1
|
Xu J, Kim DH, Wang W, Li S, Lin P, Tang G, Konoplev S, Qiu L, Fang H, Garces S, Leventaki V, E S, Medeiros LJ, Wang SA. Flow cytometric immunophenotypic features of acute myeloid leukemia with mast cell differentiation. Am J Clin Pathol 2024:aqae116. [PMID: 39244687 DOI: 10.1093/ajcp/aqae116] [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: 05/28/2024] [Accepted: 08/15/2024] [Indexed: 09/10/2024] Open
Abstract
OBJECTIVES Acute myeloid leukemia (AML) with mast cell (MC) differentiation was recently described as an aggressive subgroup of AML cases. The objectives of this study were to assess the flow cytometric immunophenotypic features of AML-MC cases. METHODS We characterized the immunophenotypic features of 21 AML-MC cases by flow cytometry and compared them to 20 reactive/regenerating bone marrow specimens. RESULTS The number of MCs detected by flow cytometry in AML-MC cases ranged from 0.4% to 21.1%, with a median of 3.5%, significantly higher than that of normal/reactive bone marrow (BM) (median, 0.01%; range, 0.000%-0.396%; P < .0001). Immunophenotypically, MCs in AML-MC cases demonstrated immaturity, differing from MCs in normal/reactive BMs, including dimmer CD45 (100% vs 0%), lower side scatter (100% vs 0%), more frequent CD34 (81% vs 20%), and CD123 (100% vs 10%) positivity, and more frequent uniform/increased CD38 expression (95% vs 20%) (all P ≤ .0001). CD2 (0/5) and CD25 (2/6, 1 uniform and 1 partial) were assessed in a subset of cases. The myeloblasts in AML-MC were typically CD34+CD117+HLA-DR+ with unusually frequent expression of CD56 (57%, all partial) and CD25 (63%, mostly partial), increased CD117 (62%), and decreased CD38 (86%). The MC percentage determined by flow cytometry correlated well with MCs detected by tryptase immunohistochemistry (r = 0.76, P < .001). CONCLUSIONS The MCs in AML-MC cases are characterized by dim CD45, low side scatter, CD34 and CD123 positivity, and uniform and increased CD38 expression. Flow cytometry is an excellent tool for identifying AML-MC cases.
Collapse
Affiliation(s)
- Jie Xu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Do Hwan Kim
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Wei Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Shaoying Li
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Pei Lin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Sergej Konoplev
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Lianqun Qiu
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Hong Fang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Sofia Garces
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Vasiliki Leventaki
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Shuyu E
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| | - Sa A Wang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, US
| |
Collapse
|
2
|
Degenfeld-Schonburg L, Sadovnik I, Smiljkovic D, Peter B, Stefanzl G, Gstoettner C, Jaksch P, Hoetzenecker K, Aigner C, Radtke C, Arock M, Sperr WR, Valent P. Coronavirus Receptor Expression Profiles in Human Mast Cells, Basophils, and Eosinophils. Cells 2024; 13:173. [PMID: 38247864 PMCID: PMC10814915 DOI: 10.3390/cells13020173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/04/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
A major problem in SARS-CoV-2-infected patients is the massive tissue inflammation in certain target organs, including the lungs. Mast cells (MC), basophils (BA), and eosinophils (EO) are key effector cells in inflammatory processes. These cells have recently been implicated in the pathogenesis of SARS-CoV-2 infections. We explored coronavirus receptor (CoV-R) expression profiles in primary human MC, BA, and EO, and in related cell lines (HMC-1, ROSA, MCPV-1, KU812, and EOL-1). As determined using flow cytometry, primary MC, BA, and EO, and their corresponding cell lines, displayed the CoV-R CD13 and CD147. Primary skin MC and BA, as well as EOL-1 cells, also displayed CD26, whereas primary EO and the MC and BA cell lines failed to express CD26. As assessed using qPCR, most cell lines expressed transcripts for CD13, CD147, and ABL2, whereas ACE2 mRNA was not detectable, and CD26 mRNA was only identified in EOL-1 cells. We also screened for drug effects on CoV-R expression. However, dexamethasone, vitamin D, and hydroxychloroquine did not exert substantial effects on the expression of CD13, CD26, or CD147 in the cells. Together, MC, BA, and EO express distinct CoV-R profiles. Whether these receptors mediate virus-cell interactions and thereby virus-induced inflammation remains unknown at present.
Collapse
Affiliation(s)
- Lina Degenfeld-Schonburg
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Irina Sadovnik
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Dubravka Smiljkovic
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Barbara Peter
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gabriele Stefanzl
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Clemens Gstoettner
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Jaksch
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria (C.A.)
| | - Konrad Hoetzenecker
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria (C.A.)
| | - Clemens Aigner
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria (C.A.)
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Michel Arock
- Laboratory of Hematology, Pitié-Salpêtrière Hospital, 75651 Paris, France;
| | - Wolfgang R. Sperr
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria; (L.D.-S.)
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
3
|
Bakhashab S, Banafea GH, Ahmed F, Alsehli H, AlShaibi HF, Bagatian N, Subhi O, Gauthaman K, Rasool M, Schulten HJ, Pushparaj PN. Characterization of human umbilical cord blood-derived mast cells using high-throughput expression profiling and next-generation knowledge discovery platforms. Exp Mol Pathol 2023; 132-133:104867. [PMID: 37634863 DOI: 10.1016/j.yexmp.2023.104867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/09/2023] [Accepted: 08/14/2023] [Indexed: 08/29/2023]
Abstract
Mast cells (MCs) are tissue-resident innate immune cells that express the high-affinity receptor for immunoglobulin E and are responsible for host defense and an array of diseases related to immune system. We aimed in this study to characterize the pathways and gene signatures of human cord blood-derived MCs (hCBMCs) in comparison to cells originating from CD34- progenitors using next-generation knowledge discovery methods. CD34+ cells were isolated from human umbilical cord blood using magnetic activated cell sorting and differentiated into MCs with rhIL-6 and rhSCF supplementation for 6-8 weeks. The purity of hCBMCs was analyzed by flow cytometry exhibiting the surface markers CD117+CD34-CD45-CD23-FcεR1αdim. Total RNA from hCBMCs and CD34- cells were isolated and hybridized using microarray. Differentially expressed genes were analyzed using iPathway Guide and Pre-Ranked Gene Set Enrichment Analysis. Next-generation knowledge discovery platforms revealed MC-specific gene signatures and molecular pathways enriched in hCBMCs and pertain the immunological response repertoire.
Collapse
Affiliation(s)
- Sherin Bakhashab
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia; Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Ghalya H Banafea
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Farid Ahmed
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Haneen Alsehli
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Centre for Stem Cells & Regenerative Medicine, King's College London, UK
| | - Huda F AlShaibi
- Biochemistry Department, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nadia Bagatian
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ohoud Subhi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kalamegam Gauthaman
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hans-Juergen Schulten
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia; Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia; Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| |
Collapse
|
4
|
Expression of CD117 (c-Kit) on Circulating B Cells in Pediatric Schistosomiasis. Infect Immun 2022; 90:e0016022. [PMID: 35862720 PMCID: PMC9387214 DOI: 10.1128/iai.00160-22] [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: 01/09/2023] Open
Abstract
Few B cells express CD27, the primary marker for memory B cells, in pediatric schistosomiasis, suggesting B cell malfunction. This study further demonstrates unexpected high expression of CD117 on circulating B cells in children highly exposed to Schistosoma mansoni infectious larvae. CD117 is expressed by immature or lymphoma B cells, but not by mature, circulating cells. We therefore sought to define the significance of CD117 on blood B cells. We found that CD117-positive (CD117+) B cells increased with the intensity of schistosome infection. In addition, CD117 expression was reduced on CD23+ B cells previously shown to correlate with resistance to infection. Stimulation with a panel of cytokines demonstrated that CD117 levels were upregulated in response to a combination of interleukin 4 (IL-4) and stem cell factor (SCF), the ligand for CD117, whereas IL-2 led to a reduction. In addition, stimulation with SCF generally reduced B cell activation levels. Upon further investigation, it was established that multiple circulating cells expressed increased levels of CD117, including monocytes, neutrophils, and eosinophils, and expression levels correlated with that of B cells. Finally, we identified a population of large circulating cells with features of reticulocytes. Overall, our results suggest that hyperexposure to intravascular parasitic worms elicits immature cells from the bone marrow. Levels of SCF were shown to reduce as children began to transition through puberty. The study results pose an explanation for the inability of children to develop significant immunity to infection until after puberty.
Collapse
|
5
|
Flow-based allergen testing: can mast cells beat basophils? Clin Chim Acta 2022; 532:64-71. [DOI: 10.1016/j.cca.2022.05.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/17/2022] [Accepted: 05/18/2022] [Indexed: 12/15/2022]
|
6
|
Berger D, Bauer K, Kornauth C, Gamperl S, Stefanzl G, Smiljkovic D, Sillaber C, Bettelheim P, Knöbl P, Schiefer AI, Greiner G, Thalhammer R, Hoermann G, Schwarzinger I, Staber PB, Sperr WR, Valent P. Secondary basophilic leukemia in Ph-negative myeloid neoplasms: A distinct subset with poor prognosis. Neoplasia 2021; 23:1183-1191. [PMID: 34731787 PMCID: PMC8572856 DOI: 10.1016/j.neo.2021.09.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 11/09/2022] Open
Abstract
During progression of myeloid neoplasms, the basophil compartment may expand substantially and in some of these patients, a basophilic leukemia is diagnosed. In patients with Ph-chromosome+ chronic myeloid leukemia, acceleration of disease is typically accompanied by marked basophilia. In other myeloid neoplasms, secondary leukemic expansion of basophils is rarely seen. We report on 5 patients who suffered from a myelodysplastic syndrome, myeloproliferative neoplasm, or acute leukemia and developed a massive expansion of basophils during disease progression. In 4 of 5 patients, peripheral blood basophil counts reached 40%, and the diagnosis “secondary basophilic leukemia” was established. As assessed by flow cytometry, neoplastic basophils expressed CD9, CD18, CD25, CD33, CD63, PD-L1, CD123, and CLL-1. In addition, basophils were found to display BB1 (basogranulin), 2D7, tryptase and KIT. In 4 of 5 patients the disease progressed quickly and treatment with azacitidine was started. However, azacitidine did not induce major clinical responses, and all patients died from progressive disease within 3 Y. In in vitro experiments, the patients´ cells and the basophilic leukemia cell line KU812 showed variable responses to targeted drugs, including azacitidine, venetoclax, hydroxyurea, and cytarabine. A combination of venetoclax and azacitidine induced cooperative antineoplastic effects in these cells. Together, secondary basophilic leukemia has a poor prognosis and monotherapy with azacitidine is not sufficient to keep the disease under control for longer time-periods. Whether drug combination, such as venetoclax+azacitidine, can induce better outcomes in these patients remains to be determined in future clinical studies.
