1
|
Vymazal O, Papatheodorou I, Andrejčinová I, Bosáková V, Vascelli G, Bendíčková K, Zelante T, Hortová-Kohoutková M, Frič J. Calcineurin-NFAT signaling controls neutrophils' ability of chemoattraction upon fungal infection. J Leukoc Biol 2024; 116:816-829. [PMID: 38648505 DOI: 10.1093/jleuko/qiae091] [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: 12/01/2023] [Revised: 03/03/2024] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
Calcineurin-nuclear factor of activated T cells (CN-NFAT) inhibitors are widely clinically used drugs for immunosuppression, but besides their required T cell response inhibition, they also undesirably affect innate immune cells. Disruption of innate immune cell function can explain the observed susceptibility of CN-NFAT inhibitor-treated patients to opportunistic fungal infections. Neutrophils play an essential role in innate immunity as a defense against pathogens; however, the effect of CN-NFAT inhibitors on neutrophil function was poorly described. Thus, we tested the response of human neutrophils to opportunistic fungal pathogens, namely Candida albicans and Aspergillus fumigatus, in the presence of CN-NFAT inhibitors. Here, we report that the NFAT pathway members were expressed in neutrophils and mediated part of the neutrophil response to pathogens. Upon pathogen exposure, neutrophils underwent profound transcriptomic changes with subsequent production of effector molecules. Importantly, genes and proteins involved in the regulation of the immune response and chemotaxis, including the chemokines CCL2, CCL3, and CCL4 were significantly upregulated. The presence of CN-NFAT inhibitors attenuated the expression of these chemokines and impaired the ability of neutrophils to chemoattract other immune cells. Our results amend knowledge about the impact of CN-NFAT inhibition in human neutrophils.
Collapse
Affiliation(s)
- Ondrej Vymazal
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Ioanna Papatheodorou
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Ivana Andrejčinová
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Veronika Bosáková
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Gianluca Vascelli
- Section of Immunology and General Pathology, Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi 1/8, Perugia, 06132, Italy
| | - Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Teresa Zelante
- Section of Immunology and General Pathology, Department of Medicine and Surgery, University of Perugia, Piazza Lucio Severi 1/8, Perugia, 06132, Italy
| | - Marcela Hortová-Kohoutková
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Pekařská 664/53, Brno, 602 00, Czech Republic
- International Clinical Research Center, Faculty of Medicine, Masaryk University, Kamenice 753/5, Brno, 625 00, Czech Republic
- Institute of Hematology and Blood Transfusion, U Nemocnice 2094/1, Prague 2, 128 00, Czech Republic
| |
Collapse
|
2
|
Chacón P, Vega-Rioja A, Doukkali B, Del Valle Rodriguez A, Fernández-Delgado L, Domínguez-Cereijo L, Segura C, Pérez-Machuca BM, Perkins JR, El Bekay R, Cornejo-García JA, Hajji N, Monteseirín J, Rivas-Pérez D. Human Neutrophils Couple Nitric Oxide Production and Extracellular Trap Formation in Allergic Asthma. Am J Respir Crit Care Med 2024; 210:593-606. [PMID: 38445953 DOI: 10.1164/rccm.202305-0889oc] [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: 05/28/2023] [Accepted: 03/06/2024] [Indexed: 03/07/2024] Open
Abstract
Rationale: Nitric oxide (NO) is increased in the airways and serum of patients with allergic asthma, suggesting an important role in asthma. NO production has been widely attributed to the canonical inducible NO synthase. Much effort has been made to inhibit this enzyme, with two outcomes: no asthma improvement and partial NO reduction, suggesting the involvement of an inducible NO synthase-independent source. Objectives: Neutrophils produce NO under inflammatory conditions, and their role in asthma has been overlooked. The present study analyzes their possible role as sources of NO. Methods: Our hypothesis was tested in 99 allergic patients with intermittent bronchial asthma and 26 healthy donors. NO production by blood and sputum neutrophils in response to allergens, anti-IgE, and anti-IgE receptor antibodies was assessed by Griess reagent, flow cytometry, and confocal microscopy. The formation of extracellular traps (ETs) as a possible consequence of NO production was quantified by Western blot and confocal microscopy, and reactive oxygen species were assessed with luminol-enhanced chemiluminescence. Measurements and Main Results: Among blood and sputum granulocytes from patients with allergic asthma, only neutrophils produce NO by an IgE-dependent mechanism. This production is independent of NO synthase, but dependent on a reaction between L-arginine and reactive oxygen species from NOX2 (NADPH oxidase). NO and ETosis are induced in parallel, and NO amplifies ET formation, which is a key mediator in asthma. Conclusions: Our findings reveal a novel role of neutrophils as the unique allergen/IgE-dependent NO source in allergic asthma, enhancing ET formation. These results suggest that NO produced by neutrophils needs further consideration in the treatment of allergic asthma.
Collapse
Affiliation(s)
- Pedro Chacón
- Laboratorio de Inmunología y Alergia-Fundación para la Gestión de la Investigación en Salud de Sevilla, Unidad de Gestión Clínica de Alergología, Hospital Universitario Virgen Macarena, Seville, Spain
- Departamento de Medicina, Facultad de Medicina, and
| | - Antonio Vega-Rioja
- Laboratorio de Inmunología y Alergia-Fundación para la Gestión de la Investigación en Salud de Sevilla, Unidad de Gestión Clínica de Alergología, Hospital Universitario Virgen Macarena, Seville, Spain
- Departamento de Medicina, Facultad de Medicina, and
| | - Bouchra Doukkali
- Laboratorio de Inmunología y Alergia-Fundación para la Gestión de la Investigación en Salud de Sevilla, Unidad de Gestión Clínica de Alergología, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Alberto Del Valle Rodriguez
- Laboratorio de Inmunología y Alergia-Fundación para la Gestión de la Investigación en Salud de Sevilla, Unidad de Gestión Clínica de Alergología, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Lourdes Fernández-Delgado
- Laboratorio de Inmunología y Alergia-Fundación para la Gestión de la Investigación en Salud de Sevilla, Unidad de Gestión Clínica de Alergología, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Leticia Domínguez-Cereijo
- Laboratorio de Inmunología y Alergia-Fundación para la Gestión de la Investigación en Salud de Sevilla, Unidad de Gestión Clínica de Alergología, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Carmen Segura
- Laboratorio de Inmunología y Alergia-Fundación para la Gestión de la Investigación en Salud de Sevilla, Unidad de Gestión Clínica de Alergología, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Beatriz María Pérez-Machuca
- Laboratorio de Inmunología y Alergia-Fundación para la Gestión de la Investigación en Salud de Sevilla, Unidad de Gestión Clínica de Alergología, Hospital Universitario Virgen Macarena, Seville, Spain
| | - James R Perkins
- Centro de Investigación Biomédica en Red de Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Investigación Biomédica de Málaga, Málaga, Spain
| | - Rajaa El Bekay
- Unidad de Gestión Clínica de Endocrinología y Nutrición, Instituto de Investigación Biomédica de Málaga Plataforma en Nanomedicina, Hospital Regional Universitario de Málaga, Málaga, Spain
- Obesity and Nutrition Centro de Investigación Biomédica en Red, Instituto de Salud Carlos III, Madrid, Spain
| | - José Antonio Cornejo-García
- Instituto de Investigación Biomédica de Málaga, Fundación Pública Andaluza para la Investigación de Málaga en Biomedicina y Salud, Málaga, Spain
