December 2016, 385 FMF patients were assessed by the Adult and Pediatric Rheumatology outpatient clinics and 297 patients had blood groups (ABO and Rh) results. The patients were grouped into two groups: colchicine-responsive patients (Group CR) and colchicine-unresponsive patients (Group CUR). RESULTS Patients with blood group A had 1.5-fold higher FMF compared with non-A blood group (OR: 1.50 [95% CI: 1.11-1.87]), particularly having a Rh (+) blood group (OR: 1.47 [95% CI: 1.13-1.91]). Furthermore, patients with blood group A had a better response to colchicine treatment than non-A blood group; (OR: 2.21 [95% CI: 1.15-4.27]). Patients with blood group O were prominently associated with colchicine resistance. CONCLUSION ABO blood phenogroups may be used in combination with other risk factors to identify FMF patients and patients at high risk for colchicine resistance.
Collapse
Affiliation(s)
- Abdulsamet Erden
- Division of Rheumatology, Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Ezgi Deniz Batu
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Berkan Armagan
- Division of Rheumatology, Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Hafize Emine Sönmez
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Alper Sarı
- Division of Rheumatology, Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Selcan Demir
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Emre Bilgin
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Esra Fırat
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Levent Kılıc
- Division of Rheumatology, Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Yelda Bilginer
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Omer Karadag
- Division of Rheumatology, Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Sedat Kiraz
- Division of Rheumatology, Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Umut Kalyoncu
- Division of Rheumatology, Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
5
|
O'Brien MJ, Shu Q, Stinson WA, Tsou PS, Ruth JH, Isozaki T, Campbell PL, Ohara RA, Koch AE, Fox DA, Amin MA. A unique role for galectin-9 in angiogenesis and inflammatory arthritis. Arthritis Res Ther 2018; 20:31. [PMID: 29433546 PMCID: PMC5809993 DOI: 10.1186/s13075-018-1519-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 01/17/2018] [Indexed: 01/25/2023] Open
Abstract
Background Galectin-9 (Gal-9) is a mammalian lectin secreted by endothelial cells that is highly expressed in rheumatoid arthritis synovial tissues and synovial fluid. Roles have been proposed for galectins in the regulation of inflammation and angiogenesis. Therefore, we examined the contribution of Gal-9 to angiogenesis and inflammation in arthritis. Methods To determine the role of Gal-9 in angiogenesis, we performed human dermal microvascular endothelial cell (HMVEC) chemotaxis, Matrigel tube formation, and mouse Matrigel plug angiogenesis assays. We also examined the role of signaling molecules in Gal-9-induced angiogenesis by using signaling inhibitors and small interfering RNA (siRNA). We performed monocyte (MN) migration assays in a modified Boyden chamber and assessed the arthritogenicity of Gal-9 by injecting Gal-9 into mouse knees. Results Gal-9 significantly increased HMVEC migration, which was decreased by inhibitors of extracellular signal-regulating kinases 1/2 (Erk1/2), p38, Janus kinase (Jnk), and phosphatidylinositol 3-kinase. Gal-9 HMVEC-induced tube formation was reduced by Erk1/2, p38, and Jnk inhibitors, and this was confirmed by siRNA knockdown. In mouse Matrigel plug assays, plugs containing Gal-9 induced significantly higher angiogenesis, which was attenuated by a Jnk inhibitor. Gal-9 also induced MN migration, and there was a marked increase in MN ingress when C57BL/6 mouse knees were injected with Gal-9 compared with the control, pointing to a proinflammatory role for Gal-9. Conclusions Gal-9 mediates angiogenesis, increases MN migration in vitro, and induces acute inflammatory arthritis in mice, suggesting a novel role for Gal-9 in angiogenesis, joint inflammation, and possibly other inflammatory diseases.