Collapse
Affiliation(s)
- Daniela Berger
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Karin Bauer
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Hematology and Oncology (LBI HO), Medical University of Vienna, Vienna, Austria
| | - Christoph Kornauth
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Hematology and Oncology (LBI HO), Medical University of Vienna, Vienna, Austria; Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Susanne Gamperl
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Gabriele Stefanzl
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Dubravka Smiljkovic
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Christian Sillaber
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Peter Bettelheim
- Division of Hematology and Oncology, Elisabethinen Hospital Linz and Europa-Platz Labor Linz, Linz, Austria
| | - Paul Knöbl
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Ana-Iris Schiefer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Georg Greiner
- Ludwig Boltzmann Institute for Hematology and Oncology (LBI HO), Medical University of Vienna, Vienna, Austria; Ihr Labor, Medical Diagnostic Laboratories, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Renate Thalhammer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Gregor Hoermann
- Ludwig Boltzmann Institute for Hematology and Oncology (LBI HO), Medical University of Vienna, Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria; Munich Leukemia Laboratory (MLL), Munich, Germany
| | - Ilse Schwarzinger
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Philipp B Staber
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang R Sperr
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
7
|
Yin Y, Bai Y, Olivera A, Metcalfe DD. Demonstration and implications of IL-3 upregulation of CD25 expression on human mast cells. J Allergy Clin Immunol 2021; 149:1392-1401.e6. [PMID: 34506850 DOI: 10.1016/j.jaci.2021.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 08/11/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND CD25+ human mast cells (huMCs) have been reported in patients with monoclonal mast cell diseases and in rare association with inflammation. However, the regulation of CD25 expression on huMCs and the possible biologic consequences remain poorly understood. OBJECTIVE We sought to identify conditions that would upregulate CD25 expression on huMCs and to explore possible functional implications. METHODS huMCs were cultured from peripheral blood progenitor cells over 6 to 8 weeks. Expression of CD25 was determined by fluorescence-activated cell sorting and soluble CD25 by ELISA. Signal transducer and activator of transcription 5 (STAT5) phosphorylation induced by IL-2 in huMCs, regulatory T (Treg) cells, or in cocultured huMCs and Treg cells was examined by fluorescence-activated cell sorting. RESULTS Addition of IL-3 to CD34+ progenitors at the initiation of huMC cultures in the presence of stem cell factor and IL-6 upregulated the expression of CD25 in developing huMCs and resulted in shedding of soluble CD25 into the media. Removal of IL-3 after the first week of culture did not affect subsequent expression of CD25. Furthermore, addition of IL-3 14 days after the initiation of the culture did not induce significant CD25 expression. Treatment with anti-IL-3 antibody or the Janus kinase inhibitor tofacitinib blocked IL-3-induced CD25 upregulation. Binding of IL-2 to CD25+ huMCs did not induce STAT5 phosphorylation. However, coincubation of Treg cells with CD25+ huMCs pretreated with IL-2 was sufficient to result in STAT5 phosphorylation in Treg cells. CONCLUSIONS IL-3 promotes CD25 expression and shedding by huMCs. Although CD25+ huMCs do not respond to IL-2, they bind IL-2 and may act as a reservoir of IL-2 to then activate lymphocytes.
Collapse
Affiliation(s)
- Yuzhi Yin
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| | - Yun Bai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Ana Olivera
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Dean D Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| |
Collapse
|
8
|
Jiménez M, Cervantes-García D, Córdova-Dávalos LE, Pérez-Rodríguez MJ, Gonzalez-Espinosa C, Salinas E. Responses of Mast Cells to Pathogens: Beneficial and Detrimental Roles. Front Immunol 2021; 12:685865. [PMID: 34211473 PMCID: PMC8240065 DOI: 10.3389/fimmu.2021.685865] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/28/2021] [Indexed: 12/19/2022] Open
Abstract
Mast cells (MCs) are strategically located in tissues close to the external environment, being one of the first immune cells to interact with invading pathogens. They are long living effector cells equipped with different receptors that allow microbial recognition. Once activated, MCs release numerous biologically active mediators in the site of pathogen contact, which induce vascular endothelium modification, inflammation development and extracellular matrix remodeling. Efficient and direct antimicrobial mechanisms of MCs involve phagocytosis with oxidative and non-oxidative microbial destruction, extracellular trap formation, and the release of antimicrobial substances. MCs also contribute to host defense through the attraction and activation of phagocytic and inflammatory cells, shaping the innate and adaptive immune responses. However, as part of their response to pathogens and under an impaired, sustained, or systemic activation, MCs may contribute to tissue damage. This review will focus on the current knowledge about direct and indirect contribution of MCs to pathogen clearance. Antimicrobial mechanisms of MCs are addressed with special attention to signaling pathways involved and molecular weapons implicated. The role of MCs in a dysregulated host response that can increase morbidity and mortality is also reviewed and discussed, highlighting the complexity of MCs biology in the context of host-pathogen interactions.
Collapse
Affiliation(s)
- Mariela Jiménez
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Daniel Cervantes-García
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico.,Cátedras CONACYT, National Council of Science and Technology, Mexico City, Mexico
| | - Laura E Córdova-Dávalos
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| | - Marian Jesabel Pérez-Rodríguez
- Department of Pharmacobiology, Centro de Investigación y de Estudios Avanzados (Cinvestav), Unidad Sede Sur, Mexico City, Mexico
| | - Claudia Gonzalez-Espinosa
- Department of Pharmacobiology, Centro de Investigación y de Estudios Avanzados (Cinvestav), Unidad Sede Sur, Mexico City, Mexico
| | - Eva Salinas
- Laboratory of Immunology, Department of Microbiology, Universidad Autónoma de Aguascalientes, Aguascalientes, Mexico
| |
Collapse
|
9
|
Sprinzl B, Greiner G, Uyanik G, Arock M, Haferlach T, Sperr WR, Valent P, Hoermann G. Genetic Regulation of Tryptase Production and Clinical Impact: Hereditary Alpha Tryptasemia, Mastocytosis and Beyond. Int J Mol Sci 2021; 22:2458. [PMID: 33671092 PMCID: PMC7957558 DOI: 10.3390/ijms22052458] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 12/12/2022] Open
Abstract
Tryptase is a serine protease that is predominantly produced by tissue mast cells (MCs) and stored in secretory granules together with other pre-formed mediators. MC activation, degranulation and mediator release contribute to various immunological processes, but also to several specific diseases, such as IgE-dependent allergies and clonal MC disorders. Biologically active tryptase tetramers primarily derive from the two genes TPSB2 (encoding β-tryptase) and TPSAB1 (encoding either α- or β-tryptase). Based on the most common gene copy numbers, three genotypes, 0α:4β, 1α:3β and 2α:2β, were defined as "canonical". About 4-6% of the general population carry germline TPSAB1-α copy number gains (2α:3β, 3α:2β or more α-extra-copies), resulting in elevated basal serum tryptase levels. This condition has recently been termed hereditary alpha tryptasemia (HαT). Although many carriers of HαT appear to be asymptomatic, a number of more or less specific symptoms have been associated with HαT. Recent studies have revealed a significantly higher HαT prevalence in patients with systemic mastocytosis (SM) and an association with concomitant severe Hymenoptera venom-induced anaphylaxis. Moreover, HαT seems to be more common in idiopathic anaphylaxis and MC activation syndromes (MCAS). Therefore, TPSAB1 genotyping should be included in the diagnostic algorithm in patients with symptomatic SM, severe anaphylaxis or MCAS.
Collapse
Affiliation(s)
- Bettina Sprinzl
- Ludwig Boltzmann Institute for Hematology and Oncology at the Hanusch Hospital, Center for Medical Genetics, Hanusch Hospital, 1140 Vienna, Austria; (B.S.); (G.U.)
- Center for Medical Genetics, Hanusch Hospital, 1140 Vienna, Austria
| | - Georg Greiner
- Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria;
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria; (W.R.S.); (P.V.)
- Ihr Labor, Medical Diagnostic Laboratories, 1220 Vienna, Austria
| | - Goekhan Uyanik
- Ludwig Boltzmann Institute for Hematology and Oncology at the Hanusch Hospital, Center for Medical Genetics, Hanusch Hospital, 1140 Vienna, Austria; (B.S.); (G.U.)
- Center for Medical Genetics, Hanusch Hospital, 1140 Vienna, Austria
- Medical School, Sigmund Freud Private University, 1020 Vienna, Austria
| | - Michel Arock
- Department of Hematology, APHP, Pitié-Salpêtrière-Charles Foix University Hospital and Sorbonne University, 75013 Paris, France;
- Centre de Recherche des Cordeliers, INSERM, Sorbonne University, Cell Death and Drug Resistance in Hematological Disorders Team, 75006 Paris, France
| | | | - Wolfgang R. Sperr
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria; (W.R.S.); (P.V.)
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Valent
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria; (W.R.S.); (P.V.)
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gregor Hoermann
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, 1090 Vienna, Austria; (W.R.S.); (P.V.)
