1
|
López-Aguilar KK, Vargas-Camaño ME, Lozano-Patiño F, Castrejón Vázquez MI. Recurrent respiratory papillomatosis: Immunological mechanisms involved in recurrence. Int Rev Immunol 2025; 44:113-126. [PMID: 39509110 DOI: 10.1080/08830185.2024.2425428] [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: 01/26/2023] [Revised: 10/12/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024]
Abstract
Recurrent respiratory papillomatosis is a benign neoplastic pathology in children, young people, and adults. It causes a significant deterioration in the quality of life, with symptoms typically referred to as dysphonia and hoarseness. This disease, with variable clinical courses ranging from spontaneous resolution to dissemination of the lower airway or airway obstruction that puts the individual's life at risk, characteristically requires multiple surgical interventions. Therapy with adjuvant drugs does not yet prove the effectiveness necessary to limit the recurrence and need for surgical reoperation in this condition. The review aimed to synthesize the immunopathogenic mechanisms of relapse in recurrent respiratory papillomatosis published in the current literature and the immunological implication of risk factors and treatment.
Collapse
Affiliation(s)
- Katya Karen López-Aguilar
- Centro Medico Nacional "20 de Noviembre" Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Ciudad de México, México
| | - María Eugenia Vargas-Camaño
- Centro Medico Nacional "20 de Noviembre" Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Ciudad de México, México
| | - Fernando Lozano-Patiño
- Centro Medico Nacional "20 de Noviembre" Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Ciudad de México, México
| | - María Isabel Castrejón Vázquez
- Centro Medico Nacional "20 de Noviembre" Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, Ciudad de México, México
| |
Collapse
|
2
|
Meng Q, Wang X, Guo D, Zhang G, Shi C, Novak A, Yang X, Luo J, Cooney RN. Sparstolonin B nano-formulation attenuates LPS-induced lung injury. Front Pharmacol 2025; 16:1532391. [PMID: 40264680 PMCID: PMC12011759 DOI: 10.3389/fphar.2025.1532391] [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: 11/21/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
Introduction Nanomedicines can improve drug delivery and efficacy while reducing side effects. Our study examines the impact of a nano-formulation of Sparstolonin B (nSsnB), a TLR-4 antagonist, on LPS-induced inflammation in RAW264.7 cells and lung injury in mice. Methods RAW264.7 cells were treated with LPS (1 μg/mL) ± nSsnB (2-64 μg/mL) for 24 h. Cell viability was assessed, cytokine levels in media were measured, and cell lysates were used to quantify NF-κB activation. C57BL/6 mice were treated with prophylactic intratracheal (IT) nSsnB (0.625 mg/kg) ± IT LPS (2.5 mg/kg). Blood and BALF were collected for cytokine, protein and cytological analysis. Lung histology was scored to evaluate lung injury. The relative abundance of MyD88 and phosphorylated NF-κB were measured in lung and HLL mice were used to measure NF-κB activation in vivo. Results nSsnB demonstrated reduced toxicity vs. free SsnB. nSsnB ameliorated the LPS-induced increase in TNF-α, IL-6 and NF-κB P65 phosphorylation in RAW264.7 cells. LPS-treated mice revealed histologic ALI, elevated BALF neutrophils/macrophages/total protein, and increased levels of TNF-α/IL-6 in both BALF and plasma. Prophylactic nSsnB attenuated all these parameters in the LPS/nSsnB group. The increased levels of MyD88 and P-NF-κB P65 in lung from LPS-treated mice were reduced in the LPS/nSsnB group and nSsnB attenuated the increase in NF-κB activation induced by IT LPS in HLL mice. Conclusion nSsnB demonstrates less toxicity than free SsnB and attenuates the effects of LPS on inflammation in RAW264.7 cells. Prophylactic nSsnB attenuates LPS-induced ALI by reducing inflammation via MyD88/NF-κB signaling pathways. Collectively these findings support the therapeutic potential of nano-formulated nSsnB for ALI treatment.
Collapse
Affiliation(s)
- Qinghe Meng
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
- Sepsis Interdisciplinary Research Center (SIRC), State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| | - Xiaojing Wang
- Department of Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| | - Dandan Guo
- Department of Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| | - Gary Zhang
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| | - Changying Shi
- Department of Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| | - Adam Novak
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| | - Xiguang Yang
- Department of Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| | - Juntao Luo
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
- Sepsis Interdisciplinary Research Center (SIRC), State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
- Department of Pharmacology, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| | - Robert N. Cooney
- Department of Surgery, State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
- Sepsis Interdisciplinary Research Center (SIRC), State University of New York (SUNY), Upstate Medical University, Syracuse, NY, United States
| |
Collapse
|
3
|
Ceccarelli S, Pasqua Marzolesi V, Vannucci J, Bellezza G, Floridi C, Nocentini G, Cari L, Traina G, Petri D, Puma F, Conte C. Toll-Like Receptor 4 and 8 are Overexpressed in Lung Biopsies of Human Non-small Cell Lung Carcinoma. Lung 2025; 203:38. [PMID: 40025339 PMCID: PMC11872755 DOI: 10.1007/s00408-025-00793-8] [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: 11/18/2024] [Accepted: 02/21/2025] [Indexed: 03/04/2025]
Abstract
PURPOSE Lung cancer is the leading cause of cancer death worldwide which includes two main types of carcinoma distinguished in non-small cell lung cancer (NSCLC) involving epithelial cells, and small cell lung cancer (SCLC) affecting neuronal cells and hormone secreting cells. Studies have shown a causal link between inflammation/innate immunity and onset of NSCLC. The present study aimed to evaluate the expression of Toll-like receptors (TLRs) 4 and TLR8 in peripheral blood mononuclear cells (PBMC) and in lung tissues of patients with NSCLC, useful for future prognostic tools for NSCLC. METHODS Patients surgically treated for NSCLC with anatomical resections and patients with benign disease were enrolled. The expression levels of TLR4 and TLR8 were determined by real time PCR and by immunohistochemical analysis in PBMC and in lung tissues, respectively. A preliminary in silico analysis including 1194 arrays from healthy and cancer tissues were extracted by Genevestigator database. The association between TLRs gene expression and survival outcome was also investigated. RESULTS Bioinformatics analysis revealed that downregulation of TLR4 and TLR8 positively impacts the survival in lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). However, no significant differences in TLR4 and TLR8 gene expression between case and control groups were observed in PBMC. A positive correlation was found in their expression levels. Interestingly, immunohistochemical analysis showed that the levels of TLR4 and TLR8 were higher in the lung tissues of patients with NSCLC than in the control group in terms of staining intensity and positive cells. CONCLUSION Albeit the precise role of TLRs is not fully defined, this study identified the potential involvement of TLR4 and TLR8 in the pathogenesis of NSCLC. Our data led us to hypothesize their potential role in overall survival which deserves to be explored further to establish whether TLR4 and TLR8 can represent positive prognostic indicators of disease in NSCLC.
Collapse
MESH Headings
- Humans
- Toll-Like Receptor 4/metabolism
- Toll-Like Receptor 4/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/surgery
- Carcinoma, Non-Small-Cell Lung/mortality
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/mortality
- Lung Neoplasms/surgery
- Male
- Female
- Middle Aged
- Aged
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/pathology
- Toll-Like Receptor 8/genetics
- Toll-Like Receptor 8/analysis
- Toll-Like Receptor 8/metabolism
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/surgery
- Biopsy
- Adenocarcinoma of Lung/pathology
- Adenocarcinoma of Lung/genetics
- Adenocarcinoma of Lung/metabolism
- Adenocarcinoma of Lung/surgery
- Adenocarcinoma of Lung/mortality
- Lung/pathology
- Lung/metabolism
- Prognosis
- Adenocarcinoma/pathology
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/mortality
- Adenocarcinoma/surgery
- Gene Expression Regulation, Neoplastic
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Case-Control Studies
- Immunohistochemistry
- Adult
Collapse
Affiliation(s)
- Silvia Ceccarelli
- Department of Surgical and Biomedical Sciences, Thoracic Surgery Unit, Medical School, University of Perugia, Perugia, Italy
| | | | - Jacopo Vannucci
- Department of Surgical and Biomedical Sciences, Thoracic Surgery Unit, Medical School, University of Perugia, Perugia, Italy
| | - Guido Bellezza
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, Perugia, Italy
| | - Claudia Floridi
- Department of Medicine and Surgery, Section of Anatomic Pathology and Histology, Medical School, University of Perugia, Perugia, Italy
| | - Giuseppe Nocentini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luigi Cari
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giovanna Traina
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Davide Petri
- Department of Environment and Health, National Institute of Health, Rome, Italy
| | - Francesco Puma
- Department of Surgical and Biomedical Sciences, Thoracic Surgery Unit, Medical School, University of Perugia, Perugia, Italy
| | - Carmela Conte
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy.
| |
Collapse
|
4
|
Viz-Lasheras S, Gómez-Carballa A, Pardo-Seco J, Bello X, Rivero-Calle I, Dacosta AI, Kaforou M, Habgood-Coote D, Cunnington AJ, Emonts M, Herberg JA, Wright VJ, Carrol ED, Paulus SC, Zenz W, Kohlfürst DS, Van der Flier M, de Groot R, Schlapbach LJ, Agyeman P, Pollard AJ, Fink C, Kuijpers TT, Anderson S, Calvo C, Martínez-Padilla MDC, Pérez-Aragón A, Gómez-Sánchez E, Valencia-Ramos J, Giménez-Sánchez F, Alonso-Quintela P, Moreno-Galarraga L, von Both U, Pokorn M, Zavadska D, Tsolia M, Vermont CL, Moll HA, Levin M, Martinón-Torres F, Salas A. A 5-transcript signature for discriminating viral and bacterial etiology in pediatric pneumonia. iScience 2025; 28:111747. [PMID: 39906557 PMCID: PMC11791257 DOI: 10.1016/j.isci.2025.111747] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 10/24/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025] Open
Abstract
Pneumonia stands as the primary cause of death among children under five, yet current diagnosis methods often result in inadequate or unnecessary treatments. Our research seeks to address this gap by identifying host transcriptomic biomarkers in the blood of children with definitive viral and bacterial pneumonia. We performed RNA sequencing on 192 prospectively collected whole blood samples, including 38 controls and 154 pneumonia cases, uncovering a 5-transcript signature (genes FAM20A, BAG3, TDRD9, MXRA7, and KLF14) that effectively distinguishes bacterial from viral pneumonia (area under the curve (AUC): 0.95 [0.88-1.00]). Initial validation using combined definitive and probable cases yielded an AUC of 0.87 [0.77-0.97], while full validation in a new prospective cohort of 32 patients achieved an AUC of 0.92 [0.83-1.00]. This robust signature holds significant potential to enhance diagnostics accuracy for pediatric pneumonia, reducing diagnostic delays and unnecessary treatments and potentially transforming clinical practice.
Collapse
Affiliation(s)
- Sandra Viz-Lasheras
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Alberto Gómez-Carballa
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Jacobo Pardo-Seco
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Xabier Bello
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Irene Rivero-Calle
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Ana Isabel Dacosta
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Myrsini Kaforou
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | | | | | - Marieke Emonts
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Jethro A. Herberg
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Victoria J. Wright
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Enitan D. Carrol
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
| | - Stephane C. Paulus
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| | - Werner Zenz
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
| | - Daniela S. Kohlfürst
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
| | - Michiel Van der Flier
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Ronald de Groot
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Luregn J. Schlapbach
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Philipp Agyeman
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andrew J. Pollard
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| | - Colin Fink
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
| | - Taco T. Kuijpers
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
| | - Suzanne Anderson
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Cristina Calvo
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
| | | | - Ana Pérez-Aragón
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
| | - Esteban Gómez-Sánchez
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
| | - Juan Valencia-Ramos
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
| | | | - Paula Alonso-Quintela
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
| | - Laura Moreno-Galarraga
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
| | - Ulrich von Both
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Marko Pokorn
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Dace Zavadska
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
| | - María Tsolia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
| | | | | | - Michael Levin
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Federico Martinón-Torres
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Antonio Salas
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - on behalf of EUCLIDS
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | - DIAMONDS
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | - GENDRES and
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | - PERFORM consortia
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
5
|
Fu Q, Liu Y, Peng C, Muluh TA, Anayyat U, Liang L. Recent Advancement in Inhaled Nano-drug Delivery for Pulmonary, Nasal, and Nose-to-brain Diseases. Curr Drug Deliv 2025; 22:3-14. [PMID: 38275044 DOI: 10.2174/0115672018268047231207105652] [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: 08/25/2023] [Revised: 10/11/2023] [Accepted: 10/31/2023] [Indexed: 01/27/2024]
Abstract
Pulmonary, nasal, and nose-to-brain diseases involve clinical approaches, such as bronchodilators, inhaled steroids, oxygen therapy, antibiotics, antihistamines, nasal steroids, decongestants, intranasal drug delivery, neurostimulation, and surgery to treat patients. However, systemic medicines have serious adverse effects, necessitating the development of inhaled formulations that allow precise drug delivery to the airways with minimum systemic drug exposure. Particle size, surface charge, biocompatibility, drug capacity, and mucoadhesive are unique chemical and physical features that must be considered for pulmonary and nasal delivery routes due to anatomical and permeability considerations. The traditional management of numerous chronic diseases has a variety of drawbacks. As a result, targeted medicine delivery systems that employ nanotechnology enhancer drug efficiency and optimize the overall outcome are created. The pulmonary route is one of the most essential targeted drug delivery systems because it allows the administering of drugs locally and systemically to the lungs, nasal cavity, and brain. Furthermore, the lungs' beneficial characteristics, such as their ability to inhibit first-pass metabolism and their thin epithelial layer, help treat several health complications. The potential to serve as noninvasive self-administration delivery sites of the lung and nasal routes is discussed in this script. New methods for treating respiratory and some systemic diseases with inhalation have been explored and highlight particular attention to using specialized nanocarriers for delivering various drugs via the nasal and pulmonary pathways. The design and development of inhaled nanomedicine for pulmonary, nasal, and respiratory medicine applications is a potential approach for clinical translation.