Collapse
Affiliation(s)
- Martin J O'Brien
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Qiang Shu
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA.,Shenzhen Research Institute of Shandong University, Shenzhen, China.,Rheumatology Department, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - W Alexander Stinson
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Pei-Suen Tsou
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Jeffrey H Ruth
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Takeo Isozaki
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Phillip L Campbell
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Ray A Ohara
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - Alisa E Koch
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA.,Department of Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - David A Fox
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA
| | - M Asif Amin
- Division of Rheumatology and Clinical Autoimmunity Center of Excellence, Department of Internal Medicine, University of Michigan Medical School, 4368 BSRB, 109 Zina Pitcher Drive, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
6
|
Monocytic thrombomodulin promotes cell adhesion through interacting with its ligand, Lewis y. Immunol Cell Biol 2016; 95:372-379. [PMID: 27808085 PMCID: PMC5415637 DOI: 10.1038/icb.2016.110] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 10/18/2016] [Accepted: 10/18/2016] [Indexed: 12/12/2022]
Abstract
The leukocyte adhesion cascade involves multiple events that efficiently localize circulating leukocytes into the injured sites to mediate inflammatory responses. From rolling to firm adhesion, the interactions between adhesion molecules have pivotal roles in increasing the avidity of leukocytes to endothelial cells. Thrombomodulin (TM), an essential anticoagulant protein in the vasculature, is also expressed on leukocytes. We previously demonstrated that Lewisy (Ley), a specific ligand of TM, is upregulated in inflamed endothelium and is involved in leukocyte adhesion. The current study aimed to investigate whether leukocyte-expressed TM promotes cell adhesion by interacting with Ley. Using human monocytic THP-1 cells as an in vitro cell model, we showed that TM increases THP-1 cell adhesion to inflamed endothelium as well as to Ley-immobilized surface. When THP-1 adhered to activated endothelium and Ley-immobilized surface, the TM distribution became polarized. Addition of soluble Ley to a suspension of THP-1 cells with TM expression triggered an increase in the level of phosphorylated p38 mitogen-activated protein kinase (MAPK), which enabled THP-1 to adhere firmly to intercellular adhesion molecule (ICAM)-1 by activating β2 integrins. In vivo, macrophage infiltration and neointima formation following arterial ligation-induced vascular injury were higher in wild-type TM (TMflox/flox) than in myeloid-specific TM-deficient (LysMcre/TMflox/flox) mice. Taken together, these results suggest a novel function for TM as an adhesion molecule in monocytes, where it enhances cell adhesion by binding Ley, leading to β2 integrin activation via p38 MAPK.
Collapse
|
7
|
Neofunctionalization of the Sec1 α1,2fucosyltransferase paralogue in leporids contributes to glycan polymorphism and resistance to rabbit hemorrhagic disease virus. PLoS Pathog 2015; 11:e1004759. [PMID: 25875017 PMCID: PMC4398370 DOI: 10.1371/journal.ppat.1004759] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/21/2015] [Indexed: 12/13/2022] Open
Abstract
RHDV (rabbit hemorrhagic disease virus), a virulent calicivirus, causes high mortalities in European rabbit populations (Oryctolagus cuniculus). It uses α1,2fucosylated glycans, histo-blood group antigens (HBGAs), as attachment factors, with their absence or low expression generating resistance to the disease. Synthesis of these glycans requires an α1,2fucosyltransferase. In mammals, there are three closely located α1,2fucosyltransferase genes rSec1, rFut2 and rFut1 that arose through two rounds of duplications. In most mammalian species, Sec1 has clearly become a pseudogene. Yet, in leporids, it does not suffer gross alterations, although we previously observed that rabbit Sec1 variants present either low or no activity. Still, a low activity rSec1 allele correlated with survival to an RHDV outbreak. We now confirm the association between the α1,2fucosyltransferase loci and survival. In addition, we show that rabbits express homogenous rFut1 and rFut2 levels in the small intestine. Comparison of rFut1 and rFut2 activity showed that type 2 A, B and H