| | - Nabil Hajji
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom; and
| | - Javier Monteseirín
- Hospital Quirón Sagrado Corazón and Hospital Quirón Infanta-Luisa, Seville, Spain
| | | |
Collapse
|
3
|
Vega-Rioja A, Chacón P, Fernández-Delgado L, Doukkali B, del Valle Rodríguez A, Perkins JR, Ranea JAG, Dominguez-Cereijo L, Pérez-Machuca BM, Palacios R, Rodríguez D, Monteseirín J, Ribas-Pérez D. Regulation and directed inhibition of ECP production by human neutrophils. Front Immunol 2022; 13:1015529. [PMID: 36518751 PMCID: PMC9744134 DOI: 10.3389/fimmu.2022.1015529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/11/2022] [Indexed: 11/30/2022] Open
Abstract
Background Neutrophils are involved in the pathophysiology of allergic asthma, where the Eosinophil Cationic Protein (ECP) is a critical inflammatory mediator. Although ECP production is attributed to eosinophils, we reported that ECP is also present in neutrophils from allergic patients where, in contrast to eosinophils, it is produced in an IgE-dependent manner. Given the key role of ECP in asthma, we investigated the molecular mechanisms involved in ECP production as well as the effects induced by agonists and widely used clinical approaches. We also analyzed the correlation between ECP production and lung function. Methods Neutrophils from allergic asthmatic patients were challenged with allergens, alone or in combination with cytokines, in the presence of cell-signaling inhibitors and clinical drugs. We analyzed ECP levels by ELISA and confocal microscopy. Lung function was assessed by spirometry. Results IgE-mediated ECP release is dependent on phosphoinositide 3-kinase, the extracellular signal-regulated kinase (ERK1/2) and the production of reactive oxygen species by NADPH-oxidase. Calcineurin phosphatase and the transcription factor NFAT are also involved. ECP release is enhanced by the cytokines interleukin (IL)-5 and granulocyte macrophage-colony stimulating factor, and inhibited by interferon-γ, IL-10, clinical drugs (formoterol, tiotropium and budesonide) and allergen-specific IT. We also found an inverse correlation between asthma severity and ECP levels. Conclusions Our results suggest the molecular pathways involved in ECP production and potential therapeutic targets. We also provide a new method to evaluate disease severity in asthmatic patients based on the quantification of in vitro ECP production by peripheral neutrophils.
Collapse
Affiliation(s)
- Antonio Vega-Rioja
- UGC de Alergología, Hospital Universitario Virgen Macarena, Sevilla, Spain,Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain,*Correspondence: Antonio Vega-Rioja, ; Pedro Chacón, ; Javier Monteseirín,
| | - Pedro Chacón
- UGC de Alergología, Hospital Universitario Virgen Macarena, Sevilla, Spain,Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain,*Correspondence: Antonio Vega-Rioja, ; Pedro Chacón, ; Javier Monteseirín,
| | | | - Bouchra Doukkali
- UGC de Alergología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | | | - James R. Perkins
- Departamento de Biología Molecular y Bioquímica. Facultad de Ciencias, Universidad de Málaga, Málaga, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | - Juan A. G. Ranea
- Departamento de Biología Molecular y Bioquímica. Facultad de Ciencias, Universidad de Málaga, Málaga, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain,Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain
| | | | | | | | | | - Javier Monteseirín
- Hospital Quirón Sagrado Corazón and Hospital Quirón Infanta-Luisa, Sevilla, Spain,*Correspondence: Antonio Vega-Rioja, ; Pedro Chacón, ; Javier Monteseirín,
| | | |
Collapse
|
4
|
Abdel-Rafei MK, Thabet NM, Rashed LA, Moustafa EM. Canagliflozin, a SGLT-2 inhibitor, relieves ER stress, modulates autophagy and induces apoptosis in irradiated HepG2 cells: Signal transduction between PI3K/AKT/GSK-3β/mTOR and Wnt/β-catenin pathways; in vitro. J Cancer Res Ther 2021; 17:1404-1418. [PMID: 34916371 DOI: 10.4103/jcrt.jcrt_963_19] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Background and Objectives Metabolic shifting from mitochondrial respiration to glycolysis characterizes malignant cells from its normal counterparts and is attributed to overactivation of oncogenic signaling pathways. Hence, this study intended to investigate the influence of canagliflozin (CAN) and/or γ-irradiation (γ-IR) on HepG2 cell proliferation, crosstalk between phosphatidylinositol 3-kinases (PI3K)/AKT/glycogen synthase kinase-3-β (GSK3-β)/mTOR and Wnt/β-catenin signaling pathways, and their regulation of diverse processes, such as endoplasmic reticulum (ER) stress, autophagy, and apoptosis. Materials and Methods HepG2 cells were treated with different doses of CAN and then exposed to different doses of γ-IR to achieve optimization that was based on cytotoxicity and clonogenic assays, respectively. The effects of CAN and/or γ-IR on glycolytic metabolism, cellular bioenergetics, oxidative stress, ER stress and autophagy biomarkers, expression of PI3K/AKT/GSK3-β/mTOR and Wnt/β-Catenin signaling pathways, and apoptotic markers were monitored. Results CAN enhanced the antitumor potential of γ-IR as displayed by a significant inhibition of clonogenic survival in HepG2 cells via inhibition of glucose uptake, lactate release, and modulation of ER stress-mediated autophagy; switched it to apoptosis; as well as disabled signaling pathways which contribute to metabolic reprogramming and tumor progression induced by γ-IR that confer radioresistance and treatment failure. Conclusion Our study sheds light on the effective combination of CAN and γ-IR in hepatocellular carcinoma treatment and necessitates CAN treatment prior to γ-IR to overcome metabolic reprogramming-associated radioresistance and improve curative outcomes.
Collapse
Affiliation(s)
- Mohamed Khairy Abdel-Rafei
- Department of Radiation Biology, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Noura Magdy Thabet
- Department of Radiation Biology, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Laila Ahmed Rashed
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Enas Mahmoud Moustafa
- Department of Radiation Biology, National Centre for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
5
|
Vymazal O, Bendíčková K, De Zuani M, Vlková M, Hortová-Kohoutková M, Frič J. Immunosuppression Affects Neutrophil Functions: Does Calcineurin-NFAT Signaling Matter? Front Immunol 2021; 12:770515. [PMID: 34795676 PMCID: PMC8593005 DOI: 10.3389/fimmu.2021.770515] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 10/19/2021] [Indexed: 11/17/2022] Open
Abstract
Neutrophils are innate immune cells with important roles in antimicrobial defense. However, impaired or dysregulated neutrophil function can result in host tissue damage, loss of homeostasis, hyperinflammation or pathological immunosuppression. A central link between neutrophil activation and immune outcomes is emerging to be the calcineurin-nuclear factor of activated T cells (NFAT) signaling pathway, which is activated by neutrophil detection of a microbial threat via pattern recognition receptors and results in inflammatory cytokine production. This potent pro-inflammatory pathway is also the target of several immunosuppressive drugs used for the treatment of autoimmune disorders, during solid organ and hematopoietic cell transplantations, and as a part of anti-cancer therapy: but what effects these drugs have on neutrophil function, and their broader consequences for immune homeostasis and microbial defense are not yet known. Here, we bring together the emerging literature describing pathology- and drug- induced neutrophil impairment, with particular focus on their effects on calcineurin-NFAT signaling in the innate immune compartment.