- MLL Munich Leukemia Laboratory, 81377 Munich, Germany;
| |
Collapse
|
10
|
Valent P, Akin C, Hartmann K, Nilsson G, Reiter A, Hermine O, Sotlar K, Sperr WR, Escribano L, George TI, Kluin-Nelemans HC, Ustun C, Triggiani M, Brockow K, Gotlib J, Orfao A, Kovanen PT, Hadzijusufovic E, Sadovnik I, Horny HP, Arock M, Schwartz LB, Austen KF, Metcalfe DD, Galli SJ. Mast cells as a unique hematopoietic lineage and cell system: From Paul Ehrlich's visions to precision medicine concepts. Am J Cancer Res 2020; 10:10743-10768. [PMID: 32929378 PMCID: PMC7482799 DOI: 10.7150/thno.46719] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
The origin and functions of mast cells (MCs) have been debated since their description by Paul Ehrlich in 1879. MCs have long been considered 'reactive bystanders' and 'amplifiers' in inflammatory processes, allergic reactions, and host responses to infectious diseases. However, knowledge about the origin, phenotypes and functions of MCs has increased substantially over the past 50 years. MCs are now known to be derived from multipotent hematopoietic progenitors, which, through a process of differentiation and maturation, form a unique hematopoietic lineage residing in multiple organs. In particular, MCs are distinguishable from basophils and other hematopoietic cells by their unique phenotype, origin(s), and spectrum of functions, both in innate and adaptive immune responses and in other settings. The concept of a unique MC lineage is further supported by the development of a distinct group of neoplasms, collectively referred to as mastocytosis, in which MC precursors expand as clonal cells. The clinical consequences of the expansion and/or activation of MCs are best established in mastocytosis and in allergic inflammation. However, MCs have also been implicated as important participants in a number of additional pathologic conditions and physiological processes. In this article, we review concepts regarding MC development, factors controlling MC expansion and activation, and some of the fundamental roles MCs may play in both health and disease. We also discuss new concepts for suppressing MC expansion and/or activation using molecularly-targeted drugs.
Collapse
|
11
|
Identification and Immunophenotypic Characterization of Normal and Pathological Mast Cells. Methods Mol Biol 2020; 2163:331-353. [PMID: 32766988 DOI: 10.1007/978-1-0716-0696-4_27] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Mast cells (MCs) are secretory cells that are central players in human allergic disease and immune responses. With the exception of a few pathological situations, MCs are usually present at relatively low frequencies in most tissues. Since their first description, MCs in tissues were identified mostly using their morphological characteristics and their typical coloration when stained with aniline dyes. However, increasing availability of highly specific antibodies now permits the use of fluorescence-based flow cytometry as the method of choice for the quantification, characterization, and purification of cells in suspension. This technique allows for a rapid analysis of thousands of events and for the identification of cells present at frequencies as low as one event in 106 unwanted cells. This method also permits for simultaneous characterization of multiple antigens at a single cell level, which is ideal in order to study rare populations of cells like MCs. Here we describe the basis of flow cytometry-based immunophenotyping applied to the study of MC. The protocol focuses on the study of human MCs present in body fluids (mainly bone marrow) but can easily be adapted to studying MCs from other tissues and species.
Collapse
|
12
|
Deciphering the differentiation trajectory from hematopoietic stem cells to mast cells. Blood Adv 2019; 2:2273-2281. [PMID: 30206100 DOI: 10.1182/bloodadvances.2018019539] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 08/18/2018] [Indexed: 01/22/2023] Open
Abstract
Hematopoietic stem cells differentiate into all types of blood cells, including peripheral tissue-resident mast cells. The early mast cell differentiation takes place in the bone marrow, after which the progenitor cells enter the circulation and mature once reaching their target organ. Early results from single-cell culture experiments and colony-forming assays have produced the classic hierarchical tree model of hematopoiesis. The introduction of high-throughput, single-cell RNA sequencing is now revolutionizing our understanding of the differentiation process, questioning the classic tree-based models. By integrating the results from early cell culture experiments with single-cell transcriptomics, we present a differentiation landscape model of hematopoiesis and discuss it with focus on mast cells. The review also describes how the hematologic neoplasm systemic mastocytosis can be used to model human hematopoiesis using naturally occurring cell barcoding by means of the common KIT D816V mutation.
Collapse
|
13
|
Abstract
Mast cell disorders comprise a heterogeneous group of rare diseases, the diagnosis of which still remains a challenge. Bone marrow analysis constitutes the most appropriate site for screening systemic involvement in mastocytosis. Morphologic, immunohistochemical, flow cytometric immunophenotyping, and molecular studies should be routinely performed for diagnostic/prognostic purposes in experienced reference centers during the diagnostic workup in suspected systemic mastocytosis. The authors review the most relevant characteristics of bone marrow expression of mast cell disorders as well as the different methodological approaches to be applied to perform an objective and reproducible diagnosis and classification of mastocytosis and other mast cell disorders.
Collapse
|
14
|
Mayado A, Teodosio C, Dasilva‐Freire N, Jara‐Acevedo M, Garcia‐Montero AC, Álvarez‐Twose I, Sánchez‐Muñoz L, Matito A, Caldas C, Muñoz‐González JI, Henriques A, Sánchez‐Gallego JI, Escribano L, Orfao A. Characterization of CD34 + hematopoietic cells in systemic mastocytosis: Potential role in disease dissemination. Allergy 2018; 73:1294-1304. [PMID: 29331029 DOI: 10.1111/all.13413] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Recent studies show that most systemic mastocytosis (SM) patients, including indolent SM (ISM) with (ISMs+) and without skin lesions (ISMs-), carry the KIT D816V mutation in PB leukocytes. We investigated the potential association between the degree of involvement of BM hematopoiesis by the KIT D816V mutation and the distribution of different maturation-associated compartments of bone marrow (BM) and peripheral blood (PB) CD34+ hematopoietic precursors (HPC) in ISM and identified the specific PB cell compartments that carry this mutation. METHODS The distribution of different maturation-associated subsets of BM and PB CD34+ HPC from 64 newly diagnosed (KIT-mutated) ISM patients and 14 healthy controls was analyzed by flow cytometry. In 18 patients, distinct FACS-purified PB cell compartments were also investigated for the KIT mutation. RESULTS ISM patients showed higher percentages of both BM and PB MC-committed CD34+ HPC vs controls, particularly among ISM cases with MC-restricted KIT mutation (ISMMC ); this was associated with progressive blockade of maturation of CD34+ HPC to the neutrophil lineage from ISMMC to multilineage KIT-mutated cases (ISMML ). Regarding the frequency of KIT-mutated cases and cell populations in PB, variable patterns were observed, the percentage of KIT-mutated PB CD34+ HPC, eosinophils, neutrophils, monocytes and T cells increasing from ISMs-MC and ISMs+MC to ISMML patients. CONCLUSION The presence of the KIT D816V mutation in PB of ISM patients is associated with (early) involvement of circulating CD34+ HPC and multiple myeloid cell subpopulations, KIT-mutated PB CD34+ HPC potentially contributing to early dissemination of the disease.
Collapse
Affiliation(s)
- A. Mayado
- Cancer Research Centre (IBMCC USAL‐CSIC) Cytometry Service (NUCLEUS) and Department of Medicine University of Salamanca Salamanca Spain
- Institute of Biomedical Research of Salamanca (IBSAL) Salamanca Spain
- Biomedical Research Networking Centre Consortium–CIBER‐CIBERONC of the Institute of Health Carlos III Madrid Spain
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
| | - C. Teodosio
- Department of Immunohematology and Blood Transfusion Leiden University Medical Center Leiden The Netherlands
| | - N. Dasilva‐Freire
- Cancer Research Centre (IBMCC USAL‐CSIC) Cytometry Service (NUCLEUS) and Department of Medicine University of Salamanca Salamanca Spain
- Institute of Biomedical Research of Salamanca (IBSAL) Salamanca Spain
- Biomedical Research Networking Centre Consortium–CIBER‐CIBERONC of the Institute of Health Carlos III Madrid Spain
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
| | - M. Jara‐Acevedo
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
- Sequencing DNA Service (NUCLEUS) University of Salamanca Salamanca Spain
| | - A. C. Garcia‐Montero
- Cancer Research Centre (IBMCC USAL‐CSIC) Cytometry Service (NUCLEUS) and Department of Medicine University of Salamanca Salamanca Spain
- Institute of Biomedical Research of Salamanca (IBSAL) Salamanca Spain
- Biomedical Research Networking Centre Consortium–CIBER‐CIBERONC of the Institute of Health Carlos III Madrid Spain
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
| | - I. Álvarez‐Twose
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) Hospital Virgen del Valle Toledo Spain
| | - L. Sánchez‐Muñoz
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) Hospital Virgen del Valle Toledo Spain
| | - A. Matito
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) Hospital Virgen del Valle Toledo Spain
| | - C. Caldas
- Cancer Research Centre (IBMCC USAL‐CSIC) Cytometry Service (NUCLEUS) and Department of Medicine University of Salamanca Salamanca Spain
- Institute of Biomedical Research of Salamanca (IBSAL) Salamanca Spain
- Biomedical Research Networking Centre Consortium–CIBER‐CIBERONC of the Institute of Health Carlos III Madrid Spain
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
| | - J. I. Muñoz‐González
- Cancer Research Centre (IBMCC USAL‐CSIC) Cytometry Service (NUCLEUS) and Department of Medicine University of Salamanca Salamanca Spain
- Institute of Biomedical Research of Salamanca (IBSAL) Salamanca Spain
- Biomedical Research Networking Centre Consortium–CIBER‐CIBERONC of the Institute of Health Carlos III Madrid Spain
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
| | - A. Henriques
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
- Instituto de Estudios de Mastocitosis de Castilla La Mancha (CLMast) Hospital Virgen del Valle Toledo Spain
| | - J. I. Sánchez‐Gallego
- Cancer Research Centre (IBMCC USAL‐CSIC) Cytometry Service (NUCLEUS) and Department of Medicine University of Salamanca Salamanca Spain
- Institute of Biomedical Research of Salamanca (IBSAL) Salamanca Spain
- Biomedical Research Networking Centre Consortium–CIBER‐CIBERONC of the Institute of Health Carlos III Madrid Spain
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
| | - L. Escribano
- Cancer Research Centre (IBMCC USAL‐CSIC) Cytometry Service (NUCLEUS) and Department of Medicine University of Salamanca Salamanca Spain
- Institute of Biomedical Research of Salamanca (IBSAL) Salamanca Spain
- Biomedical Research Networking Centre Consortium–CIBER‐CIBERONC of the Institute of Health Carlos III Madrid Spain
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
| | - A. Orfao
- Cancer Research Centre (IBMCC USAL‐CSIC) Cytometry Service (NUCLEUS) and Department of Medicine University of Salamanca Salamanca Spain
- Institute of Biomedical Research of Salamanca (IBSAL) Salamanca Spain
- Biomedical Research Networking Centre Consortium–CIBER‐CIBERONC of the Institute of Health Carlos III Madrid Spain
- Spanish Network on Mastocytosis (REMA) Toledo Salamanca Spain
| |
Collapse
|
15
|
Abstract
Mast cells are present at a low frequency in bone marrow, rendering high-sensitivity multiparametric flow cytometric analysis an ideal method to assess antigen expression on mast cells. This article discusses the normal antigen expression profile of mast cells, established criteria to identify neoplastic mast cells, and new immunophenotypic markers and approaches to identify the presence of neoplastic mast cells in cases of mastocytosis.