Collapse
Affiliation(s)
- Qiuxia Fu
- Department of General Medicine, Luzhou People's Hospital, Luzhou 646000, Sichuan, China, (PRC)
| | - Yangjie Liu
- Department of General Medicine, Luzhou People's Hospital, Luzhou 646000, Sichuan, China, (PRC)
| | - Cao Peng
- Department of General Medicine, Luzhou People's Hospital, Luzhou 646000, Sichuan, China, (PRC)
| | - Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Umer Anayyat
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Liu Liang
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan, China PRC
| |
Collapse
|
6
|
Lin X, Lin J, Ji L, Zhang J, Zhang Y, Hong J, Li G, Lin X. Protective effect of Haoqin Qingdan decoction on pulmonary and intestinal injury in mice with influenza viral pneumonia. Front Pharmacol 2024; 15:1449322. [PMID: 39712501 PMCID: PMC11658977 DOI: 10.3389/fphar.2024.1449322] [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: 06/14/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
Background Haoqin Qingdan decoction (HQQD), composed of eleven herbs, is a traditional Chinese formula widely recognized for its efficacy in treating pulmonary inflammation induced by viral infections. Despite its extensive use, the potential pulmonary and intestinal protective effects of HQQD on influenza viral pneumonia (IVP) and the underlying molecular mechanisms remain unclear. Materials and Methods Ultra-high-performance liquid chromatography coupled with mass spectrometry (UHPLC-MS) was employed to identify the major chemical constituents of the prescription. Subsequently, network analysis was conducted to predict the potential therapeutic targets of HQQD in IVP. The mechanisms by which HQQD mitigates lung and intestinal damage were further elucidated by assessing NP protein expression, inflammatory factors, TLR7/MyD88/NF-κB signaling pathway mRNAs and proteins, and through intestinal flora analysis. Results The protective effects of HQQD on pulmonary and intestinal injuries induced by IVP were thoroughly investigated using comprehensive network analysis, signaling pathway validation, and gut microflora analysis. UHPLC-MS analysis identified the primary chemical constituents. Validation experiments demonstrated a significant reduction in NP protein expression in the lungs. HQQD notably alleviated immune damage in the lungs and intestines of mice by inhibiting NP protein expression and the release of inflammatory factors such as interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ); downregulating the expression levels of TLR7, MyD88, and phospho-NF-κB p65 (p-p65); lowering serum LPS levels; and reducing the relative abundance of Proteobacteria. Conclusion HQQD exerts therapeutic effects against influenza viral pneumonia through antiviral and anti-inflammatory mechanisms and by remodeling the intestinal flora. This study provides initial insights into the "gut-lung" axis mechanism of HQQD in combating respiratory influenza virus infection.
Collapse
Affiliation(s)
- Xi Lin
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jian Lin
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lichun Ji
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiaona Zhang
- Guangzhou University of Chinese Medicine, Guangzhou, China
- Chinese Medicine Guangdong Laboratory, Guangdong, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yezi Zhang
- The Third Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junbin Hong
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Geng Li
- Chinese Medicine Guangdong Laboratory, Guangdong, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, China
- Animal Experiment Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xingdong Lin
- The Third Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
7
|
Snyder JD, Yoon TW, Lee S, Halder P, Fitzpatrick EA, Yi AK. Protein kinase D1 in myeloid lineage cells contributes to the accumulation of CXCR3 +CCR6 + nonconventional Th1 cells in the lungs and potentiates hypersensitivity pneumonitis caused by S. rectivirgula. Front Immunol 2024; 15:1403155. [PMID: 39464896 PMCID: PMC11502317 DOI: 10.3389/fimmu.2024.1403155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 09/18/2024] [Indexed: 10/29/2024] Open
Abstract
Introduction Hypersensitivity pneumonitis (HP) is an extrinsic allergic alveolitis characterized by inflammation of the interstitium, bronchioles, and alveoli of the lung that leads to granuloma formation. We previously found that activation of protein kinase D1 (PKD1) in the lungs following exposures to Saccharopolyspora rectivirgula contributes to the acute pulmonary inflammation, IL-17A expression in the lungs, and development of HP. In the present study, we investigated whether PKD1 in myeloid-lineage cells affects the pathogenic course of the S. rectivirgula-induced HP. Methods Mice were exposed intranasally to S. rectivirgula once or 3 times/week for 3 weeks. The protein and mRNA expression levels of cytokines/chemokines were detected by enzyme-linked immunosorbent assay and quantitative real-time PCR, respectively. Flow cytometry was used to detect the different types of immune cells and the levels of surface proteins. Lung tissue sections were stained with hematoxylin and eosin, digital images were captured, and immune cells influx into the interstitial lung tissue were detected. Results Compared to control PKD1-sufficient mice, mice with PKD1 deficiency in myeloid-lineage cells (PKD1mKO) showed significantly suppressed expression of TNFα, IFNγ, IL-6, CCL2, CCL3, CCL4, CXCL1, CXCL2, and CXCL10 and neutrophilic alveolitis after single intranasal exposure to S. rectivirgula. Substantially reduced levels of alveolitis and granuloma formation were observed in the PKD1mKO mice repeatedly exposed to S. rectivirgula for 3 weeks. In addition, expression levels of the Th1/Th17 polarizing cytokines and chemokines such as IFNγ, IL-17A, CXCL9, CXCL10, CXCL11, and CCL20 in lungs were significantly reduced in the PKD1mKO mice repeatedly exposed to S. rectivirgula. Moreover, accumulation of CXCR3+CCR6+ nonconventional Th1 in the lungs were significantly reduced in PKD1mKO mice repeatedly exposed to S. rectivirgula. Discussion Our results demonstrate that PKD1 in myeloid-lineage cells plays an essential role in the development and progress of HP caused by repeated exposure to S. rectivirgula by contributing Th1/Th17 polarizing proinflammatory responses, alveolitis, and accumulation of pathogenic nonconventional Th1 cells in the lungs.
Collapse
Affiliation(s)
- John D. Snyder
- Integrated Biomedical Science Graduate Program, The University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Tae Won Yoon
- Integrated Biomedical Science Graduate Program, The University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sangmin Lee
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Priyanka Halder
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Elizabeth Ann Fitzpatrick
- Integrated Biomedical Science Graduate Program, The University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ae-Kyung Yi
- Integrated Biomedical Science Graduate Program, The University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
8
|
Feng H, Zhang K, Zhang J, Wang X, Guo Z, Wang L, Chen F, Han S, Li J. The alleviating effect of Phillygenin on the regulation of respiratory microbiota and its metabolites in IBV-infected broilers by inhibiting the TLR7/MyD88/NF-κB axis. FASEB J 2024; 38:e23882. [PMID: 39143727 DOI: 10.1096/fj.202400168rr] [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: 01/22/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 08/16/2024]
Abstract
Phillygenin (PHI) is an active ingredient derived from the leaf of Forsythia suspensa that has been found to alleviate inflammation and peroxidation response. Avian infectious bronchitis (IB) is a major threat to poultry industry viral respiratory tract disease that infected with infectious bronchitis virus (IBV). This study investigated the protection of PHI to CEK cell and broiler's tracheal injury triggered by avian infectious bronchitis virus (IBV). The results showed that IBV infection did not cause serious clinical symptoms and slowing-body weight in PHI-treated broilers. The expression of virus loads, pro-inflammation factors (IL-6, TNF-α, and IL-1β) in CEK cell, and tracheas were decreased compared to the IBV group, exhibiting its potent anti-inflammation. Mechanistically, the study demonstrated that the inhibition of TLR7/MyD88/NF-κB pathway was mainly involved in the protection effect of PHI to inflammation injury. Interestingly, a higher abundance of Firmicutes and Lactobacillus in respiratory tract was observed in PHI-treated broilers than in the IBV group. Significant differences were observed between the IBV group and PHI-treated group in the Ferroptosis, Tryptophan metabolism, and Glutathione metabolism pathways. PHI exhibited potent protection effect on IBV infection and alleviated inflammation injury, mainly through inhibiting TLR7/MyD88/NF-κB pathway. The study encourages further development of PHI, paving the way to its clinical use as a new candidate drug to relieve IBV-induced respiratory symptoms.
Collapse
Affiliation(s)
- Haipeng Feng
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Kang Zhang
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Jingyan Zhang
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xuezhi Wang
- Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Zhiting Guo
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Lei Wang
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Fubing Chen
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Songwei Han
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Jianxi Li
- Engineering & Technology Research Center of Traditional Chinese Veterinary Medicine of Gansu Province, Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
9
|
Smok-Kalwat J, Mertowska P, Mertowski S, Góźdź S, Grywalska E. Toll-like Receptors: Key Players in Squamous Cell Carcinoma Progression. J Clin Med 2024; 13:4531. [PMID: 39124797 PMCID: PMC11313009 DOI: 10.3390/jcm13154531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/30/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Background/Objectives Lung squamous cell carcinoma (SCC) is one of the major subtypes of lung cancer, characterized by diverse molecular pathways and variable clinical outcomes. This study focused on assessing the levels of TLR-2, TLR-3, TLR-4, TLR-7, TLR-8, and TLR-9 on peripheral blood lymphocytes in patients with newly diagnosed SCC compared to a group of healthy controls, in the context of disease development and patient survival, conducted over three years. The study aimed to investigate the differences in TLR expression between SCC patients and healthy people and to understand their role in the development of the disease and patient survival over three years. Methods: The study included the assessment of TLR-2, TLR-3, TLR-4, TLR-7, TLR-8, and TLR-9 levels on peripheral blood lymphocytes in patients with newly diagnosed SCC and in the control group. The expression of TLRs was measured using flow cytometry, and the soluble forms of the tested TLRs were measured using enzyme-linked immunosorbent assays. All the analyses were conducted over a three-year period from the time patients were recruited to the study. The obtained test results were statistically analyzed. Results: Results showed statistically significant differences in TLR expression between the groups, with higher TLR levels correlating with an advanced stage of disease and poorer survival rates. This suggests that the deregulation of TLR levels may be involved in promoting tumor development and influencing its microenvironment. Conclusions: The research, conducted over three years, indicates the need for further research on the role of TLRs in SCC, including their potential use as therapeutic targets and biomarkers. This may help to increase the effectiveness of standard treatments and improve clinical outcomes in patients with SCC.
Collapse
Affiliation(s)
- Jolanta Smok-Kalwat
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwińskiego Street, 25-734 Kielce, Poland; (J.S.-K.); (S.G.)
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland; (P.M.); (E.G.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland; (P.M.); (E.G.)
| | - Stanisław Góźdź
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwińskiego Street, 25-734 Kielce, Poland; (J.S.-K.); (S.G.)
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland; (P.M.); (E.G.)
| |
Collapse
|
10
|
Chen YC, Chen JH, Tsai CF, Wu CY, Chang CN, Wu CT, Yeh WL. Protective effects of paeonol against cognitive impairment in lung diseases. J Pharmacol Sci 2024; 155:101-112. [PMID: 38797534 DOI: 10.1016/j.jphs.2024.04.006] [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: 02/16/2024] [Revised: 04/14/2024] [Accepted: 04/30/2024] [Indexed: 05/29/2024] Open
Abstract
Pulmonary inflammation may lead to neuroinflammation resulting in neurological dysfunction, and it is associated with a variety of acute and chronic lung diseases. Paeonol is a herbal phenolic compound with anti-inflammatory and anti-oxidative properties. The aim of this study is to understand the beneficial effects of paeonol on cognitive impairment, pulmonary inflammation and its underlying mechanisms. Pulmonary inflammation-associated cognitive deficit was observed in TNFα-stimulated mice, and paeonol mitigated the cognitive impairment by reducing the expressions of interleukin (IL)-1β, IL-6, and NOD-like receptor family pyrin domain-containing 3 (NLRP3) in hippocampus. Moreover, elevated plasma miR-34c-5p in lung-inflamed mice was also reduced by paeonol. Pulmonary inflammation induced by intratracheal instillation of TNFα in mice resulted in immune cells infiltration in bronchoalveolar lavage fluid, pulmonary edema, and acute fibrosis, and these inflammatory responses were alleviated by paeonol orally. In MH-S alveolar macrophages, tumor necrosis factor (TNF) α- and phorbol myristate acetate (PMA)-induced inflammasome activation was ameliorated by paeonol. In addition, the expressions of antioxidants were elevated by paeonol, and reactive oxygen species production was reduced. In this study, paeonol demonstrates protective effects against cognitive deficits and pulmonary inflammation by exerting anti-inflammatory and anti-oxidative properties, suggesting a powerful benefit as a potential therapeutic agent.
Collapse
Affiliation(s)
- Yen-Chang Chen
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan
| | - Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, No. 88, Sec. 1, Fengxing Road, Taichung, 427213, Taiwan
| | - Cheng-Fang Tsai
- Department of Medical Laboratory Science and Biotechnology, Asia University, No.500 Lioufeng Road, Taichung, 413305, Taiwan
| | - Chen-Yun Wu
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan
| | - Chen-Ni Chang
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan
| | - Chen-Teng Wu
- Department of Surgery, China Medical University Hospital, No. 2, Yude Road, Taichung, 404332, Taiwan
| | - Wei-Lan Yeh
- Institute of New Drug Development, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan; Department of Biochemistry, School of Medicine, China Medical University, No.91 Hsueh-Shih Road, Taichung, 404333, Taiwan.
| |
Collapse
|
11
|
Cheng ZX, Zhang J. Exploring the Role of Gut-Lung Interactions in COPD Pathogenesis: A Comprehensive Review on Microbiota Characteristics and Inflammation Modulation. CHRONIC OBSTRUCTIVE PULMONARY DISEASES (MIAMI, FLA.) 2024; 11:311-325. [PMID: 38563747 PMCID: PMC11216226 DOI: 10.15326/jcopdf.2023.0442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 04/04/2024]
Abstract
Chronic obstructive pulmonary disease (COPD) is a paramount contributor to global morbidity and mortality. Over the past decade, the concept of the "gut-lung axis" has emerged, offering a lens through which to examine the intricate interplay between the host, microbiome, and respiratory diseases, including COPD. An expanding body of evidence underscores that the composition of both the gastrointestinal and respiratory microbiome deviates in COPD patients compared to healthy individuals, leading to distinct host immune responses and clinical manifestations. The objective of this review is to provide a concise overview of the role both gut and respiratory microbiome play in the development of COPD. This was accomplished by compiling current literature on the microbiome profile in stable and exacerbated cases of COPD, as well as exploring the biological mechanisms through a discussion of relevant experiments conducted on murine models. Hallmark characteristics of the microbial profile in COPD encompass reduced Prevotella species in the respiratory microbiome, culminating in a loss of anti-inflammatory protection, and diminished Bacteroidetes in the gut microbiome, leading to a decrease in protective short-chain fatty acids. The proliferation of Proteobacteria, particularly the Haemophilus species, Moraxellaspecies, and Pseudomonas species contribute to COPD pathologies via recognition of proinflammatory lipopolysaccharide via Toll-like receptors. As a consequence, deteriorated pulmonary function, enhanced severity, increased onset of exacerbations, and elevated mortality were observed.
