1
|
Laganà A, Maglione R, Costa A, Mandelli B, Bisegna ML, Milani ML, Filipponi V, Nardacci MG, Soriano T, Santacroce E, Petrucci L, Giordano C, Martelli M, De Propris MS. Integration of CD200, CD43 and ROR1 in Multiparameter Flow Cytometry (MFC) Routine Panels for the Differential Diagnosis of B-cell lymphoproliferative Disorders (B-LPDs). Mediterr J Hematol Infect Dis 2025; 17:e2025002. [PMID: 39830795 PMCID: PMC11740909 DOI: 10.4084/mjhid.2025.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Background Clonal mature B-cell lymphoproliferative disorders (B-LPDs) are a heterogeneous group of neoplasia characterized by the proliferation of mature B lymphocytes in the peripheral blood, bone marrow and/or lymphoid tissues. B-LPDs classification into different subtypes and their diagnosis is based on a multiparametric approach. However, accurate diagnosis may be challenging, especially in cases of ambiguous interpretation. Multiparameter flow cytometry (MFC) represents an extensively used technique to detect the presence of different cellular lines in immunology and hematology. MFC results provide an essential contribution to the B-LPDs diagnostic process, even more so considering that panels are constantly integrating novel markers to improve diagnostic accuracy. Objectives The aim was to evaluate the contributing role of MFC routinary studies by analyzing the expression and the mean fluorescence intensity (MFI) of CD200, ROR1, and CD43 in various B-LPDs to evaluate their usefulness in the differential diagnosis of these diseases. Methods We retrospectively evaluated 2615 consecutive cases of newly collected samples (mostly from patients with lymphocytosis) analyzed by MFC carried out in the B-LPD diagnostic process referred to the Division of Hematology of the Sapienza University of Rome. We compared the results of CD200, ROR1, and CD43 expression percentage and their MFI between different subtypes of B-LPDs. Results In chronic lymphocytic leukemia (CLL), CD200, ROR1, and CD43 were always expressed with bright intensity. CLL samples presented high CD200 expression and MFI [CD200%, mean: 100 (range, 24-100); positivity rate: 100%; MFI, median = 125 (range, 10-1200)] statistically higher than mantle cell lymphoma (MCL) (p<0.001), which is usually negative for CD200, and variant hairy cell leukemia (vHCL, according to 2022 ICC) (p<0.001), but comparable with classic HCL (cHCL) (p>0.9). ROR1 resulted expressed in all CLL [ROR1%, mean: 100 (range, 52-100), positivity rate: 100%; MFI, median=50 (range, 10-202)] and MCL cases with comparable MFI (p>0.9). CD43 expression and MFI were significantly higher in CLL [CD43%, mean 99 (range, 59-100); positivity rate: 100%; MFI, median = 130 (range, 41-980)] than in MCL, vHCL, cHCL, and all the others mature B-cell neoplasia (p<0.001). CD200 and CD43 expression and MFI were significantly higher in cHCL compared to vHCL. Among the other mature B-cell neoplasia, CD200 was variably expressed in follicular lymphoma (FL), marginal zone lymphoma (MZL), diffuse large B-cell lymphoma (DLBCL), and lymphoplasmacytic lymphoma (LPL). ROR1 and CD43 presented a very low expression percentage in this latter group, being mostly negative. Persistent polyclonal B-cell lymphocytosis (PPBL) resulted in uniformly positive for CD200 and negative for ROR1 and CD43. Conclusions Our data suggest that evaluating CD200, ROR1, and CD43 antigens and their intensity of expression, along with commonly used markers in MFC routine panels for B-LPDs, might be extremely useful for prompt diagnostic evaluation in the differential diagnosis of these diseases.
Collapse
Affiliation(s)
- Alessandro Laganà
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Raffaele Maglione
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Alessandro Costa
- Hematology Unit, Businco Hospital, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Biancamaria Mandelli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Maria Laura Bisegna
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Maria Laura Milani
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Valeria Filipponi
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Maria Grazia Nardacci
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Tania Soriano
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Eugenio Santacroce
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Luigi Petrucci
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | - Carla Giordano
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University, Policlinico Umberto I, Rome, Italy
| | - Maurizio Martelli
- Hematology, Department of Translational and Precision Medicine, Sapienza University of Rome, Italy
| | | |
Collapse
|
2
|
Fraune C, Tazelaar HD, Butt YM, Smith ML, Larsen BT, Kelemen K. Usefulness and Limitations of Current Diagnostic Strategies for Pulmonary Mucosa-Associated Lymphoid Tissue Lymphoma: Lessons Learned From a Large Cohort. Arch Pathol Lab Med 2024; 148:419-429. [PMID: 37594899 DOI: 10.5858/arpa.2022-0521-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/27/2023] [Indexed: 08/20/2023]
Abstract
CONTEXT.— The pathologic diagnosis of pulmonary extranodal marginal zone lymphoma of mucosa-associated lymphoid tissue (MALT) is challenging. OBJECTIVE.— To evaluate the diagnostic usefulness and limitations of current diagnostic strategies for pulmonary MALT lymphoma. DESIGN.— A retrospective review of 120 cases of pulmonary MALT lymphoma from 2014 through 2021 was performed. RESULTS.— Clinicoradiologic presentations overlapped with previous observations in patients with MALT lymphoma, such as a wide age range, female predominance, frequent association with autoimmune disease or immunodeficiency, and broad imaging findings. The histopathologic diagnosis was based on a combination of morphology, immunohistochemistry, and demonstration of B-cell lineage clonality. Two-thirds (76 of 113) of MALT lymphomas had lymphoplasmacytoid cytomorphology. Occasionally, MALT lymphomas were associated with granulomas/giant cells (29%, 35 of 120) or immunoglobulin deposition disease (21%, 25 of 120), including light chain/heavy chain deposition disease, amyloidosis, and/or crystal storing histiocytosis. While CD5, CD10, Bcl-2, and Bcl-6 rarely revealed aberrancies, aberrant CD43 expression either on B-cells or on plasma cells was detected in 42% (27 of 64) of cases, including cases for which proof of clonality could not be obtained. κ/λ in situ hybridization was particularly useful for tumors with lymphoplasmacytoid morphology but performed poorly in lymphomas having no plasmacytic differentiation. κ/λ immunohistochemistry showed no additional usefulness when applied together with κ/λ in situ hybridization. Immunoglobulin gene rearrangement studies by polymerase chain reaction achieved high detection rates of clonality in all cytomorphologic subgroups. CONCLUSIONS.— Our study offers a practical evaluation of common diagnostic tests in pulmonary MALT lymphoma. We offer recommendations for a diagnostic workup that takes into consideration the usefulness and the specific limitations of the various diagnostic strategies.
Collapse
Affiliation(s)
- Christoph Fraune
- From the Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (Fraune)
| | - Henry D Tazelaar
- the Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona (Tazelaar, Butt, Smith, Larsen, Kelemen)
| | - Yasmeen M Butt
- the Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona (Tazelaar, Butt, Smith, Larsen, Kelemen)
| | - Maxwell L Smith
- the Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona (Tazelaar, Butt, Smith, Larsen, Kelemen)
| | - Brandon T Larsen
- the Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona (Tazelaar, Butt, Smith, Larsen, Kelemen)
| | - Katalin Kelemen
- the Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, Arizona (Tazelaar, Butt, Smith, Larsen, Kelemen)
| |
Collapse
|
3
|
D’Arena G, Vitale C, Pietrantuono G, Villani O, Mansueto G, D’Auria F, Statuto T, D’Agostino S, Sabetta R, Tarasco A, Innocenti I, Autore F, Fresa A, Valvano L, Tomasso A, Cafaro L, Lamorte D, Laurenti L. What Does Atypical Chronic Lymphocytic Leukemia Really Mean? A Retrospective Morphological and Immunophenotypic Study. Cancers (Basel) 2024; 16:469. [PMID: 38275909 PMCID: PMC10814247 DOI: 10.3390/cancers16020469] [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: 12/13/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Atypical chronic lymphocytic leukemia (CLL) is still defined according to morphological criteria. However, deviance from the typical surface immunological profile suggests an atypical immunological-based CLL. A large cohort of patients with CLL was retrospectively evaluated aiming at assessing morphological (FAB criteria), immunophenotypical (two or more discordances from the typical profile), and clinical-biological features of atypical CLL. Compared to typical cases, morphologically atypical CLL showed a greater percentage of unmutated IgVH and CD38 positivity, and a higher expression of CD20. Immunophenotypically atypical CLL was characterized by more advanced clinical stages, higher expression of CD20, higher rate of FMC7, CD79b and CD49d positivity, and by an intermediate-high expression of membrane surface immunoglobulin, compared to typical cases. When patients were categorized based on immunophenotypic and morphologic concordance or discordance, no difference emerged. Finally, morphological features better discriminated patients' prognosis in terms of time-to-first treatment, while concordant atypical cases showed overall a worse prognosis. Discordant cases by immunophenotype and/or morphology did not identify specific prognostic groups. Whether-in the era of molecular markers used as prognostic indicators-it does make sense to focus on morphology and immunophenotype features in CLL is still matter of debate needing further research.
Collapse
Affiliation(s)
- Giovanni D’Arena
- Immuno-Hematology and Transfusion Medicine Unit, “San Luca” Hospital, 84078 Vallo della Lucania, Italy; (R.S.); (A.T.)
| | - Candida Vitale
- A.O.U. Città della Salute e della Scienza di Torino and Department of Molecular Biotechnology and Health Sciences, Division of Hematology, University of Torino, 10125 Torino, Italy;
| | - Giuseppe Pietrantuono
- Hematology and Stem Cell Transplantation Unit, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (G.P.); (O.V.); (G.M.); (S.D.)
| | - Oreste Villani
- Hematology and Stem Cell Transplantation Unit, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (G.P.); (O.V.); (G.M.); (S.D.)
| | - Giovanna Mansueto
- Hematology and Stem Cell Transplantation Unit, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (G.P.); (O.V.); (G.M.); (S.D.)
| | - Fiorella D’Auria
- Laboratory of Clinical Pathology, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy;
| | - Teodora Statuto
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (T.S.); (L.V.)
| | - Simona D’Agostino
- Hematology and Stem Cell Transplantation Unit, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (G.P.); (O.V.); (G.M.); (S.D.)
| | - Rosalaura Sabetta
- Immuno-Hematology and Transfusion Medicine Unit, “San Luca” Hospital, 84078 Vallo della Lucania, Italy; (R.S.); (A.T.)
| | - Angela Tarasco
- Immuno-Hematology and Transfusion Medicine Unit, “San Luca” Hospital, 84078 Vallo della Lucania, Italy; (R.S.); (A.T.)
| | - Idanna Innocenti
- Hematology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (I.I.); (F.A.); (A.F.); (A.T.); (L.L.)
| | - Francesco Autore
- Hematology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (I.I.); (F.A.); (A.F.); (A.T.); (L.L.)
| | - Alberto Fresa
- Hematology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (I.I.); (F.A.); (A.F.); (A.T.); (L.L.)
| | - Luciana Valvano
- Laboratory of Clinical Research and Advanced Diagnostics, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, Italy; (T.S.); (L.V.)
| | - Annamaria Tomasso
- Hematology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (I.I.); (F.A.); (A.F.); (A.T.); (L.L.)
| | - Lorenzo Cafaro
- Immuno-Hematology and Transfusion Medicine Unit, “Immacolata” Hospital, 84073 Sapri, Italy;
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Vulture, Italy;
| | - Luca Laurenti
- Hematology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (I.I.); (F.A.); (A.F.); (A.T.); (L.L.)
| |
Collapse
|
4
|
Abstract
Both the cascade whereby a blood-borne cell enters a tissue and the anchoring of hematopoietic stem/progenitor cells (HSPCs) within bone marrow critically pivots on cell-cell interactions mediated by E-selectin binding to its canonical carbohydrate ligand, the tetrasaccharide termed "sialylated Lewis X" (sLeX). E-selectin, a member of the selectin class of adhesion molecules that is exclusively expressed by vascular endothelium, engages sLeX-bearing glycoconjugates that adorn mature leukocytes and HSPCs, as well as malignant cells, thereby permitting these cells to extravasate into various tissues. E-selectin expression is induced on microvascular endothelial cells within inflammatory loci at all tissues. However, conspicuously, E-selectin is constitutively expressed within microvessels in skin and marrow and, additionally, is inducibly expressed at these sites. Within the marrow, E-selectin receptor/ligand interactions promote lodgment of HSPCs and their malignant counterparts within hematopoietic growth-promoting microenvironments, collectively known as "vascular niches". Indeed, E-selectin receptor/ligand interactions have been reported to regulate both hematopoietic stem, and leukemic, cell proliferative dynamics. As such, signaling induced via engagement of E-selectin ligands is gaining interest as a critical mediator of homeostatic and malignant hematopoiesis, and this review will present current perspectives on the glycoconjugates mediating E-selectin receptor/ligand interactions and their currently defined role(s) in leukemogenesis.
