1
|
Mariappan V, Shanmugam L, Ranganathan Green S, Easow JM, Mutheneni SR, Thirugnanasambandhar Sivasubramanian A, Balakrishna Pillai A. Increased shedding of PECAM-1 associated with elevated serum MMP-14 levels as new blood indicators of dengue disease manifestation. Infect Dis Now 2024; 54:104964. [PMID: 39181201 DOI: 10.1016/j.idnow.2024.104964] [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: 12/22/2023] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVES Host factors that regulate plasma leakage during severe dengue (SD) are under investigation. While PECAM-1 and MMP-14 have been reported to regulate vascular integrity, their role in dengue pathogenesis remains unexplored. This study aims to assess the association of soluble PECAM-1 and MMP-14 with dengue severity symptoms. PATIENTS AND METHODS Serum levels of PECAM-1 and MMP-14 were evaluated in dengue (N-25) comprising 10 severe dengue (SD) and 15 non-severe dengue, 10 other febrile illnesses along with healthy controls (N-10) using ELISA. Protein levels were assessed using in vitro models. RESULTS From febrile to critical phase, a significant increase in PECAM-1 (P≤0.01) & MMP-14 (P≤0.001) levels were observed in SD cases compared to non-severe or other controls. Serum levels of PECAM-1 and MMP 14 were found to be positively (P≤0.001) associated. Soluble PECAM-1 levels of severe defervescence showed a positive correlation (P≤0.001) with plasma leakage and an inverse relationship (P≤0.001) with platelet count. In vitro analysis revealed elevated expression of study proteins in endothelial cells activated with severe serum samples. To the best of our knowledge, this is the first report to explore PECAM-1 or MMP-14 dynamics and their association with dengue severity. CONCLUSION Higher shedding of sPECAM-1 accompanied with increased levels of MMP-14 is strongly associated with severe dengue. However, the exact role of serum PECAM-1 in disease prognosis requires further studies.
Collapse
Affiliation(s)
- Vignesh Mariappan
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Lokesh Shanmugam
- ICMR-National Institute of Epidemiology (ICMR-NIE), Ayapakkam, Chennai 600 070, India.
| | - Siva Ranganathan Green
- Department of General Medicine, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Joshy M Easow
- Department of Microbiology, Mahatma Gandhi Medical College and Research Institute (MGMCRI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| | - Srinivasa Rao Mutheneni
- Division of Applied Biology, CSIR-Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad, Telangana 500 007, India.
| | | | - Agieshkumar Balakrishna Pillai
- Mahatma Gandhi Medical Advanced Research Institute (MGMARI), Sri Balaji Vidyapeeth (Deemed to be University), Puducherry 607 402, India.
| |
Collapse
|
2
|
Sénémaud J, Skarbek C, Hernandez B, Song R, Lefevre I, Bianchi E, Castier Y, Nicoletti A, Bureau C, Caligiuri G. Camouflaging endovascular stents with an endothelial coat using CD31 domain 1 and 2 mimetic peptides. JVS Vasc Sci 2024; 5:100213. [PMID: 39257386 PMCID: PMC11386311 DOI: 10.1016/j.jvssci.2024.100213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/17/2024] [Indexed: 09/12/2024] Open
Abstract
Objective Implantation of an endovascular device disrupts the homeostatic CD31:CD31 interactions among quiescent endothelial cells (ECs), platelets, and circulating leukocytes. The aim of this study was to design an endothelial-mimetic coating of nitinol and cobalt-chromium (CoCr) surfaces and stents using synthetic CD31 peptides, to promote device endothelialization and pacific integration within the arterial wall. Methods Peptides mimicking the domains 1 (D1) and 2 (D2) of CD31 were synthetized and immobilized onto experimental nitinol and CoCr surfaces using a three-step, dip-coating, mussel-inspired protocol using copper-free click chemistry. Human aortic EC phenotype and endothelialization assessment using parallel scratch tests were carried out using five synthetic CD31 peptides coated on 4.8-mm nitinol and CoCr flat disks and were compared with control disks. The CD31 peptide exhibiting the best results in vitro was then immobilized on clinical-grade 3 × 40-mm self-expanding nitinol and 2.5 × 20.0-mm balloon-expandable CoCr stents. Such devices were implanted in native arteries of White New Zealand rabbits, and compared with control uncoated bare metal stents (BMS) and drug-eluting stents 7 and 30 days after implantation using resin cross-sections and scanning electron microscopy (n = 2-3 per group at each time point). Results Membrane-distal CD31 D1 and D2 peptides exhibited a distinct capability to foster a healthy endothelial phenotype and to promote endothelialization in vitro. By day 7 after implantation, CD31 nitinol and CoCr stents were evenly covered by wholesome ECs, devoid of thromboinflammatory signs, in contrast with both BMS and drug-eluting stents. Such results were consistent until day 30. Conclusions Membrane-distal CD31 biomimetic peptides seem to camouflage the device surface effectively, preventing local reactions and promoting rapid and seamless endovascular integration.
Collapse
Affiliation(s)
- Jean Sénémaud
- Department of Vascular Surgery, Bichat University Hospital, Paris, France
- Université Paris Cité, Paris, France
- Laboratory for Vascular Translational Science, INSERM U1148, Paris, France
| | - Charles Skarbek
- Université Paris Cité, Paris, France
- Laboratory for Vascular Translational Science, INSERM U1148, Paris, France
| | - Belen Hernandez
- Laboratory for Vascular Translational Science, INSERM U1148, Paris, France
- Université Sorbonne Paris Nord, Paris, France
| | - Ran Song
- Sino Medical Sciences Technology Inc., Tianjin, China
| | | | - Elisabetta Bianchi
- Peptide Chemistry Unit, Peptides & Small Molecules R&D Department, IRBM SpA, Pomezia, Roma, Italy
| | - Yves Castier
- Department of Vascular Surgery, Bichat University Hospital, Paris, France
- Université Paris Cité, Paris, France
| | - Antonino Nicoletti
- Université Paris Cité, Paris, France
- Laboratory for Vascular Translational Science, INSERM U1148, Paris, France
| | - Christophe Bureau
- Sino Medical Sciences Technology Inc., Tianjin, China
- AlchiMedics S.A.S., Paris, France
| | - Giuseppina Caligiuri
- Université Paris Cité, Paris, France
- Laboratory for Vascular Translational Science, INSERM U1148, Paris, France
- Department of Cardiology, Bichat University Hospital, Paris, France
| |
Collapse
|
3
|
Chen Z, Tang L, Luo L, Luo W, Li Y, Wang X, Huang L, Hu Y, Mei H. Enhancing the Treatment of Uncontrolled Inflammation through the Targeted Delivery of TPCA-1-Loaded Nanoparticles. Pharmaceutics 2023; 15:2435. [PMID: 37896195 PMCID: PMC10609852 DOI: 10.3390/pharmaceutics15102435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023] Open
Abstract
Uncontrolled inflammation is a pathological state that underlies many diseases. Despite the development of numerous anti-inflammatory agents, the treatment of uncontrolled inflammation remains a challenging task. We developed a targeted delivery system for [5-(p-fluorophenyl)-2-ureido]thiophene-3-carboxamide (TPCA-1), a potent inhibitor of the NF-κB signaling pathway. The system comprises TPCA-1-loaded nanoparticles (NPs) functionalized with a monoclonal antibody (mAb) that specifically binds to the break point of the IgD6 region of the platelet/endothelial cell adhesion molecule-1 (PECAM-1) extracellular segment that is overexposed on the injured endothelium and activated macrophages during the pathogenesis of inflammation. In vitro binding and cellular uptake experiments revealed that the mAb modification on NPs could significantly enhance uptake by both Raw264.7 and HUVEC compared with unmodified NPs. In studies conducted at the cellular level focusing on anti-inflammatory and antioxidant effects, this formulation was found to effectively inhibit M1 polarization of macrophages, downregulate the secretion of pro-inflammatory cytokines, and reduce the production of reactive oxygen species (ROS) and nitric oxide (NO). In an animal model of vascular endothelial injury with acute inflammation, these NPs were capable of delivering TPCA-1 to inflammatory lesions in a targeted manner. Compared with the free agent-treated group, the NP-treated group exhibited reduced infiltration of inflammatory cells. In conclusion, our study demonstrates that this targeted delivery of TPCA-1-loaded NPs represents a promising strategy for improved mitigation of uncontrolled inflammation.
Collapse
Affiliation(s)
- Zhaozhao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Lili Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Wenjing Luo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Yingying Li
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Xindi Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Linlin Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Z.C.); (L.T.); (L.L.); (W.L.); (Y.L.); (X.W.); (L.H.)
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
- Key Laboratory of Biological Targeted Therapy of Hubei Province, Wuhan 430022, China
| |
Collapse
|
4
|
Andreata F, Clément M, Benson RA, Hadchouel J, Procopio E, Even G, Vorbe J, Benadda S, Ollivier V, Ho-Tin-Noe B, Le Borgne M, Maffia P, Nicoletti A, Caligiuri G. CD31 signaling promotes the detachment at the uropod of extravasating neutrophils allowing their migration to sites of inflammation. eLife 2023; 12:e84752. [PMID: 37549051 PMCID: PMC10431918 DOI: 10.7554/elife.84752] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 08/04/2023] [Indexed: 08/09/2023] Open
Abstract
Effective neutrophil migration to sites of inflammation is crucial for host immunity. A coordinated cascade of steps allows intravascular leukocytes to counteract the shear stress, transmigrate through the endothelial layer, and move toward the extravascular, static environment. Those events are tightly orchestrated by integrins, but, while the molecular mechanisms leading to their activation have been characterized, the regulatory pathways promoting their detachment remain elusive. In light of this, it has long been known that platelet-endothelial cell adhesion molecule (Pecam1, also known as CD31) deficiency blocks leukocyte transmigration at the level of the outer vessel wall, yet the associated cellular defects are controversial. In this study, we combined an unbiased proteomic study with in vitro and in vivo single-cell tracking in mice to study the dynamics and role of CD31 during neutrophil migration. We found that CD31 localizes to the uropod of migrating neutrophils along with closed β2-integrin and is required for essential neutrophil actin/integrin polarization. Accordingly, the uropod of Pecam1-/- neutrophils is unable to detach from the extracellular matrix, while antagonizing integrin binding to extracellular matrix components rescues this in vivo migratory defect. Conversely, we showed that sustaining CD31 co-signaling actively favors uropod detachment and effective migration of extravasated neutrophils to sites of inflammation in vivo. Altogether, our results suggest that CD31 acts as a molecular rheostat controlling integrin-mediated adhesion at the uropod of egressed neutrophils, thereby triggering their detachment from the outer vessel wall to reach the inflammatory sites.
Collapse
Affiliation(s)
- Francesco Andreata
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Marc Clément
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Robert A Benson
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUnited Kingdom
| | - Juliette Hadchouel
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center (PARCC)ParisFrance
| | - Emanuele Procopio
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Guillaume Even
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Julie Vorbe
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Samira Benadda
- Cell and Tissue Imaging Platform, INSERM, CNRS, ERL8252, Centre de Recherche sur l’Inflammation (CRI)ParisFrance
| | - Véronique Ollivier
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Benoit Ho-Tin-Noe
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Marie Le Borgne
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUnited Kingdom
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico IINaplesItaly
| | - Antonino Nicoletti
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
| | - Giuseppina Caligiuri
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, Laboratory for Vascular Translational Science (LVTS)ParisFrance
- Department of Cardiology and of Physiology, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Paris Nord Val-de-Seine, Site BichatParisFrance
| |
Collapse
|
5
|
von Richthofen HJ, Westerlaken GH, Gollnast D, Besteman S, Delemarre EM, Rodenburg K, Moerer P, Stapels DA, Andiappan AK, Rötzschke O, Nierkens S, Leavis HL, Bont LJ, Rooijakkers SH, Meyaard L. Soluble Signal Inhibitory Receptor on Leukocytes-1 Is Released from Activated Neutrophils by Proteinase 3 Cleavage. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:389-397. [PMID: 36637221 PMCID: PMC9915861 DOI: 10.4049/jimmunol.2200169] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 12/05/2022] [Indexed: 01/14/2023]
Abstract
Signal inhibitory receptor on leukocytes-1 (SIRL-1) is an immune inhibitory receptor expressed on human granulocytes and monocytes that dampens antimicrobial functions. We previously showed that sputum neutrophils from infants with severe respiratory syncytial virus (RSV) bronchiolitis have decreased SIRL-1 surface expression compared with blood neutrophils and that SIRL-1 surface expression is rapidly lost from in vitro activated neutrophils. This led us to hypothesize that activated neutrophils lose SIRL-1 by ectodomain shedding. Here, we developed an ELISA and measured the concentration of soluble SIRL-1 (sSIRL-1) in patients with RSV bronchiolitis and hospitalized patients with COVID-19, which are both characterized by neutrophilic inflammation. In line with our hypothesis, sSIRL-1 concentration was increased in sputum compared with plasma of patients with RSV bronchiolitis and in serum of hospitalized patients with COVID-19 compared with control serum. In addition, we show that in vitro activated neutrophils release sSIRL-1 by proteolytic cleavage and that this diminishes the ability to inhibit neutrophilic reactive oxygen species production via SIRL-1. Finally, we found that SIRL-1 shedding is prevented by proteinase 3 inhibition and by extracellular adherence protein from Staphylococcus aureus. Notably, we recently showed that SIRL-1 is activated by PSMα3 from S. aureus, suggesting that S. aureus may counteract SIRL-1 shedding to benefit from preserved inhibitory function of SIRL-1. In conclusion, we report that SIRL-1 is released from activated neutrophils by proteinase 3 cleavage and that endogenous sSIRL-1 protein is present in vivo.