Collapse
Affiliation(s)
- Zi-Xuan Cheng
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Shanghai, China
- *PhD candidate
| | - Jing Zhang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Shanghai, China
| |
Collapse
|
12
|
Bianchi F, Le Noci V, Bernardo G, Gagliano N, Colombo G, Sommariva M, Palazzo M, Dalle-Donne I, Milzani A, Pupa S, Tagliabue E, Sfondrini L. Cigarette smoke sustains immunosuppressive microenvironment inducing M2 macrophage polarization and viability in lung cancer settings. PLoS One 2024; 19:e0303875. [PMID: 38776331 PMCID: PMC11111031 DOI: 10.1371/journal.pone.0303875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 05/01/2024] [Indexed: 05/24/2024] Open
Abstract
BACKGROUND It is amply demonstrated that cigarette smoke (CS) has a high impact on lung tumor progression worsening lung cancer patient prognosis and response to therapies. Alteration of immune cell types and functions in smokers' lungs have been strictly related with smoke detrimental effects. However, the role of CS in dictating an inflammatory or immunosuppressive lung microenvironment still needs to be elucidated. Here, we investigated the effect of in vitro exposure to cigarette smoke extract (CSE) focusing on macrophages. METHODS Immortalized murine macrophages RAW 264.7 cells were cultured in the presence of CS extract and their polarization has been assessed by Real-time PCR and cytofluorimetric analysis, viability has been assessed by SRB assay and 3D-cultures and activation by exposure to Poly(I:C). Moreover, interaction with Lewis lung carcinoma (LLC1) murine cell models in the presence of CS extract were analyzed by confocal microscopy. RESULTS Obtained results indicate that CS induces macrophages polarization towards the M2 phenotype and M2-phenotype macrophages are resistant to the CS toxic activity. Moreover, CS impairs TLR3-mediated M2-M1 phenotype shift thus contributing to the M2 enrichment in lung smokers. CONCLUSIONS These findings indicate that, in lung cancer microenvironment of smokers, CS can contribute to the M2-phenotype macrophages prevalence by different mechanisms, ultimately, driving an anti-inflammatory, likely immunosuppressive, microenvironment in lung cancer smokers.
Collapse
Affiliation(s)
- Francesca Bianchi
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS San Donato, Milan, Italy
| | - Valentino Le Noci
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Giancarla Bernardo
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | - Nicoletta Gagliano
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | | | - Michele Sommariva
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Unit of Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michele Palazzo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Aldo Milzani
- Department of Biosciences, University of Milan, Milan, Italy
| | - Serenella Pupa
- Unit of Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elda Tagliabue
- Unit of Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Lucia Sfondrini
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
- Unit of Microenvironment and Biomarkers of Solid Tumors, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
13
|
Smok-Kalwat J, Mertowska P, Mertowski S, Góźdź S, Korona-Głowniak I, Kwaśniewski W, Grywalska E. Analysis of Selected Toll-like Receptors in the Pathogenesis and Advancement of Non-Small-Cell Lung Cancer. J Clin Med 2024; 13:2793. [PMID: 38792335 PMCID: PMC11122486 DOI: 10.3390/jcm13102793] [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: 04/11/2024] [Revised: 04/29/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: Non-small-cell lung cancer (NSCLC) represents a significant global health challenge, contributing to numerous cancer deaths. Despite advances in diagnostics and therapy, identifying reliable biomarkers for prognosis and therapeutic stratification remains difficult. Toll-like receptors (TLRs), crucial for innate immunity, now show potential as contributors to cancer development and progression. This study aims to investigate the role of TLR expression as potential biomarkers in the development and progression of NSCLC. (2) Materials and Methods: The study was conducted on 89 patients diagnosed with NSCLC and 40 healthy volunteers, for whom the prevalence of TLR2, TLR3, TLR4, TLR7, TLR8, and TLR9 was assessed on selected subpopulations of T and B lymphocytes in the peripheral blood of recruited patients along with the assessment of their serum concentration. (3) Result: Our study showed several significant changes in NSCLC patients at the beginning of the study. This resulted in a 5-year follow-up of changes in selected TLRs in recruited patients. Due to the high mortality rate of NSCLC patients, only 16 patients survived the 5 years. (4) Conclusions: The results suggest that TLRs may constitute real biomarker molecules that may be used for future prognostic purposes in NSCLC. However, further validation through prospective clinical and functional studies is necessary to confirm their clinical utility. These conclusions may lead to better risk stratification and tailored interventions, benefiting NSCLC patients and bringing medicine closer to precision.
Collapse
Affiliation(s)
- Jolanta Smok-Kalwat
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland; (J.S.-K.); (S.G.)
| | - Paulina Mertowska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Sebastian Mertowski
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland; (S.M.); (E.G.)
| | - Stanisław Góźdź
- Department of Clinical Oncology, Holy Cross Cancer Centre, 3 Artwinskiego Street, 25-734 Kielce, Poland; (J.S.-K.); (S.G.)
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University of Kielce, IX Wieków Kielc 19A, 25-317 Kielce, Poland
| | - Izabela Korona-Głowniak
- Department of Pharmaceutical Microbiology, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Wojciech Kwaśniewski
- Department of Gynecologic Oncology and Gynecology, Medical University of Lublin, Staszica 16 Street, 20-081 Lublin, Poland;
| | - Ewelina Grywalska
- Department of Experimental Immunology, Medical University of Lublin, 4a Chodzki Street, 20-093 Lublin, Poland; (S.M.); (E.G.)
| |
Collapse
|
14
|
Colvin KL, Wolter-Warmerdam K, Hickey F, Yeager ME. Altered peripheral blood leukocyte subpopulations, function, and gene expression in children with Down syndrome: implications for respiratory tract infection. Eur J Med Genet 2024; 68:104922. [PMID: 38325643 DOI: 10.1016/j.ejmg.2024.104922] [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: 06/06/2023] [Revised: 12/12/2023] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
OBJECTIVES We tested the hypothesis that aberrant expression of Hsa21-encoded interferon genes in peripheral blood immune cells would correlate to immune cell dysfunction in children with Down syndrome (DS). STUDY DESIGN We performed flow cytometry to quantify peripheral blood leukocyte subtypes and measured their ability to migrate and phagocytose. In matched samples, we measured gene expression levels for constituents of interferon signaling pathways. We screened 49 children, of which 29 were individuals with DS. RESULTS We show that the percentages of two peripheral blood myeloid cell subtypes (alternatively-activated macrophages and low-density granulocytes) in children with DS differed significantly from typical children, children with DS circulate a very different pattern of cytokines vs. typical individuals, and higher expression levels of type III interferon receptor Interleukin-10Rb in individuals with DS correlated with reduced migratory and phagocytic capacity of macrophages. CONCLUSIONS Increased susceptibility to severe and chronic infection in children with DS may result from inappropriate numbers and subtypes of immune cells that are phenotypically and functionally altered due to trisomy 21 associated interferonopathy.
Collapse
Affiliation(s)
- Kelley L Colvin
- Department of Bioengineering, University of Colorado Denver, Aurora, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Denver, Aurora, USA
| | | | - Francis Hickey
- Anna and John J. Sie Center for Down Syndrome, Children's Hospital Colorado, Aurora, USA; Department of Pediatrics, University of Colorado School of Medicine, Aurora, USA
| | - Michael E Yeager
- Department of Bioengineering, University of Colorado Denver, Aurora, USA; Linda Crnic Institute for Down Syndrome, University of Colorado Denver, Aurora, USA.
| |
Collapse
|
15
|
Miao S, Qiu H. The microbiome in the pathogenesis of lung cancer: The role of microbiome in lung cancer pathogenesis. APMIS 2024; 132:68-80. [PMID: 37974493 DOI: 10.1111/apm.13359] [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: 06/16/2023] [Accepted: 10/23/2023] [Indexed: 11/19/2023]
Abstract
As one of the malignant tumors with high incidence rate and high mortality, lung cancer seriously threatens the life safety of patients. Research shows that microorganisms are closely related to lung cancer. The microbiome is symbiotic with the host and plays a vital role in the functions of the human body. Microbiota dysbiosis is correlated with development of lung cancer. However, the underlying mechanisms are poorly understood. This paper summarizes the composition characteristics of the gut-lung axis microbiome and intratumoral microbiome in patients with lung cancer. We then expound five potential carcinogenic mechanisms based on microorganisms, such as genotoxicity, metabolism, inflammation, immune response, and angiogenesis. Next, we list three high-throughput sequencing methods, and finally looks forward to the prospect of microorganisms as novel targets for early diagnosis and treatment of lung cancer.
Collapse
Affiliation(s)
- Sainan Miao
- School of Nursing, Anhui Medical University, Hefei, China
| | - Huan Qiu
- School of Nursing, Anhui Medical University, Hefei, China
| |
Collapse
|
16
|
Mallek NM, Martin EM, Dailey LA, McCullough SD. Liquid application dosing alters the physiology of air-liquid interface (ALI) primary human bronchial epithelial cell/lung fibroblast co-cultures and in vitro testing relevant endpoints. FRONTIERS IN TOXICOLOGY 2024; 5:1264331. [PMID: 38464699 PMCID: PMC10922929 DOI: 10.3389/ftox.2023.1264331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 12/14/2023] [Indexed: 03/12/2024] Open
Abstract
Differentiated primary human bronchial epithelial cell (dpHBEC) cultures grown under air-liquid interface (ALI) conditions exhibit key features of the human respiratory tract and are thus critical for respiratory research as well as efficacy and toxicity testing of inhaled substances (e.g., consumer products, industrial chemicals, and pharmaceuticals). Many inhalable substances (e.g., particles, aerosols, hydrophobic substances, reactive substances) have physiochemical properties that challenge their evaluation under ALI conditions in vitro. Evaluation of the effects of these methodologically challenging chemicals (MCCs) in vitro is typically conducted by "liquid application," involving the direct application of a solution containing the test substance to the apical, air-exposed surface of dpHBEC-ALI cultures. We report that the application of liquid to the apical surface of a dpHBEC-ALI co-culture model results in significant reprogramming of the dpHBEC transcriptome and biological pathway activity, alternative regulation of cellular signaling pathways, increased secretion of pro-inflammatory cytokines and growth factors, and decreased epithelial barrier integrity. Given the prevalence of liquid application in the delivery of test substances to ALI systems, understanding its effects provides critical infrastructure for the use of in vitro systems in respiratory research as well as in the safety and efficacy testing of inhalable substances.
Collapse
Affiliation(s)
- Nicholas M. Mallek
- Curriculum in Toxicology and Environmental Medicine, University of North Carolina, Chapel Hill, NC, United States
| | - Elizabeth M. Martin
- Epigenetics and Stem Cell Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Durham, NC, United States
| | - Lisa A. Dailey
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Chapel Hill, NC, United States
| | - Shaun D. McCullough
- Public Health and Integrated Toxicology Division, Center for Public Health and Environmental Assessment, United States Environmental Protection Agency, Chapel Hill, NC, United States
- Exposure and Protection, RTI International, Durham, NC, United States
| |
Collapse
|
17
|
Ge X, Xu T, Wang M, Gao L, Tang Y, Zhang N, Zheng R, Zeng W, Chen G, Zhang B, Dai Y, Zhang Y. Chalcone-derivative L6H21 attenuates the OVA-induced asthma by targeting MD2. Eur J Med Res 2024; 29:65. [PMID: 38245791 PMCID: PMC10799361 DOI: 10.1186/s40001-023-01630-5] [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: 08/24/2023] [Accepted: 12/28/2023] [Indexed: 01/22/2024] Open
Abstract
Asthma represents a significant global challenge that affects individuals across all age groups and imposes substantial social and economic burden. Due to heterogeneity of the disease, not all patients obtain benefit with current treatments. The objective of this study was to explore the impact of MD2 on the progression of asthma using L6H21, a novel MD2 inhibitor, to identify potential targets and drug candidates for asthma treatment. To establish an asthma-related murine model and evaluate the effects of L6H21, ovalbumin (OVA) was used to sensitize and challenge mice. Pathological changes were examined with various staining techniques, such as H&E staining, glycogen staining, and Masson staining. Inflammatory cell infiltration and excessive cytokine secretion were evaluated by analyzing BALF cell count, RT-PCR, and ELISA. The TLR4/MD2 complex formation, as well as the activation of the MAPK and NF-кB pathways, was examined using western blot and co-IP. Treatment with L6H21 demonstrated alleviation of increased airway resistance, lung tissue injury, inflammatory cell infiltration and excessive cytokine secretion triggered by OVA. In addition, it also ameliorated mucus production and collagen deposition. In the L6H21 treatment group, inhibition of MAPK and NF-кB activation was observed, along with the disruption of TLR4/MD2 complex formation, in contrast to the model group. Thus, L6H21 effectively reduced the formation of the MD2 and TLR4 complex induced by OVA in a dose-dependent manner. This reduction resulted in the attenuation of MAPKs/NF-κB activation, enhanced suppression of inflammatory factor secretion, reduced excessive recruitment of inflammatory cells, and ultimately mitigated airway damage. MD2 emerges as a crucial target for asthma treatment, and L6H21, as an MD2 inhibitor, shows promise as a potential drug candidate for the treatment of asthma.
Collapse
Affiliation(s)
- Xiangting Ge
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China
| | - Tingting Xu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Division of Pulmonary Medicine, Key Laboratory of Heart and Lung, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Meiyan Wang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Lijiao Gao
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yue Tang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Ningjie Zhang
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Rui Zheng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Weimin Zeng
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Gaozhi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Bing Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China.
| | - Yuanrong Dai
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Yali Zhang
- Affiliated Yueqing Hospital, Wenzhou Medical University, Wenzhou, 325600, Zhejiang, China.
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
18
|
Casella C, Kiles F, Urquhart C, Michaud DS, Kirwa K, Corlin L. Methylomic, Proteomic, and Metabolomic Correlates of Traffic-Related Air Pollution in the Context of Cardiorespiratory Health: A Systematic Review, Pathway Analysis, and Network Analysis. TOXICS 2023; 11:1014. [PMID: 38133415 PMCID: PMC10748071 DOI: 10.3390/toxics11121014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/18/2023] [Accepted: 12/06/2023] [Indexed: 12/23/2023]
Abstract
A growing body of literature has attempted to characterize how traffic-related air pollution (TRAP) affects molecular and subclinical biological processes in ways that could lead to cardiorespiratory disease. To provide a streamlined synthesis of what is known about the multiple mechanisms through which TRAP could lead to cardiorespiratory pathology, we conducted a systematic review of the epidemiological literature relating TRAP exposure to methylomic, proteomic, and metabolomic biomarkers in adult populations. Using the 139 papers that met our inclusion criteria, we identified the omic biomarkers significantly associated with short- or long-term TRAP and used these biomarkers to conduct pathway and network analyses. We considered the evidence for TRAP-related associations with biological pathways involving lipid metabolism, cellular energy production, amino acid metabolism, inflammation and immunity, coagulation, endothelial function, and oxidative stress. Our analysis suggests that an integrated multi-omics approach may provide critical new insights into the ways TRAP could lead to adverse clinical outcomes. We advocate for efforts to build a more unified approach for characterizing the dynamic and complex biological processes linking TRAP exposure and subclinical and clinical disease and highlight contemporary challenges and opportunities associated with such efforts.