antigens recognized by RHDV strains were mainly synthesized by rFut1, and all rFut1 variants detected in wild animals were equally active. Interestingly, rSec1 RNA levels were highly variable between individuals and high expression was associated with low binding of RHDV strains to the mucosa. Co-transfection of rFut1 and rSec1 caused a decrease in rFut1-generated RHDV binding sites, indicating that in rabbits, the catalytically inactive rSec1 protein acts as a dominant-negative of rFut1. Consistent with neofunctionalization of Sec1 in leporids, gene conversion analysis showed extensive homogenization between Sec1 and Fut2 in leporids, at variance with its limited degree in other mammals. Gene conversion additionally involving Fut1 was also observed at the C-terminus. Thus, in leporids, unlike in most other mammals where it became extinct, Sec1 evolved a new function with a dominant-negative effect on rFut1, contributing to fucosylated glycan diversity, and allowing herd protection from pathogens such as RHDV. There are three members of the α1,2fucosyltransferases gene family in mammalian genomes, Fut1, Fut2 and Sec1. The encoded fucosyltransferases are key enzymes for the synthesis of glycans that can be used as ligands by pathogens. However, the polymorphism of expression of these fucosylated glycans on epithelial cell types contributes to protection at the species level. In most mammalian species Sec1 is a pseudogene and in humans, genetic variation of α1,2fucosylated glycans is provided by FUT2 polymorphisms. Rabbit haemorrhagic disease virus (RHDV) uses α1,2fucosylated glycans as attachment factors. It induces an acute disease with very high mortalities in rabbit populations. We now confirm an association between genetic markers in the rabbit Sec1-Fut2 genomic region and survival to RHDV. We show that the Fut1 gene is the main contributor to the synthesis of RHDV binding sites although individual variation is not achieved by Fut1 polymorphisms but by variation in levels of Sec1 transcription. The Sec1 protein acting as a dominant-negative of Fut1, high Sec1 expression leads to a decreased number of RHDV binding sites. Thus, unlike in other mammals, in rabbits Sec1 underwent neofunctionalization. It contributes to generate diversity of fucosylated glycans, a key mechanism for escaping pathogens such as RHDV.
Collapse
|
8
|
Isozaki T, Ruth JH, Amin MA, Campbell PL, Tsou PS, Ha CM, Haines GK, Edhayan G, Koch AE. Fucosyltransferase 1 mediates angiogenesis, cell adhesion and rheumatoid arthritis synovial tissue fibroblast proliferation. Arthritis Res Ther 2014; 16:R28. [PMID: 24467809 PMCID: PMC3978694 DOI: 10.1186/ar4456] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Accepted: 01/13/2014] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION We previously reported that sialyl Lewis(y), synthesized by fucosyltransferases, is involved in angiogenesis. Fucosyltransferase 1 (fut1) is an α(1,2)-fucosyltransferase responsible for synthesis of the H blood group and Lewis(y) antigens. However, the angiogenic involvement of fut 1 in the pathogenesis of rheumatoid arthritis synovial tissue (RA ST) has not been clearly defined. METHODS Assay of α(1,2)-linked fucosylated proteins in RA was performed by enzyme-linked lectin assay. Fut1 expression was determined in RA ST samples by immunohistological staining. We performed angiogenic Matrigel assays using a co-culture system of human dermal microvascular endothelial cells (HMVECs) and fut1 small interfering RNA (siRNA) transfected RA synovial fibroblasts. To determine if fut1 played a role in leukocyte retention and cell proliferation in the RA synovium, myeloid THP-1 cell adhesion assays and fut1 siRNA transfected RA synovial fibroblast proliferation assays were performed. RESULTS Total α(1,2)-linked fucosylated proteins in RA ST were significantly higher compared to normal (NL) ST. Fut1 expression on RA ST lining cells positively correlated with ST inflammation. HMVECs from a co-culture system with fut1 siRNA transfected RA synovial fibroblasts exhibited decreased endothelial cell tube formation compared to control siRNA transfected RA synovial fibroblasts. Fut1 siRNA also inhibited myeloid THP-1 adhesion to RA synovial fibroblasts and RA synovial fibroblast proliferation. CONCLUSIONS These data show that α(1,2)-linked fucosylated proteins are upregulated in RA ST compared to NL ST. We also show that fut1 in RA synovial fibroblasts is important in angiogenesis, leukocyte-synovial fibroblast adhesion, and synovial fibroblast proliferation, all key processes in the pathogenesis of RA.