Collapse
Affiliation(s)
- Helen J. von Richthofen
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Oncode Institute, Utrecht, the Netherlands
| | - Geertje H.A. Westerlaken
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Oncode Institute, Utrecht, the Netherlands
| | - Doron Gollnast
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Oncode Institute, Utrecht, the Netherlands
| | - Sjanna Besteman
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Department of Pediatrics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Eveline M. Delemarre
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Karlijn Rodenburg
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Petra Moerer
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Daphne A.C. Stapels
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Anand K. Andiappan
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore; and
| | - Olaf Rötzschke
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore; and
| | - Stefan Nierkens
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Helen L. Leavis
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Louis J. Bont
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Department of Pediatrics, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Suzan H.M. Rooijakkers
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Linde Meyaard
- Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands;,Oncode Institute, Utrecht, the Netherlands
| |
Collapse
|
6
|
Kogata S, Lo PC, Maeda A, Okamatsu C, Sato K, Yamamoto R, Haneda T, Yoneyama T, Toyama C, Eguchi H, Masahata K, Kamiyama M, Okuyama H, Miyagawa S. Suppression of macrophage-mediated xenogeneic rejection by the ectopic expression of human CD177. Transpl Immunol 2022; 74:101663. [PMID: 35835297 DOI: 10.1016/j.trim.2022.101663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/01/2022] [Accepted: 07/03/2022] [Indexed: 10/17/2022]
Abstract
Cellular xenogeneic rejection by the innate immune system is a major immunological obstruction that needs to be overcome for the successful clinical use of xenografts. Our focus has been on macrophage-mediated xenogeneic rejection, since suppressing macrophage function has considerable potential for practical applications in the area of xenotransplantation. We report herein on an investigation of the suppressive effect of human CD177 (hCD177) against macrophage-mediated xenogeneic rejection. Wild type swine aortic endothelial cell (SEC) and an SEC transfectant with hCD177 (SEC/hCD177) were co-cultured with macrophages, and the degree of cytotoxicity was evaluated by WST-8 assays, and phagocytosis was examined using Calcein-AM labeling methods. The expression of anti/pro-inflammatory cytokines was evaluated by RT-qPCR and the phosphorylation of SHP-1 on macrophages in co-culture was evaluated by Western blotting. The result of cytotoxicity assays indicated that hCD177 suppressed M1 macrophage-mediated xenogeneic rejection (vs. SEC, p < 0.0001). Similarly, the result of phagocytosis assays indicated that hCD177 suppressed it (vs. SEC, p < 0.05). In addition, hCD177 significantly suppressed the expression of IL-1β, a pro-inflammatory cytokine, in M1 macrophages (vs. SEC, p < 0.01). Luciferase assays using THP1-Lucia NF-kB also showed a significant difference in NF-kB activation (vs. SEC, p < 0.001). In addition, hCD177 was found to induce the phosphorylation of SHP-1 in M1 macrophages (vs. SEC, p < 0.05). These findings indicate that hCD177 suppresses M1 macrophage-mediated xenogeneic rejection, at least in part via in the phosphorylation of SHP-1.
Collapse
Affiliation(s)
- Shuhei Kogata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan; Division of Pediatric Surgery, Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Pei-Chi Lo
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akira Maeda
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Chizu Okamatsu
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazuki Sato
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Riho Yamamoto
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomoko Haneda
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Tomohisa Yoneyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Chiyoshi Toyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroshi Eguchi
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Kazunori Masahata
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Masafumi Kamiyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Hiroomi Okuyama
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Osaka, Japan; Meiji University International Institute for Bio-Resource Research, Kanagawa, Japan
| |
Collapse
|
7
|
Transfer of the longevity-associated variant of BPIFB4 gene rejuvenates immune system and vasculature by a reduction of CD38 + macrophages and NAD + decline. Cell Death Dis 2022; 13:86. [PMID: 35087020 PMCID: PMC8792139 DOI: 10.1038/s41419-022-04535-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/20/2021] [Accepted: 01/13/2022] [Indexed: 01/10/2023]
Abstract
As we age, our body experiences chronic, systemic inflammation contributing to the morbidity and mortality of the elderly. The senescent immune system has been described to have a causal role in driving systemic aging and therefore may represent a key therapeutic target to prevent pathological consequences associated with aging and extend a healthy lifespan. Previous studies from our group associated a polymorphic haplotype variant in the BPIFB4 gene (LAV-BPIFB4) with exceptional longevity. Transfer of the LAV-BPIFB4 in preclinical models halted the progression of cardiovascular diseases (CVDs) and frailty by counterbalancing chronic inflammation. In the present study, we aimed to delineate the action of systemic adeno-associated viral vector-mediated LAV-BPIFB4 gene transfer (AAV-LAV-BPIFB4) on the deleterious age-related changes of the immune system and thereby the senescence-associated events occurring in C57BL/6J mice aged 26 months. Our in vivo data showed that 26-months-old mice had a higher frequency of CD45+SA-beta Gal+ immune cells in peripheral blood than young (4-months-old) C57BL/6J mice. Notably, AAV-LAV-BPIFB4 gene transfer in aged mice reduced the pool of peripheral immunosenescent cells that were shown to be enriched in the spleen. In addition, the proper tuning of the immune secretory phenotype (IL1βlow, IL6low, IL10high) associated with a significant reduction in SA-beta Gal-positive area of aorta from AAV-LAV treated mice. At the functional level, the reduction of senescence-associated inflammation ensured sustained NAD+ levels in the plasma of AAV-LAV-BPIFB4 old mice by preventing the NADase CD38 increase in F4/80+ tissue-resident macrophages and Ly6Chigh pro-inflammatory monocytes of the spleen and bone marrow. Finally, to validate the clinical implication of our findings, we showed that Long-living-individuals (LLIs, >95 years), which delay CVDs onset, especially if LAV-carriers, were characterized by high NAD+ levels. In conclusion, the new senotherapeutic action of LAV-BPIFB4 may offer a valuable therapeutic tool to control aging and reduce the burden of its pathophysiological disorders, such as CVDs.
Collapse
|
8
|
Ding W, Chen X, Yang L, Chen Y, Song J, Bu W, Feng B, Zhang M, Luo Y, Jia X, Feng L. Combination of ShuangDan Capsule and Sorafenib Inhibits Tumor Growth and Angiogenesis in Hepatocellular Carcinoma Via PI3K/Akt/mTORC1 Pathway. Integr Cancer Ther 2022; 21:15347354221078888. [PMID: 35234063 PMCID: PMC8894619 DOI: 10.1177/15347354221078888] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a high mortality liver cancer. The existing treatments (transplantation, chemotherapy, and individualized treatment) with limitations. However, drug combination provides a viable option for hepatocellular carcinoma treatment. A Chinese patent medicine, ShuangDan Capsules (SDC), has been clinically prescribed to hepatocellular carcinoma patients as adjuvant therapy and has shown good antitumor activity. The purpose of this study was to investigate whether SDC could improve the anti-cancer effect and mitigate adverse reactions of sorafenib on HCC in vivo. Magnetic resonance imaging (MRI), immunohistochemistry, and western blot were executed to reveal the potential mechanisms of the combination of SDC and sorafenib on HCC. Tumors appeared hyperintense on T2 sequence images relative to the adjacent normal liver in MRI. Combination of SDC and sorafenib inhibited the progression of DEN (Diethylnitrosamine)-induced HCC. In the HepG2 xenografts model, sorafenib plus SDC exhibited greater suppression on tumor growth than individual treatment accompanied with decreased expression of VEGF, VEGFA, Ki67, CD31 and increased expression of caspase-3. Furthermore, PI3K/Akt/mTORC1 pathway was inhibited by co-administration. Sorafenib monotherapy elicited hepatotoxicity for specific expression in the up-regulated level of aspartate transaminase (AST) and AST/glutamic-pyruvic transaminase (ALT) ratio, but the co-administration could remedy this adverse effect. These dates indicated that the combination of SDC and sorafenib might offer a potential therapy for HCC.
Collapse
Affiliation(s)
- Wenbo Ding
- Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiuwei Chen
- Yuhuatai District Maternity and Child Care Clinic, Nanjing, P.R. China
| | - Licheng Yang
- China Pharmaceutical University, Nanjing, P.R. China
| | - Yaping Chen
- China Pharmaceutical University, Nanjing, P.R. China
| | - Jie Song
- Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Weiquan Bu
- Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Bin Feng
- Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Meng Zhang
- China Pharmaceutical University, Nanjing, P.R. China
| | - Yi Luo
- Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiaobin Jia
- China Pharmaceutical University, Nanjing, P.R. China
| | - Liang Feng
- China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
9
|
Briceño O, Peralta-Prado A, Garrido-Rodríguez D, Romero-Mora K, Chávez-Torres M, de la Barrera CA, Reyes-Terán G, Ávila-Ríos S. Characterization of CD31 expression in CD4+ and CD8+T cell subpopulations in chronic untreated HIV infection. Immunol Lett 2021; 235:22-31. [PMID: 33852965 DOI: 10.1016/j.imlet.2021.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 03/09/2021] [Accepted: 04/07/2021] [Indexed: 11/18/2022]
Abstract
BACKGROUND The platelet endothelial cell adhesion molecule-1 (PECAM-1) or CD31 has been involved in regulation of T-cell tolerance, activation, survival and homing in mice cells. However, there is limited knowledge about the expression pattern and role of this molecule in human T cells, particularly in conditions of chronic immune activation. OBJECTIVES We explored CD31 expression in T cell differentiation subsets of individuals with untreated HIV infection and in non-HIV-infected controls. We also assessed phenotypic differences between CD31+ and CD31- subsets in memory and terminally differentiated (TEMRA) CD4+ and CD8 + T cells. METHODS Forty-one individuals with untreated HIV infection and 34 non-HIV-infected controls were included in the study. We compared the expression of CD31 in CD4+ and CD8 + T cells across stages of differentiation in the two study groups by flow cytometry. We also analyzed the expression of CD57 (a marker of senescence), Ki67 (a marker of cycling cells), PD-1 (a marker of exhaustion), and CD38/HLA-DR (a marker of immune activation) on memory and TEMRA CD31+ and CD31- T cells. RESULTS CD31 expression was significantly higher in CD8 + T cells than in CD4 + T cells, measured as frequency, absolute numbers and median fluorescence intensity (MFI), in both study groups (p < 0.0001 in all cases). Intermediate differentiation subsets of CD4+ and CD8 + T cells expressed higher levels of CD31 in the context of HIV infection (p < 0.001 in all cases). CD31 expression frequency decreased with cellular differentiation of CD4+ and CD8 + T cells in both groups, but this decrease was steeper in individuals without HIV infection (CD4+: p < 0.001 and CD8+: p < 0.0001). As expected, memory and TEMRA CD4+ and CD8 + T cells expressed significantly higher levels of CD57, PD-1, Ki67 and CD38/HLA-DR in HIV-infected compared to non-HIV-infected individuals (p < 0.01 in all cases). CD31 expression was associated with lower activation of memory (but not TEMRA) CD4 + T cells in non-HIV-infected persons, an effect not observed in the HIV-infected group. CD31 expression on memory CD8 + T cells of HIV-infected individuals was associated higher levels of PD-1 (p = 0.0019) and CD38/HLADR (p = 0.0345), and higher PD-1 expression on CD8 + TEMRA (p = 0.0024), an effect not observed in non-HIV-infected individuals. CONCLUSION In the context of HIV-associated chronic immune activation, specifically on memory CD8 + T cells, CD31 expression was associated with higher PD-1 and CD38/HLA-DR co-expression, suggesting that CD31 expression may result from an insufficient attempt to contain T cell exhaustion and activation. CD31-targeted therapies may contribute to modulate these cellular responses.
Collapse
Affiliation(s)
- Olivia Briceño
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico.
| | - Amy Peralta-Prado
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| | - Daniela Garrido-Rodríguez
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| | - Karla Romero-Mora
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| | - Monserrat Chávez-Torres
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| | - Claudia-Alvarado de la Barrera
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| | - Gustavo Reyes-Terán
- Coordinating Commission of the Mexican National Institutes of Health, Mexico City, Mexico
| | - Santiago Ávila-Ríos
- Instituto Nacional de Enfermedades Respiratorias, Centro de Investigación en Enfermedades Infecciosas, México City, Mexico
| |
Collapse
|
10
|
Sluiter TJ, van Buul JD, Huveneers S, Quax PHA, de Vries MR. Endothelial Barrier Function and Leukocyte Transmigration in Atherosclerosis. Biomedicines 2021; 9:328. [PMID: 33804952 PMCID: PMC8063931 DOI: 10.3390/biomedicines9040328] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/24/2022] Open
Abstract
The vascular endothelium is a highly specialized barrier that controls passage of fluids and migration of cells from the lumen into the vessel wall. Endothelial cells assist leukocytes to extravasate and despite the variety in the specific mechanisms utilized by different leukocytes to cross different vascular beds, there is a general principle of capture, rolling, slow rolling, arrest, crawling, and ultimately diapedesis via a paracellular or transcellular route. In atherosclerosis, the barrier function of the endothelium is impaired leading to uncontrolled leukocyte extravasation and vascular leakage. This is also observed in the neovessels that grow into the atherosclerotic plaque leading to intraplaque hemorrhage and plaque destabilization. This review focuses on the vascular endothelial barrier function and the interaction between endothelial cells and leukocytes during transmigration. We will discuss the role of endothelial dysfunction, transendothelial migration of leukocytes and plaque angiogenesis in atherosclerosis.
Collapse
Affiliation(s)
- Thijs J. Sluiter
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jaap D. van Buul
- Sanquin Research and Landsteiner Laboratory, Leeuwenhoek Centre for Advanced Microscopy, Swammerdam Institute for Life Sciences, University of Amsterdam, 1066 CX Amsterdam, The Netherlands;
| | - Stephan Huveneers
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Paul H. A. Quax
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Margreet R. de Vries
- Department of Vascular Surgery, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (T.J.S.); (P.H.A.Q.)
