1
|
Frasconi TM, Kurts C, Dhana E, Kaiser R, Reichelt M, Lukacs-Kornek V, Boor P, Hauser AE, Pascual-Reguant A, Bedoui S, Turner JE, Becker-Gotot J, Ludwig-Portugall I. Renal IL-23-Dependent Type 3 Innate Lymphoid Cells Link Crystal-induced Intrarenal Inflammasome Activation with Kidney Fibrosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:865-875. [PMID: 39072698 PMCID: PMC11372247 DOI: 10.4049/jimmunol.2400041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Chronic inflammasome activation in mononuclear phagocytes (MNPs) promotes fibrosis in various tissues, including the kidney. The cellular and molecular links between the inflammasome and fibrosis are unclear. To address this question, we fed mice lacking various immunological mediators an adenine-enriched diet, which causes crystal precipitation in renal tubules, crystal-induced inflammasome activation, and renal fibrosis. We found that kidney fibrosis depended on an intrarenal inflammasome-dependent type 3 immune response driven by its signature transcription factor Rorc (retinoic acid receptor-related orphan receptor C gene), which was partially carried out by type 3 innate lymphoid cells (ILC3s). The role of ILCs in the kidney is less well known than in other organs, especially that of ILC3. In this article, we describe that depletion of ILCs or genetic deficiency for Rorc attenuated kidney inflammation and fibrosis. Among the inflammasome-derived cytokines, only IL-1β expanded ILC3 and promoted fibrosis, whereas IL-18 caused differentiation of NKp46+ ILC3. Deficiency of the type 3 maintenance cytokine, IL-23, was more protective than IL-1β inhibition, which may be explained by the downregulation of the IL-1R, but not of the IL-23R, by ILC3 early in the disease, allowing persistent sensing of IL-23. Mechanistically, ILC3s colocalized with renal MNPs in vivo as shown by multiepitope-ligand cartography. Cell culture experiments indicated that renal ILC3s caused renal MNPs to increase TGF-β production that stimulated fibroblasts to produce collagen. We conclude that ILC3s link inflammasome activation with kidney inflammation and fibrosis and are regulated by IL-1β and IL-23.
Collapse
Affiliation(s)
- Teresa M Frasconi
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Ermanila Dhana
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Romina Kaiser
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Miriam Reichelt
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Veronika Lukacs-Kornek
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Peter Boor
- Institute of Pathology, Department of Nephrology, RWTH University, Aachen, Germany
| | - Anja E Hauser
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum, Leibniz Institute, Berlin, Germany
| | - Anna Pascual-Reguant
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum, Leibniz Institute, Berlin, Germany
| | - Sammy Bedoui
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Jan-Eric Turner
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Janine Becker-Gotot
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Isis Ludwig-Portugall
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
2
|
Kim HM, Kang YM, Lee M, An HJ. Papain Suppresses Atopic Skin Inflammation through Anti-Inflammatory Activities Using In Vitro and In Vivo Models. Antioxidants (Basel) 2024; 13:928. [PMID: 39199175 PMCID: PMC11351312 DOI: 10.3390/antiox13080928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
Papain (PN) is a proteolytic enzyme derived from Carica Papaya L. While the pharmacological effects of PN have not been extensively studied compared to its enzymatic activity, PN also holds potential benefits beyond protein digestion. This study aimed to investigate the potential effects of PN against skin inflammation in house dust mite Dermatophagoides farinae body (Dfb)-exposed NC/Nga atopic dermatitis (AD) mice and human HaCaT keratinocytes and their underlying mechanisms. The effects of PN on the skin were assessed via histological examination, measurements of transepidermal water loss (TEWL), quantitative reverse transcription-polymerase chain reaction, Western blotting, and enzyme-linked immunosorbent assay. Our findings indicated that the oral intake of PN decreased the severity scores of lesions resembling AD, TEWL, and the levels of inflammatory cytokines and serum immunoglobulin E in Dfb-induced AD mice, along with a reduction in epidermal thickness and mast cell infiltration. Additionally, PN inhibited the activation of the mitogen-activated protein kinases (MAPKs) and the signal transducer and activator of transcription (STAT) pathways in Dfb-induced AD mice and HaCaT keratinocytes. Moreover, PN improved survival and reduced ROS production in H2O2-damaged HaCaT keratinocytes and enhanced the expression of antioxidant enzymes in Dfb-induced AD mice. Concludingly, the oral administration of PN suppressed inflammatory mediators and downregulated the MAPKs/STAT pathway, suggesting its potential role in AD pathogenesis.
Collapse
Affiliation(s)
- Hye-Min Kim
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Herbology, College of Korean Medicine, Sangji University, Wonju 26339, Republic of Korea;
| | - Yun-Mi Kang
- Department of Herbology, College of Korean Medicine, Sangji University, Wonju 26339, Republic of Korea;
- Korean Medicine (KM)-Application Center, Korea Institute of Oriental Medicine (KIOM), 70 Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Minho Lee
- Department of Life Science, Dongguk University-Seoul, Ilsandong-gu, Goyang-si 10326, Republic of Korea
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
3
|
Deng Y, Ding C, Yang H, Zhang M, Xiao Y, Wang H, Li J, Xiao T, Lv Z. First in vitro and in vivo evaluation of recombinant IL-1β protein as a potential immunomodulator against viral infection in fish. Int J Biol Macromol 2024; 255:128192. [PMID: 37979760 DOI: 10.1016/j.ijbiomac.2023.128192] [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: 07/08/2023] [Revised: 10/28/2023] [Accepted: 11/15/2023] [Indexed: 11/20/2023]
Abstract
IL-1β is an important proinflammatory cytokine with multifaceted modulatory roles in immune responses. In fish, recombinant IL-1β has been employed in the control of bacterial diseases, while the antiviral mechanisms of IL-1β remain largely unknown, and the efficacy of recombinant IL-1β as an immunomodulator to prevent viral diseases is still not determined. This study evaluated the immunomodulatory effects of recombinant grass carp IL-1β against grass carp reovirus (GCRV) in vitro and in vivo. Firstly, the mature form (Ser111-Lys270) of grass carp IL-1β was identified, and its recombinant protein (designated as rgcIL-1β) was prepared through prokaryotic expression. Then, an in vitro evaluation model for rgcIL-1β activity was established in the CIK cells, with the appropriate concentration (600 ng/mL) and effect time (1 h). In vitro, rgcIL-1β could not only induce the production of proinflammatory cytokines such as IL-1β, IL-6, IL-8, and TNF-α but also a series of antiviral factors including IFN-1, IFN-2, IFN-γ, and ISG15. Mechanistically, transcriptome analysis and western blotting confirmed that rgcIL-1β activated multiple transcriptional factors, including NF-κB, IRF1, IRF3, and IRF8, and the signal pathways associated with inflammatory cytokines and antiviral factors expression. Expectedly, rgcIL-1β treatment significantly inhibited GCRV replication in vitro. In vivo administration of rgcIL-1β via intraperitoneal pre-injection significantly aroused an antiviral response to restrict GCRV replication and intense tissue inflammation in grass carp, demonstrating the immunomodulatory effects of rgcIL-1β. More importantly, rgcIL-1β administrated with 10 ng/g and 1 ng/g could improve the survival rate of grass carp during GCRV infection. This study represents the first time to comprehensively reveal the immunomodulatory and antiviral mechanisms of IL-1β in fish and may also pave the way for further developing recombinant IL-1β as an immunotherapy for the prevention and control of fish viral diseases.