Collapse
Affiliation(s)
- Takeo Isozaki
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Currently Department of Internal Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Jeffrey H Ruth
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mohammad A Amin
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Phillip L Campbell
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Pei-Suen Tsou
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Christine M Ha
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | | | - Gautam Edhayan
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Alisa E Koch
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- VA Medical Service, Department of Veterans Affairs Medical Center, Ann Arbor, MI 48108, USA
| |
Collapse
|
9
|
Lin WL, Chang CF, Shi CS, Shi GY, Wu HL. Recombinant Lectin-Like Domain of Thrombomodulin Suppresses Vascular Inflammation by Reducing Leukocyte Recruitment via Interacting With Lewis Y on Endothelial Cells. Arterioscler Thromb Vasc Biol 2013; 33:2366-73. [DOI: 10.1161/atvbaha.113.301221] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Wei-Ling Lin
- From the Department of Biochemistry and Molecular Biology (W.-L.L., C.-S.S., G.-Y.S., H.-L.W.); Institute of Basic Medical Sciences (W.-L.L.); Cardiovascular Research Center (W.-L.L., G.-Y.S., H.-L.W.); Center of Bioscience and Biotechnology (H.-L.W.); and Department of Medical Laboratory Science and Biotechnology (C.-F.C.), College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chuan-Fa Chang
- From the Department of Biochemistry and Molecular Biology (W.-L.L., C.-S.S., G.-Y.S., H.-L.W.); Institute of Basic Medical Sciences (W.-L.L.); Cardiovascular Research Center (W.-L.L., G.-Y.S., H.-L.W.); Center of Bioscience and Biotechnology (H.-L.W.); and Department of Medical Laboratory Science and Biotechnology (C.-F.C.), College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chung-Sheng Shi
- From the Department of Biochemistry and Molecular Biology (W.-L.L., C.-S.S., G.-Y.S., H.-L.W.); Institute of Basic Medical Sciences (W.-L.L.); Cardiovascular Research Center (W.-L.L., G.-Y.S., H.-L.W.); Center of Bioscience and Biotechnology (H.-L.W.); and Department of Medical Laboratory Science and Biotechnology (C.-F.C.), College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Guey-Yueh Shi
- From the Department of Biochemistry and Molecular Biology (W.-L.L., C.-S.S., G.-Y.S., H.-L.W.); Institute of Basic Medical Sciences (W.-L.L.); Cardiovascular Research Center (W.-L.L., G.-Y.S., H.-L.W.); Center of Bioscience and Biotechnology (H.-L.W.); and Department of Medical Laboratory Science and Biotechnology (C.-F.C.), College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Hua-Lin Wu
- From the Department of Biochemistry and Molecular Biology (W.-L.L., C.-S.S., G.-Y.S., H.-L.W.); Institute of Basic Medical Sciences (W.-L.L.); Cardiovascular Research Center (W.-L.L., G.-Y.S., H.-L.W.); Center of Bioscience and Biotechnology (H.-L.W.); and Department of Medical Laboratory Science and Biotechnology (C.-F.C.), College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| |
Collapse
|
10
|
Rabquer BJ, Hou Y, Ruth JH, Luo W, Eitzman DT, Koch AE, Amin MA. H-2g, a glucose analog of blood group H antigen, mediates monocyte recruitment in vitro and in vivo via IL-8/CXCL8. Open Access Rheumatol 2012; 4:93-98. [PMID: 27790017 PMCID: PMC5045105 DOI: 10.2147/oarrr.s36163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective Monocyte (MN) recruitment is an essential inflammatory component of many autoimmune diseases, including rheumatoid arthritis (RA). In this study we investigated the ability of 2-fucosyllactose (H-2g), a glucose analog of blood group H antigen to induce MN migration in vivo and determined if H-2g-induced interleukin-8 (IL-8/CXCL8) plays a role in MN ingress in RA. Methods Sponge granuloma and intravital microscopy assays were performed to examine H-2g-induced in vivo MN migration and rolling, respectively. MNs were stimulated with H-2g, and the production of IL-8/CXCL8 was assessed by enzyme-linked immunosorbent assay and quantitative polymerase chain reaction. Lastly, in vitro MN migration assays and an in vivo RA synovial tissue severe combined immunodeficiency mouse model were used to determine the role of IL-8/CXCL8 in H-2g-induced MN migration. Results In vivo, H-2g induced significantly greater MN migration compared to phosphate buffered saline. Intravital microscopy revealed that H-2g mediates MN migration in vivo by inducing MN rolling. In addition, H-2g induced MN production of IL-8/CXCL8, a process that was dependent on Src kinase. Moreover, we found that H-2g mediated MN migration in vitro, and in vivo migration was inhibited by a neutralizing anti-IL-8/CXCL8 antibody. Conclusion These findings suggest that H-2g mediates MN recruitment in vitro and in vivo (in part) via IL-8/CXCL8.