Collapse
Affiliation(s)
- Cameron Casella
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Frances Kiles
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Catherine Urquhart
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Dominique S. Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
| | - Kipruto Kirwa
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
- Department of Environmental Health, Boston University School of Public Health, Boston, MA 02118, USA
| | - Laura Corlin
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA; (C.C.); (F.K.); (C.U.); (D.S.M.); (K.K.)
- Department of Civil and Environmental Engineering, Tufts University School of Engineering, Medford, MA 02155, USA
| |
Collapse
|
19
|
Pourova J, Dias P, Pour M, Bittner Fialová S, Czigle S, Nagy M, Tóth J, Balázs VL, Horváth A, Csikós E, Farkas Á, Horváth G, Mladěnka P. Proposed mechanisms of action of herbal drugs and their biologically active constituents in the treatment of coughs: an overview. PeerJ 2023; 11:e16096. [PMID: 37901462 PMCID: PMC10607228 DOI: 10.7717/peerj.16096] [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: 03/24/2023] [Accepted: 08/24/2023] [Indexed: 10/31/2023] Open
Abstract
Various medicinal plants find their use in cough treatment, based on traditions and long-term experience. Pharmacological principles of their action, however, are much less known. Herbal drugs usually contain a mixture of potentially active compounds, which can manifest diverse effects. Expectorant or antitussive effects, which can be accompanied by others, such as anti-inflammatory or antibacterial, are probably the most important in the treatment of coughs. The aim of this review is to summarize the current state of knowledge of the effects of medicinal plants or their constituents on cough, based on reliable pharmacological studies. First, a comprehensive description of each effect is provided in order to explain the possible mechanism of action in detail. Next, the results related to individual plants and substances are summarized and critically discussed based on pharmacological in vivo and in vitro investigation.
Collapse
Affiliation(s)
- Jana Pourova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University Prague, Hradec Králové, Czech Republic
| | - Patricia Dias
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University Prague, Hradec Králové, Czech Republic
| | - Milan Pour
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy, Charles University Prague, Hradec Králové, Czech Republic
| | - Silvia Bittner Fialová
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovak Republic
| | - Szilvia Czigle
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovak Republic
| | - Milan Nagy
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovak Republic
| | - Jaroslav Tóth
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University Bratislava, Bratislava, Slovak Republic
| | | | - Adrienn Horváth
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Eszter Csikós
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Ágnes Farkas
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Györgyi Horváth
- Department of Pharmacognosy, Faculty of Pharmacy, University of Pécs, Pécs, Hungary
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University Prague, Hradec Králové, Czech Republic
| |
Collapse
|
20
|
Kang C, Li X, Liu P, Liu Y, Niu Y, Zeng X, Zhao H, Liu J, Qiu S. Tolerogenic dendritic cells and TLR4/IRAK4/NF-κB signaling pathway in allergic rhinitis. Front Immunol 2023; 14:1276512. [PMID: 37915574 PMCID: PMC10616250 DOI: 10.3389/fimmu.2023.1276512] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/05/2023] [Indexed: 11/03/2023] Open
Abstract
Dendritic cells (DCs), central participants in the allergic immune response, can capture and present allergens leading to allergic inflammation in the immunopathogenesis of allergic rhinitis (AR). In addition to initiating antigen-specific immune responses, DCs induce tolerance and modulate immune homeostasis. As a special type of DCs, tolerogenic DCs (tolDCs) achieve immune tolerance mainly by suppressing effector T cell responses and inducing regulatory T cells (Tregs). TolDCs suppress allergic inflammation by modulating immune tolerance, thereby reducing symptoms of AR. Activation of the TLR4/IRAK4/NF-κB signaling pathway contributes to the release of inflammatory cytokines, and inhibitors of this signaling pathway induce the production of tolDCs to alleviate allergic inflammatory responses. This review focuses on the relationship between tolDCs and TLR4/IRAK4/NF-κB signaling pathway with AR.
Collapse
Affiliation(s)
- Chenglin Kang
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
- Department of Otolaryngology, Longgang E.N.T Hospital and Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
- Department of Otolaryngology, Second People’s Hospital of Gansu Province, Lanzhou, China
| | - Xiaomei Li
- Department of Otolaryngology, Second People’s Hospital of Gansu Province, Lanzhou, China
| | - Peng Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
| | - Yue Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
| | - Yuan Niu
- Department of Neurology, Second People’s Hospital of Gansu Province, Lanzhou, China
| | - Xianhai Zeng
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
- Department of Otolaryngology, Longgang E.N.T Hospital and Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
| | - Hailiang Zhao
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
- Department of Otolaryngology, Longgang E.N.T Hospital and Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
| | - Jiangqi Liu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
- Department of Otolaryngology, Longgang E.N.T Hospital and Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
| | - Shuqi Qiu
- Department of Graduate and Scientific Research, Zunyi Medical University Zhuhai Campus, Zhuhai, China
- Department of Otolaryngology, Longgang E.N.T Hospital and Shenzhen Key Laboratory of E.N.T, Institute of E.N.T Shenzhen, Shenzhen, China
| |
Collapse
|
21
|
Do KTH, Willenzon S, Ristenpart J, Janssen A, Volz A, Sutter G, Förster R, Bošnjak B. The effect of Toll-like receptor agonists on the immunogenicity of MVA-SARS-2-S vaccine after intranasal administration in mice. Front Cell Infect Microbiol 2023; 13:1259822. [PMID: 37854858 PMCID: PMC10580083 DOI: 10.3389/fcimb.2023.1259822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/14/2023] [Indexed: 10/20/2023] Open
Abstract
Background and aims Modified Vaccinia virus Ankara (MVA) represents a promising vaccine vector for respiratory administration to induce protective lung immunity including tertiary lymphoid structure, the bronchus-associated lymphoid tissue (BALT). However, MVA expressing the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Spike protein (MVA-SARS-2-S) required prime-boost administration to induce high titers of anti-Spike antibodies in serum and bronchoalveolar lavage (BAL). As the addition of adjuvants enables efficient tailoring of the immune responses even to live vaccines, we tested whether Toll-like receptor (TLR)-agonists affect immune responses induced by a single dose of intranasally applied MVA-SARS-2-S. Methods We intranasally immunized C57BL/6 mice with MVA-SARS-2-S vaccine in the presence of either TLR3 agonist polyinosinic polycytidylic acid [poly(I:C)], TLR4 agonist bacterial lipopolysaccharide (LPS) from Escherichia coli, or TLR9 agonist CpG oligodeoxynucleotide (CpG ODN) 1826. At different time-points after immunization, we analyzed induced immune responses using flow cytometry, immunofluorescent microscopy, and ELISA. Results TLR agonists had profound effects on MVA-SARS-2-S-induced immune responses. At day 1 post intranasal application, the TLR4 agonist significantly affected MVA-induced activation of dendritic cells (DCs) within the draining bronchial lymph nodes, increasing the ratio of CD11b+CD86+ to CD103+CD86+ DCs. Nevertheless, the number of Spike-specific CD8+ T cells within the lungs at day 12 after vaccination was increased in mice that received MVA-SARS-2-S co-administered with TLR3 but not TLR4 agonists. TLR9 agonist did neither significantly affect MVA-induced DC activation nor the induction of Spike-specific CD8+ T cells but reduced both number and size of bronchus-associated lymphoid tissue. Surprisingly, the addition of all TLR agonists failed to boost the levels of Spike-specific antibodies in serum and bronchoalveolar lavage. Conclusions Our study indicates a potential role of TLR-agonists as a tool to modulate immune responses to live vector vaccines. Particularly TLR3 agonists hold a promise to potentiate MVA-induced cellular immune responses. On the other hand, additional research is necessary to identify optimal combinations of agonists that could enhance MVA-induced humoral responses.
Collapse
Affiliation(s)
- Kim Thi Hoang Do
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | | | - Anika Janssen
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Asisa Volz
- Institute for Virology, University of Veterinary Medicine Hannover, Hannover, Germany
- German Centre for Infection Research (DZIF), Munich, Germany
| | - Gerd Sutter
- German Centre for Infection Research (DZIF), Munich, Germany
- Division of Virology, Department of Veterinary Sciences, Ludwig Maximiliam University (LMU) Munich, Munich, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Centre for Infection Research (DZIF), Hannover, Germany
| | - Berislav Bošnjak
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| |
Collapse
|
22
|
Casella C, Kiles F, Urquhart C, Michaud DS, Kirwa K, Corlin L. Methylomic, proteomic, and metabolomic correlates of traffic-related air pollution: A systematic review, pathway analysis, and network analysis relating traffic-related air pollution to subclinical and clinical cardiorespiratory outcomes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.30.23296386. [PMID: 37873294 PMCID: PMC10592990 DOI: 10.1101/2023.09.30.23296386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
A growing body of literature has attempted to characterize how traffic-related air pollution (TRAP) affects molecular and subclinical biological processes in ways that could lead to cardiorespiratory disease. To provide a streamlined synthesis of what is known about the multiple mechanisms through which TRAP could lead cardiorespiratory pathology, we conducted a systematic review of the epidemiological literature relating TRAP exposure to methylomic, proteomic, and metabolomic biomarkers in adult populations. Using the 139 papers that met our inclusion criteria, we identified the omic biomarkers significantly associated with short- or long-term TRAP and used these biomarkers to conduct pathway and network analyses. We considered the evidence for TRAP-related associations with biological pathways involving lipid metabolism, cellular energy production, amino acid metabolism, inflammation and immunity, coagulation, endothelial function, and oxidative stress. Our analysis suggests that an integrated multi-omics approach may provide critical new insights into the ways TRAP could lead to adverse clinical outcomes. We advocate for efforts to build a more unified approach for characterizing the dynamic and complex biological processes linking TRAP exposure and subclinical and clinical disease, and highlight contemporary challenges and opportunities associated with such efforts.
Collapse
Affiliation(s)
- Cameron Casella
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Frances Kiles
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Catherine Urquhart
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Dominique S. Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Kipruto Kirwa
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Environmental Health, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Laura Corlin
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA 02111, USA
- Department of Civil and Environmental Engineering, Tufts University School of Engineering, Medford, MA 02155, USA
| |
Collapse
|
23
|
Mishra S, Gandhi D, Tiwari RR, Rajasekaran S. Beneficial role of kaempferol and its derivatives from different plant sources on respiratory diseases in experimental models. Inflammopharmacology 2023; 31:2311-2336. [PMID: 37410224 DOI: 10.1007/s10787-023-01282-1] [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: 01/19/2023] [Accepted: 06/02/2023] [Indexed: 07/07/2023]
Abstract
Respiratory illnesses impose a significant health burden and cause deaths worldwide. Despite many advanced strategies to improve patient outcomes, they are often less effective. There is still considerable room for improvement in the treatment of various respiratory diseases. In recent years, alternative medicinal agents derived from food plants have shown better beneficial effects against a wide variety of disease models, including cancer. In this regard, kaempferol (KMF) and its derivatives are the most commonly found dietary flavonols. They have been found to exhibit protective effects on multiple chronic diseases like diabetes, fibrosis, and so on. A few recent articles have reviewed the pharmacological actions of KMF in cancer, central nervous system diseases, and chronic inflammatory diseases. However, there is no comprehensive review that exists regarding the beneficial effects of KMF and its derivatives on both malignant- and non-malignant respiratory diseases. Many experimental studies reveal that KMF and its derivatives are helpful in managing a wide range of respiratory diseases, including acute lung injury, fibrosis, asthma, cancer, and chronic obstructive pulmonary disease, and their underlying molecular mechanisms. In addition, we also discussed the chemistry and sources, the absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties, methods to enhance bioavailability, as well as our perspective on future research with KMF and its derivatives.
Collapse
Affiliation(s)
- Sehal Mishra
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India
| | - Deepa Gandhi
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India
| | - Rajnarayan R Tiwari
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India
| | - Subbiah Rajasekaran
- Department of Biochemistry, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, 462030, India.
| |
Collapse
|
24
|
Mukherjee S, Patra R, Behzadi P, Masotti A, Paolini A, Sarshar M. Toll-like receptor-guided therapeutic intervention of human cancers: molecular and immunological perspectives. Front Immunol 2023; 14:1244345. [PMID: 37822929 PMCID: PMC10562563 DOI: 10.3389/fimmu.2023.1244345] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/07/2023] [Indexed: 10/13/2023] Open
Abstract
Toll-like receptors (TLRs) serve as the body's first line of defense, recognizing both pathogen-expressed molecules and host-derived molecules released from damaged or dying cells. The wide distribution of different cell types, ranging from epithelial to immune cells, highlights the crucial roles of TLRs in linking innate and adaptive immunity. Upon stimulation, TLRs binding mediates the expression of several adapter proteins and downstream kinases, that lead to the induction of several other signaling molecules such as key pro-inflammatory mediators. Indeed, extraordinary progress in immunobiological research has suggested that TLRs could represent promising targets for the therapeutic intervention of inflammation-associated diseases, autoimmune diseases, microbial infections as well as human cancers. So far, for the prevention and possible treatment of inflammatory diseases, various TLR antagonists/inhibitors have shown to be efficacious at several stages from pre-clinical evaluation to clinical trials. Therefore, the fascinating role of TLRs in modulating the human immune responses at innate as well as adaptive levels directed the scientists to opt for these immune sensor proteins as suitable targets for developing chemotherapeutics and immunotherapeutics against cancer. Hitherto, several TLR-targeting small molecules (e.g., Pam3CSK4, Poly (I:C), Poly (A:U)), chemical compounds, phytocompounds (e.g., Curcumin), peptides, and antibodies have been found to confer protection against several types of cancers. However, administration of inappropriate doses of such TLR-modulating therapeutics or a wrong infusion administration is reported to induce detrimental outcomes. This review summarizes the current findings on the molecular and structural biology of TLRs and gives an overview of the potency and promises of TLR-directed therapeutic strategies against cancers by discussing the findings from established and pipeline discoveries.
