1
|
Li Y, Xing J, Qin L, Zhang C, Yang Z, Qiu M. Mechanism of isorhynchophylline in lipopolysaccharide-induced acute lung injury based on proteomic technology. Front Pharmacol 2024; 15:1397498. [PMID: 38873411 PMCID: PMC11169627 DOI: 10.3389/fphar.2024.1397498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/07/2024] [Indexed: 06/15/2024] Open
Abstract
Isorhynchophylline (IRN), a tetracyclic indole alkaloid, has anti-inflammatory and antioxidant activities against cardiovascular diseases and central nervous system disorders. Acute lung injury (ALI) is a manifestation of inflammation concentrated in the lungs and has a high incidence rate and mortality The purpose of this study is to explain the mechanism of IRN in the treatment of acute lung injury and to provide a new scheme for clinical treatment. The experimental mice were divided into three groups: CTRL, LPS, LPS+IRN. The mouse model of ALI was established by inhaling LPS solution through nose. After continuous administration of IRN solution for 7 days, the mice in LPS+IRN group were killed and the lung tissue was collected for detection. Proteomic (Data are available via ProteomeXchange with identifier PXD050432) results showed that 5727 proteins were detected in mouse lung tissues, and 16 proteins were screened out. IRN could reverse the trend of these differential proteins. In addition, IRN can act on integrin αM to reduce neutrophil recruitment and thereby produce anti-inflammatory effects and may suppress neutrophil migration through the leukocyte transendothelial migration pathway. TUNEL and RT-PCR experiments revealed that LPS-induced ALI in mice increases the apoptosis of lung tissues, damage to alveolar epithelial cells and levels of inflammatory factors. Treatment with IRN can repair tissues, improve lung tissue pathology and reduce lung inflammation.
Collapse
Affiliation(s)
- Yaru Li
- Department of Pharmacy, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Junfeng Xing
- Department of Computer Science and Technology, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Ling Qin
- First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Chuanming Zhang
- Department of Pharmacy, Baotou Medical College, Baotou, Inner Mongolia, China
| | - Zheng Yang
- First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - Min Qiu
- Department of Pharmacy, Baotou Medical College, Baotou, Inner Mongolia, China
| |
Collapse
|
2
|
Li YY, Wang XY, Li Y, Wang XM, Liao J, Wang YZ, Hong H, Yi W, Chen J. Targeting CD43 optimizes cancer immunotherapy through reinvigorating antitumor immune response in colorectal cancer. Cell Oncol (Dordr) 2023; 46:777-791. [PMID: 36920728 DOI: 10.1007/s13402-023-00794-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 03/16/2023] Open
Abstract
PURPOSE Colorectal cancer (CRC) is one of the most common malignancies worldwide, with dramatically increasing incidence and mortality for decades. However, current therapeutic strategies for CRC, including chemotherapies and immunotherapies, have only demonstrated limited efficacy. Here, we report a novel immune molecule, CD43, that can regulate the tumor immune microenvironment (TIME) and serves as a promising target for CRC immunotherapy. METHODS The correlation of CD43 expression with CRC patient prognosis was revealed by public data analysis. CD43 knockout (KO) CRC cell lines were generated by CRISPR-Cas9 technology, and a syngenetic murine CRC model was established to investigate the in vivo function of CD43. The TIME was analyzed via immunohistochemical staining, flow cytometry and RNA-seq. Immune functions were investigated by depletion of immune subsets in vivo and T-cell functional assays in vitro, including T-cell priming, cytotoxicity, and chemotaxis experiments. RESULTS In this study, we found that high expression of CD43 was correlated with poor survival of CRC patients and the limited infiltration of CD8+ T cells in human CRC tissues. Importantly, CD43 expressed on tumor cells, rather than host cells, promoted tumor progression in a syngeneic tumor model. Loss of CD43 facilitated the infiltration of immune cells and immunological memory in the TIME of CRC tumors. Mechanistically, the protumor effect of CD43 depends on T cells, thereby attenuating T-cell-mediated cytotoxicity and cDC1-mediated antigen-specific T-cell activation. Moreover, targeting CD43 synergistically improved PD-L1 blockade immunotherapy for CRC. CONCLUSION Our findings revealed that targeting tumor-intrinsic CD43 could activate the antitumor immune response and provide particular value for optimized cancer immunotherapy by regulating the TIME in CRC patients.