Collapse
Affiliation(s)
- Bradley J Rabquer
- University of Michigan Medical School, Department of Internal Medicine, Ann Arbor, MI, USA; Albion College, Biology Department, Albion, MI, USA
| | - Yong Hou
- University of Michigan Medical School, Department of Internal Medicine, Ann Arbor, MI, USA
| | - Jeffrey H Ruth
- University of Michigan Medical School, Department of Internal Medicine, Ann Arbor, MI, USA
| | - Wei Luo
- University of Michigan Medical School, Department of Internal Medicine, Ann Arbor, MI, USA
| | - Daniel T Eitzman
- University of Michigan Medical School, Department of Internal Medicine, Ann Arbor, MI, USA
| | - Alisa E Koch
- VA Medical Service, Department of Veterans Affairs, Ann Arbor, MI, USA; University of Michigan Medical School, Department of Internal Medicine, Ann Arbor, MI, USA
| | - Mohammad A Amin
- University of Michigan Medical School, Department of Internal Medicine, Ann Arbor, MI, USA
| |
Collapse
|
11
|
Abstract
Understanding the pathogenesis of joint inflammation and destruction in rheumatoid arthritis involves dissection of the cellular and molecular interactions that occur in synovial tissue. Development of effective targeted therapies has been based on progress in achieving such insights. Safer and more specific approaches to treatment could flow from discovery of cell-cell interaction pathways that are specific to inflammation of the joint and less important in the defense against systemic infection. This article highlights selected cell-cell interactions in rheumatoid arthritis synovium that may be worthy of evaluation as future therapeutic targets.
Collapse
|
12
|
Abrantes J, Posada D, Guillon P, Esteves PJ, Le Pendu J. Widespread gene conversion of alpha-2-fucosyltransferase genes in mammals. J Mol Evol 2009; 69:22-31. [PMID: 19533213 PMCID: PMC2706377 DOI: 10.1007/s00239-009-9239-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2008] [Revised: 03/13/2009] [Accepted: 04/15/2009] [Indexed: 11/25/2022]
Abstract
The alpha-2-fucosyltransferases (alpha2FTs) are enzymes involved in the biosynthesis of alpha2fucosylated glycan structures. In mammalian genomes, there are three alpha2FT genes located in tandem-FUT1, FUT2, and Sec1-each contained within a single exon. It has been suggested that these genes originated from two successive duplications, with FUT1 being generated first and FUT2 and Sec1 second. Despite gene conversion being considered the main mechanism of concerted evolution in gene families, previous studies of primates alpha2FTs failed to detect it, although the occurrence of gene conversion between FUT2 and Sec1 was recently reported in a human allele. The primary aim of our work was to initiate a broader study on the molecular evolution of mammalian alpha2FTs. Sequence comparison leads us to confirm that the three genes appeared by two rounds of duplication. In addition, we were able to detect multiple gene-conversion events at the base of primates and within several nonprimate species involving FUT2 and Sec1. Gene conversion involving FUT1 and either FUT2 or Sec1 was also detected in rabbit. The extent of gene conversion between the alpha2FTs genes appears to be species-specific, possibly related to functional differentiation of these genes. With the exception of rabbits, gene conversion was not observed in the region coding the C-terminal part of the catalytic domain. In this region, the number of amino acids that are identical between FUT1 and FUT2, but different in Sec1, is higher than in other parts of the protein. The biologic meaning of this observation may be related to functional constraints.