Collapse
Affiliation(s)
- Ondřej Vymazal
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia.,Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Kamila Bendíčková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Marco De Zuani
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Marcela Vlková
- Department of Clinical Immunology and Allergology, Faculty of Medicine, Masaryk University, Brno, Czechia.,Department of Clinical Immunology and Allergology, St. Anne´s University Hospital, Brno, Czechia
| | | | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia.,Department of Modern Immunotherapy, Institute of Hematology and Blood Transfusion, Prague, Czechia
| |
Collapse
|
6
|
Shin N, Jung N, Lee SE, Kong D, Kim NG, Kook MG, Park H, Choi SW, Lee S, Kang KS. Pimecrolimus interferes the therapeutic efficacy of human mesenchymal stem cells in atopic dermatitis by regulating NFAT-COX2 signaling. Stem Cell Res Ther 2021; 12:482. [PMID: 34454603 PMCID: PMC8399851 DOI: 10.1186/s13287-021-02547-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/08/2021] [Indexed: 11/10/2022] Open
Abstract
Background Human mesenchymal stem cells (hMSCs) therapy has recently been considered a promising treatment for atopic dermatitis (AD) due to their immunomodulation and tissue regeneration ability. In our previous studies, we demonstrated that hMSCs alleviate allergic inflammation in murine AD model by inhibiting the activation of mast cells and B cells. Also our phase I/IIa clinical trial showed clinical efficacy and safety of hMSCs in moderate-to-severe adult AD patients. However, hMSCs therapy against atopic dermatitis have had poor results in clinical field. Therefore, we investigated the reason behind this result. We hypothesized that drug–cell interaction could interfere with the therapeutic efficacy of stem cells, and investigated whether coadministration with pimecrolimus, one of the topical calcineurin inhibitors, could influence the therapeutic potential of human umbilical cord blood mesenchymal stem cells (hUCB-MSCs) in AD. Methods hUCB-MSCs were subcutaneously injected to AD-induced mice with or without pimecrolimus topical application. To examine whether pimecrolimus influenced the immunomodulatory activity of hUCB-MSCs, hUCB-MSCs were treated with pimecrolimus. Results Pimecrolimus disturbed the therapeutic effect of hUCB-MSCs when they were co-administered in murine AD model. Moreover, the inhibitory functions of hUCB-MSCs against type 2 helper T (Th2) cell differentiation and mast cell activation were also deteriorated by pimecrolimus treatment. Interestingly, we found that pimecrolimus decreased the production of PGE2, one of the most critical immunomodulatory factors in hUCB-MSCs. And we demonstrated that pimecrolimus downregulated COX2-PGE2 axis by inhibiting nuclear translocation of NFAT3. Conclusions Coadministration of pimecrolimus with hMSCs could interfere with the therapeutic efficacy of hMSCs in atopic dermatitis, and this is the first study that figured out the interaction of hMSCs with other drugs in cell therapy of atopic dermatitis. Therefore, this study might give rise to improvement of the clinical application of hMSCs therapy and facilitate the widespread application of hMSCs in clinical field. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02547-8.
Collapse
Affiliation(s)
- Nari Shin
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Namhee Jung
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd., Ace Highend Tower 8, 84, Gasan digital 1-ro, Geumcheon-gu, Seoul, 08590, Republic of Korea
| | - Seung-Eun Lee
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Dasom Kong
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Nam Gyo Kim
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Myung Geun Kook
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Hwanhee Park
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd., Ace Highend Tower 8, 84, Gasan digital 1-ro, Geumcheon-gu, Seoul, 08590, Republic of Korea
| | - Soon Won Choi
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Seunghee Lee
- Stem Cell and Regenerative Bioengineering Institute, Global R&D Center, Kangstem Biotech Co. Ltd., Ace Highend Tower 8, 84, Gasan digital 1-ro, Geumcheon-gu, Seoul, 08590, Republic of Korea.
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
7
|
Chacón P, Vega-Rioja A, Doukkali B, Del Valle Rodríguez A, Bellido V, Puente Y, Alcañiz L, Rodríguez D, Palacios R, Cornejo-García JA, Monteseirín J, Rivas-Pérez D. Targeted inhibition of allergen-induced histamine production by neutrophils. FASEB J 2021; 35:e21483. [PMID: 33788304 DOI: 10.1096/fj.202001912r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/12/2021] [Accepted: 02/15/2021] [Indexed: 11/11/2022]
Abstract
Histamine is a critical inflammatory mediator in allergic diseases. We showed in a previous work that neutrophils from allergic patients produce histamine in response to allergens to which the patients were sensitized. Here, we investigate the molecular mechanisms involved in this process using peripheral blood neutrophils. We challenged these cells in vitro with allergens and analyzed histamine release in the culture supernatants. We also explored the effect of common therapeutic drugs that ameliorate allergic symptoms, as well as allergen-specific immunotherapy. Additionally, we examined the expression of histidine decarboxylase and diamine oxidase, critical enzymes in the metabolism of histamine, under allergen challenge. We show that allergen-induced histamine release is dependent on the activation of the phosphoinositide 3-kinase, mitogen-activated protein kinase p38, and extracellular signal-regulated kinase 1/2 signaling pathways. We also found a contribution of the phosphatase calcineurin to lesser extent. Anti-histamines, glucocorticoids, anti-M3-muscarinic receptor antagonists, and mainly β2 -receptor agonists abolished the allergen-dependent histamine release. Interestingly, allergen-specific immunotherapy canceled the histamine release through the downregulation of histidine decarboxylase expression. Our observations describe novel molecular mechanisms involved in the allergen-dependent histamine release by human neutrophils and provide new targets to inhibit histamine production.
Collapse
Affiliation(s)
- Pedro Chacón
- UGC de Alergología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Antonio Vega-Rioja
- UGC de Alergología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Bouchra Doukkali
- UGC de Alergología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | | | - Virginia Bellido
- UGC de Alergología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Yolanda Puente
- UGC de Alergología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Lorena Alcañiz
- UGC de Dermatología, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | | | | | | | - Javier Monteseirín
- UGC de Alergología, Hospital Universitario Virgen Macarena, Sevilla, Spain.,Departamento de Medicina, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | | |
Collapse
|
8
|
Sánchez-Ovando S, Simpson JL, Barker D, Baines KJ, Wark PAB. Transcriptomics of biopsies identifies novel genes and pathways linked to neutrophilic inflammation in severe asthma. Clin Exp Allergy 2021; 51:1279-1294. [PMID: 34245071 DOI: 10.1111/cea.13986] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 06/03/2021] [Accepted: 06/19/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Severe asthma is a complex disease. Transcriptomic profiling has contributed to understanding the pathogenesis of asthma, especially type-2 inflammation. However, there is still poor understanding of non-type-2 asthma, and consequently, there are limited treatment options. OBJECTIVE The aim of this study was to identify differentially expressed genes (DEGs) and pathways in endobronchial biopsies associated with inflammatory phenotypes of severe asthma. METHODS This cross-sectional study examined endobronchial biopsies from 47 adults with severe asthma (neutrophilic asthma (NA) n = 9, eosinophilic asthma (EA) n = 22 and paucigranulocytic asthma (PGA) n = 16) and 13 healthy controls (HC). RNA was extracted and transcriptomic profiles generated (Illumina Humanref-12 V4) and analysed using GeneSpring GX14.9.1. Pathway identification using Ingenuity Pathway Analysis. RESULTS NA had the most distinct profile, with signature of 60 top-ranked DEGs (FC >±2) including genes associated with innate immunity response, neutrophil degranulation and IL-10 signalling. NA presented enrichment to pathways previously linked to neutrophilic inflammation; dendritic cell maturation, Th1, TREM1, inflammasome, Th17 and p38 MAPK, as well as novel links to neuroinflammation, NFAT and PKCθ signalling. EA presented similar transcriptomic profiles to PGA and HC. Despite the higher proportion of bacterial colonization in NA, no changes were observed in the transcriptomic profiles of severe asthma culture positive compared with severe asthma culture negative. CONCLUSIONS & CLINICAL RELEVANCE NA features a distinct transcriptomic profile with seven pathways enriched in NA compared to EA, PGA and HC. All those with severe asthma had significant enrichment for SUMOylation, basal cell carcinoma signalling and Wnt/β-catenin pathways compared to HC, despite high-dose inhaled corticosteroids. These findings contribute to the understanding of mechanistic pathways in endobronchial biopsies associated with NA and identify potential novel treatment targets for severe asthma.