Collapse
Affiliation(s)
- Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Ritwik Patra
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| | - Payam Behzadi
- Department of Microbiology, Shahr-e-Qods Branch, Islamic Azad University, Tehran, Iran
| | - Andrea Masotti
- Research Laboratories, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| | - Alessandro Paolini
- Research Laboratories, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children’s Hospital-IRCCS, Rome, Italy
| |
Collapse
|
25
|
Joshi I, Devine AJ, Joshi R, Smith NJ, Varisco BM. A titratable murine model of progressive emphysema using tracheal porcine pancreatic elastase. Sci Rep 2023; 13:15259. [PMID: 37709810 PMCID: PMC10502133 DOI: 10.1038/s41598-023-41527-1] [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: 07/03/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023] Open
Abstract
Progressive emphysema often leads to end-stage lung disease. Most mouse models of emphysema are typically modest (i.e. cigarette smoke exposure), and changes over time are difficult to quantify. The tracheal porcine pancreatic elastase model (PPE) produces severe injury, but the literature is conflicted as to whether emphysema improves, is stable, or progresses over time. We hypothesized a threshold of injury below which repair would occur and above which emphysema would be stable or progress. We treated 8-week-old C57BL6 mixed sex mice with 0, 0.5, 2, or 4 activity units of PPE in 100 µL PBS and performed lung stereology at 21 and 84 days. There were no significant differences in weight gain or mouse health. Despite minimal emphysema at 21-days in the 0.5 units group (2.8 µm increased mean linear intercept, MLI), MLI increased by 4.6 µm between days 21 and 84 (p = 0.0007). In addition to larger MLI at 21 days in 2- and 4-unit groups, MLI increases from day 21 to 84 were 17.2 and 34 µm respectively (p = 0.002 and p = 0.0001). Total lung volume increased, and alveolar surface area decreased with time and injury severity. Contrary to our hypothesis, we found no evidence of alveolar repair over time. Airspace destruction was both progressive and accelerative. Future mechanistic studies in lung immunity, mechano-biology, senescence, and cell-specific changes may lead to novel therapies to slow or halt progressive emphysema in humans.
Collapse
Affiliation(s)
- Imani Joshi
- College of Arts and Sciences, Xavier University, Cincinnati, OH, USA
| | - Andrew J Devine
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Rashika Joshi
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Noah J Smith
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brian M Varisco
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- University of Cincinnati College of Medicine, Cincinnati, OH, USA.
- University of Arkansas for Medical Sciences, 1 Children's Way Slot 663, Little Rock, AR, 72202, USA.
| |
Collapse
|
26
|
Song Y, Gou Y, Gao J, Chen D, Zhang H, Zhao W, Qian F, Xu A, Shen Y. Lomerizine attenuates LPS-induced acute lung injury by inhibiting the macrophage activation through reducing Ca 2+ influx. Front Pharmacol 2023; 14:1236469. [PMID: 37693893 PMCID: PMC10484514 DOI: 10.3389/fphar.2023.1236469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/14/2023] [Indexed: 09/12/2023] Open
Abstract
Acute lung injury (ALI) and acute respiratory distress syndrome (ARDS) are life-threatening lung diseases with high mortality rates, predominantly attributable to acute and severe pulmonary inflammation. Lomerizine (LMZ) is a calcium channel blocker previously used in preventing and treating migraine. Here, we found that LMZ inhibited inflammatory responses and lung pathological injury by reducing pulmonary edema, neutrophil infiltration and pro-inflammatory cytokine production in lipopolysaccharide (LPS)-induced ALI mice. In vitro experiments, upon treating with LMZ, the expression of interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α was attenuated in macrophages. The phosphorylation of p38 MAPK, ERK1/2, JNK, and NF-κB p65 was inhibited after LMZ treatment. Furthermore, LPS-induced Ca2+ influx was reduced by treating with LMZ, which correlated with inhibition of pro-inflammatory cytokine production. And L-type Ca2+ channel agonist Bay K8644 (BK) could restore cytokine generation. In conclusion, our study demonstrated that LMZ alleviates LPS-induced ALI and is a potential agent for treating ALI/ARDS.
Collapse
Affiliation(s)
- Yunduan Song
- Department of Respiratory and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Department of Clinical Laboratory, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yusen Gou
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Jiameng Gao
- Department of Respiratory and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Dongxin Chen
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Haibo Zhang
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjuan Zhao
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Qian
- Shanghai Frontiers Science Center of Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Ajing Xu
- Department of Clinical Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao Shen
- Department of Respiratory and Critical Care Medicine, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| |
Collapse
|
27
|
Brown MA, Morgan SB, Donachie GE, Horton KL, Pavord ID, Arancibia-Cárcamo CV, Hinks TSC. Epithelial immune activation and intracellular invasion by non-typeable Haemophilus influenzae. Front Cell Infect Microbiol 2023; 13:1141798. [PMID: 37180449 PMCID: PMC10167379 DOI: 10.3389/fcimb.2023.1141798] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/04/2023] [Indexed: 05/16/2023] Open
Abstract
Type-2 low asthma affects 30-50% of people with severe asthma and includes a phenotype characterized by sputum neutrophilia and resistance to corticosteroids. Airways inflammation in type-2 low asthma or COPD is potentially driven by persistent bacterial colonization of the lower airways by bacteria such as non-encapsulated Haemophilus influenzae (NTHi). Although pathogenic in the lower airways, NTHi is a commensal of the upper airways. It is not known to what extent these strains can invade airway epithelial cells, persist intracellularly and activate epithelial cell production of proinflammatory cytokines, and how this differs between the upper and lower airways. We studied NTHi infection of primary human bronchial epithelial cells (PBECs), primary nasal epithelial cells (NECs) and epithelial cell lines from upper and lower airways. NTHi strains differed in propensity for intracellular and paracellular invasion. We found NTHi was internalized within PBECs at 6 h, but live intracellular infection did not persist at 24 h. Confocal microscopy and flow cytometry showed NTHi infected secretory, ciliated and basal PBECs. Infection of PBECs led to induction of CXCL8, interleukin (IL)-1β, IL-6 and TNF. The magnitude of cytokine induction was independent of the degree of intracellular invasion, either by differing strains or by cytochalasin D inhibition of endocytosis, with the exception of the inflammasome-induced mediator IL-1β. NTHi-induced activation of TLR2/4, NOD1/2 and NLR inflammasome pathways was significantly stronger in NECs than in PBECs. These data suggest that NTHi is internalized transiently by airway epithelial cells and has capacity to drive inflammation in airway epithelial cells.
Collapse
Affiliation(s)
- Mary A. Brown
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, Experimental Medicine Division, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Sophie B. Morgan
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, Experimental Medicine Division, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Gillian E. Donachie
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, Experimental Medicine Division, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Katie L. Horton
- School of Clinical and Experimental Sciences, University of Southampton Faculty of Medicine, Southampton, United Kingdom
| | - Ian D. Pavord
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, Experimental Medicine Division, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Carolina V. Arancibia-Cárcamo
- Translational Gastroenterology Unit, Nuffield Department of Medicine, Experimental Medicine, University of Oxford, Oxford, United Kingdom
| | - Timothy S. C. Hinks
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, Experimental Medicine Division, Nuffield Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
28
|
Kheradmand F, Zhang Y, Corry DB. Contribution of adaptive immunity to human COPD and experimental models of emphysema. Physiol Rev 2023; 103:1059-1093. [PMID: 36201635 PMCID: PMC9886356 DOI: 10.1152/physrev.00036.2021] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 02/01/2023] Open
Abstract
The pathophysiology of chronic obstructive pulmonary disease (COPD) and the undisputed role of innate immune cells in this condition have dominated the field in the basic research arena for many years. Recently, however, compelling data suggesting that adaptive immune cells may also contribute to the progressive nature of lung destruction associated with COPD in smokers have gained considerable attention. The histopathological changes in the lungs of smokers can be limited to the large or small airways, but alveolar loss leading to emphysema, which occurs in some individuals, remains its most significant and irreversible outcome. Critically, however, the question of why emphysema progresses in a subset of former smokers remained a mystery for many years. The recognition of activated and organized tertiary T- and B-lymphoid aggregates in emphysematous lungs provided the first clue that adaptive immune cells may play a crucial role in COPD pathophysiology. Based on these findings from human translational studies, experimental animal models of emphysema were used to determine the mechanisms through which smoke exposure initiates and orchestrates adaptive autoreactive inflammation in the lungs. These models have revealed that T helper (Th)1 and Th17 subsets promote a positive feedback loop that activates innate immune cells, confirming their role in emphysema pathogenesis. Results from genetic studies and immune-based discoveries have further provided strong evidence for autoimmunity induction in smokers with emphysema. These new findings offer a novel opportunity to explore the mechanisms underlying the inflammatory landscape in the COPD lung and offer insights for development of precision-based treatment to halt lung destruction.
Collapse
Affiliation(s)
- Farrah Kheradmand
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| | - Yun Zhang
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
| | - David B Corry
- Department of Medicine, Baylor College of Medicine, Houston, Texas
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas
- Biology of Inflammation Center, Baylor College of Medicine, Houston, Texas
- Center for Translational Research on Inflammatory Diseases (CTRID), Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas
| |
Collapse
|
29
|
Hamza FN, Daher S, Fakhoury HMA, Grant WB, Kvietys PR, Al-Kattan K. Immunomodulatory Properties of Vitamin D in the Intestinal and Respiratory Systems. Nutrients 2023; 15:nu15071696. [PMID: 37049536 PMCID: PMC10097244 DOI: 10.3390/nu15071696] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Vitamin D plays a crucial role in modulating the innate immune response by interacting with its intracellular receptor, VDR. In this review, we address vitamin D/VDR signaling and how it contributes to the regulation of intestinal and respiratory microbiota. We additionally review some components of the innate immune system, such as the barrier function of the pulmonary and intestinal epithelial membranes and secretion of mucus, with their respective modulation by vitamin D. We also explore the mechanisms by which this vitamin D/VDR signaling mounts an antimicrobial response through the transduction of microbial signals and the production of antimicrobial peptides that constitute one of the body’s first lines of defense against pathogens. Additionally, we highlight the role of vitamin D in clinical diseases, namely inflammatory bowel disease and acute respiratory distress syndrome, where excessive inflammatory responses and dysbiosis are hallmarks. Increasing evidence suggests that vitamin D supplementation may have potentially beneficial effects on those diseases.
Collapse
Affiliation(s)
- Fatheia N. Hamza
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Sarah Daher
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Hana M. A. Fakhoury
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
- Correspondence:
| | - William B. Grant
- Sunlight, Nutrition, and Health Research Center, P.O. Box 641603, San Francisco, CA 94164-1603, USA
| | - Peter R. Kvietys
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| | - Khaled Al-Kattan
- College of Medicine, Alfaisal University, P.O. Box 50927, Riyadh 11533, Saudi Arabia
| |
Collapse
|
30
|
Ma X, Wang F, Zhen L, Cai Q. Hsa_circ_0001204 modulates inflammatory response of macrophages infected by Mycobacterium tuberculosis via TLR4/NF-κB signalling pathway. Clin Exp Pharmacol Physiol 2023; 50:132-139. [PMID: 36048566 DOI: 10.1111/1440-1681.13716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 01/05/2023]
Abstract
Circular RNAs (circRNAs) play a vital role in the regulation of Mycobacterium tuberculosis (M.tb) by macrophages. In this project, the potential role of hsa_circ_0001204 in M.tb-infected macrophages is explored. Hsa_circ_0001204 was determined in the patients with tuberculosis (TB) and M.tb-infected macrophages. Its effect on the survival of M.tb and the apoptosis and inflammation of M.tb-infected macrophages was evaluated. Toll-like receptor 4/nuclear factor-κB (TLR4/NF-κB) signalling was detected by western blotting and immunofluorescence. TB patients and M.tb-infected THP-1 cells showed the significant downregulation of hsa_circ_0001204. Upregulating hsa_circ_0001204 reduced M.tb survival and suppressed the apoptosis and inflammatory response of THP-1 cells. The TLR4/NF-κB signalling pathway could be inhibited by hsa_circ_0001204 overexpression, which was activated by M.tb-infection. Hsa_circ_0001204 confers protective effects in M.tb-infected THP-1 cells, at least partly via the inhibition of TLR4/NF-κB signalling pathway.
Collapse
Affiliation(s)
- Xiaoqing Ma
- Department of Tuberculosis, Hangzhou Chest Hospital Affiliated to Zhejiang University Medical College, Hangzhou, China
| | - Fang Wang
- The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Libo Zhen
- Department of Tuberculosis, Hangzhou Chest Hospital Affiliated to Zhejiang University Medical College, Hangzhou, China
| | - Qingshan Cai
- Department of Tuberculosis, Hangzhou Chest Hospital Affiliated to Zhejiang University Medical College, Hangzhou, China
| |
Collapse
|
31
|
Zhu W, Zhang Y, Wang Y. Immunotherapy strategies and prospects for acute lung injury: Focus on immune cells and cytokines. Front Pharmacol 2022; 13:1103309. [PMID: 36618910 PMCID: PMC9815466 DOI: 10.3389/fphar.2022.1103309] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS) is a disastrous condition, which can be caused by a wide range of diseases, such as pneumonia, sepsis, traumas, and the most recent, COVID-19. Even though we have gained an improved understanding of acute lung injury/acute respiratory distress syndrome pathogenesis and treatment mechanism, there is still no effective treatment for acute lung injury/acute respiratory distress syndrome, which is partly responsible for the unacceptable mortality rate. In the pathogenesis of acute lung injury, the inflammatory storm is the main pathological feature. More and more evidences show that immune cells and cytokines secreted by immune cells play an irreplaceable role in the pathogenesis of acute lung injury. Therefore, here we mainly reviewed the role of various immune cells in acute lung injury from the perspective of immunotherapy, and elaborated the crosstalk of immune cells and cytokines, aiming to provide novel ideas and targets for the treatment of acute lung injury.
Collapse
Affiliation(s)
- Wenfang Zhu
- Department of Respiratory Medicine, Anhui Chest Hospital, Hefei, China
| | - Yiwen Zhang
- Department of Respiratory Medicine, Anhui Chest Hospital, Hefei, China,*Correspondence: Yiwen Zhang, ; Yinghong Wang,
| | - Yinghong Wang
- Department of Pharmacy, Anhui Provincial Cancer Hospital, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China,*Correspondence: Yiwen Zhang, ; Yinghong Wang,
| |
Collapse
|
32
|
Shukla MK, Dubey A, Pandey S, Singh SK, Gupta G, Prasher P, Chellappan DK, Oliver BG, Kumar D, Dua K. Managing Apoptosis in Lung Diseases using Nano-assisted Drug Delivery System. Curr Pharm Des 2022; 28:3202-3211. [PMID: 35422206 DOI: 10.2174/1381612828666220413103831] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/25/2022] [Indexed: 01/28/2023]
Abstract
Several factors exist that limit the efficacy of lung cancer treatment. These may be tumor-specific delivery of therapeutics, airway geometry, humidity, clearance mechanisms, presence of lung diseases, and therapy against tumor cell resistance. Advancements in drug delivery using nanotechnology based multifunctional nanocarriers, have emerged as a viable method for treating lung cancer with more efficacy and fewer adverse effects. This review does a thorough and critical examination of effective nano-enabled approaches for lung cancer treatment, such as nano-assisted drug delivery systems. In addition, to therapeutic effectiveness, researchers have been working to determine several strategies to produce nanotherapeutics by adjusting the size, drug loading, transport, and retention. Personalized lung tumor therapies using sophisticated nano modalities have the potential to provide great therapeutic advantages based on individual unique genetic markers and disease profiles. Overall, this review provides comprehensive information on newer nanotechnological prospects for improving the management of apoptosis in lung cancer.