Collapse
Affiliation(s)
- Yadong Deng
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China
| | - Chunhua Ding
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China
| | - Hong Yang
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China
| | - Mengyuan Zhang
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China
| | - Yu Xiao
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China
| | - Hongquan Wang
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China
| | - Junhua Li
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China
| | - Tiaoyi Xiao
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China.
| | - Zhao Lv
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
4
|
Martínez-Gómez LE, Martinez-Armenta C, Medina-Luna D, Ordoñez-Sánchez ML, Tusie-Luna T, Ortega-Peña S, Herrera-López B, Suarez-Ahedo C, Jimenez-Gutierrez GE, Hidalgo-Bravo A, Vázquez-Cárdenas P, Vidal-Vázquez RP, Ramírez-Hinojosa JP, Martinez Matsumoto PM, Vargas-Alarcón G, Posadas-Sánchez R, Fragoso JM, Martínez-Ruiz FDJ, Zayago-Angeles DM, Mata-Miranda MM, Vázquez-Zapién GJ, Martínez-Cuazitl A, Andrade-Alvarado J, Granados J, Ramos-Tavera L, Camacho-Rea MDC, Segura-Kato Y, Rodríguez-Pérez JM, Coronado-Zarco R, Franco-Cendejas R, López-Jácome LE, Magaña JJ, Vela-Amieva M, Pineda C, Martínez-Nava GA, López-Reyes A. Implication of myddosome complex genetic variants in outcome severity of COVID-19 patients. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2023; 56:939-950. [PMID: 37365052 PMCID: PMC10273757 DOI: 10.1016/j.jmii.2023.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 03/31/2023] [Accepted: 06/10/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND/PURPOSE(S) During a viral infection, the immune response is mediated by the toll-like receptors and myeloid differentiation Factor 88 (MyD88) that play an important role sensing infections such as SARS-CoV-2 which has claimed the lives of more than 6.8 million people around the world. METHODS We carried out a cross-sectional with a population of 618 SARS-CoV-2-positive unvaccinated subjects and further classified based on severity: 22% were mild, 34% were severe, 26% were critical, and 18% were deceased. Toll Like Receptor 7 (TLR7) single-nucleotide polymorphisms (rs3853839, rs179008, rs179009, and rs2302267) and MyD88 (rs7744) were genotyped using TaqMan OpenArray. The association of polymorphisms with disease outcomes was performed by logistic regression analysis adjusted by covariates. RESULTS A significant association of rs3853839 and rs7744 of the TLR7 and MyD88 genes, respectively, was found with COVID-19 severity. The G/G genotype of the rs3853839 TLR7 was associated with the critical outcome showing an Odd Ratio = 1.98 (95% IC = 1.04-3.77). The results highlighted an association of the G allele of MyD88 gene with severe, critical and deceased outcomes. Furthermore, in the dominant model (AG + GG vs. AA), we observed an Odd Ratio = 1.70 (95% CI = 1.02-2.86) with severe, Odd Ratio = 1.82 (95% CI = 1.04-3.21) with critical, and Odd Ratio = 2.44 (95% CI = 1.21-4.9) with deceased outcomes. CONCLUSION To our knowledge this work represents an innovative report that highlights the significant association of TLR7 and MyD88 gene polymorphisms with COVID-19 outcomes and the possible implication of the MyD88 variant with D-dimer and IFN-α concentrations.
Collapse
Affiliation(s)
- Laura E Martínez-Gómez
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Carlos Martinez-Armenta
- Graduate Program in Experimental Biology, Dirección de Ciencias Biológicas y de la Salud (DCBS), Universidad Autónoma Metropolitana Iztapalapa, Ciudad de México, Mexico.
| | - Daniel Medina-Luna
- Microbiology & Immunology Department, Dalhousie University, Halifax, B3H4R2, Nova Scotia, Canada.
| | - María Luisa Ordoñez-Sánchez
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico.
| | - Tere Tusie-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico; Instituto de Investigaciones Biomédicas Universidad Nacional Autónoma de México, Mexico City, Mexico.
| | - Silvestre Ortega-Peña
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Brígida Herrera-López
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Carlos Suarez-Ahedo
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Guadalupe Elizabeth Jimenez-Gutierrez
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Alberto Hidalgo-Bravo
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Paola Vázquez-Cárdenas
- Centro de Innovación Médica Aplicada, Hospital General Dr. Manuel Gea González, Ciudad de México, Mexico.
| | - Rosa P Vidal-Vázquez
- Centro de Innovación Médica Aplicada, Hospital General Dr. Manuel Gea González, Ciudad de México, Mexico.
| | - Juan P Ramírez-Hinojosa
- Centro de Innovación Médica Aplicada, Hospital General Dr. Manuel Gea González, Ciudad de México, Mexico.
| | | | - Gilberto Vargas-Alarcón
- Departamento de Biología Molecular y Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico.
| | - Rosalinda Posadas-Sánchez
- Departamento de Biología Molecular y Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico.
| | - José-Manuel Fragoso
- Departamento de Biología Molecular y Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico.
| | | | | | - Mónica Maribel Mata-Miranda
- Laboratorio de Biología Celular y Tisular, Laboratorio de Embriología, Escuela Médico Militar, Universidad del Ejército y Fuerza Aérea, Ciudad de México, Mexico.
| | - Gustavo Jesús Vázquez-Zapién
- Laboratorio de Biología Celular y Tisular, Laboratorio de Embriología, Escuela Médico Militar, Universidad del Ejército y Fuerza Aérea, Ciudad de México, Mexico.
| | - Adriana Martínez-Cuazitl
- Laboratorio de Biología Celular y Tisular, Laboratorio de Embriología, Escuela Médico Militar, Universidad del Ejército y Fuerza Aérea, Ciudad de México, Mexico.
| | - Javier Andrade-Alvarado
- Servicio de Cirugía General, Hospital Central Norte Petróleos Mexicanos (PEMEX), Estado de México, Mexico.
| | - Julio Granados
- Departamento de Inmunogenética, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Secretaría de Salud, Mexico City, Mexico.
| | - Luis Ramos-Tavera
- Departamento de Inmunogenética, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Secretaría de Salud, Mexico City, Mexico.
| | - María Del Carmen Camacho-Rea
- Departamento de Nutrición Animal, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Secretaría de Salud, Mexico City, Mexico
| | - Yayoi Segura-Kato
- Unidad de Biología Molecular y Medicina Genómica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico.
| | - José Manuel Rodríguez-Pérez
- Departamento de Biología Molecular y Endocrinología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, Mexico.
| | - Roberto Coronado-Zarco
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Rafael Franco-Cendejas
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Luis Esau López-Jácome
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Jonathan J Magaña
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Marcela Vela-Amieva
- Laboratorio de Errores Innatos del Metabolismo y Tamiz, Instituto Nacional de Pediatría, Secretaria de Salud, Ciudad de México, Mexico.