Collapse
Affiliation(s)
- Jacqueline M Cortazar
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA
| | - David M Dorfman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Klein O, Ngo-Nyekel F, Stefanache T, Torres R, Salomonsson M, Hallgren J, Rådinger M, Bambouskova M, Campbell M, Cohen-Mor S, Dema B, Rose CG, Abrink M, Charles N, Ainooson G, Paivandy A, Pavlova VG, Serrano-Candelas E, Yu Y, Hellman L, Jensen BM, Van Anrooij B, Grootens J, Gura HK, Stylianou M, Tobio A, Blank U, Öhrvik H, Maurer M. Identification of Biological and Pharmaceutical Mast Cell- and Basophil-Related Targets. Scand J Immunol 2017; 83:465-72. [PMID: 27028428 DOI: 10.1111/sji.12436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/27/2016] [Indexed: 01/09/2023]
Affiliation(s)
- O Klein
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - F Ngo-Nyekel
- Inserm UMRS-1149, Paris, France.,CNRS ERL 8252, Paris, France.,Sorbonne Paris Cite, Laboratoire d'excellence INFLAMEX, Université Paris Diderot, Paris, France
| | - T Stefanache
- Department of Periodontology, University of Medicine and Pharmacy 'Gr. T. Popa', Iasi, Romania
| | - R Torres
- Safety and Sustainability Division, Leitat Technological Center, Barcelona, Spain
| | - M Salomonsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - J Hallgren
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - M Rådinger
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - M Bambouskova
- Department of Signal Transduction, Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - M Campbell
- Institute of Inflammation and Repair and MCCIR, University of Manchester, Manchester, UK
| | - S Cohen-Mor
- The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - B Dema
- Inserm UMRS-1149, Paris, France.,CNRS ERL 8252, Paris, France.,Sorbonne Paris Cite, Laboratoire d'excellence INFLAMEX, Université Paris Diderot, Paris, France
| | - C G Rose
- Bioengineering, Faculty of Engineering and the Environment, University of Southampton, Southampton, UK.,Immunopharmacology Group, Clinical Experimental Sciences, Faculty of Medicine, Southampton General Hospital, University of Southampton, Southampton, UK
| | - M Abrink
- Section of Immunology, Department of Biomedical Sciences and Veterinary Public Health, VHC, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - N Charles
- Inserm UMRS-1149, Paris, France.,CNRS ERL 8252, Paris, France.,Sorbonne Paris Cite, Laboratoire d'excellence INFLAMEX, Université Paris Diderot, Paris, France
| | - G Ainooson
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology, Karlsruhe, Germany
| | - A Paivandy
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - V G Pavlova
- Department of Experimental Morphology, Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - E Serrano-Candelas
- Biochemistry Unit, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Y Yu
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, the Netherlands
| | - L Hellman
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - B M Jensen
- Allergy Clinic, Copenhagen University Hospital - Gentofte Hospital, Hellerup, Denmark
| | - B Van Anrooij
- Department of Allergology, Groningen Research Institute of Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - J Grootens
- Clinical Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - H K Gura
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital and Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - M Stylianou
- Antifungal Immunity Group, Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - A Tobio
- Inserm UMRS-1149, Paris, France.,CNRS ERL 8252, Paris, France.,Sorbonne Paris Cite, Laboratoire d'excellence INFLAMEX, Université Paris Diderot, Paris, France
| | - U Blank
- Inserm UMRS-1149, Paris, France.,CNRS ERL 8252, Paris, France.,Sorbonne Paris Cite, Laboratoire d'excellence INFLAMEX, Université Paris Diderot, Paris, France
| | - H Öhrvik
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - M Maurer
- Department of Dermatology and Allergy, Allergie-Centrum-Charité, Charité -Universitätsmedizin, Berlin, Germany
| |
Collapse
|
17
|
Mayerhofer M, Aichberger KJ, Florian S, Valent P. Recognition Sites for Microbes and Components of the Immune System on Human Mast Cells: Relationship to CD Antigens and Implications for Host Defense. Int J Immunopathol Pharmacol 2016; 20:421-34. [PMID: 17880756 DOI: 10.1177/039463200702000301] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Traditionally, mast cells (MCs) have been considered to play an important role in allergic disorders and helminth infections. More recently, MCs have been implicated in a variety of different infectious diseases including life-threatening disorders caused by viruses and bacteria. Apart from recognition through specific IgE, MCs are considered to recognize such bacteria and viruses via specific cell surface binding sites. In addition, MCs interact with diverse components and cells of the immune system and thereby may facilitate the targeting and the elimination of invading microbes in the tissues. The current article provides an overview on MC antigens contributing to microbe recognition and targeting as an important element of natural host-defense.
Collapse
Affiliation(s)
- M Mayerhofer
- Division of Hematology & Hemostaseology, Medical University of Vienna, Vienna, Austria
| | | | | | | |
Collapse
|
18
|
Zotz JS, Wölbing F, Lassnig C, Kauffmann M, Schulte U, Kolb A, Whitelaw B, Müller M, Biedermann T, Huber M. CD13/aminopeptidase N is a negative regulator of mast cell activation. FASEB J 2016; 30:2225-35. [PMID: 26936360 DOI: 10.1096/fj.201600278] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 02/11/2016] [Indexed: 12/20/2022]
Abstract
Antigen-induced mast cell (MC) activation via cross-linking of IgE-bound high-affinity receptors for IgE (FcεRI) underlies type I allergy and anaphylactic shock. Comprehensive knowledge of FcεRI regulation is thus required. We have identified a functional interaction between FcεRI and CD13 in murine MCs. Antigen-triggered activation of IgE-loaded FcεRI results in cocapping and cointernalization of CD13 and equivalent internalization rates of up to 40%. Cointernalization is not unspecific, because ligand-driven KIT internalization is not accompanied by CD13 internalization. Moreover, antibody-mediated cross-linking of CD13 causes IL-6 production in an FcεRI-dependent manner. These data are indicative of a functional interaction between FcεRI and CD13 on MCs. To determine the role of this interaction, CD13-deficient bone marrow-derived MCs (BMMCs) were analyzed. Intriguingly, antigen stimulation of CD13-deficient BMMCs results in significantly increased degranulation and proinflammatory cytokine production compared to wild-type cells. Furthermore, in a low-dose model of passive systemic anaphylaxis, antigen-dependent decrease in body temperature, reflecting the anaphylactic reaction, is substantially enhanced by the CD13 inhibitor bestatin (-5.9 ± 0.6°C) and by CD13 deficiency (-8.8 ± 0.6°C) in contrast to controls (-1.2 ± 1.97°C). Importantly, bestatin does not aggravate anaphylaxis in CD13-deficient mice. Thus, we have identified CD13 as a novel negative regulator of MC activation in vitro and in vivo-Zotz, J. S., Wölbing, F., Lassnig, C., Kauffmann, M., Schulte, U., Kolb, A., Whitelaw, B., Müller, M., Biedermann, T., Huber, M. CD13/aminopeptidase N is a negative regulator of mast cell activation.
Collapse
Affiliation(s)
- Julia S Zotz
- Institute of Biochemistry and Molecular Immunology, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Florian Wölbing
- Department of Dermatology, Technical University of Munich, Munich, Germany
| | - Caroline Lassnig
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Marlies Kauffmann
- Institute of Biochemistry and Molecular Immunology, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Uwe Schulte
- Institute of Physiology II, University of Freiburg, Freiburg, Germany; Centre for Biological Signalling Studies (Bioss), Freiburg, Germany; Logopharm GmbH, March-Buchheim, Germany
| | - Andreas Kolb
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, United Kingdom; and
| | - Bruce Whitelaw
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Tilo Biedermann
- Department of Dermatology, Technical University of Munich, Munich, Germany
| | - Michael Huber
- Institute of Biochemistry and Molecular Immunology, University Hospital, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany;
| |
Collapse
|
19
|
Metcalfe DD, Pawankar R, Ackerman SJ, Akin C, Clayton F, Falcone FH, Gleich GJ, Irani AM, Johansson MW, Klion AD, Leiferman KM, Levi-Schaffer F, Nilsson G, Okayama Y, Prussin C, Schroeder JT, Schwartz LB, Simon HU, Walls AF, Triggiani M. Biomarkers of the involvement of mast cells, basophils and eosinophils in asthma and allergic diseases. World Allergy Organ J 2016; 9:7. [PMID: 26904159 PMCID: PMC4751725 DOI: 10.1186/s40413-016-0094-3] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/14/2016] [Indexed: 12/19/2022] Open
Abstract
Biomarkers of disease activity have come into wide use in the study of mechanisms of human disease and in clinical medicine to both diagnose and predict disease course; as well as to monitor response to therapeutic intervention. Here we review biomarkers of the involvement of mast cells, basophils, and eosinophils in human allergic inflammation. Included are surface markers of cell activation as well as specific products of these inflammatory cells that implicate specific cell types in the inflammatory process and are of possible value in clinical research as well as within decisions made in the practice of allergy-immunology.