Collapse
Affiliation(s)
- Yi-Yi Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Zhongshan School of Medicine, Sun Yat- sen University, Guangzhou, China.,Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xin-Yu Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Zhongshan School of Medicine, Sun Yat- sen University, Guangzhou, China.,Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yan Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Zhongshan School of Medicine, Sun Yat- sen University, Guangzhou, China.,Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiu-Mei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Zhongshan School of Medicine, Sun Yat- sen University, Guangzhou, China.,Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jing Liao
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Ying-Zhao Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hai Hong
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Wei Yi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Zhongshan School of Medicine, Sun Yat- sen University, Guangzhou, China.
| | - Jun Chen
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China. .,Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China. .,Center for Precision Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
3
|
CD43 sialoglycoprotein modulates cardiac inflammation and murine susceptibility to Trypanosoma cruzi infection. Sci Rep 2019; 9:8628. [PMID: 31197200 PMCID: PMC6565700 DOI: 10.1038/s41598-019-45138-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022] Open
Abstract
CD43 (leukosialin) is a large sialoglycoprotein abundantly expressed on the surface of most cells from the hematopoietic lineage. CD43 is directly involved in the contact between cells participating in a series of events such as signaling, adherence and host parasite interactions. In this study we examined the role of CD43 in the immune response against Trypanosoma cruzi, the protozoan parasite that causes Chagas’ disease, a potential life-threatening illness endemic in 21 Latin American countries according to the WHO. The acute stage of infection is marked by intense parasitemia and cardiac tissue parasitism, resulting in the recruitment of inflammatory cells and acute damage to the heart tissue. We show here that CD43−/− mice were more resistant to infection due to increased cytotoxicity of antigen specific CD8+ T cells and reduced inflammatory infiltration in the cardiac tissue, both contributing to lower cardiomyocyte damage. In addition, we demonstrate that the induction of acute myocarditis involves the engagement of CD43 cytoplasmic tripeptide sequence KRR to ezrin-radixin-moiesin cytoskeletal proteins. Together, our results show the participation of CD43 in different events involved in the pathogenesis of T. cruzi infection, contributing to a better overall understanding of the mechanisms underlying the pathogenesis of acute chagasic cardiomyopathy.
Collapse
|
4
|
Malaker SA, Pedram K, Ferracane MJ, Bensing BA, Krishnan V, Pett C, Yu J, Woods EC, Kramer JR, Westerlind U, Dorigo O, Bertozzi CR. The mucin-selective protease StcE enables molecular and functional analysis of human cancer-associated mucins. Proc Natl Acad Sci U S A 2019; 116:7278-7287. [PMID: 30910957 PMCID: PMC6462054 DOI: 10.1073/pnas.1813020116] [Citation(s) in RCA: 182] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mucin domains are densely O-glycosylated modular protein domains that are found in a wide variety of cell surface and secreted proteins. Mucin-domain glycoproteins are known to be key players in a host of human diseases, especially cancer, wherein mucin expression and glycosylation patterns are altered. Mucin biology has been difficult to study at the molecular level, in part, because methods to manipulate and structurally characterize mucin domains are lacking. Here, we demonstrate that secreted protease of C1 esterase inhibitor (StcE), a bacterial protease from Escherichia coli, cleaves mucin domains by recognizing a discrete peptide- and glycan-based motif. We exploited StcE's unique properties to improve sequence coverage, glycosite mapping, and glycoform analysis of recombinant human mucins by mass spectrometry. We also found that StcE digests cancer-associated mucins from cultured cells and from ascites fluid derived from patients with ovarian cancer. Finally, using StcE, we discovered that sialic acid-binding Ig-type lectin-7 (Siglec-7), a glycoimmune checkpoint receptor, selectively binds sialomucins as biological ligands, whereas the related receptor Siglec-9 does not. Mucin-selective proteolysis, as exemplified by StcE, is therefore a powerful tool for the study of mucin domain structure and function.
Collapse
Affiliation(s)
- Stacy A Malaker
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Kayvon Pedram
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | | | - Barbara A Bensing
- Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, San Francisco, CA 94143
| | - Venkatesh Krishnan
- Stanford Women's Cancer Center, Division of Gynecologic Oncology, Stanford University, Stanford, CA 94305
| | - Christian Pett
- Leibniz-Institut für Analytische Wissenschaften (ISAS), 44227 Dortmund, Germany
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden
| | - Jin Yu
- Leibniz-Institut für Analytische Wissenschaften (ISAS), 44227 Dortmund, Germany
| | - Elliot C Woods
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Jessica R Kramer
- Department of Bioengineering, University of Utah, Salt Lake City, UT 84112
| | - Ulrika Westerlind
- Leibniz-Institut für Analytische Wissenschaften (ISAS), 44227 Dortmund, Germany
- Department of Chemistry, Umeå University, 901 87 Umeå, Sweden
| | - Oliver Dorigo
- Stanford Women's Cancer Center, Division of Gynecologic Oncology, Stanford University, Stanford, CA 94305
| | - Carolyn R Bertozzi
- Department of Chemistry, Stanford University, Stanford, CA 94305;
- Howard Hughes Medical Institute, Stanford, CA 94305
| |
Collapse
|
5
|
Velázquez FE, Anastasiou M, Carrillo-Salinas FJ, Ngwenyama N, Salvador AM, Nevers T, Alcaide P. Sialomucin CD43 regulates T helper type 17 cell intercellular adhesion molecule 1 dependent adhesion, apical migration and transendothelial migration. Immunology 2019; 157:52-69. [PMID: 30690734 DOI: 10.1111/imm.13047] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 12/07/2018] [Accepted: 01/15/2019] [Indexed: 01/10/2023] Open
Abstract