| | - Carlos Pineda
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Gabriela Angélica Martínez-Nava
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| | - Alberto López-Reyes
- Laboratorio de Gerociencias, Dirección General, Medicina de Rehabilitación, Laboratorio de Infectología, Departamento de Reconstrucción Articular, Laboratorio de Medicina Genómica, Laboratorio Facilitador. Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Secretaría de Salud, Ciudad de México, Mexico.
| |
Collapse
|
5
|
Carignon S, De Moura Rodrigues D, Gosset D, Culerier E, Huot-Marchand S, Savigny F, Kaya E, Quesniaux V, Gombault A, Couillin I, Ryffel B, Le Bert M, Riteau N. Lung inflammation and interstitial fibrosis by targeted alveolar epithelial type I cell death. Front Immunol 2023; 14:1261483. [PMID: 37841243 PMCID: PMC10568624 DOI: 10.3389/fimmu.2023.1261483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Introduction The pathogenesis of chronic lung diseases is multifaceted with a major role of recurrent micro-injuries of the epithelium. While several reports clearly indicated a prominent role for surfactant-producing alveolar epithelial type 2 (AT2) cells, the contribution of gas exchange-permissive alveolar epithelial type 1 (AT1) cells has not been addressed yet. Here, we investigated whether repeated injury of AT1 cells leads to inflammation and interstitial fibrosis. Methods We chose an inducible model of AT1 cell depletion following local diphtheria toxin (DT) administration using an iDTR flox/flox (idTRfl/fl) X Aquaporin 5CRE (Aqp5CRE) transgenic mouse strain. Results We investigated repeated doses and intervals of DT to induce cell death of AT1 cells causing inflammation and interstitial fibrosis. We found that repeated DT administrations at 1ng in iDTRfl/fl X Aqp5CRE mice cause AT1 cell death leading to inflammation, increased tissue repair markers and interstitial pulmonary fibrosis. Discussion Together, we demonstrate that depletion of AT1 cells using repeated injury represents a novel approach to investigate chronic lung inflammatory diseases and to identify new therapeutic targets.
Collapse
Affiliation(s)
- Sandra Carignon
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - Dorian De Moura Rodrigues
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - David Gosset
- Center for Molecular Biophysics, CNRS Unité propre de recherche 4301, Orleans, France
| | - Elodie Culerier
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - Sarah Huot-Marchand
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - Florence Savigny
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - Eric Kaya
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - Valerie Quesniaux
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - Aurélie Gombault
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - Isabelle Couillin
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - Bernhard Ryffel
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - Marc Le Bert
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| | - Nicolas Riteau
- University of Orleans and CNRS, Immunologie et Neurogénétique Expérimentales et Moléculaires -UMR7355, Orleans, France
| |
Collapse
|
6
|
Zhu S, Zhong S, Cheng K, Zhang LS, Bai JW, Cao Z, Wang S, Chen W, Cheng S, Ma L, Ling Z, Huang Y, Gu W, Sun X, Yi C, Zhao M, Liang S, Xu JF, Sun B, Zhang Y. Vitamin B6 regulates IL-33 homeostasis to alleviate type 2 inflammation. Cell Mol Immunol 2023; 20:794-807. [PMID: 37217797 PMCID: PMC10310729 DOI: 10.1038/s41423-023-01029-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Interleukin-33 (IL-33) is a crucial nuclear cytokine that induces the type 2 immune response and maintains immune homeostasis. The fine-tuned regulation of IL-33 in tissue cells is critical to control of the type 2 immune response in airway inflammation, but the mechanism is still unclear. Here, we found that healthy individuals had higher phosphate-pyridoxal (PLP, an active form of vitamin B6) concentrations in the serum than asthma patients. Lower serum PLP concentrations in asthma patients were strongly associated with worse lung function and inflammation. In a mouse model of lung inflammation, we revealed that PLP alleviated the type 2 immune response and that this inhibitory effect relied on the activity of IL-33. A mechanistic study showed that in vivo, pyridoxal (PL) needed to be converted into PLP, which inhibited the type 2 response by regulating IL-33 stability. In mice heterozygous for pyridoxal kinase (PDXK), the conversion of PL to PLP was limited, and IL-33 levels were increased in the lungs, aggravating type 2 inflammation. Furthermore, we found that the mouse double minute 2 homolog (MDM2) protein, an E3 ubiquitin-protein ligase, could ubiquitinate the N-terminus of IL-33 and sustain IL-33 stability in epithelial cells. PLP reduced MDM2-mediated IL-33 polyubiquitination and decreased the level of IL-33 through the proteasome pathway. In addition, inhalation of PLP alleviated asthma-related effects in mouse models. In summary, our data indicate that vitamin B6 regulates MDM2-mediated IL-33 stability to constrain the type 2 response, which might help develop a potential preventive and therapeutic agent for allergy-related diseases.
Collapse
Affiliation(s)
- Songling Zhu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Shufen Zhong
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Kebin Cheng
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Li-Sha Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Jiu-Wu Bai
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Zu Cao
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Su Wang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Wen Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Shipeng Cheng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Liyan Ma
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Zhiyang Ling
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Yuying Huang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Wangpeng Gu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Xiaoyu Sun
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Chunyan Yi
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Meng Zhao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Shuo Liang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China.
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China.
| | - Bing Sun
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China.
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| | - Yaguang Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| |
Collapse
|
7
|
Chen P, Zhou Y, Li X, Yang J, Zheng Z, Zou Y, Li X, Liao J, Dai J, Xu Y, Yin L, Chen G, Gu J, Ouyang Q, Cho WJ, Tang Q, Liang G. Design, Synthesis, and Bioevaluation of Novel MyD88 Inhibitor c17 against Acute Lung Injury Derived from the Virtual Screen. J Med Chem 2023; 66:6938-6958. [PMID: 37130331 DOI: 10.1021/acs.jmedchem.3c00359] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Myeloid differentiation primary response protein 88 (MyD88) is crucial to immune cascades mediated by Toll-like receptors (TLRs) and interleukin-1 receptors (IL-1Rs). MyD88 dysregulation has been linked to a wide variety of inflammatory diseases, making it a promising new target for anti-inflammatory and cancer therapy development. In this study, 46 compounds were designed and synthesized inspired by virtual screen hit. The anti-inflammatory activity of designed compounds was evaluated biologically, and c17 was discovered to have a high binding affinity with MyD88. It inhibited the interaction of TLR4 and MyD88 and suppressed the NF-κB pathway. In addition, c17 treatment led to the accumulation in the lungs of rats and attenuated LPS-induced ALI mice model. Furthermore, c17 showed negligible toxicity in vivo. Together, these findings suggest that c17 may serve as a potential therapeutical method for the treatment of ALI and as a lead structure for the continued development of MyD88 inhibitors.