Collapse
Affiliation(s)
- Dean D. Metcalfe
- />Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - Ruby Pawankar
- />Division of Allergy, Department of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - Steven J. Ackerman
- />Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois, Chicago, IL USA
| | - Cem Akin
- />Harvard Medical School, Brigham and Women’s Hospital, Boston, MA USA
| | - Frederic Clayton
- />Department of Pathology, University of Utah Health Sciences Center, Salt Lake City, UT USA
| | - Franco H. Falcone
- />The School of Pharmacy, University of Nottingham, Nottingham, United Kingdom
| | - Gerald J. Gleich
- />Department of Dermatology, University of Utah, School of Medicine, Salt Lake City, UT USA
| | - Anne-Marie Irani
- />Virginia Commonwealth University, Children’s Hospital of Richmond, Richmond, VA USA
| | - Mats W. Johansson
- />Department of Biomolecular Chemistry, University of Wisconsin, Madison, WI USA
| | - Amy D. Klion
- />Human Eosinophil Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD USA
| | | | | | - Gunnar Nilsson
- />Clinical Immunology and Allergy, Department of Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Yoshimichi Okayama
- />Allergy and Immunology Group, Research Institute of Medical Science, Nihon University Graduate School of Medicine, Tokyo, Japan
| | - Calman Prussin
- />Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892 USA
| | - John T. Schroeder
- />Division of Allergy and Clinical Immunology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | | | - Hans-Uwe Simon
- />University of Bern, Institute of Pharmacology, Bern, Switzerland
| | - Andrew F. Walls
- />Southampton General Hospital, Immunopharmacology Group, Southampton, Hampshire UK
| | - Massimo Triggiani
- />Division of Allergy and Clinical Immunology, University of Salerno, Salerno, Italy
| |
Collapse
|
20
|
Teodosio C, Mayado A, Sánchez-Muñoz L, Morgado JM, Jara-Acevedo M, Álvarez-Twose I, García-Montero AC, Matito A, Caldas C, Escribano L, Orfao A. The immunophenotype of mast cells and its utility in the diagnostic work-up of systemic mastocytosis. J Leukoc Biol 2014; 97:49-59. [PMID: 25381388 DOI: 10.1189/jlb.5ru0614-296r] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
SM comprises a heterogeneous group of disorders, characterized by an abnormal accumulation of clonal MCs in 1 or more tissues, frequently involving the skin and BM. Despite the fact that most adult patients (>90%) carry the same genetic lesion (D816V KIT mutation), the disease presents with multiple variants with very distinct clinical and biologic features, a diverse prognosis, and different therapeutic requirements. Recent advances in the standardization of the study of BM MC by MFC allowed reproducible identification and characterization of normal/reactive MCs and their precursors, as well as the establishment of the normal MC maturational profiles. Analysis of large groups of patients versus normal/reactive samples has highlighted the existence of aberrant MC phenotypes in SM, which are essential for the diagnosis of the disease. In turn, 3 clearly distinct and altered maturation-associated immunophenotypic profiles have been reported recently in SM, which provide criteria for the distinction between ISM patients with MC-restricted and multilineage KIT mutation; thus, immunphenotyping also contributes to prognostic stratification of ISM, particularly when analysis of the KIT mutation on highly purified BM cells is not routinely available in the diagnostic work-up of the disease.
Collapse
Affiliation(s)
- Cristina Teodosio
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| | - Andrea Mayado
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| | - Laura Sánchez-Muñoz
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| | - José M Morgado
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| | - María Jara-Acevedo
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| | - Ivan Álvarez-Twose
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| | - Andrés C García-Montero
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| | - Almudena Matito
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| | - Caldas Caldas
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| | - Luis Escribano
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| | - Alberto Orfao
- *Servicio General de Citometría, Centro de Investigación del Cáncer (Instituto de Biologia Molecular y Celular del Cancer-Consejo Superior de Investigaciones Cientificas/University of Salamanca and Instituto de Investigación Biomédica de Salamanca) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain; and Centro de Estudios de Mastocitosis de Castilla La Mancha, Hospital Virgen del Valle, Toledo, Spain
| |
Collapse
|
21
|
Sánchez-Muñoz L, Teodosio C, Morgado JMT, Perbellini O, Mayado A, Alvarez-Twose I, Matito A, Jara-Acevedo M, García-Montero AC, Orfao A, Escribano L. Flow Cytometry in Mastocytosis. Immunol Allergy Clin North Am 2014; 34:297-313. [DOI: 10.1016/j.iac.2014.01.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
22
|
Servitzoglou M, Grenzelia M, Baka M, Harisi M, Pourtsidis A, Bouhoutsou D, Varvoutsi M, Doganis D, Dana H, Divane A, Kosmidis H. A novel karyotype in acute myeloid leukemia with basophilia. Pediatr Hematol Oncol 2014; 31:149-56. [PMID: 24552500 DOI: 10.3109/08880018.2014.883655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Acute basophilic leukemia is a distinct entity of Acute Myeloid Leukemia (AML) with primary differentiation to basophils. Increased basophil count has been described in AML cases with translocation t(6;9)(p23;q34) or other chromosomal abnormalities. We describe a 15-year old female teenager with AML and excess peripheral blood and bone marrow basophils. Her white blood cell count at diagnosis was 15.4 G/L with 53% basophils and 17% blasts. The bone marrow cytogenetics analysis did not reveal any of the usual abnormalities. The karyotype showed two closely related leukemic clones: the first (16 metaphases), with a total of 48 chromosomes, had an extra chromosome 8 with deletion of the long arm and an additional 21 (48,XX, +del(8)(q24.2q24.3), t21[16]), while the second clone (2 metaphases), with a total of 47 chromosomes, did not contain the extra 21 chromosome (47, sl, -21[2]). In summary, in this case of AML-M2 with excess basophils, there is a novel chromosomal abnormality, not previously reported in this entity.
Collapse
Affiliation(s)
- Marina Servitzoglou
- Oncology Department, Children's Hospital "P. & A. Kyriakou", Thivon and Levadias, Athens, Greece
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Morgado JM, Sánchez-Muñoz L, Teodósio C, Escribano L. Identification and immunophenotypic characterization of normal and pathological mast cells. Methods Mol Biol 2014; 1192:205-226. [PMID: 25149495 DOI: 10.1007/978-1-4939-1173-8_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Mast cells (MCs) are secretory cells that are central players in human allergic disease and immune responses. With the exception of a few pathological situations, MCs are usually present at relatively low frequencies in most tissues. Since their first description, MCs in tissues were identified mostly using their morphological characteristics and their typical coloration when stained with aniline dyes. However, increasing availability of highly specific antibodies now permits the use of fluorescence-based flow cytometry as the method of choice for the quantification, characterization, and purification of cells in suspension. This technique allows for a rapid analysis of thousands of events and for the identification of cells present at frequencies as low as one event in 10(6) unwanted cells. This method also permits for simultaneous characterization of multiple antigens at a single-cell level, which is ideal in order to study rare populations of cells like MCs. Here we describe the basis of flow cytometry-based immunophenotyping applied to the study of MC. The protocol focuses on the study of human MCs present in body fluids (mainly bone marrow) but can easily be adapted to study MCs from other tissues and species.
Collapse
Affiliation(s)
- José Mário Morgado
- Instituto de Estudios de Mastocitosis de Castilla La Mancha, Toledo, Spain
| | | | | | | |
Collapse
|
24
|
van Dongen JJM, Lhermitte L, Böttcher S, Almeida J, van der Velden VHJ, Flores-Montero J, Rawstron A, Asnafi V, Lécrevisse Q, Lucio P, Mejstrikova E, Szczepański T, Kalina T, de Tute R, Brüggemann M, Sedek L, Cullen M, Langerak AW, Mendonça A, Macintyre E, Martin-Ayuso M, Hrusak O, Vidriales MB, Orfao A. EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes. Leukemia 2012; 26:1908-75. [PMID: 22552007 PMCID: PMC3437410 DOI: 10.1038/leu.2012.120] [Citation(s) in RCA: 666] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 02/14/2012] [Accepted: 04/19/2012] [Indexed: 12/21/2022]
Abstract
Most consensus leukemia & lymphoma antibody panels consist of lists of markers based on expert opinions, but they have not been validated. Here we present the validated EuroFlow 8-color antibody panels for immunophenotyping of hematological malignancies. The single-tube screening panels and multi-tube classification panels fit into the EuroFlow diagnostic algorithm with entries defined by clinical and laboratory parameters. The panels were constructed in 2-7 sequential design-evaluation-redesign rounds, using novel Infinicyt software tools for multivariate data analysis. Two groups of markers are combined in each 8-color tube: (i) backbone markers to identify distinct cell populations in a sample, and (ii) markers for characterization of specific cell populations. In multi-tube panels, the backbone markers were optimally placed at the same fluorochrome position in every tube, to provide identical multidimensional localization of the target cell population(s). The characterization markers were positioned according to the diagnostic utility of the combined markers. Each proposed antibody combination was tested against reference databases of normal and malignant cells from healthy subjects and WHO-based disease entities, respectively. The EuroFlow studies resulted in validated and flexible 8-color antibody panels for multidimensional identification and characterization of normal and aberrant cells, optimally suited for immunophenotypic screening and classification of hematological malignancies.
Collapse
Affiliation(s)
- J J M van Dongen
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam (Erasmus MC), Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Kuonen F, Laurent J, Secondini C, Lorusso G, Stehle JC, Rausch T, Faes-Van't Hull E, Bieler G, Alghisi GC, Schwendener R, Andrejevic-Blant S, Mirimanoff RO, Rüegg C. Inhibition of the Kit ligand/c-Kit axis attenuates metastasis in a mouse model mimicking local breast cancer relapse after radiotherapy. Clin Cancer Res 2012; 18:4365-74. [PMID: 22711708 DOI: 10.1158/1078-0432.ccr-11-3028] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Local breast cancer relapse after breast-saving surgery and radiotherapy is associated with increased risk of distant metastasis formation. The mechanisms involved remain largely elusive. We used the well-characterized 4T1 syngeneic, orthotopic breast cancer model to identify novel mechanisms of postradiation metastasis. EXPERIMENTAL DESIGN 4T1 cells were injected in 20 Gy preirradiated mammary tissue to mimic postradiation relapses, or in nonirradiated mammary tissue, as control, of immunocompetent BALB/c mice. Molecular, biochemical, cellular, histologic analyses, adoptive cell transfer, genetic, and pharmacologic interventions were carried out. RESULTS Tumors growing in preirradiated mammary tissue had reduced angiogenesis and were more hypoxic, invasive, and metastatic to lung and lymph nodes compared with control tumors. Increased metastasis involved the mobilization of CD11b(+)c-Kit(+)Ly6G(high)Ly6C(low)(Gr1(+)) myeloid cells through the HIF1-dependent expression of Kit ligand (KitL) by hypoxic tumor cells. KitL-mobilized myeloid cells homed to primary tumors and premetastatic lungs, to give rise to CD11b(+)c-Kit(-) cells. Pharmacologic inhibition of HIF1, silencing of KitL expression in tumor cells, and inhibition of c-Kit with an anti-c-Kit-blocking antibody or with a tyrosine kinase inhibitor prevented the mobilization of CD11b(+)c-Kit(+) cells and attenuated metastasis. C-Kit inhibition was also effective in reducing mobilization of CD11b(+)c-Kit(+) cells and inhibiting lung metastasis after irradiation of established tumors. CONCLUSIONS Our work defines KitL/c-Kit as a previously unidentified axis critically involved in promoting metastasis of 4T1 tumors growing in preirradiated mammary tissue. Pharmacologic inhibition of this axis represents a potential therapeutic strategy to prevent metastasis in breast cancer patients with local relapses after radiotherapy.