Collapse
Affiliation(s)
- Evan Ales
- Department of Translational Medicine & The Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States
| | - Robert Sackstein
- Department of Translational Medicine & The Translational Glycobiology Institute, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, United States.
| |
Collapse
|
5
|
Pan X, Chen R, Zhang Y, Zhu Y, Zhao J, Yao H, Ma J. Porcine extraintestinal pathogenic Escherichia coli delivers two serine protease autotransporters coordinately optimizing the bloodstream infection. Front Cell Infect Microbiol 2023; 13:1138801. [PMID: 36875517 PMCID: PMC9978103 DOI: 10.3389/fcimb.2023.1138801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is one of the leading causes of bloodstream infections in a broad spectrum of birds and mammals, thus poses a great threat to public health, while its underlying mechanism causing sepsis is not fully understood. Here we reported a high virulent ExPEC strain PU-1, which has a robust ability to colonize within host bloodstream, while induced a low level of leukocytic activation. Two serine protease autotransporters of Enterobacteriaceae (SPATEs), VatPU-1 and TshPU-1, were found to play critical roles for the urgent blood infection of strain PU-1. Although the Vat and Tsh homologues have been identified as virulence factors of ExPEC, their contributions to bloodstream infection are still unclear. In this study, VatPU-1 and TshPU-1 were verified to interact with the hemoglobin (a well-known mucin-like glycoprotein in red blood cell), degrade the mucins of host respiratory tract, and cleave the CD43 (a major cell surface component sharing similar O-glycosylated modifications with other glycoprotein expressed on leukocytes), suggesting that these two SPATEs have the common activity to cleave a broad array of mucin-like O-glycoproteins. These cleavages significantly impaired the chemotaxis and transmigration of leukocytes, and then inhibited the activation of diverse immune responses coordinately, especially downregulated the leukocytic and inflammatory activation during bloodstream infection, thus might mediate the evasion of ExPEC from immune clearance of blood leukocytes. Taken together, these two SPATEs play critical roles to cause a heavy bacterial load within bloodstream via immunomodulation of leukocytes, which provides a more comprehensive understanding how ExPEC colonize within host bloodstream and cause severe sepsis.
Collapse
Affiliation(s)
- Xinming Pan
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- Office International Des (OIE) Reference Lab for Swine Streptococcosis, Nanjing Agricultural University, Nanjing, China
| | - Rong Chen
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- Office International Des (OIE) Reference Lab for Swine Streptococcosis, Nanjing Agricultural University, Nanjing, China
| | - Yating Zhang
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- Office International Des (OIE) Reference Lab for Swine Streptococcosis, Nanjing Agricultural University, Nanjing, China
| | - Yinchu Zhu
- Institute of Animal Husbandry and Veterinary Sciences, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Jin Zhao
- Department of Animal Science, Yuxi Agriculture Vocation-Technical College, Yuxi, China
| | - Huochun Yao
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- Office International Des (OIE) Reference Lab for Swine Streptococcosis, Nanjing Agricultural University, Nanjing, China
| | - Jiale Ma
- Ministry of Education (MOE) Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Key Lab of Animal Bacteriology, Ministry of Agriculture, Nanjing, China
- Office International Des (OIE) Reference Lab for Swine Streptococcosis, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Jiale Ma,
| |
Collapse
|
6
|
Ait-Belkacem I, Cartagena García C, Millet-Wallisky E, Izquierdo N, Loosveld M, Arnoux I, Morange PE, Galland F, Lambert N, Malergue F, Busnel JM. SARS-CoV-2 spike protein induces a differential monocyte activation that may contribute to age bias in COVID-19 severity. Sci Rep 2022; 12:20824. [PMID: 36460710 PMCID: PMC9716544 DOI: 10.1038/s41598-022-25259-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
A strong bias related to age is observed in COVID-19 patients with pediatric subjects developing a milder disease than adults. We hypothesized that a specific SARS-CoV-2 effect conjugated with preexisting differences in the immune systems may explain this. Using flow cytometry, we investigated basal immune differences in a cohort consisting of 16 non-infected young and 16 aged individuals and further leveraged an in vitro whole blood model of SARS-CoV-2 infection so that functional differences could be mined as well. In short, blood diluted in culture media was incubated 5 or 24 h with the trimeric spike protein or controls. Following unsupervised analysis, we first confirmed that the immune lymphoid and myeloid systems in adults are less efficient and prone to develop higher inflammation than those in children. We notably identified in adults a higher CD43 lymphocyte expression, known for its potentially inhibitory role. The spike protein induced different responses between adults and children, notably a higher increase of inflammatory markers together with lower monocyte and B cell activation in adults. Interestingly, CD169, a CD43 ligand overexpressed in COVID-19 patients, was confirmed to be strongly modulated by the spike protein. In conclusion, the spike protein exacerbated the preexisting lower immune responsiveness and higher inflammatory potential in adults. Altogether, some of the markers identified may explain the marked age bias and be predictive of severity.
Collapse
Affiliation(s)
- Ines Ait-Belkacem
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France
- Aix Marseille Université CNRS INSERM CIML Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Celia Cartagena García
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France
- INSERM UMRs 1097, Aix Marseille University, Marseille, France
| | - Ewa Millet-Wallisky
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France
| | - Nicolas Izquierdo
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France
| | - Marie Loosveld
- Hematology Laboratory, Timone University Hospital, APHM, Marseille, France
| | - Isabelle Arnoux
- Hematology Laboratory, Timone University Hospital, APHM, Marseille, France
| | | | - Franck Galland
- Aix Marseille Université CNRS INSERM CIML Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | | | - Fabrice Malergue
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France.
| | - Jean-Marc Busnel
- Department of Research and Development, Beckman Coulter Life Sciences-Immunotech, Marseille, France.
| |
Collapse
|
7
|
Hyun SW, Feng C, Liu A, Lillehoj EP, Trotta R, Kingsbury TJ, Passaniti A, Lugkey KN, Chauhan S, Cipollo JF, Luzina IG, Atamas SP, Cross AS, Goldblum SE. Altered sialidase expression in human myeloid cells undergoing apoptosis and differentiation. Sci Rep 2022; 12:14173. [PMID: 35986080 PMCID: PMC9390117 DOI: 10.1038/s41598-022-18448-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 08/11/2022] [Indexed: 11/10/2022] Open
Abstract
To gain insight into sialic acid biology and sialidase/neuraminidase (NEU) expression in mature human neutrophil (PMN)s, we studied NEU activity and expression in PMNs and the HL60 promyelocytic leukemic cell line, and changes that might occur in PMNs undergoing apoptosis and HL60 cells during their differentiation into PMN-like cells. Mature human PMNs contained NEU activity and expressed NEU2, but not NEU1, the NEU1 chaperone, protective protein/cathepsin A(PPCA), NEU3, and NEU4 proteins. In proapoptotic PMNs, NEU2 protein expression increased > 30.0-fold. Granulocyte colony-stimulating factor protected against NEU2 protein upregulation, PMN surface desialylation and apoptosis. In response to 3 distinct differentiating agents, dimethylformamide, dimethylsulfoxide, and retinoic acid, total NEU activity in differentiated HL60 (dHL60) cells was dramatically reduced compared to that of nondifferentiated cells. With differentiation, NEU1 protein levels decreased > 85%, PPCA and NEU2 proteins increased > 12.0-fold, and 3.0-fold, respectively, NEU3 remained unchanged, and NEU4 increased 1.7-fold by day 3, and then returned to baseline. In dHL60 cells, lectin blotting revealed decreased α2,3-linked and increased α2,6-linked sialylation. dHL60 cells displayed increased adhesion to and migration across human bone marrow-derived endothelium and increased bacterial phagocytosis. Therefore, myeloid apoptosis and differentiation provoke changes in NEU catalytic activity and protein expression, surface sialylation, and functional responsiveness.
Collapse
|
8
|
Ning X, Wei X, Chen B, Li Z, Zheng Z, Yi Z, Wei Q, Guo X, Kang Q, Feng R, Wei Y. CD43 is an adverse prognostic factor in newly diagnosed multiple myeloma. Leuk Lymphoma 2022; 63:2573-2578. [PMID: 35819872 DOI: 10.1080/10428194.2022.2092854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Currently, the expression pattern and prognostic value of CD43 expression in multiple myeloma (MM) remain unknown. 109 newly diagnosed MM patients were recruited and CD43 expression was determined by multiparameter flow cytometry, of which 77 (70.6%) were CD43 positive. Patients with positive CD43 expression were more likely to present with, hemoglobin < 85 g/L (p = 0.008), International Staging System (ISS) stage III (p = 0.044), 13q14 deletion (p = 0.034) and more monoclonal plasma cells (p = 0.003). Patients with CD43 positive had significantly poor treatment response (p = 0.021), progression-free survival (PFS) (p = 0.012), and overall survival (OS) (p = 0.023) than those without CD43. The poorer prognosis of CD43-positive patients was retained in multivariate analysis (p = 0.005 for PFS; p = 0.013 for OS). Our study indicated that CD43 was an independent adverse prognostic factor in multiple myeloma.
Collapse
Affiliation(s)
- Xueqin Ning
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaolei Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingyuan Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhen Li
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhongxin Zheng
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhengshan Yi
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xutao Guo
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiaoxi Kang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ru Feng
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongqiang Wei
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Tang J, Gong Y, Ma X. Bispecific Antibodies Progression in Malignant Melanoma. Front Pharmacol 2022; 13:837889. [PMID: 35401191 PMCID: PMC8984188 DOI: 10.3389/fphar.2022.837889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
The discovery of oncogenes and immune checkpoints has revolutionized the treatment of melanoma in the past 10 years. However, the current PD-L1 checkpoints lack specificity for tumors and target normal cells expressing PD-L1, thus reducing the efficacy on malignant melanoma and increasing the side effects. In addition, the treatment options for primary or secondary drug-resistant melanoma are limited. Bispecific antibodies bind tumor cells and immune cells by simultaneously targeting two antigens, enhancing the anti-tumor targeting effect and cytotoxicity and reducing drug-resistance in malignant melanoma, thus representing an emerging strategy to improve the clinical efficacy. This review focused on the treatment of malignant melanoma by bispecific antibodies and summarized the effective results of the experiments that have been conducted, also discussing the different aspects of these therapies. The role of the melanoma epitopes, immune cell activation, cell death and cytotoxicity induced by bispecific antibodies were evaluated in the clinical or preclinical stage, as these therapies appear to be the most suitable in the treatment of malignant melanoma.
Collapse
Affiliation(s)
- Juan Tang
- Department of Oncology, West China Hospital of Sichuan University, Chengdu, China
| | - Youling Gong
- Department of Oncology, West China Hospital of Sichuan University, Chengdu, China
| | - Xuelei Ma
- Department of Oncology, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
10
|
van Houtum EJH, Büll C, Cornelissen LAM, Adema GJ. Siglec Signaling in the Tumor Microenvironment. Front Immunol 2021; 12:790317. [PMID: 34966391 PMCID: PMC8710542 DOI: 10.3389/fimmu.2021.790317] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022] Open
Abstract
Sialic acid-binding immunoglobulin-like lectins (Siglecs) are a family of receptors that recognize sialoglycans - sialic acid containing glycans that are abundantly present on cell membranes. Siglecs are expressed on most immune cells and can modulate their activity and function. The majority of Siglecs contains immune inhibitory motifs comparable to the immune checkpoint receptor PD-1. In the tumor microenvironment (TME), signaling through the Siglec-sialoglycan axis appears to be enhanced through multiple mechanisms favoring tumor immune evasion similar to the PD-1/PD-L1 signaling pathway. Siglec expression on tumor-infiltrating immune cells appears increased in the immune suppressive microenvironment. At the same time, enhanced Siglec ligand expression has been reported for several tumor types as a result of aberrant glycosylation, glycan modifications, and the increased expression of sialoglycans on proteins and lipids. Siglec signaling has been identified as important regulator of anti-tumor immunity in the TME, but the key factors contributing to Siglec activation by tumor-associated sialoglycans are diverse and poorly defined. Among others, Siglec activation and signaling are co-determined by their expression levels, cell surface distribution, and their binding preferences for cis- and trans-ligands in the TME. Siglec binding preference are co-determined by the nature of the proteins/lipids to which the sialoglycans are attached and the multivalency of the interaction. Here, we review the current understanding and emerging conditions and factors involved in Siglec signaling in the TME and identify current knowledge gaps that exist in the field.