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
11
|
Sannier A, Stroumza N, Atlan M, Even G, Guedj K, Sénémaud J, Coscas R, Caligiuri G, Nicoletti A. A CD31-Derived Peptide Prevents the Development of Antibody-Mediated Lesions in a Rat Model of Aortic Allograft. Transplant Proc 2021; 53:746-749. [PMID: 33549347 DOI: 10.1016/j.transproceed.2021.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 01/08/2021] [Indexed: 12/01/2022]
Abstract
BACKGROUND Antibody-mediated rejection (AMR) is a major cause of graft loss. The development of donor-specific antibodies (DSAs) directed against the allogeneic HLA molecules expressed by the graft also leads to accelerated arteriosclerosis. CD31 is a protein expressed on endothelial and immune cells, ensuring homeostasis at this interface. When strong immune stimulation occurs, the regulatory function of CD31 is lost owing to cleavage of its extracellular portion. P8RI, a synthetic peptide that binds to the ectodomain of CD31, is able to restore the CD31 immunomodulatory function. In this study, we hypothesized that CD31 could represent an attractive molecular target in AMR and investigated whether P8RI could prevent the development of vascular antibody-mediated lesions. MATERIALS AND METHODS A rat model of orthotopic aortic allograft was used, and P8RI was administered for 28 days. Circulating DSAs were quantified to assess the alloimmune humoral response, and histologic and immunohistochemical analyses of aortic allografts were performed to estimate antibody-mediated lesions in the allograft. RESULTS Aorta-allografted rats receiving P8RI developed fewer DSAs than control animals (mean fluorescence intensity 344 vs 741). The density of nuclei in the media (3.4 x 10-5 vs 2.2 x 10-5 nuclei/px2) and media surface area (2.33 x 106 vs 2.02 x 106 px2) were higher in animals treated with P8RI than in control animals. CONCLUSIONS These data support a therapeutic potential for molecules able to restore the CD31 signaling to fight AMR. P8RI, an agonist synthetic peptide targeting CD31, might prevent DSA production and have a beneficial effect in limiting arterial antibody-mediated lesions. CD31 agonists may become therapeutic tools to prevent and treat solid organ transplant arteriosclerosis.
Collapse
Affiliation(s)
- Aurélie Sannier
- University of Paris, Institut national de la santé et de la recherche médicale, UMRS1148, Paris, France; Department of Pathology, Assistance publique - hôpitaux de Paris, Bichat Hospital, Paris, France.
| | - Nathaniel Stroumza
- Department of Reconstructive and Aesthetic Plastic Surgery, Assistance publique - hôpitaux de Paris, Tenon Hospital, Paris, France
| | - Michael Atlan
- Department of Reconstructive and Aesthetic Plastic Surgery, Assistance publique - hôpitaux de Paris, Tenon Hospital, Paris, France; Sorbonne University, Paris, France
| | - Guillaume Even
- University of Paris, Institut national de la santé et de la recherche médicale, UMRS1148, Paris, France
| | - Kevin Guedj
- University of Paris, Institut national de la santé et de la recherche médicale, UMRS1148, Paris, France
| | - Jean Sénémaud
- University of Paris, Institut national de la santé et de la recherche médicale, UMRS1148, Paris, France; Vascular and Thoracic Surgery Department, Assistance publique - hôpitaux de Paris, Bichat Hospital, Paris, France
| | - Raphaël Coscas
- Vascular Surgery Department, Assistance publique - hôpitaux de Paris, Ambroise Paré Hospital, Boulogne-Billancourt, France; Versailles Saint-Quentin-en-Yvelines University, Versailles, France
| | - Giuseppina Caligiuri
- University of Paris, Institut national de la santé et de la recherche médicale, UMRS1148, Paris, France
| | - Antonino Nicoletti
- University of Paris, Institut national de la santé et de la recherche médicale, UMRS1148, Paris, France
| |
Collapse
|
12
|
Abstract
The potential of CD31 as a therapeutic target in atherosclerosis has been considered ever since its cloning in the 1990s, but the exact role played by this molecule in the biologic events underlying atherosclerosis has remained controversial, resulting in the stalling of any therapeutic perspective. Due to the supposed cell adhesive properties of CD31, specific monoclonal antibodies and recombinant proteins were regarded as blocking agents because their use prevented the arrival of leukocytes at sites of acute inflammation. However, the observed effect of those compounds likely resulted from the engagement of the immunomodulatory function of CD31 signaling. This was acknowledged only later though, upon the discovery of CD31's 2 intracytoplasmic tyrosine residues called immunoreceptor tyrosine inhibitory motifs. A growing body of evidence currently points at a therapeutic potential for CD31 agonists in atherothrombosis. Clinical observations show that CD31 expression is altered at the surface of leukocytes infiltrating unhealed atherothrombotic lesions and that the physiological immunomodulatory functions of CD31 are lost at the surface of blood leukocytes in patients with acute coronary syndromes. On the contrary, translational studies using candidate therapeutic molecules in laboratory animals have provided encouraging results: synthetic peptides administered to atherosclerotic mice as systemic drugs in the acute phases of atherosclerotic complications favor the healing of wounded arteries, whereas the immobilization of CD31 agonist peptides onto coronary stents implanted in farm pigs favors their peaceful integration within the coronary arterial wall.
Collapse
Affiliation(s)
- Giuseppina Caligiuri
- From the Laboratory for Vascular Translational Science, Inserm U1148, Université de Paris, France; and Department of Cardiology, Assistance Publique-Hôpitaux de Paris, Hôpitaux Universitaires Paris Nord Val-de-Seine, Site Bichat, France
| |
Collapse
|
13
|
Kared H, Tan SW, Lau MC, Chevrier M, Tan C, How W, Wong G, Strickland M, Malleret B, Amoah A, Pilipow K, Zanon V, Govern NM, Lum J, Chen JM, Lee B, Florian MC, Geiger H, Ginhoux F, Ruiz-Mateos E, Fulop T, Rajasuriar R, Kamarulzaman A, Ng TP, Lugli E, Larbi A. Immunological history governs human stem cell memory CD4 heterogeneity via the Wnt signaling pathway. Nat Commun 2020; 11:821. [PMID: 32041953 PMCID: PMC7010798 DOI: 10.1038/s41467-020-14442-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 01/09/2020] [Indexed: 12/21/2022] Open
Abstract
The diversity of the naïve T cell repertoire drives the replenishment potential and capacity of memory T cells to respond to immune challenges. Attrition of the immune system is associated with an increased prevalence of pathologies in aged individuals, but whether stem cell memory T lymphocytes (TSCM) contribute to such attrition is still unclear. Using single cells RNA sequencing and high-dimensional flow cytometry, we demonstrate that TSCM heterogeneity results from differential engagement of Wnt signaling. In humans, aging is associated with the coupled loss of Wnt/β-catenin signature in CD4 TSCM and systemic increase in the levels of Dickkopf-related protein 1, a natural inhibitor of the Wnt/β-catenin pathway. Functional assays support recent thymic emigrants as the precursors of CD4 TSCM. Our data thus hint that reversing TSCM defects by metabolic targeting of the Wnt/β-catenin pathway may be a viable approach to restore and preserve immune homeostasis in the context of immunological history.
Collapse
Affiliation(s)
- Hassen Kared
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore.
| | - Shu Wen Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Mai Chan Lau
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Marion Chevrier
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Crystal Tan
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Wilson How
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Glenn Wong
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Marie Strickland
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Benoit Malleret
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Amanda Amoah
- Institute of Molecular Medicine, University of Ulm, Ulm, Germany
| | - Karolina Pilipow
- Humanitas Clinical and Research Center, Laboratory of Translational Immunology (LTI), Rozzano, Italy
| | - Veronica Zanon
- Humanitas Clinical and Research Center, Laboratory of Translational Immunology (LTI), Rozzano, Italy
| | - Naomi Mc Govern
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Josephine Lum
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Jin Miao Chen
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | | | - Hartmut Geiger
- Institute of Molecular Medicine, University of Ulm, Ulm, Germany
- Experimental Hematology and Cancer Biology, CCHMC, Cincinnati, OH, USA
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore
| | - Ezequiel Ruiz-Mateos
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, Seville, Spain
| | - Tamas Fulop
- Department of Medicine, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Reena Rajasuriar
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Adeeba Kamarulzaman
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Tze Pin Ng
- Gerontology Research Programme and Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Enrico Lugli
- Humanitas Clinical and Research Center, Laboratory of Translational Immunology (LTI), Rozzano, Italy
| | - Anis Larbi
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Immunos Building, 8A Biomedical Grove, Biopolis, Republic of Singapore.
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore.
- Department of Medicine, Faculty of Medicine, University of Sherbrooke, Sherbrooke, Quebec, Canada.
| |
Collapse
|
14
|
Cleaved CD31 as a target for in vivo molecular imaging of inflammation. Sci Rep 2019; 9:19560. [PMID: 31863037 PMCID: PMC6925130 DOI: 10.1038/s41598-019-56163-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/02/2019] [Indexed: 01/16/2023] Open
Abstract
There is a need for new targets to specifically localize inflammatory foci, usable in a wide range of organs. Here, we hypothesized that the cleaved molecular form of CD31 is a suitable target for molecular imaging of inflammation. We evaluated a bioconjugate of D-P8RI, a synthetic peptide that binds all cells with cleaved CD31, in an experimental rat model of sterile acute inflammation. Male Wistar rats were injected with turpentine oil into the gastrocnemius muscle two days before 99mTc-HYNIC-D-P8RI (or its analogue with L-Proline) SPECT/CT or [18F]FDG PET/MRI. Biodistribution, stability study, histology, imaging and autoradiography of 99mTc-HYNIC-D-P8RI were further performed. Biodistribution studies revealed rapid elimination of 99mTc-HYNIC-D-P8RI through renal excretion with almost no uptake from most organs and excellent in vitro and in vivo stability were observed. SPECT/CT imaging showed a significant higher 99mTc-HYNIC-D-P8RI uptake compared with its analogue with L-Proline (negative control) and no significant difference compared with [18F]FDG (positive control). Moreover, autoradiography and histology revealed a co-localization between 99mTc-HYNIC-D-P8RI uptake and inflammatory cell infiltration. 99mTc-HYNIC-D-P8RI constitutes a new tool for the detection and localization of inflammatory sites. Our work suggests that targeting cleaved CD31 is an attractive strategy for the specific in vivo imaging of inflammatory processes.
Collapse
|
15
|
Angelini G, Flego D, Vinci R, Pedicino D, Trotta F, Ruggio A, Piemontese GP, Galante D, Ponzo M, Biasucci LM, Liuzzo G, Crea F. Matrix metalloproteinase-9 might affect adaptive immunity in non-ST segment elevation acute coronary syndromes by increasing CD31 cleavage on CD4+ T-cells. Eur Heart J 2019; 39:1089-1097. [PMID: 29211854 PMCID: PMC5915953 DOI: 10.1093/eurheartj/ehx684] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/02/2017] [Indexed: 12/22/2022] Open
Abstract
Aims In patients with acute coronary syndrome (ACS), the higher activity of effector T-cells suggests that mechanisms involving adaptive immunity dysregulation might play a role in coronary instability. The shedding of the functional CD31 domain 1–5 leads to uncontrolled lymphocyte activation. In experimental models, matrix metalloproteinase-9 (MMP-9) has been implicated in endothelial CD31 cleavage. Interestingly, higher serum levels of MMP-9 have been observed in ACS. We aim to investigate the mechanisms underlying CD31 dysregulation in ACS. Methods and results To assess CD31 cleavage on CD4+ T-cells, we analysed by flow cytometry CD4+ T-cells of 30 ACS, 25 stable angina (SA) patients, and 28 controls (CTRL) using two different CD31 antibodies that specifically recognize domain 1–5 or the non-functional membrane-proximal domain 6. The ratio between the domains was significantly lower in ACS than in SA and CTRL (P = 0.002 ACS vs. SA; P = 0.002 ACS vs. CTRL). After stimulation with anti-CD3/CD28, the 1–5/6 domain ratio was significantly lower in ACS than in SA (P = 0.005). ELISA of supernatants obtained from T-cell receptor-stimulated CD4+ T-cells showed higher production of MMP-9 in ACS than in SA (P < 0.001). CD31 domain 1–5 expression in activated CD4+ T-cells from ACS patients increased after treatment with a specific MMP-9 inhibitor (P = 0.042). Conclusion Our study suggest that enhanced MMP-9 release plays a key role in determining the cleavage and shedding of the functional CD31 domain 1–5 in CD4+ T-cells of ACS patients. This mechanism might represent an important therapeutic target to modulate T-cell dysregulation in ACS. ![]()
Collapse
Affiliation(s)
- Giulia Angelini
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Davide Flego
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Ramona Vinci
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Daniela Pedicino
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Francesco Trotta
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Aureliano Ruggio
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Giuseppe P Piemontese
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Domenico Galante
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Myriana Ponzo
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Luigi M Biasucci
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Giovanna Liuzzo
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| | - Filippo Crea
- Department of Cardiovascular and Thoracic Sciences, Catholic University of the Sacred Heart, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli, 8-00168 Rome, Italy
| |
Collapse
|
16
|
Fike AJ, Kumova OK, Carey AJ. Dissecting the defects in the neonatal CD8 + T-cell response. J Leukoc Biol 2019; 106:1051-1061. [PMID: 31260598 DOI: 10.1002/jlb.5ru0319-105r] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 05/24/2019] [Accepted: 06/10/2019] [Indexed: 12/26/2022] Open
Abstract
The neonatal period presents a complex scenario where the threshold of reactivity toward colonizing microbiota, maternal antigens, autoantigens, and pathogens must be carefully moderated and balanced. CD8+ T cells are critical for the response against intracellular bacteria and viruses, but this immune compartment maintains altered function relative to adult counterparts because of the unique challenges which infants face. Here, we review our current understanding of the factors which may promote the attenuation and altered function of the neonatal CD8+ T-cell response and potential avenues for future study. Specifically, we have focused on the neonatal CD8+ T-cell ontogeny, memory formation, TCR structure and repertoire, TCR inhibitory receptors, and the clinical implications of altered neonatal CD8+ T-cell function. Special emphasis has been placed on examining the response of preterm neonates relative to term neonates and adults.