T helper type 17 lymphocytes (Th17 cells) infiltrate the central nervous system (CNS), induce inflammation and demyelination and play a pivotal role in the pathogenesis of multiple sclerosis. Sialomucin CD43 is highly expressed in Th17 cells and mediates adhesion to endothelial selectin (E-selectin), an initiating step in Th17 cell recruitment to sites of inflammation. CD43-/- mice have impaired Th17 cell recruitment to the CNS and are protected from experimental autoimmune encephalomyelitis (EAE), the mouse model of multiple sclerosis. However, E-selectin is dispensable for the development of EAE, in contrast to intercellular and vascular cell adhesion molecules (ICAM-1 and VCAM-1). We report that CD43-/- mice have decreased demyelination and T-cell infiltration, but similar up-regulation of ICAM-1 and VCAM-1 in the spinal cord, compared with wild-type (WT) mice, at the initiation of EAE. CD43-/- Th17 cells have impaired adhesion to ICAM-1 under flow conditions in vitro, despite having similar expression of LFA-1, the main T-cell ligand for ICAM-1, as WT Th17 cells. Regardless of the route of integrin activation, CD43-/- Th17 cell firm arrest on ICAM-1 was comparable to that of WT Th17 cells, but CD43-/- Th17 cells failed to optimally apically migrate on immobilized ICAM-1-coated coverslips and endothelial cells, and to transmigrate under shear flow conditions in an ICAM-1-dependent manner. Collectively, these findings unveil novel roles for CD43, facilitating adhesion of Th17 cells to ICAM-1 and modulating apical and transendothelial migration, as mechanisms potentially responsible for Th17 cell recruitment to sites of inflammation such as the CNS.
Collapse
Affiliation(s)
| | - Marina Anastasiou
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA.,Laboratory of Autoimmunity and Inflammation, University of Crete Medical School, Crete, Greece
| | | | - Njabulo Ngwenyama
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Ane M Salvador
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Tania Nevers
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Pilar Alcaide
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| |
Collapse
|
6
|
Fay KT, Chihade DB, Chen CW, Klingensmith NJ, Lyons JD, Ramonell K, Liang Z, Coopersmith CM, Ford ML. Increased mortality in CD43-deficient mice during sepsis. PLoS One 2018; 13:e0202656. [PMID: 30226896 PMCID: PMC6143188 DOI: 10.1371/journal.pone.0202656] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/07/2018] [Indexed: 11/18/2022] Open
Abstract
CD43 is a large transmembrane protein involved in T cell activation. Previous studies of CD43-/- mice in viral models have demonstrated a role for CD43 in Th1/Th2 skewing, activation of Foxp3+ Treg, and T cell apoptosis. However, the role of CD43 during sepsis has never been tested. Thus, we interrogated the role of CD43 during sepsis using a murine cecal ligation and puncture (CLP) model, and found that CD43-/- mice demonstrated significantly worsened mortality compared to B6 mice following CLP. Phenotypic analysis of splenocytes isolated 24 h after septic insult revealed significantly increased apoptosis of central memory cells in both CD4+ and CD8+ T cell compartments in CD43-/- septic mice compared to WT septic mice. Furthermore, CD43-/-septic mice exhibited a prominent Th2 skewing following sepsis relative to WT septic mice, as evidenced by a significant decrease in the frequency of IL-2+ CXCR3+ TH1 cells as a significant increase in the frequency of IL-4+ CCR4+ TH2 cells. Finally, septic CD43-/- animals contained significantly fewer CD25+ Foxp3+ TReg cells as compared to WT septic animals. Importantly, depleting CD25+ Treg eliminated the increased mortality observed in CD43-/- mice. Taken together, these data demonstrate an important role of CD43 in modulating immune dysregulation and mortality following sepsis.
Collapse
Affiliation(s)
- Katherine T. Fay
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Deena B. Chihade
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Ching-Wen Chen
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Nathan J. Klingensmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - John D. Lyons
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Kimberly Ramonell
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Zhe Liang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Craig M. Coopersmith
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Critical Care Center, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Mandy L. Ford
- Department of Surgery, Emory University School of Medicine, Atlanta, GA, United States of America
- Emory Transplant Center, Emory University School of Medicine, Atlanta, GA, United States of America
- * E-mail:
| |
Collapse
|
7
|
Margoles LM, Mittal R, Klingensmith NJ, Lyons JD, Liang Z, Serbanescu MA, Wagener ME, Coopersmith CM, Ford ML. Chronic Alcohol Ingestion Delays T Cell Activation and Effector Function in Sepsis. PLoS One 2016; 11:e0165886. [PMID: 27861506 PMCID: PMC5115670 DOI: 10.1371/journal.pone.0165886] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 10/19/2016] [Indexed: 12/21/2022] Open
Abstract
Sepsis is the leading cause of death in intensive care units in the US, and it is known that chronic alcohol use is associated with higher incidence of sepsis, longer ICU stays, and higher mortality from sepsis. Both sepsis and chronic alcohol use are associated with immune deficits such as decreased lymphocyte numbers, impaired innate immunity, delayed-type hypersensitivity reactions, and susceptibility to infections; however, understanding of specific pathways of interaction or synergy between these two states of immune dysregulation is lacking. This study therefore sought to elucidate mechanisms underlying the immune dysregulation observed during sepsis in the setting of chronic alcohol exposure. Using a murine model of chronic ethanol ingestion followed by sepsis induction via cecal ligation and puncture, we determined that while CD4+ and CD8+ T cells isolated from alcohol fed mice eventually expressed the same cellular activation markers (CD44, CD69, and CD43) and effector molecules (IFN-γ, TNF) as their water fed counterparts, there was an overall delay in the acquisition of these phenotypes. This early lag in T cell activation was associated with significantly reduced IL-2 production at a later timepoint in both the CD4+ and CD8+ T cell compartments in alcohol sepsis, as well as with a reduced accumulation of CD8dim activated effectors. Taken together, these data suggest that delayed T cell activation may result in qualitative differences in the immune response to sepsis in the setting of chronic alcohol ingestion.