Collapse
Affiliation(s)
- Eline J. H. van Houtum
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Christian Büll
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW), Utrecht, Netherlands
- Copenhagen Center for Glycomics, Departments of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lenneke A. M. Cornelissen
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gosse J. Adema
- Radiotherapy & OncoImmunology Laboratory, Department of Radiation Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
11
|
Genome-wide CRISPR screens reveal a specific ligand for the glycan-binding immune checkpoint receptor Siglec-7. Proc Natl Acad Sci U S A 2021; 118:2015024118. [PMID: 33495350 DOI: 10.1073/pnas.2015024118] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Glyco-immune checkpoint receptors, molecules that inhibit immune cell activity following binding to glycosylated cell-surface antigens, are emerging as attractive targets for cancer immunotherapy. Defining biologically relevant ligands that bind and activate such receptors, however, has historically been a significant challenge. Here, we present a CRISPRi genomic screening strategy that allowed unbiased identification of the key genes required for cell-surface presentation of glycan ligands on leukemia cells that bind the glyco-immune checkpoint receptors Siglec-7 and Siglec-9. This approach revealed a selective interaction between Siglec-7 and the mucin-type glycoprotein CD43. Further work identified a specific N-terminal glycopeptide region of CD43 containing clusters of disialylated O-glycan tetrasaccharides that form specific Siglec-7 binding motifs. Knockout or blockade of CD43 in leukemia cells relieves Siglec-7-mediated inhibition of immune killing activity. This work identifies a potential target for immune checkpoint blockade therapy and represents a generalizable approach to dissection of glycan-receptor interactions in living cells.
Collapse
|
12
|
Soday L, Potts M, Hunter LM, Ravenhill BJ, Houghton JW, Williamson JC, Antrobus R, Wills MR, Matheson NJ, Weekes MP. Comparative Cell Surface Proteomic Analysis of the Primary Human T Cell and Monocyte Responses to Type I Interferon. Front Immunol 2021; 12:600056. [PMID: 33628210 PMCID: PMC7897682 DOI: 10.3389/fimmu.2021.600056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 01/13/2021] [Indexed: 12/28/2022] Open
Abstract
The cellular response to interferon (IFN) is essential for antiviral immunity, IFN-based therapy and IFN-related disease. The plasma membrane (PM) provides a critical interface between the cell and its environment, and is the initial portal of entry for viruses. Nonetheless, the effect of IFN on PM proteins is surprisingly poorly understood, and has not been systematically investigated in primary immune cells. Here, we use multiplexed proteomics to quantify IFNα2a-stimulated PM protein changes in primary human CD14+ monocytes and CD4+ T cells from five donors, quantifying 606 and 482 PM proteins respectively. Comparison of cell surface proteomes revealed a remarkable invariance between donors in the overall composition of the cell surface from each cell type, but a marked donor-to-donor variability in the effects of IFNα2a. Furthermore, whereas only 2.7% of quantified proteins were consistently upregulated by IFNα2a at the surface of CD4+ T cells, 6.8% of proteins were consistently upregulated in primary monocytes, suggesting that the magnitude of the IFNα2a response varies according to cell type. Among these differentially regulated proteins, we found the viral target Endothelin-converting enzyme 1 (ECE1) to be an IFNα2a-stimulated protein exclusively upregulated at the surface of CD4+ T cells. We therefore provide a comprehensive map of the cell surface of IFNα2a-stimulated primary human immune cells, including previously uncharacterized interferon stimulated genes (ISGs) and candidate antiviral factors.
Collapse
Affiliation(s)
- Lior Soday
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Martin Potts
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Leah M. Hunter
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Benjamin J. Ravenhill
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Jack W. Houghton
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - James C. Williamson
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| | - Mark R. Wills
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Nicholas J. Matheson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), University of Cambridge, Cambridge, United Kingdom
- NHS Blood and Transplant, Cambridge, United Kingdom
| | - Michael P. Weekes
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
13
|
Abstract
Objectives: CD43 can be useful in routine flow cytometry. We conducted a systematic review aiming to describe when CD43 is used by flow cytometry in malignant hematology and to determine its value in these settings. Methods: Systematic review of MEDLINE (search 'CD43' AND 'flow cytometry,' starting in 2010). Results: Twenty-one of 103 entries retrieved were included in this systematic review. CD43 is used in three settings: 1) in the classification of mature B cell lymphoproliferative disorders, 2) as part of a strategy to quantify residual disease in chronic lymphocytic leukemia (CLL) and 3) to help classify CD10-positive B cell populations. In this section, the published data is summarized, the clinical usefulness in each of these settings is evaluated and illustrative cases are shown. Conclusion: CD43 has a growing role in the diagnosis and management of B cell malignancies; it has become essential for the classification of B cell lymphoproliferative disorders and may be of help in the differential diagnosis of CD10-positive lymphomas by FC. It is also required for optimal quantification of CLL residual disease, which will soon be used to guide therapeutic decisions.
Collapse
Affiliation(s)
- Marc Sorigue
- Hematology Laboratory, ICO-Hospital Germans Trias I Pujol, Functional Cytomics- IJC, Universitat Autònoma De Barcelona , Badalona, Spain
| |
Collapse
|
14
|
de Jong G, Bartels L, Kedde M, Verdegaal EME, Gillissen MA, Levie SE, Cercel MG, van Hal-van Veen SE, Fatmawati C, van de Berg D, Yasuda E, Claassen YB, Bakker AQ, van der Burg SH, Schotte R, Villaudy J, Spits H, Hazenberg MD, van Helden PM, Wagner K. Melanoma cells can be eliminated by sialylated CD43 × CD3 bispecific T cell engager formats in vitro and in vivo. Cancer Immunol Immunother 2020; 70:1569-1581. [PMID: 33225419 DOI: 10.1007/s00262-020-02780-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 10/22/2020] [Indexed: 01/13/2023]
Abstract
Targeted cancer therapy with monoclonal antibodies has proven successful for different cancer types but is limited by the availability of suitable antibody targets. CD43s, a unique sialylated form of CD43 expressed by hematologic malignancies, is a recently identified target and antibodies interacting with CD43s may have therapeutic potential against acute myeloid leukemia (AML) and myelodysplastic syndrome. CD43s is recognized by the human antibody AT1413, that was derived from a high-risk AML patient who successfully cleared leukemia after allogeneic stem cell transplantation. Here we observed that AT1413 binds also to certain non-hematopoietic tumor cells, particularly melanoma and breast cancer. AT1413 immune precipitated CD43s from melanoma cells confirming that it recognizes the same target on melanoma as on AML. AT1413 induced antibody-dependent cellular cytotoxicity against short-term cultured patient-derived melanoma samples. However, AT1413 was unable to affect the growth of melanoma cells in vivo. To increase the efficacy of AT1413 as a therapeutic antibody, we generated two different formats of bispecific T-cell engaging antibodies (TCEs): one binding bivalently (bTCE) and the other monovalently (knob-in-hole; KiH) to both CD43s and CD3ε. In vitro, these TCEs redirected T-cell cytotoxicity against melanoma cells with differences in potencies. To investigate their effects in vivo, we grafted mice that harbor a human immune system with the melanoma cell line A375. Treatment with both AT1413 bTCE and AT1413 KiH significantly reduced tumor outgrowth in these mice. These data indicate a broad therapeutic potential of AT1413 that includes AML and CD43s-expressing solid tumors that originate from CD43-negative tissues.
Collapse
Affiliation(s)
- G de Jong
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Department of Hematology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands
| | - L Bartels
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands
| | - M Kedde
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - E M E Verdegaal
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - M A Gillissen
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Department of Hematology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands
| | - S E Levie
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - M G Cercel
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | | | - C Fatmawati
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - D van de Berg
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - E Yasuda
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - Y B Claassen
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - A Q Bakker
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - S H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - R Schotte
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - J Villaudy
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| | - H Spits
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands.,Department of Experimental Immunology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands
| | - M D Hazenberg
- Department of Hematology, Amsterdam UMC, Location AMC, Amsterdam, The Netherlands.,Cancer Center Amsterdam (CCA), Amsterdam, The Netherlands.,Amsterdam Infection and Immunity Institute (AI&II), Amsterdam, The Netherlands.,Department of Hematopoiesis, Sanquin Research, Amsterdam, The Netherlands
| | - P M van Helden
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands.
| | - K Wagner
- AIMM Therapeutics, Meibergdreef 59, 1105 BA, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Bacterial sialoglycosidases in Virulence and Pathogenesis. Pathogens 2019; 8:pathogens8010039. [PMID: 30909660 PMCID: PMC6471121 DOI: 10.3390/pathogens8010039] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/18/2019] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
Human oral microbiome and dysbiotic infections have been recently evidently identified. One of the major reasons for such dysbiosis is impairment of the immune system. Periodontitis is a chronic inflammatory disease affecting the tissues that surround and support the teeth. In the United States., approximately 65 million people are affected by this condition. Its occurrence is also associated with many important systemic diseases such as cardiovascular disease, rheumatoid arthritis, and Alzheimer’s disease. Among the most important etiologies of periodontitis is Porphyromonas gingivalis, a keystone bacterial pathogen. Keystone pathogens can orchestrate inflammatory disease by remodeling a normally benign microbiota causing imbalance between normal and pathogenic microbiota (dysbiosis). The important characteristics of P. gingivalis causing dysbiosis are its virulence factors which cause effective subversion of host defenses to its advantage allowing other pathogens to grow. Some of the mechanisms involved in these processes are still not well-understood. However, various microbial strategies target host sialoglycoproteins for immune dysregulation. In addition, the enzymes that break down sialoglycoproteins and sialoglycans are the “sialoglycoproteases”, resulting in exposed terminal sialic acid. This process could lead to pathogen-toll like receptor (TLR) interactions mediated through sialic acid receptor ligand mechanisms. Assessing the function of P. gingivalis sialoglycoproteases, could pave the way to designing carbohydrate analogues and sialic acid mimetics to serve as drug targets.
Collapse
|
16
|
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: 13] [Impact Index Per Article: 2.2] [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
|
17
|
Kalergis AM, Anegon I, González PA. FOCIS goes south: advances in translational and clinical immunology. Immunotherapy 2017; 9:789-792. [PMID: 28877630 DOI: 10.2217/imt-2017-0079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
FOCIS goes South: Advances in Translational and Clinical Immunology was the first Federation of Clinical Immunology Societies (FOCIS) ( www.focisnet.org ) meeting held in Latin America (May 15-17, 2017, Santiago de Chile, Chile). The meeting was organized as a 3-day workshop and was fostered by the Millennium Institute on Immunology and Immunotherapy, a recently nominated FOCIS Center of Excellence. The workshop brought together FOCIS associates, such as members of the FOCIS Board of Directors, Directors of different Centers of Excellence, regional speakers and 350 attendees. The Meeting covered aspects of immune regulation and modulation, as well as immunotherapy in areas of autoimmunity, transplantation, cancer and infectious diseases, among others. The activity also had a full-day immunology course and a day-long flow cytometry course.