Collapse
Affiliation(s)
- Adam J Fike
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Ogan K Kumova
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Alison J Carey
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
Fike AJ, Kumova OK, Tardif VJ, Carey AJ. Neonatal influenza-specific effector CTLs retain elevated CD31 levels at the site of infection and have decreased IFN-γ production. J Leukoc Biol 2018; 105:539-549. [PMID: 30536476 DOI: 10.1002/jlb.4a0518-191r] [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: 05/11/2018] [Revised: 10/08/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022] Open
Abstract
The underlying mechanisms that regulate neonatal immune suppression are poorly characterized. CD31 (PECAM1) is highly expressed on neonatal lymphocytes and is a known modulator of TCR signaling. To further characterize the role of CD31 in the neonatal CTL response, 3-d and 7-d-old murine neonates were infected with influenza virus and compared to adults. The majority of the pulmonary viral-specific CTLs in the 3-d-old murine neonate retain CD31 expression, whereas adult CTLs have decreased CD31 expression. In addition, CD31+ neonatal viral-specific CTLs demonstrate decreased IFN-γ production, decreased proliferative capacity, and increased likelihood of death. At the peak of infection, sorted neonatal effector CTLs continue to transcribe CD31, indicating a developmental regulation of expression. To explore potential mechanisms for this reduced function, we compared the expression of the transcription factors Eomesodermin (Eomes) and T-bet; there was a significant increase in Eomes paired with a reduction in T-bet in CD31+ neonatal effector CTLs in the lung. Furthermore, in vitro stimulated neonatal CTLs significantly reduce IFN-γ production upon CD31 signaling. Altogether, these data indicate that neonatal CTLs may retain elevated levels of CD31 to maintain peripheral T cell suppression during the bridge to ex utero life.
Collapse
Affiliation(s)
- Adam J Fike
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.,Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Ogan K Kumova
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Virginie J Tardif
- Division of Infectious Diseases and HIV Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Alison J Carey
- Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.,Pediatrics, St. Christopher's Hospital for Children, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Newman DK, Fu G, McOlash L, Schauder D, Newman PJ, Cui W, Rao S, Johnson BD, Gershan JA, Riese MJ. Frontline Science: PECAM-1 (CD31) expression in naïve and memory, but not acutely activated, CD8 + T cells. J Leukoc Biol 2018; 104:883-893. [PMID: 30063264 PMCID: PMC6195461 DOI: 10.1002/jlb.2hi0617-229rrr] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 06/20/2017] [Accepted: 07/01/2018] [Indexed: 01/16/2023] Open
Abstract
Inhibitory cell surface proteins on T cells are often dynamically regulated, which contributes to their physiologic function. PECAM-1 (CD31) is an inhibitory receptor that facilitates TGF-β-mediated suppression of T cell activity. It is well established in CD4+ T cells that PECAM-1 is expressed in naïve recent thymic emigrants, but is down-regulated after acute T cell activation and absent from memory cells. The extent to which PECAM-1 expression is similarly regulated in CD8+ T cells is much less well characterized. We evaluated T cells recovered from mice after infection with a model intracellular pathogen and determined that, in CD8+ T cells, PECAM-1 expression was strongly down-regulated during acute infection but re-expressed to intermediate levels in memory cells. Down-regulation of PECAM-1 expression in CD8+ T cells was transcriptionally regulated and affected by the strength and nature of TCR signaling. PECAM-1 was also detected on the surface of human activated/memory CD8+ , but not CD4+ T cells. These data demonstrate that PECAM-1 expression is dynamically regulated, albeit differently, in both CD4+ and CD8+ T cells. Furthermore, unlike memory CD4+ T cells, memory CD8+ T cells retain PECAM-1 expression and have the potential to be modulated by this inhibitory receptor.
Collapse
Affiliation(s)
- Debra K. Newman
- Blood Research Institute, Blood Center of Wisconsin, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
| | - Guoping Fu
- Blood Research Institute, Blood Center of Wisconsin, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
| | - Laura McOlash
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
| | - David Schauder
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
| | - Peter J. Newman
- Blood Research Institute, Blood Center of Wisconsin, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
| | - Weiguo Cui
- Blood Research Institute, Blood Center of Wisconsin, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
| | - Sridhar Rao
- Blood Research Institute, Blood Center of Wisconsin, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
| | - Bryon D. Johnson
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
| | - Jill A. Gershan
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
| | - Matthew J. Riese
- Blood Research Institute, Blood Center of Wisconsin, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
- Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, 8733 Watertown Plank Road, Milwaukee, WI, 53226
| |
Collapse
|
19
|
Villar J, Zhang H, Slutsky AS. Lung Repair and Regeneration in ARDS: Role of PECAM1 and Wnt Signaling. Chest 2018; 155:587-594. [PMID: 30392791 DOI: 10.1016/j.chest.2018.10.022] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Revised: 10/18/2018] [Accepted: 10/18/2018] [Indexed: 01/08/2023] Open
Abstract
ARDS is an acute inflammatory pulmonary process triggered by severe pulmonary and systemic insults to the alveolar-capillary membrane. This causes increased vascular permeability and the development of interstitial and alveolar protein-rich edema, leading to acute hypoxemic respiratory failure. Supportive treatment includes the use of lung-protective ventilatory strategies that decrease the work of breathing, can improve oxygenation, and minimize ventilator-induced lung injury. Despite substantial advances in supportive measures, there are no specific pharmacologic treatments for ARDS, and the overall hospital mortality rate remains about 40% in most series. The pathophysiology of ARDS involves interactions among multiple mechanisms, including immune cell infiltration, cytokine storm, alveolar-capillary barrier disruption, cell apoptosis, and the development of fibrosis. Here we review some new developments in the molecular basis of lung injury, with a focus on possible novel pharmacologic interventions aimed at improving the outcomes of patients with ARDS. Our focus is on platelet-endothelial cell adhesion molecule-1, which contributes to the maintenance and restoration of vascular integrity following barrier disruption. We also highlight the wingless-related integration site signaling pathway, which appears to be a central mechanism for lung healing as well as for fibrotic development.
Collapse
Affiliation(s)
- Jesús Villar
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain; Multidisciplinary Organ Dysfunction Evaluation Research Network, Research Unit, Hospital Universitario Dr Negrin, Las Palmas de Gran Canaria, Spain; Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada
| | - Haibo Zhang
- Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada; Department of Anesthesia and Department of Physiology, University of Toronto, Toronto, Canada
| | - Arthur S Slutsky
- Keenan Research Center for Biomedical Sciences at the Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, Canada; Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
20
|
Hoang QT, Nuzzo A, Louedec L, Delbosc S, Andreata F, Khallou-Laschet J, Assadi M, Montravers P, Longrois D, Corcos O, Caligiuri G, Nicoletti A, Michel JB, Tran-Dinh A. Peptide binding to cleaved CD31 dampens ischemia/reperfusion-induced intestinal injury. Intensive Care Med Exp 2018; 6:27. [PMID: 30112663 PMCID: PMC6093833 DOI: 10.1186/s40635-018-0192-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Accepted: 07/30/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND CD31 is a key transmembrane neutrophil immunoregulatory receptor. Mesenteric ischemia/reperfusion-induced neutrophil activation leads to a massive cleavage and shedding of the most extracellular domains of CD31 into plasma, enhancing the deleterious effect of neutrophil activation. We have evaluated the preventive therapeutic potential of an engineered synthetic octapeptide (P8RI), which restores the inhibitory intracellular signaling of cleaved CD31, in an experimental model of acute mesenteric ischemia/reperfusion. METHODS In a randomized, controlled, and experimenter-blinded preclinical study, mesenteric ischemia/reperfusion (I/R) was induced in Wistar rats by superior mesenteric artery occlusion for 30 min followed by 4 h of reperfusion. Three groups of rats were compared: I/R + saline perfusion (I/R controls group, n = 7), I/R + preventive P8RI perfusion (P8RI group, n = 7), and sham-operated rats + saline perfusion (sham group, n = 7). RESULTS Compared with I/R controls, P8RI perfusion significantly decreased intestinal ischemia/reperfusion injury (Chiu's score, P = 0.01; epithelial area, P = 0.001) and bacterial translocation (plasma Escherichia coli DNA, P = 0.04) and could limit intestinal bleeding (P = 0.09). P8RI decreased neutrophil activation assessed by matrix metalloproteinase-9 release in plasma (P < 0.001) and in the intestinal wall, albeit without statistical significance (P = 0.06 and P = 0.058 for myeloperoxydase). Inhibition of CD31 cleavage from neutrophils could play a major role in the protective effects of P8RI (P < 0.0001). CONCLUSIONS Preventive administration of P8RI, a CD31-agonist peptide, could decrease I/R-induced intestinal injury by potentially limiting neutrophil activation.
Collapse
Affiliation(s)
- Quoc Thang Hoang
- INSERM LVTS U1148, Paris-Diderot, Université Sorbonne, Paris, France.,Department of Anesthesiology and Surgical Critical Care, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | - Alexandre Nuzzo
- INSERM LVTS U1148, Paris-Diderot, Université Sorbonne, Paris, France.,Structure d'URgences Vasculaires Intestinales (SURVI), CHU Beaujon, Clichy, France
| | - Liliane Louedec
- INSERM LVTS U1148, Paris-Diderot, Université Sorbonne, Paris, France
| | - Sandrine Delbosc
- INSERM LVTS U1148, Paris-Diderot, Université Sorbonne, Paris, France
| | | | | | - Maksud Assadi
- INSERM LVTS U1148, Paris-Diderot, Université Sorbonne, Paris, France.,Département d'anesthésie-réanimation, CHU Bichat-Claude Bernard, 46, rue Henri Huchard, 75877, Paris Cedex 18, France
| | - Philippe Montravers
- Département d'anesthésie-réanimation, CHU Bichat-Claude Bernard, 46, rue Henri Huchard, 75877, Paris Cedex 18, France.,INSERM UMR 1152, Paris-Diderot, Université Sorbonne, Paris, France
| | - Dan Longrois
- INSERM LVTS U1148, Paris-Diderot, Université Sorbonne, Paris, France.,Département d'anesthésie-réanimation, CHU Bichat-Claude Bernard, 46, rue Henri Huchard, 75877, Paris Cedex 18, France
| | - Olivier Corcos
- INSERM LVTS U1148, Paris-Diderot, Université Sorbonne, Paris, France.,Structure d'URgences Vasculaires Intestinales (SURVI), CHU Beaujon, Clichy, France
| | | | | | | | - Alexy Tran-Dinh
- INSERM LVTS U1148, Paris-Diderot, Université Sorbonne, Paris, France. .,Département d'anesthésie-réanimation, CHU Bichat-Claude Bernard, 46, rue Henri Huchard, 75877, Paris Cedex 18, France.
| |
Collapse
|
21
|
|
22
|
Scheible KM, Emo J, Laniewski N, Baran AM, Peterson DR, Holden-Wiltse J, Bandyopadhyay S, Straw AG, Huyck H, Ashton JM, Tripi KS, Arul K, Werner E, Scalise T, Maffett D, Caserta M, Ryan RM, Reynolds AM, Ren CL, Topham DJ, Mariani TJ, Pryhuber GS. T cell developmental arrest in former premature infants increases risk of respiratory morbidity later in infancy. JCI Insight 2018; 3:96724. [PMID: 29467329 DOI: 10.1172/jci.insight.96724] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 01/17/2018] [Indexed: 12/31/2022] Open
Abstract
The inverse relationship between gestational age at birth and postviral respiratory morbidity suggests that infants born preterm (PT) may miss a critical developmental window of T cell maturation. Despite a continued increase in younger PT survivors with respiratory complications, we have limited understanding of normal human fetal T cell maturation, how ex utero development in premature infants may interrupt normal T cell development, and whether T cell development has an effect on infant outcomes. In our longitudinal cohort of 157 infants born between 23 and 42 weeks of gestation, we identified differences in T cells present at birth that were dependent on gestational age and differences in postnatal T cell development that predicted respiratory outcome at 1 year of age. We show that naive CD4+ T cells shift from a CD31-TNF-α+ bias in mid gestation to a CD31+IL-8+ predominance by term gestation. Former PT infants discharged with CD31+IL8+CD4+ T cells below a range similar to that of full-term born infants were at an over 3.5-fold higher risk for respiratory complications after NICU discharge. This study is the first to our knowledge to identify a pattern of normal functional T cell development in later gestation and to associate abnormal T cell development with health outcomes in infants.
Collapse
Affiliation(s)
| | | | | | - Andrea M Baran
- Department of Biostatistics and Computational Biology, and
| | | | | | | | - Andrew G Straw
- Department of Biostatistics and Computational Biology, and
| | | | | | | | - Karan Arul
- Undergraduate Campus, University of Rochester, Rochester, New York, USA
| | | | | | | | | | - Rita M Ryan
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Anne Marie Reynolds
- Department of Pediatrics, State University of New York, University at Buffalo, Buffalo, New York, USA
| | - Clement L Ren
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | | | | | | |
Collapse
|
23
|
Early M, Schroeder WG, Unnithan R, Gilchrist JM, Muller WA, Schenkel A. Differential effect of Platelet Endothelial Cell Adhesion Molecule-1 (PECAM-1) on leukocyte infiltration during contact hypersensitivity responses. PeerJ 2017; 5:e3555. [PMID: 28713655 PMCID: PMC5507171 DOI: 10.7717/peerj.3555] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 06/18/2017] [Indexed: 11/26/2022] Open
Abstract
Background 2′–4′ Dinitrofluorobenzene (DNFB) induced contact hypersensitivity is an established model of contact sensitivity and leukocyte migration. Platelet Endothelial Cell Adhesion Molecule-1 (PECAM-1) deficient mice were used to examine the role of PECAM-1 in the migration capacity of several different leukocyte populations after primary and secondary application. Results γδ T lymphocytes, granulocytes, and Natural Killer cells were most affected by PECAM-1 deficiency at the primary site of application. γδ T lymphocytes, granulocytes, DX5+ Natural Killer cells, and, interestingly, effector CD4+ T lymphocytes were most affected by the loss of PECAM-1 at the secondary site of application. Conclusions PECAM-1 is used by many leukocyte populations for migration, but there are clearly differential effects on the usage by each subset. Further, the overall kinetics of each population varied between primary and secondary application, with large relative increases in γδ T lymphocytes during the secondary response.