Collapse
Affiliation(s)
- Pan Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, China
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Ying Zhou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Xiaobo Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Jun Yang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Zhiwei Zheng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yu Zou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Xiang Li
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Jing Liao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Jintian Dai
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yuye Xu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Lina Yin
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, China
| | - Gaozhi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jing Gu
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing 400038, China
| | - Won-Jea Cho
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea
| | - Qidong Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 310053, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| |
Collapse
|
8
|
Sylvester M, Son A, Schwartz DM. The Interactions Between Autoinflammation and Type 2 Immunity: From Mechanistic Studies to Epidemiologic Associations. Front Immunol 2022; 13:818039. [PMID: 35281022 PMCID: PMC8907424 DOI: 10.3389/fimmu.2022.818039] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 02/02/2022] [Indexed: 12/30/2022] Open
Abstract
Autoinflammatory diseases are a group of clinical syndromes characterized by constitutive overactivation of innate immune pathways. This results in increased production of or responses to monocyte- and neutrophil-derived cytokines such as interleukin-1β (IL-1β), Tumor Necrosis Factor-α (TNF-α), and Type 1 interferon (IFN). By contrast, clinical allergy is caused by dysregulated type 2 immunity, which is characterized by expansion of T helper 2 (Th2) cells and eosinophils, as well as overproduction of the associated cytokines IL-4, IL-5, IL-9, and IL-13. Traditionally, type 2 immune cells and autoinflammatory effectors were thought to counter-regulate each other. However, an expanding body of evidence suggests that, in some contexts, autoinflammatory pathways and cytokines may potentiate type 2 immune responses. Conversely, type 2 immune cells and cytokines can regulate autoinflammatory responses in complex and context-dependent manners. Here, we introduce the concepts of autoinflammation and type 2 immunity. We proceed to review the mechanisms by which autoinflammatory and type 2 immune responses can modulate each other. Finally, we discuss the epidemiology of type 2 immunity and clinical allergy in several monogenic and complex autoinflammatory diseases. In the future, these interactions between type 2 immunity and autoinflammation may help to expand the spectrum of autoinflammation and to guide the management of patients with various autoinflammatory and allergic diseases.
Collapse
Affiliation(s)
- McKella Sylvester
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Aran Son
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniella M Schwartz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
9
|
Matsushita K, Tanaka H, Yasuda K, Adachi T, Fukuoka A, Akasaki S, Koida A, Kuroda E, Akira S, Yoshimoto T. Regnase-1 degradation is crucial for IL-33- and IL-25-mediated ILC2 activation. JCI Insight 2020; 5:131480. [PMID: 31990689 DOI: 10.1172/jci.insight.131480] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 01/22/2020] [Indexed: 12/22/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are a critical innate source of type 2 cytokines in allergic inflammation. Although ILC2s are recognized as a critical cell population in the allergic inflammation, the regulatory mechanism(s) of ILC2s are less well understood. Here, we show that Regnase-1, an immune regulatory RNAse that degrades inflammatory mRNAs, negatively regulates ILC2 function and that IκB kinase (IKK) complex-mediated Regnase-1 degradation is essential for IL-33- and IL-25-induced ILC2 activation. ILC2s from Regnase-1AA/AA mice expressing a Regnase-1 S435A/S439A mutant resistant to IKK complex-mediated degradation accumulated Regnase-1 protein in response to IL-33 and IL-25. IL-33- and IL-25-stimulated Regnase-1AA/AA ILC2s showed reduced cell proliferation and type 2 cytokine (IL-5, IL-9, and IL-13) production and increased cell death. In addition, Il2ra and Il1rl1, but not Il5, Il9, or Il13, mRNAs were destabilized in IL-33-stimulated Regnase-1AA/AA ILC2s. In vivo, Regnase-1AA/AA mice showed attenuated acute type 2 pulmonary inflammation induced by the instillation of IL-33, IL-25, or papain. Furthermore, the expulsion of Nippostrongylus brasiliensis was significantly delayed in Regnase-1AA/AA mice. These results demonstrate that IKK complex-mediated Regnase-1 degradation is essential for ILC2-mediated type 2 responses both in vitro and in vivo. Therefore, controlling Regnase-1 degradation is a potential therapeutic target for ILC2-contributed allergic disorders.
Collapse
Affiliation(s)
- Kazufumi Matsushita
- Laboratory of Allergic Diseases, Institute for Advanced Medical Sciences, and.,Department of Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hiroki Tanaka
- Laboratory of Host Defense, World Premier International Immunology Frontier Research Center, and.,Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Koubun Yasuda
- Department of Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Takumi Adachi
- Department of Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Ayumi Fukuoka
- Department of Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Shoko Akasaki
- Laboratory of Allergic Diseases, Institute for Advanced Medical Sciences, and
| | - Atsuhide Koida
- Department of Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Etsushi Kuroda
- Department of Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, World Premier International Immunology Frontier Research Center, and.,Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Tomohiro Yoshimoto
- Laboratory of Allergic Diseases, Institute for Advanced Medical Sciences, and.,Department of Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
10
|
Freitas KM, Silva ACAE, Veloso ES, Ferreira Ê, Barcelos LS, Caliari MV, Salas CE, Lopes MTP. P1G10, the Proteolytic Fraction from Vasconcellea cundinamarcensis, Stimulates Tissue Repair after Acute Exposure to Ultraviolet B Radiation. Int J Mol Sci 2019; 20:E4373. [PMID: 31489890 PMCID: PMC6770601 DOI: 10.3390/ijms20184373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2019] [Revised: 08/16/2019] [Accepted: 08/28/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND P1G10 is a cysteine proteolytic fraction from Vasconcellea cundinamarcensis latex, obtained by chromatographic separation on Sephadex-G10 and ultrafiltration. This fraction enhances healing in different models of skin lesions, and displays a protective/healing effect against gastric ulcers, where it was suggested an antioxidant role. METHODS We evaluated here the effect of topical treatment with P1G10, in mice lesions induced by UVB. RESULTS After single exposure to 2.4 J cm-2 UVB, P1G10 reduced erythema, increased cellularity of hypodermis, enhanced MPO activity and IL1β, and inhibited COX2 levels. These results point to an anti-inflammatory effect by P1G10. This fraction displayed antioxidant activity by reversing the depletion of glutathione (GSH), glutathione peroxidase (GSH-Px), superoxide dismutase (SOD) and reducing the catalase activity increased by UVB. These changes may be related to a reduction in MDA observed in groups treated with P1G10. P1G10 also inhibited MMP-9, caspase-3 and pkat while increasing p53 levels.
Collapse
Affiliation(s)
- Kátia M Freitas
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antônio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil.
| | - Ana C Araújo E Silva
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antônio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil.