Collapse
Affiliation(s)
- Alexis M Kalergis
- Departamento de GenéticaMolecular y Microbiología, Facultad de Ciencias Biológicas, PontificiaUniversidad Católica de Chilee.,Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago-Chile. Av. Portugal 49, Santiago E-8330025, Chile.,Departamento de Endocrinología, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago-Chile.,Centre de Recherche en Transplantation et ImmunologieUMR1064, INSERM, Université de Nantes, Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et ImmunologieUMR1064, INSERM, Université de Nantes, Nantes, France.,Institut deTransplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Pablo A González
- Departamento de GenéticaMolecular y Microbiología, Facultad de Ciencias Biológicas, PontificiaUniversidad Católica de Chilee.,Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago-Chile. Av. Portugal 49, Santiago E-8330025, Chile
| |
Collapse
|
18
|
Batdorf BH, Kroft SH, Hosking PR, Harrington AM, Mackinnon AC, Olteanu H. Evaluation of CD43 expression in non-hematopoietic malignancies. Ann Diagn Pathol 2017; 29:23-27. [PMID: 28807337 DOI: 10.1016/j.anndiagpath.2017.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 04/25/2017] [Indexed: 12/30/2022]
Abstract
OBJECTIVES CD43 is normally expressed only on the surface of leukocytes, and is considered a sensitive and specific marker for hematologic malignancies. As such, it may have diagnostic utility in confirming hematolymphoid lineage in cases that are negative for CD45. Aberrant CD43 expression has been described in non-hematopoietic tumors, although literature data on this topic is variable and sometimes contradictory. To clarify and expand on existing literature findings, we evaluated CD43 expression by immunohistochemistry (IHC) in a large cohort (307) of non-hematopoietic neoplasms, including poorly differentiated malignancies. METHODS 17 tissue microarrays and sections from 19 individual cases were stained with CD43 (clone DF-T1) monoclonal antibody. The proportion of positive cells, stain localization (nuclear, cytoplasmic or membranous), and intensity (compared to internal leukocyte controls) were recorded in all cases. RESULTS There were 98/307 (32%) positive cases, that showed focal weak nuclear staining in 1-25% of cells, including 23/25 (92%) pancreatic ductal adenocarcinomas; 31/34 (91%) breast invasive ductal carcinomas; 13/15 (87%) papillary thyroid carcinomas; 3/4 (75%) follicular thyroid carcinomas; 6/15 (40%) renal cell carcinomas; 9/28 (32%) lung adenocarcinomas; 1/13 (8%) lung squamous cell carcinomas (SCCs); 2/8 (25%) prostate adenocarcinomas; 8/62 (13%) colon adenocarcinomas; and 2/21 (10%) neuroendocrine neoplasms. None of the positive cases demonstrated strong, membranous CD43 expression comparable to that seen in background mature lymphocytes or segmented neutrophils. Negative cases included 11 cervical SCCs, 12 cervical adenocarcinomas, 19 urothelial carcinomas, 10 lung small cell carcinomas, 11 sarcomas, and 19 poorly differentiated carcinomas from various tissue sites. CONCLUSIONS In our cohort, most non-hematopoietic neoplasms are negative for CD43 expression, with a subset showing focal, weak nuclear positivity. This data indicates that uniform and strong membranous staining appears to be specific to hematopoietic neoplasms.
Collapse
Affiliation(s)
- Bjorn H Batdorf
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Steven H Kroft
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Paul R Hosking
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | | | - Horatiu Olteanu
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, United States.
| |
Collapse
|
19
|
|
20
|
Weber C, Shantsila E, Hristov M, Caligiuri G, Guzik T, Heine GH, Hoefer IE, Monaco C, Peter K, Rainger E, Siegbahn A, Steffens S, Wojta J, Lip GYH. Role and analysis of monocyte subsets in cardiovascular disease. Joint consensus document of the European Society of Cardiology (ESC) Working Groups "Atherosclerosis & Vascular Biology" and "Thrombosis". Thromb Haemost 2016; 116:626-37. [PMID: 27412877 DOI: 10.1160/th16-02-0091] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/02/2016] [Indexed: 12/21/2022]
Abstract
Monocytes as cells of the innate immunity are prominently involved in the development of atherosclerotic lesions. The heterogeneity of blood monocytes has widely been acknowledged by accumulating experimental and clinical data suggesting a differential, subset-specific contribution of the corresponding subpopulations to the pathology of cardiovascular and other diseases. This document re-evaluates current nomenclature and summarises key findings on monocyte subset biology to propose a consensus statement about phenotype, separation and quantification of the individual subsets.
Collapse
Affiliation(s)
- Christian Weber
- Dr. Christian Weber, LMU Munich - Cardiovascular Prevention, Pettenkoferstr. 9, 80336 Munich, Germany, Tel.: +49 89 4400 54350, Fax: +49 89 4400 54352, E-mail:
| | | | - Michael Hristov
- Dr. Michael Hristov, LMU Munich - Cardiovascular Prevention, Pettenkoferstr. 9, 80336 Munich, Germany, Tel.: +49 89 4400 54350, Fax: +49 89 4400 54352, E-mail:
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Daum M, Broszeit F, Hoffmann-Röder A. Synthesis of a Fluorinated Sialophorin Hexasaccharide-Threonine Conjugate for Fmoc Solid-Phase Glycopeptide Synthesis. European J Org Chem 2016. [DOI: 10.1002/ejoc.201600523] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Affiliation(s)
- Markus Daum
- Center for Integrated Protein Science Munich (CIPS ) at the Department of Chemistry; Ludwig-Maximilians-Universität; Butenandtstr. 5-13 81377 Munich Germany
| | - Frederik Broszeit
- Center for Integrated Protein Science Munich (CIPS ) at the Department of Chemistry; Ludwig-Maximilians-Universität; Butenandtstr. 5-13 81377 Munich Germany
| | - Anja Hoffmann-Röder
- Center for Integrated Protein Science Munich (CIPS ) at the Department of Chemistry; Ludwig-Maximilians-Universität; Butenandtstr. 5-13 81377 Munich Germany
| |
Collapse
|
22
|
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
|
23
|
Meiler S, Baumer Y, McCurdy S, Lee BH, Kitamoto S, Boisvert WA. Cluster of differentiation 43 deficiency in leukocytes leads to reduced atherosclerosis--brief report. Arterioscler Thromb Vasc Biol 2014; 35:309-11. [PMID: 25524774 DOI: 10.1161/atvbaha.114.304513] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the role of cluster of differentiation 43 (CD43), an integral membrane glycoprotein with both proadhesive and antiadhesive activities, in atherosclerosis. APPROACH AND RESULTS Low-density lipoprotein receptor-deficient mice were lethally irradiated and reconstituted with either bone marrow from CD43(-/-) mice or from wild-type controls. We found that mice lacking the CD43 on their leukocytes had significantly less severe atherosclerosis and that, contrary to our expectation, macrophage infiltration into the vessel wall was not affected by the lack of CD43 in the leukocytes. However, we found that CD43 mediates cholesterol homeostasis in macrophages by facilitating cholesterol efflux. This resulted in a significant reduction in storage of cholesterol in the aorta of mice lacking CD43 in the leukocytes. CONCLUSIONS CD43 may be an important mediator of macrophage lipid homeostasis, thereby affecting macrophage foam cell formation and ultimately atherosclerotic plaque development.
Collapse
Affiliation(s)
- Svenja Meiler
- From the Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu (S.M., Y.B., S.M., W.A.B.); Department of Food and Nutrition, School of Food Science and Technology, Chung-Ang University, Seoul, Korea (B.-H.L.); Departments of Cardiovascular Medicine and Advanced Therapeutics for Cardiovascular Diseases, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (S.K.); and Kazan Federal University, Kazan, Russia (W.A.B.)
| | - Yvonne Baumer
- From the Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu (S.M., Y.B., S.M., W.A.B.); Department of Food and Nutrition, School of Food Science and Technology, Chung-Ang University, Seoul, Korea (B.-H.L.); Departments of Cardiovascular Medicine and Advanced Therapeutics for Cardiovascular Diseases, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (S.K.); and Kazan Federal University, Kazan, Russia (W.A.B.)
| | - Sara McCurdy
- From the Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu (S.M., Y.B., S.M., W.A.B.); Department of Food and Nutrition, School of Food Science and Technology, Chung-Ang University, Seoul, Korea (B.-H.L.); Departments of Cardiovascular Medicine and Advanced Therapeutics for Cardiovascular Diseases, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (S.K.); and Kazan Federal University, Kazan, Russia (W.A.B.)
| | - Bog-Hieu Lee
- From the Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu (S.M., Y.B., S.M., W.A.B.); Department of Food and Nutrition, School of Food Science and Technology, Chung-Ang University, Seoul, Korea (B.-H.L.); Departments of Cardiovascular Medicine and Advanced Therapeutics for Cardiovascular Diseases, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (S.K.); and Kazan Federal University, Kazan, Russia (W.A.B.)
| | - Shiro Kitamoto
- From the Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu (S.M., Y.B., S.M., W.A.B.); Department of Food and Nutrition, School of Food Science and Technology, Chung-Ang University, Seoul, Korea (B.-H.L.); Departments of Cardiovascular Medicine and Advanced Therapeutics for Cardiovascular Diseases, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (S.K.); and Kazan Federal University, Kazan, Russia (W.A.B.)
| | - William A Boisvert
- From the Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, Honolulu (S.M., Y.B., S.M., W.A.B.); Department of Food and Nutrition, School of Food Science and Technology, Chung-Ang University, Seoul, Korea (B.-H.L.); Departments of Cardiovascular Medicine and Advanced Therapeutics for Cardiovascular Diseases, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan (S.K.); and Kazan Federal University, Kazan, Russia (W.A.B.).
| |
Collapse
|
24
|
Roles of major histocompatibility complex class II in inducing protective immune responses to influenza vaccination. J Virol 2014; 88:7764-75. [PMID: 24760891 DOI: 10.1128/jvi.00748-14] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Major histocompatibility complex class II-deficient (MHC-II KO; Aβ(-/-)) mice were used to assess the roles of MHC-II molecules in inducing protective immune responses to vaccination. After vaccination with influenza A/PR8 virus-like particle (VLP) vaccine, in vivo and in vitro vaccine antigen-specific IgG isotype antibodies were not detected in MHC-II KO mice, which is quite different from CD4 T cell-deficient mice that induced vaccine-specific IgG antibodies. The deficiency in MHC-II did not significantly affect the induction of antigen-specific IgM antibody in sera. MHC-II KO mice that were vaccinated with influenza VLP, whole inactivated influenza virus, or live attenuated influenza virus vaccines were not protected against lethal infection with influenza A/PR8 virus. Adoptive transfer of fractionated spleen cells from wild-type mice to MHC-II KO mice indicated that CD43(+) cell populations with MHC-II contributed more significantly to producing vaccine-specific IgG antibodies than CD43(-) B220(+) conventional B cell or CD4 T cell populations, as well as conferring protection against lethal infection. Bone marrow-derived dendritic cells from MHC-II KO mice showed a significant defect in producing interleukin-6 and tumor necrosis factor alpha cytokines. Thus, results indicate that MHC-II molecules play multiple roles in inducing protective immunity to influenza vaccination. Importance: Major histocompatibility complex class II (MHC-II) has been known to activate CD4 T helper immune cells. A deficiency in MHC-II was considered to be equivalent to the lack of CD4 T cells in developing host immune responses to pathogens. However, the roles of MHC-II in inducing protective immune responses to vaccination have not been well understood. In the present study, we demonstrate that MHC-II-deficient mice showed much more significant defects in inducing protective antibody responses to influenza vaccination than CD4 T cell-deficient mice. Further analysis showed that CD43 marker-positive immune cells with MHC-II, as well as an innate immunity-simulating adjuvant, could rescue some defects in inducing protective immune responses in MHC-II-deficient mice. These results have important implications for our understanding of host immunity-inducing mechanisms to vaccination, as well as in developing effective vaccines and adjuvants.