Collapse
Affiliation(s)
- Merideth Early
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States of America
| | - William G Schroeder
- Department of Pediatrics, University of Colorado Health Sciences Center, Aurora, CO, United States of America
| | - Ranajana Unnithan
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States of America
| | - John M Gilchrist
- Department of Physiology, University of California, San Francisco, United States of America
| | - William A Muller
- Department of Pathology, Northwestern University, Chicago, IL, United States of America
| | - Alan Schenkel
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, United States of America
| |
Collapse
|
24
|
Characterization of CD31 expression on murine and human neonatal T lymphocytes during development and activation. Pediatr Res 2017; 82:133-140. [PMID: 28355204 PMCID: PMC5509503 DOI: 10.1038/pr.2017.81] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 03/11/2017] [Indexed: 01/26/2023]
Abstract
BackgroundCD31, expressed by the majority of the neonatal T-cell pool, is involved in modulation of T-cell receptor signaling by increasing the threshold for T-cell activation. Therefore, CD31 could modulate neonatal tolerance and adaptive immune responses.MethodsLymphocytes were harvested from murine neonates at different ages, human late preterm and term cord blood, and adult peripheral blood. Human samples were activated over a 5-day period to simulate acute inflammation. Mice were infected with influenza; lungs and spleens were harvested at days 6 and 9 post infection and analyzed by flow cytometry.ResultsCD31-expressing neonatal murine CD4+ and CD8a+ T cells increase over the first week of life. Upon in vitro stimulation, human infants' CD4+ and CD8a+ T cells shed CD31 faster in comparison with adults. In the context of acute infection, mice infected at 3 days of age have an increased number of naive and activated CD31+ T lymphocytes at the site of infection at days 6 and 9 post infection, as compared with those infected at 7 days of age; however, the opposite is true in the periphery.ConclusionDifferences in trafficking of CD31+ cytotoxic T lymphocytes (CTLs) during acute influenza infection could modulate tolerance and contribute to a dampened adaptive immune response in neonates.
Collapse
|
25
|
Sass FA, Schmidt-Bleek K, Ellinghaus A, Filter S, Rose A, Preininger B, Reinke S, Geissler S, Volk HD, Duda GN, Dienelt A. CD31+ Cells From Peripheral Blood Facilitate Bone Regeneration in Biologically Impaired Conditions Through Combined Effects on Immunomodulation and Angiogenesis. J Bone Miner Res 2017; 32:902-912. [PMID: 27976803 DOI: 10.1002/jbmr.3062] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 12/07/2016] [Accepted: 12/12/2016] [Indexed: 12/30/2022]
Abstract
Controlled revascularization and inflammation are key elements regulating endogenous regeneration after (bone) tissue trauma. Peripheral blood-derived cell subsets, such as regulatory T-helper cells and circulating (endothelial) progenitor cells, respectively, can support endogenous tissue healing, whereas effector T cells that are associated with an aged immune system can hinder bone regeneration. CD31 is expressed by diverse leukocytes and is well recognized as a marker of circulating endothelial (precursor) cells; however, CD31 is absent from the surface of differentiated effector T cells. Thus, we hypothesized that by separating the inhibitory fractions from the supportive fractions of circulating cells within the peripheral blood (PB) using the CD31 marker, bone regeneration in biologically compromised conditions, such as those observed in aged patients, could be improved. In support of our hypothesis, we detected an inverse correlation between CD31+ cells and effector T cells in the hematomas of human fracture patients, dependent on the age of the patient. Furthermore, we demonstrated the regenerative capacity of human PB-CD31+ cells in vitro. These findings were translated to a clinically relevant rat model of impaired bone healing. The transplantation of rat PB-CD31+ cells advanced bone tissue restoration in vivo and was associated with an early anti-inflammatory response, the stimulation of (re)vascularization, and reduced fibrosis. Interestingly, the depletion or enrichment of the highly abundant CD31+/14+ monocytes from the mixed CD31+ cell population diminished tissue regeneration at different levels, suggesting combined effects within the PB-CD31+ subsets. In summary, an intraoperative enrichment of PB-CD31+ cells might be a novel option to facilitate endogenous regeneration under biologically impaired situations by supporting immunomodulation and vascularization. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- F Andrea Sass
- Julius Wolff Institute (JWI) and Center for Musculoskeletal Surgery, Charité - University Medicine Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - University Medicine Berlin, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute (JWI) and Center for Musculoskeletal Surgery, Charité - University Medicine Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - University Medicine Berlin, Berlin, Germany
| | - Agnes Ellinghaus
- Julius Wolff Institute (JWI) and Center for Musculoskeletal Surgery, Charité - University Medicine Berlin, Berlin, Germany
| | - Sebastian Filter
- Julius Wolff Institute (JWI) and Center for Musculoskeletal Surgery, Charité - University Medicine Berlin, Berlin, Germany
| | - Alexander Rose
- Julius Wolff Institute (JWI) and Center for Musculoskeletal Surgery, Charité - University Medicine Berlin, Berlin, Germany
| | - Bernd Preininger
- Center for Musculoskeletal Surgery, Charité - University Medicine Berlin, Berlin, Germany
| | - Simon Reinke
- Julius Wolff Institute (JWI) and Center for Musculoskeletal Surgery, Charité - University Medicine Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - University Medicine Berlin, Berlin, Germany
| | - Sven Geissler
- Julius Wolff Institute (JWI) and Center for Musculoskeletal Surgery, Charité - University Medicine Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - University Medicine Berlin, Berlin, Germany
| | - Hans-Dieter Volk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - University Medicine Berlin, Berlin, Germany.,Institute of Medical Immunology, Charité - University Medicine Berlin, Berlin, Germany
| | - Georg N Duda
- Julius Wolff Institute (JWI) and Center for Musculoskeletal Surgery, Charité - University Medicine Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - University Medicine Berlin, Berlin, Germany
| | - Anke Dienelt
- Julius Wolff Institute (JWI) and Center for Musculoskeletal Surgery, Charité - University Medicine Berlin, Berlin, Germany.,Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - University Medicine Berlin, Berlin, Germany
| |
Collapse
|
26
|
Boeckh-Behrens T, Kleine J, Kaesmacher J, Zimmer C, Schirmer L, Simon S, Poppert H. The CD31 molecule: a possible neuroprotective agent in acute ischemic stroke? Thromb J 2017; 15:11. [PMID: 28413360 PMCID: PMC5390341 DOI: 10.1186/s12959-017-0134-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Accepted: 04/08/2017] [Indexed: 01/01/2023] Open
Abstract
Background The transmembrane receptor molecule CD31 is known to have immunomodulatory functions, suggesting a possible neuroprotective effect in the context of acute ischemic stroke by restricting an over-activation of secondary immunological processes. This study examines the density of CD31+ cells in mechanically extracted thrombi of stroke patients with the aim to test whether the occurrence of CD31+ cells was associated with a beneficial clinical outcome in those patients. Methods Thrombi of 122 consecutive patients with large anterior circulation stroke were collected during intracranial mechanical recanalization. Out of these, 86 immunostained specimens of adequate quality could be analysed. The density of CD31+ cells was quantified and compared with clinical outcome data of the affected patients. Results The density of CD31+ cells was positively related to early patient improvement (ΔNIHSS, r = 0.283, p = 0,012) with an even clearer relationship after exclusion of patients who died in the early hospital phase (r = 0.371, p = 0.001). This finding stayed stable also in the multivariate analysis after corrrection for other outcome-influencing factors (p = 0.049). Conclusion This study shows a stable relation between CD31+ cells and early clinical improvement of patients with acute ischemic stroke. This finding is in line with recent reports showing immunomodulatory and potential neuroprotective effects of CD31, suggesting that CD31 may be a promising neuroprotective agent in stroke patients.
Collapse
Affiliation(s)
- Tobias Boeckh-Behrens
- Department of Neuroradiology, University Hospital Rechts der Isar, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Justus Kleine
- Department of Neuroradiology, Vivantes Klinikum Neukölln, Rudowerstr. 48, 12351 Berlin, Germany
| | - Johannes Kaesmacher
- Department of Neuroradiology, University Hospital Rechts der Isar, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Claus Zimmer
- Department of Neuroradiology, University Hospital Rechts der Isar, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Lucas Schirmer
- Department of Neurology, University Hospital Rechts der Isar, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Sophie Simon
- Department of Neurology, University Hospital Rechts der Isar, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany
| | - Holger Poppert
- Department of Neurology, University Hospital Rechts der Isar, Technical University Munich, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
27
|
Batorov EV, Tikhonova MA, Kryuchkova IV, Sergeevicheva VV, Sizikova SA, Ushakova GY, Batorova DS, Gilevich AV, Ostanin AA, Shevela EY, Chernykh ER. CD4 + memory T cells retain surface expression of CD31 independently of thymic function in patients with lymphoproliferative disorders following autologous hematopoietic stem-cell transplantation. Int J Hematol 2017; 106:108-115. [PMID: 28293817 DOI: 10.1007/s12185-017-2214-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/09/2017] [Accepted: 03/10/2017] [Indexed: 11/30/2022]
Abstract
High-dose chemotherapy with autologous hematopoietic stem-cell transplantation (AHSCT) causes severe and long-lasting immunodeficiency in patients with lymphoproliferative disorders. The thymus begins to restore the T-cell repertoire approximately from the sixth month post-transplant. We assessed the dynamics of post-transplant recovery of CD4+CD45RA+CD31+ T cells, "recent thymic emigrants" (RTEs), and a poorly described subtype of CD4+CD45RA-CD31+ T cells in 90 patients with lymphoproliferative disorders following high-dose chemotherapy with AHSCT. Relative and absolute counts of CD4+CD31+ naïve and memory T cells were evaluated before AHSCT, at the day of engraftment, and 6- and 12-month post-transplant. The pre-transplant count of CD4+CD45RA+CD31+ T cells was lower than in healthy controls, and did not reach donors' values during the 12-month period. The pre-transplant number of CD4+CD45RA-CD31+ T cells was higher than in healthy controls and was restored rapidly following AHSCT. Post-transplant mediastinal radiotherapy reduced counts of RTEs and elongated recovery period. Non-thymic tissue irradiation did not reduce this subset. The obtained data indicate that homeostatic proliferation may decrease the significance of CD31 expression on CD4+CD45RA+ T cells as a marker of RTEs, and suggest that evaluation of RTEs recovery by flow cytometry requires an accurate gating strategy to exclude CD31+ memory T cells.
Collapse
Affiliation(s)
- Egor V Batorov
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation.
| | - Marina A Tikhonova
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation
| | - Irina V Kryuchkova
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation
| | - Vera V Sergeevicheva
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation
| | - Svetlana A Sizikova
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation
| | - Galina Y Ushakova
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation
| | - Dariya S Batorova
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation
| | - Andrey V Gilevich
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation
| | - Alexander A Ostanin
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation
| | - Ekaterina Y Shevela
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation
| | - Elena R Chernykh
- Research Institute of Fundamental and Clinical Immunology, 14 Yadrintsevskaya St, 630099, Novosibirsk, Russian Federation
| |
Collapse
|
28
|
Berthel A, Zoernig I, Valous NA, Kahlert C, Klupp F, Ulrich A, Weitz J, Jaeger D, Halama N. Detailed resolution analysis reveals spatial T cell heterogeneity in the invasive margin of colorectal cancer liver metastases associated with improved survival. Oncoimmunology 2017; 6:e1286436. [PMID: 28405518 PMCID: PMC5384380 DOI: 10.1080/2162402x.2017.1286436] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 01/13/2017] [Accepted: 01/20/2017] [Indexed: 12/15/2022] Open
Abstract
On a broader scale, T cell density and localization in colorectal cancer liver metastases have prognostic and predictive implications. As T cell distribution at higher resolutions has not been fully investigated, a detailed resolution analysis of T cell distribution was performed. Patient tissues were divided into 10 µm distance classes between the tumor border and adjacent normal liver. Thereby, distinct density patterns of T cell localization in relation to the malignant tissue could be detected. At a distance of 20 to 30 µm to the tumor, a decrease of CD3 T cells is common. Within this area, cytotoxic Granzyme B and CD8+ T cells were found to be significantly reduced as well as CD163 macrophages were increased and identified to be in close contact with T cells. Our data suggests a physical or functional border within this region. Survival analysis revealed improved overall survival in patients with high T cells numbers at the direct tumor border. Interestingly, the decreased T cells in the 20 to 30 µm region were also found to be significantly associated with improved survival. Consequently, the detailed localization of T cells, despite blockade, could be associated with improved clinical outcome. The high-resolution analysis represents new insights into relevant heterogenous T cell distributions especially related to clinical responses. As the paradoxical observation of localization-dependent prognostic relevance of T cell densities is only detectable by detailed spatial analyses, this investigation of spatial profiles at higher resolutions is suggested as a new biomarker for survival and response to therapies.