Collapse
Affiliation(s)
- Monu K Shukla
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan-173229, Himachal Pradesh, India
| | - Amit Dubey
- Computational Chemistry and Drug Discovery Division, Quanta Calculus Pvt. Ltd., Kushinagar-274203, India.,Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 602105, India
| | - Sadanand Pandey
- Department of Chemistry, College of Natural Sciences, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, South Korea
| | - Sachin K Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara-144411, Punjab, India.,Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Gaurav Gupta
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu 602105, India.,School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Jaipur, 302017, India.,Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, 248007, India
| | - Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Dehradun 248007, India
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil 57000, Kuala Lumpur, Malaysia
| | - Brian G Oliver
- School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia.,Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, NSW 2037, Australia
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan-173229, Himachal Pradesh, India
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo NSW 2007, Australia.,Woolcock Institute of Medical Research, University of Sydney, Sydney, New South Wales, NSW 2037, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW 2007, Australia
| |
Collapse
|
33
|
Antiviral Activity against Respiratory Syncytial Virus of Polysaccharide from Jerusalem Artichoke (Helianthus tuberosus L.). BIOMED RESEARCH INTERNATIONAL 2022; 2022:1809879. [PMID: 36193325 PMCID: PMC9526606 DOI: 10.1155/2022/1809879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022]
Abstract
Jerusalem artichoke (Helianthus tuberosus L.) polysaccharide (JAP) is a chain polysaccharide composed of D-fructose connected by β (1-2) glycosidic bonds, which is a kind of inulin. This study evaluated the anti-respiratory syncytial virus (RSV) activity of JAP in vivo and in vitro. To investigate its antiviral activity, an MTT assay, q-PCR, enzyme-linked immunosorbent assay (ELISA), and lung histological observation were performed. The results showed that JAP showed anti-RSV activity in vitro with a half maximal inhibitory concentration (IC50) of approximately 29.15 μg/mL. In vivo results suggested that JAP could effectively inhibit RSV proliferation in the lungs and improve lung tissue lesions in RSV-infected mice. Additionally, JAP could also reduce the expression of TLR3 and TLR4 in the lungs, increase serum anti-inflammatory factors IL-4 levels, and reduce pro-inflammatory factors TNF-α and TNF-β levels, which may be related to its anti-RSV activity. This study provides a new approach to anti-RSV therapy and enriches the potential applications of JAP.
Collapse
|
34
|
Silva IAN, Gvazava N, Bölükbas DA, Stenlo M, Dong J, Hyllen S, Pierre L, Lindstedt S, Wagner DE. A Semi-quantitative Scoring System for Green Histopathological Evaluation of Large Animal Models of Acute Lung Injury. Bio Protoc 2022; 12:e4493. [PMID: 36199700 PMCID: PMC9486691 DOI: 10.21769/bioprotoc.4493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/03/2022] [Accepted: 06/28/2022] [Indexed: 12/29/2022] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a life-threatening, high mortality pulmonary condition characterized by acute lung injury (ALI) resulting in diffuse alveolar damage. Despite progress regarding the understanding of ARDS pathophysiology, there are presently no effective pharmacotherapies. Due to the complexity and multiorgan involvement typically associated with ARDS, animal models remain the most commonly used research tool for investigating potential new therapies. Experimental models of ALI/ARDS use different methods of injury to acutely induce lung damage in both small and large animals. These models have historically played an important role in the development of new clinical interventions, such as fluid therapy and the use of supportive mechanical ventilation (MV). However, failures in recent clinical trials have highlighted the potential inadequacy of small animal models due to major anatomical and physiological differences, as well as technical challenges associated with the use of clinical co-interventions [e.g., MV and extracorporeal membrane oxygenation (ECMO)]. Thus, there is a need for larger animal models of ALI/ARDS, to allow the incorporation of clinically relevant measurements and co-interventions, hopefully leading to improved rates of clinical translation. However, one of the main challenges in using large animal models of preclinical research is that fewer species-specific experimental tools and metrics are available for evaluating the extent of lung injury, as compared to rodent models. One of the most relevant indicators of ALI in all animal models is evidence of histological tissue damage, and while histological scoring systems exist for small animal models, these cannot frequently be readily applied to large animal models. Histological injury in these models differs due to the type and severity of the injury being modeled. Additionally, the incorporation of other clinical support devices such as MV and ECMO in large animal models can lead to further lung damage and appearance of features absent in the small animal models. Therefore, semi-quantitative histological scoring systems designed to evaluate tissue-level injury in large animal models of ALI/ARDS are needed. Here we describe a semi-quantitative scoring system to evaluate histological injury using a previously established porcine model of ALI via intratracheal and intravascular lipopolysaccharide (LPS) administration. Additionally, and owing to the higher number of samples generated from large animal models, we worked to implement a more sustainable and greener histopathological workflow throughout the entire process.
Collapse
Affiliation(s)
- Iran A. N. Silva
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
,
Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
,
Stem Cell Center, Lund University, Lund, Sweden
| | - Nika Gvazava
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
,
Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
,
Stem Cell Center, Lund University, Lund, Sweden
| | - Deniz A. Bölükbas
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
,
Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
,
Stem Cell Center, Lund University, Lund, Sweden
| | - Martin Stenlo
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
,
Stem Cell Center, Lund University, Lund, Sweden
,
Department of Cardiothoracic Anesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
| | - Jiao Dong
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
,
Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
,
Stem Cell Center, Lund University, Lund, Sweden
,
Department of Oral and Maxillofacial Surgery, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Snejana Hyllen
- Department of Cardiothoracic Anesthesia and Intensive Care, Skåne University Hospital, Lund, Sweden
,
Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Leif Pierre
- Department of Cardiothoracic Surgery, Heart and Lung Transplantation, Skåne University Hospital, Lund, Sweden
,
Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Sandra Lindstedt
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
,
Stem Cell Center, Lund University, Lund, Sweden
,
Department of Cardiothoracic Surgery, Heart and Lung Transplantation, Skåne University Hospital, Lund, Sweden
,
Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Darcy E. Wagner
- Lung Bioengineering and Regeneration, Department of Experimental Medical Sciences, Lund University, Lund, Sweden
,
Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
,
Stem Cell Center, Lund University, Lund, Sweden
,
NanoLund, Lund University, Lund, Sweden
,
*For correspondence:
| |
Collapse
|
35
|
Li X, Wei Y, Li S, Liang J, Liu Z, Cui Y, Gao J, Yang Z, Li L, Zhou H, Chen S, Yang C. Zanubrutinib ameliorates lipopolysaccharide-induced acute lung injury via regulating macrophage polarization. Int Immunopharmacol 2022; 111:109138. [PMID: 35973369 DOI: 10.1016/j.intimp.2022.109138] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/31/2022] [Accepted: 08/05/2022] [Indexed: 12/24/2022]
Abstract
Acute lung injury (ALI) is a disease characterized by pulmonary diffusion dysfunction and its exacerbation stage is acute respiratory distress syndrome (ARDS), which may develop to multiple organ failure and seriously threatens human health. ALI has high mortality rates and few effective treatments, thus effective protection measures for ALI are becoming increasingly important. Macrophages play a key regulatory role in the pathogenesis of ALI, and the degree of macrophage polarization is closely related to the severity and prognosis of ALI. In this study, we evaluated the effects of Zanubrutinib (ZB), a BTK small molecule inhibitor approved by the FDA for the treatment of cell lymphoma, on macrophage polarization and acute lung injury. In the in vivo study, we constructed a mouse model of Lipopolysaccharide (LPS)-induced acute lung injury and found that ZB could improve the acute injury of mouse lungs by inhibiting the secretion of proinflammatory factors and promoting the secretion of anti-inflammatory factors, reduce the number of inflammatory cells in alveolar lavage fluid, and then alleviate the inflammatory response. In vivo and in vitro studies have shown that ZB could inhibit the M1 macrophage polarization and promote the M2 macrophage polarization. Subsequent mechanistic studies revealed that ZB could inhibit the macrophage M1 polarization via targeting BTK activation and inhibiting JAK2/STAT1 and TLR4/MyD88/NF-κB signaling pathways, and promote the macrophage M2 polarization by promoting the activation of STAT6 and PI3K / Akt signaling pathways. In summary, ZB has shown therapeutic effect in LPS-induced acute lung injury in mice, which provides a potential candidate drug to treat acute lung injury.
Collapse
Affiliation(s)
- Xiaohe Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China
| | - Yuli Wei
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China
| | - Shimeng Li
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China
| | - Jingjing Liang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China
| | - Zhichao Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China
| | - Yunyao Cui
- Tianjin Jikun Technology Co., Ltd., Tianjin 301700, People's Republic of China
| | - Jingjing Gao
- Tianjin Jikun Technology Co., Ltd., Tianjin 301700, People's Republic of China
| | - Zhongyi Yang
- Tianjin Jikun Technology Co., Ltd., Tianjin 301700, People's Republic of China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, People's Republic of China
| | - Lei Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Tianjin 300192, People's Republic of China
| | - Honggang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China.
| | - Shanshan Chen
- The First Affiliated Hospital of Zhengzhou University, People's Republic of China.
| | - Cheng Yang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Haihe Education Park, 38 Tongyan Road, Tianjin 300353, People's Republic of China; Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin 300457, People's Republic of China.
| |
Collapse
|
36
|
Ni L, Jing S, Zhu L, Yang X, Wang X, Tu S. The Immune Change of the Lung and Bowel in an Ulcerative Colitis Rat Model and the Protective Effect of Sodium Houttuyfonate Combined With Matrine. Front Immunol 2022; 13:888918. [PMID: 35844499 PMCID: PMC9280623 DOI: 10.3389/fimmu.2022.888918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/30/2022] [Indexed: 01/19/2023] Open
Abstract
Objective To explore the immune change of lung injury of Ulcerative colitis (UC) by observing the changes of inherent immunity and adaptive immunity of the lung and bowel in UC rat models after the treatment of Sodium Houttuyfonate combined with Matrine. Method UC rat models were established with the mucous membrane of colon allergize combined with TNBS-alcohol enteroclysis for 1 week and 5 weeks. 1-week experimental rats were divided into normal group and model group, 5/each group. 5-weeks experimental rats were divided into normal group, model group, Sodium Houttuyfonate (2.9mg/ml) combined with Matrine (1.47mg/ml), and positive control sulfasalazine (10mg/ml), 5/each group. All rats were administered by gavage for 5 weeks. The histopathological and fibrotic changes in the lung and bowel were observed, and the expressions of Tumor Necrosis Factor (TNF)- α, interleukin (IL)-8 in the lung, bowel, and serum were detected by radio-immunity and immunohistochemistry, and the mRNA expressions of Toll-like receptor (TLR)-4, nuclear factor kappa (NF-κB), Macrophage migration inhibitory factor (MIF), Mucosal addressing cell adhesion molecule-1 (MadCAM1) and Pulmonary surfactant protein-A (SP-A) in the lung and bowel were detected by Real time-PCR. Result Compared with the normal group, the model rats had significant histopathological and fibrotic changes both in the lung and bowel, and all treatment groups were improved. After treatment, TLR4, IL-8, MIF, and TNF-α in the lung decreased (P<0.05); NF-KB, IL-8, and MIF in the bowel increased (P<0.05); MadCAM1 both in lung and bowel decreased (P<0.05); SP-A decreased in bowel and increased in the lung (P<0.05). Conclusion The cause of lung injury in this model was found to be related to inherent immunity and adaptive immunity, while the cause of bowel injury in this model was found to be mainly related to adaptive immunity. Sodium Houttuyfonate combined with Matrine could improve bowel and lung injury.
Collapse
Affiliation(s)
- Lulu Ni
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Shan Jing
- Department of Internal Medicine of Traditional Chinese Medicine (TCM), Nantong Hospital, Nantong, China
| | - Li Zhu
- Department of Internal Medicine of Traditional Chinese Medicine (TCM), Dong- zhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Yang
- Department of Internal Medicine of Traditional Chinese Medicine (TCM), Third Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China
| | - Xinyue Wang
- Department of Internal Medicine of Traditional Chinese Medicine (TCM), Dong- zhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Su Tu
- Department of Emergency, the Affiliated Wuxi NO 2 People’s Hospital of Nanjing Medical University, Wuxi, China
- *Correspondence: Su Tu,
| |
Collapse
|
37
|
Martín-Medina A, Cerón-Pisa N, Martinez-Font E, Shafiek H, Obrador-Hevia A, Sauleda J, Iglesias A. TLR/WNT: A Novel Relationship in Immunomodulation of Lung Cancer. Int J Mol Sci 2022; 23:6539. [PMID: 35742983 PMCID: PMC9224119 DOI: 10.3390/ijms23126539] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 02/07/2023] Open
Abstract
The most frequent cause of death by cancer worldwide is lung cancer, and the 5-year survival rate is still very poor for patients with advanced stage. Understanding the crosstalk between the signaling pathways that are involved in disease, especially in metastasis, is crucial to developing new targeted therapies. Toll-like receptors (TLRs) are master regulators of the immune responses, and their dysregulation in lung cancer is linked to immune escape and promotes tumor malignancy by facilitating angiogenesis and proliferation. On the other hand, over-activation of the WNT signaling pathway has been reported in lung cancer and is also associated with tumor metastasis via induction of Epithelial-to-mesenchymal-transition (EMT)-like processes. An interaction between both TLRs and the WNT pathway was discovered recently as it was found that the TLR pathway can be activated by WNT ligands in the tumor microenvironment; however, the implications of such interactions in the context of lung cancer have not been discussed yet. Here, we offer an overview of the interaction of TLR-WNT in the lung and its potential implications and role in the oncogenic process.