Collapse
Affiliation(s)
- François Kuonen
- Department of Radio-Oncology, Centre Hospitalier Universitaire Vaudois, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Köberle M, Kaesler S, Kempf W, Wölbing F, Biedermann T. Tetraspanins in mast cells. Front Immunol 2012; 3:106. [PMID: 22783251 PMCID: PMC3346162 DOI: 10.3389/fimmu.2012.00106] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 04/17/2012] [Indexed: 01/01/2023] Open
Abstract
Mast cells (MC) are key mediators of the immune system, most prominently known for their role in eliciting harmful allergic reactions. Mast cell mediator release (e.g. by degranulation) is triggered by FcεRI recognition of antigen – IgE complexes. Until today no therapeutic targeting of this and other mast cell activation pathways is established. Among possible new candidates there are tetraspanins that have been described on MC already several years ago. Tetraspanins are transmembrane proteins acting as scaffolds, mediating local clustering of their interaction partners, and thus amplify their activities. More recently, tetraspanins were also found to exert intrinsic receptor functions. Tetraspanins have been found to be crucial components of fundamental biological processes like cell motility and adhesion. In immune cells, they not only boost the effectiveness of antigen presentation by clustering MHC molecules, they are also key players in all kinds of degranulation events and immune receptor clustering. This review focuses on the contribution of tetraspanins clustered with FcεRI or residing in granule membranes to classical MC functions but also undertakes an outlook on the possible contribution of tetraspanins to newly described mast cell functions and discusses possible targets for drug development.
Collapse
Affiliation(s)
- Martin Köberle
- Department of Dermatology, Eberhard Karls University Tübingen Tübingen, Germany
| | | | | | | | | |
Collapse
|
27
|
Schaefer T, Zajonz A, Lorentz P, Bohnacker T, Wymann MP, Schweighoffer T. Luminal decoration of blood vessels by activated perivasal mast cells in allergic rhinitis. Allergy 2012; 67:510-20. [PMID: 22313335 DOI: 10.1111/j.1398-9995.2012.02790.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/30/2011] [Indexed: 12/14/2022]
Abstract
BACKGROUND In allergic diseases, like in rhinitis, antigen challenge induces rapid degranulation of tissue resident mast cells and subsequent recruitment of leukocytes in response to soluble immunmodulators. The fate of mast cell-derived, membrane associated factors in inflamed tissue remained however unresolved. METHODS Components of the mast cell granular membrane, including the unique marker CD63var, were examined by FACS and by confocal laser scanning microscopy in cell culture and in diseased human tissue. RESULTS We discovered that selected mast cell membrane components appeared on the surface of distinct bystander cells. Acceptor cells did not acquire these molecules simply by uptake of soluble material or in the form of exosomes. Instead, physically stable cell-to-cell contact was required for transfer, in which a Notch2-Jagged1 interaction played a decisive role. This process is activation-dependent, unidirectional, and involves a unique membrane topology. Endothelial cells were particularly efficient acceptors. In organotypic 3D in vitro cultures we found that transferred mast cell molecules traversed an endothelial monolayer, and reappeared focally compacted on its distal surface, away from the actual contact zone. Moreover, we observed that such mast cell-derived membrane patches decorate microcapillaries in the nasal mucosa of allergic rhinitis patients. CONCLUSION Direct membrane transfer from perivasal mast cells into nearby blood vessels constitutes a novel mechanism to modulate endothelial surface features with apparent significance in allergic diseases.
Collapse
Affiliation(s)
- T. Schaefer
- Novartis Institutes for Biomedical Research (NIBR); Basel; Switzerland
| | - A. Zajonz
- Novartis Institutes for Biomedical Research (NIBR); Basel; Switzerland
| | - P. Lorentz
- Institute of Biochemistry and Genetics; Department of Biomedicine; University of Basel; Basel; Switzerland
| | - T. Bohnacker
- Institute of Biochemistry and Genetics; Department of Biomedicine; University of Basel; Basel; Switzerland
| | - M. P. Wymann
- Institute of Biochemistry and Genetics; Department of Biomedicine; University of Basel; Basel; Switzerland
| | - T. Schweighoffer
- Novartis Institutes for Biomedical Research (NIBR); Basel; Switzerland
| |
Collapse
|
28
|
Sugawara K, Bíró T, Tsuruta D, Tóth BI, Kromminga A, Zákány N, Zimmer A, Funk W, Gibbs BF, Zimmer A, Paus R. Endocannabinoids limit excessive mast cell maturation and activation in human skin. J Allergy Clin Immunol 2012; 129:726-738.e8. [PMID: 22226549 DOI: 10.1016/j.jaci.2011.11.009] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 11/01/2011] [Accepted: 11/08/2011] [Indexed: 12/19/2022]
Abstract
BACKGROUND Mast cells (MCs) crucially contribute to many inflammatory diseases. However, the physiological controls preventing excessive activities of MCs in human skin are incompletely understood. OBJECTIVE Since endocannabinoids are important neuroendocrine MC modifiers, we investigated how stimulation/inhibition of cannabinoid 1 (CB1) receptors affect the biology of human skin MCs in situ. METHODS This was investigated in the MC-rich connective tissue sheath of organ-cultured human scalp hair follicles by quantitative (immuno)histomorphometry, ultrastructural, and quantitative PCR techniques with the use of CB1 agonists or antagonists, CB1 knockdown, or CB1 knockout mice. RESULTS Kit+ MCs within the connective tissue sheath of human hair follicles express functional CB1 receptors, whose pharmacological blockade or gene silencing significantly stimulated both the degranulation and the maturation of MCs from resident progenitor cells in situ (ie, enhanced the number of tryptase+, FcεRIα, or chymase+ connective tissue sheath-MCs). This was, at least in part, stem cell factor-dependent. CB1 agonists counteracted the MC-activating effects of classical MC secretagogues. Similar phenomena were observed in CB1 knockout mice, attesting to the in vivo relevance of this novel MC-inhibitory mechanism. CONCLUSION By using human hair follicle organ culture as an unconventional, but clinically relevant model system for studying the biology of MCs in situ, we show that normal skin MCs are tightly controlled by the endocannabinoid system. This limits excessive activation and maturation of MCs from resident progenitors via "tonic" CB1 stimulation by locally synthesized endocannabinoids. The excessive numbers and activation of MCs in allergic and other chronic inflammatory skin diseases may partially arise from resident intracutaneous MC progenitors, for example, because of insufficient CB1 stimulation. Therefore, CB1 stimulation is a promising strategy for the future management of allergy and MC-dependent skin diseases.
Collapse
Affiliation(s)
- Koji Sugawara
- Department of Dermatology, University of Lübeck, Lübeck, Germany; Department of Dermatology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Teodosio C, García-Montero AC, Jara-Acevedo M, Álvarez-Twose I, Sánchez-Muñoz L, Almeida J, Morgado JM, Matito A, Escribano L, Orfao A. An immature immunophenotype of bone marrow mast cells predicts for multilineage D816V KIT mutation in systemic mastocytosis. Leukemia 2011; 26:951-8. [DOI: 10.1038/leu.2011.293] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
30
|
Laidlaw TM, Steinke JW, Tiñana AM, Feng C, Xing W, Lam BK, Paruchuri S, Boyce JA, Borish L. Characterization of a novel human mast cell line that responds to stem cell factor and expresses functional FcεRI. J Allergy Clin Immunol 2011; 127:815-22.e1-5. [PMID: 21281958 DOI: 10.1016/j.jaci.2010.12.1101] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Revised: 11/12/2010] [Accepted: 12/15/2010] [Indexed: 11/24/2022]
Abstract
BACKGROUND Studies of human mast cells (MCs) are constrained by the paucity of functional cell lines, the expense of maintaining MCs in culture, and technical complexities. OBJECTIVE We derived and characterized a human MC line that arose spontaneously from a culture of nontransformed hematopoietic progenitor cells. METHODS CD34(+) enriched mononuclear cells derived from a donor with aspirin-exacerbated respiratory disease were cultured for 8 weeks with stem cell factor and IL-6 and with IL-3 for the first week only. The cells (termed LUVA cells) survived and proliferated without further addition of any growth factors and have been maintained in culture for approximately 2 years. RESULTS LUVA cells possess metachromatic cytoplasmic granules that are immunoreactive for tryptase, cathepsin G, and carboxypeptidase A3. They express transcripts encoding FcεRI, c-kit, chymase, tryptase, histidine decarboxylase, carboxypeptidase A3, and the type 1 receptor for cysteinyl leukotrienes. Flow cytometry confirmed uniform expression of FcεRI, c-kit, and FcγRII. FcεRI cross-linkage induced the release of β-hexosaminidase, prostaglandin D(2), thromboxane A(2), and macrophage inflammatory protein 1β. Immortalization was not associated with either a known genomic mutation of c-kit in the donor or a somatic mutation of c-kit within the cells, and it was not associated with c-kit autophosphorylation. CONCLUSIONS LUVA cells are an immortalized human MC line that can be maintained without stem cell factor and display high levels of normally signaling c-kit and FcεRI. These cells will prove valuable for functional human MC studies.