Collapse
Affiliation(s)
- Lindsay M. Margoles
- Division of Infectious Diseases, Emory University, Atlanta, GA, United States of America
| | - Rohit Mittal
- Department of Surgery, Emory University, Atlanta, GA, United States of America
| | | | - John D. Lyons
- Department of Surgery, Emory University, Atlanta, GA, United States of America
| | - Zhe Liang
- Department of Surgery, Emory University, Atlanta, GA, United States of America
| | - Mara A. Serbanescu
- Department of Surgery, Emory University, Atlanta, GA, United States of America
| | - Maylene E. Wagener
- Department of Surgery, Emory University, Atlanta, GA, United States of America
- Emory Transplant Center, Emory University, Atlanta, GA, United States of America
| | - Craig M. Coopersmith
- Department of Surgery, Emory University, Atlanta, GA, United States of America
- Emory Critical Care Center, Emory University, Atlanta, GA, United States of America
| | - Mandy L. Ford
- Department of Surgery, Emory University, Atlanta, GA, United States of America
- Emory Transplant Center, Emory University, Atlanta, GA, United States of America
| |
Collapse
|
8
|
Modak M, Majdic O, Cejka P, Jutz S, Puck A, Gerwien JG, Steinberger P, Zlabinger GJ, Strobl H, Stöckl J. Engagement of distinct epitopes on CD43 induces different co-stimulatory pathways in human T cells. Immunology 2016; 149:280-296. [PMID: 27392084 PMCID: PMC5046061 DOI: 10.1111/imm.12642] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Revised: 06/07/2016] [Accepted: 06/24/2016] [Indexed: 12/30/2022] Open
Abstract
Co‐receptors, being either co‐stimulatory or co‐inhibitory, play a pivotal role in T‐cell immunity. Several studies have indicated that CD43, one of the abundant T‐cell surface glycoproteins, acts not only as a potent co‐receptor but also as a negative regulator for T‐cell activation. Here we demonstrate that co‐stimulation of human peripheral blood (PB) T cells through two distinct CD43 epitopes recognized by monoclonal antibodies (mAb) CD43‐6E5 (T6E5‐act) and CD43‐10G7 (T10G7‐act) potently induced T‐cell proliferation. However, T‐cell co‐stimulation through two CD43 epitopes differentially regulated activation of nuclear factor of activated T cells (NFAT) and nuclear factor‐κB (NF‐κB) transcription factors, T‐cell cytokine production and effector function. T6E5‐act produced high levels of interleukin‐22 (IL‐22) and interferon‐γ (IFN‐γ) similar to T cells activated via CD28 (TCD28‐act), whereas T10G7‐act produced low levels of inflammatory cytokines but higher levels of regulatory cytokines transforming growth factor‐β (TGF‐β) and interleukin‐35 (IL‐35). Compared with T6E5‐act or to TCD28‐act, T10G7‐act performed poorly in response to re‐stimulation and further acquired a T‐cell suppressive function. T10G7‐act did not directly inhibit proliferation of responder T cells, but formed stable heterotypic clusters with dendritic cells (DC) via CD2 to constrain activation of responder T cells. Together, our data demonstrate that CD43 is a unique and polarizing regulator of T‐cell function.