Collapse
|
25
|
Aberrant glycosylation as biomarker for cancer: focus on CD43. BIOMED RESEARCH INTERNATIONAL 2014; 2014:742831. [PMID: 24689054 PMCID: PMC3943294 DOI: 10.1155/2014/742831] [Citation(s) in RCA: 280] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 12/10/2013] [Indexed: 11/23/2022]
Abstract
Glycosylation is a posttranslational modification of proteins playing a major role in cell signalling, immune recognition, and cell-cell interaction because of their glycan branches conferring structure variability and binding specificity to lectin ligands. Aberrant expression of glycan structures as well as occurrence of truncated structures, precursors, or novel structures of glycan may affect ligand-receptor interactions and thus interfere with regulation of cell adhesion, migration, and proliferation. Indeed, aberrant glycosylation represents a hallmark of cancer, reflecting cancer-specific changes in glycan biosynthesis pathways such as the altered expression of glycosyltransferases and glycosidases. Most studies have been carried out to identify changes in serum glycan structures. In most cancers, fucosylation and sialylation are significantly modified. Thus, aberrations in glycan structures can be used as targets to improve existing serum cancer biomarkers. The ability to distinguish differences in the glycosylation of proteins between cancer and control patients emphasizes glycobiology as a promising field for potential biomarker identification. In this review, we discuss the aberrant protein glycosylation associated with human cancer and the identification of protein glycoforms as cancer biomarkers. In particular, we will focus on the aberrant CD43 glycosylation as cancer biomarker and the potential to exploit the UN1 monoclonal antibody (UN1 mAb) to identify aberrant CD43 glycoforms.
Collapse
|
26
|
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
|
27
|
Intracellular patterns of sialophorin expression define a new molecular classification of breast cancer and represent new targets for therapy. Br J Cancer 2013; 110:146-55. [PMID: 24281005 PMCID: PMC3887278 DOI: 10.1038/bjc.2013.526] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Revised: 08/09/2013] [Accepted: 08/13/2013] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Sialophorin is a transmembrane sialoglycoprotein. Normally, the molecule is only produced by white blood cells where it regulates functions such as intercellular adhesion, intracellular signalling, apoptosis, migration and proliferation. METHODS Normal breast tissue and primary breast tumours were analysed by immunohistochemistry for sialophorin expression. The sialophorin-positive breast cancer cell line MCF7 was engineered to stably express either non-targeted or sialophorin-targeted small interfering RNA (siRNA). Assays were then performed in vitro to assess apoptosis, intracellular adhesion, transendothelial migration and cytotoxicity. An orthotopic mouse model assayed ability to produce tumours in vivo. RESULTS Normal breast epithelial cells exhibit expression of the N-terminal domain of sialophorin in the cytoplasm but not the nucleus. The majority of these normal cells are also negative for expression of the C-terminal domain. In contrast, malignant breast epithelial cells exhibit N-terminal expression both in the cytoplasm and nucleus and the majority express the C-terminus in the nucleus. Using differential patterns of intracellular expression of the N and C termini of sialophorin, we define six subtypes of breast cancer that are independent of histological and receptor status classification. Targeting sialophorin with siRNA resulted in the MCF7 breast cancer cell line exhibiting increased homotypic adhesion, decreased transendothelial migration, increased susceptibility to apoptosis, increased vulnerability to lysis by natural killer cells and decreased ability to produce tumours in mice. CONCLUSION Our results indicate that intracellular patterns of sialophorin expression define a new molecular classification of breast cancer and that sialophorin represents a novel therapeutic target.
Collapse
|
28
|
Kim MY, Cho JY. 20S-dihydroprotopanaxatriol modulates functional activation of monocytes and macrophages. J Ginseng Res 2013; 37:300-7. [PMID: 24198655 PMCID: PMC3818956 DOI: 10.5142/jgr.2013.37.300] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 03/18/2013] [Accepted: 03/18/2013] [Indexed: 11/18/2022] Open
Abstract
20S-dihydroprotopanaxatriol (2H-PPT) is a derivative of protopanaxatrol from ginseng. Unlike other components from Panax ginseng, the pharmacological activity of this compound has not been fully elucidated. In this study, we investigated the modulatory activity of 2H-PPT on the cellular responses of monocytes and macrophages to understand its immunoregulatory actions. 2H-PPT strongly upregulated the release of radicals in sodium nitroprusside-treated RAW264.7 cells and the surface levels of costimulatory molecule CD86. More importantly, this compound remarkably suppressed nitric oxide production, morphological changes, phagocytic uptake, cell-cell aggregation, and cell-matrix adhesion in RAW264.7 and U937 cells in the presence or absence of lipopolysaccharide, anti-CD43 antibody, fibronectin, and phorbal 12-myristate 13-acetate. Therefore, our results suggest that 2H-PPT can be applied as a novel functional immunoregulator of macrophages and monocytes.
Collapse
Affiliation(s)
- Mi-Yeon Kim
- School of Systems Biological Science, Soongsil University, Seoul 156-743, Korea
| | | |
Collapse
|
29
|
Ohnishi H, Sasaki H, Nakamura Y, Kato S, Ando K, Narimatsu H, Tachibana K. Regulation of cell shape and adhesion by CD34. Cell Adh Migr 2013; 7:426-33. [PMID: 24036614 DOI: 10.4161/cam.25957] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
We previously reported that expression of CD43/leukosialin induces cell rounding and microvillus formation via inhibition of cell adhesion. Here, we found that CD34, a cell surface sialomucin and marker for hematopoietic progenitor cells, also inhibited cell adhesion and induced cell rounding and microvillus formation. Forced expression of CD34-induced cell rounding, microvillus formation, and phosphorylation of ezrin/radixin/moesin (ERM) proteins in HEK293T cells, while inhibiting integrin-mediated cell re-attachment. Furthermore, CD34+ blood cells and KG-1 cells, which express endogenous CD34 on their surface, were spherical in shape, surrounded by microvilli, and non-adherent to substrata. In addition, cleavage of O-sialomucin augmented integrin-mediated cell adhesion of KG-1 cells. These results suggest the involvement of CD34 in the inhibition of integrin-mediated cell adhesion and formation of the cell surface structure. The inhibitory function of CD34 in cell adhesion may affect cell shape organization via phosphorylation of ERM proteins. Cellular structures such as the spherical shape and microvilli of CD34+ cells may also contribute to regulation of cell adhesion.
Collapse
Affiliation(s)
- Hiroe Ohnishi
- Research Center for Medical Glycoscience; National Institute of Advanced Industrial Science and Technology (AIST); Umezono, Tsukuba, Ibaraki, Japan; Health Research Institute; AIST; Nakouji, Amagasaki, Hyogo, Japan
| | - Hiroyuki Sasaki
- Department of Molecular Cell Biology, Institute of DNA Medicine, The Jikei University School of Medicine, Nishi-Shinbashi, Minato-ku, Tokyo 105, Japan
| | - Yoshihiko Nakamura
- Research Center for Regenerative Medicine; Tokai University School of Medicine; Bohseidai, Isehara, Kanagawa, Japan
| | - Shunichi Kato
- Department of Cell Transplantation and Regenerative Medicine; Tokai University School of Medicine; Bohseidai, Isehara, Kanagawa, Japan
| | - Kiyoshi Ando
- Research Center for Regenerative Medicine; Tokai University School of Medicine; Bohseidai, Isehara, Kanagawa, Japan; Department of Hematology/Oncology; Tokai University School of Medicine; Bohseidai, Isehara, Kanagawa, Japan
| | - Hisashi Narimatsu
- Research Center for Medical Glycoscience; National Institute of Advanced Industrial Science and Technology (AIST); Umezono, Tsukuba, Ibaraki, Japan
| | - Kouichi Tachibana
- Research Center for Medical Glycoscience; National Institute of Advanced Industrial Science and Technology (AIST); Umezono, Tsukuba, Ibaraki, Japan; Health Research Institute; AIST; Nakouji, Amagasaki, Hyogo, Japan
| |
Collapse
|
30
|
Ruiz-Perez F, Nataro JP. Bacterial serine proteases secreted by the autotransporter pathway: classification, specificity, and role in virulence. Cell Mol Life Sci 2013; 71:745-70. [PMID: 23689588 DOI: 10.1007/s00018-013-1355-8] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 04/25/2013] [Accepted: 04/29/2013] [Indexed: 01/07/2023]
Abstract
Serine proteases exist in eukaryotic and prokaryotic organisms and have emerged during evolution as the most abundant and functionally diverse group. In Gram-negative bacteria, there is a growing family of high molecular weight serine proteases secreted to the external milieu by a fascinating and widely employed bacterial secretion mechanism, known as the autotransporter pathway. They were initially found in Neisseria, Shigella, and pathogenic Escherichia coli, but have now also been identified in Citrobacter rodentium, Salmonella, and Edwardsiella species. Here, we focus on proteins belonging to the serine protease autotransporter of Enterobacteriaceae (SPATEs) family. Recent findings regarding the predilection of serine proteases to host intracellular or extracellular protein-substrates involved in numerous biological functions, such as those implicated in cytoskeleton stability, autophagy or innate and adaptive immunity, have helped provide a better understanding of SPATEs' contributions in pathogenesis. Here, we discuss their classification, substrate specificity, and potential roles in pathogenesis.
Collapse
Affiliation(s)
- Fernando Ruiz-Perez
- Department of Pediatrics, School of Medicine, University of Virginia, P.O.Box 800326, MR4 Room 4012C, 409 Lane Road, Charlottesville, VA, 22908, USA,
| | | |
Collapse
|
31
|
Mullaly SC, Oudhoff MJ, Min PH, Burrows K, Antignano F, Rattray DG, Chenery A, McNagny KM, Ziltener HJ, Zaph C. Requirement for core 2 O-glycans for optimal resistance to helminth infection. PLoS One 2013; 8:e60124. [PMID: 23555902 PMCID: PMC3612062 DOI: 10.1371/journal.pone.0060124] [Citation(s) in RCA: 7] [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: 01/27/2013] [Accepted: 02/21/2013] [Indexed: 11/19/2022] Open
Abstract
The migration of lymphocytes to the small intestine is controlled by expression of the integrin α4β7 and the chemokine receptor CCR9. However, the molecules that specifically regulate migration to the large intestine remain unclear. Immunity to infection with the large intestinal helminth parasite Trichuris muris is dependent upon CD4+ T cells that migrate to the large intestine. We examine the role of specific chemokine receptors, adhesion molecules and glycosyltransferases in the development of protective immunity to Trichuris. Mice deficient in expression of the chemokine receptors CCR2 or CCR6 were resistant to infection with Trichuris. Similarly, loss of CD34, CD43, CD44 or PSGL-1 had no effect on resistance to infection. In contrast, simultaneous deletion of the Core2 β1,6-N-acetylglucosaminyltransferase (C2GnT) enzymes C2GnT1 and C2Gnt2 resulted in delayed expulsion of worms. These results suggest that C2GnT-dependent modifications may play a role in migration of protective immune cells to the large intestine.
Collapse
MESH Headings
- Animals
- Antigens, CD34/genetics
- Antigens, CD34/metabolism
- CD4-Positive T-Lymphocytes/metabolism
- Hyaluronan Receptors/genetics
- Hyaluronan Receptors/metabolism
- Intestine, Large/metabolism
- Intestine, Large/parasitology
- Leukosialin/genetics
- Leukosialin/metabolism
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- N-Acetylglucosaminyltransferases/genetics
- N-Acetylglucosaminyltransferases/metabolism
- Polysaccharides/metabolism
- Real-Time Polymerase Chain Reaction
- Receptors, CCR2/genetics
- Receptors, CCR2/metabolism
- Receptors, CCR6/genetics
- Receptors, CCR6/metabolism
- Trichuriasis/genetics
- Trichuriasis/metabolism
- Trichuris/pathogenicity
Collapse
Affiliation(s)
- Sarah C. Mullaly
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Menno J. Oudhoff
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Paul H. Min
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kyle Burrows
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Frann Antignano
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - David G. Rattray
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alistair Chenery
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Kelly M. McNagny
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Hermann J. Ziltener
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Colby Zaph
- The Biomedical Research Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
32
|
Fu Q, Cash SE, Andersen JJ, Kennedy CR, Oldenburg DG, Zander VB, Foley GR, Simon Shelley C. CD43 in the nucleus and cytoplasm of lung cancer is a potential therapeutic target. Int J Cancer 2012; 132:1761-70. [PMID: 23015282 DOI: 10.1002/ijc.27873] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 08/31/2012] [Indexed: 12/27/2022]
Abstract
CD43 is a transmembrane sialoglycoprotein. Normally the molecule is only produced by white blood cells where it regulates functions such as intercellular adhesion, intracellular signaling, apoptosis, migration and proliferation. Two CD43 antibodies were used to interrogate 66 cases of non-small cell lung cancer (NSCLC) and 24 cases of small cell lung cancer (SCLC). In addition, we engineered the CD43-positive lung cancer cell line A549 to stably express either non-targeted or CD43-targeted small-interfering RNA (siRNA). These lines were then subjected to in vitro assays of apoptosis, natural killer (NK) cell cytotoxicity, intercellular adhesion and transendothelial migration. A xenograft mouse model evaluated the ability of the lines to grow primary tumors in vivo. CD43 was found to be expressed in the majority of both SCLC and NSCLC. Inclusive of CD43-negative tumors, differential patterns of nuclear and cytoplasmic expression of CD43 define four molecular subcategories of lung cancer. Targeting CD43 in A549 lung cancer cells, increased homotypic adhesion, decreased heterotypic adhesion and transendothelial migration, increased susceptibility to apoptosis and increased vulnerability to lysis by NK cells. Furthermore, targeting inhibited the growth of primary tumors in nude mice.