- Faculdade de Medicina do Mucuri, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Rua do Cruzeiro, nº 01, Bairro Jardim São Paulo, Teófilo Otoni 39803-371, MG, Brazil.
| | - Emerson S Veloso
- Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antônio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil.
| | - Ênio Ferreira
- Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antônio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil.
| | - Lucíola S Barcelos
- Departamento Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antônio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil.
| | - Marcelo V Caliari
- Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antônio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil.
| | - Carlos E Salas
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antônio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil.
| | - Miriam T P Lopes
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antônio Carlos 6627, Belo Horizonte 31270-901, MG, Brazil.
| |
Collapse
|
11
|
Nagashima R, Kosai H, Masuo M, Izumiyama K, Noshikawaji T, Morimoto M, Kumaki S, Miyazaki Y, Motohashi H, Yamamoto M, Tanaka N. Nrf2 Suppresses Allergic Lung Inflammation by Attenuating the Type 2 Innate Lymphoid Cell Response. THE JOURNAL OF IMMUNOLOGY 2019; 202:1331-1339. [PMID: 30674574 DOI: 10.4049/jimmunol.1801180] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/24/2018] [Indexed: 12/13/2022]
Abstract
The Keap1-Nrf2 system plays a pivotal role in the oxidative stress response by inducing a number of cytoprotective genes. Under stress, damaged epithelial cells release cytokines that activate type 2 innate lymphoid cells (ILC2s), which mediate the allergic immune response. In this article, we investigated the role of the Keap1-Nrf2 pathway in ILC2 homeostasis and allergic inflammation using Nrf2 knockout mice. ILC2s from Nrf2-deficient mice showed a transient, upregulated IL-33 response and underwent hyperproliferation in response to a combined stimulation of IL-33 with IL-2, IL-7, or TSLP. This enhanced proliferation was correlated with an increased activation of downstream signals, including JAK1, Akt, and Erk1/2. In contrast, activating Nrf2 with a chemical inducer (CDDO-Im) decreased the viability of the wild-type but not of the Nrf2-deficient ILC2s. This effect on viability resembled that exerted by the corticosteroid dexamethasone; however, unlike the latter, the Nrf2-dependent cell death was mediated by neither caspase 3-dependent apoptosis nor necroptosis. Using a mouse intratracheal IL-33 administration allergy model, we found that the activation of Nrf2 by CDDO-Im in vivo decreased the number of pulmonary ILC2s and eosinophils. These findings indicated that Nrf2 is an important regulator of the allergic response by determining the survival and death of ILC2s, and these findings suggest that Nrf2 activation is a potential therapeutic strategy for steroid-resistant allergy alleviation.
Collapse
Affiliation(s)
- Ryuichi Nagashima
- Division of Cancer Biology and Therapeutics, Miyagi Cancer Center Research Institute, Medeshima-Shiode, Natori 981-1293, Japan.,Division of Tumor Immunobiology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Japan
| | - Hitomi Kosai
- Division of Cancer Biology and Therapeutics, Miyagi Cancer Center Research Institute, Medeshima-Shiode, Natori 981-1293, Japan
| | - Masahiro Masuo
- Department of Respiratory Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Tokyo 113-8510, Japan
| | - Keiko Izumiyama
- Division of Cancer Biology and Therapeutics, Miyagi Cancer Center Research Institute, Medeshima-Shiode, Natori 981-1293, Japan
| | - Taketo Noshikawaji
- Division of Cancer Biology and Therapeutics, Miyagi Cancer Center Research Institute, Medeshima-Shiode, Natori 981-1293, Japan.,Division of Tumor Immunobiology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Japan
| | - Motoko Morimoto
- School of Food, Agricultural and Environmental Sciences, Miyagi University, 2-2-1 Hatadate, Sendai 982-0215, Japan
| | - Satoru Kumaki
- Department of Pediatrics, Sendai Medical Center, 2-8-8 Miyagino, Sendai 983-8520, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, 1-5-45 Yushima, Tokyo 113-8510, Japan
| | - Hozumi Motohashi
- Division of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University 4-1 Seiryo-machi, Sendai 980-8575, Japan; and
| | - Masayuki Yamamoto
- Division of Medical Biochemistry, Tohoku University Graduate School of Medicine 2-1 Seiryo-machi, Sendai 980-8575, Japan
| | - Nobuyuki Tanaka
- Division of Cancer Biology and Therapeutics, Miyagi Cancer Center Research Institute, Medeshima-Shiode, Natori 981-1293, Japan; .,Division of Tumor Immunobiology, Tohoku University Graduate School of Medicine, 2-1 Seiryo-machi, Sendai 980-8575, Japan
| |
Collapse
|
12
|
Chen L, Chen H, Chen P, Zhang W, Wu C, Sun C, Luo W, Zheng L, Liu Z, Liang G. Development of 2-amino-4-phenylthiazole analogues to disrupt myeloid differentiation factor 88 and prevent inflammatory responses in acute lung injury. Eur J Med Chem 2019; 161:22-38. [DOI: 10.1016/j.ejmech.2018.09.068] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/11/2018] [Accepted: 09/26/2018] [Indexed: 11/16/2022]
|
13
|
Bueter CL, Deepe GS. Aeroallergens Exacerbate Histoplasma capsulatum Infection. THE JOURNAL OF IMMUNOLOGY 2018; 201:3352-3361. [PMID: 30348735 DOI: 10.4049/jimmunol.1800644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/20/2018] [Indexed: 11/19/2022]
Abstract
Allergens such as house dust mites (HDM) and papain induce strong Th2 responses, including elevated IL-4, IL-5, and IL-13 and marked eosinophilia in the airways. Histoplasma capsulatum is a dimorphic fungal pathogen that induces a strong Th1 response marked by IFN-γ and TNF-α production, leading to rapid clearance in nonimmunocompromised hosts. Th1 responses are generally dominant and overwhelm the Th2 response when stimuli for both are present, although there are instances when Th2 stimuli downregulate a Th1 response. We determined if the Th2 response to allergens prevents the host from mounting a Th1 response to H. capsulatum in vivo. C57BL/6 mice exposed to HDM or papain and infected with H. capsulatum exhibited a dominant Th2 response early, characterized by enhanced eosinophilia and elevated Th2 cytokines in lungs. These mice manifested exacerbated fungal burdens, suggesting that animals skewed toward a Th2 response by an allergen are less able to clear the H. capsulatum infection despite an intact Th1 response. In contrast, secondary infection is not exacerbated by allergen exposure, indicating that the memory response may suppress the Th2 response to HDM and quickly clear the infection. In conclusion, an in vivo skewing toward Th2 by allergens exacerbates fungal infection, even though there is a concurrent and unimpaired Th1 response to H. capsulatum.
Collapse
Affiliation(s)
- Chelsea L Bueter
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH 45267; and
| | - George S Deepe
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH 45267; and .,Veterans Affairs Hospital, Cincinnati, OH 45220
| |
Collapse
|
14
|
Secher T, Maillet I, Mackowiak C, Le Bérichel J, Philippeau A, Panek C, Boury M, Oswald E, Saoudi A, Erard F, Le Bert M, Quesniaux V, Couturier-Maillard A, Ryffel B. The probiotic strain Escherichia coli Nissle 1917 prevents papain-induced respiratory barrier injury and severe allergic inflammation in mice. Sci Rep 2018; 8:11245. [PMID: 30050168 PMCID: PMC6062509 DOI: 10.1038/s41598-018-29689-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 07/16/2018] [Indexed: 02/07/2023] Open
Abstract
Allergic asthma is characterized by a strong Th2 and Th17 response with inflammatory cell recruitment, airways hyperreactivity and structural changes in the lung. The protease allergen papain disrupts the airway epithelium triggering a rapid eosinophilic inflammation by innate lymphoid cell type 2 (ILC2) activation, leading to a Th2 immune response. Here we asked whether the daily oral administrations of the probiotic Escherichia coli strain Nissle 1917 (ECN) might affect the outcome of the papain protease induced allergic lung inflammation in BL6 mice. We find that ECN gavage significantly prevented the severe allergic response induced by repeated papain challenges and reduced lung inflammatory cell recruitment, Th2 and Th17 response and respiratory epithelial barrier disruption with emphysema and airway hyperreactivity. In conclusion, ECN administration attenuated severe protease induced allergic inflammation, which may be beneficial to prevent allergic asthma.