Collapse
Affiliation(s)
- Stephany Sánchez-Ovando
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, University of Newcastle, NSW, Australia
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, University of Newcastle, NSW, Australia
| | - Daniel Barker
- Faculty of Health and Medicine, University of Newcastle, NSW, Australia
| | - Katherine J Baines
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, University of Newcastle, NSW, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, Faculty of Health and Medicine, University of Newcastle, NSW, Australia.,Respiratory and Sleep Medicine, John Hunter Hospital, NSW, Australia
| |
Collapse
|
9
|
Li Y, Zhang YY, Yang LT, Liu JQ, Zhou C, Liu ZQ, Yang G, Mo LH, Liu ZG, Feng BS, Yang PC. FcγRI plays a critical role in patients with ulcerative colitis relapse. Eur J Immunol 2020; 51:459-470. [PMID: 33078845 DOI: 10.1002/eji.202048622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 09/10/2020] [Accepted: 10/19/2020] [Indexed: 01/16/2023]
Abstract
Ulcerative colitis (UC) is a disease that frequently relapses and affects more than 0.1% general population; the underlying mechanism is poorly understood. Published data show that polymorphonuclear neutrophils (PMN) contribute to the pathogenesis of UC. This study aims to identify antigen (Ag)-specific PMNs and investigate their role in UC relapse. In this study, the correlation between PMN activities and UC relapse was assessed in a group of UC patients. A UC mouse model was developed to expand the findings of UC patient study. The results showed that a positive correlation was detected between the high PMN activities and the food Ag-specific IgG amounts in colon biopsies of UC patients. UC patient-derived Ag-specific PMNs could be activated upon exposure to food specific Ag. The Ag/FcγRI complexes were detected on the surface of PMNs in UC patients. Re-exposure of sensitized PMNs to specific Ag triggered PMN activation and induced UC-like inflammation in the mouse colon. We conclude that FcγRI plays a critical role in UC relapse. Inhibition of FcγRI can efficiently inhibits experimental UC.
Collapse
Affiliation(s)
- Yan Li
- Department of Gastroenterology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuan-Yi Zhang
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Department of Respirology & Allergy, Affiliated Luohu Hospital of Shenzhen University, Shenzhen, China
| | - Li-Teng Yang
- Department of Respirology & Allergy, Affiliated Luohu Hospital of Shenzhen University, Shenzhen, China
| | - Jiang-Qi Liu
- Longgang ENT Hospital and Shenzhen ENT Institute, Shenzhen, China
| | - Chuan Zhou
- Department of Gastroenterology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhi-Qiang Liu
- Longgang ENT Hospital and Shenzhen ENT Institute, Shenzhen, China
| | - Gui Yang
- Department of Otolaryngology, Longgang Central Hospital, Shenzhen, China
| | - Li-Hua Mo
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Zhi-Gang Liu
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Bai-Sui Feng
- Department of Gastroenterology, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ping-Chang Yang
- Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Shenzhen, China
| |
Collapse
|
10
|
Ram BM, Dolpady J, Kulkarni R, Usha R, Bhoria U, Poli UR, Islam M, Trehanpati N, Ramakrishna G. Human papillomavirus (HPV) oncoprotein E6 facilitates Calcineurin-Nuclear factor for activated T cells 2 (NFAT2) signaling to promote cellular proliferation in cervical cell carcinoma. Exp Cell Res 2018; 362:132-141. [DOI: 10.1016/j.yexcr.2017.11.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 11/06/2017] [Accepted: 11/08/2017] [Indexed: 12/13/2022]
|
11
|
Wang B, Zhao XH. Apigenin induces both intrinsic and extrinsic pathways of apoptosis in human colon carcinoma HCT-116 cells. Oncol Rep 2016; 37:1132-1140. [PMID: 27959417 DOI: 10.3892/or.2016.5303] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 11/24/2016] [Indexed: 11/06/2022] Open
Abstract
Apigenin is one of the plant-originated flavones with anticancer activities. In this study, apigenin was assessed for its in vitro effects on a human colon carcinoma line (HCT‑116 cells) in terms of anti-proliferation, cell cycle progression arrest, apoptosis and intracellular reactive oxygen species (ROS) generation, and then outlined its possible apoptotic mechanism for the cells. Apigenin exerted cytotoxic effect on the cells via inhibiting cell growth in a dose-time-dependent manner and causing morphological changes, arrested cell cycle progression at G0/G1 phase, and decreased mitochondrial membrane potential of the treated cells. Apigenin increased respective ROS generation and Ca2+ release and thereby, caused ER stress in the treated cells. Apigenin shows apoptosis induction towards the cells, resulting in enhanced portion of apoptotic cells. A mechanism involved ROS generation and endoplasmic reticulum stress was outlined for the apigenin-mediated apoptosis via both intrinsic mitochondrial and extrinsic pathways, based on the assayed mRNA and protein expression levels in the cells. With this mechanism, apigenin resulted in the HCT-116 cells with enhanced intracellular ROS generation and Ca2+ release together with damaged mitochondrial membrane, and upregulated protein expression of CHOP, DR5, cleaved BID, Bax, cytochrome c, cleaved caspase-3, cleaved caspase-8 and cleaved caspase-9, which triggered apoptosis of the cells.
Collapse
Affiliation(s)
- Bo Wang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China
| | - Xin-Huai Zhao
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin, Heilongjiang 150030, P.R. China
| |
Collapse
|
12
|
Park J, Choi H, Kim B, Chae U, Lee DG, Lee SR, Lee S, Lee HS, Lee DS. Peroxiredoxin 5 (Prx5) decreases LPS-induced microglial activation through regulation of Ca 2+/calcineurin-Drp1-dependent mitochondrial fission. Free Radic Biol Med 2016; 99:392-404. [PMID: 27585948 DOI: 10.1016/j.freeradbiomed.2016.08.030] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 07/24/2016] [Accepted: 08/28/2016] [Indexed: 12/30/2022]
Abstract
Microglial activation is a hallmark of neurodegenerative diseases. ROS activates microglia by regulating transcription factors to express pro-inflammatory genes and is associated with disruption of Ca2+ homeostasis through thiol redox modulation. Recently, we reported that Prx5 can regulate activation of microglia cells by governing ROS. In addition, LPS leads to excessive mitochondrial fission, and regulation of mitochondrial dynamics involved in a pro-inflammatory response is important for the maintenance of microglial activation. However, the precise relationship among these signals and the role of Prx5 in mitochondrial dynamics and microglial activation is still unknown. In this study, we demonstrated that Ca2+/calcineurin-dependent de-phosphorylation of Drp1 induces mitochondrial fission and regulates mitochondrial ROS production, which influences the expression of pro-inflammatory mediators in LPS-induced microglia cells. Moreover, it is likely that cytosolic and Nox-derived ROS were upstream of mitochondrial fission and mitochondrial ROS generation in activated microglia cells. Prx5 regulates LPS-induced mitochondrial fission through modulation of Ca2+/calcineurin-dependent Drp1 de-phosphorylation by eliminating Nox-derived and cytosolic ROS. Therefore, we suggest that mitochondrial dynamics may be essential for understanding pro-inflammatory responses and that Prx5 may be used as a new therapeutic target to prevent neuroinflammation and neurodegenerative diseases.