Collapse
Affiliation(s)
- Anna Berthel
- Clinical Cooperation Unit "Applied Tumor Immunity," National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Inka Zoernig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and University Hospital Heidelberg , Heidelberg, Germany
| | - Nektarios A Valous
- Clinical Cooperation Unit "Applied Tumor Immunity," National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ) , Heidelberg, Germany
| | - Christoph Kahlert
- Department of Surgery, University Hospital Dresden , Dresden, Germany
| | - Fee Klupp
- Department of Surgery, University Hospital Heidelberg , Heidelberg, Germany
| | - Alexis Ulrich
- Department of Surgery, University Hospital Heidelberg , Heidelberg, Germany
| | - Juergen Weitz
- Department of Surgery, University Hospital Dresden , Dresden, Germany
| | - Dirk Jaeger
- Clinical Cooperation Unit "Applied Tumor Immunity," National Center for Tumor Diseases (NCT) and German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Medical Oncology, National Center for Tumor Diseases (NCT) and University Hospital Heidelberg, Heidelberg, Germany
| | - Niels Halama
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and University Hospital Heidelberg , Heidelberg, Germany
| |
Collapse
|
29
|
Douaisi M, Resop RS, Nagasawa M, Craft J, Jamieson BD, Blom B, Uittenbogaart CH. CD31, a Valuable Marker to Identify Early and Late Stages of T Cell Differentiation in the Human Thymus. THE JOURNAL OF IMMUNOLOGY 2017; 198:2310-2319. [PMID: 28159903 DOI: 10.4049/jimmunol.1500350] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 01/06/2017] [Indexed: 12/21/2022]
Abstract
Although CD31 expression on human thymocytes has been reported, a detailed analysis of CD31 expression at various stages of T cell development in the human thymus is missing. In this study, we provide a global picture of the evolution of CD31 expression from the CD34+ hematopoietic precursor to the CD45RA+ mature CD4+ and CD8+ single-positive (SP) T cells. Using nine-color flow cytometry, we show that CD31 is highly expressed on CD34+ progenitors and stays high until the early double-positive stage (CD3-CD4+CD8α+β-). After β-selection, CD31 expression levels become low to undetectable. CD31 expression then increases and peaks on CD3highCD4+CD8+ double-positive thymocytes. However, following positive selection, CD31 expression differs dramatically between CD4+ and CD8+ lineages: homogeneously high on CD8 SP but lower or negative on CD4 SP cells, including a subset of CD45RA+CD31- mature CD4+ thymocytes. CD31 expression on TCRγδ thymocytes is very similar to that of CD4 SP cells. Remarkably, there is a substantial subset of semimature (CD45RA-) CD4 SP thymocytes that lack CD31 expression. Moreover, FOXP3+ and ICOS+ cells are overrepresented in this CD31- subpopulation. Despite this CD31-CD45RA- subpopulation, most egress-capable mature CD45RA+ CD4 SP thymocytes express CD31. The variations in CD31 expression appear to coincide with three major selection processes occurring during thymopoiesis: β-selection, positive selection, and negative selection. Considering the ability of CD31 to modulate the TCR's activation threshold via the recruitment of tyrosine phosphatases, our results suggest a significant role for CD31 during T cell development.
Collapse
Affiliation(s)
- Marc Douaisi
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095
| | - Rachel S Resop
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095
| | - Maho Nagasawa
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Joshua Craft
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095
| | - Beth D Jamieson
- Department of Medicine, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095.,University of California Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles, CA 90024.,University of California Los Angeles AIDS Institute and Center for AIDS Research, Los Angeles, CA 90095; and
| | - Bianca Blom
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Christel H Uittenbogaart
- Department of Microbiology, Immunology and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095; .,University of California Los Angeles Jonsson Comprehensive Cancer Center, Los Angeles, CA 90024.,University of California Los Angeles AIDS Institute and Center for AIDS Research, Los Angeles, CA 90095; and.,Department of Pediatrics, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA 90095
| |
Collapse
|
30
|
Završnik M, Kariž S, Makuc J, Šeruga M, Cilenšek I, Petrovič D. PECAM-1 Leu125Val (rs688) Polymorphism and Diabetic Nephropathy in Caucasians with Type 2 Diabetes Mellitus. Anal Cell Pathol (Amst) 2016; 2016:3152967. [PMID: 28116228 PMCID: PMC5225318 DOI: 10.1155/2016/3152967] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/07/2016] [Indexed: 12/30/2022] Open
Abstract
Objectives. Platelet endothelial cell adhesion molecule-1 (PECAM-1) plays a key role in the transendothelial migration of circulating leukocytes during inflammation and in the maintenance of vascular endothelial integrity. We hypothesized that genetic variation in PECAM-1 gene could be associated with diabetic nephropathy (DN) and with the level of soluble PECAM-1 in Caucasians with type 2 diabetes mellitus (T2DM). Design and Methods. We analyzed the rs688 single nucleotide polymorphism of PECAM-1 gene C373G (Leu125Val) at exon 3, which encodes the first extracellular Ig-like domain that mediates the homophilic binding of PECAM-1, in 276 T2DM subjects with documented DN (cases) and 375 T2DM subjects without DN (controls), using a polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) strategy. Level of plasma soluble PECAM-1 (sPECAM-1) was measured by ELISA in a subpopulation of 120 diabetics with DN. Results. We found no association between the Leu125Val polymorphism and DN in subjects with T2DM. Likewise, the Leu125Val polymorphism was not associated with serum sPECAM-1 levels in a subpopulation of 120 diabetics with DN. Conclusion. The Leu125Val polymorphism of PECAM-1 and the level of sPECAM-1 are not associated with DN in T2DM subjects of Slovenian origin.
Collapse
Affiliation(s)
- Matej Završnik
- University Medical Centre Maribor, Clinic for Internal Medicine, Department for Diabetes and Metabolic Diseases, Maribor, Slovenia
| | - Stojan Kariž
- General Hospital Izola, Department of Internal Medicine, Izola, Slovenia
| | - Jana Makuc
- General Hospital Slovenj Gradec, Department of Internal Medicine, Slovenj Gradec, Slovenia
| | - Maja Šeruga
- General Hospital Murska Sobota, Department of Internal Medicine, Murska Sobota, Slovenia
| | - Ines Cilenšek
- Faculty of Medicine, Institute of Histology and Embryology, University of Ljubljana, Ljubljana, Slovenia
| | - Daniel Petrovič
- Faculty of Medicine, Institute of Histology and Embryology, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
31
|
Castañeda DM, Salgado DM, Narváez CF. B cells naturally induced during dengue virus infection release soluble CD27, the plasma level of which is associated with severe forms of pediatric dengue. Virology 2016; 497:136-145. [DOI: 10.1016/j.virol.2016.07.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 07/15/2016] [Accepted: 07/18/2016] [Indexed: 01/13/2023]
|
32
|
Immune checkpoints and rheumatic diseases: what can cancer immunotherapy teach us? Nat Rev Rheumatol 2016; 12:593-604. [DOI: 10.1038/nrrheum.2016.131] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
33
|
Newman DK, Fu G, Adams T, Cui W, Arumugam V, Bluemn T, Riese MJ. The adhesion molecule PECAM-1 enhances the TGF-β-mediated inhibition of T cell function. Sci Signal 2016; 9:ra27. [PMID: 26956486 DOI: 10.1126/scisignal.aad1242] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Transforming growth factor-β (TGF-β) is an immunosuppressive cytokine that inhibits the proinflammatory functions of T cells, and it is a major factor in abrogating T cell activity against tumors. Canonical TGF-β signaling results in the activation of Smad proteins, which are transcription factors that regulate target gene expression. We found that the cell surface molecule platelet endothelial cell adhesion molecule-1 (PECAM-1) facilitated noncanonical (Smad-independent) TGF-β signaling in T cells. Subcutaneously injected tumor cells that are dependent on TGF-β-mediated suppression of immunity for growth grew more slowly in PECAM-1(-/-) mice than in their wild-type counterparts. T cells isolated from PECAM-1(-/-) mice demonstrated relative insensitivity to the TGF-β-dependent inhibition of interferon-γ (IFN-γ) production, granzyme B synthesis, and cellular proliferation. Similarly, human T cells lacking PECAM-1 demonstrated decreased sensitivity to TGF-β in a manner that was partially restored by reexpression of PECAM-1. Co-incubation of T cells with TGF-β and a T cell-activating antibody resulted in PECAM-1 phosphorylation on an immunoreceptor tyrosine-based inhibitory motif (ITIM) and the recruitment of the inhibitory Src homology 2 (SH2) domain-containing tyrosine phosphatase-2 (SHP-2). Such conditions also induced the colocalization of PECAM-1 with the TGF-β receptor complex as identified by coimmunoprecipitation, confocal microscopy, and proximity ligation assays. These studies indicate a role for PECAM-1 in enhancing the inhibitory functions of TGF-β in T cells and suggest that therapeutic targeting of the PECAM-1-TGF-β inhibitory axis represents a means to overcome TGF-β-dependent immunosuppression within the tumor microenvironment.
Collapse
Affiliation(s)
- Debra K Newman
- Blood Research Institute, BloodCenter of Wisconsin, 8727 Watertown Plank Road, Milwaukee, WI 53226, USA. Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA. Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Guoping Fu
- Blood Research Institute, BloodCenter of Wisconsin, 8727 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Tamara Adams
- Blood Research Institute, BloodCenter of Wisconsin, 8727 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Weiguo Cui
- Blood Research Institute, BloodCenter of Wisconsin, 8727 Watertown Plank Road, Milwaukee, WI 53226, USA. Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Vidhyalakshmi Arumugam
- Blood Research Institute, BloodCenter of Wisconsin, 8727 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Theresa Bluemn
- Blood Research Institute, BloodCenter of Wisconsin, 8727 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Matthew J Riese
- Blood Research Institute, BloodCenter of Wisconsin, 8727 Watertown Plank Road, Milwaukee, WI 53226, USA. Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, Milwaukee, WI 53226, USA. Division of Hematology and Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
34
|
Altered Profile of Circulating Endothelial-Derived Microparticles in Ventilator-Induced Lung Injury. Crit Care Med 2016; 43:e551-9. [PMID: 26308427 DOI: 10.1097/ccm.0000000000001280] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Pulmonary endothelial cell injury is central to the pathophysiology of acute lung injury. Mechanical ventilation can cause endothelial disruption and injury, even in the absence of preexisting inflammation. Platelet-endothelial cell adhesion molecule-1 is a transmembrane protein connecting adjacent endothelial cells. We hypothesized that injurious mechanical ventilation will increase circulating lung endothelial-derived microparticles, defined as microparticles positive for platelet-endothelial cell adhesion molecule-1, which could serve as potential biomarkers and mediators of ventilator-induced lung injury. DESIGN Prospective randomized, controlled, animal investigation. SETTING A hospital preclinical animal laboratory. SUBJECTS Forty-eight Sprague-Dawley rats. INTERVENTIONS Animals were randomly allocated to one of the three following ventilatory protocols for 4 hours: spontaneous breathing (control group), mechanical ventilation with low tidal volume (6 mL/kg), and mechanical ventilation with high tidal volume (20 mL/kg). In both mechanical ventilation groups, positive end-expiratory pressure of 2 cm H2O was applied. MEASUREMENTS AND MAIN RESULTS We analyzed histologic lung damage, gas exchange, wet-to-dry lung weight ratio, serum cytokines levels, circulating endothelial-derived microparticles, platelet-endothelial cell adhesion molecule-1 lung protein content, and immunohistochemistry. When compared with low-tidal volume mechanical ventilation, high-tidal volume ventilation increased lung edema score and caused gas-exchange deterioration. These changes were associated with a marked increased of circulating endothelial-derived microparticles and a reduction of platelet-endothelial cell adhesion molecule-1 protein levels in the high-tidal volume lungs (p < 0.0001). CONCLUSIONS There is an endothelial-derived microparticle profile associated with disease-specific features of ventilator-induced lung injury. This profile could serve both as a biomarker of acute lung injury and, potentially, as a mediator of systemic propagation of pulmonary inflammatory response.
Collapse
|
35
|
Abstract
Constitutive resistance to cell death induced by inflammatory stimuli activating the extrinsic pathway of apoptosis is a key feature of vascular endothelial cells (ECs). Although this property is central to the maintenance of the endothelial barrier during inflammation, the molecular mechanisms of EC protection from cell-extrinsic, proapoptotic stimuli have not been investigated. We show that the Ig-family member CD31, which is expressed by endothelial but not epithelial cells, is necessary to prevent EC death induced by TNF-α and cytotoxic T lymphocytes in vitro. Combined quantitative RT-PCR array and biochemical analysis show that, upon the engagement of the TNF receptor with TNF-α on ECs, CD31 becomes activated and, in turn, counteracts the proapoptotic transcriptional program induced by TNF-α via activation of the Erk/Akt pathway. Specifically, Akt activation by CD31 signals prevents the localization of the forkhead transcription factor FoxO3 to the nucleus, thus inhibiting transcription of the proapoptotic genes CD95/Fas and caspase 7 and de-repressing the expression of the antiapoptotic gene cFlar. Both CD31 intracellular immunoreceptor tyrosine-based inhibition motifs are required for its prosurvival function. In vivo, CD31 gene transfer is sufficient to recapitulate the cytoprotective mechanisms in CD31(-) pancreatic β cells, which become resistant to immune-mediated rejection when grafted in fully allogeneic recipients.