Collapse
Affiliation(s)
- Thomas Secher
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France. .,INSERM, UMR 1100, Research Center for Respiratory Diseases, and University of Tours, Tours, France.
| | - Isabelle Maillet
- CNRS, UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Claire Mackowiak
- CNRS, UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Jessica Le Bérichel
- CNRS, UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Amandine Philippeau
- CNRS, UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Corinne Panek
- CNRS, UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Michèle Boury
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France
| | - Eric Oswald
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.,CHU Toulouse, Hôpital Purpan, Service de Bactériologie-Hygiène, Toulouse, France
| | - Abdelhadi Saoudi
- Centre de Physiopathologie de Toulouse Purpan (CPTP), Université de Toulouse, UPS, Inserm, CNRS, Toulouse, France
| | - Francois Erard
- CNRS, UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Marc Le Bert
- CNRS, UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Valérie Quesniaux
- CNRS, UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France.,University of Orleans, Orleans, France
| | | | - Bernhard Ryffel
- CNRS, UMR7355, Experimental and Molecular Immunology and Neurogenetics, Orleans, France. .,University of Orleans, Orleans, France. .,University of Cape Town, IDM, Cape Town, Republic of South Africa.
| |
Collapse
|
15
|
Bassoy EY, Towne JE, Gabay C. Regulation and function of interleukin-36 cytokines. Immunol Rev 2018; 281:169-178. [PMID: 29247994 DOI: 10.1111/imr.12610] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The interleukin (IL)-36 cytokines include 3 agonists, IL-36α, IL-36β, and IL-36γ that bind to a common receptor composed of IL-36R and IL-1RAcP to stimulate inflammatory responses. IL-36Ra is a natural antagonist that binds to IL-36R, but does not recruit the co-receptor IL-1RAcP and does not stimulate any intracellular responses. The IL-36 cytokines are expressed predominantly by epithelial cells and act on a number of cells including immune cells, epithelial cells, and fibroblasts. Processing of the N-terminus is required for full agonist or antagonist activity for all IL-36 members. The role of IL-36 has been extensively demonstrated in the skin where it can act on keratinocytes and immune cells to induce a robust inflammatory response that has been implicated in psoriatic disorders. Emerging data also suggest a role for this cytokine family in pulmonary and intestinal physiology and pathology.
Collapse
Affiliation(s)
- Esen Yonca Bassoy
- Division of Rheumatology, Department of Internal Medicine Specialties & Department of Pathology-Immunology, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
| | - Jennifer E Towne
- Immunology Discovery, Janssen Research and Development, San Diego, CA, USA
| | - Cem Gabay
- Division of Rheumatology, Department of Internal Medicine Specialties & Department of Pathology-Immunology, University Hospitals of Geneva and University of Geneva, Geneva, Switzerland
| |
Collapse
|
16
|
Functional and morphological differences of the lung upon acute and chronic ozone exposure in mice. Sci Rep 2018; 8:10611. [PMID: 30006538 PMCID: PMC6045627 DOI: 10.1038/s41598-018-28261-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 06/14/2018] [Indexed: 02/07/2023] Open
Abstract
Environmental air pollutants including ozone cause severe lung injury and aggravate respiratory diseases such as asthma and COPD. Here we compared the effect of ozone on respiratory epithelium injury, inflammation, hyperreactivity and airway remodeling in mice upon acute (1ppm, 1 h) and chronic exposure (1.5ppm, 2 h, twice weekly for 6 weeks). Acute ozone exposure caused respiratory epithelial disruption with protein leak and neutrophil recruitment in the broncho-alveolar space, leading to lung inflammation and airway hyperresponsiveness (AHR) to methacholine. All these parameters were increased upon chronic ozone exposure, including collagen deposition. The structure of the airways as assessed by automatic numerical image analysis showed significant differences: While acute ozone exposure increased bronchial and lumen circularity but decreased epithelial thickness and area, chronic ozone exposure revealed epithelial injury with reduced height, distended bronchioles, enlarged alveolar space and increased collagen deposition, indicative of peribronchiolar fibrosis and emphysema as characterized by a significant increase in the density and diameter of airspaces with decreased airspace numbers. In conclusion, morphometric numerical analysis enables an automatic and unbiased assessment of small airway remodeling. The structural changes of the small airways correlated with functional changes allowing to follow the progression from acute to chronic ozone induced respiratory pathology.
Collapse
|
17
|
Xiao Y, Xu W, Su W. NLRP3 inflammasome: A likely target for the treatment of allergic diseases. Clin Exp Allergy 2018; 48:1080-1091. [PMID: 29900602 DOI: 10.1111/cea.13190] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 05/25/2018] [Accepted: 06/11/2018] [Indexed: 12/28/2022]
Abstract
Allergic diseases, such as asthma, rhinitis, dermatitis, conjunctivitis, and anaphylaxis, have recently become a global public health concern. According to previous studies, the NLRP3 inflammasome is a multi-protein complex known to be associated with many inflammatory conditions. In response to allergens or allergen/damage-associated molecular signals, NLRP3 changes its conformation to allow the assembly of the NLRP3 inflammasome complex and activates caspase-1, which is an evolutionarily conserved enzyme that proteolytically cleaves other proteins, such as the precursors of the inflammatory cytokines IL-1β and IL-18. Subsequently, active caspase-1 cleaves pro-IL-1 and pro-IL-18. Recently, accumulating human and mouse experimental evidence has demonstrated that the NLRP3 inflammasome, IL-1β, and IL-18 are critically involved in the development of allergic diseases. Furthermore, the application of specific NLRP3 inflammasome inhibitors has been demonstrated in animal models. Therefore, these inhibitors may represent potential therapeutic methods for the management of clinical allergic disorders. This review summarizes findings related to the NLRP3 inflammasome and its related factors and concludes that specific NLRP3 inflammasome inhibitors may be potential therapeutic agents for allergic diseases.