Collapse
Affiliation(s)
- Junghyung Park
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hoonsung Choi
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeollabuk-do, Republic of Korea
| | - Bokyung Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Unbin Chae
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Dong Gil Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Chungcheongbuk-do, Republic of Korea
| | - Seunghoon Lee
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Jeollabuk-do, Republic of Korea
| | - Hyun-Shik Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea; College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
13
|
Fernández-Delgado L, Vega-Rioja A, Ventura I, Chamorro C, Aroca R, Prados M, Bobadilla P, Rodríguez D, Palacios R, Monteseirín J. Allergens Induce the Release of Lactoferrin by Neutrophils from Asthmatic Patients. PLoS One 2015; 10:e0141278. [PMID: 26488881 PMCID: PMC4619071 DOI: 10.1371/journal.pone.0141278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 10/05/2015] [Indexed: 11/26/2022] Open
Abstract
Background Despite the evidence that Lactoferrin (Lf) is involved in allergic asthma processes, it is unknown whether neutrophils can be one of the main cellular sources of this key inflammatory mediator directly in response of an IgE mediated stimulus. The present study was undertaken to analyze this question. Methods Neutrophils from healthy subjects (n = 34) and neutrophils from allergic asthmatic patients (n = 102) were challenged in vitro with specific allergens to which the patients were sensitized, PAF, or agonist mAbs against IgE-receptors, and the levels of Lf were measured in the culture supernatant. The levels of serum IgE together with the severity of symptoms were also analyzed. Results Lf was released into the culture supernatant of neutrophils from allergic asthmatic patients in response to allergens and PAF. This response was highly allergen-specific, and did not happen in neutrophils from healthy donors. Allergen effect was mimicked by Abs against FcεRI and galectin-3 but not by FcεRII. The levels of released Lf correlated well with the levels of serum specific IgE and severity of asthma symptoms. These observations represent a novel view of neutrophils as an important source of Lf in allergic asthma. Importantly, the levels of released Lf by neutrophils could therefore be used to evaluate disease severity in allergic asthmatic patients.
Collapse
Affiliation(s)
- Lourdes Fernández-Delgado
- Unidad de Gestión Clínica de Alergia Intercentros, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Antonio Vega-Rioja
- Unidad de Gestión Clínica de Alergia Intercentros, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Inmaculada Ventura
- Unidad de Gestión Clínica de Alergia Intercentros, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Cristina Chamorro
- Unidad de Gestión Clínica de Alergia Intercentros, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Rocío Aroca
- Unidad de Gestión Clínica de Alergia Intercentros, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | - Manuel Prados
- Unidad de Gestión Clínica de Alergia Intercentros, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | | | | | | | - Javier Monteseirín
- Unidad de Gestión Clínica de Alergia Intercentros, Hospital Universitario Virgen Macarena, Sevilla, Spain
- Departamento de Medicina. Facultad de Medicina. Universidad de Sevilla, Sevilla, Spain
- * E-mail:
| |
Collapse
|
14
|
Gupta AK, Giaglis S, Hasler P, Hahn S. Efficient neutrophil extracellular trap induction requires mobilization of both intracellular and extracellular calcium pools and is modulated by cyclosporine A. PLoS One 2014; 9:e97088. [PMID: 24819773 PMCID: PMC4018253 DOI: 10.1371/journal.pone.0097088] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 04/14/2014] [Indexed: 12/21/2022] Open
Abstract
Excessive or aberrant generation of neutrophil extracellular traps (NETs) has recently become implicated in the underlying aetiology of a number of human pathologies including preeclampsia, systemic lupus erythromatosus, rheumatoid arthritis, auto-antibody induced small vessel vasculitis, coagulopathies such as deep vein thrombosis or pulmonary complications. These results imply that effective pharmacological therapeutic strategies will need to be developed to counter overt NETosis in these and other inflammatory disorders. As calcium flux is implicated in the generation of reactive oxygen species and histone citrullination, two key events in NETosis, we analysed the roles of both extra- and intracellular calcium pools and their modulation by pharmacological agents in the NETotic process in detail. Interleukin-8 (IL-8) was used as a physiological stimulus of NETosis. Our data demonstrate that efficient induction of NETosis requires mobilisation of both extracellular and intracellular calcium pools. Since modulation of the calcineurin pathway by cyclosporine A has been described in neutrophils, we investigated its influence on NETosis. Our data indicate that IL-8 induced NETosis is reduced by ascomycin and cyclosporine A, antagonists of the calcineurin pathway, but not following treatment with rapamycin, which utilizes the mTOR pathway. The action of the G protein coupled receptor phospholipase C pathway appears to be essential for the induction of NETs by IL-8, as NETosis was diminished by treatment with either pertussis toxin, a G-protein inhibitor, the phospholipase C inhibitor, U73122, or staurosporine, an inhibitor of protein kinase C. The data regarding the calcineurin antagonists, ascomycin and cyclosporine A, open the possibility to therapeutically supress or modulate NETosis. They also provide new insight into the mechanism whereby such immune suppressive drugs render transplant patients susceptible to opportunistic fungal infections.
Collapse
Affiliation(s)
- Anurag Kumar Gupta
- Laboratory for Prenatal Medicine, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Stavros Giaglis
- Laboratory for Prenatal Medicine, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
- Department of Rheumatology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Paul Hasler
- Department of Rheumatology, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Sinuhe Hahn
- Laboratory for Prenatal Medicine, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
15
|
Ventura I, Vega A, Chamorro C, Aroca R, Gómez E, Pineda F, Palacios R, Blanca M, Monteseirín J. Allergen immunotherapy decreases LPS-induced NF-κB activation in neutrophils from allergic patients. Pediatr Allergy Immunol 2014; 25:129-35. [PMID: 24118194 DOI: 10.1111/pai.12145] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND Allergen-specific immunotherapy (IT) is widely used to treat allergic diseases. The molecular mechanisms have not been clarified yet completely. The present work was undertaken to analyze the effect of IT in the activation of NF-κB. METHODS Neutrophils from 15 pollen-allergic IT-treated patients, 10 untreated pollen-allergic patients, and 10 healthy donors were in vitro stimulated with LPS. NF-κB activation (p65/p52) was measured in their nuclear extracts by enzyme-linked immunosorbent assay (ELISA). IκBα phosphorylation, NF-κB-repressing factor (NRF) activation, and thromboxane A2 (TXA2 ) and Interleukin-8 (IL-8) release were measured by ELISA. RESULTS There was a positive correlation between the score of symptoms and NF-κB activation in human neutrophils. IT significantly decreased NF-κB activation levels in neutrophils compared with neutrophils from untreated patients. IκBα phosphorylation and NRF activation levels were, respectively, significantly lower and higher in neutrophils from IT-treated patients than from untreated patients. IL-8 and TXA2 release were significantly lower in neutrophils from IT-treated patients than from untreated patients. CONCLUSIONS IT positive effects are at least in part mediated by the negative regulation of NF-κB activation in human neutrophils. These observations represent a novel view of neutrophils as possible cell target to treat IgE-dependent diseases through NF-κB downmodulation.