Collapse
Affiliation(s)
- Tanya M Laidlaw
- Department of Medicine, Harvard Medical School, Boston, Mass, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Sánchez-Muñoz L, Teodósio C, Morgado JM, Escribano L. Immunophenotypic Characterization of Bone Marrow Mast Cells in Mastocytosis and Other Mast Cell Disorders. Methods Cell Biol 2011; 103:333-59. [DOI: 10.1016/b978-0-12-385493-3.00014-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
32
|
Brooks SA, Bailey E. RT-qPCR comparison of mast cell populations in whole blood from healthy horses and those with laminitis. Anim Genet 2010; 41 Suppl 2:16-22. [PMID: 21070271 DOI: 10.1111/j.1365-2052.2010.02093.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Inflammatory damage to the digital laminae, a structure responsible for suspension of the distal skeleton within the hoof capsule, results in a painful and often life-threatening disease in horses called laminitis. There can be many diverse causes of laminitis; however, previous work in the horse has suggested that in each case, the inflammation and resulting tissue damage is consistent with the action of mediators released from mast cells (MC), as well as the downstream consequences of their activation. The recent development of molecular genetics tools to characterize cells based on their transcriptional activity makes a new approach for measuring MCs possible. Healthy thoroughbred horses from a variety of age groups were used to assess the amount of variation in KIT (encoding mast cell growth factor receptor) and TPSB2 (encoding mast cell tryptase beta 2) gene expression present in the population and to establish "normal" values. Horses (n=9) with a wider range of body condition scores (3-8), because of a more lax management setting that could predispose them to laminitis, had significantly higher KIT expression in circulating peripheral blood cells than horses under individualized management conditions (n=10) that produced ideal body condition scores (4-6) (mean 2.573-fold, P<0.0005). Likewise, horses affected with acute laminitis (n=11) had elevated expression of TPSB2 (2.760-fold, P=0.0011) relative to control horses (n=15). These data suggest that investigation of MC-related genes KIT and TPSB2 may be effective to assay MC population and activity. More work is needed to refine the diagnostic criteria to better describe at what point MC activation occurs and illustrate the use of gene expression assays in clinical cases of laminitis. Additionally, MC activation is associated with inflammatory disease in several mammalian species and may prove a valuable therapeutic target in the horse.
Collapse
Affiliation(s)
- S A Brooks
- Department of Animal Science, 129 Morrison Hall, Cornell University, Ithaca, NY 14853, USA.
| | | |
Collapse
|
33
|
Schäfer T, Starkl P, Allard C, Wolf RM, Schweighoffer T. A granular variant of CD63 is a regulator of repeated human mast cell degranulation. Allergy 2010; 65:1242-55. [PMID: 20337613 DOI: 10.1111/j.1398-9995.2010.02350.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Mast cells are secretory immune cells whose degranulation can provoke acute allergic reactions. It is presently unclear, however, whether an individual mast cell can repeatedly degranulate or turns dysfunctional after a single antigen stimulus. This work thus aims to better define the mast cell life cycle, with particular focus on new target structures for therapeutic or diagnostic approaches in allergy. METHODS Monoclonal antibodies were raised against degranulated cord blood-derived human mast cells. A subset of these antibodies that exclusively recognized degranulated mast cells, but did not cross-react with quiescent mast cells or other hematopoietic cell types, became key reagents in subsequent experiments. RESULTS We identified a granular variant of tetraspanin CD63 as an exclusive molecular marker of degranulated human mast cells. Mutant analyses indicate that a cysteine cluster around residue C170 and protein glycosylation at residue N172 account for the antibody specificity. Here, we show that mast cells, which underwent an initial FcεRI-mediated degranulation, can be degranulated for at least another cycle in vitro. Repeated degranulation, however, requires an IgE/antigen stimulus that differs from the preceding one. Furthermore, the new variant-specific anti-CD63 antibodies effectively impair repeated cycles of mast cell degranulation. CONCLUSION Our findings indicate that mast cells are stable, multiple-use cells, which are capable of surviving and delivering several consecutive hits. Surface expression of the novel CD63 variant is a distinguishing feature of such primed cells. Reagents directed against this molecular hallmark may thus become valuable diagnostic and therapeutic agents.
Collapse
Affiliation(s)
- Thorsten Schäfer
- Novartis Institutes for Biomedical Research (NIBR), Basel, Switzerland.
| | | | | | | | | |
Collapse
|
34
|
Valent P, Cerny-Reiterer S, Herrmann H, Mirkina I, George TI, Sotlar K, Sperr WR, Horny HP. Phenotypic heterogeneity, novel diagnostic markers, and target expression profiles in normal and neoplastic human mast cells. Best Pract Res Clin Haematol 2010; 23:369-78. [PMID: 21112036 DOI: 10.1016/j.beha.2010.07.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mast cells (MC) are specialized immune cells that play a key role in anaphylactic reactions. Growth, differentiation, and function of these cells are regulated by a complex network of cytokines, surface receptors, signaling molecules, the microenvironment, and the genetic background. A number of previous and more recent data suggest that MC are heterogeneous in terms of cytokine-regulation, expression of cytoplasmic and cell surface antigens, and response to ligands. MC heterogeneity is often organ-specific and is considered to be related to MC plasticity, disease-associated factors, and the maturation stage of the cells. The stem cell factor (SCF) receptor KIT (CD117) is expressed on all types of MC independent of maturation and activation-status. In systemic mastocytosis (SM), KIT is often expressed in MC in a mutated and constitutively activated form. In these patients, MC aberrantly display CD2 and CD25, diagnostic markers of neoplastic MC in all SM variants. In advanced SM, MC co-express substantial amounts of CD30, whereas CD2 expression on MC may be decreased compared to indolent SM. Other surface molecules, such as CD63 or CD203c, are overexpressed on neoplastic MC in SM, and are further upregulated upon cross-linking of the IgE receptor. Some of the cell surface antigens expressed on MC or their progenitors may serve as therapeutic targets in the future. These targets include CD25, CD30, CD33, CD44, and CD117/KIT. The current article provides an overview on cell surface antigens and target receptors expressed by MC in physiologic and reactive tissues, and in patients with SM, with special reference to phenotypic heterogeneity and clinical implications.
Collapse
Affiliation(s)
- Peter Valent
- Ludwig Boltzmann Cluster Oncology, Vienna, Austria.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Teodosio C, García-Montero AC, Jara-Acevedo M, Sánchez-Muñoz L, Alvarez-Twose I, Núñez R, Schwartz LB, Walls AF, Escribano L, Orfao A. Mast cells from different molecular and prognostic subtypes of systemic mastocytosis display distinct immunophenotypes. J Allergy Clin Immunol 2010; 125:719-26, 726.e1-726.e4. [PMID: 20061010 DOI: 10.1016/j.jaci.2009.10.020] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 10/19/2009] [Accepted: 10/20/2009] [Indexed: 02/04/2023]
Abstract
BACKGROUND Systemic mastocytosis (SM) is a heterogeneous group of disorders with distinct clinical and biological behavior. Despite this, little is known about the immunophenotypic features of the distinct diagnostic categories of SM. OBJECTIVE To analyze the immunophenotypic characteristics of bone marrow (BM) mast cells (MCs) of different subtypes of SM. METHODS Bone marrow samples from 123 patients with different subtypes of SM and 92 controls were analyzed for a broad panel of immunophenotypic markers by flow cytometry. RESULTS Three clearly different maturation-associated immunophenotypic profiles were found for BMMCs in SM. These different profiles were associated with both genetic markers of the disease and its clinical behavior. BMMCs from poor-prognosis categories of SM (aggressive SM and MC leukemia) typically showed an immature phenotype with clonal involvement of all myeloid lineages by the D816V stem cell growth factor receptor gene (KIT) mutation. In turn, a mature activated versus resting BMMC immunophenotype was commonly found among patients with good-prognosis subtypes of SM depending on whether they carried (indolent SM and clonal MC activation disorders) or not (well differentiated SM) the D816V KIT mutation. CONCLUSION Bone marrow MCs from SM show 3 different maturation-related immunophenotypic profiles that are associated with both the genetic markers of the disease and its clinical behavior.
Collapse
Affiliation(s)
- Cristina Teodosio
- Servicio General de Citometría, Instituto de Biología Molecular y Celular del Cáncer, Centro de Investigación del Cáncer/IBMCC (CSIC-USAL) and Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Identification of proapoptotic Bim as a tumor suppressor in neoplastic mast cells: role of KIT D816V and effects of various targeted drugs. Blood 2009; 114:5342-51. [PMID: 19850739 DOI: 10.1182/blood-2008-08-175190] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Systemic mastocytosis (SM) is a myeloid neoplasm involving mast cells (MCs) and their progenitors. In most cases, neoplastic cells display the D816V-mutated variant of KIT. KIT D816V exhibits constitutive tyrosine kinase (TK) activity and has been implicated in increased survival and growth of neoplastic MCs. Recent data suggest that the proapoptotic BH3-only death regulator Bim plays a role as a tumor suppressor in various myeloid neoplasms. We found that KIT D816V suppresses expression of Bim in Ba/F3 cells. The KIT D816-induced down-regulation of Bim was rescued by the KIT-targeting drug PKC412/midostaurin. Both PKC412 and the proteasome-inhibitor bortezomib were found to decrease growth and promote expression of Bim in MC leukemia cell lines HMC-1.1 (D816V negative) and HMC-1.2 (D816V positive). Both drugs were also found to counteract growth of primary neoplastic MCs. Furthermore, midostaurin was found to cooperate with bortezomib and with the BH3-mimetic obatoclax in producing growth inhibition in both HMC-1 subclones. Finally, a Bim-specific siRNA was found to rescue HMC-1 cells from PKC412-induced cell death. Our data show that KIT D816V suppresses expression of proapoptotic Bim in neoplastic MCs. Targeting of Bcl-2 family members by drugs promoting Bim (re)-expression, or by BH3-mimetics such as obatoclax, may be an attractive therapy concept in SM.
Collapse
|
37
|
Lindberg RL, Achtnichts L, Hoffmann F, Kuhle J, Kappos L. Natalizumab alters transcriptional expression profiles of blood cell subpopulations of multiple sclerosis patients. J Neuroimmunol 2008; 194:153-64. [DOI: 10.1016/j.jneuroim.2007.11.007] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2007] [Revised: 11/01/2007] [Accepted: 11/12/2007] [Indexed: 11/16/2022]
|
38
|
Abstract
Two types of mast cells, MC(T) and MC(TC), exist in humans. MC(T) and MC(TC) are different in their granular neutral proteases, tissue localizations, and functions. This article describes the differences between the cutaneous mast cell receptors.