Collapse
Affiliation(s)
- Joana Abrantes
- CIBIO-UP, Centro de Investigação em Biodiversidade e Recursos Genéticos, Campus Agrário de Vairão, Rua Padre Armando Quintas, Vairao, Portugal
- INSERM, U892, Institut de Biologie, Université de Nantes, 9 Quai Moncousu, 44093 Nantes, Cedex 01, France
- Departamento de Zoologia e Antropologia, Faculdade de Ciências da Universidade do Porto, Porto, Portugal
| | - David Posada
- Departamento de Bioquímica, Genética e Inmunología, Facultad de Biología, Campus Lagoas-Marcosende, Universidad de Vigo, Vigo, 36310 Spain
| | - Patrice Guillon
- INSERM, U892, Institut de Biologie, Université de Nantes, 9 Quai Moncousu, 44093 Nantes, Cedex 01, France
| | - Pedro J. Esteves
- CIBIO-UP, Centro de Investigação em Biodiversidade e Recursos Genéticos, Campus Agrário de Vairão, Rua Padre Armando Quintas, Vairao, Portugal
- CITS, Centro de Investigação em Tecnologias da Saúde, IPSN, CESPU, Gandra, Portugal
| | - Jacques Le Pendu
- INSERM, U892, Institut de Biologie, Université de Nantes, 9 Quai Moncousu, 44093 Nantes, Cedex 01, France
| |
Collapse
|
13
|
Gayer CP, Chaturvedi LS, Wang S, Alston B, Flanigan TL, Basson MD. Delineating the signals by which repetitive deformation stimulates intestinal epithelial migration across fibronectin. Am J Physiol Gastrointest Liver Physiol 2009; 296:G876-85. [PMID: 19179620 PMCID: PMC2670672 DOI: 10.1152/ajpgi.90648.2008] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Repetitive strain stimulates intestinal epithelial migration across fibronectin via focal adhesion kinase (FAK), Src, and extracellular signal-related kinase (ERK) although how these signals act and interact remains unclear. We hypothesized that PI3K is central to this pathway. We subjected Caco-2 and intestinal epithelial cell-6 cells to 10 cycles/min deformation on flexible fibronectin-coated membranes, assayed migration by wound closure, and signaling by immunoblots. Strain stimulated PI3K, AKT, glycogen synthase kinase (GSK), and p38 phosphorylation. Blocking each kinase prevented strain stimulation of migration. Blocking PI3K prevented strain-stimulated ERK and p38 phosphorylation. Blocking AKT did not. Downstream, blocking PI3K, AKT, or ERK inhibited strain-induced GSK-Ser9 phosphorylation. Upstream of AKT, reducing FAK or Rac1 by siRNA blocked strain-stimulated AKT phosphorylation, but inhibiting Src by PP2 or siRNA did not. Transfection with FAK point mutants at Tyr397, Tyr576/577, or Tyr925 demonstrated that only FAK925 phosphorylation is required for strain-stimulated AKT phosphorylation. Myosin light chain activation by strain required FAK, Rac1, PI3K, AKT, GSK, and ERK but not Src or p38. Finally, blebbistatin, a nonmuscle myosin II inhibitor, blocked the motogenic effect of strain downstream of myosin light chain. Thus strain stimulates intestinal epithelial migration across fibronectin by a complex pathway including Src, FAK, Rac1, PI3K, AKT, GSK, ERK, p38, myosin light chain, and myosin II.
Collapse
Affiliation(s)
- Christopher P. Gayer
- Departments of Surgery, Anatomy, and Cell Biology, John D. Dingell VA Medical Center, Wayne State University, Detroit and Michigan State University, Lansing, Michigan
| | - Lakshmi S. Chaturvedi
- Departments of Surgery, Anatomy, and Cell Biology, John D. Dingell VA Medical Center, Wayne State University, Detroit and Michigan State University, Lansing, Michigan
| | - Shouye Wang
- Departments of Surgery, Anatomy, and Cell Biology, John D. Dingell VA Medical Center, Wayne State University, Detroit and Michigan State University, Lansing, Michigan
| | - Brittany Alston
- Departments of Surgery, Anatomy, and Cell Biology, John D. Dingell VA Medical Center, Wayne State University, Detroit and Michigan State University, Lansing, Michigan
| | - Thomas L. Flanigan
- Departments of Surgery, Anatomy, and Cell Biology, John D. Dingell VA Medical Center, Wayne State University, Detroit and Michigan State University, Lansing, Michigan
| | - Marc D. Basson
- Departments of Surgery, Anatomy, and Cell Biology, John D. Dingell VA Medical Center, Wayne State University, Detroit and Michigan State University, Lansing, Michigan
| |
Collapse
|