Collapse
Affiliation(s)
- Inmaculada Ventura
- Servicio de Inmunología y Alergia, Hospital Universitario Virgen Macarena, Sevilla, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Vandewalle A, Tourneur E, Bens M, Chassin C, Werts C. Calcineurin/NFAT signaling and innate host defence: a role for NOD1-mediated phagocytic functions. Cell Commun Signal 2014; 12:8. [PMID: 24479879 PMCID: PMC3910266 DOI: 10.1186/1478-811x-12-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 01/23/2014] [Indexed: 12/12/2022] Open
Abstract
The calcineurin/nuclear factor of activated T cells (NFATs) signaling pathway plays a central role in T cell mediated adaptive immune responses, but a number of recent studies demonstrated that calcineurin/NFAT signaling also plays a key role in the control of the innate immune response by myeloid cells. Calcineurin inhibitors, such as cyclosporine A (CsA) and tacrolimus (FK506), are commonly used in organ transplantation to prevent graft rejection and in a variety of immune diseases. These immunosuppressive drugs have adverse effects and significantly increase host's susceptibility towards bacterial or fungal infections. Recent studies highlighted the role of NFAT signaling in fungal infection and in the control of the pattern recognition receptor nucleotide-binding oligomerization domain-containing protein 1 (NOD1), which predominantly senses invasive Gram-negative bacteria and mediates neutrophil phagocytic functions. This review summarises some of the current knowledge concerning the role of NFAT signaling in the innate immune response and the recent advances on NFAT-dependent inhibition of NOD1-mediated innate immune response caused by CsA, which may contribute to sensitizing transplant recipients to bacterial infection.
Collapse
Affiliation(s)
- Alain Vandewalle
- Centre de Recherche sur l'Inflammation (CRI), UMRS 1149 et Groupe ATIP-AVENIR, Université Denis Diderot - Paris 7, Paris, France.
| | | | | | | | | |
Collapse
|
17
|
Liu H, Zhao Q, Song Q, Zhou FH, Kang HJ, Pan L, Yao YM. Release of High Mobility Protein Box-1 is Greatly Regulated by Nuclear Factor of Activated T Cell-2 in Human Monocytes. EUR J INFLAMM 2014. [DOI: 10.1177/1721727x1401200110] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Close talk between inflammatory mediators and immunological cytokines has been discovered and reported. In this study, the role of nuclear factor of activated T cell-2 (NFAT2) in regulation of high mobility group box-1 (HMGB1) release was investigated. THP-1 cell and HEK293T cell were incubated and stimulated by lipopolysaccharide (LPS). Firstly, binding site between HMGB1 and NFAT2 was identified by co-immunoprecipitation (IP). Box A, Box B and CT domain of HMGB1 were constructed, as well as Rel-homology-domain (RHD), pre-RHD and pro-RHD of NFAT2. THP-1 cell was harvested, cell lysate and culture medium were collected at appointed times. Binding between HMGB1 and NFAT2 was measured, HMGB1 protein level in culture medium was analyzed at the same time. Secondly, the role of NFAT2 in regulating HMGB1 release was investigated. When THP-1 cell was cultured for 24 h, HMGB1 protein level was measured at appointed times with or without siRNA to inhibit NFAT2 expression. Our data show that HMGB1 bound to NFAT2 in THP-1 cell cytoplasm. Further experiments showed that box B domain of HMGB1 could bind to pre-RHD of NFAT2. After stimulation by LPS, interaction between HMGB1 and NFAT2 was discovered decreasing gradually. However, HMGB1 protein level increased in culture medium at the same time. Furthermore, HMGB1 release could be enhanced by NFAT2 inhibition. Taken together, release of HMGB1 could be regulated by NFAT2 in human monocytes.
Collapse
Affiliation(s)
- H. Liu
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Q. Zhao
- Gastroenterology Department of Nanlou, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Q. Song
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - F-H. Zhou
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - H-J. Kang
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - L. Pan
- Critical Care Medicine Department, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Y-M. Yao
- Department of Microbiology and Immunology, Burns Institute, First Hospital Affiliated to the Chinese PLA General Hospital (formerly 304th Hospital), Beijing, People's Republic of China
| |
Collapse
|
18
|
Yamaguchi T, Omori M, Tanaka N, Fukui N. Distinct and additive effects of sodium bicarbonate and continuous mild heat stress on fiber type shift via calcineurin/NFAT pathway in human skeletal myoblasts. Am J Physiol Cell Physiol 2013; 305:C323-33. [PMID: 23703530 DOI: 10.1152/ajpcell.00393.2012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ingestion of sodium bicarbonate (NaHCO3) is known to enhance athletic performance, probably via increased extracellular buffering capacity. At present, little is known about the direct effects of NaHCO3 on myogenesis, especially in vitro. Here, we examined the effects of NaHCO3 and the combined effects of NaHCO3 and continuous mild heat stress (CMHS) at 39°C on the differentiation of human skeletal muscle myoblasts (HSMMs). Levels of myosin heavy chain (MyHC) type I mRNA increased with increasing NaHCO3 concentrations; in contrast, those of MyHC IIx decreased. The NaHCO3-induced fast-to-slow shift was additively enhanced by CMHS. Likewise, intracellular calcium levels and expression of three factors, nuclear factor of activated T cells c2 (NFATc2), NFATc4, and peroxisome-proliferator-activated receptor-γ coactivator-1α, were upregulated with increasing NaHCO3 concentrations; moreover, these effects of NaHCO3 were additively enhanced by CMHS. Overexpression experiments and small interfering RNA-mediated knockdown experiments confirmed that NFATc2 and NFATc4 were involved in MyHC I regulation. The present study provided evidence that NaHCO3 and CMHS distinctly and additively induced a fast-to-slow fiber type shift through changes in intracellular calcium levels and the modulation of calcium signaling.
Collapse
Affiliation(s)
- Tetsuo Yamaguchi
- Clinical Research Center, National Hospital Organization Sagamihara Hospital, Minami-ku, Sagamihara City, Kanagawa, Japan.
| | | | | | | |
Collapse
|
19
|
Alcañiz L, Vega A, Chacón P, El Bekay R, Ventura I, Aroca R, Blanca M, Bergstralh DT, Monteseirin J. Histamine production by human neutrophils. FASEB J 2013; 27:2902-10. [PMID: 23572231 DOI: 10.1096/fj.12-223867] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Histamine is an important mediator in the development of allergic reactions. Only a small subset of human cell types is able to produce histamine. No previous studies have shown that human neutrophils are among them. The present work was undertaken to analyze whether human neutrophils produce histamine, and to determine what agonists are involved in histamine production by human neutrophils. The expression of histidine decarboxylase in human neutrophils was established by quantitative PCR, Western blotting, and flow cytometry analysis. The activity of the enzyme was determined by ELISA, which measured histamine in the culture supernatant of neutrophils stimulated with a set of classical agonists. Human neutrophils are bona fide histamine-producing cells. Neutrophils store ∼0.29 pg/cell and release ∼50% of the histamine content in an antigen-dependent manner and on stimulation with other neutrophil agonists. Basal expression of histidine decarboxylase, the rate-limiting enzyme in histamine production, is higher in neutrophils from patients with allergies than from healthy donors. Our results cannot be ascribed to cell contamination for several reasons. LPS failed to induce histamine release by basophils, whereas it induced histamine release by neutrophils; and we did not detect basophils, monocytes, or lymphocytes in our neutrophil preparations. Eosinophils, albeit detected, were only 0.001-0.004% of the final cell population, and they did not store or release histamine on antigen or LPS stimulation. Antigens to which patients with allergies were sensitized stimulated release of histamine from neutrophils. These observations represent a novel view of neutrophils as possible source of histamine in the allergic diseases.