Collapse
Affiliation(s)
- Aina Martín-Medina
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
| | - Noemi Cerón-Pisa
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
| | - Esther Martinez-Font
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
- Medical Oncology Department, Hospital Universitario Son Espases, 07120 Palma, Spain
| | - Hanaa Shafiek
- Chest Diseases Department, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Antònia Obrador-Hevia
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
- Molecular Diagnosis Unit, Hospital Universitario Son Espases, 07120 Palma, Spain
| | - Jaume Sauleda
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
- Department of Respiratory Medicine, Hospital Universitario Son Espases, 07120 Palma, Spain
- Centro de Investigación Biomédica en Red in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| | - Amanda Iglesias
- Instituto de Investigación Sanitaria de les Illes Balears (IdISBa), 07120 Palma, Spain
- Centro de Investigación Biomédica en Red in Respiratory Diseases (CIBERES), 28029 Madrid, Spain
| |
Collapse
|
38
|
Suzuki M, Cooksley C, Suzuki T, Ramezanpour M, Nakazono A, Nakamaru Y, Homma A, Vreugde S. TLR Signals in Epithelial Cells in the Nasal Cavity and Paranasal Sinuses. FRONTIERS IN ALLERGY 2022; 2:780425. [PMID: 35387020 PMCID: PMC8974762 DOI: 10.3389/falgy.2021.780425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/01/2021] [Indexed: 12/24/2022] Open
Abstract
The respiratory tract is constantly at risk of invasion by microorganisms such as bacteria, viruses, and fungi. In particular, the mucosal epithelium of the nasal cavity and paranasal sinuses is at the very forefront of the battles between the host and the invading pathogens. Recent studies have revealed that the epithelium not only constitutes a physical barrier but also takes an essential role in the activation of the immune system. One of the mechanisms equipped in the epithelium to fight against microorganisms is the Toll-like receptor (TLR) response. TLRs recognize common structural components of microorganisms and activate the innate immune system, resulting in the production of a plethora of cytokines and chemokines in the response against microbes. As the epithelia-derived cytokines are deeply involved in the pathogenesis of inflammatory conditions in the nasal cavity and paranasal sinuses, such as chronic rhinosinusitis (CRS) and allergic rhinitis (AR), the molecules involved in the TLR response may be utilized as therapeutic targets for these diseases. There are several differences in the TLR response between nasal and bronchial epithelial cells, and knowledge of the TLR signals in the upper airway is sparse compared to that in the lower airway. In this review, we provide recent evidence on TLR signaling in the upper airway, focusing on the expression, regulation, and responsiveness of TLRs in human nasal epithelial cells (HNECs). We also discuss how TLRs in the epithelium are involved in the pathogenesis of, and possible therapeutic targeting, for CRS and AR.
Collapse
Affiliation(s)
- Masanobu Suzuki
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Clare Cooksley
- Department of Surgery-Otorhinolaryngology Head and Neck Surgery, Central Adelaide Local Health Network and the University of Adelaide, Adelaide, SA, Australia
| | - Takayoshi Suzuki
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mahnaz Ramezanpour
- Department of Surgery-Otorhinolaryngology Head and Neck Surgery, Central Adelaide Local Health Network and the University of Adelaide, Adelaide, SA, Australia
| | - Akira Nakazono
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yuji Nakamaru
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Homma
- Department of Otolaryngology-Head and Neck Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Sarah Vreugde
- Department of Surgery-Otorhinolaryngology Head and Neck Surgery, Central Adelaide Local Health Network and the University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
39
|
Chen M, Deng H, Zhao Y, Miao X, Gu H, Bi Y, Zhu Y, Guo Y, Shi S, Xu J, Zhao D, Liu F. Toll-Like Receptor 2 Modulates Pulmonary Inflammation and TNF-α Release Mediated by Mycoplasma pneumoniae. Front Cell Infect Microbiol 2022; 12:824027. [PMID: 35372108 PMCID: PMC8968444 DOI: 10.3389/fcimb.2022.824027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Objectives To investigate the roles that Toll-like receptors (TLRs) play in lung inflammation mediated by Mycoplasma pneumoniae (MP). Methods The changes in TLRs and tumor necrosis factor alpha (TNF-α) in peripheral blood of children with M. pneumoniae pneumonia (MPP) were monitored, and the interactions of signaling molecules regulating TNF-α release in A549 cells and neutrophils after M. pneumoniae stimulation were investigated. In TLR2 knockout (TLR2-/-) mice, the levels of TNF-α in bronchial alveolar lavage fluid (BALF) and peripheral blood after mycoplasma infection and the pathological changes in the lung tissue of mice were detected. Results TNF-α levels in peripheral blood of children with MPP were higher than those in non-infected children, and children with refractory MPP had the highest levels of TNF-α and TLR2. TNF-α secretion and TLR2, myeloid differentiation primary response 88 (MyD88) and phospho-p65(p-p65) levels were increased in stimulated cells. TNF-α secretion was suppressed upon siRNA-mediated TLR2 silencing. Pharmacological inhibition of nuclear factor-kappa B (NF-κB) and MyD88 effectively reduced TNF-α expression. Compared with wild-type mice, the TNF-α in serum and BALF decreased, and lung pro-inflammatory response was partially suppressed in TLR2-/- mice. Conclusion We concluded that TLR2 regulates M. pneumoniae-mediated lung inflammation and TNF-α release through the TLR2-MyD88-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ming Chen
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Department of Respiratory Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Huan Deng
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yue Zhao
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Xueqing Miao
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Haiyan Gu
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Bi
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yifan Zhu
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yun Guo
- Department of Respiratory Medicine, The Affiliated Wuxi Children’s Hospital of Nanjing Medical University, Wuxi, China
| | - Shuang Shi
- Department of Respiratory Medicine, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jiejing Xu
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Deyu Zhao
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Deyu Zhao, ; Feng Liu,
| | - Feng Liu
- Department of Respiratory Medicine, Children’s Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Deyu Zhao, ; Feng Liu,
| |
Collapse
|
40
|
HD-13 Induces Swine Pneumonia Progression via Activation of TLR9. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:8660752. [PMID: 35132333 PMCID: PMC8817872 DOI: 10.1155/2022/8660752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/12/2021] [Accepted: 12/16/2021] [Indexed: 12/25/2022]
Abstract
Swine pneumonia commonly known as swine pasteurellosis is an infectious disease of swine caused by Pasteurella multocida infection. It has been reported that Toll-like receptors (TLRs) play a vital role in swine pneumonia progression. However, the underlying mechanism has not been elucidated. This research was aimed at investigating the molecular mechanism by which TLR9 regulates swine pneumonia progression. Our findings illustrated that the HD-13 strain of Pasteurella multocida D (HD-13) accelerated TLR9 expression in porcine alveolar macrophage 3D4/21 cells; HD-13 activated the inflammatory response via accelerating TLR9 expression. Mechanistically, HD-13 activated mitogen-activated protein kinase (MAPK) and nuclear factor kB (NF-κB) signals. In conclusion, HD-13 may activate MAPK and NF-κB pathways via accelerating TLR9 expression, thereby accelerating the inflammatory response in the progression of swine pneumonia. TLR9 may serve as a novel therapeutic target for swine pneumonia. Our research may provide a theoretical basis for the prevention and treatment of swine pneumonia.
Collapse
|
41
|
The role of immunotherapy in treating lung cancer: current status and future perspective. FUTURE DRUG DISCOVERY 2021. [DOI: 10.4155/fdd-2021-0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Lung cancers have the worst incident and mortality rates. Cancers such as advanced non-small-cell lung carcinomas are inoperable and often the only treatment available is chemo-radiotherapy. There has been little improvement in long-term survival recently, prompting research into novel treatments. Immune checkpoint inhibitors (ICIs) are a form of immunotherapy used in lung cancer. The efficacy of ICIs is dependent on: the part of the pathway affected; the presence of prognostic biomarkers; the method of efficacy assessment; the stage of the disease and other drugs involved. Monoclonal antibodies, Toll-like receptor agonists and cancer vaccines have shown modest effects on survival. Refinement of treatment regimens and prognostic biomarkers will help improve the survival of patients in the future.
Collapse
|
42
|
Li A, Chan HP, Gan PX, Liew MF, Wong WF, Lim HF. Eosinophilic endotype of chronic obstructive pulmonary disease: similarities and differences from asthma. Korean J Intern Med 2021; 36:1305-1319. [PMID: 34634855 PMCID: PMC8588979 DOI: 10.3904/kjim.2021.180] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/01/2021] [Indexed: 11/27/2022] Open
Abstract
Approximately 25% to 40% of patients with chronic obstructive pulmonary disease (COPD) have the eosinophilic endotype. It is important to identify this group accurately because they are more symptomatic and are at increased risk for exacerbations and accelerated decline in forced expiratory volume in the 1st second. Importantly, this endotype is a marker of treat ment responsiveness to inhaled corticosteroid (ICS), resulting in decreased mortality risk. In this review, we highlight differences in the biology of eosinophils in COPD compared to asthma and the different definitions of the COPD eosinophilic endotype based on sputum and blood eosinophil count (BEC) with the corresponding limitations. Although BEC is useful as a biomarker for eosinophilic COPD endotype, optimal BEC cut-offs can be combined with clinical characteristics to improve its sensitivity and specificity. A targeted approach comprising airway eosinophilia and appropriate clinical and physiological features may improve identification of subgroups of patients who would benefit from biologic therapy or early use of ICS for disease modification.
Collapse
Affiliation(s)
- Andrew Li
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Hospital, National University Health System,
Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore,
Singapore
| | - Hiang Ping Chan
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Hospital, National University Health System,
Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore,
Singapore
| | - Phyllis X.L. Gan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System,
Singapore
- Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore,
Singapore
| | - Mei Fong Liew
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Hospital, National University Health System,
Singapore
- FAST and Chronic Programmes, Alexandra Hospital, National University Health System,
Singapore
| | - W.S. Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System,
Singapore
- Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore,
Singapore
| | - Hui-Fang Lim
- Division of Respiratory and Critical Care Medicine, Department of Medicine, National University Hospital, National University Health System,
Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore,
Singapore
| |
Collapse
|
43
|
Danielsen PH, Bendtsen KM, Knudsen KB, Poulsen SS, Stoeger T, Vogel U. Nanomaterial- and shape-dependency of TLR2 and TLR4 mediated signaling following pulmonary exposure to carbonaceous nanomaterials in mice. Part Fibre Toxicol 2021; 18:40. [PMID: 34717665 PMCID: PMC8557558 DOI: 10.1186/s12989-021-00432-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 10/12/2021] [Indexed: 12/18/2022] Open
Abstract
Background Pulmonary exposure to high doses of engineered carbonaceous nanomaterials (NMs) is known to trigger inflammation in the lungs paralleled by an acute phase response. Toll-like receptors (TLRs), particularly TLR2 and TLR4, have recently been discussed as potential NM-sensors, initiating inflammation. Using Tlr2 and Tlr4 knock out (KO) mice, we addressed this hypothesis and compared the pattern of inflammation in lung and acute phase response in lung and liver 24 h after intratracheal instillation of three differently shaped carbonaceous NMs, spherical carbon black (CB), multi-walled carbon nanotubes (CNT), graphene oxide (GO) plates and bacterial lipopolysaccharide (LPS) as positive control.
Results The LPS control confirmed a distinct TLR4-dependency as well as a pronounced contribution of TLR2 by reducing the levels of pulmonary inflammation to 30 and 60% of levels in wild type (WT) mice. At the doses chosen, all NM caused comparable neutrophil influxes into the lungs of WT mice, and reduced levels were only detected for GO-exposed Tlr2 KO mice (35%) and for CNT-exposed Tlr4 KO mice (65%). LPS-induced gene expression was strongly TLR4-dependent. CB-induced gene expression was unaffected by TLR status. Both GO and MWCNT-induced Saa1 expression was TLR4-dependent. GO-induced expression of Cxcl2, Cxcl5, Saa1 and Saa3 were TLR2-dependent. NM-mediated hepatic acute phase response in terms of liver gene expression of Saa1 and Lcn2 was shown to depend on TLR2 for all three NMs. TLR4, in contrast, was only relevant for the acute phase response caused by CNTs, and as expected by LPS. Conclusion TLR2 and TLR4 signaling was not involved in the acute inflammatory response caused by CB exposure, but contributed considerably to that of GO and CNTs, respectively. The strong involvement of TLR2 in the hepatic acute phase response caused by pulmonary exposure to all three NMs deserves further investigations. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-021-00432-z.
Collapse
Affiliation(s)
| | | | | | - Sarah Søs Poulsen
- National Research Centre for the Working Environment, Copenhagen, Denmark
| | - Tobias Stoeger
- Comprehensive Pneumology Center (CPC)/Institute of Lung Biology and Disease (ILBD) Helmholtz Zentrum München, Neuherberg, Germany
| | - Ulla Vogel
- National Research Centre for the Working Environment, Copenhagen, Denmark. .,DTU Food, Technical University of Denmark, Kgs. Lyngby, Denmark.
| |
Collapse
|
44
|
Hernandez-Ramirez G, Barber D, Tome-Amat J, Garrido-Arandia M, Diaz-Perales A. Alternaria as an Inducer of Allergic Sensitization. J Fungi (Basel) 2021; 7:jof7100838. [PMID: 34682259 PMCID: PMC8539034 DOI: 10.3390/jof7100838] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 01/02/2023] Open
Abstract
Alternaria alternata is a saprophytic mold whose spores are disseminated in warm dry air, the typical weather of the Mediterranean climate region (from 30° to 45°), with a peak during the late summer and early autumn. Alternaria spores are known to be biological contaminants and a potent source of aeroallergens. One consequence of human exposure to Alternaria is an increased risk of developing asthma, with Alt a 1 as its main elicitor and a marker of primary sensitization. Although the action mechanism needs further investigation, a key role of the epithelium in cytokine production, TLR-activated alveolar macrophages and innate lymphoid cells in the adaptive response was demonstrated. Furthermore, sensitization to A. alternata seems to be a trigger for the development of co-sensitization to other allergen sources and may act as an exacerbator of symptoms and an elicitor of food allergies. The prevalence of A. alternata allergy is increasing and has led to expanding research on the role of this fungal species in the induction of IgE-mediated respiratory diseases. Indeed, recent research has allowed new perspectives to be considered in the assessment of exposure and diagnosis of fungi-induced allergies, although more studies are needed for the standardization of immunotherapy formulations.
Collapse
Affiliation(s)
- Guadalupe Hernandez-Ramirez
- Centro de Biotecnología Y Genómica de Plantas (CBGP, UPM-INIA), Universidad Politécnica de Madrid (UPM), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28223 Madrid, Spain; (G.H.-R.); (J.T.-A.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain
| | - Domingo Barber
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Instituto de Medicina Molecular Aplicada (IMMA), Universidad San Pablo CEU, CEU Universities, 28925 Madrid, Spain;
| | - Jaime Tome-Amat
- Centro de Biotecnología Y Genómica de Plantas (CBGP, UPM-INIA), Universidad Politécnica de Madrid (UPM), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28223 Madrid, Spain; (G.H.-R.); (J.T.-A.); (M.G.-A.)
| | - Maria Garrido-Arandia
- Centro de Biotecnología Y Genómica de Plantas (CBGP, UPM-INIA), Universidad Politécnica de Madrid (UPM), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28223 Madrid, Spain; (G.H.-R.); (J.T.-A.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain
| | - Araceli Diaz-Perales
- Centro de Biotecnología Y Genómica de Plantas (CBGP, UPM-INIA), Universidad Politécnica de Madrid (UPM), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), 28223 Madrid, Spain; (G.H.-R.); (J.T.-A.); (M.G.-A.)