Collapse
Affiliation(s)
- Madhura Modak
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Otto Majdic
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Petra Cejka
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Sabrina Jutz
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Alexander Puck
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Jens G Gerwien
- Biopharmaceuticals Research Unit, Inflammation Biology, Novo Nordisk A/S, Måløv, Denmark
| | - Peter Steinberger
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Gerhard J Zlabinger
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Institute of Pathophysiology and Immunology, Centre of Molecular Medicine, Medical University of Graz, Graz, Austria
| | - Johannes Stöckl
- Institute of Immunology, Centre for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
9
|
Velázquez F, Grodecki-Pena A, Knapp A, Salvador AM, Nevers T, Croce K, Alcaide P. CD43 Functions as an E-Selectin Ligand for Th17 Cells In Vitro and Is Required for Rolling on the Vascular Endothelium and Th17 Cell Recruitment during Inflammation In Vivo. THE JOURNAL OF IMMUNOLOGY 2015; 196:1305-1316. [PMID: 26700769 DOI: 10.4049/jimmunol.1501171] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 11/10/2015] [Indexed: 12/20/2022]
Abstract
Endothelial E- and P-selectins mediate lymphocyte trafficking in inflammatory processes by interacting with lymphocyte selectin ligands. These are differentially expressed among different T cell subsets and function alone or in cooperation to mediate T cell adhesion. In this study, we characterize the expression and functionality of E-selectin ligands in Th type 17 lymphocytes (Th17 cells) and report that CD43 functions as a Th17 cell E-selectin ligand in vitro that mediates Th17 cell rolling on the vascular endothelium and recruitment in vivo. We demonstrate Th17 cells express CD44, P-selectin glycoprotein ligand (PSGL)-1, and CD43. Few PSGL-1(-/-)CD43(-/-) Th17 cells accumulated on E-selectin under shear flow conditions compared with wild-type cells. CD43(-/-) Th17 cell accumulation on E-selectin was impaired as compared with wild-type and PSGL-1(-/-), and similar to that observed for PSGL-1(-/-)CD43(-/-) Th17 cells, indicating that CD43 alone is a dominant ligand for E-selectin. Notably, this finding is Th17 cell subset specific because CD43 requires cooperation with PSGL-1 in Th1 cells for binding to E-selectin. In vivo, Th17 cell recruitment into the air pouch was reduced in CD43(-/-) mice in response to CCL20 or TNF-α, and intravital microscopy studies demonstrated that CD43(-/-) Th17 cells had impaired rolling on TNF-α-treated microvessels. Furthermore, CD43(-/-) mice were protected from experimental autoimmune encephalomyelitis and had impaired recruitment of Th17 cells in the spinal cord. Our findings demonstrate that CD43 is a major E-selectin ligand in Th17 cells that functions independent of PSGL-1, and they suggest that CD43 may hold promise as a therapeutic target to modulate Th17 cell recruitment.
Collapse
Affiliation(s)
- Francisco Velázquez
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111.,Sackler School of Biomedical Sciences Immunology program, Tufts University School of Medicine, Boston, MA 02111
| | - Anna Grodecki-Pena
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111
| | - Andrew Knapp
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111
| | - Ane M Salvador
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111
| | - Tania Nevers
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111
| | - Kevin Croce
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115
| | - Pilar Alcaide
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA 02111.,Sackler School of Biomedical Sciences Immunology program, Tufts University School of Medicine, Boston, MA 02111.,800 Washington St, Box #80, Boston, MA 02111
| |
Collapse
|
10
|
Dang PT, Bui Q, D'Souza CS, Orian JM. Modelling MS: Chronic-Relapsing EAE in the NOD/Lt Mouse Strain. Curr Top Behav Neurosci 2015; 26:143-177. [PMID: 26126592 DOI: 10.1007/7854_2015_378] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Modelling complex disorders presents considerable challenges, and multiple sclerosis (MS) is no exception to this rule. The aetiology of MS is unknown, and its pathophysiology is poorly understood. Moreover, the last two decades have witnessed a dramatic revision of the long-held view of MS as an inflammatory demyelinating white matter disease. Instead, it is now regarded as a global central nervous system (CNS) disorder with a neurodegenerative component. Currently, there is no animal model recapitulating MS immunopathogenesis. Available models are based on autoimmune-mediated demyelination, denoted experimental autoimmune encephalomyelitis (EAE) or virally or chemically induced demyelination. Of these, the EAE model has been the most commonly used. It has been extensively improved since its first description and now exists as a number of variants, including genetically modified and humanized versions. Nonetheless, EAE is a distinct disease, and each variant models only certain facets of MS. Whilst the search for more refined MS models must continue, it is important to further explore where mechanisms underlying EAE provide proof-of-principle for those driving MS pathogenesis. EAE variants generated with the myelin component myelin oligodendrocyte glycoprotein (MOG) have emerged as the preferred ones, because in this particular variant disease is associated with both T- and B-cell effector mechanisms, together with demyelination. MOG-induced EAE in the non-obese diabetic (NOD) mouse strain exhibits a chronic-relapsing EAE clinical profile and high disease incidence. We describe the generation of this variant, its contribution to the understanding of MS immune and pathogenetic mechanisms and potential for evaluation of candidate therapies.
Collapse
Affiliation(s)
- Phuc T Dang
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Quyen Bui
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Claretta S D'Souza
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - Jacqueline M Orian
- Department of Biochemistry and La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia.
| |
Collapse
|
11
|
Galindo-Albarrán AO, Ramírez-Pliego O, Labastida-Conde RG, Melchy-Pérez EI, Liquitaya-Montiel A, Esquivel-Guadarrama FR, Rosas-Salgado G, Rosenstein Y, Santana MA. CD43 signals prepare human T cells to receive cytokine differentiation signals. J Cell Physiol 2014; 229:172-80. [PMID: 24328034 DOI: 10.1002/jcp.24430] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
T cells are increasingly used for passive immunotherapy and bone marrow transplantation. Proper ex-vivo management of the cells is important for the desired therapeutic effects. For differentiation into effector cells of the Th1 and Th2 phenotypes, T-cells require signals from IFNγ and IL-4, respectively. Naïve cells have an extremely low expression of the specific receptors that recognize these cytokines, indicating that in order to differentiate, cells need to perceive other signals that will enable them to sense the cytokine milieu. CD43 has been proposed as one of the molecules that make the initial contacts with antigen presenting cells. We report here that in cord blood, adult naïve and total human T cells, CD43 signals induced the expression of both IFNγ and IL-4 receptors, mediate their capping, increased their signaling and augmented differentiation mediated by these receptors. CD43 signals also stimulated the expression of IFNγ and in neonatal cells that of IL-4 as well. These data demonstrate an important role for CD43 signals in T-cell preparedness for differentiation into effector cells.