Collapse
Affiliation(s)
- Lorena Alcañiz
- Servicio Regional de Inmunología y Alergia, Hospital Universitario Virgen Macarena, Seville, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Tourneur E, Ben Mkaddem S, Chassin C, Bens M, Goujon JM, Charles N, Pellefigues C, Aloulou M, Hertig A, Monteiro RC, Girardin SE, Philpott DJ, Rondeau E, Elbim C, Werts C, Vandewalle A. Cyclosporine A impairs nucleotide binding oligomerization domain (Nod1)-mediated innate antibacterial renal defenses in mice and human transplant recipients. PLoS Pathog 2013; 9:e1003152. [PMID: 23382681 PMCID: PMC3561241 DOI: 10.1371/journal.ppat.1003152] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 12/08/2012] [Indexed: 12/12/2022] Open
Abstract
Acute pyelonephritis (APN), which is mainly caused by uropathogenic Escherichia coli (UPEC), is the most common bacterial complication in renal transplant recipients receiving immunosuppressive treatment. However, it remains unclear how immunosuppressive drugs, such as the calcineurin inhibitor cyclosporine A (CsA), decrease renal resistance to UPEC. Here, we investigated the effects of CsA in host defense against UPEC in an experimental model of APN. We show that CsA-treated mice exhibit impaired production of the chemoattractant chemokines CXCL2 and CXCL1, decreased intrarenal recruitment of neutrophils, and greater susceptibility to UPEC than vehicle-treated mice. Strikingly, renal expression of Toll-like receptor 4 (Tlr4) and nucleotide-binding oligomerization domain 1 (Nod1), neutrophil migration capacity, and phagocytic killing of E. coli were significantly reduced in CsA-treated mice. CsA inhibited lipopolysaccharide (LPS)-induced, Tlr4-mediated production of CXCL2 by epithelial collecting duct cells. In addition, CsA markedly inhibited Nod1 expression in neutrophils, macrophages, and renal dendritic cells. CsA, acting through inhibition of the nuclear factor of activated T-cells (NFATs), also markedly downregulated Nod1 in neutrophils and macrophages. Silencing the NFATc1 isoform mRNA, similar to CsA, downregulated Nod1 expression in macrophages, and administration of the 11R-VIVIT peptide inhibitor of NFATs to mice also reduced neutrophil bacterial phagocytosis and renal resistance to UPEC. Conversely, synthetic Nod1 stimulating agonists given to CsA-treated mice significantly increased renal resistance to UPEC. Renal transplant recipients receiving CsA exhibited similar decrease in NOD1 expression and neutrophil phagocytosis of E. coli. The findings suggest that such mechanism of NFATc1-dependent inhibition of Nod1-mediated innate immune response together with the decrease in Tlr4-mediated production of chemoattractant chemokines caused by CsA may contribute to sensitizing kidney grafts to APN.
Collapse
Affiliation(s)
- Emilie Tourneur
- INSERM U773, Centre de Recherche Biomédicale Bichat Beaujon, Université Paris 7 - Denis Diderot, Paris, France
| | - Sanae Ben Mkaddem
- INSERM U699, Paris, France; Université Paris 7 - Denis Diderot, Paris, France
| | - Cécilia Chassin
- INSERM U699, Paris, France; Université Paris 7 - Denis Diderot, Paris, France
| | - Marcelle Bens
- INSERM U773, Centre de Recherche Biomédicale Bichat Beaujon, Université Paris 7 - Denis Diderot, Paris, France
| | - Jean-Michel Goujon
- Université de Poitiers, CHU Poitiers; Service d'Anatomie et Cytologie Pathologiques, Poitiers, France
| | - Nicolas Charles
- INSERM U699, Paris, France; Université Paris 7 - Denis Diderot, Paris, France
| | | | - Meryem Aloulou
- INSERM U699, Paris, France; Université Paris 7 - Denis Diderot, Paris, France
| | - Alexandre Hertig
- Service Urgences Néphrologiques et Transplantation Rénale and INSERM U702, Hôpital Tenon; Université Paris 6 - Pierre et Marie Curie, Paris, France
| | - Renato C. Monteiro
- INSERM U699, Paris, France; Université Paris 7 - Denis Diderot, Paris, France
| | - Stephen E. Girardin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada
| | - Dana J. Philpott
- Department of Immunology, University of Toronto, Toronto, Canada
| | - Eric Rondeau
- Service Urgences Néphrologiques et Transplantation Rénale and INSERM U702, Hôpital Tenon; Université Paris 6 - Pierre et Marie Curie, Paris, France
| | - Carole Elbim
- INSERM UMR-S 945, Hôpital Pitié-Salpêtrière, Université Paris 6 - Pierre et Marie Curie, Paris, France
| | - Catherine Werts
- Institut Pasteur, G5 Biologie et Génétique des Parois Bactériennes, Paris, France
| | - Alain Vandewalle
- INSERM U773, Centre de Recherche Biomédicale Bichat Beaujon, Université Paris 7 - Denis Diderot, Paris, France
- * E-mail:
| |
Collapse
|
21
|
Abstract
The calcineurin/nuclear factor of activated T cells (NFAT) signaling pathway mediates multiple adaptive T-cell functions, but recent studies have shown that calcineurin/NFAT signaling also contributes to innate immunity and regulates the homeostasis of innate cells. Myeloid cells, including granulocytes and dendritic cells, can promote inflammation, regulate adaptive immunity, and are essential mediators of early responses to pathogens. Microbial ligation of pattern-recognition receptors, such as TLR4, CD14, and dectin 1, is now known to induce the activation of calcineurin/NFAT signaling in myeloid cells, a finding that has provided new insights into the molecular pathways that regulate host protection. Inhibitors of calcineurin/NFAT binding, such as cyclosporine A and FK506, are broadly used in organ transplantation and can act as potent immunosuppressive drugs in a variety of different disorders. There is increasing evidence that these agents influence innate responses as well as inhibiting adaptive T-cell functions. This review focuses on the role of calcineurin/NFAT signaling in myeloid cells, which may contribute to the various unexplained effects of immunosuppressive drugs already being used in the clinic.