Collapse
|
36
|
Teteloshvili N, Kluiver J, van der Geest KSM, van der Lei RJ, Jellema P, Pawelec G, Brouwer E, Kroesen BJ, Boots AMH, van den Berg A. Age-Associated Differences in MiRNA Signatures Are Restricted to CD45RO Negative T Cells and Are Associated with Changes in the Cellular Composition, Activation and Cellular Ageing. PLoS One 2015; 10:e0137556. [PMID: 26360056 PMCID: PMC4567287 DOI: 10.1371/journal.pone.0137556] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 08/19/2015] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs (miRNAs) have emerged as important players in the regulation of T-cell functionality. However, comprehensive insight into the extent of age-related miRNA changes in T cells is lacking. We established miRNA expression patterns of CD45RO- naïve and CD45RO+ memory T-cell subsets isolated from peripheral blood cells from young and elderly individuals. Unsupervised clustering of the miRNA expression data revealed an age-related clustering in the CD45RO- T cells, while CD45RO+ T cells clustered based on expression of CD4 and CD8. Seventeen miRNAs showed an at least 2-fold up- or downregulation in CD45RO- T cells obtained from young as compared to old donors. Validation on the same and independent samples revealed a statistically significant age-related upregulation of miR-21, miR-223 and miR-15a. In a T-cell subset analysis focusing on known age-related phenotypic changes, we showed significantly higher miR-21 and miR-223 levels in CD8+CD45RO-CCR7- TEMRA compared to CD45RO-CCR7+ TNAIVE-cells. Moreover, miR-21 but not miR-223 levels were significantly increased in CD45RO-CD31- post-thymic TNAIVE cells as compared to thymic CD45RO-CD31+ TNAIVE cells. Upon activation of CD45RO- TNAIVE cells we observed a significant induction of miR-21 especially in CD4+ T cells, while miR-223 levels significantly decreased only in CD4+ T cells. Besides composition and activation-induced changes, we showed a borderline significant increase in miR-21 levels upon an increasing number of population doublings in CD4+ T-cell clones. Together, our results show that ageing related changes in miRNA expression are dominant in the CD45RO- T-cell compartment. The differential expression patterns can be explained by age related changes in T-cell composition, i.e. accumulation of CD8+ TEMRA and CD4+ post-thymic expanded CD31- T cells and by cellular ageing, as demonstrated in a longitudinal clonal culture model.
Collapse
Affiliation(s)
- Nato Teteloshvili
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Initiative on Healthy Ageing and Immune Longevity (GRAIL), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Joost Kluiver
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Kornelis S. M. van der Geest
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Initiative on Healthy Ageing and Immune Longevity (GRAIL), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Roelof Jan van der Lei
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Pytrick Jellema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Graham Pawelec
- Department of Internal Medicine II, Centre for Medical Research, University of Tübingen, Tübingen, Germany
- School of Science and Technology, Nottingham Trent University, Nottingham, United KIngdom
| | - Elisabeth Brouwer
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Initiative on Healthy Ageing and Immune Longevity (GRAIL), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart-Jan Kroesen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Initiative on Healthy Ageing and Immune Longevity (GRAIL), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annemieke M. H. Boots
- Department of Rheumatology and Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Initiative on Healthy Ageing and Immune Longevity (GRAIL), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anke van den Berg
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Groningen Research Initiative on Healthy Ageing and Immune Longevity (GRAIL), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
37
|
LI GANG, HAN ZONGLIN, DONG HEGUI, ZHANG XIA, KONG XIANGQIAN, JIN XING. Platelet endothelial cell adhesion molecule-1 gene 125C/G polymorphism is associated with deep vein thrombosis. Mol Med Rep 2015; 12:2203-10. [DOI: 10.3892/mmr.2015.3586] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Accepted: 12/19/2014] [Indexed: 11/06/2022] Open
|
38
|
Collier FM, Tang MLK, Martino D, Saffery R, Carlin J, Jachno K, Ranganathan S, Burgner D, Allen KJ, Vuillermin P, Ponsonby AL. The ontogeny of naïve and regulatory CD4(+) T-cell subsets during the first postnatal year: a cohort study. Clin Transl Immunology 2015; 4:e34. [PMID: 25859389 PMCID: PMC4386616 DOI: 10.1038/cti.2015.2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 01/21/2015] [Indexed: 12/26/2022] Open
Abstract
As there is limited knowledge regarding the longitudinal development and early ontogeny of naïve and regulatory CD4(+) T-cell subsets during the first postnatal year, we sought to evaluate the changes in proportion of naïve (thymic and central) and regulatory (resting and activated) CD4(+) T-cell populations during the first postnatal year. Blood samples were collected and analyzed at birth, 6 and 12 months of age from a population-derived sample of 130 infants. The proportion of naïve and regulatory CD4(+) T-cell populations was determined by flow cytometry, and the thymic and central naïve populations were sorted and their phenotype confirmed by relative expression of T cell-receptor excision circle DNA (TREC). At birth, the majority (94%) of CD4(+) T cells were naïve (CD45RA(+)), and of these, ~80% had a thymic naïve phenotype (CD31(+) and high TREC), with the remainder already central naïve cells (CD31(-) and low TREC). During the first year of life, the naïve CD4(+) T cells retained an overall thymic phenotype but decreased steadily. From birth to 6 months of age, the proportion of both resting naïve T regulatory cells (rTreg; CD4(+)CD45RA(+)FoxP3(+)) and activated Treg (aTreg, CD4(+)CD45RA(-)FoxP3(high)) increased markedly. The ratio of thymic to central naïve CD4(+) T cells was lower in males throughout the first postnatal year indicating early sexual dimorphism in immune development. This longitudinal study defines proportions of CD4(+) T-cell populations during the first year of postnatal life that provide a better understanding of normal immune development.
Collapse
Affiliation(s)
- Fiona M Collier
- Child Health Research Unit and Barwon Biomedical Research, University Hospital, Barwon Health , Geelong, Victoria, Australia ; School of Medicine, Deakin University , Waurn Ponds, Victoria, Australia ; Murdoch Childrens Research Institute , Parkville, Victoria, Australia
| | - Mimi L K Tang
- Murdoch Childrens Research Institute , Parkville, Victoria, Australia ; The Royal Childrens Hospital , Parkville, Victoria, Australia ; The University of Melbourne , Parkville, Victoria, Australia
| | - David Martino
- Murdoch Childrens Research Institute , Parkville, Victoria, Australia ; The University of Melbourne , Parkville, Victoria, Australia
| | - Richard Saffery
- Murdoch Childrens Research Institute , Parkville, Victoria, Australia ; The University of Melbourne , Parkville, Victoria, Australia
| | - John Carlin
- Murdoch Childrens Research Institute , Parkville, Victoria, Australia ; The University of Melbourne , Parkville, Victoria, Australia
| | - Kim Jachno
- Murdoch Childrens Research Institute , Parkville, Victoria, Australia
| | - Sarath Ranganathan
- Murdoch Childrens Research Institute , Parkville, Victoria, Australia ; The Royal Childrens Hospital , Parkville, Victoria, Australia ; The University of Melbourne , Parkville, Victoria, Australia
| | - David Burgner
- Murdoch Childrens Research Institute , Parkville, Victoria, Australia ; The University of Melbourne , Parkville, Victoria, Australia
| | - Katrina J Allen
- Murdoch Childrens Research Institute , Parkville, Victoria, Australia ; The Royal Childrens Hospital , Parkville, Victoria, Australia ; The University of Melbourne , Parkville, Victoria, Australia
| | - Peter Vuillermin
- Child Health Research Unit and Barwon Biomedical Research, University Hospital, Barwon Health , Geelong, Victoria, Australia ; School of Medicine, Deakin University , Waurn Ponds, Victoria, Australia ; Murdoch Childrens Research Institute , Parkville, Victoria, Australia
| | - Anne-Louise Ponsonby
- Murdoch Childrens Research Institute , Parkville, Victoria, Australia ; The Royal Childrens Hospital , Parkville, Victoria, Australia ; The University of Melbourne , Parkville, Victoria, Australia
| |
Collapse
|
39
|
Abstract
A successful pregnancy depends on a complex process that establishes fetomaternal tolerance. Seminal plasma is known to induce maternal immune tolerance to paternal alloantigens, but the seminal factors that regulate maternal immunity have yet to be characterized. Here, we show that a soluble form of CD38 (sCD38) released from seminal vesicles to the seminal plasma plays a crucial role in inducing tolerogenic dendritic cells and CD4(+) forkhead box P3(+) (Foxp3(+)) regulatory T cells (Tregs), thereby enhancing maternal immune tolerance and protecting the semiallogeneic fetus from resorption. The abortion rate in BALB/c females mated with C57BL/6 Cd38(-/-) males was high compared with that in females mated with Cd38(+/+) males, and this was associated with a reduced proportion of Tregs within the CD4(+) T-cell pool. Direct intravaginal injection of sCD38 to CBA/J pregnant mice at preimplantation increased Tregs and pregnancy rates in mice under abortive sonic stress from 48 h after mating until euthanasia. Thus, sCD38 released from seminal vesicles to the seminal plasma acts as an immunoregulatory factor to protect semiallogeneic fetuses from maternal immune responses.
Collapse
|
40
|
Resop RS, Uittenbogaart CH. Human T-Cell Development and Thymic Egress: An Infectious Disease Perspective. FORUM ON IMMUNOPATHOLOGICAL DISEASES AND THERAPEUTICS 2015; 6:33-49. [PMID: 28670486 PMCID: PMC5489135 DOI: 10.1615/forumimmundisther.2015014226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Emigration of mature naïve CD4 SP T cells from the human thymus to the periphery is not fully understood, although elucidation of the mechanisms that govern egress of T cells is crucial to understanding both basic immunology and the immune response in diseases such as HIV infection. Recent work has brought to light the requirement for sphingosine-1-phosphate (S1P) and its receptors in a variety of fields including mature naïve T-cell egress from the thymus of mice. We are examining the expression and function of this novel requisite T-cell egress receptor within the human thymus, characterizing changes observed in the expression and function of this receptor in infectious diseases. To perform this work, we use a variety of humanized murine models reviewed in this article. Future work in the field of T-cell egress, especially as it pertains to S1P receptors, should advance the fields of basic T-cell immunology and immunopathology and open new avenues for exploration into novel therapeutics.
Collapse
Affiliation(s)
- Rachel S. Resop
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
- Department of Pediatrics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
| | - Christel H. Uittenbogaart
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
- Department of Pediatrics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
- University of California at Los Angeles AIDS Institute, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, David Geffen Medical School at UCLA, Los Angeles, CA 90095
| |
Collapse
|
41
|
Flego D, Severino A, Trotta F, Previtero M, Ucci S, Zara C, Pedicino D, Massaro G, Biasucci LM, Liuzzo G, Crea F. Altered CD31 expression and activity in helper T cells of acute coronary syndrome patients. Basic Res Cardiol 2014; 109:448. [PMID: 25344833 DOI: 10.1007/s00395-014-0448-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 10/13/2014] [Accepted: 10/15/2014] [Indexed: 12/20/2022]
Abstract
In acute coronary syndrome (ACS), T cell abnormalities are associated to a worse outcome. Loss of inhibitory activity of CD31, an Ig-like adhesion molecule, on peripheral leukocytes has been found to enhance atherosclerosis in experimental models. In this study, we examined the expression of CD31 on T cells, and its role on TCR signaling in 35 patients with non-ST elevation ACS, in 35 patients with stable angina (SA), and in 35 controls. Furthermore, 10 ACS and 10 SA patients were re-analyzed at 1-year follow-up. Flow-cytometry analysis showed that in ACS patients, CD31 expression was reduced on total CD4(+) and CD4(+)CD28(null) (P < 0.001, ACS vs. SA), on naïve (P < 0.001, ACS vs. SA) and on central-memory and effector-memory CD4(+) T cells (P < 0.05, ACS vs. SA and controls). The immunomodulatory effect of CD31 on TCR signaling of CD4(+) and CD4(+)CD28(null) T cells, was lower in ACS than SA patients (P < 0.05, for both comparisons). At 1-year follow-up, CD31 expression and function increased in ACS becoming similar to that found in SA. CD31 recruitment in the immunological synapse was lower in ACS than controls (P = 0.012). Moreover, CD31 modulated MAPK signaling and reduced the expression of T bet and Rorγ-t, necessary for Th1 and Th17 differentiation. Finally, we studied TCR signaling in CD31(+) naïve and primed T cell subsets observing a different pattern of protein phosphorylation. A CD31-mediated regulatory pathway is enhanced in SA and temporarily downregulated in ACS. As CD31 modulates both T cell activation, by increasing the threshold for TCR stimulation, and T cell differentiation, it might represent a novel molecular target to treat T cell abnormalities in ACS.
Collapse
Affiliation(s)
- Davide Flego
- Institute of Cardiology, Catholic University, Largo A. Gemelli, 8, 00168, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Clement M, Fornasa G, Loyau S, Morvan M, Andreata F, Guedj K, Khallou-Laschet J, Larghi P, Le Roux D, Bismuth G, Chiocchia G, Hivroz C, Newman DK, Nicoletti A, Caligiuri G. Upholding the T cell immune-regulatory function of CD31 inhibits the formation of T/B immunological synapses in vitro and attenuates the development of experimental autoimmune arthritis in vivo. J Autoimmun 2014; 56:23-33. [PMID: 25277651 DOI: 10.1016/j.jaut.2014.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 09/03/2014] [Accepted: 09/06/2014] [Indexed: 11/29/2022]
Abstract
CD31, a trans-homophilic inhibitory receptor expressed on both T- and B-lymphocytes, drives the mutual detachment of interacting leukocytes. Intriguingly, T cell CD31 molecules relocate to the immunological synapse (IS), where the T and B cells establish a stable interaction. Here, we show that intact CD31 molecules, which are able to drive an inhibitory signal, are concentrated at the periphery of the IS but are excluded from the center of the IS. At this site, were the cells establish the closest contact, the CD31 molecules are cleaved, and most of the extracellular portion of the protein, including the trans-homophilic binding sites, is shed from the cell surface. T cells lacking CD31 trans-homophilic binding sites easily establish stable interactions with B cells; at the opposite, CD31 signaling agonists inhibit T/B IS formation as well as the ensuing helper T cell activation and function. Confocal microscopy and flow cytometry analysis of experimental T/B IS shows that the T cell inhibitory effects of CD31 agonists depend on SHP-2 signaling, which reduces the phosphorylation of ZAP70. The analysis of synovial tissue biopsies from patients affected by rheumatoid arthritis showed that T cell CD31 molecules are excluded from the center of the T/B cell synapses in vivo. Interestingly, the administration of CD31 agonists in vivo significantly attenuated the development of the clinical signs of collagen-induced arthritis in DBA1/J mice. Altogether, our data indicate that the T cell co-inhibitory receptor CD31 prevents the formation of functional T/B immunological synapses and that therapeutic strategies aimed at sustaining CD31 signaling will attenuate the development of autoimmune responses in vivo.