Collapse
|
12
|
Nestor CE, Barrenäs F, Wang H, Lentini A, Zhang H, Bruhn S, Jörnsten R, Langston MA, Rogers G, Gustafsson M, Benson M. DNA methylation changes separate allergic patients from healthy controls and may reflect altered CD4+ T-cell population structure. PLoS Genet 2014; 10:e1004059. [PMID: 24391521 PMCID: PMC3879208 DOI: 10.1371/journal.pgen.1004059] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 11/11/2013] [Indexed: 12/30/2022] Open
Abstract
Altered DNA methylation patterns in CD4+ T-cells indicate the importance of epigenetic mechanisms in inflammatory diseases. However, the identification of these alterations is complicated by the heterogeneity of most inflammatory diseases. Seasonal allergic rhinitis (SAR) is an optimal disease model for the study of DNA methylation because of its well-defined phenotype and etiology. We generated genome-wide DNA methylation (Npatients = 8, Ncontrols = 8) and gene expression (Npatients = 9, Ncontrols = 10) profiles of CD4+ T-cells from SAR patients and healthy controls using Illumina's HumanMethylation450 and HT-12 microarrays, respectively. DNA methylation profiles clearly and robustly distinguished SAR patients from controls, during and outside the pollen season. In agreement with previously published studies, gene expression profiles of the same samples failed to separate patients and controls. Separation by methylation (Npatients = 12, Ncontrols = 12), but not by gene expression (Npatients = 21, Ncontrols = 21) was also observed in an in vitro model system in which purified PBMCs from patients and healthy controls were challenged with allergen. We observed changes in the proportions of memory T-cell populations between patients (Npatients = 35) and controls (Ncontrols = 12), which could explain the observed difference in DNA methylation. Our data highlight the potential of epigenomics in the stratification of immune disease and represents the first successful molecular classification of SAR using CD4+ T cells. T-cells, a type of white blood cell, are an important part of the immune-system in humans. T-cells allow us to adapt our immune-response to the various infectious agents we encounter during life. However, T-cells can also cause disease when they target the body's own cells, e.g. Psoriasis, or when they react to a harmless particle or ‘antigen’, i.e. allergy. Much evidence supports an environmental, or ‘epigenetic’, component to allergy. Surprisingly, although allergy is viewed as a T-cell disease with an epigenetic component, no studies have identified epigenetic differences between healthy individuals and allergic individuals. Using a state-of-the-art genome-wide approach, we found that we could clearly and robustly separate allergic patients from healthy controls. It is often assumed that these changes reflect changes in DNA methylation in a given type of cell; however such differences can also result from different mixtures of T-cell subtypes in the samples. Indeed, we found that allergic patients had different proportions of T-cell sub-types compared to healthy controls. These changes in T-cell proportions may explain the difference in DNA methylation profile we observed between patients and controls. Our study is the first successful molecular classification of allergy using CD4+ T cells.