Collapse
|
22
|
Kawarabayashi Y, Hai L, Honda A, Horiuchi S, Tsujioka H, Ichikawa J, Inoue R. Critical role of TRPC1-mediated Ca²⁺ entry in decidualization of human endometrial stromal cells. Mol Endocrinol 2012; 26:846-58. [PMID: 22474110 PMCID: PMC5417103 DOI: 10.1210/me.2011-1259] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 02/28/2012] [Indexed: 12/23/2022] Open
Abstract
Decidualization is an ovarian steroid-induced remodeling/differentiation process of uterus essential for embryo implantation and placentation. Here, we investigated the possible involvement of enhanced Ca²⁺ dynamics in the decidualization process in human endometrial stromal cells (hESC) in its connection with a recently emerging nonvoltage-gated Ca²⁺ entry channel superfamily, the transient receptor potential (TRP) protein. Combined application of 17β-estradiol (E₂) (10 nM) and progesterone (P₄) (1 μM) for 7-14 d resulted in morphological changes of hESC characteristic of decidualization (i.e. cell size increase), whereas sole application of E₂ exerted little effects. A 7- to 14-d E₂/P₄ treatment greatly increased the expression level of decidualization markers IGF binding protein-1 (IGFBP-1) and prolactin and also up-regulated the expression of TRPC1, a canonical TRP subfamily member that has been implicated in store-operated Ca²⁺ influx (SOC) in other cell types. In parallel with this up-regulation, SOC activity in hESC, the nuclear translocation of phosphorylated cAMP responsive element binding protein (p-CREB) and the expression of Forkhead box protein 01 were enhanced significantly. Small interfering RNA knockdown of TRPC1 counteracted the E₂/P₄-induced up-regulation of IGFBP-1 and prolactin and enhancement of SOC activity together with the inhibition of hESC size increase, p-CREB nuclear translocation, and FOXO1 up-regulation. Coadministration of SOC inhibitors SK&F96365 or Gd³⁺ with E₂/P₄ also suppressed the up-regulation of IGFBP-1 and hESC size increase. Similar inhibitory effects were observed with extracellularly applied TRPC1 extracellular loop 3-directed antibody, which is known to bind a near-pore domain of TRPC1 channel and block its Ca²⁺ transporting activity. These results strongly suggest that up-regulation of TRPC1 protein and consequent enhancement of SOC-mediated Ca²⁺ influx may serve as a crucial step for the decidualization process of hESC probably via p-CREB-dependent transcriptional activity associated with FOXO1 activation.
Collapse
|
23
|
Interleukin(IL)-4 promotion of CXCL-8 gene transcription is mediated by ERK1/2 pathway in human pulmonary artery endothelial cells. Mol Immunol 2011; 48:1784-92. [PMID: 21645924 DOI: 10.1016/j.molimm.2011.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 04/27/2011] [Accepted: 05/03/2011] [Indexed: 01/07/2023]
Abstract
Interleukin-4 is central to allergic pulmonary inflammatory responses, but its contribution to airway neutrophilia remains controversial. The endothelium plays a critical role in regulating leukocyte recruitment and migration during inflammation. However, its response to IL-4 is reported to either increase or decrease the production of neutrophil chemotactic factors. We hypothesized that these conflicting findings may be due to the tissue and the size of the vessels from which endothelial cells have been derived. The expression of CXCL-8 by human primary culture umbilical veins endothelial cells (HUVECs), human pulmonary artery endothelial cells (HPAECs), and human pulmonary microvascular endothelial cells (HPMECs) when stimulated with recombinant human IL-4 (rhIL-4) was studied. The chemoattractant property of the cells' supernatants for neutrophils was evaluated using Boyden chambers. The role of the nuclear factor-κB (NF-κB), and mitogen-activated protein kinases (MAPK) in IL-4-induced HPAECs was studied using Western blotting and electrophoretic mobility shift assay (EMSA). We demonstrated that IL-4 increased the mRNA expression and the protein production of CXCL-8 in HPAECs, but not in HUVECs and HPMECs. The supernatants of HAPECs stimulated by IL-4 significantly promoted neutrophils migration in a dose-dependent manner, and was significantly attenuated by an inhibitor of CXCL-8. We also found that extracellular-regulated protein kinase1/2 (ERK1/2) is activated by IL-4 in HPAECs, but not JUN-N-terminal protein kinase (JNK) or p38 MAPK pathway. Furthermore, NF-κB-DNA binding activity, phosphorylation of IκBα and p65 levels were not affected by rhIL-4 in HAPECs. These findings indicate marked functional differences in the response of micro and macro-ECs to IL-4. ERK1/2, rather than NF-κB, JNK and p38 MAPK signaling, plays a role in IL-4 induced chemokine activation. Our results suggest that inhibition of ERK1/2 may be a possible target for airway neutrophilia in allergic lung diseases.
Collapse
|
24
|
Post interventional cardiology urinary thromboxane correlates with PlateletMapping® detected aspirin resistance. Thromb Res 2010; 125:e118-22. [DOI: 10.1016/j.thromres.2009.11.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 10/14/2009] [Accepted: 11/18/2009] [Indexed: 11/17/2022]
|
25
|
Mora J, Riggs EK, Fu J, MacGlashan DW, Fox SA, Yu B, Tobin MC, Thomas LL. Expression of the high affinity IgE receptor by neutrophils of individuals with allergic asthma is both minimal and insensitive to regulation by serum IgE. Clin Immunol 2009; 132:132-40. [PMID: 19359220 PMCID: PMC2739590 DOI: 10.1016/j.clim.2009.03.513] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2009] [Revised: 02/26/2009] [Accepted: 03/16/2009] [Indexed: 01/08/2023]
Abstract
We evaluated the hypothesis that serum IgE regulates neutrophil FcepsilonRI expression in the same manner as described for other FcepsilonRI+ cells. FcepsilonRI expression by neutrophils of 40 asthma subjects and 20 control subjects did not correlate with serum IgE levels, whereas FcepsilonRI expression by basophils of the same subjects showed a highly significant correlation. The level of FcepsilonRI expression by neutrophils of both asthma and control subjects was approximately 1% of that for basophil FcepsilonRI expression. IgE+ neutrophils were minimally detectable, and FcepsilonRI alpha-subunit was not detected in Western blots of neutrophil membranes and cytosol. The neutrophil FcepsilonRI did not support anti-IgE stimulated superoxide release or IgE-induced increase in neutrophil survival. We conclude that FcepsilonRI expression by neutrophils of both asthma patients and control individuals is minimal at best and that, if present, neutrophil FcepsilonRI expression, unlike that of other human FcepsilonRI+ cells, is not regulated by serum IgE.
Collapse
Affiliation(s)
- Juanita Mora
- Department of Immunology/Microbiology, Section of Allergy and Immunology, Rush University Medical Center, Chicago, IL
| | - Emily K. Riggs
- Department of Immunology/Microbiology, Section of Allergy and Immunology, Rush University Medical Center, Chicago, IL
| | - Jun Fu
- Department of Immunology/Microbiology, Section of Allergy and Immunology, Rush University Medical Center, Chicago, IL
| | - Donald W. MacGlashan
- Department of Medicine, Division of Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Susan A. Fox
- Department of Immunology/Microbiology, Section of Allergy and Immunology, Rush University Medical Center, Chicago, IL
| | - Byung Yu
- Department of Immunology/Microbiology, Section of Allergy and Immunology, Rush University Medical Center, Chicago, IL
- John H. Stroger Hospital of Cook County, Chicago, IL
| | - Mary C. Tobin
- Department of Immunology/Microbiology, Section of Allergy and Immunology, Rush University Medical Center, Chicago, IL
| | - Larry L. Thomas
- Department of Immunology/Microbiology, Section of Allergy and Immunology, Rush University Medical Center, Chicago, IL
| |
Collapse
|