Collapse
Affiliation(s)
- Yichen Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenna Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Li J, Saruta K, Dumouchel JP, Magat JM, Thomas JL, Ajami D, Rebek M, Rebek J, Bigby TD. Small Molecule Mimetics of α-Helical Domain of IRAK2 Attenuate the Proinflammatory Effects of IL-33 in Asthma-like Mouse Models. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:4036-4043. [PMID: 29728508 PMCID: PMC5988972 DOI: 10.4049/jimmunol.1700693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 04/10/2018] [Indexed: 01/04/2023]
Abstract
IL-33 and its receptor ST2 play important roles in airway inflammation and contribute to asthma onset and exacerbation. The IL-33/ST2 signaling pathway recruits adapter protein myeloid differentiation primary response 88 (MyD88) to transduce intracellular signaling. MyD88 forms a complex with IL-R-associated kinases (IRAKs), IRAK4 and IRAK2, called the Myddosome (MyD88-IRAK4-IRAK2). The myddosome subsequently activates downstream NF-κB and MAPKs p38 and JNK. We established an asthma-like mouse model by intratracheal administration of IL-33. The IL-33 model has a very similar phenotype compared with the OVA-induced mouse asthma model. The importance of MyD88 in the IL-33/ST2 signaling transduction was demonstrated by the MyD88 knockout mice, which were protected from the IL-33-induced asthma. We synthesized small molecule mimetics of the α-helical domain of IRAK2 with drug-like characteristics based on the recent advances in the designing of α-helix compounds. The mimetics can competitively interfere in the protein-protein interaction between IRAK2 and IRAK4, leading to disruption of Myddosome formation. A series of small molecules were screened using an NF-κB promoter assay in vitro. The lead compound, 7004, was further studied in the IL-33-induced and OVA-induced asthma mouse models in vivo. Compound 7004 can inhibit the IL-33-induced NF-κB activity, disrupt Myddosome formation, and attenuate the proinflammatory effects in asthma-like models. Our data indicate that the Myddosome may represent a novel intracellular therapeutic target for diseases in which IL-33/ST2 plays important roles, such as asthma and other inflammatory diseases.
Collapse
Affiliation(s)
- Jinghong Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093;
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | - Kunio Saruta
- The Scripps Research Institute, La Jolla, CA 92037
| | - Justin P Dumouchel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | - Jenna M Magat
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | - Joanna L Thomas
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | | | - Mitra Rebek
- The Scripps Research Institute, La Jolla, CA 92037
| | - Julius Rebek
- The Scripps Research Institute, La Jolla, CA 92037
| | - Timothy D Bigby
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| |
Collapse
|
19
|
Michaudel C, Maillet I, Fauconnier L, Quesniaux V, Chung KF, Wiegman C, Peter D, Ryffel B. Interleukin-1α Mediates Ozone-Induced Myeloid Differentiation Factor-88-Dependent Epithelial Tissue Injury and Inflammation. Front Immunol 2018; 9:916. [PMID: 29867931 PMCID: PMC5950844 DOI: 10.3389/fimmu.2018.00916] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/13/2018] [Indexed: 02/04/2023] Open
Abstract
Air pollution associated with ozone exposure represents a major inducer of respiratory disease in man. In mice, a single ozone exposure causes lung injury with disruption of the respiratory barrier and inflammation. We investigated the role of interleukin-1 (IL-1)-associated cytokines upon a single ozone exposure (1 ppm for 1 h) using IL-1α-, IL-1β-, and IL-18-deficient mice or an anti-IL-1α neutralizing antibody underlying the rapid epithelial cell death. Here, we demonstrate the release of the alarmin IL-1α after ozone exposure and that the acute respiratory barrier injury and inflammation and airway hyperreactivity are IL-1α-dependent. IL-1α signaling via IL-1R1 depends on the adaptor protein myeloid differentiation factor-88 (MyD88). Importantly, epithelial cell signaling is critical, since deletion of MyD88 in lung type I alveolar epithelial cells reduced ozone-induced inflammation. In addition, intratracheal injection of recombinant rmIL-1α in MyD88acid mice led to reduction of inflammation in comparison with wild type mice treated with rmIL-1α. Therefore, a major part of inflammation is mediated by IL-1α signaling in epithelial cells. In conclusion, the alarmin IL-1α released upon ozone-induced tissue damage and inflammation is mediated by MyD88 signaling in epithelial cells. Therefore, IL-1α may represent a therapeutic target to attenuate ozone-induced lung inflammation and hyperreactivity.
Collapse
Affiliation(s)
- Chloé Michaudel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans, France
| | - Isabelle Maillet
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans, France
| | | | - Valérie Quesniaux
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans, France
| | - Kian Fan Chung
- Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom.,NIHR Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, London, United Kingdom
| | - Coen Wiegman
- Airways Disease, National Heart and Lung Institute, Imperial College London, London, United Kingdom.,NIHR Respiratory Biomedical Research Unit, Royal Brompton & Harefield NHS Foundation Trust, London, United Kingdom
| | - Daniel Peter
- Immunology and Respiratory Diseases Research, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Bernhard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS-University of Orleans, Orleans, France
| |
Collapse
|
20
|
Di Padova F, Quesniaux VFJ, Ryffel B. MyD88 as a therapeutic target for inflammatory lung diseases. Expert Opin Ther Targets 2018; 22:401-408. [PMID: 29658361 DOI: 10.1080/14728222.2018.1464139] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Myeloid differentiation primary response protein 88 (MyD88) is a critical adaptor protein involved in Toll-like and IL-1 receptor family signaling controlling innate immune responses and inflammation. Genetic deletion of MyD88 function results in profound suppression of inflammation and reduced resistance of the host to pathogens indicating non-redundant roles of MyD88. The TIR domain is critical for MyD88 dimerization and signaling for TLR and IL-1R family receptor. Areas covered: Emerging evidence suggests that chemical disruption of the TIR domain attenuates cell activation and inhibits in vivo MyD88-dependent inflammation. We review the development of MyD88 dimerization disruptors as a novel therapeutic approach of respiratory diseases with a focus on COPD. Expert opinion: There is a proof of concept that therapeutic targeting of MyD88 is feasible and first preclinical data are highly promising. This opens a great opportunity to treat exacerbations of COPD and other chronic respiratory diseases. However, extensive preclinical investigations and risk analyses are required with carefully evaluation of reduced host resistance and opportunistic infections.
Collapse
Affiliation(s)
| | - Valerie F J Quesniaux
- b Laboratory of experimental and molecular immunology and neurogenetics (INEM) , UMR 7355 CNRS-University of Orleans , Orleans , France
| | - Bernhard Ryffel
- b Laboratory of experimental and molecular immunology and neurogenetics (INEM) , UMR 7355 CNRS-University of Orleans , Orleans , France.,c IDM, Institute of Infectious Disease and Molecular Medicine , University of Cape Town , Cape Town , RSA
| |
Collapse
|
21
|
Mei B, Yang S, Yue Y, Hou J, Wang K, Chen G, Liang M, Wu Z. Acute adrenal cortex injury during cardiopulmonary bypass in a canine model. J Thorac Cardiovasc Surg 2018; 156:696-706. [PMID: 29753511 DOI: 10.1016/j.jtcvs.2018.03.151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 03/04/2018] [Accepted: 03/05/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Cardiopulmonary bypass (CPB) might induce systemic inflammatory responses that cause acute injuries to multiple organs. However, no direct evidence exists to determine whether CPB leads to adrenal cortex injury or to describe its underlying mechanism. METHODS Twelve healthy adult beagles were randomly assigned into control and CPB groups. After cannulation, mild hypothermia CPB was performed in the CPB group but not in the control group. The serum concentrations of various cytokines, cortisol, and aldosterone were assessed. Adrenal cortex injuries were evaluated using standard histological methods. Steroidogenic enzymes and the nucleotide-binding oligomerization domain-like receptor containing pyrin domain 3 (NLRP3) inflammasome pathway were detected using quantitative polymerase chain reaction and Western blot analysis. RESULTS During CPB, serum interleukin (IL)-6, IL-8, IL-10, tumor necrosis factor α, cortisol, and aldosterone levels were significantly higher in the CPB group. The pathologic study revealed higher injury scores (3.6 ± 0.6 vs 0.7 ± 0.7) and significantly more severe edema, inflammatory cell infiltration (lymphocytes and neutrophils), and apoptosis in the CPB group. The electron microscopic examination showed swollen mitochondria, ruptured mitochondrial cristae, reduced lipid droplets, and increased secondary lysosomes in the CPB group. The mRNA expression levels of NLRP3 and the protein levels of 17α-hydroxylase and IL-1β in adrenal tissue were significantly upregulated in the CPB group. CONCLUSIONS CPB induces significant systemic and local inflammation in the adrenal cortex and results in cytological architectural and ultrastructural alterations in adrenocorticocytes. In addition, the NLRP3 inflammasome pathway might promote adrenal gland injury during CPB and might represent a novel potential therapeutic target.