Collapse
Affiliation(s)
- Colm E. Nestor
- The Centre for Individualized Medicine, Linköping University Hospital, Linköping University, Linköping, Sweden
- * E-mail:
| | - Fredrik Barrenäs
- The Centre for Individualized Medicine, Linköping University Hospital, Linköping University, Linköping, Sweden
| | - Hui Wang
- The Centre for Individualized Medicine, Linköping University Hospital, Linköping University, Linköping, Sweden
- Department of Pediatrics, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Antonio Lentini
- The Centre for Individualized Medicine, Linköping University Hospital, Linköping University, Linköping, Sweden
| | - Huan Zhang
- The Centre for Individualized Medicine, Linköping University Hospital, Linköping University, Linköping, Sweden
| | - Sören Bruhn
- The Centre for Individualized Medicine, Linköping University Hospital, Linköping University, Linköping, Sweden
| | - Rebecka Jörnsten
- Mathematical Sciences, Chalmers University of Technology, University of Gothenburg, Gothenburg, Sweden
| | - Michael A. Langston
- Department of Electrical Engineering and Computer Science, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Gary Rogers
- National Institute for Computational Sciences, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Mika Gustafsson
- The Centre for Individualized Medicine, Linköping University Hospital, Linköping University, Linköping, Sweden
| | - Mikael Benson
- The Centre for Individualized Medicine, Linköping University Hospital, Linköping University, Linköping, Sweden
| |
Collapse
|
13
|
Dozmorov MG, Wren JD, Alarcón-Riquelme ME. Epigenomic elements enriched in the promoters of autoimmunity susceptibility genes. Epigenetics 2013; 9:276-85. [PMID: 24213554 DOI: 10.4161/epi.27021] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Genome-wide association studies have identified a number of autoimmune disease-susceptibility genes. Whether or not these loci share any regulatory or functional elements, however, is an open question. Finding such common regulators is of considerable research interest in order to define systemic therapeutic targets. The growing amount of experimental genomic annotations, particularly those from the ENCODE project, provide a wealth of opportunities to search for such commonalities. We hypothesized that regulatory commonalities might not only delineate a regulatory landscape predisposing to autoimmune diseases, but also define functional elements distinguishing specific diseases. We further investigated if, and how, disease-specific epigenomic elements can identify novel genes yet to be associated with the diseases. We evaluated transcription factors, histone modifications, and chromatin state data obtained from the ENCODE project for statistically significant over- or under-representation in the promoters of genes associated with Systemic Lupus Erythematosus (SLE), Rheumatoid Arthritis (RA), and Systemic Sclerosis (SSc). We identified BATF, BCL11A, IRF4, NFkB, PAX5, and PU.1 as transcription factors over-represented in SLE- and RA-susceptibility gene promoters. H3K4me1 and H3K4me2 epigenomic marks were associated with SLE susceptibility genes, and H3K9me3 was common to both SLE and RA. In contrast to a transcriptionally active signature in SLE and RA, SSc-susceptibility genes were depleted in activating epigenomic elements. Using epigenomic elements enriched in SLE and RA, we identified additional immune and B cell signaling-related genes with the same elements in their promoters. Our analysis suggests common and disease-specific epigenomic elements that may define novel therapeutic targets for controlling aberrant activation of autoimmune susceptibility genes.
Collapse
Affiliation(s)
- Mikhail G Dozmorov
- Oklahoma Medical Research Foundation; Arthritis and Clinical Immunology Research Program; Oklahoma City, OK USA
| | - Jonathan D Wren
- Oklahoma Medical Research Foundation; Arthritis and Clinical Immunology Research Program; Oklahoma City, OK USA; University of Oklahoma Health Sciences Center; Department of Biochemistry and Molecular Biology; Oklahoma City, OK USA
| | - Marta E Alarcón-Riquelme
- Oklahoma Medical Research Foundation; Arthritis and Clinical Immunology Research Program; Oklahoma City, OK USA; GENYO; Centre for Genomics and Oncological Research; Pfizer; University of Granada; Andalusian Regional Government; Granada, Spain
| |
Collapse
|
14
|
Zhou HF, Yan H, Cannon JL, Springer LE, Green JM, Pham CTN. CD43-mediated IFN-γ production by CD8+ T cells promotes abdominal aortic aneurysm in mice. THE JOURNAL OF IMMUNOLOGY 2013; 190:5078-85. [PMID: 23585675 DOI: 10.4049/jimmunol.1203228] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD43 is a glycosylated surface protein abundantly expressed on lymphocytes. Its role in immune responses has been difficult to clearly establish, with evidence supporting both costimulatory and inhibitory functions. In addition, its contribution to disease pathogenesis remains elusive. Using a well-characterized murine model of elastase-induced abdominal aortic aneurysm (AAA) that recapitulates many key features of the human disease, we established that the presence of CD43 on T cells is required for AAA formation. Moreover, we found that IFN-γ-producing CD8(+) T cells, but not CD4(+) T cells, promote the development of aneurysm by enhancing cellular apoptosis and matrix metalloprotease activity. Reconstitution with IFN-γ-producing CD8(+) T cells or recombinant IFN-γ promotes the aneurysm phenotype in CD43(-/-) mice, whereas IFN-γ antagonism abrogates disease in wild-type animals. Furthermore, we showed that the presence of CD43 with an intact cytoplasmic domain capable of binding to ezrin-radixin-moesin cytoskeletal proteins is essential for optimal in vivo IFN-γ production by T cells and aneurysm formation. We have thus identified a robust physiologic role for CD43 in a relevant animal model and established an important in vivo function for CD43-dependent regulation of IFN-γ production. These results further suggest that IFN-γ antagonism or selective blockade of CD43(+)CD8(+) T cell activities merits further investigation for immunotherapy in AAA.