Collapse
Affiliation(s)
- Michihiro Hide
- Department of Dermatology, Programs for Biomedical Research, Division of Molecular Medical Science, Graduate School of Biomedical Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan.
| | | | | |
Collapse
|
39
|
Lappalainen J, Lindstedt KA, Kovanen PT. A protocol for generating high numbers of mature and functional human mast cells from peripheral blood. Clin Exp Allergy 2007; 37:1404-14. [PMID: 17845422 DOI: 10.1111/j.1365-2222.2007.02778.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Mast cells (MCs) are multi-functional effector cells with an essential role in innate immunity and host defence, and under several pathological conditions, such as allergy. Here, we aimed at defining the culture conditions that would allow efficient generation of mature and functional human MCs from their progenitor cells. METHODS Human peripheral blood-derived CD34(+) progenitor cells were cultured in vitro under serum-free conditions with human stem cell factor for 9 weeks. Growth and differentiation of the cells into MCs were optimized by selected cytokines and a combination of hypoxic and normoxic conditions. MCs were phenotypically characterized by immunocytochemistry, their preformed mediators were quantified, and their functional ability to degranulate and release histamine was tested. RESULTS On average, 20 x 10(6) mature MCs were generated from 0.5 x 10(6) progenitor cells during 9 weeks of culture, i.e. at least a 40-fold increase in cell number was achieved. The mature MCs had oval-shaped non-lobular nuclei, contained histamine, heparin, tryptase, chymase, and cathepsin G in their secretory granules, and strongly expressed c-kit (CD117) and Fc epsilon receptor I on their surface. Histamine release from the cells could be brought about by IgE-anti-IgE cross-linkage, compound 48/80, substance P, and anaphylatoxin C3a. The MCs remained functional for several weeks after their maturation. CONCLUSION This study describes an efficient protocol for generating mature MCs from human peripheral blood with a functional phenotype of connective tissue-type MCs. Use of these cultured human MCs will increase our knowledge and understanding about human MC development and biology in human disease.
Collapse
|
40
|
Yamaguchi M, Azuma H, Fujihara M, Hamada H, Ikeda H. Generation of a considerable number of functional mast cells with a high basal level of FcepsilonRI expression from cord blood CD34+ cells by co-culturing them with bone marrow stromal cell line under serum-free conditions. Scand J Immunol 2007; 65:581-8. [PMID: 17523952 DOI: 10.1111/j.1365-3083.2007.01937.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The number of mast cells (MC) that can be obtained from tissue is limited, making it difficult to study the role of MC. Cultured MC derived from cord blood (CB)-CD34(+) cells proliferate well compared with those derived from adult CD34(+) cells; however, they have been reported to be phenotypically or functionally immature regardless of culture system. For example, very few cells express FcepsilonRI. To resolve this problem, we addressed the effect of human bone marrow stromal cell line on the development of cultured MC. CB-CD34(+) (1 x 10(4)) cells were cultured for 8 weeks in a serum-free medium containing rhIL-6 and rhSCF with or without a human bone marrow stromal cell line, namely, co-culture and liquid culture, and were compared in various regards. MC were basically determined by metachromatic staining of granules. The number of MC obtained (60.3 +/- 15.8 x 10(5) versus 2.0 +/- 1.0 x 10(5)), percentage of FcepsilonRI(+) cells (29.3 +/- 9.4% versus 1.9 +/- 0.8%), histamine content (9.7 +/- 2.8 pg/cell versus 5.8 +/- 2.3 pg/cell), and IgE-mediated histamine release (46 +/- 10% versus 17 +/- 7%) were higher (P < 0.01 and P < 0.05) in the co-culture than in the liquid culture. When CB-CD34(+) cells were developed in liquid culture with the co-culture supernatant (CM), a significant increase (P < 0.01) in the percentage of FcepsilonRI(+) cells and in cell number was observed but these values were lower than those of co-cultured MC. We concluded that this co-culture system was useful for obtaining a considerable number of mature MC with a high basal level of functional FcepsilonRI expression from CB-CD34(+) cells. Yet unknown humoral factors in CM may partly mediate this effect.
Collapse
Affiliation(s)
- M Yamaguchi
- Hokkaido Red Cross Blood Centre, Sapporo, Japan
| | | | | | | | | |
Collapse
|
41
|
Krauth MT, Böhm A, Agis H, Sonneck K, Samorapoompichit P, Florian S, Sotlar K, Valent P. Effects of the CD33-targeted drug gemtuzumab ozogamicin (Mylotarg) on growth and mediator secretion in human mast cells and blood basophils. Exp Hematol 2007; 35:108-16. [PMID: 17198879 DOI: 10.1016/j.exphem.2006.09.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2006] [Revised: 09/11/2006] [Accepted: 09/13/2006] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Mylotarg (gemtuzumab ozogamicin [GO]) has recently been introduced as a novel CD33-targeting drug in clinical hematology. However, despite efficacy, GO produces significant side effects including an infusion syndrome. We have recently shown that mast cells (MCs) and basophils (BAs) express CD33. In the present study, we investigated the effects of GO on growth and mediator secretion in MCs and BAs. METHODS Growth-inhibitory effects of GO on neoplastic MCs (HMC-1) and BAs (KU812) as well as cord blood-derived MC and BA progenitor cells were determined by counting cell numbers and the numbers of apoptotic cells. The amount of histamine secreted from primary MCs and BAs was measured by radioimmunoassay after incubation of cells with GO alone or GO together with an anti-immunoglobulin E (IgE) antibody. RESULTS MCs and BAs as well as HMC-1 cells and KU812 cells were found to express CD33 mRNA and the CD33 protein. GO was found to inhibit the growth of HMC-1 cells and KU812 cells as well as stem cell factor-dependent differentiation of MCs and interleukin-3-induced growth of BAs from their cord blood-derived progenitors. The GO-induced inhibition of growth of neoplastic cells was found to be associated with induction of apoptosis. GO neither induced secretion of histamine from MCs or BAs nor upregulated the anti-IgE-induced release of histamine in these cells. CONCLUSIONS GO counteracts growth of normal and neoplastic MCs and BAs without inducing rapid release of histamine. The exact value of GO as a targeted drug for the treatment of high-grade MC or BA neoplasms remains to be determined.
Collapse
MESH Headings
- Aminoglycosides/pharmacology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Antigens, CD/analysis
- Antigens, CD/drug effects
- Antigens, CD/genetics
- Antigens, Differentiation, Myelomonocytic/analysis
- Antigens, Differentiation, Myelomonocytic/drug effects
- Antigens, Differentiation, Myelomonocytic/genetics
- Apoptosis/drug effects
- Basophils/cytology
- Basophils/drug effects
- Basophils/metabolism
- Blood Cells
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cells, Cultured
- Fetal Blood/cytology
- Gemtuzumab
- Histamine/metabolism
- Humans
- Mast Cells/cytology
- Mast Cells/drug effects
- Mast Cells/metabolism
- Sialic Acid Binding Ig-like Lectin 3
- Stem Cells
Collapse
Affiliation(s)
- Maria-Theresa Krauth
- Department of Internal Medicine I, Division of Hematology & Hemostaseology, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Bischoff SC. Role of mast cells in allergic and non-allergic immune responses: comparison of human and murine data. Nat Rev Immunol 2007; 7:93-104. [PMID: 17259966 DOI: 10.1038/nri2018] [Citation(s) in RCA: 455] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The versatile role of mast cells in allergy, in innate immune responses and in the regulation of tissue homeostasis is well recognized. However, it is often not made clear that most mast-cell data derive solely from experiments in mice or rats, species that obviously never suffer from allergic and most other mast-cell-associated human diseases. Data on human mast cells are limited, and the mast-cell source and species from which findings derive are frequently not indicated in the titles and summaries of research publications. This Review summarizes recent data on human mast cells, discusses differences with murine mast cells, and describes new tools to study this increasingly meaningful cell type in humans.
Collapse
Affiliation(s)
- Stephan C Bischoff
- Department of Nutritional Medicine & Immunology, University of Hohenheim, Fruwirthstr. 12, D-70593 Stuttgart, Germany.
| |
Collapse
|
43
|
Florian S, Sonneck K, Czerny M, Hennersdorf F, Hauswirth AW, Bühring HJ, Valent P. Detection of novel leukocyte differentiation antigens on basophils and mast cells by HLDA8 antibodies. Allergy 2006; 61:1054-62. [PMID: 16918507 DOI: 10.1111/j.1398-9995.2006.01171.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Basophils (BA) and mast cells (MC) are important effector cells in allergic reactions. Development, growth and effector cell functions are regulated by a network of cytokines, other ligands, and respective cell surface antigens. METHODS We examined the expression of novel CD antigens on human BA, lung MC, the BA cell line KU-812, and the MC line HMC-1. Expression of surface antigens was analyzed by monoclonal antibodies (mAb) of the HLDA8 workshop and flow cytometry. RESULTS Basophils were found to stain positive for CXCR1 (CD181), CCR1 (CD191), CCR2 (CD192), CCR7 (CD197), IL-18Ralpha (CDw218a), IL-18Rbeta (CDw218b), TRAIL-R1 (CD261), TRAIL-R2 (CD262), TACI (CD267), TLR-4 (CD284), LAIR1 (CD305), EMR-2 (CD312), JAM1 (CD321), and JAM2 (CD322). Lung MC were found to react with mAb against EMR-2 (CD312) and JAM1 (CD321). KU-812 cells were found to stain positive for CXCR1 (CD181), TRAIL-R2 (CD262), B7H2 (CD275), TLR-4 (CD284), JAM1 (CD321), and E-Cadherin (CD324). HMC-1 cells were recognized by mAb against TRAIL-R2 (CD262), B7H2 (CD275), LAIR1 (CD305), EMR-2 (CD312), JAM1 (CD321), and Siglec-6 (CDw327). CONCLUSIONS Extensive phenotyping with antibodies against novel CD antigens provides further evidence that BA and MC represent two separate hematopoietic cell lineages with unique phenotypic properties observed in mature cells as well as malignant immature cells. Further studies are required to define the functional role of these CD antigens expressed in BA and MC.
Collapse
Affiliation(s)
- S Florian
- Department of Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | |
Collapse
|
44
|
|