Collapse
Affiliation(s)
- Bo Mei
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Song Yang
- Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China; Department of Cardiosurgery Intensive Care Unit, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuan Yue
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Jian Hou
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Keke Wang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Guangxian Chen
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Mengya Liang
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Zhongkai Wu
- Department of Cardiac Surgery, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China.
| |
Collapse
|
22
|
Abstract
Innate lymphoid cells are functionally diverse subsets of immune cells including the conventional natural killer cells, lymphoid tissue inducers, type 1, 2, and 3 with significant roles in immunity and pathogenesis of inflammatory diseases. Type 2 innate lymphoid cells (ILC2s) resemble type 2 helper (Th2) cells in cytokine production and contribute to anti-helminth immunity, maintaining mucosal tissue integrity, and adipose tissue browning. ILC2s play important roles in the pathogenesis of allergic diseases and asthma. Studying the pathways of activation and regulation of ILC2s are currently a priority for giving a better understanding of pathogenesis of diseases with immunological roots. Recently, our laboratory and others have shown several pathways of regulation of ILC2s by co-stimulatory molecules such as ICOS, regulatory T cells and by compounds such as nicotine. In this review, we summarize the current understanding of the mechanisms of activation and regulation of ILC2s and the role of these cells in health and disease.
Collapse
Affiliation(s)
- Hadi Maazi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, California 90033, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, Los Angeles, California 90033, USA
| |
Collapse
|
23
|
Li C, Maillet I, Mackowiak C, Viala C, Di Padova F, Li M, Togbe D, Quesniaux V, Lai Y, Ryffel B. Experimental atopic dermatitis depends on IL-33R signaling via MyD88 in dendritic cells. Cell Death Dis 2017; 8:e2735. [PMID: 28383552 PMCID: PMC5477596 DOI: 10.1038/cddis.2017.90] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 01/08/2023]
Abstract
Atopic dermatitis (AD) is a chronic Th2 type inflammatory skin disorder. Here we report that MyD88 signaling is crucial in the pathogenesis of experimental AD induced by vitamin D3 analog MC903. The clinical signs and inflammation caused by MC903 are drastically reduced in MyD88−/− mice with diminished eosinophil, neutrophil infiltration and Th2 cytokine expression. The biological effect of interleukin-1 (IL-1) family members relies on MyD88 signaling. We observed a strong upregulation of IL-1 family cytokine expression, including IL-1α, IL-1β, IL-33, IL-18, IL-36α, IL-36β, IL-36γ and IL-36Ra. Therefore, we asked which cytokine of the IL-1 family would be essential for MC903-induced AD syndrome. We find a significant reduction of AD in IL-33−/− and IL-33R/ST2−/− mice, only a minor reduction in double IL-1αβ−/− mice and no difference in IL-36R−/− and IL-36Ra−/− mice. IL-33 is expressed in keratinocytes, and MyD88 signaling in dendritic cells (DCs) is crucial for AD development as inflammation was drastically reduced in DC-specific MyD88−/− mice (CD11c-cre × MyD88-floxed). Taken together, the data demonstrate a critical role of MyD88 in DCs and of IL-33 signaling via ST2 in MC903-induced AD. These data suggest that IL-33/IL-33R may be a therapeutic target of AD.
Collapse
Affiliation(s)
- Changwei Li
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS and University of Orleans, F-45071 Orleans-Cedex 2, France.,Shanghai Institute of Traumatology and Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Ruijin Hospital, Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Isabelle Maillet
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS and University of Orleans, F-45071 Orleans-Cedex 2, France
| | - Claire Mackowiak
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS and University of Orleans, F-45071 Orleans-Cedex 2, France
| | - Camille Viala
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS and University of Orleans, F-45071 Orleans-Cedex 2, France
| | - Franco Di Padova
- Novartis Institutes for Biomedical Research WSJ 386.11.48.38 4002, Basel, Switzerland
| | - Mei Li
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Centre National de la Recherche Scientifique UMR7104/Institut National de la Santé et de la Recherche Médicale U964/Université de Strasbourg, Illkirch, France.,University of Strasbourg Institute for Advanced Study, Strasbourg, France.,Freiburg Institute for Advanced Studies, Freiburg, Germany
| | | | - Valérie Quesniaux
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS and University of Orleans, F-45071 Orleans-Cedex 2, France
| | - Yuping Lai
- IDM, Institute of Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Bernhard Ryffel
- Experimental and Molecular Immunology and Neurogenetics (INEM), UMR 7355 CNRS and University of Orleans, F-45071 Orleans-Cedex 2, France.,Shanghai Key Laboratory of Regulatory Biology, School of Life Sciences, East China Normal University, Shanghai 200241, China
| |
Collapse
|
24
|
Walsh PT, Fallon PG. The emergence of the IL-36 cytokine family as novel targets for inflammatory diseases. Ann N Y Acad Sci 2016; 1417:23-34. [PMID: 27783881 DOI: 10.1111/nyas.13280] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 09/22/2016] [Accepted: 09/26/2016] [Indexed: 12/13/2022]
Abstract
The recently discovered interleukin (IL)-36 family of cytokines form part of the broader IL-1 family and are emerging as important mediators of inflammatory disease. The IL-36 subfamily consists of three ligands-IL-36α, IL-36β, and IL-36γ-and the natural antagonist IL-36Ra. The cytokines exert their effects through a specific IL-36 receptor consisting of IL-36R and IL-1RAcP chains. IL-36 cytokines can direct both innate and adaptive immune responses by acting on parenchymal, stromal, and specific immune cell subsets. In humans, inactivating mutations in the gene encoding the IL-36R antagonist, which lead to unregulated IL-36R signaling, lead to an autoinflammatory condition termed deficiency of the IL-36R antagonist, which primarily manifests as a severe form of pustular psoriasis. While such discoveries have prompted deeper mechanistic studies highlighting the important role of IL-36 cytokines in psoriatic skin inflammation, it is now evident that IL-36 cytokines can also play important roles in inflammatory disorders in other organs, such as the gastrointestinal tract and the lungs. Given these emerging roles, strategies to specifically target the expression and activity of the IL-36 family have the potential to uncover novel therapeutic approaches aimed at treating inflammatory diseases in humans.
Collapse
Affiliation(s)
- Patrick T Walsh
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland.,School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Padraic G Fallon
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland.,School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|