Collapse
Affiliation(s)
- Hui-fang Zhou
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | |
Collapse
|
15
|
Donos N, Hamlet S, Lang NP, Salvi GE, Huynh-Ba G, Bosshardt DD, Ivanovski S. Gene expression profile of osseointegration of a hydrophilic compared with a hydrophobic microrough implant surface. Clin Oral Implants Res 2011; 22:365-72. [DOI: 10.1111/j.1600-0501.2010.02113.x] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
16
|
Cannon JL, Mody PD, Blaine KM, Chen EJ, Nelson AD, Sayles LJ, Moore TV, Clay BS, Dulin NO, Shilling RA, Burkhardt JK, Sperling AI. CD43 interaction with ezrin-radixin-moesin (ERM) proteins regulates T-cell trafficking and CD43 phosphorylation. Mol Biol Cell 2011; 22:954-63. [PMID: 21289089 PMCID: PMC3069020 DOI: 10.1091/mbc.e10-07-0586] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
CD43 interaction with ERM proteins regulates CD43 phosphorylation and T-cell migration. CD43 phosphorylation can also drive CD43 localization in T-cells independently of ERM association. Cell polarization is a key feature of cell motility, driving cell migration to tissues. CD43 is an abundantly expressed molecule on the T-cell surface that shows distinct localization to the migrating T-cell uropod and the distal pole complex (DPC) opposite the immunological synapse via association with the ezrin-radixin-moesin (ERM) family of actin regulatory proteins. CD43 regulates multiple T-cell functions, including T-cell activation, proliferation, apoptosis, and migration. We recently demonstrated that CD43 regulates T-cell trafficking through a phosphorylation site at Ser-76 (S76) within its cytoplasmic tail. Using a phosphorylation-specific antibody, we now find that CD43 phosphorylation at S76 is enhanced by migration signals. We further show that CD43 phosphorylation and normal T-cell trafficking depend on CD43 association with ERM proteins. Interestingly, mutation of S76 to mimic phosphorylation enhances T-cell migration and CD43 movement to the DPC while blocking ERM association, showing that CD43 movement can occur in the absence of ERM binding. We also find that protein kinase Cθ can phosphorylate CD43. These results show that while CD43 binding to ERM proteins is crucial for S76 phosphorylation, CD43 movement and regulation of T-cell migration can occur through an ERM-independent, phosphorylation–dependent mechanism.
Collapse
Affiliation(s)
- J L Cannon
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Garcia GG, Miller RA. Age-related defects in the cytoskeleton signaling pathways of CD4 T cells. Ageing Res Rev 2011; 10:26-34. [PMID: 19941976 DOI: 10.1016/j.arr.2009.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Revised: 11/18/2009] [Accepted: 11/18/2009] [Indexed: 11/25/2022]
Abstract
It has been postulated that the cytoskeleton controls many aspects of T cell function, including activation, proliferation and apoptosis. Recent advances in our understanding of F-actin polymerization and the Ezrin-Radixin-Moesin (ERM) family of cytoskeleton signal proteins have provided new insights into immunological synapse formation during T cell activation. During aging there is a significant decline of T cell function largely attributable to declines in activation of CD4 T cells and defects in the formation of the immunological synapse. Here we discuss recent progress in the understanding of how aging alters F-actin and ERM proteins in mouse CD4 T cells, and the implications of these changes for the T cell activation process.
Collapse
|
18
|
Zhao Y, Tong J, He D, Pendyala S, Evgeny B, Chun J, Sperling AI, Natarajan V. Role of lysophosphatidic acid receptor LPA2 in the development of allergic airway inflammation in a murine model of asthma. Respir Res 2009; 10:114. [PMID: 19930563 PMCID: PMC2788521 DOI: 10.1186/1465-9921-10-114] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2009] [Accepted: 11/20/2009] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Lysophosphatidic acid (LPA) plays a critical role in airway inflammation through G protein-coupled LPA receptors (LPA1-3). We have demonstrated that LPA induced cytokine and lipid mediator release in human bronchial epithelial cells. Here we provide evidence for the role of LPA and LPA receptors in Th2-dominant airway inflammation. METHODS Wild type, LPA1 heterozygous knockout mice (LPA1+/-), and LPA2 heterozygous knockout mice (LPA2+/-) were sensitized with inactivated Schistosoma mansoni eggs and local antigenic challenge with Schistosoma mansoni soluble egg Ag (SEA) in the lungs. Bronchoalveolar larvage (BAL) fluids and lung tissues were collected for analysis of inflammatory responses. Further, tracheal epithelial cells were isolated and challenged with LPA. RESULTS BAL fluids from Schistosoma mansoni egg-sensitized and challenged wild type mice (4 days of challenge) showed increase of LPA level (approximately 2.8 fold), compared to control mice. LPA2+/- mice, but not LPA1+/- mice, exposed to Schistosoma mansoni egg revealed significantly reduced cell numbers and eosinophils in BAL fluids, compared to challenged wild type mice. Both LPA2+/- and LPA1+/- mice showed decreases in bronchial goblet cells. LPA2+/- mice, but not LPA1+/- mice showed the decreases in prostaglandin E2 (PGE2) and LPA levels in BAL fluids after SEA challenge. The PGE2 production by LPA was reduced in isolated tracheal epithelial cells from LPA2+/- mice. These results suggest that LPA and LPA receptors are involved in Schistosoma mansoni egg-mediated inflammation and further studies are proposed to understand the role of LPA and LPA receptors in the inflammatory process.
Collapse
Affiliation(s)
- Yutong Zhao
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Jiankun Tong
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Donghong He
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Srikanth Pendyala
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Berdyshev Evgeny
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | - Jerold Chun
- Department of Molecular Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Anne I Sperling
- Department of Medicine, The University of Chicago, Chicago, Illinois, USA
| | | |
Collapse
|