Collapse
Affiliation(s)
- Qiangwei Fu
- Department of Hematology/Oncology, Gundersen Medical Foundation, La Crosse, WI 54601, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Jeon YK, Min HS, Lee YJ, Kang BH, Kim EJ, Park HJ, Bae Y, Lee HG, Park WS, Song HG, Jung KC, Park SH. Targeting of a developmentally regulated epitope of CD43 for the treatment of acute leukemia. Cancer Immunol Immunother 2011; 60:1697-706. [PMID: 21710258 PMCID: PMC11028941 DOI: 10.1007/s00262-011-1066-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 06/14/2011] [Indexed: 02/02/2023]
Abstract
Previously, we developed a JL1 mouse monoclonal antibody that specifically recognizes the leukemic cells of T, B, and myeloid lineages, but not the peripheral blood cells and pluripotent hematopoietic stem cells. Here, we identified that JL1 mAb recognized a specific epitope of human CD43 and validated its potential as an anti-leukemic targeting agent. After the comprehensive screening of JL1 Ag in the human thymocyte cDNA library, multiple fusion gene constructs encoding human CD43 were generated to identify its specific epitope to JL1 antibody. JL1 antibody interacted with a developmentally regulated and non-glycosylated epitope of the human CD43 extracellular domain (AA 73-81, EGSPLWTSI). In an in vivo leukemia model using NOD/SCID mice injected with CCRF-CEM7 cells, JL1 antibody induced effective cytotoxicity in tumor cells and prolonged survival (p < 0.05). Saporin conjugation to JL1 antibody effectively depleted tumor cells in in vitro cytotoxic assays and also prolonged survival in a leukemic mouse model (p < 0.001). These preclinical results further support the therapeutic potential of the JL1 antibody in the management of acute leukemia.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Blotting, Western
- Cell Separation
- Disease Models, Animal
- Epitopes, B-Lymphocyte/immunology
- Flow Cytometry
- Humans
- Leukemia, Biphenotypic, Acute/drug therapy
- Leukemia, Biphenotypic, Acute/immunology
- Leukosialin/immunology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Real-Time Polymerase Chain Reaction
Collapse
Affiliation(s)
- Yoon Kyung Jeon
- Department of Pathology, Seoul National University College of Medicine, Seoul, 110-799 Korea
| | - Hye Sook Min
- Department of Pathology, Seoul National University College of Medicine, Seoul, 110-799 Korea
- Dinona Inc., Seoul, Korea
| | - Yoo Jeong Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, 110-799 Korea
- Graduate Program of Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Byung Hyun Kang
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Ji Kim
- Graduate Program of Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Hyo Jin Park
- Department of Pathology, Seoul National University Bundang Hospital, Bundang, Korea
| | - Youngmee Bae
- Department of Parasitology and Tropical Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Gyu Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, 110-799 Korea
- Graduate Program of Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Weon Seo Park
- Department of Pathology and Research Center for Hematopoietic Tumor, National Cancer Center, Goyang, Korea
| | - Hyung Geun Song
- Dinona Inc., Seoul, Korea
- Department of Pathology, Chungbuk National University, Cheongju, Korea
| | - Kyeong Cheon Jung
- Department of Pathology, Seoul National University College of Medicine, Seoul, 110-799 Korea
- Dinona Inc., Seoul, Korea
- Graduate Program of Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Seong Hoe Park
- Department of Pathology, Seoul National University College of Medicine, Seoul, 110-799 Korea
- Graduate Program of Immunology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
34
|
Serine protease autotransporters from Shigella flexneri and pathogenic Escherichia coli target a broad range of leukocyte glycoproteins. Proc Natl Acad Sci U S A 2011; 108:12881-6. [PMID: 21768350 DOI: 10.1073/pnas.1101006108] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The serine protease autotransporters of Enterobacteriaceae (SPATEs) are secreted by pathogenic Gram-negative bacteria through the autotransporter pathway. We previously classified SPATE proteins into two classes: cytotoxic (class 1) and noncytotoxic (class 2). Here, we show that Pic, a class 2 SPATE protein produced by Shigella flexneri 2a, uropathogenic and enteroaggregative Escherichia coli strains, targets a broad range of human leukocyte adhesion proteins. Substrate specificity was restricted to glycoproteins rich in O-linked glycans, including CD43, CD44, CD45, CD93, CD162 (PSGL-1; P-selectin glycoprotein ligand 1), and the surface-attached chemokine fractalkine, all implicated in leukocyte trafficking, migration, and inflammation. N-terminal sequencing of proteolytic products revealed Pic (protease involved in colonization) cleavage sites to occur before Thr or Ser residues. The purified carbohydrate sLewis-X implied in inflammation and malignancy inhibited cleavage of PSGL-1 by Pic. Exposure of human leukocytes to purified Pic resulted in polymorphonuclear cell activation, but impaired chemotaxis and transmigration; Pic-treated T cells underwent programmed cell death. We also show that the Pic-related protease Tsh/Hbp, implicated in extraintestinal infections, exhibited a spectrum of substrates similar to those cleaved by Pic. In the guinea pig keratoconjunctivitis model, a Shigella pic mutant induced greater inflammation than its parent strain. We suggest that the class-2 SPATEs represent unique immune-modulating bacterial virulence factors.
Collapse
|
35
|
Yamane J, Ohnishi H, Sasaki H, Narimatsu H, Ohgushi H, Tachibana K. Formation of microvilli and phosphorylation of ERM family proteins by CD43, a potent inhibitor for cell adhesion: cell detachment is a potential cue for ERM phosphorylation and organization of cell morphology. Cell Adh Migr 2011; 5:119-32. [PMID: 21045567 DOI: 10.4161/cam.5.2.13908] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
CD43/sialophorin/leukosialin, a common leukocyte antigen, is known as an inhibitor for cell adhesion. The ectodomain of CD43 is considered as a molecular barrier for cell adhesion, while the cytoplasmic domain has a binding site for Ezrin/Radixin/Moesin (ERM). We found expression of CD43 induced cell rounding, inhibition of cell re-attachment, augmentation of microvilli, and phosphorylation of ERM in HEK293T cells. Mutant studies revealed the ectodomain of CD43, but not the intracellular domain, essential and sufficient for all these phenomena. We also found that forced cell detachment by itself induced phosphorylation of ERM in HEK293T cells. Taken together, these findings indicate that inhibition of cell adhesion by the ectodomain of CD43 induces phosphorylation of ERM, microvilli formation, and eventual cell rounding. Furthermore, our study suggests a novel possibility that cell detachment itself induces activation of ERM and modification of cell shape.
Collapse
Affiliation(s)
- Junko Yamane
- Health Research Institute, Tissue Engineering Research Group, National Institute of Advanced Industrial Science and Technology, Nakouji, Amagasaki, Hyogo, Japan
| | | | | | | | | | | |
Collapse
|
36
|
Seyerl M, Kirchberger S, Majdic O, Seipelt J, Jindra C, Schrauf C, Stöckl J. Human rhinoviruses induce IL-35-producing Treg via induction of B7-H1 (CD274) and sialoadhesin (CD169) on DC. Eur J Immunol 2010; 40:321-9. [PMID: 19950173 DOI: 10.1002/eji.200939527] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
IL-35 is a heterodimer of EBV-induced gene 3 and of the p35 subunit of IL-12, and recently identified as an inhibitory cytokine produced by natural Treg in mice, but not in humans. Here we demonstrate that DC activated by human rhinoviruses (R-DC) induce IL-35 production and release, as well as a suppressor function in CD4(+) and CD8(+) T cells derived from human peripheral blood but not in naïve T cells from cord blood. The induction of IL-35-producing T cells by R-DC was FOXP3-independent, but blocking of B7-H1 (CD274) and sialoadhesin (CD169) on R-DC with mAb against both receptors prevented the induction of IL-35. Thus, the combinatorial signal delivered by R-DC to T cells via B7-H1 and sialoadhesin is crucial for the induction of human IL-35(+) Treg. These results demonstrate a novel pathway and its components for the induction of immune-inhibitory T cells.
Collapse
Affiliation(s)
- Maria Seyerl
- Institute of Immunology, Medical University of Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
37
|
Benton KD, Hermann RJ, Vomhof-DeKrey EE, Haring JS, Van der Steen T, Smith J, Dovat S, Dorsam GP. A transcriptionally permissive epigenetic landscape at the vasoactive intestinal peptide receptor-1 promoter suggests a euchromatin nuclear position in murine CD4 T cells. REGULATORY PEPTIDES 2009; 158:68-76. [PMID: 19729043 PMCID: PMC2771554 DOI: 10.1016/j.regpep.2009.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/03/2009] [Revised: 07/02/2009] [Accepted: 08/16/2009] [Indexed: 01/19/2023]
Abstract
T cells express receptors for neuropeptides that mediate immunological activities. Vasoactive intestinal peptide receptor-1 (VPAC1), the prototypical group II G protein coupled receptor, binds two neuropeptides with high-affinity, called vasoactive intestinal peptide and pituitary adenylate cyclase activating polypeptide. During T cell signaling, VPAC1 mRNA expression levels are significantly downregulated through a Src kinase dependent mechanism, thus altering the sensitivity for these neuropeptides during an immune reaction. Presently, it is unknown whether the mechanism that regulates VPAC1 during T cell signaling involves epigenetic changes. Therefore, we hypothesized that the epigenetic landscape consisting of diacetylation at H3K9/14 and trimethylation at H3K4, two transcriptionally permissive histone modifications, would parallel VPAC1 expression showing high enrichment in untreated T cells, but lower enrichment in alpha-CD3 treated T cells. To this end, quantitative chromatin immunoprecipitation (ChIP) analysis of H3K9/14ac and H3K4me3 was conducted using purified CD4(+) T cells, with CD45R(+) B cells as a negative control. Our data revealed that these histone modifications at the VPAC1 promoter did indeed parallel its mRNA levels between T and B lymphocytes, but did not decrease during T cell signaling. Collectively, these data strongly imply a euchromatin nuclear position for the VPAC1 locus irrespective of the activation status of T cells.
Collapse
Affiliation(s)
- K D Benton
- Department of Chemistry and Molecular Biology, North Dakota State University, Fargo, ND 58108, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Ramírez-Pliego O, Escobar-Zárate DL, Rivera-Martínez GM, Cervantes-Badillo MG, Esquivel-Guadarrama FR, Rosas-Salgado G, Rosenstein Y, Santana MA. CD43 signals induce Type One lineage commitment of human CD4+ T cells. BMC Immunol 2007; 8:30. [PMID: 18036228 PMCID: PMC2235884 DOI: 10.1186/1471-2172-8-30] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2007] [Accepted: 11/23/2007] [Indexed: 12/20/2022] Open
Abstract
Background The activation and effector phenotype of T cells depend on the strength of the interaction of the TcR with its cognate antigen and additional signals provided by cytokines and by co-receptors. Lymphocytes sense both the presence of an antigen and also clues from antigen-presenting cells, which dictate the requisite response. CD43 is one of the most abundant molecules on the surface of T cells; it mediates its own signalling events and cooperates with those mediated by the T cell receptor in T cell priming. We have examined the role of CD43 signals on the effector phenotype of adult CD4+ and CD8+ human T cells, both alone and in the presence of signals from the TcR. Results CD43 signals direct the expression of IFNγ in human T cells. In freshly isolated CD4+ T cells, CD43 signals potentiated expression of the IFNγ gene induced by TcR activation; this was not seen in CD8+ T cells. In effector cells, CD43 signals alone induced the expression of the IFNγ gene in CD4+ T cells and to a lesser extent in CD8+ cells. The combined signals from CD43 and the TcR increased the transcription of the T-bet gene in CD4+ T cells and inhibited the transcription of the GATA-3 gene in both populations of T cells, thus predisposing CD4+ T cells to commitment to the T1 lineage. In support of this, CD43 signals induced a transient membrane expression of the high-affinity chains of the receptors for IL-12 and IFNγ in CD4+ T cells. CD43 and TcR signals also cooperated with those of IL-12 in the induction of IFNγ expression. Moreover, CD43 signals induced the co-clustering of IFNγR and the TcR and cooperated with TcR and IL-12 signals, triggering a co-capping of both receptors in CD4+ populations, a phenomenon that has been associated with a T1 commitment. Conclusion Our results suggest a key role for CD43 signals in the differentiation of human CD4+ T cells into a T1 pattern.