- Departamento de Biotecnología-Biología Vegetal, Escuela Técnica Superior de Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid (UPM), 28040 Madrid, Spain
- Correspondence:
| |
Collapse
|
45
|
Liu G, Lu Y, Shi L, Kong J, Hu H, Liu W. Trace endotoxin in reclaimed water is only one of the risk sources in subchronic inhalation exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 285:117479. [PMID: 34090073 DOI: 10.1016/j.envpol.2021.117479] [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: 03/23/2021] [Revised: 05/12/2021] [Accepted: 05/25/2021] [Indexed: 06/12/2023]
Abstract
Pulmonary injury and inflammation have been detected in cases of subchronic inhalation exposure to reclaimed water, but the mechanism remains unclear. Endotoxin has been verified as the key risk factor in acute inhalation exposure through the TLR4 (Toll-like receptor 4) signalling pathway. In long-term exposure, endotoxin may also be a risk factor in reclaimed water, but the contribution of other health risk factors should not be underestimated. Wild-type C57BL/6J and TLR4-signalling-pathway-defective mice were used in this study to assess the risk of subchronic inhalation exposure to reclaimed water. Two types of reclaimed water, i.e., secondary effluent and MBR (membrane bioreactor) effluent, were found to induce pulmonary injury and inflammation in the wild-type mice and Tlr4-/- mutants. However, when both mice were exposed to the same concentrations of pure endotoxin in reclaimed water, only the wild-type mice that were treated with high-dose endotoxin showed pulmonary injury. In summary, non-TLR4 signalling pathways are related to lung inflammation caused by long-term exposure to reclaimed water. It is highly possible that pollutants in addition to endotoxin in the reclaimed water can induce chronic inflammation in the lung.
Collapse
Affiliation(s)
- Gang Liu
- State Environment Protection Key Laboratory of Microorganism Application and Rish Control, School of Environment, Tsinghua University, Beijing, 100084, China
| | - Yun Lu
- State Environment Protection Key Laboratory of Microorganism Application and Rish Control, School of Environment, Tsinghua University, Beijing, 100084, China.
| | - Liangliang Shi
- State Environment Protection Key Laboratory of Microorganism Application and Rish Control, School of Environment, Tsinghua University, Beijing, 100084, China
| | - Jiayang Kong
- State Environment Protection Key Laboratory of Microorganism Application and Rish Control, School of Environment, Tsinghua University, Beijing, 100084, China
| | - Hongying Hu
- State Environment Protection Key Laboratory of Microorganism Application and Rish Control, School of Environment, Tsinghua University, Beijing, 100084, China
| | - Wanli Liu
- School of Life Science, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
46
|
First case of low-dose umbilical cord blood therapy for pediatric acute respiratory distress syndrome induced by Pneumocystis carinii pneumonia. Eur J Med Res 2021; 26:100. [PMID: 34454624 PMCID: PMC8397599 DOI: 10.1186/s40001-021-00548-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/07/2021] [Indexed: 01/02/2023] Open
Abstract
Objective This study aimed to present the case of a boy with acute distress syndrome (ARDS) treated with low-dose umbilical cord blood (UCB) therapy and explore the underlying possible mechanism. Methods A 7-year-old boy with severe Pneumocystis carinii pneumonia and severe ARDS was treated with allogeneic UCB as salvage therapy. Results The patient did not improve after being treated with lung protective ventilation, pulmonary surfactant replacement, and extracorporeal membrane oxygenation (ECMO) for 30 days. However, his disease reversed 5 days after allogeneic UCB infusion, and he weaned from ECMO after 7 days of infusion. Bioinformatics confirmed that his Toll-like receptor (TLR) was abnormal before UCB infusion. However, after the infusion, his immune system was activated and repaired, and the TLR4/MyD88/NF-κB signaling pathway was recovered. Conclusion Allogenic UCB could treat ARDS by repairing the TLR4/MyD88/NF-κB signaling pathway, thereby achieving stability of the immune system.
Collapse
|
47
|
Role of Toll-Like Receptor 2 in Regulation of T-Helper Immune Response in Chronic Obstructive Pulmonary Disease. Can Respir J 2021; 2021:5596095. [PMID: 34426754 PMCID: PMC8380179 DOI: 10.1155/2021/5596095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/29/2021] [Accepted: 08/10/2021] [Indexed: 11/23/2022] Open
Abstract
Objective According to modern views, the differences in the clinical course of chronic obstructive pulmonary disease (COPD) are associated with certain types of T-helper (Th) immune response. Recent data have shown that toll-like receptor 2 (TLR2) is involved in the development of Th immune response. However, TLR2-mediated regulation of Th subpopulation balance in COPD needs to be elucidated. The aim of our work is to determine the mechanisms of TLR2-mediated regulation of Th immune response in COPD of varying severity. Methods The study included 323 smokers/ex-smokers with stable COPD (GOLD I, GOLD II, and GOLD III) and 97 healthy nonsmokers (control group). Serum levels of Th1 (TNF-α and IFN-γ), Th2 (IL-4), Th17 (IL-6 and IL-17A), Treg (IL-10) cytokines, and the percentage of peripheral blood Th cells expressing TLR2 (CD4+CD282+) were assessed by flow cytometry. Serum concentrations of IL-21 (Th17) and TGF-β1 (Treg) were measured using the ELISA method. The predominant Th cytokine profile in serum was determined by calculating the ratios between levels of Th1 and Th17 cytokines. Spearman's correlation test was performed. Results Patients with COPD GOLD II and III with Th1 and Th17 cytokine profiles exhibited an increase in the percentage of CD4+CD282+ cells compared to the control group. In COPD GOLD I–III, positive correlations between CD4+CD282+ cell frequency and Th17 cytokine levels (IL-6, IL-17A, and IL-21) were found. In COPD GOLD I, IL-10 concentration was negatively correlated with the percentages of studied cells; in COPD GOLD II, a positive correlation between these parameters was noted. Conclusions Enhanced TLR2 expression on CD4+ cells shifts cytokine profile toward Th17 phenotype that plays a crucial role in COPD progression. The level of TLR2 expression on peripheral blood CD4+ cells may be considered as a biomarker for diagnosing and predicting the progression of COPD.
Collapse
|
48
|
Arora S, Singh P, Ahmad S, Ahmad T, Dohare R, Almatroodi SA, Alrumaihi F, Rahmani AH, Syed MA. Comprehensive Integrative Analysis Reveals the Association of KLF4 with Macrophage Infiltration and Polarization in Lung Cancer Microenvironment. Cells 2021; 10:cells10082091. [PMID: 34440860 PMCID: PMC8392240 DOI: 10.3390/cells10082091] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 01/05/2023] Open
Abstract
Macrophage polarization and infiltration to the tumor microenvironment (TME) is a critical determining factor for tumor progression. Macrophages are polarized into two states—M1 (pro-inflammatory, anti-tumorigenic and stimulated by LPS or IFN-γ) and M2 (anti-inflammatory pro-tumorigenic and stimulated by IL-4) phenotypes. Specifically, M2 macrophages enhance tumor cell growth and survival. Recent evidences suggest the pivotal role of microRNAs in macrophage polarization during the development of Non-small cell lung cancer (NSCLC), thus proposing a new therapeutic option to target lung cancer. In silico analysis determined cogent upregulation of KLF4, downregulation of IL-1β and miR-34a-5p in NSCLC tissues, consequently worsening the overall survival of NSCLC patients. We observed a significant association of KLF4 with macrophage infiltration and polarization in NSCLC. We found that KLF4 is critically implicated in M2 polarization of macrophages, which, in turn, promotes tumorigenesis. KLF4 expression correlated with miR-34a-5p and IL-1β in a feed-forward loop (FFL), both of which are implicated in immune regulation. Mechanistic overexpression of miR-34a-5p in macrophages (IL-4 stimulated) inhibits KLF4, along with downregulation of ARG1, REL-1MB (M2 macrophage specific markers), and upregulation of IL-1β, IL-6, (M1 macrophage specific markers), demonstrating macrophage polarization switch from M2 to M1 phenotype. Moreover, co-culture of these macrophages with NSCLC cells reduces their proliferation, wound healing, clonogenic capacity and enhanced NO-mediated apoptosis. Further, transfection of miR-34a-5p in NSCLC cells, also degrades KLF4, but enhances the expression of KLF4 regulated genes—IL-1β, IL-6 (pro-inflammatory mediators), which is further enhanced upon co-culture with IL-4 stimulated macrophages. Additionally, we observed a significant increase in i-NOS/NO content upon co-culture, suggesting polarization reversion of macrophages from M2 to M1, and eventually leading to anti-tumor effects. Our findings thus show a significant role of KLF4 in tumorigenesis and TAM polarization of NSCLC. However, miR-34a-5p mediated targeting of these molecular networks will provide a better therapeutic intervention for NSCLC.
Collapse
Affiliation(s)
- Shweta Arora
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Prithvi Singh
- Centre for Interdisciplinary Research in Basic Sciences, Srinivasa Ramanujan Block, Jamia Millia Islamia, New Delhi 110025, India
| | - Shaniya Ahmad
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advance Research and Studies, Jamia Millia Islamia, New Delhi 110025, India
| | - Ravins Dohare
- Centre for Interdisciplinary Research in Basic Sciences, Srinivasa Ramanujan Block, Jamia Millia Islamia, New Delhi 110025, India
| | - Saleh A Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Mansoor Ali Syed
- Translational Research Lab, Department of Biotechnology, Faculty of Natural Sciences, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
49
|
Riikonen R, Korppi M, Törmänen S, Koponen P, Nuolivirta K, Helminen M, He Q, Lauhkonen E. Risk factors for irreversible airway obstruction after infant bronchiolitis. Respir Med 2021; 187:106545. [PMID: 34332337 DOI: 10.1016/j.rmed.2021.106545] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 06/03/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Increasing evidence shows that environmental factors in childhood play a role in development of irreversible airway obstruction. We evaluated early-life and preschool-age risk factors for irreversible airway obstruction in adolescence after bronchiolitis in infancy. METHODS This study is a secondary analysis of data collected during prospective long-term follow-up of our post-bronchiolitis cohort. Risk factor data were collected during hospitalisation and on follow-up visits at 5-7 and 10-13 years of ages. Lung function was measured from 103 participants with impulse oscillometry at 5-7 years of age and from 89 participants with flow-volume spirometry at 10-13 years of age. RESULTS Asthma diagnosis at <12 months of age showed a significant association with irreversible airway obstruction at 10-13 years of age independently from current asthma. Irreversible airway obstruction was less frequent in children with variant than wild genotype of the Toll-like receptor 4(TLR4) rs4986790, but the significance was lost in logistic regression adjusted for current asthma and weight status. Higher post-bronchodilator respiratory system resistance at 5 Hz and lower baseline and post-bronchodilator reactance at 5 Hz by impulse oscillometry at 5-7 years of age were associated with irreversible airway obstruction at 10-13 years of age. CONCLUSION Asthma diagnosis during the first living year and worse lung function at preschool age increased the risk for irreversible airway obstruction at 10-13 years of age after bronchiolitis. TLR4 rs4986790 polymorphism may be protective for development of irreversible airway obstruction after bronchiolitis.
Collapse
Affiliation(s)
- Riikka Riikonen
- Center for Child Health Research, Tampere University and University Hospital, Tampere, Finland.
| | - Matti Korppi
- Center for Child Health Research, Tampere University and University Hospital, Tampere, Finland
| | - Sari Törmänen
- Center for Child Health Research, Tampere University and University Hospital, Tampere, Finland
| | - Petri Koponen
- Center for Child Health Research, Tampere University and University Hospital, Tampere, Finland
| | - Kirsi Nuolivirta
- Department of Paediatrics, Seinäjoki Central Hospital, Seinäjoki, Finland
| | - Merja Helminen
- Department of Paediatrics, Tampere University Hospital, Tampere, Finland
| | - Qiushui He
- Institute of Biomedicine, University of Turku, Turku, Finland; Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Eero Lauhkonen
- Center for Child Health Research, Tampere University and University Hospital, Tampere, Finland; Department of Paediatrics, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
50
|
Zhang Y, Wang R, Shi W, Zheng Z, Wang X, Li C, Zhang S, Zhang P. Antiviral effect of fufang yinhua jiedu (FFYH) granules against influenza A virus through regulating the inflammatory responses by TLR7/MyD88 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 275:114063. [PMID: 33813013 PMCID: PMC9759603 DOI: 10.1016/j.jep.2021.114063] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 05/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fufang-Yinhua-Jiedu Granules (FFYH) optimized from a Yin-Qiao-San, as traditional Chinese medicine (TCM), was used to treat influenza and upper respiratory tract infection and was recommended for the prevention and treatment of SARS in 2003 and current COVID-19 in Anhui Province in 2020. AIM OF STUDY In the clinical studies, FFYH was very effective for the treatment of influenza, but the mechanism of action against influenza A virus remains unclear. In the present study, we investigated the antiviral effect of FFYH against influenza A virus in vitro and vivo. Moreover, the potential mechanism of FFYH against influenza A virus in vivo was investigated for the first time. MATERIALS AND METHODS CPE inhibition assay and HA assay were used to evaluate the in vitro antiviral effects of FFYH against influenza A virus H1N1, H3N2, H5N1, H7N9 and H9N2. Mice were used to evaluate the antiviral effect of FFYH in vivo with ribavirin and lianhuaqingwen as positive controls. RT-PCR was used to quantify the mRNA transcription of TNF-α, IL-6, IFN-γ, IP10, and IL-1β mRNA. ELISA was used to examine the expression of inflammatory factors such as TNF-α, IL-6, IFN-γ, IP10, and IL-1β in sera. The blood parameters were analyzed with auto hematology analyzer. Moreover, the potential mechanism of FFYH against influenza A virus in vivo was also investigated. RESULTS FFYH showed a broad-spectrum of antiviral activity against H1N1, H3N2, H5N1, H7N9, and H9N2 influenza A viruses. Furthermore, FFYH dose-dependently increased the survival rate, significantly prolonged the median survival time of mice, and markedly reduced lung injury caused by influenza A virus. Also, FFYH significantly improve the sick signs, food taken, weight loss, blood parameters, lung index, and lung pathological changes. Moreover, FFYH could markedly inhibit the inflammatory cytokine expression of TNF-α, IL-6, IFN-γ, IP10, IL-10, and IL-1β mRNA or protein via inhibition of the TLR7/MyD88/NF-κB signaling pathway in vivo. CONCLUSION FFYH not only showed a broad-spectrum of anti-influenza virus activity in vitro, but also exhibited a significant protective effect against lethal influenza virus infection in vivo. Furthermore, our results indicated that the in vivo antiviral effect of FFYH against influenza virus may be attributed to suppressing the expression of inflammatory cytokines via regulating the TLR7/MyD88/NF-κB signaling pathway. These findings provide evidence for the clinical treatment of influenza A virus infection with FFYH.
Collapse
Affiliation(s)
- Yuqian Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Ronghua Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Weiqing Shi
- Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, 210009, China
| | - Zhihui Zheng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Xiaoquan Wang
- College of Veterinary Medicine & Jiangsu Provincial Key Laboratory of Human Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Cheng Li
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Shuofeng Zhang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Pinghu Zhang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China; College of Veterinary Medicine & Jiangsu Provincial Key Laboratory of Human Zoonosis, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|