Collapse
Affiliation(s)
- Marc Clement
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Giulia Fornasa
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Stéphane Loyau
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Marion Morvan
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Francesco Andreata
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Kevin Guedj
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Jamila Khallou-Laschet
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Paola Larghi
- Institut Curie, Section Recherche, Inserm U932, F-75005 Paris, France
| | - Delphine Le Roux
- Inserm U1016, Institut Cochin, F-75014 Paris, France; Centre National de la Recherche Scientifique, UMR8104, F-75014 Paris, France; Université Paris Descartes, Paris F-75014, France
| | - Georges Bismuth
- Inserm U1016, Institut Cochin, F-75014 Paris, France; Centre National de la Recherche Scientifique, UMR8104, F-75014 Paris, France; Université Paris Descartes, Paris F-75014, France
| | - Gilles Chiocchia
- Inserm U987, Faculté des Sciences de la Santé "Simone Veil", F-78180 Saint-Quentin-en-Yvelines, France; Laboratoire d'Excellence "Inflamex", F-75018 Paris, France; Université Versailles-Saint-Quentin, F-78180 Saint-Quentin-en-Yvelines, France
| | - Claire Hivroz
- Institut Curie, Section Recherche, Inserm U932, F-75005 Paris, France
| | - Debra K Newman
- Blood Research Institute, Blood Center of Wisconsin, Milwaukee, WI 53226, USA
| | - Antonino Nicoletti
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France
| | - Giuseppina Caligiuri
- Institut National de la Santé et de la Recherche Médicale (Inserm), U1148, "Laboratory of Vascular Translational Science", F-75018 Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Faculté de Médecine, Site Xavier Bichat, F-75018 Paris, France; Département Hospitalo-Universitaire DHU "FIRE", F-75018 Paris, France.
| |
Collapse
|
43
|
CD31 is a key coinhibitory receptor in the development of immunogenic dendritic cells. Proc Natl Acad Sci U S A 2014; 111:E1101-10. [PMID: 24616502 DOI: 10.1073/pnas.1314505111] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
CD31 is a transhomophilic tyrosine-based inhibitory motif receptor and is expressed by both dendritic cells (DCs) and T lymphocytes. Previous studies have established that the engagement of CD31 drives immune-inhibitory signaling in T lymphocytes, but the effect exerted by CD31 signaling in DCs remains elusive. Here, we show that CD31 is a key coinhibitory receptor on stimulated DCs, favoring the development of tolerogenic functions and finally resulting in T-cell tolerance. The disruption of CD31 signaling favored the immunogenic maturation and migration of resident DCs to the draining lymph nodes. In contrast, sustaining the CD31/SHP-1 signaling during DC maturation resulted in reduced NF-κB nuclear translocation, expression of costimulatory molecules, and production of immunogenic cytokines (e.g., IL-12, IL-6), whereas the expression of TGF-β and IL-10 were increased. More importantly, CD31-conditioned DCs purified from the draining lymph nodes of ovalbumin-immunized mice favored the generation of antigen-specific regulatory T cells (CD25(+) forkhead box P3(+)) at the expense of effector (IFN-γ(+)) cells upon coculture with naive ovalbumin-specific CD4(+) T lymphocytes ex vivo. Finally, the adoptive transfer of CD31-conditioned myelin oligodendrocyte glycoprotein-loaded DCs carried immune tolerance against the subsequent development of MOG-induced experimental autoimmune encephalomyelitis in vivo. The key coinhibitory role exerted by CD31 on DCs highlighted by the present study may have important implications both in settings where the immunogenic function of DCs is desirable, such as infection and cancer, and in settings where tolerance-driving DCs are preferred, such as autoimmune diseases and transplantation.
Collapse
|
44
|
Soluble platelet-endothelial cell adhesion molecule-1, a biomarker of ventilator-induced lung injury. Crit Care 2014; 18:R41. [PMID: 24588994 PMCID: PMC4057495 DOI: 10.1186/cc13754] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 02/25/2014] [Indexed: 11/27/2022] Open
Abstract
Introduction Endothelial cell injury is an important component of acute lung injury. Platelet-endothelial cell adhesion molecule-1 (PECAM1) is a transmembrane protein that connects endothelial cells to one another and can be detected as a soluble, truncated protein (sPECAM1) in serum. We hypothesized that injurious mechanical ventilation (MV) leads to shedding of PECAM1 from lung endothelial cells resulting in increasing sPECAM1 levels in the systemic circulation. Methods We studied 36 Sprague–Dawley rats in two prospective, randomized, controlled studies (healthy and septic) using established animal models of ventilator-induced lung injury. Animals (n = 6 in each group) were randomized to spontaneous breathing or two MV strategies: low tidal volume (VT) (6 ml/kg) and high-VT (20 ml/kg) on 2 cmH2O of positive end-expiratory pressure (PEEP). In low-VT septic animals, 10 cmH2O of PEEP was applied. We performed pulmonary histological and physiological evaluation and measured lung PECAM1 protein content and serum sPECAM1 levels after four hours ventilation period. Results High-VT MV caused severe lung injury in healthy and septic animals, and decreased lung PECAM1 protein content (P < 0.001). Animals on high-VT had a four- to six-fold increase of mean sPECAM1 serum levels than the unventilated counterpart (35.4 ± 10.4 versus 5.6 ± 1.7 ng/ml in healthy rats; 156.8 ± 47.6 versus 35.6 ± 12.6 ng/ml in septic rats) (P < 0.0001). Low-VT MV prevented these changes. Levels of sPECAM1 in healthy animals on high-VT MV paralleled the sPECAM1 levels of non-ventilated septic animals. Conclusions Our findings suggest that circulating sPECAM1 may represent a promising biomarker for the detection and monitoring of ventilator-induced lung injury.
Collapse
|
45
|
Marelli-Berg FM, Clement M, Mauro C, Caligiuri G. An immunologist's guide to CD31 function in T-cells. J Cell Sci 2013; 126:2343-52. [PMID: 23761922 DOI: 10.1242/jcs.124099] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Although it is expressed by all leukocytes, including T-, B-lymphocytes and dendritic cells, the immunoglobulin-like receptor CD31 is generally regarded by immunologists as a marker of endothelial cell lineage that lacks an established functional role in adaptive immunity. This perception has recently been challenged by studies that reveal a key role for this molecule in the regulation of T-cell homeostasis, effector function and trafficking. The complexity of the biological functions of CD31 results from the integration of its adhesive and signaling functions in both the immune and vascular systems. Signaling by means of CD31 is induced by homophilic engagement during the interactions of immune cells and is mediated by phosphatase recruitment or activation through immunoreceptor tyrosine inhibitory motifs (ITIMs) that are located in its cytoplasmic tail. Loss of CD31 function is associated with excessive immunoreactivity and susceptibility to cytotoxic killing. Here, we discuss recent findings that have brought to light a non-redundant, complex role for this molecule in the regulation of T-cell-mediated immune responses, with large impact on our understanding of immunity in health and disease.
Collapse
Affiliation(s)
- Federica M Marelli-Berg
- William Harvey Research Institute, Barts' and The London School of Medicine, Queen Mary, University of London, Charterhouse Square, London EC1M 6BQ, UK.
| | | | | | | |
Collapse
|
46
|
Platelet endothelial cell adhesion molecule 1 deficiency misguides venous thrombus resolution. Blood 2013; 122:3376-84. [DOI: 10.1182/blood-2013-04-499558] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Key Points
PECAM-1 deficiency misguides venous thrombus resolution. PECAM-1 cell surface shedding occurs at the site of venous thrombosis.
Collapse
|
47
|
Saunders JAH, Estes KA, Kosloski LM, Allen HE, Dempsey KM, Torres-Russotto DR, Meza JL, Santamaria PM, Bertoni JM, Murman DL, Ali HH, Standaert DG, Mosley RL, Gendelman HE. CD4+ regulatory and effector/memory T cell subsets profile motor dysfunction in Parkinson's disease. J Neuroimmune Pharmacol 2012; 7:927-38. [PMID: 23054369 PMCID: PMC3515774 DOI: 10.1007/s11481-012-9402-z] [Citation(s) in RCA: 218] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 08/27/2012] [Indexed: 02/06/2023]
Abstract
Animal models and clinical studies have linked the innate and adaptive immune system to the pathology of Parkinson’s disease (PD). Despite such progress, the specific immune responses that influence disease progression have eluded investigators. Herein, we assessed relationships between T cell phenotype and function with PD progression. Peripheral blood lymphocytes from two separate cohorts, a discovery cohort and a validation cohort, totaling 113 PD patients and 96 age- and environment-matched caregivers were examined by flow cytometric analysis and T cell proliferation assays. Increased effector/memory T cells (Tem), defined as CD45RO+ and FAS+ CD4+ T cells and decreased CD31+ and α4β7+ CD4+ T cells were associated with progressive Unified Parkinson’s Disease Rating Scale III scores. However, no associations were seen between immune biomarkers and increased age or disease duration. Impaired abilities of regulatory T cells (Treg) from PD patients to suppress effector T cell function was observed. These data support the concept that chronic immune stimulation, notably Tem activation and Treg dysfunction is linked to PD pathobiology and disease severity, but not disease duration. The association of T cell phenotypes with motor symptoms provides fresh avenues for novel biomarkers and therapeutic designs.
Collapse
Affiliation(s)
- Jessica A Hutter Saunders
- Department of Pharmacology and Experimental Neuroscience, Center for Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Kishore M, Ma L, Cornish G, Nourshargh S, Marelli-Berg FM. Primed T cell responses to chemokines are regulated by the immunoglobulin-like molecule CD31. PLoS One 2012; 7:e39433. [PMID: 22724015 PMCID: PMC3378580 DOI: 10.1371/journal.pone.0039433] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Accepted: 05/22/2012] [Indexed: 01/22/2023] Open
Abstract
CD31, an immunoglobulin-like molecule expressed by leukocytes and endothelial cells, is thought to contribute to the physiological regulation T cell homeostasis due to the presence of two immunotyrosine-based inhibitory motifs in its cytoplasmic tail. Indeed, loss of CD31 expression leads to uncontrolled T cell-mediated inflammation in a variety of experimental models of disease and certain CD31 polymorphisms correlate with increased disease severity in human graft-versus-host disease and atherosclerosis. The molecular mechanisms underlying CD31-mediated regulation of T cell responses have not yet been clarified. We here show that CD31-mediated signals attenuate T cell chemokinesis both in vitro and in vivo. This effect selectively affects activated/memory T lymphocytes, in which CD31 is clustered on the cell membrane where it segregates to the leading edge. We provide evidence that this molecular segregation, which does not occur in naïve T lymphocytes, might lead to cis-CD31 engagement on the same membrane and subsequent interference with the chemokine-induced PI3K/Akt signalling pathway. We propose that CD31-mediated modulation of memory T cell chemokinesis is a key mechanism by which this molecule contributes to the homeostatic regulation of effector T cell immunity.
Collapse
Affiliation(s)
- Madhav Kishore
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | | | | | | | | |
Collapse
|
49
|
|
50
|
Fornasa G, Clement M, Groyer E, Gaston AT, Khallou-Laschet J, Morvan M, Guedj K, Kaveri SV, Tedgui A, Michel JB, Nicoletti A, Caligiuri G. A CD31-derived peptide prevents angiotensin II-induced atherosclerosis progression and aneurysm formation. Cardiovasc Res 2012; 94:30-7. [PMID: 22293851 DOI: 10.1093/cvr/cvs076] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
AIMS The loss of the inhibitory receptor CD31 on peripheral T lymphocytes is associated with the incidence of atherosclerotic complications such as abdominal aortic aneurysms (AAA) in patients and plaque thrombosis in mice. However, we have recently discovered that a small fragment of extracellular CD31 remains expressed on the surface of the apparently 'CD31-negative' T-cells and that it is possible to restore the CD31-mediated T-cell inhibition in vivo by using a synthetic CD31-derived peptide. Here, we wanted to evaluate the therapeutic potential of the peptide in an experimental model of accelerated atherosclerosis and AAA formation. METHODS AND RESULTS The effect of the murine CD31-derived peptide (aa 551-574, 1.5 mg/kg/day, sc) was evaluated on the extent of atherosclerotic plaques and the incidence of AAA in 28-week-old apolipoprotein E knockout mice (male, n ≥ 8/group) submitted to chronic angiotensin II infusion. The therapeutic mechanisms of the peptide were assessed by evaluating its effect on immune cell functions in vivo and in vitro. The prevalence of angiotensin II-induced AAA correlated with the loss of extracellular CD31 on T-cells. CD31 peptide treatment reduced both aneurysm formation and plaque size (P < 0.05 vs. control). Protection was associated with reduced perivascular leucocyte infiltration and T-cell activation in vivo. Functional in vitro studies showed that the peptide is able to suppress both T-cell and macrophage activation. CONCLUSION CD31 peptides could represent a new class of drugs intended to prevent the inflammatory cell processes, such as those underlying progression of atherosclerosis and development of AAA.
Collapse
Affiliation(s)
- Giulia Fornasa
- INSERM, UMRS 698, 46 rue Henri Huchard, F-75018 Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|