Collapse
Affiliation(s)
- Oscar Ramírez-Pliego
- Facultad de Ciencias, Universidad Autónoma del Estado de Morelos, Av, Universidad 1001, Col, Chamilpa, Cuernavaca, 62210, Mexico.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
The vast majority of peripheral T cells exist as resting lymphocytes until a signal for activation has been received. In response to antigen, this activation involves ligation of the T-cell receptor (TCR) and signal transmission through the CD3 complex, which then initiates a cascade of intracellular events that lead to the expression of genes used in T-cell activation. T-cell activation also requires soluble mediators in the form of cytokines and chemokines that regulate the process in both positive and negative ways, and costimulatory signals received in conjunction with TCR/CD3 signaling are important in the activation of T cells. Unlike T cells in other peripheral immune compartments, small and large intestinal intraepithelial lymphocytes (IELs) bear some but not all properties of activated T cells, suggesting that they constitute a large population of 'partially activated' effector cells. Thus, regulation of the IEL activation process must be held in tight check, yet it must be ready to respond to foreign antigen rapidly and effectively. We discuss how costimulatory molecules may hold the key to controlling IEL activation through a multiphase process beginning with cells that have already entered into the early stage of activation.
Collapse
Affiliation(s)
- Dina Montufar-Solis
- Department of Diagnostic Sciences, Dental Branch, University of Texas Health Science Center at Houston, Houston, TX, USA
| | | | | |
Collapse
|
40
|
Jiang HR, Hwenda L, Makinen K, Oetke C, Crocker PR, Forrester JV. Sialoadhesin promotes the inflammatory response in experimental autoimmune uveoretinitis. THE JOURNAL OF IMMUNOLOGY 2006; 177:2258-64. [PMID: 16887986 DOI: 10.4049/jimmunol.177.4.2258] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Macrophages are a prominent component of the effector cell compartment in a number of CD4+ T cell-mediated organ-specific autoimmune diseases. In this study, we investigated the role of the sialic acid binding Ig-like lectin sialoadhesin (Sn, Siglec-1) in a model of interphotoreceptor retinal binding protein peptide-induced experimental autoimmune uveoretinitis in mice with targeted deletion of Sn. Our data show that compared with wild-type mice, experimental autoimmune uveoretinitis is reduced in severity in the initial stages in the Sn knockout (KO) mice. In addition, there is a reduction in the proliferative capacity of T cells from the KO mice draining lymph nodes after immunization with interphotoreceptor retinal binding protein peptides, which is manifest some days before disease onset and persists for the duration of disease. Furthermore, activated T cells from the draining lymph nodes of Sn KO mice secrete lower levels of IFN-gamma. The data suggest a role for Sn in "fine tuning" the immune response to autoantigens by modulating T cell priming.
Collapse
Affiliation(s)
- Hui-Rong Jiang
- Department of Ophthalmology, Medical School Foresterhill, Aberdeen, United Kingdom
| | | | | | | | | | | |
Collapse
|
41
|
Kirchberger S, Vetr H, Majdic O, Stockinger H, Stöckl J. Engagement of ICAM-1 by major group rhinoviruses activates the LFA-1/ICAM-3 cell adhesion pathway in mononuclear phagocytes. Immunobiology 2006; 211:537-47. [PMID: 16920492 DOI: 10.1016/j.imbio.2006.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cell-cell interactions are critical at key points of immune responses and are mediated by a complex array of adhesion receptors. One of the most important adhesion molecules on leukocytes is intercellular adhesion molecule 1 (ICAM-1, CD54). Here we demonstrate that engagement of ICAM-1 with human major group rhinoviruses (HRV) enhances adhesiveness and homotypic aggregation of human monocytes and monocyte-derived dendritic cells (DC). Cluster formation upon engagement of ICAM-1 with HRV14 represents an active process. It is temperature and energy dependent, requires divalent cations, an intact cytoskeleton and protein de novo synthesis. Homotypic interaction between monocytes induced by HRV14 can be inhibited with blocking mAbs against LFA-1 (CD11a/CD18) and ICAM-3 (CD50) as well as with a mAb against the first immunoglobulin (Ig)-domain of PECAM-1 (CD31). Induction of enhanced cytoadhesiveness by HRV14 was not accompanied with an upregulation of LFA-1, ICAM-3 or PECAM-1 expression. Binding studies with recombinant PECAM-1 proteins indicated, however, that monocyte clustering upon engagement of ICAM-1 with HRV was accompanied with increased homophilic PECAM-1 interactions. Taken together the results of our study demonstrate that signalling via ICAM-1 induces adhesiveness of mononuclear phagocytes, which critically involves PECAM-1 and is mediated via LFA-1/ICAM-3.
Collapse
Affiliation(s)
- Stefanie Kirchberger
- Institute of Immunology, Medical University of Vienna, Borschkegasse 8a, A-1090, Austria
| | | | | | | | | |
Collapse
|
42
|
Fierro NA, Pedraza-Alva G, Rosenstein Y. TCR-Dependent Cell Response Is Modulated by the Timing of CD43 Engagement. THE JOURNAL OF IMMUNOLOGY 2006; 176:7346-53. [PMID: 16751378 DOI: 10.4049/jimmunol.176.12.7346] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Binding of Ag by the Ag receptor in combination with other stimuli provided by costimulatory receptors triggers the expansion and differentiation of T lymphocytes. However, it is unclear whether the time when costimulatory molecules interact with their counterreceptors with regards to Ag recognition leads to different T cell responses. Provided that the coreceptor molecule CD43 is a very abundant molecule evenly distributed on the membrane of T cell surface protruding 45 nm from the cell, we hypothesized that CD43 is one of the first molecules that interacts with the APC and thus modulates TCR activation. We show that engaging CD43 before or simultaneously with the TCR inhibited Lck-Src homology 2 domain containing phosphatase-1 interaction, preventing the onset of a negative feedback loop on TCR signals, favoring high levels of IL-2, cell proliferation, and secretion of proinflammatory cytokines and chemokines. In contrast, the intracellular signals resulting of engaging the TCR before CD43 were insufficient to induce IL-2 production and cell proliferation. Interestingly, when stimulated through the TCR and CD28, cells proliferated vigorously, independent of the order with which molecules were engaged. These results indicate that CD43 induces a signaling cascade that prolongs the duration of TCR signaling and support the temporal summation model for T cell activation. In addition to the strength and duration of intracellular signals, our data underscore temporality with which certain molecules are engaged as yet another mechanism to fine tune T cell signal quality, and ultimately immune function.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal/metabolism
- Antibodies, Monoclonal/pharmacology
- Cell Proliferation
- Cells, Cultured
- Clonal Anergy/immunology
- Cytokines/biosynthesis
- Humans
- Intracellular Signaling Peptides and Proteins/metabolism
- JNK Mitogen-Activated Protein Kinases/physiology
- Leukosialin/immunology
- Leukosialin/metabolism
- Leukosialin/physiology
- Ligands
- Lymphocyte Activation/immunology
- MAP Kinase Signaling System/immunology
- Membrane Proteins/metabolism
- Mitogen-Activated Protein Kinase 1/physiology
- Phosphorylation
- Protein Phosphatase 1
- Protein Tyrosine Phosphatase, Non-Receptor Type 6
- Protein Tyrosine Phosphatases/metabolism
- Receptors, Antigen, T-Cell/antagonists & inhibitors
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/physiology
- T-Lymphocytes/enzymology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Time Factors
Collapse
Affiliation(s)
- Nora A Fierro
- Instituto de Biotecnología and Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Cuernavaca, Morelos
| | | | | |
Collapse
|
43
|
Montufar-Solis D, Garza T, Teng BB, Klein JR. Upregulation of ICOS on CD43+ CD4+ murine small intestinal intraepithelial lymphocytes during acute reovirus infection. Biochem Biophys Res Commun 2006; 342:782-90. [PMID: 16500623 PMCID: PMC2894703 DOI: 10.1016/j.bbrc.2006.02.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Accepted: 02/08/2006] [Indexed: 11/24/2022]
Abstract
Murine intestinal intraepithelial lymphocytes (IELs) can be classified according to expression of a CD43 glycoform recognized by the S7 monoclonal antibody. In this study, we examined the response of S7+ and S7- IELs in mice during acute reovirus serotype 3 (Dearing strain) infection, which was confirmed by virus-specific real-time PCR. In vivo proliferation increased significantly for both S7- and S7+ IELs on day 4 post-infection as determined by BrdU incorporation; however, expression of the inducible costimulatory (ICOS) molecule, which peaked on day 7 post-infection, was upregulated on S7+ CD4+ T cells, most of which were CD4+8- IELs. In vitro ICOS stimulation by syngeneic peritoneal macrophages induced IFN-gamma secretion from IELs from day 7 infected mice, and was suppressed by treatment with anti-ICOS mAb. Additionally, IFN-gamma mRNA increased in CD4+ IELs on day 6 post-infection. These findings indicate that S7- and S7+ IELs are differentially mobilized during the immune response to reovirus infection; that the regulated expression of ICOS is associated with S7+ IELs; and that stimulation of IELs through ICOS enhances IFN-gamma synthesis during infection.
Collapse
Affiliation(s)
- Dina Montufar-Solis
- Department of Diagnostic Sciences, Dental Branch, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
44
|
Montufar-Solis D, Garza T, Klein JR. Selective upregulation of immune regulatory and effector cytokine synthesis by intestinal intraepithelial lymphocytes following CD43 costimulation. Biochem Biophys Res Commun 2005; 338:1158-63. [PMID: 16246302 PMCID: PMC2894695 DOI: 10.1016/j.bbrc.2005.10.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2005] [Accepted: 10/12/2005] [Indexed: 10/25/2022]
Abstract
The involvement of the CD43 molecule in the activation of mouse small intestinal intraepithelial lymphocytes (IELs) has been studied using a panel of twenty-two regulatory and effector immune response analytes. In the absence of stimulation in vitro, IELs produced low levels of CCL5 only. Upon CD3 stimulation, the activity of seven of twenty-two analytes was elevated relative to unstimulated cultures, including several proinflammatory cytokines and chemokines. Notably, CD3 stimulation in the presence of CD43 costimulation resulted in elevated levels of five analytes (interleukin-2, interferon-gamma, CCL5, granulocyte colony-stimulating factor, and granulocyte-monocyte colony-stimulating factor) above that produced by CD3 stimulation alone. That CD43 costimulation was responsible for elevated cytokine/chemokine activity was confirmed at the transcriptional level by real-time PCR for IFN-gamma and CCL5, and by ELISA for IFN-gamma. These findings open the way to a better understanding of the process by which T cells are activated in the intestinal epithelium.
Collapse
Affiliation(s)
- Dina Montufar-Solis
- Department of Diagnostic Sciences, Dental Branch, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tomas Garza
- Department of Diagnostic Sciences, Dental Branch, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - John R. Klein
- Department of Diagnostic Sciences, Dental Branch, The University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
45
|
Randhawa AK, Ziltener HJ, Merzaban JS, Stokes RW. CD43 is required for optimal growth inhibition of Mycobacterium tuberculosis in macrophages and in mice. THE JOURNAL OF IMMUNOLOGY 2005; 175:1805-12. [PMID: 16034122 DOI: 10.4049/jimmunol.175.3.1805] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We explored the role of macrophage (Mphi) CD43, a transmembrane glycoprotein, in the pathogenesis of Mycobacterium tuberculosis. Using gene-deleted mice (CD43-/-), we assessed the association of the bacterium with distinct populations of Mphi and found that CD43-/- Mphi bound less M. tuberculosis than CD43+/+ Mphi. Increased infective doses did not abrogate this difference. However, reduced association due to the absence of CD43 could be overcome by serum components. Mphi from heterozygote mice, which express 50% of wild-type CD43, bound more bacteria than CD43-/- but less than CD43+/+, proving that the gene dose of CD43 correlates with binding of M. tuberculosis. Furthermore, the reduced ability of CD43-/- Mphi to bind bacteria was restricted to mycobacterial species. We also found that the survival and replication of M. tuberculosis within Mphi was enhanced significantly in the absence of CD43, making this the first demonstration that the mechanism of mycobacterial entry influences its subsequent growth. Most importantly, we show here that the absence of CD43 in mice aerogenically infected with M. tuberculosis results in an increased bacterial load during both the acute and chronic stages of infection and more rapid development of granulomas, with greater lung involvement and distinctive cellularity.
Collapse
MESH Headings
- Acute Disease
- Administration, Inhalation
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/physiology
- Bacterial Adhesion/genetics
- Bacterial Adhesion/immunology
- Bone Marrow Cells/immunology
- Bone Marrow Cells/microbiology
- Cell Adhesion/genetics
- Cell Adhesion/immunology
- Cell Membrane/immunology
- Cell Membrane/metabolism
- Cell Membrane/microbiology
- Chronic Disease
- Gene Dosage
- Growth Inhibitors/biosynthesis
- Growth Inhibitors/deficiency
- Growth Inhibitors/genetics
- Growth Inhibitors/physiology
- Immunophenotyping
- Leukosialin
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Macrophages, Alveolar/immunology
- Macrophages, Alveolar/microbiology
- Macrophages, Alveolar/pathology
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/microbiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mycobacterium tuberculosis/growth & development
- Mycobacterium tuberculosis/immunology
- Mycobacterium tuberculosis/pathogenicity
- Opsonin Proteins/metabolism
- Salmonella typhimurium/genetics
- Salmonella typhimurium/immunology
- Sialoglycoproteins/biosynthesis
- Sialoglycoproteins/deficiency
- Sialoglycoproteins/genetics
- Sialoglycoproteins/physiology
- Tuberculosis, Pulmonary/genetics
- Tuberculosis, Pulmonary/immunology
- Tuberculosis, Pulmonary/microbiology
- Tuberculosis, Pulmonary/pathology
Collapse
Affiliation(s)
- April K Randhawa
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
46
|
Kirchberger S, Majdic O, Steinberger P, Blüml S, Pfistershammer K, Zlabinger G, Deszcz L, Kuechler E, Knapp W, Stöckl J. Human Rhinoviruses Inhibit the Accessory Function of Dendritic Cells by Inducing Sialoadhesin and B7-H1 Expression. THE JOURNAL OF IMMUNOLOGY 2005; 175:1145-52. [PMID: 16002716 DOI: 10.4049/jimmunol.175.2.1145] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Dendritic cells (DC) are professional APCs with an unmatched ability to interact with and activate T cells. There is accumulating evidence that DC not only efficiently stimulate T cell activation but also regulate T cell responses. However, little is known about cell surface structures on DC involved in the regulation of T cell responses. We demonstrate that human rhinoviruses (HRV) can efficiently inhibit the accessory function of DC through induction of inhibitory cell surface receptors. We observed that treatment of DC with HRV14 (R-DC), a member of the major group HRV family, diminished their T cell stimulatory capacity and induced a promiscuous and deep anergic state in cocultured T cells despite high levels of MHC molecules as well as costimulatory molecules, e.g., B7-1 (CD80) and B7-2 (CD86), and independent of inhibitory soluble factors such as IL-10. In contrast, expression of inhibitory B7-H1 molecules was up-regulated and R-DC de novo expressed sialoadhesin (Sn). Most importantly, blocking of B7-H1 and Sn on R-DC with specific mAbs against both receptors reverted the inhibitory phenotype. Thus, inhibitory signals delivered from R-DC to T cells via B7-H1 and Sn were critical for the induction of anergy. These observations suggest that an altered accessory molecule repertoire on DC upon interaction with HRV down-modulates adaptive immune responses during the viral infection.
Collapse
Affiliation(s)
- Stefanie Kirchberger
- Institute of Immunology and Institute of Biochemistry, Medical University of Vienna, Borschkegasse 8a, A-1090 Vienna, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee PS, Beneck D, Weisberger J, Gorczyca W. Coexpression of CD43 by Benign B Cells in the Terminal Ileum. Appl Immunohistochem Mol Morphol 2005; 13:138-41. [PMID: 15894925 DOI: 10.1097/01.pai.0000126115.09998.6c] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Expression of CD43 by B cells is often used as a diagnostic criterion in favor of a B-cell lymphoproliferative disorder, including small lymphocytic lymphoma/chronic lymphocytic leukemia, mantle cell lymphoma, Burkitt lymphoma, precursor B-lymphoblastic lymphoma, and a subset of marginal zone B-cell lymphomas. Benign B cells generally do not coexpress CD43. The authors analyzed 20 biopsies of the terminal ileum for nonneoplastic disease for expression of CD43 and compared them with other sites and with CD20, CD138, and CD3 reactivity. The majority of cases (85%) showed strong coexpression of CD43 by benign perifollicular B cells. The presence of CD43 coexpression in B-cell populations of the terminal ileum, including those of Peyer's patches, should not be used as a diagnostic parameter to differentiate extranodal marginal zone B-cell lymphoma of MALT type from reactive processes.
Collapse
Affiliation(s)
- Po-Shing Lee
- From Division of Hematopathology, IMPATH Inc, New York, NY 10019, USA
| | | | | | | |
Collapse
|
48
|
del Rio R, Rincón M, Layseca-Espinosa E, Fierro NA, Rosenstein Y, Pedraza-Alva G. PKCtheta is required for the activation of human T lymphocytes induced by CD43 engagement. Biochem Biophys Res Commun 2005; 325:133-43. [PMID: 15522211 DOI: 10.1016/j.bbrc.2004.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2004] [Indexed: 12/12/2022]
Abstract
The turnover of phosphoinositides leading to PKC activation constitutes one of the principal axes of intracellular signaling. In T lymphocytes, the enhanced and prolonged PKC activation resulting from the engagement of the TcR and co-receptor molecules ensures a productive T cell response. The CD43 co-receptor promotes activation and proliferation, by inducing IL-2 secretion and CD69 expression. CD43 engagement has been shown to promote phosphoinositide turnover and DAG production. Moreover, PKC activation was found to be required for the activation of the MAP kinase pathway in response to CD43 ligation. Here we show that CD43 engagement led to the membrane translocation and enzymatic activity of specific PKC isoenzymes: cPKC (alpha/beta), nPKC (epsilon and theta;), aPKC (zeta) and PKCmu. We also show that activation of PKCtheta; resulting from CD43 ligation induced CD69 expression through an ERK-dependent pathway leading to AP-1, NF-kappaB activation and an ERK independent pathway promoting NFAT activation. Together, these data suggest that PKCtheta; plays a critical role in the co-stimulatory functions of CD43 in human T cells.
Collapse
Affiliation(s)
- Roxana del Rio
- Instituto de Biotecnología/Universidad Nacional Autónoma de México, AP 510-3 Cuernavaca, Mor. 62250, Mexico
| | | | | | | | | | | |
Collapse
|
49
|
Wang HC, Montufar-Solis D, Teng BB, Klein JR. Maximum Immunobioactivity of Murine Small Intestinal Intraepithelial Lymphocytes Resides in a Subpopulation of CD43+ T Cells. THE JOURNAL OF IMMUNOLOGY 2004; 173:6294-302. [PMID: 15528368 DOI: 10.4049/jimmunol.173.10.6294] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD43 has been linked to many function-associated T cell activities. Using mAbs that recognize two different CD43 determinants, we show that, although mouse small intestinal intraepithelial lymphocytes (IELs) expressed the CD43 core molecule reactive with mAb R2/60, only about one-half of the total IELs-including some but not all of the TCRalphabeta and TCRgammadelta cells-expressed the CD43 S7(-) reactive determinant. CD43 S7(+) IELs secreted more IL-2, IL-4, IL-10, IL-17, and IFN-gamma following anti-CD3 stimulation, and were >4-fold more cytotoxic in fresh isolates and >16-fold more cytotoxic after anti-CD3 stimulation, than S7(-) IELs. S7(+) but not S7(-) IELs from the ileum of IL-10(-/-) mice spontaneously produced IFN-gamma. In vivo BrdU uptake by IELs in non-Ag-primed mice was greatest in the S7(+) population, indicating that significantly more S7(+) IELs than S7(-) IELs undergo cell expansion under normal homeostatic conditions. DNA microarray analyses showed that S7(+) IELs expressed higher levels of genes associated with activated T cells, whereas S7(-) IELs expressed genes used in the regulation of NK cells. These findings define two functionally distinct populations of IELs based on CD43 expression independent of TCR class, and they identify a subset of IELs that may serve as a target to better control intestinal inflammation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/metabolism
- Antigens, CD/biosynthesis
- Antigens, CD/immunology
- CD3 Complex/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Proliferation
- Cytokines/metabolism
- Cytotoxicity Tests, Immunologic
- Dose-Response Relationship, Immunologic
- Female
- Interferon-gamma/biosynthesis
- Interleukin-10/deficiency
- Interleukin-10/genetics
- Intestinal Mucosa/cytology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestine, Small/cytology
- Intestine, Small/immunology
- Intestine, Small/metabolism
- Leukosialin
- Lymphocyte Activation/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Oligonucleotide Array Sequence Analysis/methods
- Protein Isoforms/biosynthesis
- Protein Isoforms/immunology
- Sialoglycoproteins/biosynthesis
- Sialoglycoproteins/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Th1 Cells/immunology
- Th2 Cells/immunology
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Heuy-Ching Wang
- Department of Diagnostic Sciences, Dental Branch, University of Texas Health Science Center at Houston, 6516 M.D. Anderson Boulevard, Houston, TX 77030, USA
| | | | | | | |
Collapse
|
50
|
Kyoizumi S, Ohara T, Kusunoki Y, Hayashi T, Koyama K, Tsuyama N. Expression characteristics and stimulatory functions of CD43 in human CD4+ memory T cells: analysis using a monoclonal antibody to CD43 that has a novel lineage specificity. THE JOURNAL OF IMMUNOLOGY 2004; 172:7246-53. [PMID: 15187099 DOI: 10.4049/jimmunol.172.12.7246] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We have used HSCA-2, an mAb that recognizes a sialic acid-dependent epitope on the low molecular mass (approximately 115-kDa) glycoform of CD43 that is expressed in resting T and NK cells, to examine the expression characteristics and stimulatory functions of CD43 in human CD4+ memory T cells. Having previously reported that the memory cells that respond to recall Ags in a CD4+ CD45RO+ T cell population almost all belong to a subset whose surface CD43 expression levels are elevated, we now find that exposing these same memory T cells to HSCA-2 mAb markedly increases their proliferative responsiveness to recall Ags. We think it unlikely that this increase in responsiveness is a result of CD43-mediated monocyte activation, especially given that the HSCA-2 mAb differs from all previously used CD43 mAbs in having no obvious binding specificity for monocyte CD43. Predictably, treatment with HSCA-2 mAb did not lead to significant recall responses in CD4+ CD45RO+ T cells, whose CD43 expression levels were similar to or lower than those of naive cells. Other experiments indicated that the HSCA-2 mAb was capable of enhancing the proliferative responsiveness of CD4+ memory T cells that had been exposed to polyclonal stimulation by monocyte-bound CD3 mAb and could also act in synergy with CD28 mAb to enhance the responsiveness of CD4+ T cells to CD3 stimulation. Taken together, these findings suggest that the CD43 molecules expressed on CD4+ memory T cells may be capable of enhancing the costimulatory signaling and hence providing accessory functions to TCR-mediated activation processes.
Collapse
Affiliation(s)
- Seishi Kyoizumi
- Laboratory of Immunology, Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, 5-2 Hijiyama Park, Minami Ward, Hiroshima 732-0815, Japan.
| | | | | | | | | | | |
Collapse
|